1
|
Tian R, Song H, Li J, Yuan T, Liu J, Wang Y, Li Y, Song X. PINCH-1 promotes tumor growth and metastasis by enhancing DRP1-mediated mitochondrial fission in head and neck squamous cell carcinoma. Cancer Biol Ther 2025; 26:2477365. [PMID: 40065703 PMCID: PMC11901378 DOI: 10.1080/15384047.2025.2477365] [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: 10/16/2024] [Revised: 02/25/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
PURPOSE Abnormal expression of PINCH-1 has been observed in various types of human cancers. However, the clinical importance and mechanism underlying its role in head and neck squamous cell carcinoma (HNSCC) is yet to be fully elucidated. METHODS This study evaluated the expression of PINCH-1 in HNSCC samples through immunohistochemical staining and Western blotting. AMC-HN-8, Cal27, and SCC7 cell lines were utilized for cellular function experiments, both in vivo and in vitro. CCK8, colony-formation assay, flow cytometry, wound-healing assay, and transwell assay were employed to investigate the effects of alterations in target proteins on the growth and metastasis of cancer cells. Mito-Tracker Deep Red FM was used to track mitochondrial morphology. RESULTS PINCH-1 was found to be overexpressed in HNSCC and closely associated with lymph node metastasis and poor pathologic differentiation. Its upregulation promoted proliferation, inhibited apoptosis, and enhanced migration and invasion in HNSCC cells. It also promoted mitochondrial fission. We conducted a mechanism analysis, which showed that PINCH-1 knockdown inhibited mitochondrial fission by reducing the expression of DRP1. Furthermore, inhibition of mitochondrial fission could impede the proliferation and metastasis of HNSCC cells. Re-expression of DRP1 reversed the inhibitory effect of PINCH-1 knockdown on mitochondrial fission, cell proliferation, and metastasis in HNSCC cells. CONCLUSIONS PINCH-1 plays a critical oncogenic role in HNSCC by enhancing DRP1-mediated mitochondrial fission, which may serve as a novel therapeutic target for HNSCC.
Collapse
Affiliation(s)
- Ruxian Tian
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Hao Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Jiaxuan Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Ting Yuan
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Jiahui Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yaqi Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Yumei Li
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, Shandong, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai, Shandong, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai, Shandong, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai, Shandong, China
| |
Collapse
|
2
|
Xu Y, Wang T, Liang X, Yang J, Zhang Y, Bao S. Global research trends and focus on immunotherapy for endometrial cancer: a comprehensive bibliometric insight and visualization analysis (2012-2024). Front Immunol 2025; 16:1571800. [PMID: 40264788 PMCID: PMC12011754 DOI: 10.3389/fimmu.2025.1571800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/24/2025] [Indexed: 04/24/2025] Open
Abstract
Background This study conducted a novel systematic bibliometric and visualization analysis of global literature on immunotherapy for endometrial cancer (EC) to explore dynamic trends, research hotspots, and emerging topics, providing valuable references for future research. Methods Articles and reviews on EC immunotherapy published between 2012 and August 2024 were retrieved from the Web of Science Core Collection (WoSCC). Bibliometric tools, CiteSpace and VOSviewer, were used to analyze clustering patterns and research dynamics. Results A total of 861 articles were contributed by 5,331 authors from 1,392 institutions across 58 countries or regions, involving 1,823 keywords. China demonstrated outstanding performance in this field, contributing over 40% of the total publications and ranking first in publication volume. However, the total citation counts for publications from China lags that of the United States, highlighting the latter's leading position and areas for further improvement in China's research efforts. The University of Texas Medical Anderson Cancer Center and Nanjing Medical University were the two institutions with the highest number of publications. In terms of authorship, research teams led by Bosse, Tjalling, and Creutzberg, Carien L made significant contributions to advancing the field. Among individual publications, the work by Talhouk et al. achieved the highest average annual citation count of 70.88, demonstrating its profound impact. In terms of journals, Gynecologic Oncology emerged as a pivotal academic platform, publishing numerous articles and achieving the highest co-citation frequency. Additionally, Frontiers in Oncology, Frontiers in Immunology, and Frontiers in Genetics have become some of the most active and rapidly developing journals in recent years. Research hotspots are concentrated on themes such as the "Tumor Immune Microenvironment", "Immune Checkpoint Inhibitors", and "Targeted Therapy". Recent trends and frontier research focus on the combined application of immune checkpoint inhibitors with other therapies, research on the application of nanotechnology in immunotherapy, and the integration of artificial intelligence to enhance precision medicine. Additionally, efforts are increasingly directed toward advancing various immunotherapy strategies from basic research to clinical applications. Conclusions This comprehensive analysis reveals rapid advancements and significant potential in EC immunotherapy. Strengthening international collaboration and addressing barriers in the translation of research to clinical practice will drive further progress in this promising field.
Collapse
Affiliation(s)
- Yachen Xu
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Tao Wang
- School of Public Health, Hainan Medical University, Haikou, China
| | - Xiaojing Liang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Jie Yang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Yuxiang Zhang
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| | - Shan Bao
- Department of Gynecology and Obstetrics, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
- Key Laboratory of Reproductive Health Diseases Research and Translation (Hainan Medical University), Ministry of Education, Haikou, China
- Hainan Provincial Key Laboratory for Human Reproductive Medicine and Genetic Research, The First Affiliated Hospital of Hainan Medical University, Hainan Medical University, Haikou, China
- Medical Laboratory Center, Hainan Affiliated Hospital of Hainan Medical University, Hainan General Hospital, Haikou, China
| |
Collapse
|
3
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [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: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
4
|
Pei Y, Liang H, Guo Y, Wang B, Wu H, Jin Z, Lin S, Zeng F, Wu Y, Shi Q, Xu J, Huang Y, Ren T, Liu J, Guo W. Liquid-liquid phase separation drives immune signaling transduction in cancer: a bibliometric and visualized study from 1992 to 2024. Front Oncol 2025; 15:1509457. [PMID: 40104511 PMCID: PMC11913689 DOI: 10.3389/fonc.2025.1509457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Accepted: 01/28/2025] [Indexed: 03/20/2025] Open
Abstract
Background Liquid-liquid phase separation (LLPS) is a novel concept that could explain how living cells precisely modulate internal spatial and temporal functions. However, a comprehensive bibliometric analysis on LLPS and immune signaling processes in cancer is still scarce. This study aims to perform a bibliometric assessment of research to explore the landscape of LLPS research in immune signaling pathways for cancer. Methods Utilizing the Web of Science Core Collection database and multiple analysis software, we performed quantitative and qualitative analyses of the study situation between LLPS and immune signaling in cancer from 1992 to 2024. Results The corresponding authors were primarily from China and the USA. The most relevant references were the "International Journal of Molecular Sciences", "Proteomics". The annual number of publications exhibited a fast upward tendency from 2020 to 2024. The most frequent key terms included expression, separation, activation, immunotherapy, and mechanisms. Qualitative evaluation emphasized the TCR, BCR, cGAS-STING, RIG-1, NF-κB signaling pathways associated with LLPS processes. Conclusion This research is the first to integratively map out the knowledge structure and forward direction in the area of immune transduction linked with LLPS over the past 30 years. In summary, although this research area is still in its infancy, illustrating the coordinated structures and communications between cancer and immune signaling with LLPS within a spatial framework will offer deeper insights into the molecular mechanisms of cancer development and further enhance the effectiveness of existing immunotherapies.
Collapse
Affiliation(s)
- Yanhong Pei
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Haijie Liang
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Yu Guo
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Boyang Wang
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Han Wu
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Zhijian Jin
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Shanyi Lin
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Fanwei Zeng
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Yifan Wu
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Qianyu Shi
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Jiuhui Xu
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Yi Huang
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Tingting Ren
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| | - Jiarui Liu
- Department of Anatomy, Histology and Embryology, School of Basic Medical Sciences, Health Science Center, Peking University, Beijing, China
- Neuroscience Research Institute, Peking University, Beijing, China
| | - Wei Guo
- Department of Bone Tumor, Peking University People's Hospital, Beijing, China
| |
Collapse
|
5
|
Kuang L, Wu L, Li Y. Extracellular vesicles in tumor immunity: mechanisms and novel insights. Mol Cancer 2025; 24:45. [PMID: 39953480 PMCID: PMC11829561 DOI: 10.1186/s12943-025-02233-w] [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: 09/30/2024] [Accepted: 01/14/2025] [Indexed: 02/17/2025] Open
Abstract
Extracellular vesicles (EVs), nanoscale vesicles secreted by cells, have attracted considerable attention in recent years due to their role in tumor immunomodulation. These vesicles facilitate intercellular communication by transporting proteins, nucleic acids, and other biologically active substances, and they exhibit a dual role in tumor development and immune evasion mechanisms. Specifically, EVs can assist tumor cells in evading immune surveillance and attack by impairing immune cell function or modulating immunosuppressive pathways, thereby promoting tumor progression and metastasis. Conversely, they can also transport and release immunomodulatory factors that stimulate the activation and regulation of the immune system, enhancing the body's capacity to combat malignant diseases. This dual functionality of EVs presents promising avenues and targets for tumor immunotherapy. By examining the biological characteristics of EVs and their influence on tumor immunity, novel therapeutic strategies can be developed to improve the efficacy and relevance of cancer treatment. This review delineates the complex role of EVs in tumor immunomodulation and explores their potential implications for cancer therapeutic approaches, aiming to establish a theoretical foundation and provide practical insights for the advancement of future EVs-based cancer immunotherapy strategies.
Collapse
Affiliation(s)
- Liwen Kuang
- School of Medicine, Chongqing University, Chongqing, China
| | - Lei Wu
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yongsheng Li
- School of Medicine, Chongqing University, Chongqing, China.
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
6
|
Finan JM, Guo Y, Bartlett AQ, Reyer M, Hawthorne K, Haerr M, Halamish H, Lamikanra O, Calvert V, Chen C, Xia Z, Petricoin EF, Sears RC, Byrne KT, Brody JR. Pancreatic cancer-intrinsic HuR regulates the pro-tumorigenic properties of extracellular vesicles. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.08.637077. [PMID: 39975239 PMCID: PMC11839136 DOI: 10.1101/2025.02.08.637077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) tumors contain chaotic vasculature that limits immune surveillance and promotes early events in the metastatic cascade. However, current antiangiogenic therapies have failed in PDAC, and thus, it remains important to uncover mechanisms by which cancer cells signal to endothelial cells to increase angiogenesis. Our lab has shown that the tumor-intrinsic RNA-binding protein HuR ( ELAVL1 ) plays an important role re-shaping the tumor microenvironment (TME) by regulating the stability and translation of cytokine encoding transcripts. Herein, we demonstrate that PDAC-intrinsic HuR influences endothelial cell function in the TME via extracellular vesicle (EV) signaling, an underexplored signaling axis in tumor progression. We found that HuR knockout (KO) tumors have impaired growth in an immunocompetent mouse model, and that administering purified wildtype (WT) EVs can increase tumor growth. Further, we observed that PDAC EVs contain HuR-dependent mRNA and protein cargoes relating to endothelial cell function and angiogenesis. Treatment of endothelial cells with HuR WT EVs strongly increased the expression of genes involved in barrier function and endothelial cell development, and directly increased their migratory and tube forming functions. In an immunocompetent orthotopic mouse model of PDAC, we showed that HuR increases endothelial cell presence and sprouting, while decreasing ICAM-1 expression. Importantly, we found utilizing a genetic EV reporter, that decreased ICAM-1 within WT tumors occurs in endothelial cells that have imported PDAC EVs, suggesting that this signaling axis is directly modulating endothelial cell behavior in vivo . Collectively, our data reveal a new role of HuR in EV signaling to endothelial cells, promoting angiogenesis while restricting endothelial cell leukocyte trafficking behavior.
Collapse
|
7
|
Singer M, Valerin J, Zhang Z, Zhang Z, Dayyani F, Yaghmai V, Choi A, Imagawa D, Abi-Jaoudeh N. Promising Cellular Immunotherapy for Colorectal Cancer Using Classical Dendritic Cells and Natural Killer T Cells. Cells 2025; 14:166. [PMID: 39936958 PMCID: PMC11817869 DOI: 10.3390/cells14030166] [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: 12/30/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 02/13/2025] Open
Abstract
Colorectal cancer (CRC) remains one of the leading causes of cancer-related morbidity and mortality around the world. Despite advances in surgery, chemotherapy, and targeted therapies, the prognosis for patients with metastatic or advanced CRC remains poor. Immunotherapies comprising immune checkpoint inhibitors showed disappointing responses in metastatic CRC (mCRC). However, cellular immunotherapy, specifically using classical dendritic cells (cDCs), may hold unique promise in immune recognition for CRC antigens. cDCs are substantial players in immune recognition and are instrumental in orchestrating innate and adaptive immune responses by processing and presenting tumor antigens to effector cells. Natural killer T (NKT) cells are insufficiently studied but unique effector cells because of their ability to bridge innate and adaptive immune reactions and the crosstalk with dendritic cells in cancer. This review explores the therapeutic potential of using both cDCs and NKT cells as a synergistic therapy in CRC, focusing on their biological roles, strategies for harnessing their capabilities, clinical applications, and the challenges within the tumor microenvironment. Both cDCs and NKT cells can be used as a new effective approach for cell-based therapies in cancers to provide a new hope for CRC patients that are challenging to treat.
Collapse
Affiliation(s)
- Mahmoud Singer
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Jennifer Valerin
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - Zhuoli Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Zigeng Zhang
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - Farshid Dayyani
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - Vahid Yaghmai
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| | - April Choi
- Department of Medicine, Chao Family Comprehensive Cancer Center, University of California, Irvine, CA 92697, USA (A.C.)
| | - David Imagawa
- Department of Surgery, University of California Irvine, Orange, CA 92697, USA
| | - Nadine Abi-Jaoudeh
- Department of Radiological Sciences, School of Medicine, University of California, Irvine, CA 92617, USA
| |
Collapse
|
8
|
Sierra J, de León UAP, Padilla-Longoria P. Tumor microenvironment noise-induced polarization: the main challenge in macrophages' immunotherapy for cancer. Mol Cell Biochem 2025:10.1007/s11010-025-05205-2. [PMID: 39827422 DOI: 10.1007/s11010-025-05205-2] [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: 10/02/2024] [Accepted: 01/04/2025] [Indexed: 01/22/2025]
Abstract
Disturbance of epigenetic processes can lead to altered gene function and malignant cellular transformation. In particular, changes in the epigenetic landscape are a central topic in cancer biology. The initiation and progression of cancer are now recognized to involve both epigenetic and genetic alterations. In this paper, we study the epigenetic mechanism (related to the tumor microenvironment) responsible for increasing tumor-associated macrophages that promote the occurrence and metastasis of tumor cells, support tumor angiogenesis, inhibit T-cell-mediated anti-tumor immune response, and lead to tumor progression. We show that the tumor benefits from the macrophages' high degree of plasticity and larger epigenetic basins corresponding to phenotypes that favor cancer development through a process that we call noise-induced polarization. Moreover, we propose a mechanism to promote the appropriate epigenetic stability for immunotherapies involving macrophages, which includes p53 and APR-246 (eprenetapopt). Our results show that a combination therapy may be necessary to ensure the proper epigenetic stability of macrophages, which otherwise will contribute to cancer progression. On the other hand, we conclude that macrophages may remain in the anti-tumoral state in types of cancer that exhibit less TP53 mutation, like colorectal cancer; in these cases, macrophages' immunotherapy may be more suitable. We finally mention the relevance of the epigenetic potential (Waddington's landscape) as the backbone for our study, which encapsulates the biological information of the system.
Collapse
Affiliation(s)
- Jesus Sierra
- CIMAT, De Jalisco s/n, Gto., 36023, Guanajuato, Mexico
| | - Ugo Avila-Ponce de León
- Schiffer Group, Vaccine and Infectious Disease, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Pablo Padilla-Longoria
- IIMAS, Universidad Nacional Autonoma de Mexico (UNAM), Ciudad Universitaria, 04510, Mexico City, Mexico.
| |
Collapse
|
9
|
Koca T, Gocen Vardar N, Aksoy RA, Korcum AF. Comprehensive Evaluation of Inflammatory Biomarkers in Cervical Cancer Treated with Chemoradiotherapy. Curr Oncol 2025; 32:39. [PMID: 39851955 PMCID: PMC11763994 DOI: 10.3390/curroncol32010039] [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: 11/20/2024] [Revised: 12/31/2024] [Accepted: 01/08/2025] [Indexed: 01/26/2025] Open
Abstract
Objective: Inflammatory biomarkers have been shown to possess both prognostic and predictive significance in various cancers. Among the emerging biomarkers, the pan-immune-inflammation value (PIV) has recently been introduced as a novel indicator representing both the immune response and the systemic inflammatory state. This study aims to comprehensively evaluate the predictive value of inflammatory biomarkers on survival outcomes in cervical cancer patients undergoing chemoradiotherapy. Methods: A total of 90 patients who had undergone chemoradiotherapy for cervical cancer were included. Data on demographics, treatment protocols, pre-treatment blood parameters, and survival outcomes were collected. The association between inflammatory biomarkers and survival outcomes was investigated through univariate and multivariate analyses. Results: The univariate analysis identified the following as predictors of progression-free survival (PFS): neutrophil-lymphocyte ratio (NLR), platelet-lymphocyte ratio (PLR), monocyte-lymphocyte ratio (MLR), systemic immune-inflammation index (SII), PIV, C-reactive protein (CRP), albumin, and tumor size. Multivariate analysis revealed that only the PIV significantly predicted PFS (HR 3.05, 95% CI 1.0 to 9.3, p = 0.04). In the univariate analysis, several variables were predictive of overall survival (OS), including NLR, PLR, MLR, SII, PIV, CRP, LDH, albumin, tumor size, and Eastern Cooperative Oncology Group Performance Status (ECOG PS). Multivariate analysis revealed CRP (HR 3.41, 95% CI 1.5 to 7.7, p = 0.003) and ECOG PS (HR 4.78, 95% CI 1.3 to 17.3, p = 0.01) predictive of OS, with PIV approaching statistical significance (HR 2.56, 95% CI 0.8 to 7.6, p = 0.09). Conclusions: This study provides the first comprehensive analysis of the association between cervical cancer and various inflammatory biomarkers. Many of these biomarkers have demonstrated predictive value for survival outcomes in patients with cervical cancer undergoing definitive chemoradiotherapy. Among the biomarkers evaluated, CRP and PIV were identified as the most predictive, warranting further exploration in future research.
Collapse
Affiliation(s)
- Timur Koca
- Department of Radiation Oncology, Akdeniz University, 07070 Antalya, Turkey; (N.G.V.); (A.F.K.)
| | - Nurcihan Gocen Vardar
- Department of Radiation Oncology, Akdeniz University, 07070 Antalya, Turkey; (N.G.V.); (A.F.K.)
| | - Rahmi Atıl Aksoy
- Department of Radiation Oncology, Izmir City Hospital, 35540 Izmir, Turkey;
| | - Aylin Fidan Korcum
- Department of Radiation Oncology, Akdeniz University, 07070 Antalya, Turkey; (N.G.V.); (A.F.K.)
| |
Collapse
|
10
|
Giuliani G, Stewart W, Li Z, Jayaprakash C, Das J. Spatial organization and stochastic fluctuations of immune cells impact clinical responsiveness to immunotherapy in melanoma patients. PNAS NEXUS 2024; 3:pgae539. [PMID: 39677361 PMCID: PMC11642613 DOI: 10.1093/pnasnexus/pgae539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 11/11/2024] [Indexed: 12/17/2024]
Abstract
High-dimensional, spatial single-cell technologies, such as CyTOF imaging mass cytometry (IMC), provide detailed information regarding locations of a large variety of cancer and immune cells in microscopic scales in tumor microarray slides obtained from patients prior to immune checkpoint inhibitor (ICI) therapy. An important question is how the initial spatial organization of these cells in the tumor microenvironment (TME) changes with time and regulates tumor growth and eventually outcomes as patients undergo ICI therapy. Utilizing IMC data of melanomas of patients who later underwent ICI therapy, we develop a spatially resolved interacting cell system model that is calibrated against patient response data to address the above question. We find that the tumor fate in these patients is determined by the spatial organization of activated CD8+ T cells, macrophages, and melanoma cells and the interplay between these cells that regulate exhaustion of CD8+ T cells. We find that fencing of tumor cell boundaries by exhausted CD8+ T cells is dynamically generated from the initial conditions that can play a protumor role. Furthermore, we find that specific spatial features such as co-clustering of activated CD8+ T cells and macrophages in the pretreatment samples determine the fate of the tumor progression, despite stochastic fluctuations and changes over the treatment course. Our framework enables the determination of mechanisms of interplay between a key subset of tumor and immune cells in the TME that regulate clinical response to ICIs.
Collapse
Affiliation(s)
- Giuseppe Giuliani
- Department of Physics, The Ohio State University, Columbus, OH 43210, USA
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH 43205, USA
| | | | - Zihai Li
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Department of Internal Medicine, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| | | | - Jayajit Das
- Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH 43205, USA
- Pelotonia Institute for Immuno-Oncology, The Ohio State University, Columbus, OH 43210, USA
- Department of Pediatrics, College of Medicine, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
11
|
Anton A, Hutchinson R, Hovens CM, Christie M, Ryan A, Gibbs P, Costello A, Peters J, Neeson PJ, Corcoran NM, Tran B. An immune suppressive tumor microenvironment in primary prostate cancer promotes tumor immune escape. PLoS One 2024; 19:e0301943. [PMID: 39602457 PMCID: PMC11602054 DOI: 10.1371/journal.pone.0301943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Accepted: 03/25/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Immunotherapy has demonstrated limited activity in prostate cancer to date. This likely reflects an immune suppressive tumor microenvironment (TME), with previous studies suggesting low PD-L1 expression and a sparse immune cell infiltrate. We aimed to further characterise the immune TME in primary prostate cancer and correlate immune subset densities with clinical outcomes. METHODS Two distinct cohorts of patients treated with radical prostatectomy were identified, based on the development of biochemical recurrence (BCR), one subgroup with high International Society of Urological Pathologists (ISUP) grade group, recurrent disease and a second with low grade, non-recurrent disease. A prostate immunohistochemical (IHC) antibody cocktail was used to differentiate tumor and peritumoral benign tissue. Specific CD8+, CD4+, FoxP3+, CD20+ and CD68+ cell subsets were identified using IHC staining of consecutive slides. PD-L1 and CD8/PD-L1 dual staining were also performed. Cell subset densities were quantified within tumor and peritumoral regions. We used descriptive statistics to report cell subset densities and T-tests to compare groups by age, grade and the development of BCR. Univariable and multivariable logistic regression were used to analyse risk factors for BCR and the development of metastatic disease. RESULTS A total of 175 patients were included, with a median age of 63 years and median pre-operative PSA of 8.2ng/ml. BCR occurred in 115 patients (66%) and 56 (32%) developed metastatic disease. CD68+ cells were the most abundant (median 648.8/mm2 intratumoral, 247.6/mm2 peritumoral), while PD-L1+ and PD-L1/CD8+ cell density was low overall (PD-L1+ median 162.4/mm2 intratumoral, 141.7/mm2 peritumoral; PD-L1/CD8+ (median 5.52/mm2 intratumoral, 3.41/mm2 peritumoral). Overall, grade group and T-stage were independently associated with BCR and metastatic disease. Higher density of peritumoral PD-L1+ cells was an independent risk factor for BCR (OR 5.33, 95%CI 1.31-21.61, p = 0.019).Although higher densities of CD8+ and CD4+ cells were observed in higher grade group 3-5 tumors, these were not associated with the development of BCR or metastasis. CONCLUSIONS In our cohort of prostate cancer patients who underwent radical prostatectomy, higher grade group and T-stage were independent predictors of BCR and metastasis. Despite higher grade group being associated with higher CD8+ cell density, PD-L1+ and PD-L1/CD8+ cell densities were low overall, suggesting lower T cell receptor recognition of tumor antigens. Further understanding of this phenomenon would influence development of future immunotherapeutic strategies in prostate cancer.
Collapse
Affiliation(s)
- Angelyn Anton
- Division of personalised oncology, Walter and Eliza Hall Institute, Melbourne, Australia
| | - Ryan Hutchinson
- Division of personalised oncology, Walter and Eliza Hall Institute, Melbourne, Australia
| | - Christopher M. Hovens
- Royal Melbourne Hospital, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
| | | | - Andrew Ryan
- TissuPath, Mount Waverley, Melbourne, Australia
| | - Peter Gibbs
- Division of personalised oncology, Walter and Eliza Hall Institute, Melbourne, Australia
- Western Health, Melbourne, Australia
| | - Anthony Costello
- Royal Melbourne Hospital, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
- Epworth Healthcare, Melbourne, Australia
| | - Justin Peters
- Royal Melbourne Hospital, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
- Epworth Healthcare, Melbourne, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Australia
| | - Niall M. Corcoran
- Royal Melbourne Hospital, Melbourne, Australia
- Department of Surgery, University of Melbourne, Melbourne, Australia
- Western Health, Melbourne, Australia
| | - Ben Tran
- Division of personalised oncology, Walter and Eliza Hall Institute, Melbourne, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Australia
- Department of Medical Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
12
|
Su C, Lin Z, Ye Z, Liang J, Yu R, Wan Z, Hou J. Development of a prognostic model for early-stage gastric cancer-related DNA methylation-driven genes and analysis of immune landscape. Front Mol Biosci 2024; 11:1455890. [PMID: 39575189 PMCID: PMC11579923 DOI: 10.3389/fmolb.2024.1455890] [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: 06/27/2024] [Accepted: 09/13/2024] [Indexed: 11/24/2024] Open
Abstract
Background and Aims This study aimed to develop a prognostic model based on DNA methylation-driven genes for patients with early-stage gastric cancer and to examine immune infiltration and function across varying risk levels. Methods We analyzed data from stage I/II gastric cancer patients in The Cancer Genome Atlas which included clinical details, mRNA expression profiles, and level 3 DNA methylation array data. Using the empirical Bayes method of the limma package, we identified differentially expressed genes (DEGs), and the MethylMix package facilitated the identification of DNA methylation-driven genes (DMGs). Univariate Cox regression and LASSO (least absolute shrinkage and selector operation) analyses were utilized to pinpoint critical genes. A risk score prediction model was formulated using two genes that demonstrated the most significant hazard ratios (HRs). Model performance was evaluated within the initial cohort and verified in the GSE84437 cohort; a nomogram was also constructed based on these genes. We further examined 50 methylation sites associated with three CpG islands in C1orf35 and 14 methylation sites linked to one CpG island in FAAH. The CIBERSORT package was employed to identify immune cell clusters in the prediction model. Results A total of 176 DNA methylation-driven genes were refined down to a four-gene signature (ZC3H12A was hypermethylated; GATA3, C1orf35, and FAAH were hypomethylated), which exhibited a significant correlation with overall survival (OS), as evidenced by p-values below 0.05 following univariate Cox regression and LASSO analysis. Specifically, for the risk score prediction model, C1orf35, which had the highest hazard ratio (HR = 2.035, p = 0.028), and FAAH, with the lowest hazard ratio (HR = 0.656, p = 0.012), were selected. The Kaplan-Meier analysis demonstrated distinct survival outcomes between the high-risk and low-risk score groups. The model's predictive accuracy was confirmed with an area under the curve (AUC) of 0.611 for 3-year survival and 0.564 for 5-year survival. Notably, the hypomethylation of the three CpG islands in C1orf35 and the single CpG island in FAAH was significantly different in stage I/II gastric cancer patients compared to normal tissues. Additionally, the high-risk score group showed a notable association with resting CD4 memory T cells. Conclusion Promoter hypomethylation of C1orf35 and FAAH in early-stage gastric cancer underscores their potential as biomarkers for accurate diagnosis and treatment. The developed predictive model employing genes affected by DNA methylation serves as a crucial independent prognostic factor in early-stage gastric cancer.
Collapse
Affiliation(s)
- Chen Su
- The School of Clinical Medical, Fujian Medical University, Fuzhou, Fujian, China
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen, China
| | - Zeyang Lin
- Department of Pathology, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Zhijian Ye
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen, China
| | - Jing Liang
- Department of Pathology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Rong Yu
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen, China
| | - Zheng Wan
- Department of Minimally Invasive and Interventional Therapy for Cancer, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen, China
| | - Jingjing Hou
- The School of Clinical Medical, Fujian Medical University, Fuzhou, Fujian, China
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen, China
| |
Collapse
|
13
|
Vallejos R, Zhantuyakova A, Negri GL, Martin SD, Spencer SE, Thornton S, Leung S, Lynch B, Qin Y, Chow C, Liang B, Zdravko S, Douglas JM, Milne K, Mateyko B, Nelson BH, Howitt BE, Kommoss FK, Horn LC, Hoang L, Singh N, Morin GB, Huntsman DG, Cochrane D. Changes in the tumour microenvironment mark the transition from serous borderline tumour to low-grade serous carcinoma. J Pathol 2024; 264:197-211. [PMID: 39081243 DOI: 10.1002/path.6338] [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: 01/10/2024] [Revised: 05/06/2024] [Accepted: 06/22/2024] [Indexed: 09/04/2024]
Abstract
Low-grade serous ovarian carcinoma (LGSC) is a rare and lethal subtype of ovarian cancer. LGSC is pathologically, biologically, and clinically distinct from the more common high-grade serous ovarian carcinoma (HGSC). LGSC arises from serous borderline ovarian tumours (SBTs). The mechanism of transformation for SBTs to LGSC remains poorly understood. To better understand the biology of LGSC, we performed whole proteome profiling of formalin-fixed, paraffin-embedded tissue blocks of LGSC (n = 11), HGSC (n = 19), and SBTs (n = 26). We identified that the whole proteome is able to distinguish between histotypes of the ovarian epithelial tumours. Proteins associated with the tumour microenvironment were differentially expressed between LGSC and SBTs. Fibroblast activation protein (FAP), a protein expressed in cancer-associated fibroblasts, is the most differentially abundant protein in LGSC compared with SBT. Multiplex immunohistochemistry (IHC) for immune markers (CD20, CD79a, CD3, CD8, and CD68) was performed to determine the presence of B cells, T cells, and macrophages. The LGSC FAP+ stroma was associated with greater abundance of Tregs and M2 macrophages, features not present in SBTs. Our proteomics cohort reveals that there are changes in the tumour microenvironment in LGSC compared with its putative precursor lesion, SBT. These changes suggest that the tumour microenvironment provides a supportive environment for LGSC tumourigenesis and progression. Thus, targeting the tumour microenvironment of LGSC may be a viable therapeutic strategy. © 2024 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Rodrigo Vallejos
- Department of Genome Sciences and Technology, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Almira Zhantuyakova
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
| | | | - Spencer D Martin
- Diagnostic and Molecular Pathology, University of British Columbia, Vancouver, BC, Canada
| | | | - Shelby Thornton
- Molecular and Advanced Pathology Core, University of British Columbia, Vancouver, BC, Canada
| | - Samuel Leung
- Molecular and Advanced Pathology Core, University of British Columbia, Vancouver, BC, Canada
| | - Branden Lynch
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| | - Yimei Qin
- Molecular and Advanced Pathology Core, University of British Columbia, Vancouver, BC, Canada
| | - Christine Chow
- Molecular and Advanced Pathology Core, University of British Columbia, Vancouver, BC, Canada
| | - Brooke Liang
- Stanford University School of Medicine, Stanford, CA, USA
| | | | | | - Katy Milne
- Deeley Research Centre, BC Cancer, Victoria, Canada
| | | | | | | | - Felix Kf Kommoss
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
- Diagnostic and Molecular Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Lars-Christian Horn
- Division of Gynecologic, Breast and Perinatal Pathology, Institute of Pathology, University Hospital Leipzig, Leipzig, Germany
| | - Lien Hoang
- Diagnostic and Molecular Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Naveena Singh
- Diagnostic and Molecular Pathology, University of British Columbia, Vancouver, BC, Canada
| | - Gregg B Morin
- Genome Sciences Centre, BC Cancer, Vancouver, BC, Canada
| | - David G Huntsman
- Department of Genome Sciences and Technology, University of British Columbia, Vancouver, BC, Canada
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
- Department of Obstetrics and Gynaecology, University of British Columbia, Vancouver, BC, Canada
- Diagnostic and Molecular Pathology, University of British Columbia, Vancouver, BC, Canada
- Molecular and Advanced Pathology Core, University of British Columbia, Vancouver, BC, Canada
| | - Dawn Cochrane
- Department of Molecular Oncology, BC Cancer, Vancouver, BC, Canada
| |
Collapse
|
14
|
Liang J, Zhu L, Li J, Wu K, Zhang M, Ma S, Chen X, Xia B. Comprehensive analysis to identify IL7R as a immunotherapy biomarker from pan-cancer analysis to in vitro validation. Discov Oncol 2024; 15:509. [PMID: 39347891 PMCID: PMC11442881 DOI: 10.1007/s12672-024-01357-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Immunotherapy faces a major challenge in treatment resistance, highlighting the need for efficacy biomarkers identification. The tumor microenvironment (TME) significantly influences treatment outcomes, necessitating molecular TME exploration to address immunotherapy resistance. METHODS The study initially pinpointed IL7R as a pivotal TME gene and then examined its impact on TME's CD8 + T cells at the single-cell level. Bulk-RNA analysis investigated IL7R function, immune cell infiltration related to IL7R in TCGA pan-cancer samples with its expression verified in clinical samples through immunohistochemistry. Genome instability and immune-related molecular expression associated with IL7R were also assessed. Furthermore, the clinical efficacy of IL7R was evaluated in various immunotherapy treatment cohorts. RESULTS Our single-cell analyses and cell-cased experiment revealed that T cells with high IL7R expression tended to be non-terminal and correlated with favorable immunotherapy responses. High IL7R expression corresponded to increased immune and stromal cell signiture, immune pathway enrichment, and an immune-inflamed environment in Bulk-RNA analysis and immunohistochemistry verification. These patients exhibited higher proportions of memory T cells and M1 cells within the TME, along with frequent genome instability and immune molecular upregulation. While IL7R had varied prognostic impact across the TCGA dataset, patients with high IL7R expression showed extended survival under immunotherapy. CONCLUSION IL7R plays a critical role in shaping TME diversity across cancer types and holds promise as a relevant biomarker for predicting immunotherapy benefits.
Collapse
Affiliation(s)
- Jiafeng Liang
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Lucheng Zhu
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Jiawei Li
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Kan Wu
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Minna Zhang
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Shenglin Ma
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China
| | - Xueqin Chen
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China.
| | - Bing Xia
- Department of Thoracic Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China.
| |
Collapse
|
15
|
Shi JX, Zhang ZC, Yin HZ, Piao XJ, Liu CH, Liu QJ, Zhang JC, Zhou WX, Liu FC, Yang F, Wang YF, Liu H. RNA m6A modification in ferroptosis: implications for advancing tumor immunotherapy. Mol Cancer 2024; 23:213. [PMID: 39342168 PMCID: PMC11437708 DOI: 10.1186/s12943-024-02132-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 09/19/2024] [Indexed: 10/01/2024] Open
Abstract
The pursuit of innovative therapeutic strategies in oncology remains imperative, given the persistent global impact of cancer as a leading cause of mortality. Immunotherapy is regarded as one of the most promising techniques for systemic cancer therapies among the several therapeutic options available. Nevertheless, limited immune response rates and immune resistance urge us on an augmentation for therapeutic efficacy rather than sticking to conventional approaches. Ferroptosis, a novel reprogrammed cell death, is tightly correlated with the tumor immune environment and interferes with cancer progression. Highly mutant or metastasis-prone tumor cells are more susceptible to iron-dependent nonapoptotic cell death. Consequently, ferroptosis-induction therapies hold the promise of overcoming resistance to conventional treatments. The most prevalent post-transcriptional modification, RNA m6A modification, regulates the metabolic processes of targeted RNAs and is involved in numerous physiological and pathological processes. Aberrant m6A modification influences cell susceptibility to ferroptosis, as well as the expression of immune checkpoints. Clarifying the regulation of m6A modification on ferroptosis and its significance in tumor cell response will provide a distinct method for finding potential targets to enhance the effectiveness of immunotherapy. In this review, we comprehensively summarized regulatory characteristics of RNA m6A modification on ferroptosis and discussed the role of RNA m6A-mediated ferroptosis on immunotherapy, aiming to enhance the effectiveness of ferroptosis-sensitive immunotherapy as a treatment for immune-resistant malignancies.
Collapse
Affiliation(s)
- Jun-Xiao Shi
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Zhi-Chao Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Hao-Zan Yin
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China
| | - Xian-Jie Piao
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Cheng-Hu Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Qian-Jia Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Jia-Cheng Zhang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Wen-Xuan Zhou
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu-Chen Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Fu Yang
- The Department of Medical Genetics, Naval Medical University, Shanghai, 200433, China.
- Key Laboratory of Biosafety Defense, Ministry of Education, Shanghai, 200433, China.
- Shanghai Key Laboratory of Medical Biodefense, Shanghai, 200433, China.
| | - Yue-Fan Wang
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| | - Hui Liu
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China.
| |
Collapse
|
16
|
Li Y, Luo Y. STdGCN: spatial transcriptomic cell-type deconvolution using graph convolutional networks. Genome Biol 2024; 25:206. [PMID: 39103939 DOI: 10.1186/s13059-024-03353-0] [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: 10/01/2023] [Accepted: 07/26/2024] [Indexed: 08/07/2024] Open
Abstract
Spatially resolved transcriptomics integrates high-throughput transcriptome measurements with preserved spatial cellular organization information. However, many technologies cannot reach single-cell resolution. We present STdGCN, a graph model leveraging single-cell RNA sequencing (scRNA-seq) as reference for cell-type deconvolution in spatial transcriptomic (ST) data. STdGCN incorporates expression profiles from scRNA-seq and spatial localization from ST data for deconvolution. Extensive benchmarking on multiple datasets demonstrates that STdGCN outperforms 17 state-of-the-art models. In a human breast cancer Visium dataset, STdGCN delineates stroma, lymphocytes, and cancer cells, aiding tumor microenvironment analysis. In human heart ST data, STdGCN identifies changes in endothelial-cardiomyocyte communications during tissue development.
Collapse
Affiliation(s)
- Yawei Li
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
- Center for Collaborative AI in Healthcare, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA
| | - Yuan Luo
- Department of Preventive Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
- Center for Collaborative AI in Healthcare, Feinberg School of Medicine, Northwestern University, Chicago, IL, 60611, USA.
| |
Collapse
|
17
|
Rodrigues-Jesus J, Canadas-Sousa A, Oliveira P, Figueira AC, Marrinhas C, Petrucci GN, Gregório H, Tinoco F, Goulart A, Felga H, Vilhena H, Dias-Pereira P. Distribution of Inflammatory Infiltrate in Feline Mammary Lesions: Relationship With Clinicopathological Features. Vet Comp Oncol 2024. [PMID: 38863270 DOI: 10.1111/vco.12987] [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: 01/30/2024] [Revised: 03/25/2024] [Accepted: 05/21/2024] [Indexed: 06/13/2024]
Abstract
Inflammation is a frequent finding in feline mammary neoplasms. Recent research suggests that the presence and location of tumour-associated immune cells might play a significant role in the clinical outcome of feline mammary carcinomas. The present study aimed to characterise the overall inflammatory infiltrates in healthy, hyperplastic/dysplastic, benign and malignant lesions of the feline mammary gland, and to evaluate its association with clinicopathological features. Perilesional and intralesional inflammatory foci were evaluated in 307 lesions from 185 queens, and categorised according to its distribution and intensity. The presence, location and density of tertiary lymphoid structures were also assessed. A control group included 24 queens without mammary changes. The presence of intralesional and perilesional inflammatory infiltrate was observed in a majority of the lesions (80.8% and 90.2%, respectively), but differed according to the type of mammary lesion, being more remarkable in malignant neoplasms. Only scarce individual cells were observed in 28.1% of the normal mammary glands. Data analysis revealed statistically significant associations (p < 0.05) between the presence of a more prominent intralesional and perilesional inflammatory reaction and several clinicopathological features associated with worse prognosis, including clinical stage, tumour size, mitotic count, lymphovascular invasion and lymph node metastasis. Furthermore, tertiary lymphoid structures were significantly more frequent in tumours with an infiltrative growth and lymph node metastasis. According to our results, the inflammatory reaction present in different types of feline mammary lesions is associated with the development of more aggressive tumours.
Collapse
Affiliation(s)
- Joana Rodrigues-Jesus
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, Porto, Portugal
| | - Ana Canadas-Sousa
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, Porto, Portugal
| | - Pedro Oliveira
- Department of Populations Studies, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, Porto, Portugal
| | - Ana Catarina Figueira
- OneVet Veterinary University Hospital of Coimbra (HVUC), Coimbra, Portugal
- Centre for Investigation Vasco da Gama (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, Coimbra, Portugal
| | - Carla Marrinhas
- Centre for Investigation Vasco da Gama (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, Coimbra, Portugal
- OneVet Veterinary Hospital of Baixo Vouga (HVBV), Águeda, Portugal
| | - Gonçalo N Petrucci
- Centre for Investigation Vasco da Gama (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, Coimbra, Portugal
- OneVet Veterinary Hospital of Porto (HVP), Porto, Portugal
- Department of Animal and Veterinary Sciences, University Institute for Health Sciences, CESPU, CRL, Gandra, Portugal
| | - Hugo Gregório
- Department of Animal and Veterinary Sciences, University Institute for Health Sciences, CESPU, CRL, Gandra, Portugal
- AniCura Veterinary Hospital Centre (CHV), Porto, Portugal
| | - Flora Tinoco
- Dra. Flora Tinoco Veterinary Clinic, Maia, Portugal
| | | | - Helena Felga
- Clínica dos Gatos Veterinary Clinic, Porto, Portugal
| | - Hugo Vilhena
- OneVet Veterinary University Hospital of Coimbra (HVUC), Coimbra, Portugal
- Centre for Investigation Vasco da Gama (CIVG), Department of Veterinary Sciences, Vasco da Gama University School, Coimbra, Portugal
- Animal and Veterinary Research Centre (CECAV), University of Trás-os-Montes e Alto Douro, Vila Real, Portugal
- Associate Laboratory of Animal and Veterinary Sciences AL4AnimaLS, Lisbon, Portugal
| | - Patrícia Dias-Pereira
- Department of Pathology and Molecular Immunology, School of Medicine and Biomedical Sciences, ICBAS-UP, University of Porto, Porto, Portugal
| |
Collapse
|
18
|
Zeng J, Ke C, Tian K, Nie J, Huang S, Song X, Xian Z. Highly expressed of BID indicates poor prognosis and mediates different tumor microenvironment characteristics in clear cell renal cell carcinoma. Discov Oncol 2024; 15:176. [PMID: 38767695 PMCID: PMC11106230 DOI: 10.1007/s12672-024-01035-8] [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: 02/16/2024] [Accepted: 05/12/2024] [Indexed: 05/22/2024] Open
Abstract
BACKGROUND Studies have found that BH3 interacting domain death agonist (BID) is closely related to the occurrence and development of many kinds of tumors. However, little attention has been paid to the situation of BID in clear cell renal cell carcinoma (ccRCC). So, our aim was to explore the effect of BID in ccRCC. METHODS Survival analysis, ROC curve, correlation analysis and Cox regression analysis were executed to analyze the prognostic value and clinical correlation of BID in ccRCC. The risk prognosis model was constructed in the training cohort and further validated in the internal testing cohort, ICGC cohort, and GEO cohort. Transcriptome sequencing and immunohistochemical staining of clinical specimens were used to validate the results of bioinformatics analysis. The GSEA, ESTIMATE algorithm, CIBERSORT algorithm, ssGSEA, TIDE score, correlation and difference analysis were used to analyze the effects of BID on immune infiltration in tumor microenvironment (TME). RESULTS BID was highly expressed in ccRCC tissues, which was verified by transcriptome sequencing and immunohistochemical staining of clinical specimens. Patients with high expression of BID had a worse prognosis. BID is an independent prognostic factor for ccRCC. The prognostic model based on BID can accurately predict the prognosis of patients in different cohorts. In addition, the expression levels of BID was closely related to immunomodulatory molecules such as PD-1, LAG3, and CTLA4. Enrichment analysis indicated that BID was significantly enriched in immune-related responses and cancer-related pathways. The change of BID expression mediates different characteristics of immune infiltration in TME. CONCLUSIONS BID is highly expressed in ccRCC, which is a reliable biomarker of ccRCC prognosis. It is closely related to TME, and may be a potential target for immunotherapy in patients with ccRCC.
Collapse
Affiliation(s)
- Jiayi Zeng
- Department of Urology, Guangdong Provincial People's Hospital's Nanhai Hospital, Foshan, China
| | - Chuangbo Ke
- Department of Urology, Guangdong Provincial People's Hospital's Nanhai Hospital, Foshan, China
| | - Kaiwen Tian
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jianru Nie
- Department of Urology, Guangdong Provincial People's Hospital's Nanhai Hospital, Foshan, China
| | - Shaoming Huang
- Department of Urology, Ganzhou Hospital of Guangdong Provincial People's Hospital, Ganzhou Municipal Hospital, Ganzhou, China
| | - Xiaosong Song
- Department of Urology, Guangdong Provincial People's Hospital's Nanhai Hospital, Foshan, China
| | - Zhiyong Xian
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Department of Urology, Guangdong Provincial People's Hospital's Nanhai Hospital, Foshan, China.
| |
Collapse
|
19
|
Wu GH, He C, Che G, Zhou Z, Chen BY, Wu HM, Chen JF, Zhu WP, Yang Y, Zhou Z, Teng LS, Wang HY. The role of FERMT2 in the tumor microenvironment and immunotherapy in pan-cancer using comprehensive single-cell and bulk sequencing. Heliyon 2024; 10:e30505. [PMID: 38726194 PMCID: PMC11079299 DOI: 10.1016/j.heliyon.2024.e30505] [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: 12/07/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
FERMT2 has been identified as a participant in integrin-linked kinase signaling pathways, influencing epithelial-mesenchymal transition and thereby affecting tumor initiation, progression, and invasion. While the character of FERMT2 in the tumor microenvironment (TME) as well as its implications for immunotherapy remain unclear. Thus, we conducted a comprehensive analysis to assess the prognostic significance of FERMT2 using Kaplan-Meier analysis. In addition, we employed enrichment analysis to uncover potential underlying molecular mechanisms. Using "Immunedeconv" package, we evaluated the immune characteristics of FERMT2 within TME. Furthermore, we determined the expression levels of FERMT2 in various cell types within TME, based on single-cell sequencing data. To confirm the co-expression of FERMT2 and markers of cancer-associated fibroblasts (CAFs), we performed multiplex immunofluorescence staining on tissue paraffin sections across various cancer types. Our analysis disclosed a significant correlation between elevated FERMT2 expression and unfavorable prognosis in specific cancer types. Furthermore, we identified a strong correlation between FERMT2 expression and diverse immune-related factors, including immune checkpoint molecules, immune cell infiltration, microsatellite instability (MSI), and tumor mutational burden (TMB). Additionally, there was a significant correlation between FERMT2 expression and immune-related pathways, particularly those associated with activating, migrating, and promoting the growth of fibroblasts in diverse cancer types. Interestingly, we observed consistent co-expression of FERMT2 in both malignant tumor cells and stromal cells, particularly within CAFs. Notably, our findings also indicated that FERMT2, in particular, exhibited elevated expression levels within tumor tissues and co-expressed with α-SMA in CAFs based on the multiplex immunofluorescence staining results.
Collapse
Affiliation(s)
- Guang-hao Wu
- School of Clinical Medicine, Hangzhou Normal University Medical College, Hangzhou, China
| | - Chao He
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Gang Che
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zheng Zhou
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Bi-ying Chen
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hai-ming Wu
- Department of Gastrointestinal Surgery, Yiwu Central Hospital, Jinhua, China
| | - Jian-feng Chen
- Department of Gastrointestinal Surgery, Yiwu Central Hospital, Jinhua, China
| | - Wei-pu Zhu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| | - Yan Yang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Zhan Zhou
- Institute of Drug Metabolism and Pharmaceutical Analysis and Zhejiang Provincial Key Laboratory of Anti-Cancer Drug Research, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Li-song Teng
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Hai-yong Wang
- Department of Surgical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| |
Collapse
|
20
|
Zhang J, Shi X, Wang M, Zhai R, Wang M, Gong Z, Ni Z, Xu T, Zhu W, Liu L. Identification of immunogenic cell death-related damage-related molecular patterns (DAMPs) to predict outcomes in patients with head and neck squamous cell carcinoma. J Cancer Res Clin Oncol 2024; 150:240. [PMID: 38713284 PMCID: PMC11076381 DOI: 10.1007/s00432-024-05779-2] [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: 11/18/2023] [Accepted: 05/03/2024] [Indexed: 05/08/2024]
Abstract
PURPOSE Head and neck cancer is the sixth most common type of cancer worldwide, wherein the immune responses are closely associated with disease occurrence, development, and prognosis. Investigation of the role of immunogenic cell death-related genes (ICDGs) in adaptive immune response activation may provide cues into the mechanism underlying the outcome of HNSCC immunotherapy. METHODS ICDGs expression patterns in HNSCC were analyzed, after which consensus clustering in HNSCC cohort conducted. A 4-gene prognostic model was constructed through LASSO and Cox regression analyses to analyze the prognostic index using the TCGA dataset, followed by validation with two GEO datasets. The distribution of immune cells and the response to immunotherapy were compared between different risk subtypes through multiple algorithms. Moreover, immunohistochemical (IHC) analyses were conducted to validate the prognostic value of HSP90AA1 as a predictor of HNSCC patient prognosis. In vitro assays were performed to further detect the effect of HSP90AA1 in the development of HNSCC. RESULTS A novel prognostic index based on four ICDGs was constructed and proved to be useful as an independent factor of HNSCC prognosis. The risk score derived from this model grouped patients into high- and low-risk subtypes, wherein the high-risk subtype had worse survival outcomes and poorer immunotherapy response. IHC analysis validated the applicability of HSP90AA1 as a predictor of prognosis of HNSCC patients. HSP90AA1 expression in tumor cells promotes the progression of HNSCC. CONCLUSIONS Together, these results highlight a novel four-gene prognostic signature as a valuable tool to assess survival status and prognosis of HNSCC patients.
Collapse
Affiliation(s)
- Jiayi Zhang
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Xinzhan Shi
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Mengqi Wang
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Rundong Zhai
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Mengyao Wang
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Zizhen Gong
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Zihui Ni
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Teng Xu
- Department of Oral and Maxillofacial Surgery, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Weiwen Zhu
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China
| | - Laikui Liu
- Department of Basic Science of Stomatology, The Affiliated Stomatological Hospital of Nanjing Medical University, Jiangsu, China.
- State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, Nanjing Medical University, Jiangsu, China.
- Jiangsu Province Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Jiangsu, China.
| |
Collapse
|
21
|
Seager RJ, Ko H, Pabla S, Senosain MF, Kalinski P, Van Roey E, Gao S, Strickland KC, Previs RA, Nesline MK, Hastings S, Zhang S, Conroy JM, Jensen TJ, Eisenberg M, Caveney B, Severson EA, Ramkissoon S, Gandhi S. Immunologic Factors Associated with Differential Response to Neoadjuvant Chemoimmunotherapy in Triple-Negative Breast Cancer. J Pers Med 2024; 14:481. [PMID: 38793063 PMCID: PMC11122407 DOI: 10.3390/jpm14050481] [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: 03/29/2024] [Revised: 04/17/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Background: KEYNOTE-522 resulted in FDA approval of the immune checkpoint inhibitor pembrolizumab in combination with neoadjuvant chemotherapy for patients with early-stage, high-risk, triple-negative breast cancer (TNBC). Unfortunately, pembrolizumab is associated with several immune-related adverse events (irAEs). We aimed to identify potential tumor microenvironment (TME) biomarkers which could predict patients who may attain pathological complete response (pCR) with chemotherapy alone and be spared the use of anti-PD-1 immunotherapy. Methods: Comprehensive immune profiling, including RNA-seq gene expression assessment of 395 immune genes, was performed on matched FFPE tumor samples from 22 stage I-III TNBC patients (14 patients treated with neoadjuvant chemotherapy alone (NAC) and 8 treated with neoadjuvant chemotherapy combined with pembrolizumab (NAC+I)). Results: Differential gene expression analysis revealed that in the NAC group, IL12B and IL13 were both significantly associated with pCR. In the NAC+I group, LCK and TP63 were significantly associated with pCR. Patients in both treatment groups exhibiting pCR tended to have greater tumor inflammation than non-pCR patients. In the NAC+I group, patients with pCR tended to have greater cell proliferation and higher PD-L1 expression, while in the NAC group, patients with pCR tended to have lower cancer testis antigen expression. Additionally, the NAC+I group trended toward a lower relative dose intensity averaged across all chemotherapy drugs, suggesting that more dose reductions or treatment delays occurred in the NAC+I group than the NAC group. Conclusions: A comprehensive understanding of immunologic factors could potentially predict pCR to chemotherapy alone, enabling the avoidance of the unnecessary treatment of these patients with checkpoint inhibitors.
Collapse
Affiliation(s)
- Robert J. Seager
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Heidi Ko
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Sarabjot Pabla
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Maria-Fernanda Senosain
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Pawel Kalinski
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| | - Erik Van Roey
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Shuang Gao
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Kyle C. Strickland
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Department of Pathology, Duke University Medical Center, Duke Cancer Institute, Durham, NC 27710, USA
| | - Rebecca Ann Previs
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Department of Obstetrics & Gynecology, Duke University Medical Center, Duke Cancer Institute, Division of Gynecologic Oncology, Durham, NC 27710, USA
| | - Mary K. Nesline
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Stephanie Hastings
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Shengle Zhang
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Jeffrey M. Conroy
- Labcorp Oncology, Buffalo, NY 14263, USA; (S.P.); (M.-F.S.); (E.V.R.); (S.G.); (S.Z.); (J.M.C.)
| | - Taylor J. Jensen
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | | | | | - Eric A. Severson
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
| | - Shakti Ramkissoon
- Labcorp Oncology, Durham, NC 27710, USA; (H.K.); (K.C.S.); (R.A.P.); (M.K.N.); (S.H.); (T.J.J.); (E.A.S.); (S.R.)
- Wake Forest Comprehensive Cancer Center and Department of Pathology, Wake Forest School of Medicine, Winston-Salem, NC 27710, USA
| | - Shipra Gandhi
- Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, USA;
| |
Collapse
|
22
|
Srbova L, Arasalo O, Lehtonen AJ, Pokki J. Measuring mechanical cues for modeling the stromal matrix in 3D cell cultures. SOFT MATTER 2024; 20:3483-3498. [PMID: 38587658 DOI: 10.1039/d3sm01425h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/09/2024]
Abstract
A breast-cancer tumor develops within a stroma, a tissue where a complex extracellular matrix surrounds cells, mediating the cancer progression through biomechanical and -chemical cues. Current materials partially mimic the stromal matrix in 3D cell cultures but methods for measuring the mechanical properties of the matrix at cell-relevant-length scales and stromal-stiffness levels are lacking. Here, to address this gap, we developed a characterization approach that employs probe-based microrheometry and Bayesian modeling to quantify length-scale-dependent mechanics and mechanical heterogeneity as in the stromal matrix. We examined the interpenetrating network (IPN) composed of alginate scaffolds (for adjusting mechanics) and type-1 collagen (a stromal-matrix constituent). We analyzed viscoelasticity: absolute-shear moduli (stiffness/elasticity) and phase angles (viscous and elastic characteristics). We determined the relationship between microrheometry and rheometry information. Microrheometry reveals lower stiffness at cell-relevant scales, compared to macroscale rheometry, with dependency on the length scale (10 to 100 μm). These data show increasing IPN stiffness with crosslinking until saturation (≃15 mM of Ca2+). Furthermore, we report that IPN stiffness can be adjusted by modulating collagen concentration and interconnectivity (by polymerization temperature). The IPNs are heterogeneous structurally (in SEM) and mechanically. Interestingly, increased alginate crosslinking changes IPN heterogeneity in stiffness but not in phase angle, until the saturation. In contrast, such changes are undetectable in alginate scaffolds. Our nonlinear viscoelasticity analysis at tumor-cell-exerted strains shows that only the softer IPNs stiffen with strain, like the stromal-collagen constituent. In summary, our approach can quantify the stromal-matrix-related viscoelasticity and is likely applicable to other materials in 3D culture.
Collapse
Affiliation(s)
- Linda Srbova
- Department of Electrical Engineering and Automation, Aalto University, Espoo, FI-02150, Finland.
| | - Ossi Arasalo
- Department of Electrical Engineering and Automation, Aalto University, Espoo, FI-02150, Finland.
| | - Arttu J Lehtonen
- Department of Electrical Engineering and Automation, Aalto University, Espoo, FI-02150, Finland.
| | - Juho Pokki
- Department of Electrical Engineering and Automation, Aalto University, Espoo, FI-02150, Finland.
| |
Collapse
|
23
|
Soeiro JF, Sousa FL, Monteiro MV, Gaspar VM, Silva NJO, Mano JF. Advances in screening hyperthermic nanomedicines in 3D tumor models. NANOSCALE HORIZONS 2024; 9:334-364. [PMID: 38204336 PMCID: PMC10896258 DOI: 10.1039/d3nh00305a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 01/03/2024] [Indexed: 01/12/2024]
Abstract
Hyperthermic nanomedicines are particularly relevant for tackling human cancer, providing a valuable alternative to conventional therapeutics. The early-stage preclinical performance evaluation of such anti-cancer treatments is conventionally performed in flat 2D cell cultures that do not mimic the volumetric heat transfer occurring in human tumors. Recently, improvements in bioengineered 3D in vitro models have unlocked the opportunity to recapitulate major tumor microenvironment hallmarks and generate highly informative readouts that can contribute to accelerating the discovery and validation of efficient hyperthermic treatments. Leveraging on this, herein we aim to showcase the potential of engineered physiomimetic 3D tumor models for evaluating the preclinical efficacy of hyperthermic nanomedicines, featuring the main advantages and design considerations under diverse testing scenarios. The most recent applications of 3D tumor models for screening photo- and/or magnetic nanomedicines will be discussed, either as standalone systems or in combinatorial approaches with other anti-cancer therapeutics. We envision that breakthroughs toward developing multi-functional 3D platforms for hyperthermia onset and follow-up will contribute to a more expedited discovery of top-performing hyperthermic therapies in a preclinical setting before their in vivo screening.
Collapse
Affiliation(s)
- Joana F Soeiro
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
- Department of Physics, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Filipa L Sousa
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Maria V Monteiro
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| | - Nuno J O Silva
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
- Department of Physics, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193, Aveiro, Portugal.
| |
Collapse
|
24
|
Scholl JN, Weber AF, Dias CK, Lima VP, Grun LK, Zambonin D, Anzolin E, Dos Santos Dias WW, Kus WP, Barbé-Tuana F, Battastini AMO, Worm PV, Figueiró F. Characterization of purinergic signaling in tumor-infiltrating lymphocytes from lower- and high-grade gliomas. Purinergic Signal 2024; 20:47-64. [PMID: 36964277 PMCID: PMC10828327 DOI: 10.1007/s11302-023-09931-4] [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: 12/08/2022] [Accepted: 03/06/2023] [Indexed: 03/26/2023] Open
Abstract
Malignant gliomas are highly heterogeneous glia-derived tumors that present an aggressive and invasive nature, with a dismal prognosis. The multi-dimensional interactions between glioma cells and other tumor microenvironment (TME) non-tumoral components constitute a challenge to finding successful treatment strategies. Several molecules, such as extracellular purines, participate in signaling events and support the immunosuppressive TME of glioma patients. The purinergic signaling and the ectoenzymes network involved in the metabolism of these extracellular nucleotides are still unexplored in the glioma TME, especially in lower-grade gliomas (LGG). Also, differences between IDH-mutant (IDH-Mut) versus wild-type (IDH-WT) gliomas are still unknown in this context. For the first time, to our knowledge, this study characterizes the TME of LGG, high-grade gliomas (HGG) IDH-Mut, and HGG IDH-WT patients regarding purinergic ectoenzymes and P1 receptors, focusing on tumor-infiltrating lymphocytes. Here, we show that ectoenzymes from both canonical and non-canonical pathways are increased in the TME when compared to the peripheral blood. We hypothesize this enhancement supports extracellular adenosine generation, hence increasing TME immunosuppression.
Collapse
Affiliation(s)
- Juliete Nathali Scholl
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Augusto Ferreira Weber
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Camila Kehl Dias
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Vinícius Pierdoná Lima
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Lucas Kich Grun
- Programa de Pós-Graduação Em Pediatria E Saúde da Criança, Escola de Medicina, PUCRS, Porto Alegre, RS, Brazil
| | - Diego Zambonin
- Departamento de Neurocirurgia, Hospital Cristo Redentor, Porto Alegre, Brazil
| | - Eduardo Anzolin
- Departamento de Neurocirurgia, Hospital Cristo Redentor, Porto Alegre, Brazil
| | | | | | - Florencia Barbé-Tuana
- Programa de Pós-Graduação Em Biologia Celular E Molecular, Escola de Ciências da Saúde E da Vida, PUCRS, Porto Alegre, RS, Brazil
| | - Ana Maria Oliveira Battastini
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil
| | - Paulo Valdeci Worm
- Departamento de Neurocirurgia, Hospital Cristo Redentor, Porto Alegre, Brazil
- Departmento de Cirurgia, Universidade Federal de Ciências da Saúde de Porto Alegre, Rio Grande Do Sul, Porto Alegre, Brazil
| | - Fabrício Figueiró
- Programa de Pós-Graduação Em Ciências Biológicas: Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil.
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, UFRGS, Porto Alegre, RS, Brazil.
| |
Collapse
|
25
|
Gostomczyk K, Marsool MDM, Tayyab H, Pandey A, Borowczak J, Macome F, Chacon J, Dave T, Maniewski M, Szylberg Ł. Targeting circulating tumor cells to prevent metastases. Hum Cell 2024; 37:101-120. [PMID: 37874534 PMCID: PMC10764589 DOI: 10.1007/s13577-023-00992-6] [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: 09/11/2023] [Accepted: 10/03/2023] [Indexed: 10/25/2023]
Abstract
Circulating tumor cells (CTCs) are cancer cells that detach from the primary tumor, enter the bloodstream or body fluids, and spread to other body parts, leading to metastasis. Their presence and characteristics have been linked to cancer progression and poor prognosis in different types of cancer. Analyzing CTCs can offer valuable information about tumors' genetic and molecular diversity, which is crucial for personalized therapy. Epithelial-mesenchymal transition (EMT) and the reverse process, mesenchymal-epithelial transition (MET), play a significant role in generating and disseminating CTCs. Certain proteins, such as EpCAM, vimentin, CD44, and TGM2, are vital in regulating EMT and MET and could be potential targets for therapies to prevent metastasis and serve as detection markers. Several devices, methods, and protocols have been developed for detecting CTCs with various applications. CTCs interact with different components of the tumor microenvironment. The interactions between CTCs and tumor-associated macrophages promote local inflammation and allow the cancer cells to evade the immune system, facilitating their attachment and invasion of distant metastatic sites. Consequently, targeting and eliminating CTCs hold promise in preventing metastasis and improving patient outcomes. Various approaches are being explored to reduce the volume of CTCs. By investigating and discussing targeted therapies, new insights can be gained into their potential effectiveness in inhibiting the spread of CTCs and thereby reducing metastasis. The development of such treatments offers great potential for enhancing patient outcomes and halting disease progression.
Collapse
Affiliation(s)
- Karol Gostomczyk
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Torun, Poland.
- University Hospital No. 2 Im. Dr Jan Biziel, Ujejskiego 75, 85-168, Bydgoszcz, Poland.
| | | | | | | | - Jędrzej Borowczak
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Torun, Poland
| | - Facundo Macome
- Universidad del Norte Santo Tomás de Aquino, San Miquel de Tucuman, Argentina
| | - Jose Chacon
- American University of Integrative Sciences, Cole Bay, Saint Martin, Barbados
| | - Tirth Dave
- Bukovinian State Medical University, Chernivtsi, Ukraine
| | - Mateusz Maniewski
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Torun, Poland
| | - Łukasz Szylberg
- Department of Obstetrics, Gynaecology and Oncology, Chair of Pathomorphology and Clinical Placentology, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University, Torun, Poland
- Department of Tumor Pathology and Pathomorphology, Oncology Centre, Prof. Franciszek Łukaszczyk Memorial Hospital, Bydgoszcz, Poland
- Chair of Pathology, Dr Jan Biziel Memorial University Hospital No. 2, Bydgoszcz, Poland
| |
Collapse
|
26
|
Kouthouridis S, Sotra A, Khan Z, Alvarado J, Raha S, Zhang B. Modeling the Progression of Placental Transport from Early- to Late-Stage Pregnancy by Tuning Trophoblast Differentiation and Vascularization. Adv Healthc Mater 2023; 12:e2301428. [PMID: 37830445 PMCID: PMC11468690 DOI: 10.1002/adhm.202301428] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 09/18/2023] [Indexed: 10/14/2023]
Abstract
The early-stage placental barrier is characterized by a lack of fetal circulation and by a thick trophoblastic barrier, whereas the later-stage placenta consists of vascularized chorionic villi encased in a thin, differentiated trophoblast layer, ideal for nutrient transport. In this work, predictive models of early- and late-stage placental transport are created using blastocyst-derived placental stem cells (PSCs) by modulating PSC differentiation and model vascularization. PSC differentiation results in a thinner, fused trophoblast layer, as well as an increase in human chorionic gonadotropin secretion, barrier permeability, and secretion of certain inflammatory cytokines, which are consistent with in vivo findings. Further, gene expression confirms this shift toward a differentiated trophoblast subtype. Vascularization results in a molecule type- and size-dependent change in dextran and insulin permeability. These results demonstrate that trophoblast differentiation and vascularization have critical effects on placental barrier permeability and that this model can be used as a predictive measure to assess fetal toxicity of xenobiotic substances at different stages of pregnancy.
Collapse
Affiliation(s)
- Sonya Kouthouridis
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Alexander Sotra
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Zaim Khan
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Justin Alvarado
- Department of Biochemistry and Biomedical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Sandeep Raha
- Department of Pediatrics and the Graduate Programme in Medical SciencesMcMaster UniversityHamiltonONL8S 4L8Canada
| | - Boyang Zhang
- Department of Chemical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
- School of Biomedical EngineeringMcMaster UniversityHamiltonONL8S 4L8Canada
| |
Collapse
|
27
|
Li Z, Liu Y, Guo P, Wei Y. Construction and validation of a novel angiogenesis pattern to predict prognosis and immunotherapy efficacy in colorectal cancer. Aging (Albany NY) 2023; 15:12413-12450. [PMID: 37938164 PMCID: PMC10683615 DOI: 10.18632/aging.205189] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 10/02/2023] [Indexed: 11/09/2023]
Abstract
BACKGROUND Evidence suggests that the tumor microenvironment (TME) affects the tumor active response to immunotherapy. Tumor angiogenesis is closely related to the TME. Nonetheless, the effects of angiogenesis on the TME of colorectal cancer (CRC) remain unknown. METHODS We comprehensively assessed the angiogenesis patterns in CRC based on 36 angiogenesis-related genes (ARGs). Subsequently, we evaluated the prognostic values and therapeutic sensitivities of angiogenesis patterns using multiple methods. We then performed the machine learning algorithm and functional experiments to identify the prognostic key ARGs. Ultimately, the regulation of gut microbiota on the expression of ARGs was further investigated by using whole genome sequencing. RESULTS Two angiogenesis clusters were identified and angiogenesis cluster B was characterized by increased stromal and immunity activation with unfavorable odds of survival. Further, an ARG_score including 9 ARGs to predict recurrence-free survival (RFS) was established and its predominant predictive ability was confirmed. The low ARG_score patients were characterized by a high mutation burden, high microsatellite instability, and immune activation with better prognosis. Moreover, patients with high KLK10 expression were associated with a hot tumor immune microenvironment, poorer immune checkpoint blocking treatment, and shorter survival. The in vitro experiments also indicated that Fusobacterium nucleatum (F.n) infection significantly induced KLK10 expression in CRC. CONCLUSIONS The quantification of angiogenesis patterns could contribute to predict TME characteristics, prognosis, and individualized immunotherapy strategies. Furthermore, our findings suggest that F.n may influence CRC progression through ARGs, which could serve as a clinical biomarker and therapeutic target for F.n-infected CRC patients.
Collapse
Affiliation(s)
- Zhiyong Li
- Department of Emergency Surgery, Peking University People’s Hospital, Xicheng, Beijing 100044, China
| | - Yang Liu
- Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo Second Hospital, Ningbo, Zhejiang 315010, China
| | - Peng Guo
- Department of Emergency Surgery, Peking University People’s Hospital, Xicheng, Beijing 100044, China
- Department of Emergency Medicine, Peking University People’s Hospital, Xicheng, Beijing 100044, China
- Laboratory of Surgery Oncology, Peking University People’s Hospital, Xicheng, Beijing 100044, China
| | - Yunwei Wei
- Department of Pancreatic and Gastrointestinal Surgery Division, Ningbo Second Hospital, Ningbo, Zhejiang 315010, China
- Ningbo Key Laboratory of Intestinal Microecology and Human Major Diseases, Ningbo, Zhejiang 315010, China
| |
Collapse
|
28
|
Shinada M, Kato D, Motegi T, Tsuboi M, Ikeda N, Aoki S, Iguchi T, Li T, Kodera Y, Ota R, Hashimoto Y, Takahashi Y, Chambers J, Uchida K, Kato Y, Nishimura R, Nakagawa T. Podoplanin Drives Amoeboid Invasion in Canine and Human Mucosal Melanoma. Mol Cancer Res 2023; 21:1205-1219. [PMID: 37493578 DOI: 10.1158/1541-7786.mcr-22-0929] [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: 11/19/2022] [Revised: 04/09/2023] [Accepted: 07/13/2023] [Indexed: 07/27/2023]
Abstract
Mucosal melanoma metastasizes at an early stage of the disease in human and dog. We revealed that overexpression of podoplanin in tumor invasion fronts (IF) was related to poor prognosis of dogs with mucosal melanoma. Moreover, podoplanin expressed in canine mucosal melanoma cells promotes proliferation and aggressive amoeboid invasion by activating Rho-associated kinase (ROCK)-myosin light chain 2 (MLC2) signaling. PDPN-ROCK-MLC2 signaling plays a role in cell-cycle arrest and cellular senescence escape as a mechanism for regulating proliferation. Podoplanin induces amoeboid invasion in the IFs of mouse xenografted tumor tissues, similar to canine mucosal melanoma clinical samples. We further identified that podoplanin expression was related to poor prognosis of human patients with mucosal melanoma, and human mucosal melanoma with podoplanin-high expression enriched gene signatures related to amoeboid invasion, similar to canine mucosal melanoma. Overall, we propose that podoplanin promotes canine and human mucosal melanoma metastasis by inducing aggressive amoeboid invasion and naturally occurring canine mucosal melanoma can be a novel research model for podoplanin expressing human mucosal melanoma. IMPLICATIONS Podoplanin could be a new therapeutic target to restrict the metastatic dissemination of canine and human mucosal melanoma.
Collapse
Affiliation(s)
- Masahiro Shinada
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Daiki Kato
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Tomoki Motegi
- Veterinary Medical Center, The University of Tokyo, Tokyo, Japan
| | - Masaya Tsuboi
- Veterinary Medical Center, The University of Tokyo, Tokyo, Japan
| | - Namiko Ikeda
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Susumu Aoki
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takaaki Iguchi
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Toshio Li
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuka Kodera
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Ryosuke Ota
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yuko Hashimoto
- Veterinary Medical Center, The University of Tokyo, Tokyo, Japan
| | - Yosuke Takahashi
- Veterinary Medical Center, The University of Tokyo, Tokyo, Japan
| | - James Chambers
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Kazuyuki Uchida
- Laboratory of Veterinary Pathology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Yukinari Kato
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Miyagi, Japan
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Miyagi, Japan
| | - Ryohei Nishimura
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| | - Takayuki Nakagawa
- Laboratory of Veterinary Surgery, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan
| |
Collapse
|
29
|
Sartorius D, Blume ML, Fleischer JR, Ghadimi M, Conradi LC, De Oliveira T. Implications of Rectal Cancer Radiotherapy on the Immune Microenvironment: Allies and Foes to Therapy Resistance and Patients' Outcome. Cancers (Basel) 2023; 15:5124. [PMID: 37958298 PMCID: PMC10650490 DOI: 10.3390/cancers15215124] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/16/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Aside from surgical resection, locally advanced rectal cancer is regularly treated with neoadjuvant chemoradiotherapy. Since the concept of cancer treatment has shifted from only focusing on tumor cells as drivers of disease progression towards a broader understanding including the dynamic tumor microenvironment (TME), the impact of radiotherapy on the TME and specifically the tumor immune microenvironment (TIME) is increasingly recognized. Both promoting as well as suppressing effects on anti-tumor immunity have been reported in response to rectal cancer (chemo-)radiotherapy and various targets for combination therapies are under investigation. A literature review was conducted searching the PubMed database for evidence regarding the pleiotropic effects of (chemo-)radiotherapy on the rectal cancer TIME, including alterations in cytokine levels, immune cell populations and activity as well as changes in immune checkpoint proteins. Radiotherapy can induce immune-stimulating and -suppressive alterations, potentially mediating radioresistance. The response is influenced by treatment modalities, including the dosage administered and the highly individual intrinsic pre-treatment immune status. Directly addressing the main immune cells of the TME, this review aims to highlight therapeutical implications since efficient rectal cancer treatment relies on personalized strategies combining conventional therapies with immune-modulating approaches, such as immune checkpoint inhibitors.
Collapse
Affiliation(s)
| | | | | | | | - Lena-Christin Conradi
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075 Göttingen, Germany; (D.S.); (M.L.B.); (J.R.F.); (M.G.)
| | - Tiago De Oliveira
- Department of General, Visceral and Pediatric Surgery, University Medical Center Göttingen, Robert-Koch-Straβe 40, 37075 Göttingen, Germany; (D.S.); (M.L.B.); (J.R.F.); (M.G.)
| |
Collapse
|
30
|
Zeng J, Zhu P, Tang Y, Zhang C, Ye C, Cheng S, Tian K, Yang B, Zeng W, Liu Y, Xian Z, Yu Y. Identification of pyroptosis-related subtypes and comprehensive analysis of characteristics of the tumor microenvironment infiltration in clear cell renal cell carcinoma. Sci Rep 2023; 13:16055. [PMID: 37749171 PMCID: PMC10519968 DOI: 10.1038/s41598-023-43023-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: 02/05/2023] [Accepted: 09/18/2023] [Indexed: 09/27/2023] Open
Abstract
Pyroptosis is a kind of programmed cell death triggered by the inflammasome. Growing evidence has revealed the crucial utility of pyroptosis in tumors. However, the potential mechanism of pyroptosis in clear cell renal cell carcinoma (ccRCC) is still unclear. In this research, we systematically analyze the genetic and transcriptional alterations of pyroptosis-related genes (PRGs) in ccRCC, identify pyroptosis-related subtypes, analyze the clinical and microenvironmental differences among different subtypes, develop a corresponding prognostic model to predict the prognosis of patients, and interpret the effect of pyroptosis on ccRCC microenvironment. This study provides a new perspective for a comprehensive understanding of the role of pyroptosis in ccRCC and its impact on the immune microenvironment, and a reliable scoring system was established to predict patients' prognosis.
Collapse
Affiliation(s)
- Jiayi Zeng
- Department of Urology, Guangdong Provincial People's Hospital's Nanhai Hospital, Foshan, China
| | - Ping Zhu
- Department of Immunology, School of Basic Medical Science, Southern Medical University, Guangzhou, China
| | - Yanlin Tang
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Changzheng Zhang
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Chujin Ye
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shouyu Cheng
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Kaiwen Tian
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Bowen Yang
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Weinan Zeng
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yanjun Liu
- Department of Immunology, School of Basic Medical Science, Southern Medical University, Guangzhou, China.
| | - Zhiyong Xian
- Department of Urology, Guangdong Provincial People's Hospital's Nanhai Hospital, Foshan, China.
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
| | - Yuming Yu
- Department of Urology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China.
| |
Collapse
|
31
|
Cao F, Qi Y, Wu W, Li X, Yang C. Single-cell and genetic multi-omics analysis combined with experiments confirmed the signature and potential targets of cuproptosis in hepatocellular carcinoma. Front Cell Dev Biol 2023; 11:1240390. [PMID: 37745297 PMCID: PMC10516581 DOI: 10.3389/fcell.2023.1240390] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/24/2023] [Indexed: 09/26/2023] Open
Abstract
Background: Cuproptosis, as a recently discovered type of programmed cell death, occupies a very important role in hepatocellular carcinoma (HCC) and provides new methods for immunotherapy; however, the functions of cuproptosis in HCC are still unclear. Methods: We first analyzed the transcriptome data and clinical information of 526 HCC patients using multiple algorithms in R language and extensively described the copy number variation, prognostic and immune infiltration characteristics of cuproptosis related genes (CRGs). Then, the hub CRG related genes associated with prognosis through LASSO and Cox regression analyses and constructed a prognostic prediction model including multiple molecular markers and clinicopathological parameters through training cohorts, then this model was verified by test cohorts. On the basis of the model, the clinicopathological indicators, immune infiltration and tumor microenvironment characteristics of HCC patients were further explored via bioinformation analysis. Then, We further explored the key gene biological function by single-cell analysis, cell viability and transwell experiments. Meantime, we also explored the molecular docking of the hub genes. Results: We have screened 5 hub genes associated with HCC prognosis and constructed a prognosis prediction scoring model. And the model results showed that patients in the high-risk group had poor prognosis and the expression levels of multiple immune markers, including PD-L1, CD276 and CTLA4, were higher than those patients in the low-risk group. We found a significant correlation between risk score and M0 macrophages and memory CD4+ T cells. And the single-cell analysis and molecular experiments showed that BEX1 were higher expressed in HCC tissues and deletion inhibited the proliferation, invasion and migration and EMT pathway of HCC cells. Finally, it was observed that BEX1 could bind to sorafenib to form a stable conformation. Conclusion: The study not only revealed the multiomics characteristics of CRGs in HCC but also constructed a new high-accuracy prognostic prediction model. Meanwhile, BEX1 were also identified as hub genes that can mediate the cuproptosis of hepatocytes as potential therapeutic targets for HCC.
Collapse
Affiliation(s)
- Feng Cao
- Department of General, Visceral and Transplantation Surgery, University Hospital RWTH Aachen, Aachen, Germany
| | - Yong Qi
- Department of General Surgery, The First Hospital of Anhui Medical University, Hefei, China
| | - Wenyong Wu
- Department of General Surgery, The First Hospital of Anhui Medical University, Hefei, China
| | - Xutong Li
- Department of Infectious Diseases, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Chuang Yang
- Department of Visceral, Transplant, Thoracic and Vascular Surgery, University Hospital of Leipzig, Leipzig, Germany
| |
Collapse
|
32
|
Zhang D, Chen S, Cao W, Geng N, Feng C. HALP score based on hemoglobin, albumin, lymphocyte and platelet can predict the prognosis of tongue squamous cell carcinoma patients. Heliyon 2023; 9:e20126. [PMID: 37809958 PMCID: PMC10559844 DOI: 10.1016/j.heliyon.2023.e20126] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2023] [Revised: 09/01/2023] [Accepted: 09/12/2023] [Indexed: 10/10/2023] Open
Abstract
Objective The preoperative hemoglobin, albumin, lymphocyte, and platelet (HALP) score, a comprehensive marker of nutritional and immunological status, has been found to be robust for tumor prognosis prediction. Here, we evaluated the use of HALP in the prognostic prediction of tongue squamous cell carcinoma (TSCC). Study design Patients with TSCC were retrospectively recruited from the years 2009-2019. Patient clinicopathological characteristics, along with preoperative blood parameters, were recorded on admission, and the cut-off HALP value was determined by X-tile software. Kaplan-Meier curves and Cox regression analyses were used to evaluate the predictive value of HALP for patient overall survival (OS) and disease-free survival (DFS). Results A total of 339 TSCC patients were enrolled. The optimal HALP threshold was 56 and the patients were divided into two groups according to their scores. The Kaplan-Meier analysis showed that patients in the high-HALP group experienced longer OS (p = 0.007) and DFS (p = 0.006) than those in the low-HALP group. Multivariate analysis showed that elevated HALP (p = 0.038) was an independent predictor of OS, while age (p = 0.008), T stage (p < 0.001), N stage (p = 0.020), and degree of tumor differentiation (p < 0.001) were risk factors. Conclusion The findings showed that the preoperative HALP score was an independent predictor of prognosis in patients with TSCC.
Collapse
Affiliation(s)
| | | | - Wei Cao
- Department of Stomatology, The Frist Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Ningbo Geng
- Department of Stomatology, The Frist Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, People's Republic of China
| | - Chongjin Feng
- Department of Stomatology, The Frist Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510080, People's Republic of China
| |
Collapse
|
33
|
Su C, Yu R, Hong X, Zhang P, Guo Y, Cai JC, Hou J. CXCR4 Expressed by Tumor-Infiltrating B Cells in Gastric Cancer Related to Survival in the Tumor Microenvironment: An Analysis Combining Single-Cell RNA Sequencing with Bulk RNA Sequencing. Int J Mol Sci 2023; 24:12890. [PMID: 37629071 PMCID: PMC10454711 DOI: 10.3390/ijms241612890] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/27/2023] Open
Abstract
According to the World Health Organization (WHO), gastric cancer (GC) is the fourth leading cause of tumor-related mortality globally and one of the most prevalent malignant tumors. To better understand the role of tumor-infiltrating B cells (TIBs) in GC, this work used single-cell RNA sequencing (scRNA-Seq) and bulk RNA sequencing (bulk RNA-Seq) data to identify candidate hub genes. Both scRNA-Seq and bulk RNA-Seq data for stomach adenocarcinoma (STAD) were obtained from the GEO and TCGA databases, respectively. Using scRNA-seq data, the FindNeighbors and FindClusters tools were used to group the cells into distinct groups. Immune cell clusters were sought in the massive RNA-seq expression matrix using the single-sample gene set enrichment analysis (ssGSEA). The expression profiles were used in Weighted Gene Coexpression Network Analysis (WGCNA) to build TCGA's gene coexpression networks. Next, univariate Cox regression, LASSO regression, and Kaplan-Meier analyses were used to identify hub genes in scRNA-seq data from sequential B-cell analyses. Finally, we examined the correlation between the hub genes and TIBs utilizing the TISIDB database. We confirmed the immune-related markers in clinical validation samples using reverse transcriptase polymerase chain reaction (RT-PCR) and immunohistochemistry (IHC). 15 cell clusters were classified in the scRNA-seq database. According to the WGCNA findings, the green module is most associated with cancer and B cells. The intersection of 12 genes in two separate datasets (scRNA and bulk) was attained for further analysis. However, survival studies revealed that increased C-X-C motif chemokine receptor 4 (CXCR4) expression was linked to worse overall survival. CXCR4 expression is correlated with active, immature, and memory B cells in STAD were identified. Finally, RT-PCR and IHC assays verified that in GC, CXCR4 is overexpressed, and its expression level correlates with TIBs. We used scRNA-Seq and bulk RNA-Seq to study STAD's cellular composition. We found that CXCR4 is highly expressed by TIBs in GC, suggesting that it may serve as a hub gene for these cells and a starting point for future research into the molecular mechanisms by which these immune cells gain access to tumors and potentially identify therapeutic targets.
Collapse
Affiliation(s)
- Chen Su
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China; (C.S.); (R.Y.)
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361102, China; (X.H.); (P.Z.); (Y.G.)
| | - Rong Yu
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China; (C.S.); (R.Y.)
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361102, China; (X.H.); (P.Z.); (Y.G.)
| | - Xiaoquan Hong
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361102, China; (X.H.); (P.Z.); (Y.G.)
- Department of General Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China
| | - Panpan Zhang
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361102, China; (X.H.); (P.Z.); (Y.G.)
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Yingying Guo
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361102, China; (X.H.); (P.Z.); (Y.G.)
- State Key Laboratory of Stress Cell Biology, School of Life Sciences, Xiamen University, Xiamen 361102, China
| | - Jian-Chun Cai
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China; (C.S.); (R.Y.)
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361102, China; (X.H.); (P.Z.); (Y.G.)
| | - Jingjing Hou
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361102, China; (C.S.); (R.Y.)
- Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361102, China; (X.H.); (P.Z.); (Y.G.)
| |
Collapse
|
34
|
Panaampon J, Zhou Y, Saengboonmee C. Metformin as a booster of cancer immunotherapy. Int Immunopharmacol 2023; 121:110528. [PMID: 37364322 DOI: 10.1016/j.intimp.2023.110528] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/12/2023] [Accepted: 06/15/2023] [Indexed: 06/28/2023]
Abstract
Metformin, a biguanide antidiabetic, has been studied for its repurposing effects in oncology. Although a modest effect was observed in a single-agent regimen, metformin can synergize the anti-tumor effects of other modalities. The promising combination for cancer treatment is with immunotherapy. Despite high efficacy for some cancers, immunotherapy could be limited by modulation of the tumor immune microenvironment and the immune exhaustion of cytotoxic immune cells. Combining immunotherapy with metformin, thus, exerted a rescuing effect of immunotherapy and potentiated the anti-tumor effects of each other. Although not fully understood, metformin shows promoting effects of immunotherapy by several mechanisms. Those proposed mechanisms have been partially proven and are suggested for possible therapeutic strategies for cancer treatment. In this review, a state-of-the-art of metformin's boosting effects on immunotherapy is reviewed and discussed. The future directions for metformin research in preclinical and clinical immunotherapy are also suggested.
Collapse
Affiliation(s)
- Jutatip Panaampon
- Division of Hematologic Neoplasia, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02215, USA; Division of Hematopoiesis, Joint Research Center for Human Retrovirus Infection, Kumamoto University, Kumamoto 8600811, Japan
| | - Yubin Zhou
- Department of Thoracic Surgery, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan 610072, China; Chinese Academy of Sciences Sichuan Translational Medicine Research Hospital, Chengdu, Sichuan 610072, China
| | - Charupong Saengboonmee
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen 40002, Thailand; Center for Translational Medicine, Faculty of Medicine, Khon Kaen University 40002, Thailand.
| |
Collapse
|
35
|
Gautam SK, Batra SK, Jain M. Molecular and metabolic regulation of immunosuppression in metastatic pancreatic ductal adenocarcinoma. Mol Cancer 2023; 22:118. [PMID: 37488598 PMCID: PMC10367391 DOI: 10.1186/s12943-023-01813-y] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/23/2023] [Indexed: 07/26/2023] Open
Abstract
Immunosuppression is a hallmark of pancreatic ductal adenocarcinoma (PDAC), contributing to early metastasis and poor patient survival. Compared to the localized tumors, current standard-of-care therapies have failed to improve the survival of patients with metastatic PDAC, that necessecitates exploration of novel therapeutic approaches. While immunotherapies such as immune checkpoint blockade (ICB) and therapeutic vaccines have emerged as promising treatment modalities in certain cancers, limited responses have been achieved in PDAC. Therefore, specific mechanisms regulating the poor response to immunotherapy must be explored. The immunosuppressive microenvironment driven by oncogenic mutations, tumor secretome, non-coding RNAs, and tumor microbiome persists throughout PDAC progression, allowing neoplastic cells to grow locally and metastasize distantly. The metastatic cells escaping the host immune surveillance are unique in molecular, immunological, and metabolic characteristics. Following chemokine and exosomal guidance, these cells metastasize to the organ-specific pre-metastatic niches (PMNs) constituted by local resident cells, stromal fibroblasts, and suppressive immune cells, such as the metastasis-associated macrophages, neutrophils, and myeloid-derived suppressor cells. The metastatic immune microenvironment differs from primary tumors in stromal and immune cell composition, functionality, and metabolism. Thus far, multiple molecular and metabolic pathways, distinct from primary tumors, have been identified that dampen immune effector functions, confounding the immunotherapy response in metastatic PDAC. This review describes major immunoregulatory pathways that contribute to the metastatic progression and limit immunotherapy outcomes in PDAC. Overall, we highlight the therapeutic vulnerabilities attributable to immunosuppressive factors and discuss whether targeting these molecular and immunological "hot spots" could improve the outcomes of PDAC immunotherapies.
Collapse
Affiliation(s)
- Shailendra K Gautam
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| | - Surinder K Batra
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA
| | - Maneesh Jain
- Department of Biochemistry and Molecular Biology, College of Medicine, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
- Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE, 68198, USA.
| |
Collapse
|
36
|
Borella R, Paolini A, Aramini B, Gibellini L, Masciale V, Lo Tartaro D, Dominici M, De Biasi S, Cossarizza A. Multiparametric analysis of tumor infiltrating lymphocytes in solid tumors. Methods Cell Biol 2023; 195:39-70. [PMID: 40180454 DOI: 10.1016/bs.mcb.2023.03.006] [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] [Indexed: 04/05/2025]
Abstract
The use of single-cell technologies in characterizing the interactions between immune and cancer cells is in continuous expansion. Indeed, the combination of different single-cell approaches enables the definition of novel phenotypic and functional aspects of the immune cells infiltrating the tumor microenvironment (TME). This approach is promoting the discovery of relevant and reliable predictive biomarkers, along with the development of new promising treatments. In this chapter, we describe the main subsets of tumor-infiltrating lymphocytes from a phenotypical and functional point of view and discuss the use of single-cell technologies used to characterize these cell populations within TME.
Collapse
Affiliation(s)
- Rebecca Borella
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum-University of Bologna and G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Masciale
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy.
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy; Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy
| |
Collapse
|
37
|
Zhang Y, Xue W, Xu C, Nan Y, Mei S, Ju D, Wang S, Zhang X. Innate Immunity in Cancer Biology and Therapy. Int J Mol Sci 2023; 24:11233. [PMID: 37510993 PMCID: PMC10379825 DOI: 10.3390/ijms241411233] [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: 05/21/2023] [Revised: 07/05/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Immunotherapies including adaptive immune checkpoint inhibitors (ICIs) and chimeric antigen receptor (CAR) T cells, have developed the treatment of cancer in clinic, and most of them focus on activating T cell immunity. Although these strategies have obtained unprecedented clinical responses, only limited subsets of cancer patients could receive long-term benefits, highlighting the demand for identifying novel targets for the new era of tumor immunotherapy. Innate immunity has been demonstrated to play a determinative role in the tumor microenvironment (TME) and influence the clinical outcomes of tumor patients. A thorough comprehension of the innate immune cells that infiltrate tumors would allow for the development of new therapeutics. In this review, we outline the role and mechanism of innate immunity in TME. Moreover, we discuss innate immunity-based cancer immunotherapy in basic and clinical studies. Finally, we summarize the challenges in sufficiently motivating innate immune responses and the corresponding strategies and measures to improve anti-tumor efficacy. This review could aid the comprehension of innate immunity and inspire the creation of brand-new immunotherapies for the treatment of cancer.
Collapse
Affiliation(s)
- Yuxia Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Wenjing Xue
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Caili Xu
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yanyang Nan
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shuang Mei
- Shanghai Tinova Immune Therapeutics Co., Ltd., Shanghai 201413, China
| | - Dianwen Ju
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Shaofei Wang
- Department of Cellular and Genetic Medicine, School of Basic Medical Sciences, Fudan University, Shanghai 200032, China
| | - Xuyao Zhang
- Department of Biological Medicines & Shanghai Engineering Research Center of Immunotherapeutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| |
Collapse
|
38
|
Li Y, Luo Y. Spatial Transcriptomic Cell-type Deconvolution Using Graph Neural Networks. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.10.532112. [PMID: 37333198 PMCID: PMC10274700 DOI: 10.1101/2023.03.10.532112] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/20/2023]
Abstract
Spatially resolved transcriptomics performs high-throughput measurement of transcriptomes while preserving spatial information about the cellular organizations. However, many spatially resolved transcriptomic technologies can only distinguish spots consisting of a mixture of cells instead of working at single-cell resolution. Here, we present STdGCN, a graph neural network model designed for cell type deconvolution of spatial transcriptomic (ST) data that can leverage abundant single-cell RNA sequencing (scRNA-seq) data as reference. STdGCN is the first model incorporating the expression profiles from single cell data as well as the spatial localization information from the ST data for cell type deconvolution. Extensive benchmarking experiments on multiple ST datasets showed that STdGCN outperformed 14 published state-of-the-art models. Applied to a human breast cancer Visium dataset, STdGCN discerned spatial distributions between stroma, lymphocytes and cancer cells for tumor microenvironment dissection. In a human heart ST dataset, STdGCN detected the changes of potential endothelial-cardiomyocyte communications during tissue development.
Collapse
Affiliation(s)
- Yawei Li
- Department of Preventive Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Collaborative AI in Healthcare, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| | - Yuan Luo
- Department of Preventive Medicine, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
- Center for Collaborative AI in Healthcare, Northwestern University, Feinberg School of Medicine, Chicago, IL 60611, USA
| |
Collapse
|
39
|
Xiao Y, Zhou H, Chen Y, Liu L, Wu Q, Li H, Lin P, Li J, Wu J, Tang L. A novel anoikis-related gene prognostic signature and its correlation with the immune microenvironment in colorectal cancer. Front Genet 2023; 14:1186862. [PMID: 37323657 PMCID: PMC10265740 DOI: 10.3389/fgene.2023.1186862] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 05/15/2023] [Indexed: 06/17/2023] Open
Abstract
Background: Anoikis is a type of apoptosis associated with cell detachment. Resistance to anoikis is a focal point of tumor metastasis. This study aimed to explore the relationship among anoikis-related genes (ARGs), immune infiltration, and prognosis in colorectal cancer (CRC). Methods: The transcriptome profile and clinical data on patients with CRC were retrieved from The Cancer Genome Atlas and Gene Expression Omnibus databases. Patients were divided into two clusters based on the expression of ARGs. Differences between the two ARG molecular subtypes were analyzed in terms of prognosis, functional enrichment, gene mutation frequency, and immune cell infiltration. An ARG-related prognostic signature for predicting overall survival in patients with CRC was developed and validated using absolute value convergence and selection operator (LASSO) regression analysis. The correlation between the signature risk score and clinicopathological features, immune cell infiltration, immune typing, and immunotherapy response was analyzed. The risk score combined with clinicopathological characteristics was used to construct a nomogram to assess CRC patients' prognosis. Results: Overall, 151 ARGs were differentially expressed in CRC. Two ARG subtypes, namely, ARG-high and ARG-low groups, were identified and correlated with CRC prognosis. The gene mutation frequency and immune, stromal, and ESTIMATE scores of the ARG-high group were higher than those of the ARG-low group. Moreover, CD8, natural killer cells, M1 macrophages, human leukocyte antigen (HLA), and immune checkpoint-related genes were significantly increased in the ARG-high group. An optimized 25-gene CRC prognostic signature was successfully constructed, and its prognostic predictive ability was validated. The high-risk score was correlated with T, N, M, and TNM stages. Risk scores were negatively correlated with dendritic cells, eosinophils, and CD4 cells, and significantly positively correlated with regulatory T cells. Patients in the high-risk group were more likely to exhibit immune unresponsiveness. Finally, the nomogram model was constructed and showed good prognostic predictive power. Conclusion: ARGs are associated with clinicopathological features and the prognosis of CRC, and play important roles in the immune microenvironment. Herein, we underpinned the usefulness of ARGs in CRC to develop more effective immunotherapy techniques.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Junxin Wu
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lirui Tang
- Department of Radiation Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| |
Collapse
|
40
|
Selvaggi F, Catalano T, Lattanzio R, Cotellese R, Aceto GM. Wingless/It/β-catenin signaling in liver metastasis from colorectal cancer: A focus on biological mechanisms and therapeutic opportunities. World J Gastroenterol 2023; 29:2764-2783. [PMID: 37274070 PMCID: PMC10237106 DOI: 10.3748/wjg.v29.i18.2764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 02/28/2023] [Accepted: 04/17/2023] [Indexed: 05/11/2023] Open
Abstract
The liver is the most common site of metastases in patients with colorectal cancer. Colorectal liver metastases (CRLMs) are the result of molecular mechanisms that involve different cells of the liver microenvironment. The aberrant activation of Wingless/It (Wnt)/β-catenin signals downstream of Wnt ligands initially drives the oncogenic transformation of the colon epithelium, but also the progression of metastatization through the epithelial-mesenchymal transition/mesenchymal-epithelial transition interactions. In liver microenvironment, metastatic cells can also survive and adapt through dormancy, which makes them less susceptible to pro-apoptotic signals and therapies. Treatment of CRLMs is challenging due to its variability and heterogeneity. Advances in surgery and oncology have been made in the last decade and a pivotal role for Wnt/β-catenin pathway has been re-cognized in chemoresistance. At the state of art, there is a lack of clear understanding of why and how this occurs and thus where exactly the opportunities for developing anti-CRLMs therapies may lie. In this review, current knowledge on the involvement of Wnt signaling in the development of CRLMs was considered. In addition, an overview of useful biomarkers with a revision of surgical and non-surgical therapies currently accepted in the clinical practice for colorectal liver metastasis patients were provided.
Collapse
Affiliation(s)
- Federico Selvaggi
- Department of Surgical, ASL2 Lanciano-Vasto-Chieti, Ospedale Clinicizzato SS Annunziata of Chieti, Chieti 66100, Italy
| | - Teresa Catalano
- Department of Clinical and Experimental Medicine, University of Messina, Messina 98125, Italy
| | - Rossano Lattanzio
- Department of Innovative Technologies in Medicine & Dentistry, University “G. d’Annunzio” Chieti-Pescara, Chieti 66100, Italy
| | - Roberto Cotellese
- Department of Medical, Oral and Biotechnological Sciences, University “G. d’Annunzio” Chieti-Pescara, Chieti 66100, Italy
- Villa Serena Foundation for Research, Villa Serena - Del Dott. L. Petruzzi, Città Sant’Angelo 65013, Pescara, Italy
| | - Gitana Maria Aceto
- Department of Medical, Oral and Biotechnological Sciences, “G. d’Annunzio” University of Chieti-Pescara, Chieti 66100, Italy
| |
Collapse
|
41
|
Genduso S, Freytag V, Schetler D, Kirchner L, Schiecke A, Maar H, Wicklein D, Gebauer F, Bröker K, Stürken C, Milde-Langosch K, Oliveira-Ferrer L, Ricklefs FL, Ewald F, Wolters-Eisfeld G, Riecken K, Unrau L, Krause L, Bohnenberger H, Offermann A, Perner S, Sebens S, Lamszus K, Diehl L, Linder S, Jücker M, Schumacher U, Lange T. Tumor cell integrin β4 and tumor stroma E-/P-selectin cooperatively regulate tumor growth in vivo. J Hematol Oncol 2023; 16:23. [PMID: 36932441 PMCID: PMC10022201 DOI: 10.1186/s13045-023-01413-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2022] [Accepted: 02/13/2023] [Indexed: 03/19/2023] Open
Abstract
BACKGROUND The immunological composition of the tumor microenvironment has a decisive influence on the biological course of cancer and is therefore of profound clinical relevance. In this study, we analyzed the cooperative effects of integrin β4 (ITGB4) on tumor cells and E-/P-selectin on endothelial cells within the tumor stroma for regulating tumor growth by shaping the local and systemic immune environment. METHODS We used several preclinical mouse models for different solid human cancer types (xenograft and syngeneic) to explore the role of ITGB4 (shRNA-mediated knockdown in tumor cells) and E-/P-selectins (knockout in mice) for tumor growth; effects on apoptosis, proliferation and intratumoral signaling pathways were determined by histological and biochemical methods and 3D in vitro experiments; changes in the intratumoral and systemic immune cell composition were determined by flow cytometry and immunohistochemistry; chemokine levels and their attracting potential were measured by ELISA and 3D invasion assays. RESULTS We observed a very robust synergism between ITGB4 and E-/P-selectin for the regulation of tumor growth, accompanied by an increased recruitment of CD11b+ Gr-1Hi cells with low granularity (i.e., myeloid-derived suppressor cells, MDSCs) specifically into ITGB4-depleted tumors. ITGB4-depleted tumors undergo apoptosis and actively attract MDSCs, well-known to promote tumor growth in several cancers, via increased secretion of different chemokines. MDSC trafficking into tumors crucially depends on E-/P-selectin expression. Analyses of clinical samples confirmed an inverse relationship between ITGB4 expression in tumors and number of tumor-infiltrating leukocytes. CONCLUSIONS These findings suggest a distinct vulnerability of ITGB4Lo tumors for MDSC-directed immunotherapies.
Collapse
Affiliation(s)
- Sandra Genduso
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Vera Freytag
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Daniela Schetler
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Lennart Kirchner
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Alina Schiecke
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Hanna Maar
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Institute of Anatomy I, Cancer Center Central Germany, Jena University Hospital, Teichgraben 7, 07743, Jena, Germany
| | - Daniel Wicklein
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of Anatomy and Cell Biology, University of Marburg, Robert-Koch-Strasse 8, 35037, Marburg, Germany
| | - Florian Gebauer
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of General, Visceral and Thoracic Surgery, University Hospital Cologne, Kerpener Strasse 62, 50937, Cologne, Germany
| | - Katharina Bröker
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
| | - Christine Stürken
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Faculty of Medicine, MSH Medical School Hamburg, Medical University, 20251, Hamburg, Germany
| | - Karin Milde-Langosch
- Department of Gynecology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | - Franz L Ricklefs
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Florian Ewald
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Gerrit Wolters-Eisfeld
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Department of Pediatric Hematology and Oncology, Research Institute Childrens' Cancer Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Kristoffer Riecken
- Research Department Cell and Gene Therapy, Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Ludmilla Unrau
- Institue of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Krause
- Institute of Medical Biometry and Epidemiology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hanibal Bohnenberger
- Institute of Pathology, University Medical Center Göttingen, Robert-Koch-Strasse 40, 37075, Göttingen, Germany
| | - Anne Offermann
- Institute of Pathology, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Sven Perner
- Institute of Pathology, University of Lübeck and University Medical Center Schleswig-Holstein, Campus Lübeck, Ratzeburger Allee 160, 23538, Lübeck, Germany
| | - Susanne Sebens
- Institute for Experimental Cancer Research, Kiel University (CAU) and University Medical Center Schleswig-Holstein, Campus Kiel, Arnold-Heller-Strasse 3, 24105, Kiel, Germany
| | - Katrin Lamszus
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Linda Diehl
- Institue of Experimental Immunology and Hepatology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Stefan Linder
- Institute of Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manfred Jücker
- Institute of Biochemistry and Signal Transduction, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Udo Schumacher
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany
- Medical School Berlin, Leipziger Platz 10, 10117, Berlin, Germany
| | - Tobias Lange
- Institute of Anatomy and Experimental Morphology, University Medical Center Hamburg-Eppendorf, Martinistrasse 52, 20246, Hamburg, Germany.
- Institute of Anatomy I, Cancer Center Central Germany, Jena University Hospital, Teichgraben 7, 07743, Jena, Germany.
| |
Collapse
|
42
|
Bray J, Eward W, Breen M. Evaluating the relevance of surgical margins. Part one: The problems with current methodology. Vet Comp Oncol 2023; 21:1-11. [PMID: 36308442 DOI: 10.1111/vco.12865] [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: 05/10/2022] [Revised: 10/11/2022] [Accepted: 10/24/2022] [Indexed: 11/28/2022]
Abstract
The goal of cancer surgery is to achieve a "clean" microscopic resection, with no residual tumour remaining in the wound. To achieve that goal, the surgeon typically incorporates a measured buffer of grossly normal tissue about the entire circumference of the tumour. Microscopic analysis of the resection boundaries is then performed to determine if all traces of the tumour have been completely removed. This analysis is thought to provide a surrogate indication as to the likelihood for that tumour to recur after surgery. However, it is recognised that tumour recurrence may not occur even when microscopic evidence of tumour has been identified at the resection margins, and recurrence can also occur when conventional histology has considered the tumour to have been completely removed. The explanations for this dichotomy are numerous and include technical and practical limitations of the processing methodology, and also several surgeon-related and tumour-related reasons. Ultimately, the inability to confidently determine when a tumour has been removed sufficiently to prevent recurrence can impact on the ability to provide owners with confident treatment advice. In this article, the authors describe the challenges with defining the true extent of the tumour margin from the perspective of the surgeon, the pathologist and the tumour. The authors also provide an analysis of why our current efforts to ensure that all traces of the local tumour have been successfully removed may provide an imperfect assessment of the risk of recurrence.
Collapse
Affiliation(s)
| | - Will Eward
- Duke Cancer Center, Durham, North Carolina, USA
| | - Matthew Breen
- College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
43
|
Szczerba K, Stokowa-Soltys K. What Is the Correlation between Preeclampsia and Cancer? The Important Role of Tachykinins and Transition Metal Ions. Pharmaceuticals (Basel) 2023; 16:366. [PMID: 36986466 PMCID: PMC10058266 DOI: 10.3390/ph16030366] [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: 12/15/2022] [Revised: 02/09/2023] [Accepted: 02/24/2023] [Indexed: 03/06/2023] Open
Abstract
Metal ions are irreplaceable in many biological processes. They are components of numerous metalloproteins and serve as cofactors or structural elements for enzymes. Interestingly, iron, copper and zinc play important roles in accelerating or preventing neoplastic cell transformation. Noteworthily, a lot of proliferative and invasive mechanisms are exploited by both malignant tumors and pregnancy. Cancer cells, as well as developing placenta cells, create a microenvironment supportive of immunologic privilege and angiogenesis. Therefore, pregnancy and cancer progression share many similarities. Moreover, during preeclampsia and cancer, significant changes in relevant trace element concentrations, tachykinin levels, expressions of neurokinin receptors, oxidative stress and angiogenic imbalance are observed. This sheds a new light on the role of metal ions and tachykinins in cancer progression and pregnancy, especially in preeclamptic women.
Collapse
Affiliation(s)
| | - Kamila Stokowa-Soltys
- Faculty of Chemistry, University of Wroclaw, F. Joliot-Curie 14, 50-383 Wroclaw, Poland
| |
Collapse
|
44
|
Identification of Cuproptosis-Related Subtypes in Lung Cancer, Characterization of Tumor Microenvironment Infiltration, and Establishment of a Prognostic Model. BIOMED RESEARCH INTERNATIONAL 2022; 2022:7406636. [PMID: 36588537 PMCID: PMC9797313 DOI: 10.1155/2022/7406636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 10/21/2022] [Accepted: 11/07/2022] [Indexed: 12/24/2022]
Abstract
Cuproptosis, a recently found kind of programmed cell death, has been linked to tumor development, prognosis, and therapeutic response. The roles of cuproptosis-related genes (CRG) in the tumor microenvironment (TME) are, nevertheless, unknown. We evaluated alterations in CRG and assessed the related expression patterns in 1445 lung cancer (LC) samples from three separate datasets, analyzing genetic, and transcriptional domains. We discovered two separate molecular subtypes of CRG and discovered that various subtypes of CRG were connected with patient clinical features and prognosis. Furthermore, we discovered connections between distinct CRG subtypes and TME cell infiltration features. The CRG_score was then developed and validated for predicting overall survival (OS). Following that, we investigated the relationship between CRG_score and the cancer stem cell (CSC) index and chemotherapeutic treatment sensitivity. In addition, we created a very accurate nomogram to increase the clinical usefulness of CRG_score. The potential roles of CRG in the tumor-immune-microenvironment, clinical characteristics, and prognosis in LC are demonstrated by our multiplex study. These findings expand our understanding of CRG in LC and may open up new options for assessing LC patients' prognosis and generating more effective immunotherapeutic treatments.
Collapse
|
45
|
Liu Z, Liu B, Feng C, Li C, Wang H, Zhang H, Liu P, Li Z, He S, Tu C. Molecular characterization of immunogenic cell death indicates prognosis and tumor microenvironment infiltration in osteosarcoma. Front Immunol 2022; 13:1071636. [PMID: 36569869 PMCID: PMC9780438 DOI: 10.3389/fimmu.2022.1071636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Accepted: 11/25/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Osteosarcoma (OS) is a highly aggressive bone malignancy with a poor prognosis, mainly in children and adolescents. Immunogenic cell death (ICD) is classified as a type of programmed cell death associated with the tumor immune microenvironment, prognosis, and immunotherapy. However, the feature of the ICD molecular subtype and the related tumor microenvironment (TME) and immune cell infiltration has not been carefully investigated in OS. Methods The ICD-related genes were extracted from previous studies, and the RNA expression profiles and corresponding data of OS were downloaded from The Cancer Genome Atlas and Gene Expression Omnibus database. The ICD-related molecular subtypes were classed by the "ConsensusclusterPlus" package and the construction of ICD-related signatures through univariate regression analysis. ROC curves, independent analysis, and internal validation were used to evaluate signature performance. Moreover, a series of bioinformatic analyses were used for Immunotherapy efficacy, tumor immune microenvironments, and chemotherapeutic drug sensitivity between the high- and low-risk groups. Results Herein, we identified two ICD-related subtypes and found significant heterogeneity in clinical prognosis, TME, and immune response signaling among distinct ICD subtypes. Subsequently, a novel ICD-related prognostic signature was developed to determine its predictive performance in OS. Also, a highly accurate nomogram was then constructed to improve the clinical applicability of the novel ICD-related signature. Furthermore, we observed significant correlations between ICD risk score and TME, immunotherapy response, and chemotherapeutic drug sensitivity. Notably, the in vitro experiments further verified that high GALNT14 expression is closely associated with poor prognosis and malignant progress of OS. Discussion Hence, we identified and validated that the novel ICD-related signature could serve as a promising biomarker for the OS's prognosis, chemotherapy, and immunotherapy response prediction, providing guidance for personalized and accurate immunotherapy strategies for OS.
Collapse
Affiliation(s)
- Zhongyue Liu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Binfeng Liu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chengyao Feng
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Chenbei Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Haixia Zhang
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Ping Liu
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhihong Li
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shasha He
- Department of Oncology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,*Correspondence: Shasha He, ; Chao Tu,
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China,*Correspondence: Shasha He, ; Chao Tu,
| |
Collapse
|
46
|
STEAP3 can predict the prognosis and shape the tumor microenvironment of clear cell renal cell carcinoma. BMC Cancer 2022; 22:1204. [PMID: 36424540 PMCID: PMC9686107 DOI: 10.1186/s12885-022-10313-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Accepted: 11/14/2022] [Indexed: 11/25/2022] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a common malignant tumor of the urinary system characterized by poor prognosis and difficult treatment. It has been reported that iron metabolism dysregulation is a common phenomenon in ccRCC and is closely related to the process of ccRCC. But still now, the exact function and underlying mechanisms of iron metabolism dysregulation in ccRCC have not been fully elucidated. In this study, we comprehensively investigated the prognostic value and potential role of STEAP3 (an iron metabolism-related gene) in ccRCC. STEAP3 is significantly up-regulated in ccRCC. High STEAP3 expression is associated with gender, hemoglobin level, pathological grade, tumor stage and significantly predicts an unfavorable prognosis of ccRCC patients. Functional enrichment analysis and evaluation of the tumor microenvironment indicated that STEAP3 was involved in the remodeling of tumor extracellular matrix and the shaping of an immune-suppressive tumor microenvironment to promote tumor metastasis and evade immune killing. Besides, the expression of STEAP3 is also associated with the expression of various immune checkpoint molecules and the IC50 of targeted drugs. Finally, we verified STEAP3 by RT-qPCR and IHC staining. In conclusion, we found that STEAP3 can serve as a candidate prognostic biomarker for ccRCC, and targeting STEAP3 and its biological processes may provide new references for the individualized treatment of ccRCC.
Collapse
|
47
|
Mirzayans R, Murray D. What Are the Reasons for Continuing Failures in Cancer Therapy? Are Misleading/Inappropriate Preclinical Assays to Be Blamed? Might Some Modern Therapies Cause More Harm than Benefit? Int J Mol Sci 2022; 23:13217. [PMID: 36362004 PMCID: PMC9655591 DOI: 10.3390/ijms232113217] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 10/28/2022] [Accepted: 10/28/2022] [Indexed: 07/30/2023] Open
Abstract
Over 50 years of cancer research has resulted in the generation of massive amounts of information, but relatively little progress has been made in the treatment of patients with solid tumors, except for extending their survival for a few months at best. Here, we will briefly discuss some of the reasons for this failure, focusing on the limitations and sometimes misunderstanding of the clinical relevance of preclinical assays that are widely used to identify novel anticancer drugs and treatment strategies (e.g., "synthetic lethality"). These include colony formation, apoptosis (e.g., caspase-3 activation), immunoblotting, and high-content multiwell plate cell-based assays, as well as tumor growth studies in animal models. A major limitation is that such assays are rarely designed to recapitulate the tumor repopulating properties associated with therapy-induced cancer cell dormancy (durable proliferation arrest) reflecting, for example, premature senescence, polyploidy and/or multinucleation. Furthermore, pro-survival properties of apoptotic cancer cells through phoenix rising, failed apoptosis, and/or anastasis (return from the brink of death), as well as cancer immunoediting and the impact of therapeutic agents on interactions between cancer and immune cells are often overlooked in preclinical studies. A brief review of the history of cancer research makes one wonder if modern strategies for treating patients with solid tumors may sometimes cause more harm than benefit.
Collapse
|
48
|
Zhou Y, Li X, Long G, Tao Y, Zhou L, Tang J. Identification and validation of a tyrosine metabolism-related prognostic prediction model and characterization of the tumor microenvironment infiltration in hepatocellular carcinoma. Front Immunol 2022; 13:994259. [PMID: 36341373 PMCID: PMC9633179 DOI: 10.3389/fimmu.2022.994259] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 10/04/2022] [Indexed: 08/05/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is an aggressive and heterogeneous disease characterized by high morbidity and mortality. The liver is the vital organ that participates in tyrosine catabolism, and abnormal tyrosine metabolism could cause various diseases, including HCC. Besides, the tumor immune microenvironment is involved in carcinogenesis and can influence the patients' clinical outcomes. However, the potential role of tyrosine metabolism pattern and immune molecular signature is poorly understood in HCC. METHODS Gene expression, somatic mutations, copy number variation data, and clinicopathological information of HCC were downloaded from The Cancer Genome Atlas (TCGA) database. GSE14520 from the Gene Expression Omnibus (GEO) databases was used as a validation dataset. We performed unsupervised consensus clustering of tyrosine metabolism-related genes (TRGs) and classified patients into distinct molecular subtypes. We used ESTIMATE algorithms to evaluate the immune infiltration. We then applied LASSO Cox regression to establish the TRGs risk model and validated its predictive performance. RESULTS In this study, we first described the alterations of 42 TRGs in HCC cohorts and characterized the clinicopathological characteristics and tumor microenvironmental landscape of the two distinct subtypes. We then established a tyrosine metabolism-related scoring system and identified five TRGs, which were highly correlated with prognosis and representative of this gene set, namely METTL6, GSTZ1, ADH4, ADH1A, and LCMT1. Patients in the high-risk group had an inferior prognosis. Univariate and multivariate Cox proportional hazards regression analysis also showed that the tyrosine metabolism-related signature was an independent prognostic indicator. Besides, receiver operating characteristic curve (ROC) analysis demonstrated the predictive accuracy of the TRGs signature that could reliably predict 1-, 3-, and 5-year survival in both TCGA and GEO cohorts. We also got consistent results by performing clone formation and invasion analysis, and immunohistochemical (IHC) assays. Moreover, we also discovered that the TRGs signature was significantly associated with the different immune landscapes and therapeutic drug sensitivity. CONCLUSION Our comprehensive analysis revealed the potential molecular signature and clinical utilities of TRGs in HCC. The model based on five TRGs can accurately predict the survival outcomes of HCC, improving our knowledge of TRGs in HCC and paving a new path for guiding risk stratification and treatment strategy development for HCC patients.
Collapse
Affiliation(s)
- Yangying Zhou
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xuanxuan Li
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Guo Long
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yongguang Tao
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- Key Laboratory of Carcinogenesis of the Ministry of Health, Cancer Research Institute, School of Basic Medicine, Central South University, Changsha, China
- Department of Thoracic Surgery, Hunan Key Laboratory of Tumor Models and Individualized Medicine, Second Xiangya Hospital, Central South University, Changsha, China
| | - Ledu Zhou
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Jianing Tang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
49
|
Xie J, Zheng S, Zou Y, Tang Y, Tian W, Wong CW, Wu S, Ou X, Zhao W, Cai M, Xie X. Turning up a new pattern: Identification of cancer-associated fibroblast-related clusters in TNBC. Front Immunol 2022; 13:1022147. [PMID: 36275659 PMCID: PMC9583405 DOI: 10.3389/fimmu.2022.1022147] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2022] [Accepted: 09/16/2022] [Indexed: 11/17/2022] Open
Abstract
Growing evidence indicates a connection between cancer-associated fibroblasts (CAFs) and tumor microenvironment (TME) remodeling and tumor progression. Nevertheless, how patterns of CAFs impact TME and immunotherapy responsiveness in triple-negative breast cancer (TNBC) remains unclear. Here, we systematically investigate the relationship between TNBC progression and patterns of CAFs. By using unsupervised clustering methods in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) dataset, we identified two distinct CAF-associated clusters that were related to clinical features, characteristics of TME, and prognosis of patients. Then, we established a CAF-related prognosis index (CPI) by the least absolute shrinkage and selection operator (LASSO)-Cox regression method. CPI showed prognostic accuracy in both training and validation cohorts (METABRIC, GSE96058, and GSE21653). Consequently, we constructed a nomogram with great predictive performance. Moreover, the CPI was verified to be correlated with the responsiveness of immunotherapy in three independent cohorts (GSE91061, GSE165252, and GSE173839). Taken together, the CPI might help us improve our recognition of the TME of TNBC, predict the prognosis of TNBC patients, and offer more immunotherapy strategies in the future.
Collapse
Affiliation(s)
- Jindong Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Shaoquan Zheng
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yutian Zou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Yuhui Tang
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wenwen Tian
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Chau-Wei Wong
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Song Wu
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Xueqi Ou
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| | - Wanzhen Zhao
- Department of Radiotherapy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Manbo Cai
- Department of Radiotherapy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Xiaoming Xie
- Department of Breast Oncology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China
| |
Collapse
|
50
|
Nie S, Huili Y, Yao A, Liu J, Wang Y, Wang L, Zhang L, Kang S, Cao F. Identification of subtypes of clear cell renal cell carcinoma and construction of a prognostic model based on fatty acid metabolism genes. Front Genet 2022; 13:1013178. [PMID: 36186450 PMCID: PMC9523225 DOI: 10.3389/fgene.2022.1013178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 08/22/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The effects of fatty acid metabolism in many tumors have been widely reported. Due to the diversity of lipid synthesis, uptake, and transformation in clear cell renal cell carcinoma (ccRCC) cells, many studies have shown that ccRCC is associated with fatty acid metabolism. The study aimed was to explore the impact of fatty acid metabolism genes on the prognosis and immunotherapy of ccRCC.Methods: Two subtypes were distinguished by unsupervised clustering analysis based on the expression of 309 fatty acid metabolism genes. A prognostic model was constructed by lasso algorithm and multivariate COX regression analysis using fatty acid metabolism genes as the signatures. The tumor microenvironment between subtypes and between risk groups was further analyzed. The International Cancer Genome Consortium cohort was used for external validation of the model.Results: The analysis showed that subtype B had a poorer prognosis and a higher degree of immune infiltration. The high-risk group had a poorer prognosis and higher tumor microenvironment scores. The nomogram could accurately predict patient survival.Conclusion: Fatty acid metabolism may affect the prognosis and immune infiltration of patients with ccRCC. The analysis was performed to understand the potential role of fatty acid metabolism genes in the immune infiltration and prognosis of patients. These findings have implications for individualized treatment, prognosis, and immunization for patients with ccRCC.
Collapse
Affiliation(s)
- Shiwen Nie
- North China University of Science and Technology, Tangshan, China
| | - Youlong Huili
- North China University of Science and Technology, Tangshan, China
| | - Anliang Yao
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Jian Liu
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Yong Wang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Lei Wang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Liguo Zhang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Shaosan Kang
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
| | - Fenghong Cao
- Department of Urology, North China University of Science and Technology Affiliated Hospital, Tangshan, China
- *Correspondence: Fenghong Cao,
| |
Collapse
|