1
|
Hu Z, Li W, Wei L, Ma J. Lactoferrin in cancer: Focus on mechanisms and translational medicine. Biochim Biophys Acta Rev Cancer 2025; 1880:189330. [PMID: 40274081 DOI: 10.1016/j.bbcan.2025.189330] [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: 12/05/2024] [Revised: 04/16/2025] [Accepted: 04/21/2025] [Indexed: 04/26/2025]
Abstract
Lactoferrin is an iron-binding glycoprotein that provides natural protective effects to the human body. Its biological properties, including antibacterial, antiviral, anti-inflammatory, immune-regulatory, and iron metabolism-regulating functions, have been extensively studied. With further research, lactoferrin's impact on tumorigenesis and tumor microenvironment has become increasingly evident, as it inhibits tumor proliferation, invasion, and metastasis through multiple pathways. This article summarizes the molecular mechanisms underlying lactoferrin's anticancer effects, explores its association with the malignant progression of various cancers, and highlights its clinical translational potential as a potential cancer biomarker and drug delivery carrier to enhance anticancer therapy efficiency. Due to the high safety profile of lactoferrin, its widespread application in the field of cancer treatment is highly anticipated.
Collapse
Affiliation(s)
- Zhengyu Hu
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Wenchao Li
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China
| | - Lingyu Wei
- Laboratory of Clinical Research Center, Department of Pathology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, Shanxi, China.
| | - Jian Ma
- Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China; Cancer Research Institute, School of Basic Medicine Sciences, Xiangya School of Medicine, Central South University, Changsha, China; Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, NHC Key Laboratory of Carcinogenesis, Hunan Key Laboratory of Nonresolving Inflammation and Cancer, Hunan Key Laboratory of Cancer Metabolism, Changsha, China.
| |
Collapse
|
2
|
Issa H, Singh L, Lai KS, Parusheva-Borsitzky T, Ansari S. Dynamics of inflammatory signals within the tumor microenvironment. World J Exp Med 2025; 15:102285. [DOI: 10.5493/wjem.v15.i2.102285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/31/2024] [Accepted: 01/11/2025] [Indexed: 04/16/2025] Open
Abstract
Tumor stroma, or tumor microenvironment (TME), has been in the spotlight during recent years for its role in tumor development, growth, and metastasis. It consists of a myriad of elements, including tumor-associated macrophages, cancer-associated fibroblasts, a deregulated extracellular matrix, endothelial cells, and vascular vessels. The release of proinflammatory molecules, due to the inflamed microenvironment, such as cytokines and chemokines is found to play a pivotal role in progression of cancer and response to therapy. This review discusses the major key players and important chemical inflammatory signals released in the TME. Furthermore, the latest breakthroughs in cytokine-mediated crosstalk between immune cells and cancer cells have been highlighted. In addition, recent updates on alterations in cytokine signaling between chronic inflammation and malignant TME have also been reviewed.
Collapse
Affiliation(s)
- Hala Issa
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Lokjan Singh
- Department of Microbiology, Karnali Academy of Health Sciences, Jumla 21200, Karnali, Nepal
| | - Kok-Song Lai
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Tina Parusheva-Borsitzky
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| | - Shamshul Ansari
- Division of Health Sciences, Higher Colleges of Technology, Abu Dhabi 25026, United Arab Emirates
| |
Collapse
|
3
|
Shimagaki T, Sugimachi K, Tomino T, Onishi E, Koga N, Kasagi Y, Sugiyama M, Kimura Y, Morita M. Cachexia index as a prognostic marker in patients undergoing biliary tract cancer resection. Surg Today 2025:10.1007/s00595-025-03073-3. [PMID: 40493171 DOI: 10.1007/s00595-025-03073-3] [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: 03/28/2025] [Accepted: 05/21/2025] [Indexed: 06/12/2025]
Abstract
PURPOSE This study investigated the relationship between the preoperative cachexia index (CXI) and long-term outcomes in patients who underwent radical resection for biliary tract cancer (BTC). METHODS We analyzed 128 patients who underwent BTC resection at a single institute between 2014 and 2022. The diagnoses of the patients included intrahepatic cholangiocarcinoma (n = 27), perihilar cholangiocarcinoma (n = 17), distal cholangiocarcinoma (n = 36), gallbladder carcinoma (n = 20), and ampullary cholangiocarcinoma (n = 28). The relationship between CXI and long-term outcomes after BTC resection was investigated. CXI was calculated on the basis of the preoperative skeletal muscle index, serum albumin level, and neutrophil-to-lymphocyte ratio. RESULTS The low-CXI group (36/69, 52.1%) had a significantly higher rate of lymph node metastasis than the high-CXI group (14/59, 23.7%). The multivariate analysis confirmed that lymph node metastasis, poor differentiation (poor), and inclusion in the low-CXI group were independently associated with disease-free survival (DFS) and overall survival (OS) in patients who underwent radical resection for BTC. After propensity score matching, the low-CXI group had a significantly worse prognosis than the high-CXI group in terms of both DFS and OS. CONCLUSION CXI may be a useful prognostic factor for DFS and OS after resection of BTC.
Collapse
Affiliation(s)
- Tomonari Shimagaki
- Department of Hepatobiliary and Pancreatic Surgery, NHO Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan.
| | - Keishi Sugimachi
- Department of Hepatobiliary and Pancreatic Surgery, NHO Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| | - Takahiro Tomino
- Department of Hepatobiliary and Pancreatic Surgery, NHO Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| | - Emi Onishi
- Department of Hepatobiliary and Pancreatic Surgery, NHO Kyushu Cancer Center, 3-1-1 Notame, Minami-ku, Fukuoka, 811-1395, Japan
| | - Naomichi Koga
- Department of Gastroenterological Surgery, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Yuta Kasagi
- Department of Gastroenterological Surgery, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Masahiko Sugiyama
- Department of Gastroenterological Surgery, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Yasue Kimura
- Department of Gastroenterological Surgery, NHO Kyushu Cancer Center, Fukuoka, Japan
| | - Masaru Morita
- Department of Gastroenterological Surgery, NHO Kyushu Cancer Center, Fukuoka, Japan
| |
Collapse
|
4
|
Toghraie FS, Bayat M, Hosseini MS, Ramezani A. Tumor-infiltrating myeloid cells; mechanisms, functional significance, and targeting in cancer therapy. Cell Oncol (Dordr) 2025; 48:559-590. [PMID: 39998754 PMCID: PMC12119771 DOI: 10.1007/s13402-025-01051-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/20/2025] [Indexed: 02/27/2025] Open
Abstract
Tumor-infiltrating myeloid cells (TIMs), which encompass tumor-associated macrophages (TAMs), tumor-associated neutrophils (TANs), myeloid-derived suppressor cells (MDSCs), and tumor-associated dendritic cells (TADCs), are of great importance in tumor microenvironment (TME) and are integral to both pro- and anti-tumor immunity. Nevertheless, the phenotypic heterogeneity and functional plasticity of TIMs have posed challenges in fully understanding their complexity roles within the TME. Emerging evidence suggested that the presence of TIMs is frequently linked to prevention of cancer treatment and improvement of patient outcomes and survival. Given their pivotal function in the TME, TIMs have recently been recognized as critical targets for therapeutic approaches aimed at augmenting immunostimulatory myeloid cell populations while depleting or modifying those that are immunosuppressive. This review will explore the important properties of TIMs related to immunity, angiogenesis, and metastasis. We will also document the latest therapeutic strategies targeting TIMs in preclinical and clinical settings. Our objective is to illustrate the potential of TIMs as immunological targets that may improve the outcomes of existing cancer treatments.
Collapse
Affiliation(s)
- Fatemeh Sadat Toghraie
- Institute of Biotechnology, Faculty of the Environment and Natural Sciences, Brandenburg University of Technology, Cottbus, Germany
| | - Maryam Bayat
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahsa Sadat Hosseini
- Regenerative Medicine Research Center, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Amin Ramezani
- Shiraz Institute for Cancer Research, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran.
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
5
|
Fiste O, Mavrothalassitis E, Kokkalis A, Anagnostakis M, Gomatou G, Kontogiannis A, Karaviti D, Karaviti E, Syrigos NK, Kotsakis A, Kotteas EA. Inflammation-related biomarkers as predictors of pathological complete response in early-stage breast cancer. Clin Transl Oncol 2025; 27:2453-2460. [PMID: 39668275 DOI: 10.1007/s12094-024-03814-9] [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: 11/12/2024] [Accepted: 11/23/2024] [Indexed: 12/14/2024]
Abstract
BACKGROUND Neoadjuvant systemic therapy (NAT) represents an attractive option for improved outcomes of early-stage breast cancer (BC) patients, as it can significantly reduce tumor burden thus permitting breast-conserving resections. Equally important, the eradication of viable cancer cells post-NAT, also known as pathological complete response (pCR), has emerged as a strong prognostic biomarker, reflecting tumor's biology and subsequent treatment responses. Yet to date, no validated markers predictive of pCR have been identified. METHODS The present retrospective study aimed to explore the value of neutrophil-tolymphocyte ratio (NLR) and platelet-to-lymphocyte ratio (PLR) as potential predictors of pCR. RESULTS Despite no statistically significant associations have been reported, NLR and PLR dynamics during NAT, as longitudinal inflammatory phenotypes, merit further investigation in larger cohorts. CONCLUSION In the future, the integration of a comprehensive inflammatory biomarker panel into clinical practice could assist in a priori treatment selection process.
Collapse
Affiliation(s)
- Oraianthi Fiste
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece.
| | - Evangelos Mavrothalassitis
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece
| | - Alexandros Kokkalis
- Department of Medical Oncology, University Hospital of Larissa, 41334, Larissa, Greece
| | - Maximilian Anagnostakis
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece
| | - Georgia Gomatou
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece
| | - Athanasios Kontogiannis
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece
| | - Dimitra Karaviti
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece
| | - Eleftheria Karaviti
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece
| | - Nikolaos Konstantinos Syrigos
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece
| | - Athanasios Kotsakis
- Department of Medical Oncology, University Hospital of Larissa, 41334, Larissa, Greece
| | - Elias Alexandros Kotteas
- Oncology Unit, Third Department of Internal Medicine and Laboratory, National and Kapodistrian University of Athens, Sotiria General Hospital, 11527, Athens, Greece
| |
Collapse
|
6
|
Pentimalli TM, Schallenberg S, León-Periñán D, Legnini I, Theurillat I, Thomas G, Boltengagen A, Fritzsche S, Nimo J, Ruff L, Dernbach G, Jurmeister P, Murphy S, Gregory MT, Liang Y, Cordenonsi M, Piccolo S, Coscia F, Woehler A, Karaiskos N, Klauschen F, Rajewsky N. Combining spatial transcriptomics and ECM imaging in 3D for mapping cellular interactions in the tumor microenvironment. Cell Syst 2025; 16:101261. [PMID: 40220761 DOI: 10.1016/j.cels.2025.101261] [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: 07/12/2024] [Revised: 12/13/2024] [Accepted: 03/19/2025] [Indexed: 04/14/2025]
Abstract
Tumors are complex ecosystems composed of malignant and non-malignant cells embedded in a dynamic extracellular matrix (ECM). In the tumor microenvironment, molecular phenotypes are controlled by cell-cell and ECM interactions in 3D cellular neighborhoods (CNs). While their inhibition can impede tumor progression, routine molecular tumor profiling fails to capture cellular interactions. Single-cell spatial transcriptomics (ST) maps receptor-ligand interactions but usually remains limited to 2D tissue sections and lacks ECM readouts. Here, we integrate 3D ST with ECM imaging in serial sections from one clinical lung carcinoma to systematically quantify molecular states, cell-cell interactions, and ECM remodeling in CN. Our integrative analysis pinpointed known immune escape and tumor invasion mechanisms, revealing several druggable drivers of tumor progression in the patient under study. This proof-of-principle study highlights the potential of in-depth CN profiling in routine clinical samples to inform microenvironment-directed therapies. A record of this paper's transparent peer review process is included in the supplemental information.
Collapse
Affiliation(s)
- Tancredi Massimo Pentimalli
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin
| | - Simon Schallenberg
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany
| | - Daniel León-Periñán
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Ivano Legnini
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Human Technopole, Milan, Italy
| | - Ilan Theurillat
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Gwendolin Thomas
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Anastasiya Boltengagen
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Sonja Fritzsche
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | - Jose Nimo
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany; Humboldt-Universität zu Berlin, Institute of Biology, 10099 Berlin, Germany
| | | | - Gabriel Dernbach
- Institute of Pathology, Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität Berlin, Berlin, Germany; Aignostics GmbH, Berlin, Germany; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany
| | | | | | | | - Yan Liang
- NanoString® Technologies, Inc, Seattle, WA, USA
| | | | - Stefano Piccolo
- Department of Molecular Medicine, University of Padua, Padua, Italy; IFOM ETS, the AIRC Institute of Molecular Oncology, Milan, Italy
| | - Fabian Coscia
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Spatial Proteomics Group, Berlin, Germany
| | - Andrew Woehler
- Systems Biology Imaging Platform, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Howard Hughes Medical Institute, Janelia Research Campus, Ashburn, VA, USA
| | - Nikos Karaiskos
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
| | - Frederick Klauschen
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; BIFOLD - Berlin Institute for the Foundations of Learning and Data, Berlin, Germany; Institute of Pathology, Ludwig Maximilians Universität, Munich, Germany
| | - Nikolaus Rajewsky
- Laboratory for Systems Biology of Regulatory Elements, Berlin Institute for Medical Systems Biology (BIMSB), Max-Delbrück-Centrum for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany; Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin; German Center for Cardiovascular Research (DZHK), Site Berlin, Berlin, Germany; NeuroCure Cluster of Excellence, Berlin, Germany; German Cancer Consortium (DKTK), Berlin, Germany; National Center for Tumor Diseases (NCT), Site Berlin, Berlin, Germany.
| |
Collapse
|
7
|
Meng C, Lin K, Shi W, Teng H, Wan X, DeBruine A, Wang Y, Liang X, Leo J, Chen F, Gu Q, Zhang J, Van V, Maldonado KL, Gan B, Ma L, Lu Y, Zhao D. Histone methyltransferase ASH1L primes metastases and metabolic reprogramming of macrophages in the bone niche. Nat Commun 2025; 16:4681. [PMID: 40394007 PMCID: PMC12092585 DOI: 10.1038/s41467-025-59381-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 04/22/2025] [Indexed: 05/22/2025] Open
Abstract
Bone metastasis is a major cause of cancer death; however, the epigenetic determinants driving this process remain elusive. Here, we report that histone methyltransferase ASH1L is genetically amplified and is required for bone metastasis in men with prostate cancer. ASH1L rewires histone methylations and cooperates with HIF-1α to induce pro-metastatic transcriptome in invading cancer cells, resulting in monocyte differentiation into lipid-associated macrophage (LA-TAM) and enhancing their pro-tumoral phenotype in the metastatic bone niche. We identified IGF-2 as a direct target of ASH1L/HIF-1α and mediates LA-TAMs' differentiation and phenotypic changes by reprogramming oxidative phosphorylation. Pharmacologic inhibition of the ASH1L-HIF-1α-macrophages axis elicits robust anti-metastasis responses in preclinical models. Our study demonstrates epigenetic alterations in cancer cells reprogram metabolism and features of myeloid components, facilitating metastatic outgrowth. It establishes ASH1L as an epigenetic driver priming metastasis and macrophage plasticity in the bone niche, providing a bona fide therapeutic target in metastatic malignancies.
Collapse
Affiliation(s)
- Chenling Meng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kevin Lin
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Wei Shi
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Hongqi Teng
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xinhai Wan
- Department of Endocrine Neoplasia & Hormonal Disorders, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Anna DeBruine
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Yin Wang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Xin Liang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Javier Leo
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
- The University of Texas MD Anderson Cancer Center UTHealth Graduate School of Biomedical Sciences, Houston, TX, 77030, USA
| | - Feiyu Chen
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Qianlin Gu
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Jie Zhang
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Vivien Van
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Kiersten L Maldonado
- Department of Imaging Physics, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Boyi Gan
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Li Ma
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| | - Yue Lu
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| | - Di Zhao
- Department of Experimental Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA.
| |
Collapse
|
8
|
Pagano CA, Masini MA, Sabbatini M, Gribaudo G, Manfredi M, Caprì FG, Bonetto V, Magnelli V, Donadelli M, Corino R, Belay MH, Robotti E, Marengo E. Simulated Microgravity-Induced Alterations in PDAC Cells: A Potential Role for Trichostatin A in Restoring Cellular Phenotype. Int J Mol Sci 2025; 26:4758. [PMID: 40429900 PMCID: PMC12112487 DOI: 10.3390/ijms26104758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) accounts for 90% of all pancreatic malignancies. Despite the remarkable improvement concerning treatment, late detection and resistance to clinically used chemotherapeutic agents remain major challenges. Trichostatin A (TSA), a histone deacetylase inhibitor, has been recognized as an effective therapeutic agent against PDAC by inhibiting proliferation, inducing apoptosis, and sensitizing PDAC cells to chemotherapeutic agents such as gemcitabine. Microgravity has become a useful tool in cancer research due to its effects on various cellular processes. This paper presents a deep molecular and proteomic analysis investigating cell growth, the modulation of cytokeratins, and proteins related to apoptosis, cellular metabolism, and protein synthesis after TSA treatment in simulated microgravity (SMG)-exposed PaCa44 3D cells. Our analysis concerns the effects of TSA treatment on cell proliferation: the impairment of the cell cycle with the downregulation of proteins involved in Cdc42 signaling and G1/G2- and G2/M-phase transitions. Thus, we observed modification of survival pathways and proteins related to autophagy and apoptosis. We also observed changes in proteins involved in the regulation of transcription and the repair of damaged DNA. TSA treatment promotes the downregulation of some markers involved in the maintenance of the potency of stem cells, while it upregulates proteins involved in the induction and modulation of the differentiation process. Our data suggest that TSA treatment restores the cell phenotype prior to simulated microgravity exposure, and exerts an intriguing activity on PDAC cells by reducing proliferation and inducing cell death via multiple pathways.
Collapse
Affiliation(s)
- Corinna Anais Pagano
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Maria Angela Masini
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Maurizio Sabbatini
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Giorgia Gribaudo
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Marcello Manfredi
- Department of Translational Medicine (DIMET), University of Eastern Piedmont, 28100 Novara, Italy;
- Institute for Molecular and Translational Cardiology (IMTC), IRCCS Policlinico San Donato, 20097 Milan, Italy
| | - Flavia Giusy Caprì
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Valentina Bonetto
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Valeria Magnelli
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Massimo Donadelli
- Department of Neurosciences, Biomedicine and Movement Sciences (DNBM), University of Verona, 37134 Verona, Italy;
| | - Roberto Corino
- Centro Italiano di Diagnostica Medica Ultrasonica (CIDIMU), 10128 Turin, Italy;
| | - Masho Hilawie Belay
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Elisa Robotti
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| | - Emilio Marengo
- Department of Science and Innovation Technology (DISIT), Università del Piemonte Orientale, 15121 Alessandria, Italy; (C.A.P.); (M.A.M.); (G.G.); (F.G.C.); (V.B.); (V.M.); (M.H.B.); (E.R.); (E.M.)
| |
Collapse
|
9
|
Israeli Dangoor S, Khoury R, Salomon K, Pozzi S, Shahar S, Miari A, Leichtmann-Bardoogo Y, Bar-Hai N, Frommer N, Yeini E, Winkler T, Balint Lahat N, Kamer I, Hadad O, Laue K, Brem H, Hyde TM, Bar J, Barshack I, Ben-David U, Ishay-Ronen D, Maoz BM, Satchi-Fainaro R. CCL2 blockade combined with PD-1/P-selectin immunomodulators impedes breast cancer brain metastasis. Brain 2025; 148:1740-1756. [PMID: 39450648 PMCID: PMC12073999 DOI: 10.1093/brain/awae347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 09/11/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
Over the last two decades, the diagnosis and treatment of breast cancer patients have improved considerably. However, brain metastases remain a major clinical challenge and a leading cause of mortality. Thus, a better understanding of the pathways involved in the metastatic cascade is essential. To this end, we have investigated the reciprocal effects of astrocytes and breast cancer cells, employing traditional 2D cell culture and our unique 3D multicellular tumouroid models. Our findings revealed that astrocytes enhance the proliferation, migration and invasion of breast cancer cells, suggesting a supportive role for astrocytes in breast cancer outgrowth to the brain. Elucidating the key players in astrocyte-breast cancer cells crosstalk, we found that CCL2 is highly expressed in breast cancer brain metastases tissue sections from both patients and mice. Our in vitro and in vivo models further confirmed that CCL2 has a functional role in brain metastasis. Given their aggressive nature, we sought additional immune checkpoints for rationale combination therapy. Among the promising candidates were the adhesion molecule P-selectin, which we have recently shown to play a key role in the crosstalk with microglia cells and the co-inhibitory receptor PD-1, the main target of currently approved immunotherapies. Finally, combining CCL2 inhibition with immunomodulators targeting either PD-1/PD-L1 or P-selectin/P-Selectin Ligand-1 axes in our human 3D tumouroid models and in vivo presented more favourable outcomes than each monotherapy. Taken together, we propose that CCL2-CCR2/CCR4 is a key pathway promoting breast cancer brain metastases and a promising target for an immunotherapeutic combination approach.
Collapse
Affiliation(s)
- Sahar Israeli Dangoor
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Rami Khoury
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Koren Salomon
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Sabina Pozzi
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Shir Shahar
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Adan Miari
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | | | - Neta Bar-Hai
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
- Affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Neta Frommer
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Eilam Yeini
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tom Winkler
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Nora Balint Lahat
- Department of Pathology, Sheba Medical Center, Tel Hashomer 5262000, Israel
| | - Iris Kamer
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
| | - Ori Hadad
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Kathrin Laue
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Henry Brem
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Thomas M Hyde
- Lieber Institute for Brain Development, Johns Hopkins Medical Campus, Baltimore, MD 21218, USA
- Department of Psychiatry & Behavioral Science, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21218, USA
| | - Jair Bar
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Tel Hashomer 5262000, Israel
- Department of Pathology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Uri Ben-David
- Department of Human Molecular Genetics and Biochemistry, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Dana Ishay-Ronen
- Cancer Research Center, Oncology Institute, Sheba Medical Center, Tel-Hashomer 5262000, Israel
- Affiliated with Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ben M Maoz
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol Center for Regenerative Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Ronit Satchi-Fainaro
- Department of Physiology and Pharmacology, Faculty of Medicine, Tel Aviv University, Tel Aviv 6997801, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
- The Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
10
|
Bayrak O, Alper M, Basbinar Y, Bayrak S. The role of thrombin in the paradoxical interplay of cancer metastasis and the vascular system: A driving dynamic. Biomed Pharmacother 2025; 186:118031. [PMID: 40215647 DOI: 10.1016/j.biopha.2025.118031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/26/2025] [Accepted: 03/28/2025] [Indexed: 04/25/2025] Open
Abstract
The coagulation system plays a complex role in cancer therapy. Endothelial damage and tissue factor increased by chemotherapy initiate the coagulation cascade, producing active FXa and releasing thrombin. Thrombin triggers tumor growth and metastasis, leading to severe thromboembolic events in cancer patients. Direct thrombin inhibitors do not have the expected anti-metastatic effect as PAR-2 remains active and increases the risk of bleeding. Therefore, dual inhibition of thrombin by FXa inhibition and plasmin inhibition, which converts fibrin to fibrinogen, is targeted. Clinical studies show that the use of tranexamic acid in patients on NOAC therapy may be beneficial without increasing the risk of bleeding. This approach offers a promising strategy to provide an anti-metastatic effect in cancer treatment.
Collapse
Affiliation(s)
- Ozge Bayrak
- Dokuz Eylul University, Institute of Health Sciences, Department of Oncology, Izmir, Turkey
| | - Meltem Alper
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey
| | - Yasemin Basbinar
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey
| | - Serdar Bayrak
- Dokuz Eylul University, Institute of Oncology, Department of Translational Oncology, Izmir, Turkey; Dokuz Eylul University, Faculty of Medicine, Department of Cardiovascular Surgery, Izmir, Turkey.
| |
Collapse
|
11
|
Hanitrarimalala V, Prgomet Z, Hedhammar M, Tassidis H, Wingren AG. In vitro 3D modeling of colorectal cancer: the pivotal role of the extracellular matrix, stroma and immune modulation. Front Genet 2025; 16:1545017. [PMID: 40376304 PMCID: PMC12078225 DOI: 10.3389/fgene.2025.1545017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/23/2025] [Indexed: 05/18/2025] Open
Abstract
Colorectal cancer (CRC) is a leading global cancer with high mortality, especially in metastatic cases, with limited therapeutic options. The tumor microenvironment (TME), a network comprising various immune cells, stromal cells and extracellular (ECM) components plays a crucial role in influencing tumor progression and therapy outcome. The genetic heterogeneity of CRC and the complex TME complicates the development of effective, personalized treatment strategies. The prognosis has slowly improved during the past decades, but metastatic CRC (mCRC) is common among patients and is still associated with low survival. The therapeutic options for CRC differ from those for mCRC and include surgery (mostly for CRC), chemotherapy, growth factor receptor signaling pathway targeting, as well as immunotherapy. Malignant CRC cells are established in the TME, which varies depending on the primary or metastatic site. Herein, we review the role and interactions of several ECM components in 3D models of CRC and mCRC tumor cells, with an emphasis on how the TME affects tumor growth and treatment. This comprehensive summary provides support for the development of 3D models that mimic the interactions within the TME, which will be essential for the development of novel anticancer therapies.
Collapse
Affiliation(s)
- Veroniaina Hanitrarimalala
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Zdenka Prgomet
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - My Hedhammar
- KTH Royal Institute of Technology, Division of Protein Technology, Stockholm, Sweden
| | - Helena Tassidis
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| | - Anette Gjörloff Wingren
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| |
Collapse
|
12
|
Zhou Y, Cao P, Zhu Q. The regulatory role of m6A in cancer metastasis. Front Cell Dev Biol 2025; 13:1539678. [PMID: 40356596 PMCID: PMC12066624 DOI: 10.3389/fcell.2025.1539678] [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: 12/04/2024] [Accepted: 04/16/2025] [Indexed: 05/15/2025] Open
Abstract
Metastasis remains a primary cause of cancer-related mortality, with its intricate mechanisms continuing to be uncovered through advancing research. Among the various regulatory processes involved, RNA modification has emerged as a critical epitranscriptomic mechanism influencing cancer metastasis. N6-methyladenosine (m6A), recognized as one of the most prevalent and functionally significant RNA modifications, plays a central role in the regulation of RNA metabolism. In this review, we explore the multifaceted role of m6A in the different stages of cancer metastasis, including epithelial-mesenchymal transition, invasion, migration, and colonization. In addition to summarizing the current state of our understanding, we offer insights into how m6A modifications modulate key oncogenic pathways, highlighting the implications of recent discoveries for therapeutic interventions. Furthermore, we critically assess the limitations of previous studies and propose areas for future research, including the potential for targeting m6A as a novel approach in anti-metastatic therapies. Our analysis provides a comprehensive understanding of the regulatory landscape of m6A in metastasis, offering directions for continued exploration in this rapidly evolving field.
Collapse
Affiliation(s)
- Ying Zhou
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Peng Cao
- Department of Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Qing Zhu
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
13
|
Tan W, Zhu Y, Chen S. Innovative approach to the detection of circulating tumor biomarkers: multi-dimensional application of liposome technology. Lipids Health Dis 2025; 24:160. [PMID: 40295973 PMCID: PMC12036244 DOI: 10.1186/s12944-025-02578-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 04/19/2025] [Indexed: 04/30/2025] Open
Abstract
Malignant tumors represent a significant worldwide health challenge, with elevated morbidity and mortality rates necessitating enhanced early identification and individualized treatment. Liposomes, as biomimetic lipid-based nanovesicles, have developed as a multifaceted platform for detecting and treating malignant tumors due to their excellent biocompatibility, stability, and membrane fusion properties. Circulating tumor markers, such as circulating tumor cells (CTCs), extracellular vesicles (EVs), circulating tumor proteins (CTPs), and circulating tumor nucleic acids (ctNAs), play a key role in early cancer diagnosis, disease progression monitoring, and personalized therapy. Liposome-based platforms enable effective molecular recognition, targeted detection, and signal amplification by targeting circulating tumor biomarkers, significantly increasing the potential for early tumor diagnosis and treatment. This review systematically summarizes advancements in the study of liposomes concerning circulating tumor markers, including applications in targeted recognition, early detection, and disease diagnosis, while discussing present problems and prospective applications of existing technology.
Collapse
Affiliation(s)
- Weichu Tan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Yitong Zhu
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China
| | - Siting Chen
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, School of Basic Medical Sciences, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Provincial Key Laboratory of Single-Cell and Extracellular Vesicles, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, People's Republic of China.
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong, 511518, People's Republic of China.
| |
Collapse
|
14
|
Zhou X, Li R, Lai M, Lai C. Exploring molecular and cellular mechanisms of Pre-Metastatic niche in renal cell carcinoma. Mol Cancer 2025; 24:121. [PMID: 40264130 PMCID: PMC12012986 DOI: 10.1186/s12943-025-02315-9] [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: 12/21/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025] Open
Abstract
Renal cell carcinoma (RCC) is among the most frequently occurring types of cancer, and its metastasis is a major contributor to its elevated mortality. Before the primary tumor metastasizes to secondary or distant organs, it remodels the microenvironment of these sites, creating a pre-metastatic niche (PMN) conducive to the colonization and growth of metastatic tumors. RCC releases a variety of biomolecules that induce angiogenesis, alter vascular permeability, modulate immune cells to create an immunosuppressive microenvironment, affect extracellular matrix remodeling and metabolic reprogramming, and determine the organotropism of metastasis through different signaling pathways. This review summarizes the principal processes and mechanisms underlying the formation of the premetastatic niche in RCC. Additionally, we emphasize the significance and potential of targeting PMNs for the prevention and treatment of tumor metastasis in future therapeutic approaches. Finally, we summarized the currently potential targeted strategies for detecting and treating PMN in RCC and provide a roadmap for further in-depth studies on PMN in RCC.
Collapse
Affiliation(s)
- Xiao Zhou
- Department of Pathology, and Department of Pathology Sir Run Run Shaw Hospital, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Science (2019RU042), Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Ruirui Li
- Institute of Immunology, Department of Respiratory Disease of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Maode Lai
- Department of Pathology, and Department of Pathology Sir Run Run Shaw Hospital, Research Unit of Intelligence Classification of Tumor Pathology and Precision Therapy, Chinese Academy of Medical Science (2019RU042), Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China.
| | - Chong Lai
- Department of Urology, First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310006, China.
| |
Collapse
|
15
|
Figols M, Chekhun S, Fernández-Saorin M, Pérez-Criado I, Bautista A, Font A, Ruiz de Porras V. Tumor-Educated Platelets in Urological Tumors: A Novel Biosource in Liquid Biopsy. Int J Mol Sci 2025; 26:3595. [PMID: 40332071 PMCID: PMC12026913 DOI: 10.3390/ijms26083595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 04/02/2025] [Accepted: 04/09/2025] [Indexed: 05/08/2025] Open
Abstract
Platelets, traditionally recognized for their role in hemostasis, have emerged as pivotal players in cancer biology. They actively contribute to tumor proliferation, angiogenesis, immune evasion, and metastasis and thus play a significant role in cancer progression. Tumor-educated platelets (TEPs) acquire protumorigenic phenotypes through RNA, protein, and receptor profile alterations driven by interactions with tumors and their microenvironment. These modifications enable TEPs to enhance tumor growth and dissemination and to play a critical role throughout the metastatic process. Moreover, TEPs are promising biomarkers that can easily be analyzed in liquid biopsies. Since they dynamically mirror tumor activity through transcriptomic and proteomic changes, their analysis offers a non-invasive method for determining cancer detection and diagnosis, patient prognosis, therapy monitoring, and personalization of treatment. Their demonstrated accuracy in identifying cancer types and predicting treatment responses underscores their ability to provide real-time insights into tumor biology, including in urological malignancies. Their diagnostic potential and their accessibility as blood-sourced biomarkers position TEPs as transformative tools in advancing personalized oncology. Here, we focus on the role of TEPs in urological tumors, exploring their applications in early cancer detection, disease monitoring, and the design of tailored therapeutic strategies.
Collapse
Affiliation(s)
- Mariona Figols
- Medical Oncology Department, Althaia Xarxa Assistencial Universitària de Manresa, C/ Dr. Joan Soler, 1-3, 08243 Manresa, Spain; (M.F.); (I.P.-C.); (A.B.)
- PhD Programme in Medicine and Biomedical Sciences, Doctoral School, University of Vic, Central University of Catalonia (UVic-UCC), C/ Dr. Junyent, 1, 08500 Vic, Spain
- Faculty of Medicine, University of Vic, Central University of Catalonia (UVicUCC), Can Baumann, Ctra, de Roda, 70, 08500 Vic, Spain
| | - Sviatoslav Chekhun
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916 Badalona, Spain; (S.C.); (M.F.-S.); (A.F.)
- Badalona Applied Research Group in Oncology (B⋅ARGO), Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
- Medical Oncology Department, Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
| | - Maria Fernández-Saorin
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916 Badalona, Spain; (S.C.); (M.F.-S.); (A.F.)
- Badalona Applied Research Group in Oncology (B⋅ARGO), Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
| | - Ignacio Pérez-Criado
- Medical Oncology Department, Althaia Xarxa Assistencial Universitària de Manresa, C/ Dr. Joan Soler, 1-3, 08243 Manresa, Spain; (M.F.); (I.P.-C.); (A.B.)
| | - Ana Bautista
- Medical Oncology Department, Althaia Xarxa Assistencial Universitària de Manresa, C/ Dr. Joan Soler, 1-3, 08243 Manresa, Spain; (M.F.); (I.P.-C.); (A.B.)
| | - Albert Font
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916 Badalona, Spain; (S.C.); (M.F.-S.); (A.F.)
- Badalona Applied Research Group in Oncology (B⋅ARGO), Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
- Medical Oncology Department, Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
| | - Vicenç Ruiz de Porras
- CARE Program, Germans Trias i Pujol Research Institute (IGTP), Camí de les Escoles, s/n, 08916 Badalona, Spain; (S.C.); (M.F.-S.); (A.F.)
- Badalona Applied Research Group in Oncology (B⋅ARGO), Catalan Institute of Oncology, Camí de les Escoles, s/n, 08916 Badalona, Spain
- GRET and Toxicology Unit, Department of Pharmacology, Toxicology and Therapeutic Chemistry, Faculty of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
16
|
Wang L, Chen Z, Dai Z, Li M, Chen B, Wang B. Membrane-intercalating conjugated oligoelectrolytes for lipid membrane imaging. Biomater Sci 2025; 13:1923-1938. [PMID: 40084474 DOI: 10.1039/d5bm00028a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
Membrane-intercalating conjugated oligoelectrolytes (MICOEs), a class of phospholipid bilayer mimics, demonstrate an exceptional ability to spontaneously integrate into biological membranes through a combination of electrostatic and hydrophobic interactions. This unique property makes them promising candidates for membrane imaging applications. Over the past decade, MICOEs have been successfully applied to imaging and tracking a wide range of biological membranes, including microbial membranes, mammalian plasma membranes, intracellular membranes, extracellular vesicles, and artificial liposomes. Recent advancements have shed light on the imaging mechanisms of MICOEs and highlighted their potential as fluorescent probes, with a focus on structural optimization to enhance their performance. Building on these developments, this review will explore the intercalation mechanisms of MICOEs, analyze the structure-activity relationships governing their molecular design and imaging capabilities, and discuss the future challenges and emerging opportunities for their application as advanced membrane dyes.
Collapse
Affiliation(s)
- Lingna Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Zehua Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Zhaohui Dai
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Meng Li
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Bo Chen
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| | - Bing Wang
- Ningbo Key Laboratory of Biomedical Imaging Probe Materials and Technology, Laboratory of Advanced Theranostic Materials and Technology, Ningbo Institute of Materials Technology and Engineering, Chinese Academy of Sciences, Ningbo, Zhejiang 315201, China
| |
Collapse
|
17
|
Chen C, Wang Z, Lin Q, Li M, Xu L, Fu Y, Zhao X, Ma Z, Xu J, Zhou S, Zhang M, Qian Y, Bao L, Wang B, Wang M, Ding Q, Wang Q, Wang S. NAT10 Promotes Gastric Cancer Liver Metastasis by Modulation of M2 Macrophage Polarization and Metastatic Tumor Cell Hepatic Adhesion. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410263. [PMID: 39985269 PMCID: PMC12005778 DOI: 10.1002/advs.202410263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/21/2024] [Indexed: 02/24/2025]
Abstract
The relationship between patterns of RNA modifications and gastric cancer (GC) liver metastasis (GCLM) remains unclear. Here, by single-cell sequencing, clinical sample analysis, and mouse model studies, an abnormal increase in the expression of the RNA acetyltransferase N-acetyltransferase 10 (NAT10) in liver metastatic GC cells is identified. NAT10-mediated N4-acetylcytidine modification of CXCL2 and KLF5 mRNA increases their stability. Then, secreted CXCL2 is found to promote the infiltration and polarization of M2-like macrophages to produce oncostatin M, which transcriptionally activates NAT10 expression via STAT3 signaling. In addition, organoid models confirm that NAT10 promotes the adhesion of GC cells to hepatocytes. Mechanistically, KLF5 transcriptionally activates ITGαV, facilitating GC cell attachment to hepatocytes. Intriguingly, high expression of NAT10/KLF5 axis is associated with poor prognosis of GC patients and targeting this axis significantly reduces GCLM in preclinical murine models. Collectively, these findings suggest the clinical significance of NAT10 in developing targeted therapies for GC patients with liver metastasis.
Collapse
Affiliation(s)
- Chen Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
| | - Zhangding Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
| | - Qingfeng Lin
- Department of OncologyJiangyin Clinical College of Xuzhou Medical UniversityJiangyin Hospital Affiliated to Nantong UniversityJiangyin People's HospitalJiangyin214400China
| | - Mengmeng Li
- Medical School of Nanjing UniversityNanjing210093China
| | - Lei Xu
- Department of GastroenterologyThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Yao Fu
- Department of PathologyThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Xiaoya Zhao
- Medical School of Nanjing UniversityNanjing210093China
| | - Zhuang Ma
- Medical School of Nanjing UniversityNanjing210093China
| | - Jiawen Xu
- Medical School of Nanjing UniversityNanjing210093China
| | - Shimeng Zhou
- Medical School of Nanjing UniversityNanjing210093China
| | - Mingyue Zhang
- Medical School of Nanjing UniversityNanjing210093China
| | - Yun Qian
- Medical School of Nanjing UniversityNanjing210093China
| | - Linsen Bao
- Division of Gastric SurgeryDepartment of General SurgeryThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Bo Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
| | - Meng Wang
- Division of Gastric SurgeryDepartment of General SurgeryThe Affiliated Drum Tower Hospital of Nanjing University Medical SchoolNanjing210008China
| | - Qingqing Ding
- Department of Geriatric OncologyThe First Affiliated Hospital of Nanjing Medical UniversityNanjing210029China
| | - Qiang Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
| | - Shouyu Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Anhui Medical University; MOE Innovation Center for Basic Research in Tumor ImmunotherapyAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefei230022China
- Medical School of Nanjing UniversityNanjing210093China
| |
Collapse
|
18
|
Huang S, Cui Y, Wang B, Liu FJ, Liu JY, Shao W, Wang XQ, Guo Q. Bisboronic Ester-Enabled Redox-Targeted Cancer Therapy: A Cascade ROS Consumption Approach for Efficient Elimination of Cancer Cells. J Med Chem 2025; 68:6656-6664. [PMID: 40051089 DOI: 10.1021/acs.jmedchem.5c00038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/28/2025]
Abstract
Elevated levels of reactive oxygen species (ROS) in cancer cells, arising from their altered metabolism, represent a distinctive therapeutic target. Here, we introduce the first example of bisboronic esters as potent agents for rapidly disrupting redox homeostasis, selectively inducing oxidative stress to eliminate cancer cells. Structure-activity relationship studies revealed that both the electronic and steric factors significantly influence their cytotoxicity, with the most effective compounds achieving IC50 values as low as 0.63 μM. Confocal imaging confirmed the rapid intracellular depletion of ROS, leading to a severe metabolic imbalance and efficient cancer cell death. This study highlights the potential of boronate-based therapeutics for redox-targeted cancer therapy, which may provide a promising foundation for the development of innovative anticancer strategies.
Collapse
Affiliation(s)
- Silin Huang
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P. R. China
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| | - Yue Cui
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, Hangzhou 310022, China
| | - Boqiang Wang
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, P. R. China
| | - Fang-Jie Liu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, Hangzhou 310022, China
| | - Jia-Yi Liu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, Hangzhou 310022, China
| | - Wen Shao
- Key Laboratory for Green Organic Synthesis and Application of Hunan Province, Key Laboratory of Environmentally Friendly Chemistry and Application of Ministry of Education, College of Chemistry, Xiangtan University, Xiangtan 411105, P. R. China
| | - Xue-Qiang Wang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Zhejiang, Hangzhou 310022, China
| | - Qiang Guo
- Faculty of Life Science and Technology, Kunming University of Science and Technology, Kunming, Yunnan 650500, P. R. China
- Department of Gastroenterology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan 650500, P.R. China
| |
Collapse
|
19
|
Nishida A, Andoh A. The Role of Inflammation in Cancer: Mechanisms of Tumor Initiation, Progression, and Metastasis. Cells 2025; 14:488. [PMID: 40214442 PMCID: PMC11987742 DOI: 10.3390/cells14070488] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/14/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Inflammation is an essential component of the immune response that protects the host against pathogens and facilitates tissue repair. Chronic inflammation is a critical factor in cancer development and progression. It affects every stage of tumor development, from initiation and promotion to invasion and metastasis. Tumors often create an inflammatory microenvironment that induces angiogenesis, immune suppression, and malignant growth. Immune cells within the tumor microenvironment interact actively with cancer cells, which drives progression through complex molecular mechanisms. Chronic inflammation is triggered by factors such as infections, obesity, and environmental toxins and is strongly linked to increased cancer risk. However, acute inflammatory responses can sometimes boost antitumor immunity; thus, inflammation presents both challenges and opportunities for therapeutic intervention. This review examines how inflammation contributes to tumor biology, emphasizing its dual role as a critical factor in tumorigenesis and as a potential therapeutic target.
Collapse
Affiliation(s)
- Atsushi Nishida
- Department of Medicine, Shiga University of Medical Science, Seta-Tsukinowa, Otsu 520-2192, Shiga, Japan;
| | | |
Collapse
|
20
|
Alsheleh T, Zraikat M, Daoud F, Alqudah DA, Abdelghany S, Abu Siniyeh A, Alshaer W. In vitro cellular interaction of drug-loaded liposomes with 2D and 3D cell culture of U87-MG cell line. PLoS One 2025; 20:e0320374. [PMID: 40131912 PMCID: PMC11936199 DOI: 10.1371/journal.pone.0320374] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 02/18/2025] [Indexed: 03/27/2025] Open
Abstract
The distinctive physiological and physical properties of 3D cultures that mimic tumor microenvironments in vivo make them more suitable for assessing the efficacy of drugs and nanoparticles compared to 2D culture models. Therefore, this study aims to examine and contrast how liposomes interact with cell cultures in both 2D and 3D models. Hanging drop technique was used to generate 3D spheroids. Cellular toxicity of Doxorubicin and Doxil®-liposomes was tested using an MTT assay. Cellular uptake of Doxil®-liposomes was investigated in 3D and 2D cell culture models using flow cytometry and confocal microscopy. Finally, migration and invasion assays were used to investigate the Doxil®-liposomes interaction with the two models 2D model and 3D model, respectively. Our findings show that cells were able to form spheroid structures when a specific cell ratio was maintained. Flow cytometry analysis revealed that 2D cells exhibited higher Doxil®-liposome uptake than 3D cells. The data obtained from confocal and fluorescent microscopy supported the findings of the flow cytometry analysis. Furthermore, the MTT assay showed that Doxil®-liposomes induced less metabolic-disruption compared to free Doxorubicin. Our results also demonstrated that Doxil®-liposomes interacted more loosely with the 3D model than 2D cells, which was further confirmed by measurements of the total migration and invasion areas. Therefore, a 3D model replicating the in vivo conditions of tumor structure and extracellular matrix to assess the delivery of liposomal-nanoparticles to spheroids through a collagen matrix can be more informative and recapitulate the in vivo microenvironment than the 2D model.
Collapse
Affiliation(s)
- Tasneem Alsheleh
- Department of Biology, Faculty of Science, The University of Jordan, Amman, Jordan
| | - Manar Zraikat
- Department of Pharmacology, Faculty of Medicine, The University of Jordan, Amman, Jordan
| | - Fadwa Daoud
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Dana A. Alqudah
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| | - Sharif Abdelghany
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The University of Jordan, Amman, Jordan
| | - Ahmed Abu Siniyeh
- Department of Clinical Laboratory Sciences, Faculty of Science, The University of Jordan, Amman, Jordan
| | - Walhan Alshaer
- Cell Therapy Center, The University of Jordan, Amman, Jordan
| |
Collapse
|
21
|
Lin YY, Lan HY, Teng HW, Wang YP, Lin WC, Hwang WL. Colorectal cancer stem cells develop NK cell resistance via homotypic cell-in-cell structures suppressed by Stathmin1. Theranostics 2025; 15:4308-4324. [PMID: 40225568 PMCID: PMC11984389 DOI: 10.7150/thno.110379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 03/05/2025] [Indexed: 04/15/2025] Open
Abstract
Rationale: Advances in cancer therapies have significantly improved patient survival; however, tumors enriched in cancer stem cells (CSCs) have poor treatment responses. CSCs are a key source of tumor heterogeneity, contributing to therapeutic resistance and unfavorable patient outcomes. In the tumor microenvironment (TME), cell-in-cell (CIC) structures, where one cell engulfs another, have been identified as markers of poor prognosis. Despite their clinical relevance, the mechanisms underlying CIC formation across different tumor cell subpopulations remain largely unknown. Elucidating these processes could provide novel insights and therapeutic opportunities to address aggressive, treatment-resistant cancers. Method: Fluorescent mCherry-carrying colorectal cancer stem cells (CRCSCs) were expanded as spheroids in serum-free media and cocultured with either parental cancer cell-expressing Venus fluorescent protein or CFSE dye-stained immune cells (T cells, M1/M2 macrophages, neutrophils, and NK cells) or treated with EGFR- or PD-L1-targeting antibodies to assess the formation of CIC structures. Genes potentially crucial for the formation of CIC structures were knocked down or overexpressed, and their effects on CIC formation were evaluated. The clinical relevance of the in vitro findings was confirmed through analysis of formalin-fixed, paraffin-embedded (FFPE) human colorectal cancer (CRC) specimens. Results: CRCSCs have a strong predilection for serving as the outer cell in a CIC structure and forming homotypic CIC structures predominantly with parental CRC cells. The frequency of CIC structure formation increased when the cells were exposed to anti-PD-L1 antibody treatment. Both the outer CRCSC in a CIC structure and CRCSCs released from a homotypic CIC structure showed enhanced resistance to the cytotoxicity of NK-92MI cells. Restoration of Stathmin1 (STMN1) expression but not RAC1 knockdown in CRCSCs reduced the homotypic CIC frequency, disrupted the outer cell fate in CIC structures, and increased cell susceptibility to NK-92MI cytotoxicity. In CRC patients, CIC structures are associated with poor tumor differentiation, negative STMN1 expression, and poor prognosis. Conclusion: CSCs play a crucial role in informing CIC structures in CRC. CIC structure formation partially depends on low STMN1 expression and confers a survival advantage under NK cytotoxicity. Targeting this pathway may significantly improve immunotherapy's efficacy for CRC patients.
Collapse
Affiliation(s)
- Yen-Yu Lin
- Department of Pathology, Fu Jen Catholic University Hospital, Fu Jen Catholic University, New Taipei City 24352, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei City 24205, Taiwan
| | - Hsin-Yi Lan
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Hao-Wei Teng
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei 112, Taiwan
- School of Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Ya-Pei Wang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wen-Chun Lin
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
| | - Wei-Lun Hwang
- Department of Biotechnology and Laboratory Science in Medicine, National Yang Ming Chiao Tung University, Taipei 112, Taiwan
- Cancer and Immunology Research Center, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| |
Collapse
|
22
|
Wang Y, Tian W, Li R, Zhou D, Ding K, Feng S, Ge Y, Luo Y, Chen Z, Hou H. Platelet FcRγ inhibits tumor metastasis by preventing the colonization of circulating tumor cells. Eur J Pharmacol 2025; 990:177286. [PMID: 39848529 DOI: 10.1016/j.ejphar.2025.177286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 12/18/2024] [Accepted: 01/20/2025] [Indexed: 01/25/2025]
Abstract
Fc receptor γ subunit (FcRγ) activation plays a crucial role in cancer carcinogenesis. Here, we aimed to uncover the impact of FcRγ on circulating tumor cells (CTC) colonization and the underlying mechanism. FcRγ deficient (FcRγ-/-) mice were used to investigate the functional effects of FcRγ in cancer metastasis, and the results demonstrated that FcRγ deficiency significantly promotes metastasis. The tumor metastasis effect, antiplatelet, platelet or neutrophil infusion experiments were conducted with FcRγ deficient (FcRγ-/-) mice and wild type mice (WT), bearing B16F10 or LCC tumor cells. Blood routine test, flow cytometry, immunofluorescent staining and in vivo image were applied for analysis. Platelet adhesion and neutrophil chemotaxis were analyzed by flow cytometry and ELISA in vitro. Platelet adoptive model was used for mimicing early colonization stage. Our results indicated FcRγ deficiency significantly promoted tumor metastasis accompanied with increased number of platelet and neutrophil in the lung. Further investigation showed that FcRγ-/- platelet infusion, rather than FcRγ-/- neutrophils, promoted CTC colonization. While platelet inhibitor Aspirin abrogated the platelet-mediated infiltration of neutrophil in the lung. Mechanistically, platelet FcRγ deficiency facilitated the adhesion of platelets and cancer cells and increased secretion of CXCL5 and CXCL7 which triggered the platelet-induced neutrophil recruitment. In sum, our study indicates that FcRγ is a restrainer in controlling cancer metastasis through regulating the adhesion of platelets and cancer cells and recruiting more neutrophils, which provides a potential target for anti-metastatic therapies. The level of FcRγ expression in platelets could act as a novel biomarker for cancer metastasis.
Collapse
Affiliation(s)
- Yun Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Wei Tian
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China; School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Rui Li
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Dewang Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Kaiqiang Ding
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Shuang Feng
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Yao Ge
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Yan Luo
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Zhen Chen
- School of Pharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China
| | - Hui Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, PR China.
| |
Collapse
|
23
|
Hussein L, Moaness M, Mabrouk M, Farahat MG, Beherei HH. Advancements in mesoporous bioactive glasses for effective bone cancer therapy: Recent developments and future perspectives. BIOMATERIALS AND BIOSYSTEMS 2025; 17:100108. [PMID: 40083816 PMCID: PMC11904600 DOI: 10.1016/j.bbiosy.2025.100108] [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: 12/30/2024] [Revised: 02/04/2025] [Accepted: 02/14/2025] [Indexed: 03/16/2025] Open
Abstract
This review focuses on recent advancements in the effective use of mesoporous bioactive glasses (MBG) in the treatment of bone cancer, focusing on Osteosarcoma (OS). Bone cancers are rare but are associated with significant morbidity and mortality; often, aggressive treatment is required. Conventional treatments such as surgery, radiation, and chemotherapy are often not enough. This is because surgery cannot completely remove the tumor, without creating a critical size which are defects larger than 2 cm that cannot be repaired by physiological mechanisms. As a result, patients often face the additional burden of radiation and chemotherapy. Scientists have been exploring new treatments, including hyperthermia-targeted therapy, polymeric nanoparticles, and stem cell therapy. This could potentially negatively impact healthy tissues and organs. MBG offers a promising alternative to chemotherapeutic agents and ions for disease treatment as it acts as a multifunctional drug delivery system (DDS). In addition, MBG can also be engineered into scaffolds to facilitate local delivery of growth factors and drugs, thus promoting the efficiency of bone healing and restoration. Therefore, the current review highlights various MBG types reported in the past decade and explores potential future paths to enhance their use in bone cancer treatment while also giving insight on the already commercially available BGs that are used in different bone-related disease.
Collapse
Affiliation(s)
- Laila Hussein
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mona Moaness
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mostafa Mabrouk
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| | - Mohamed G. Farahat
- Botany and Microbiology Department, Faculty of Science, Cairo University, Giza, Egypt
- Biotechnology Department, Faculty of Postgraduate Studies for Nanotechnology, Sheikh Zayed Branch Campus, Cairo University, Sheikh Zayed City 12588, Egypt
| | - Hanan H. Beherei
- Refractories, Ceramics and Building Materials Department, Advanced Materials, Technology and Mineral Resources Research Institute, National Research Centre, 33 El Bohouth St., Dokki, PO Box 12622, Cairo, Egypt
| |
Collapse
|
24
|
Sun J, Hosen MB, Deng WM, Tian A. Epithelial Polarity Loss and Multilayer Formation: Insights Into Tumor Growth and Regulatory Mechanisms. Bioessays 2025; 47:e202400189. [PMID: 39737681 DOI: 10.1002/bies.202400189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 01/01/2025]
Abstract
Epithelial tissues serve as critical barriers in metazoan organisms, maintaining structural integrity and facilitating essential physiological functions. Epithelial cell polarity regulates mechanical properties, signaling, and transport, ensuring tissue organization and homeostasis. However, the barrier function is challenged by cell turnover during development and maintenance. To preserve tissue integrity while removing dying or unwanted cells, epithelial tissues employ cell extrusion. This process removes both dead and live cells from the epithelial layer, typically causing detached cells to undergo apoptosis. Transformed cells, however, often resist apoptosis, leading to multilayered structures and early carcinogenesis. Malignant cells may invade neighboring tissues. Loss of cell polarity can lead to multilayer formation, cell extrusion, and invasion. Recent studies indicate that multilayer formation in epithelial cells with polarity loss involves a mixture of wild-type and mutant cells, leading to apical or basal accumulation. The directionality of accumulation is regulated by mutations in polarity complex genes. This phenomenon, distinct from traditional apical or basal extrusion, exhibits similarities to the endophytic or exophytic growth observed in human tumors. This review explores the regulation and implications of these phenomena for tissue biology and disease pathology.
Collapse
Affiliation(s)
- Jie Sun
- Department of Biochemistry and Molecular Biology, Louisiana Cancer Research Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Md Biplob Hosen
- Department of Biochemistry and Molecular Biology, Louisiana Cancer Research Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Wu-Min Deng
- Department of Biochemistry and Molecular Biology, Louisiana Cancer Research Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Aging Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Aiguo Tian
- Department of Biochemistry and Molecular Biology, Louisiana Cancer Research Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Cancer Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
- Tulane Aging Center, Tulane University School of Medicine, New Orleans, Louisiana, USA
| |
Collapse
|
25
|
Yu Z, Fu J, Mantareva V, Blažević I, Wu Y, Wen D, Battulga T, Wang Y, Zhang J. The role of tumor-derived exosomal LncRNA in tumor metastasis. Cancer Gene Ther 2025; 32:273-285. [PMID: 40011710 DOI: 10.1038/s41417-024-00852-x] [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: 04/24/2024] [Revised: 10/22/2024] [Accepted: 11/05/2024] [Indexed: 02/28/2025]
Abstract
Tumor metastasis regulated by multiple complicated pathways is closely related to variations in the tumor microenvironment. Exosomes can regulate the tumor microenvironment through various mechanisms. Exosomes derived from tumor cells carry a variety of substances, including long non-coding RNAs (lncRNAs), play important roles in intercellular communication and act as critical determinants influencing tumor metastasis. In this review, we elaborate on several pivotal processes through which lncRNAs regulate tumor metastasis, including the regulation of epithelial‒mesenchymal transition, promotion of angiogenesis and lymphangiogenesis, enhancement of the stemness of tumor cells, and evasion of immune clearance. Additionally, we comprehensively summarized a diverse array of potential tumor-derived exosomal lncRNA biomarkers to facilitate accurate diagnosis and prognosis in a clinical setting.
Collapse
Affiliation(s)
- Zhile Yu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Jiali Fu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Vanya Mantareva
- Institute of Organic Chemistry with Centre of Phytochemistry, Bulgarian Academy of Sciences, Bld. 9, 1113, Sofia, Bulgaria
| | - Ivica Blažević
- Department of Organic Chemistry, Faculty of Chemistry and Technology, University of Split, Ruđera Boškovića 35, 21000, Split, Croatia
| | - Yusong Wu
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Dianchang Wen
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China
| | - Tungalag Battulga
- School of Pharmacy, Mongolian National University of Medical Sciences, Ulaanbaatar, Mongolia.
| | - Yuqing Wang
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China.
- The Affiliated Traditional Chinese Medicine Hospital, Guangzhou Medical University, Guangzhou, 510140, PR China.
| | - Jianye Zhang
- The Fifth Affiliated Hospital, Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, The NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences, Guangzhou Medical University, Guangzhou, 510700, PR China.
- The Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, 511518, PR China.
| |
Collapse
|
26
|
Martínez CG, Therapontos S, Lorente JA, Lucena MA, Ortega FG, Serrano MJ. Evaluating MicroRNAs as diagnostic tools for lymph node metastasis in breast cancer: Findings from a systematic review and meta-analysis. Crit Rev Oncol Hematol 2025; 207:104598. [PMID: 39732303 DOI: 10.1016/j.critrevonc.2024.104598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 12/11/2024] [Accepted: 12/12/2024] [Indexed: 12/30/2024] Open
Abstract
Lymph node metastasis (LNM) significantly affects the prognosis and clinical management of breast cancer (BC) patients. This systematic review and meta-analysis aim to identify microRNAs (miRNAs) associated with LNM in BC and evaluate their potential diagnostic and prognostic value. Following PRISMA guidelines, a comprehensive literature search was conducted in PubMed, Web of Science, and SCOPUS databases, to assess the role of miRNAs in LNM BC. The Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool was used to evaluate the quality of included studies. A total of 84 miRNAs were identified as differentially expressed in BC patients with LNM. Of these, a meta-analysis was performed in two microRNAs that were present in at least 3 different articles with a coherent expression direction: miR-155 and miR-34a. The meta-analysis returned a pooled a Log2 fold change of 1.50 for miR-155 (upregulated) and -0.53 for miR-34a (downregulated) with no evidence of publication bias, and a low risk of bias and applicability concerns. To conclude, this study names miR-155 and miR-34a as potential diagnostic biomarkers for LNM in BC, although further experimental validation is necessary to confirm these findings and develop non-invasive diagnostic tools for clinical use.
Collapse
Affiliation(s)
- Coral González Martínez
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain; Biomedical Research Institute IBS-Granada, Avda. de Madrid, 15, Granada 18012, Spain; Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, Granada 18071, Spain; Integral Oncology Division, Virgen de las Nieves University Hospital, Av. Dr. Olóriz 16, Granada 18012, Spain; Molecular Pathology Lab. Pathological Anatomy Unit, University Hospital Virgen de las Nieves, Granada 18016, Spain
| | - Stavros Therapontos
- Utrecht University, Heidelberglaan 8, Utrecht 3584 CS, Netherlands; Integral Oncology Division, Virgen de las Nieves University Hospital, Av. Dr. Olóriz 16, Granada 18012, Spain; Molecular Pathology Lab. Pathological Anatomy Unit, University Hospital Virgen de las Nieves, Granada 18016, Spain
| | - Jose A Lorente
- Laboratory of Genetic Identification, Legal Medicine and Toxicology Department, Faculty of Medicine, University of Granada, Avenida de la Investigación 11, Granada 18071, Spain; Integral Oncology Division, Virgen de las Nieves University Hospital, Av. Dr. Olóriz 16, Granada 18012, Spain; Molecular Pathology Lab. Pathological Anatomy Unit, University Hospital Virgen de las Nieves, Granada 18016, Spain
| | - Miriam Alcaide Lucena
- Unidad de Patología Mamaria, Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario San Cecilio, Granada, Spain; Integral Oncology Division, Virgen de las Nieves University Hospital, Av. Dr. Olóriz 16, Granada 18012, Spain; Molecular Pathology Lab. Pathological Anatomy Unit, University Hospital Virgen de las Nieves, Granada 18016, Spain
| | - F Gabriel Ortega
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain; Biomedical Research Institute IBS-Granada, Avda. de Madrid, 15, Granada 18012, Spain; Integral Oncology Division, Virgen de las Nieves University Hospital, Av. Dr. Olóriz 16, Granada 18012, Spain; Molecular Pathology Lab. Pathological Anatomy Unit, University Hospital Virgen de las Nieves, Granada 18016, Spain.
| | - M Jose Serrano
- GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, Liquid biopsy and Cancer Interception Group, PTS Granada, Avenida de la Ilustración 114, Granada 18016, Spain; Biomedical Research Institute IBS-Granada, Avda. de Madrid, 15, Granada 18012, Spain; Unidad de Patología Mamaria, Servicio de Cirugía General y Aparato Digestivo, Hospital Universitario San Cecilio, Granada, Spain; Integral Oncology Division, Virgen de las Nieves University Hospital, Av. Dr. Olóriz 16, Granada 18012, Spain; Molecular Pathology Lab. Pathological Anatomy Unit, University Hospital Virgen de las Nieves, Granada 18016, Spain.
| |
Collapse
|
27
|
Luo Y, Zhang N, Ye J, Wang Z, Zhou X, Liu J, Cai J, Li C, Chen L. Unveiling lactylation modification: A new hope for cancer treatment. Biomed Pharmacother 2025; 184:117934. [PMID: 39986235 DOI: 10.1016/j.biopha.2025.117934] [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: 12/29/2024] [Revised: 02/13/2025] [Accepted: 02/18/2025] [Indexed: 02/24/2025] Open
Abstract
This review article delves into the multifaceted role of lactylation modification in antitumor therapy, revealing the complex interplay between lactylation modification and the tumor microenvironment (TME), metabolic reprogramming, gene expression, and immunotherapy. As an emerging epigenetic modification, lactylation has a significant impact on the metabolic pathways of tumor cells, immune evasion, gene expression regulation, and sensitivity to chemotherapy drugs. Studies have shown that lactylation modification significantly alters the development and therapeutic response of tumors by affecting metabolites in the TME, immune cell functions, and signaling pathways. In the field of immunotherapy, the regulatory role of lactylation modification provides a new perspective and potential targets for tumor treatment, including modulating the sensitivity of tumors to immunotherapy by affecting the expression of immune checkpoint molecules and the infiltration of immune cells. Moreover, research progress on lactylation modification in various types of tumors indicates that it may serve as a biomarker to predict patients' responses to chemotherapy and immunotherapy. Overall, research on lactylation modification provides a theoretical foundation for the development of new tumor treatment strategies and holds promise for improving patient prognosis and quality of life. Future research will further explore the application potential of lactylation modification in tumor therapy and how to improve treatment efficacy by targeting lactylation modification.
Collapse
Affiliation(s)
- Yuxiang Luo
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Ning Zhang
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Zuao Wang
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Xinchi Zhou
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jipeng Liu
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Jing Cai
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China.
| | - Chen Li
- Department of Orthopedic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Institute of Orthopedics of Jiangxi Province, Nanchang, Jiangxi 330006, China; Jiangxi Provincial Key Laboratory of Spine and Spinal Cord Disease, Jiangxi 330006, China; Institute of Minimally Invasive Orthopedics, Nanchang University, Jiangxi 330006, China.
| | - Leifeng Chen
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, China; Precision Oncology Medicine Center,The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi Province, 330006, People's Republic of China.
| |
Collapse
|
28
|
Zhang M, Zhou G, Xu Y, Wei B, Liu Q, Zhang G, Chang R. Immunogenic cell death signature predicts survival and reveals the role of VEGFA + Mast cells in lung adenocarcinoma. Sci Rep 2025; 15:7213. [PMID: 40021802 PMCID: PMC11871002 DOI: 10.1038/s41598-025-91401-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 02/20/2025] [Indexed: 03/03/2025] Open
Abstract
Lung cancer is prevalent worldwide and is a major cause of cancer-related mortality. Despite being the primary model for immunotherapy research, the response rates of lung cancer patients to immunotherapy are unsatisfactory. Furthermore, research on immunogenic cell death (ICD) in lung cancer is limited, which limits the development of strategies that combine ICD-related therapies with immunotherapy. In this study, we compiled and summarized 69 genes associated with ICD and developed an IRS. Across seven independent datasets, the IRS was identified as an independent prognostic factor. IRS was positively associated with multiple tumor proliferation pathways and negatively associated with immune-related pathways. Additionally, IRS negatively correlated with the infiltration of various immune cells, supporting its association with survival outcomes. Based on the correlation between IRS and immune activity, we validated the ability of IRS to predict immunotherapy efficacy across seven immunotherapy datasets and demonstrated that patients who respond to immunotherapy tend to have a lower IRS. Moreover, utilizing single-cell RNA sequencing, we revealed the role of mast cells in the TME with the highest IRS. Through interactions with various receptors on macrophages, endothelial cells, and tumor cells, mast cells promote tumor progression, providing a comprehensive explanation for poor prognosis and lack of response to immunotherapy in patients with high IRS. Our study offers new guidance for combination therapies in lung adenocarcinoma patients and elucidated the mechanism by which mast cells contribute to cancer development within the TME.
Collapse
Affiliation(s)
- Meng Zhang
- The Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
| | - Guowei Zhou
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yantao Xu
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Benliang Wei
- Big Data Institute, Central South University, Changsha, Hunan, China
| | - Qian Liu
- Big Data Institute, Central South University, Changsha, Hunan, China
| | - Guanxiong Zhang
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
- The Department of Dermatology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, Changsha, Hunan, China.
- Furong Laboratory, Changsha, Hunan, China.
| | - Ruimin Chang
- The Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
- Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Changsha, Hunan, China.
| |
Collapse
|
29
|
Ma M, Liu C, Jiang L, Liu D, Zhang P, Tao M, Zhang M, Gong J, Peng Z, Zhang X, Li J, Zheng C, Deng M, Shen L, Qi C. Exploring the therapeutic efficacy difference in claudin18.2-targeted cell therapy revealed by single-cell sequencing. iScience 2025; 28:111768. [PMID: 39925434 PMCID: PMC11804729 DOI: 10.1016/j.isci.2025.111768] [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: 01/26/2024] [Revised: 09/02/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
Chimeric antigen receptor T cell (CAR T) therapy has been successfully used to treat hematological malignancies. Nonetheless, its application to solid tumors remains challenging. Our previous analysis of the ongoing clinical trial (NCT03874897) demonstrated promising results in patients with advanced CLDN18.2-positive gastric cancer who received CT041 CAR T treatment. Here, we collected peripheral blood and ascites from five patients from the clinical trial 3 and 7 days (d) after CT041 infusion. Patients with a high proportion of naïve-like T cells were more likely to benefit from CT041 treatment. We found that high expression of CLDN18 in ascites epithelial cells correlated with a favorable prognosis, whereas ascites epithelial cells with high MYC expression and strong interactions between tumor cells and T cells were adverse prognostic factors for CT041 treatment. These findings may provide theoretical evidence for the screening of populations that can benefit from CAR T therapy and improve the efficacy of CAR T therapy.
Collapse
Affiliation(s)
- Mingyang Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Chang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Lei Jiang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Dan Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Panpan Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Min Tao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Miao Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
| | - Jifang Gong
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Zhi Peng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Xiaotian Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Jian Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Chunhong Zheng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
- International Cancer Institute, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Mi Deng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, 52 Fucheng Road, Beijing 100142, China
- International Cancer Institute, Peking University, No. 38 Xueyuan Road, Beijing 100191, China
- State Key Laboratory of Molecular Oncology, Beijing Key Laboratory of Carcinogenesis and Translational Research, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | - Changsong Qi
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Early Drug Development Centre, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
30
|
Zhu XR, Zhu JQ, Gu QH, Liu N, Lu JJ, Li XH, Liu YY, Zheng X, Chen MB, Ji Y. A novel identified epithelial ligand-receptor-associated gene signature highlights POPDC3 as a potential therapy target for non-small cell lung cancer. Cell Death Dis 2025; 16:114. [PMID: 39971925 PMCID: PMC11840029 DOI: 10.1038/s41419-025-07410-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 01/13/2025] [Accepted: 01/30/2025] [Indexed: 02/21/2025]
Abstract
The tumor microenvironment (TME) is pivotal in non-small cell lung cancer (NSCLC) progression, influencing drug resistance and immune cell behavior through complex ligand-receptor (LR) interactions. This study developed an epithelial LR-related prognostic risk score (LRrisk) to identify biomarkers and targets in NSCLC. We identified twenty epithelial LRs with significant prognostic implications and delineated three molecular NSCLC subtypes with distinct outcomes, pathological characteristics, biological pathways, and immune profiles. The LRrisk model was constructed using twelve differentially expressed ligand-receptor interaction-related genes (LRGs), with a focus on POPDC3 (popeye domain-containing protein 3), which was overexpressed in NSCLC cells. Functional assays revealed that POPDC3 knockdown reduced cell proliferation, migration, invasion, and epithelial-mesenchymal transition (EMT), while its overexpression promoted cancerous activities. In vivo, POPDC3 silencing hindered, and its overexpression accelerated the growth of NSCLC xenografts in nude mice. Additionally, high expression levels of POPDC3 in NSCLC tissues were associated with enhanced CD4+ T cell infiltration and increased PD-1 expression within the TME. Moreover, ectopic POPDC3 overexpression in C57BL/6 J mouse Lewis lung carcinoma (LLC) xenografts enhanced CD4+ T cell infiltration and PD-1 expression in the TME. This research establishes a robust epithelial LR-related signature, highlighting POPDC3 as a critical facilitator of NSCLC progression and a potential therapeutic target.
Collapse
Affiliation(s)
- Xiao-Ren Zhu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Jia-Qi Zhu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Qian-Hui Gu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
| | - Na Liu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Jing-Jing Lu
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Xiao-Hong Li
- Department of Clinical Laboratory, The First People's Hospital of Taicang, Taicang, China
| | - Yuan-Yuan Liu
- Clinical Research and Lab Center, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China
- Medical School of Jiangsu University, Zhenjiang, China
| | - Xian Zheng
- Medical School of Jiangsu University, Zhenjiang, China.
- Department of Pharmacy, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
| | - Min-Bin Chen
- Department of Radiotherapy and Oncology, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, China.
- Medical School of Jiangsu University, Zhenjiang, China.
| | - Yong Ji
- Department of Thoracic Surgery, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi People's Hospital, Wuxi Medical Center, Nanjing Medical University, Wuxi, China.
| |
Collapse
|
31
|
Akyol D, Özcan FG. The effect of preoperative laboratory values on prognostic factors following cytoreduction surgery in ovarian cancer: Neutrophil lymphocyte ratio, platelet lymphocyte ratio, and lymphocyte monocyte ratio. Medicine (Baltimore) 2025; 104:e41504. [PMID: 39960969 PMCID: PMC11835068 DOI: 10.1097/md.0000000000041504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Accepted: 01/23/2025] [Indexed: 02/20/2025] Open
Abstract
In cancer patients, prognostic markers are needed to improve the management and clinical course of both the cancer itself and surgery therefor. Elevated systemic inflammatory markers are associated with morbidity and mortality in most cancer types. In this study, we aimed to determine the prognostic value of inflammatory markers such as neutrophil to lymphocyte ratio (NLR), platelet to lymphocyte ratio (PLR), and lymphocyte to monocyte ratio (LMR) in patients undergoing cytoreductive surgery for ovarian cancer. The data of 188 patients who underwent surgery for ovarian cancer between December 2022 and December 2023 were retrospectively analyzed. Receiver operating characteristic (ROC) curves were constructed to evaluate the correlation between complications and the inflammatory prognostic factors NLR, PLR, and LMR. Optimal cutoff values were determined as the points where the Youden index (sensitivity + specificity - 1) was maximal. Patients were compared according to the presence of complications. As a result of the ROC curve analysis, patients were divided as high and low NLR and PLR groups. The difference of preoperative and postoperative inflammatory prognostic factors was compared according to the presence of complications. In this study in which a total of 90 patients were evaluated, the cutoff value for NLR was 2.04 (areas under the ROC curve: 0.655; P < .05) and the cutoff value for PLR was 145.3 (areas under the ROC curve: 0.740; P < .05) according to the presence of complications. In the group with complications, lymphopenia and thrombocytosis were more common preoperatively, while patients were more anemic postoperatively (P < .05). Patients in the high NLR group were younger and received less neoadjuvant chemotherapy. In the high PLR group, the number of patients receiving neoadjuvant chemotherapy was lower, and although the patients were more anemic and lymphopenic, higher rates of neutrophilia and thrombocytosis were observed. The analysis of preoperative and postoperative NLR, PLR, and LMR differences revealed an increase in NLR and PLR values and a decrease in LMR values (P < .05). The preoperative systemic inflammatory biomarkers NLR and PLR may be considered as prognostic predictors of poor postoperative outcomes. Therefore, consideration of these biomarkers may have an important role in clinical course management.
Collapse
Affiliation(s)
- Duygu Akyol
- Department of Anesthesiology and Reanimation, Başakşehir Çam and Sakura City Hospital, Republic of Turkey Ministry of Health, İstanbul, Turkey
| | - Funda Gümüş Özcan
- Department of Anesthesiology and Reanimation, Başakşehir Çam and Sakura City Hospital, Republic of Turkey Ministry of Health, İstanbul, Turkey
| |
Collapse
|
32
|
Ilg MM, Lapthorn AR, Harding SL, Minhas T, Koduri G, Bustin SA, Cellek S. Development of a phenotypic screening assay to measure activation of cancer-associated fibroblasts. Front Pharmacol 2025; 16:1526495. [PMID: 40017592 PMCID: PMC11865240 DOI: 10.3389/fphar.2025.1526495] [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: 11/11/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Background In cancer metastasis, tumor cells condition distant tissues to create a supportive environment, or metastatic niche, by driving the activation of cancer-associated fibroblasts (CAFs). These CAFs remodel the extracellular matrix, creating a microenvironment that supports tumor growth and compromises immune cell function, enabling cancer cells to evade immune detection. Consequently, targeting the activation of CAFs has been proposed as a therapeutic strategy to hinder metastatic spread. Our objective was to develop the first in vitro phenotypic screening assay capable of assessing this activation process. Methods Human primary lung fibroblasts were co-cultured with highly invasive breast cancer cells (MDA-MB-231) to identify changes in the expression of selected genes using RT-qPCR. An In-Cell ELISA (ICE)-based assay using human lung fibroblasts, MDA-MB-231 cells and human monocytes (THP-1 cells) was developed to measure the activation of CAFs. Another ELISA assay was used to measure released osteopontin. Results When lung fibroblast were co-cultured with MDA-MB-231 cells, among the 10 selected genes, the genes for osteopontin (SPP1), insulin like growth factor 1 (IGF1), periostin (POSTN) and α-smooth muscle actin (α-SMA, ACTA2) elicited the greatest fold change (55-, 37-, 8- and 5-fold respectively). Since osteopontin, IGF-1 and periostin are secreted proteins and α-SMA is an intracellular cytoskeleton protein, α-SMA was chosen to be the readout biomarker for the ICE assay. When fibroblasts were co-cultured with MDA-MB-231 cells and monocytes in the 96 well ICE assay, α-SMA expression was increased 2.3-fold yielding a robust Z' of 0.56. A secondary, low throughput assay was developed by measuring the release of osteopontin which showed a 6-fold increase when fibroblasts were co-cultured with MDA-MB-231 cells and monocytes. Discussion This phenotypic assay is the first to measure the activation of CAFs in a 96-well format, making it suitable for medium-to high-throughput screening of potential therapeutic compounds. By focusing on observable cellular phenotypic changes rather than targeting specific molecular pathways, this assay allows for a broader and unbiased identification of compounds capable of modulating CAF activation.
Collapse
Affiliation(s)
- Marcus M. Ilg
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Alice R. Lapthorn
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Sophie L. Harding
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Tariq Minhas
- The Essex Cardiothoracic Centre, Basildon University Hospital, Basildon, United Kingdom
| | - Gouri Koduri
- Southend University Hospital NHS Foundation Trust, Westcliff-on-Sea, United Kingdom
| | - Stephen A. Bustin
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| | - Selim Cellek
- Fibrosis Research Group, Medical Technology Research Centre, Anglia Ruskin University, Chelmsford, United Kingdom
| |
Collapse
|
33
|
Niitsu H, Nakahara H, Ishida K, Kaneko Y, Hayes CN, Arihiro K, Hinoi T. Real-world data analysis for factors influencing the quality check status in FoundationOne CDx cancer genomic profiling tests. Sci Rep 2025; 15:5167. [PMID: 39939621 PMCID: PMC11822185 DOI: 10.1038/s41598-025-85846-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 01/06/2025] [Indexed: 02/14/2025] Open
Abstract
Comprehensive genomic profiling (CGP) testing is used worldwide for personalized cancer treatment. Unstained slides from formalin-fixed paraffin-embedded (FFPE) blocks are required for tissue-based CGP testing. However, the use of low quality FFPE specimens can result in testing failure or invalid results. Although several previous studies have indicated that the quality of genomic testing is affected by various factors, the factor(s) that has the greatest influence on CGP testing has not been well studied in real-world data. Thus, we conducted a large-scale multi-institutional study in Hiroshima, Japan to investigate the factors associated with quality check status in FoundationOne CDx CGP testing using real-world data from 1,204 participants from an area with a population of approximately 2.7 million. This study revealed low percentage of tumor nuclei in FFPE specimen, long-term storage of FFPE block, and pancreatic cancer as independent risk factors for qualified status. Of the three factors, percentage of tumor nuclei had the largest effect on quality check status, while the magnitudes of the effects of storage time of FFPE or pancreatic cancer were minor. Collectively, our real-world data indicate that tumor purity is the most important factor for successful CGP, and we would suggest greater than 35% as an ideal percentage of tumor nuclei for CGP submission.
Collapse
Affiliation(s)
- Hiroaki Niitsu
- Department of Clinical and Molecular Genetics, Hiroshima University Hospital, Hiroshima, Japan
| | - Hikaru Nakahara
- Department of Clinical and Molecular Genetics, Hiroshima University Hospital, Hiroshima, Japan
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Katsunari Ishida
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| | - Yoshie Kaneko
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| | - C Nelson Hayes
- Department of Gastroenterology, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Koji Arihiro
- Department of Anatomical Pathology, Hiroshima University Hospital, Hiroshima, Japan
| | - Takao Hinoi
- Department of Clinical and Molecular Genetics, Hiroshima University Hospital, Hiroshima, Japan.
- Department of Clinical and Molecular Genetics, Hiroshima University Hospital Address, 1-2-3 Kasumi, Minami-ku, Hiroshima City, 734-8551, Hiroshima, Japan.
| |
Collapse
|
34
|
Starikova EA, Mammedova JT, Rubinstein AA, Sokolov AV, Kudryavtsev IV. Activation of the Coagulation Cascade as a Universal Danger Sign. Curr Issues Mol Biol 2025; 47:108. [PMID: 39996829 PMCID: PMC11854423 DOI: 10.3390/cimb47020108] [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: 12/28/2024] [Revised: 01/29/2025] [Accepted: 02/02/2025] [Indexed: 02/26/2025] Open
Abstract
Hemostasis is a mechanism that stops bleeding from an injured vessel, involves multiple interlinked steps, culminating in the formation of a "clot" sealing the damaged area. Moreover, it has long been recognized that inflammation also provokes the activation of the coagulation system. However, there has been an increasing amount of evidence revealing the immune function of the hemostasis system. This review collects and analyzes the results of the experimental studies and data from clinical observations confirming the inflammatory function of hemostasis. Here, we summarize the latest knowledge of the pathways in immune system activation under the influence of coagulation factors. The data analyzed allow us to consider the components of hemostasis as receptors recognizing «foreign» or damaged «self» or/and as «self» damage signals that initiate and reinforce inflammation and affect the direction of the adaptive immune response. To sum up, the findings collected in the review allow us to classify the coagulation factors, such as Damage-Associated Molecular Patterns that break down the conventional concepts of the coagulation system.
Collapse
Affiliation(s)
- Eleonora A. Starikova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
- Department of Microbiology and Virology, Institute of Medical Education Almazov National Medical Research Centre, 2 Akkuratova Street, 197341 Saint Petersburg, Russia
| | - Jennet T. Mammedova
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Department of Molecular Biotechnology, Chemical and Biotechnology Faculty, Saint Petersburg State Institute of Technology, Moskovski Ave., 26, 190013 Saint Petersburg, Russia
| | - Artem A. Rubinstein
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
| | - Alexey V. Sokolov
- Laboratory of Systemic Virology, Department of Molecular Biology of Viruses, Smorodintsev Research Institute of Influenza, 15/17, Prof. Popova Str., 197376 Saint Petersburg, Russia;
| | - Igor V. Kudryavtsev
- Laboratory of Cellular Immunology, Department of Immunology, Institute of Experimental Medicine, Akademika Pavlova 12, 197376 Saint Petersburg, Russia (I.V.K.)
- Medical Faculty, First Saint Petersburg State I. Pavlov Medical University, L’va Tolstogo St. 6-8, 197022 Saint Petersburg, Russia
| |
Collapse
|
35
|
Zhang W, Ge L, Zhang Y, Zhang Z, Zhang W, Song F, Huang P, Xu T. Targeted intervention of tumor microenvironment with HDAC inhibitors and their combination therapy strategies. Eur J Med Res 2025; 30:69. [PMID: 39905506 PMCID: PMC11792708 DOI: 10.1186/s40001-025-02326-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 01/23/2025] [Indexed: 02/06/2025] Open
Abstract
Histone deacetylation represents a significant epigenetic mechanism that involves the removal of acetyl groups from histones, subsequently influencing gene transcription. Overexpression of histone deacetylases (HDACs) is prevalent across various cancer types, positioning HDAC inhibitors as broadly applicable therapeutic agents. These inhibitors are known to enhance tumor immune antigenicity, potentially slowing tumor progression. Furthermore, the tumor microenvironment, which is intricately linked to cancer development, acts as a mediator in the proliferation of numerous cancers and presents a viable target for oncological therapies. This paper primarily explores how HDAC inhibitors can regulate cancer progression via the tumor microenvironment and suppress tumor growth through multiple pathways, in addition to examining the synergistic effects of combined drug therapies involving HDAC inhibitors.
Collapse
Affiliation(s)
- Wanli Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Luqi Ge
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Yiwen Zhang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China
- Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, China
| | - Zhentao Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Wen Zhang
- Department of Pharmacology, College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - Feifeng Song
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| | - Ping Huang
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
- Clinical Research Center for Cancer of Zhejiang Province, Hangzhou, China.
| | - Tong Xu
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People's Hospital(Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, Zhejiang, China.
- Key Laboratory of Endocrine Gland Diseases of Zhejiang Province, Hangzhou, 310014, China.
| |
Collapse
|
36
|
Song K, Chen C, Xu H, Chen L, Xu H, Han X, Chen H, Qin Z. Prediction of Survival in the Elderly Patients with Glioblastoma using Cumulative Inflammatory Markers Score. J Neurol Surg B Skull Base 2025; 86:98-105. [PMID: 39881741 PMCID: PMC11774615 DOI: 10.1055/s-0044-1779050] [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: 08/04/2023] [Accepted: 12/21/2023] [Indexed: 01/31/2025] Open
Abstract
Objectives This retrospective study aimed to explore the prognostic effect of cumulative score based on neutrophil-to-lymphocyte ratio (NLR), platelet-to-lymphocyte ratio (PLR), and fibrinogen in older adults diagnosed with glioblastoma (GBM). Design Retrospective study. Setting Huashan Hospital. Participants Patients aged over 60 years and diagnosed with GBM between 2010 and 2017. Main Outcome Measures Results of preoperative routine biochemistry and coagulation blood examinations were reviewed from medical records. Overall survival (OS) was considered a period from first resection surgery until death. Progression-free survival (PFS) was considered a period from initial operation until the date of tumor progression demonstrated in brain magnetic resonance imaging or death from any cause. If no event occurred, the last follow-up appointment was the end of the observation for OS or PFS. The Kaplan-Meier method was used to evaluate survival curves, and prognostic factors were analyzed by the Cox proportional hazards model. Results A total of 289 patients were included. Patients with higher levels of fibrinogen, NLR, and PLR had significantly shorter median OS ( p = 0.001, p = 0.016, and p = 0.002, respectively) and PFS ( p = 0.004, p = 0.022, and p = 0.009, respectively) compared with those with lower levels. Multivariate analyses showed a significant association between higher F-NLR-PLR score and reduced OS (adjusted hazard ratios [aHRs]: 1.356, 95% confidence interval [CI] 1.009-1.822 for scores 1-2 compared with 0; 5.974, 95% CI 2.811-12.698 for score 3 compared with 0). Similarly, a significant association between higher F-NLR-PLR score and reduced PFS was observed (aHR: 1.428, 95% CI 1.066-1.912 for scores 1-2 compared with 0; aHR: 2.860, 95% CI 1.315-6.223 for score 3 compared with 0). Conclusion Higher F-NLR-PLR score is associated with reduced OS and PFS in older adults with GBM, which helps identify patients at high risk and guide the individualized treatment in clinical practice.
Collapse
Affiliation(s)
- Kun Song
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| | - Chunjui Chen
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| | - Hao Xu
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| | - Lingchao Chen
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| | - Hongzhi Xu
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| | - Xi Han
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| | - Hong Chen
- Department of Neuropathology, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| | - Zhiyong Qin
- Department of Neurosurgery, Huashan Hospital Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
37
|
To A, Yu Z, Sugimura R. Recent advancement in the spatial immuno-oncology. Semin Cell Dev Biol 2025; 166:22-28. [PMID: 39705969 DOI: 10.1016/j.semcdb.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Recent advancements in spatial transcriptomics and spatial proteomics enabled the high-throughput profiling of single or multi-cell types and cell states with spatial information. They transformed our understanding of the higher-order architectures and paired cell-cell interactions within a tumor microenvironment (TME). Within less than a decade, this rapidly emerging field has discovered much crucial fundamental knowledge and significantly improved clinical diagnosis in the field of immuno-oncology. This review summarizes the conceptual frameworks to understand spatial omics data and highlights the updated knowledge of spatial immuno-oncology.
Collapse
Affiliation(s)
- Alex To
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Zou Yu
- School of Biomedical Sciences, University of Hong Kong, Hong Kong
| | - Ryohichi Sugimura
- School of Biomedical Sciences, University of Hong Kong, Hong Kong; Centre for Translational Stem Cell Biology, Hong Kong.
| |
Collapse
|
38
|
Mano Y, Igarashi Y, Komori K, Hashimoto I, Watanabe H, Takahashi K, Kano K, Fujikawa H, Yamada T, Himuro H, Kouro T, Wei F, Tsuji K, Horaguchi S, Komahashi M, Oshima T, Sasada T. Characteristics and clinical significance of immune cells in omental milky spots of patients with gastric cancer. Front Immunol 2025; 16:1521278. [PMID: 39949777 PMCID: PMC11821591 DOI: 10.3389/fimmu.2025.1521278] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 01/02/2025] [Indexed: 02/16/2025] Open
Abstract
The omentum is a common site of peritoneal metastasis in various cancers, including gastric cancer. It contains immune cell aggregates known as milky spots, which provide a microenvironment for peritoneal immunity by regulating innate and adaptive immune responses. In this study, we investigated gene expression profiles in cells from omental milky spots of patients with gastric cancer (n = 37) by RNA sequencing analysis and classified the patients into four groups (G1-4). Notably, significant differences were observed between the groups in terms of macroscopic type, lymphatic invasion, venous invasion, and pathological stage (pStage). G3, which was enriched in genes related to acquired immunity, showed earlier tumor stages (macroscopic type 0, Ly0, V0, and pStage I) and a better prognosis. In contrast, G4 showed enrichment of genes related to neutrophils and innate immunity; G1 and G2 showed no enrichment of innate or adaptive immune-related genes, suggesting an immune desert microenvironment. Cytometric analysis revealed significantly more T and B cells and fewer neutrophils in G3. Accordingly, the immune microenvironment in omental milky spots may vary depending on the stage of gastric cancer progression. When univariate Cox proportional hazards regression models were used to search for prognostically relevant genes specific to G3, 23 potential prognostic genes were identified as common genes associated with relapse-free survival and overall survival. In addition, the multivariate Cox proportional hazards model using these prognostic genes and clinicopathological information showed that combining the B cell marker CD19 and Ly had a high predictive accuracy for prognosis. Based on this study's results, it is possible that tumor progression, such as lymphatic and/or venous infiltration of tumor cells, may affect the immune cell composition and proportions in omental milky spots of patients with gastric cancer and analysis of gene expression in omental milky spots may help to predict gastric cancer prognosis.
Collapse
Affiliation(s)
- Yasunobu Mano
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Keisuke Komori
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
- Department of Surgery, Yokohama City University, Yokohama, Japan
| | - Itaru Hashimoto
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
- Department of Surgery, Yokohama City University, Yokohama, Japan
| | - Hayato Watanabe
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Kosuke Takahashi
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Kazuki Kano
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Hirohito Fujikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Takanobu Yamada
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Hidetomo Himuro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Taku Kouro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Kayoko Tsuji
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shun Horaguchi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Mitsuru Komahashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Takashi Oshima
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| |
Collapse
|
39
|
Liao S, Gao X, Zhou K, Kang Y, Ji L, Zhong X, Lv J. Exploration of metastasis-related signatures in osteosarcoma based on tumor microenvironment by integrated bioinformatic analysis. Heliyon 2025; 11:e41358. [PMID: 39844989 PMCID: PMC11750479 DOI: 10.1016/j.heliyon.2024.e41358] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 11/21/2024] [Accepted: 12/18/2024] [Indexed: 01/24/2025] Open
Abstract
Background The present study aims to explore the metastasis-related signatures in connection with tumor microenvironment (TME), revealing new molecular targets promising in improving osteosarcoma (OS) patients' outcomes. Methods The high-throughput sequencing data was downloaded from the TARGET database and performed the ESTIMATE algorithm. Metastasis-related information was obtained from the GSE21257 dataset. Differentially expressed genes (DEGs) associated with the stromal and immune cell infiltration patterns were identified. DEGs with similar biological functions were grouped into the same module by Gene Ontology (GO) analysis and MCODE analysis. Prognostic DEGs were selected in two datasets through survival analysis. Weighted gene co-expression network analysis (WGCNA) was performed to find metastasis-related modules and genes. RT-PCR was utilized to evaluate the expression of the key prognostic DEGs associated with metastasis in OS patients. Results The median scores of the stromal and immune groups of OS samples were 58 and -416, and a total of 200 overlapping DEGs were identified. These DEGs basically played fundamental roles in immune response relevant GO terms and were clustered into 9 different modules. Among them, 24 metastasis-related DEGs were selected from the GSE21257 dataset which contains the stromal and immune cell infiltration patterns. Finally, IRF8, HLA-DMA, and HLA-DMB were proved to exhibit significant higher expression levels in cancerous tissues than in para-cancerous tissues for OS patients. Conclusion We identified three principal genes as promising signatures for predicting the survival the prognosis of OS patients. Exploration of metastasis-related signatures in TME may be valuable for enhancing treatment strategies for OS.
Collapse
Affiliation(s)
- Shiyao Liao
- Center for Plastic & Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Xing Gao
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215000, China
| | - Kai Zhou
- The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325006, China
| | - Yao Kang
- Center for Plastic & Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Lichen Ji
- Center for Plastic & Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Xugang Zhong
- Center for Plastic & Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
- Qingdao University, Qingdao, Shandong, 266000, China
| | - Jun Lv
- Center for Plastic & Reconstructive Surgery, Department of Orthopedics, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| |
Collapse
|
40
|
Zhao X, Yang L, Pan J, Zeng Z, Zhang T, Yang Y, Zhang J, Chen T, Xiao Z, Pan W. CXCL8 modulates M0 macrophage proliferation and polarization to influence tumor progression in cervical cancer. Sci Rep 2025; 15:790. [PMID: 39755693 PMCID: PMC11700176 DOI: 10.1038/s41598-024-81726-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: 08/28/2024] [Accepted: 11/28/2024] [Indexed: 01/06/2025] Open
Abstract
Cervical cancer (CESC) presents significant clinical challenges due to its complex tumor microenvironment (TME) and varied treatment responses. This study identified undifferentiated M0 macrophages as high-risk immune cells critically involved in CESC progression. Co-culture experiments further demonstrated that M0 macrophages significantly promoted HeLa cell proliferation, migration, and invasion, underscoring their pivotal role in modulating tumor cell behavior within the TME. A nine-gene prognostic model constructed from immune gene signatures highlighted CXCL8 as a key regulator of M0 macrophage behavior. Functional experiments demonstrated that CXCL8 knockdown in M0 macrophages inhibited their proliferation, shifted polarization toward an M1-dominant phenotype, and reduced tumor-promoting M2 polarization. Co-culture experiments with CXCL8-deficient M0 macrophages further revealed a suppression of HeLa cell proliferation, migration, and invasion. These findings position M0 macrophages as central regulators within the TME and suggest that targeting pathways like CXCL8 could provide novel therapeutic strategies for improving outcomes in CESC patients.
Collapse
Affiliation(s)
- Xiyan Zhao
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang, 550009, China
- Guizhou Institute of Precision Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550009, China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang, 550009, China
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guizhou, Guiyang, 550025, China
| | - Li Yang
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang, 550009, China
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang, 550009, China
- Department of Medical Laboratory science, Guizhou Medical University, Guizhou, Guiyang, 550004, China
| | - Jigang Pan
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guizhou, Guiyang, 550025, China
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, Guiyang, 550025, China
| | - Zhirui Zeng
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guizhou, Guiyang, 550025, China
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, Guiyang, 550025, China
| | - Tuo Zhang
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guizhou, Guiyang, 550025, China
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, Guiyang, 550025, China
| | - Yushi Yang
- Department of Pathology, The Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang, 550025, China
| | - Jingjing Zhang
- Affiliated Children's Hospital, Nanjing Medical University School of Pediatrics, Nanjing, Jiangsu, 210008, China
| | - Tengxiang Chen
- Guizhou Institute of Precision Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550009, China.
- Transformation Engineering Research Center of Chronic Disease Diagnosis and Treatment, Department of Physiology, College of Basic Medicine, Guizhou Medical University, Guizhou, Guiyang, 550025, China.
- Guizhou Provincial Key Laboratory of Pathogenesis & Drug Research on Common Chronic Diseases, Guizhou Medical University, Guizhou, Guiyang, 550025, China.
| | - Ziwen Xiao
- Guizhou Institute of Precision Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550009, China.
- Department of Obstetrics and Gynecology, The Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang, 550009, China.
| | - Wei Pan
- Prenatal Diagnosis Center in Guizhou Province, The Affiliated Hospital of Guizhou Medical University, Guizhou, Guiyang, 550009, China.
- Guizhou Institute of Precision Medicine, the Affiliated Hospital of Guizhou Medical University, Guiyang, 550009, China.
| |
Collapse
|
41
|
Qian BZ, Ma RY. Immune Microenvironment in Breast Cancer Metastasis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1464:413-432. [PMID: 39821036 DOI: 10.1007/978-3-031-70875-6_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Metastatic disease is the final stage of breast cancer that accounts for vast majority of patient death. Mounting data over recent years strongly support the critical roles of the immune microenvironment in determining breast cancer metastasis. The latest single-cell studies provide further molecular evidence illustrating the heterogeneity of this immune microenvironment. This chapter summarizes major discoveries on the role of various immune cells in metastasis progression and discusses future research opportunities. Studies investigating immune heterogeneity within primary breast cancer and across different metastasis target organs can potentially lead to more precise treatment strategies with improved efficacy.
Collapse
Affiliation(s)
- Bin-Zhi Qian
- Department of Oncology, Fudan University Shanghai Cancer Center, Zhangjiang-Fudan International Innovation Center, Shanghai Medical College, The Human Phenome Institute, Fudan University, Shanghai, China.
| | - Ruo-Yu Ma
- Department of Oncology, Fudan University Shanghai Cancer Center, Zhangjiang-Fudan International Innovation Center, Shanghai Medical College, The Human Phenome Institute, Fudan University, Shanghai, China
| |
Collapse
|
42
|
Isaguliants M, Zhitkevich A, Petkov S, Gorodnicheva T, Mezale D, Fridrihsone I, Kuzmenko Y, Kostyushev D, Kostyusheva A, Gordeychuk I, Bayurova E. Enzymatic activity of HIV-1 protease defines migration of tumor cells in vitro and enhances their metastatic activity in vivo. Biochimie 2025; 228:32-43. [PMID: 39128490 DOI: 10.1016/j.biochi.2024.08.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 07/09/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
Overexpression of aspartic proteases, as cathepsin D, is an independent marker of poor prognosis in breast cancer, correlated with the incidence of clinical metastasis. We aimed to find if HIV-1 aspartic protease (PR) can play a similar role. Murine adenocarcinoma 4T1luc2 cells were transduced with lentivirus encoding inactivated drug-resistant PR, generating subclones PR20.1 and PR20.2. Subclones were assessed for production of reactive oxygen species (ROS), expression of epithelial-mesenchymal transition (EMT) factors, and in vitro migratory activity in the presence or absence of antioxidant N-acetyl cysteine and protease inhibitors. Tumorigenic activity was evaluated by implanting cells into BALB/c mice and following tumor growth by calipering and bioluminescence imaging in vivo, and metastases, by organ imaging ex vivo. Both subclones expressed PR mRNA, and PR20.2, also the protein detected by Western blotting. PR did not induce production of ROS, and had no direct effect on cell migration rate, however, treatment with inhibitors of drug-resistant PR suppressed the migratory activity of both subclones. Furthermore, expression of N-cadherin and Vimentin in PR20.2 cells and their migration were enhanced by antioxidant treatment. Sensitivity of in vitro migration to protease inhibitors and to antioxidant, known to restore PR activity, related the effects to the enzymatic activity of PR. In vivo, PR20.2 cells demonstrated higher tumorigenic and metastatic activity than PR20.1 or parental cells. Thus, HIV-1 protease expressed in breast cancer cells determines their migration in vitro and metastatic activity in vivo. This effect may aggravate clinical course of cancers in people living with HIV-1.
Collapse
Affiliation(s)
- M Isaguliants
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - A Zhitkevich
- Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| | - S Petkov
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - T Gorodnicheva
- Institute of Translational Medicine, Pirogov Russian National Research Medical University, 117997, Moscow, Russia.
| | - D Mezale
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - I Fridrihsone
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden.
| | - Y Kuzmenko
- Engelhardt Institute of Molecular Biology, Academy of Sciences of the Russian Federation, 119991, Moscow, Russia.
| | - D Kostyushev
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119991, Moscow, Russia.
| | - A Kostyusheva
- Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, Sechenov University, 119991, Moscow, Russia.
| | - I Gordeychuk
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden; Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| | - E Bayurova
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, 17177, Stockholm, Sweden; Chumakov Federal Scientific Center for Research and Development of Immune-and-Biological Products of Russian Academy of Sciences, 108819, Moscow, Russia.
| |
Collapse
|
43
|
Hałas-Wiśniewska M, Arendt W, Grzanka A, Izdebska M. Downregulation of Ezrin Suppresses Migration Potential in Cervical Cancer Cells. Pharmaceuticals (Basel) 2024; 18:3. [PMID: 39861066 PMCID: PMC11769092 DOI: 10.3390/ph18010003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 12/16/2024] [Accepted: 12/20/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND The literature reports that ezrin (EZR) is important as a linker between microfilaments and cellular environments. Moreover, it affects cancer cell migration, but the exact mechanism is not fully understood. In this study, we aimed to investigate the role of EZR in the migration of two different types of cervical cancer cells-from primary lesion (SiHa) and lymph node metastases (HT-3). In addition, we showed for the first time that a reduced EZR protein level affects the cellular response to the routinely used treatment with cisplatin. METHODS The most important stage of the study consisted of conducting a series of tests enabling the assessment of the migration potential of cervical cancer cells without altered EZR expression and with silenced protein expression. RESULTS Reducing the EZR level resulted in a decrease in the invasive and migration potential of SiHa and HT-3 cells' inhibition of colony formation, a decrease in adhesive properties, and a strong reorganization of F-actin with a dominance of cells with a mitotic catastrophe phenotype. A lower level of protein significantly reduces the motor skills of SiHa and HT-3 cervical cancer cells. CONCLUSIONS This significantly affects the assessment of EZR as a potential factor that can limit the development of metastases in targeted cancer therapy of cervical cancer.
Collapse
Affiliation(s)
- Marta Hałas-Wiśniewska
- Department of Histology and Embryology, Faculty of Medicine, Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Toruń, Karłowicza 24, 85-092 Bydgoszcz, Poland; (W.A.); (A.G.); (M.I.)
| | | | | | | |
Collapse
|
44
|
Choi H, Hwang W. Anesthetic Approaches and Their Impact on Cancer Recurrence and Metastasis: A Comprehensive Review. Cancers (Basel) 2024; 16:4269. [PMID: 39766169 PMCID: PMC11674873 DOI: 10.3390/cancers16244269] [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: 10/21/2024] [Revised: 11/10/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
Cancer recurrence and metastasis remain critical challenges following surgical resection, influenced by complex perioperative mechanisms. This review explores how surgical stress triggers systemic changes, such as neuroendocrine responses, immune suppression, and inflammation, which promote the dissemination of residual cancer cells and circulating tumor cells. Key mechanisms, such as epithelial-mesenchymal transition and angiogenesis, further enhance metastasis, while hypoxia-inducible factors and inflammatory responses create a microenvironment conducive to tumor progression. Anesthetic agents and techniques modulate these mechanisms in distinct ways. Inhaled anesthetics, such as sevoflurane, may suppress immune function by increasing catecholamines and cytokines, thereby promoting cancer progression. In contrast, propofol-based total intravenous anesthesia mitigates stress responses and preserves natural killer cell activity, supporting immune function. Opioids suppress immune surveillance and promote angiogenesis through the activation of the mu-opioid receptor. Opioid-sparing strategies using NSAIDs show potential in preserving immune function and reducing recurrence risk. Regional anesthesia offers benefits by reducing systemic stress and immune suppression, though the clinical outcomes remain inconsistent. Additionally, dexmedetomidine and ketamine exhibit dual effects, both enhancing and inhibiting tumor progression depending on the dosage and context. This review emphasizes the importance of individualized anesthetic strategies to optimize long-term cancer outcomes. While retrospective studies suggest potential benefits of propofol-based total intravenous anesthesia and regional anesthesia, further large-scale trials are essential to establish the definitive role of anesthetic management in cancer recurrence and survival.
Collapse
Affiliation(s)
| | - Wonjung Hwang
- Department of Anesthesiology and Pain Medicine, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea;
| |
Collapse
|
45
|
Liao AQ, Wen J, Wei JC, Xu BB, Jin N, Lin HY, Qin XY. Syntheses, crystal structures of copper (II)-based complexes of sulfonamide derivatives and their anticancer effects through the synergistic effect of anti-angiogenesis, anti-inflammation, pro-apoptosis and cuproptosis. Eur J Med Chem 2024; 280:116954. [PMID: 39406115 DOI: 10.1016/j.ejmech.2024.116954] [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: 04/27/2024] [Revised: 10/07/2024] [Accepted: 10/08/2024] [Indexed: 11/25/2024]
Abstract
Three novel copper(II)-based complexes Cu-1, Cu-2, and Cu-3 containing sulfamethoxazole or sulfamethazine ligand were obtained, and their single structures were characterized. Both Cu-1 and Cu-3 show a broad spectrum of cytotoxicity than Cu-2, and Cu-1 is more cytotoxic than Cu-3. What's interesting is that Cu-1 can exhibit obvious inhibitory effect on the growth of human triple-negative breast cancer in vivo and vitro through anti-proliferative, anti-angiogenic, anti-inflammatory, pro-apoptotic and cuproptotic synergistic effects. Though Cu-3 shows no significant cytotoxicity against MDA-MB-231 cells, it can significantly inhibit the growth of SKOV3 cells in vitro by down-regulating the expression of some key proteins in the VEGF/VEGFR2 signaling pathway and the expression of some pro-inflammatory cytokines, and by disrupting the balance of intracellular reactive oxygen species levels.
Collapse
Affiliation(s)
- Ai-Qiu Liao
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Juan Wen
- Department of Pharmacy, The Affiliated Hospital of Guilin Medical University, Guangxi, Guilin, 541001, China
| | - Jing-Chen Wei
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Bing-Bing Xu
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Nan Jin
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Hong-Yu Lin
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China
| | - Xiu-Ying Qin
- College of Pharmacy, Guilin Medical University, Guangxi, Guilin, 541004, China.
| |
Collapse
|
46
|
Shen H, Zuo F. Prognostic role of systemic inflammation response index (SIRI) in patients with pancreatic cancer: a meta-analysis. Front Oncol 2024; 14:1465279. [PMID: 39723376 PMCID: PMC11668680 DOI: 10.3389/fonc.2024.1465279] [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: 07/16/2024] [Accepted: 11/26/2024] [Indexed: 12/28/2024] Open
Abstract
Background The significance of the systemic inflammation response index (SIRI) in predicting the prognosis of patients with pancreatic cancer (PC) has been extensively explored; however, findings remain controversial. As such, this meta-analysis was performed to more precisely determine the utility of the SIRI in predicting PC prognosis. Methods A comprehensive literature search of the PubMed, Web of Science, Embase, and Cochrane Library databases for relevant studies, published up to June 25, 2024, was performed. The primary and secondary endpoints were overall survival (OS) and progression-free survival (PFS), respectively. The prognostic utility of the SIRI in predicting PC prognosis was estimated by calculating pooled hazard ratios (HRs) and corresponding 95% confidence intervals (CIs). Results Seven studies comprising 1160 patients were included in the present meta-analysis. Pooled findings revealed that elevated SIRI was as a prominent prognostic marker of OS (HR 2.40 [95% CI 1.88-3.05]; p<0.001) and PFS (HR 1.95 [95% CI 1.19-3.21]; p=0.008) in patients diagnosed with PC. According to subgroup analysis, the SIRI remained an outstanding prognostic marker for OS, irrespective of region, sample size, study center, study design, TNM stage, cancer type, cut-off value, treatment, or survival analysis type (all p<0.05). Moreover, based on subgroup analysis, the SIRI demonstrated significant utility in predicting PFS, irrespective of region and threshold value (p<0.05). Conclusion Results of the present meta-analysis revealed that an increased SIRI significantly predicted OS and PFS in patients diagnosed with PC. Considering its cost-effectiveness and availability, the SIRI may be a promising biomarker for predicting prognosis in patients with PC.
Collapse
Affiliation(s)
- Huifen Shen
- Department of Neurology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| | - Fei Zuo
- Department of Gastroenterology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
47
|
Cheung EC, Strathdee D, Stevenson D, Coomes J, Blyth K, Vousden KH. Regulation of ROS signaling by TIGAR induces cancer-modulating responses in the tumor microenvironment. Proc Natl Acad Sci U S A 2024; 121:e2416076121. [PMID: 39636862 DOI: 10.1073/pnas.2416076121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 11/03/2024] [Indexed: 12/07/2024] Open
Abstract
The consequences of reactive oxygen species (ROS) in cancer cells are complex and have been shown to both promote and retard tumorigenesis in different models. In mouse models of pancreatic ductal adenocarcinoma (PDAC), loss of the antioxidant defense gene Tigar results in both a reduction in the development of early pancreatic intraepithelial neoplasia and an increase in invasive and metastatic capacity, accompanied by decreased survival of mice lacking pancreatic TIGAR. We previously demonstrated that increased ROS following loss of TIGAR promotes various cancer cell-intrinsic changes that contribute to metastatic capacity, including epithelial to mesenchymal transition, enhanced migration and invasion, and an increase in ERK signaling. In this study, we show that pancreatic overexpression of TIGAR decreases metastatic capacity and migratory phenotypes in an aggressive model of PDAC, consistent with the concept that dynamic modulation of TIGAR in PDAC contributes to the development and progression of these tumors. Using TIGAR deficient and overexpressing mouse models, we find that the impact of modulation of TIGAR and ROS in PDAC cells also has a profound effect on the normal stromal cells surrounding the tumor. Loss of TIGAR promotes the production of cytokines by cancer cells that induce changes in the surrounding fibroblasts to adopt a tumor-supportive phenotype. Furthermore, these cytokines also attract macrophages that support PDAC dissemination and metastasis. Taken together our work shows that TIGAR-modulated ROS in PDAC can control cell intrinsic and extrinsic changes to impact tumor aggression.
Collapse
Affiliation(s)
- Eric C Cheung
- The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Douglas Strathdee
- Cancer Research UK Scotland Institute Scotland Institute, Glasgow G61 1BD, Scotland
| | - David Stevenson
- Cancer Research UK Scotland Institute Scotland Institute, Glasgow G61 1BD, Scotland
| | - Jack Coomes
- The Francis Crick Institute, London NW1 1AT, United Kingdom
| | - Karen Blyth
- Cancer Research UK Scotland Institute Scotland Institute, Glasgow G61 1BD, Scotland
- School of Cancer Sciences, University of Glasgow, Glasgow G61 1QH, Scotland
| | | |
Collapse
|
48
|
Okamoto Y, Tsumoto K. Cardiac Arrhythmia: Molecular Mechanisms and Therapeutic Strategies. Int J Mol Sci 2024; 25:13253. [PMID: 39769015 PMCID: PMC11676050 DOI: 10.3390/ijms252413253] [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: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
Arrhythmias are divided into supraventricular and ventricular, depending on where they originate [...].
Collapse
Affiliation(s)
- Yosuke Okamoto
- Department of Cell Physiology, Akita University Graduate School of Medicine, 1-1-1, Hondo, Akita 010-8543, Japan
| | - Kunichika Tsumoto
- Department of Physiology II, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa 920-0293, Japan
| |
Collapse
|
49
|
Li S, Lin Y, Gao X, Zeng D, Cen W, Su Y, Su J, Zeng C, Huang Z, Zeng H, Huang S, Tang M, Li X, Luo M, Huang Z, Liang R, Ye J. Integrative multi-omics analysis reveals a novel subtype of hepatocellular carcinoma with biological and clinical relevance. Front Immunol 2024; 15:1517312. [PMID: 39712016 PMCID: PMC11659151 DOI: 10.3389/fimmu.2024.1517312] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a highly heterogeneous tumor, and the development of accurate predictive models for prognosis and drug sensitivity remains challenging. Methods We integrated laboratory data and public cohorts to conduct a multi-omics analysis of HCC, which included bulk RNA sequencing, proteomic analysis, single-cell RNA sequencing (scRNA-seq), spatial transcriptomics sequencing (ST-seq), and genome sequencing. We constructed a tumor purity (TP) and tumor microenvironment (TME) prognostic risk model. Proteomic analysis validated the TP-TME-related signatures. Joint analysis of scRNA-seq and ST-seq revealed characteristic clusters associated with TP high-risk subtypes, and immunohistochemistry confirmed the expression of key genes. We conducted functional enrichment analysis, transcription factor activity inference, cell-cell interaction, drug efficacy analysis, and mutation information analysis to identify a novel subtype of HCC. Results Our analyses constructed a robust HCC prognostic risk prediction model. The patients with TP-TME high-risk subtypes predominantly exhibit hypoxia and activation of the Wnt/beta-catenin, Notch, and TGF-beta signaling pathways. Furthermore, we identified a novel subtype, XPO1+Epithelial. This subtype expresses signatures of the TP risk subtype and aligns with the biological behavior of high-risk patients. Additional analyses revealed that XPO1+Epithelial is influenced primarily by fibroblasts via ligand-receptor interactions, such as FN1-(ITGAV+ITGB1), and constitute a significant component of the TP-TME subtype. Moreover, XPO1+Epithelial interact with monocytes/macrophages, T/NK cells, and endothelial cells through ligand-receptor pairs, including MIF-(CD74+CXCR4), MIF-(CD74+CD44), and VEGFA-VEGFR1R2, respectively, thereby promoting the recruitment of immune-suppressive cells and angiogenesis. The ST-seq cohort treated with Tyrosine Kinase Inhibitors (TKIs) and Programmed Cell Death Protein 1 (PD-1) presented elevated levels of TP and TME risk subtype signature genes, as well as XPO1+Epithelial, T-cell, and endothelial cell infiltration in the treatment response group. Drug sensitivity analyses indicated that TP-TME high-risk subtypes, including sorafenib and pembrolizumab, were associated with sensitivity to multiple drugs. Further exploratory analyses revealed that CTLA4, PDCD1, and the cancer antigens MSLN, MUC1, EPCAM, and PROM1 presented significantly increase expression levels in the high-risk subtype group. Conclusions This study constructed a robust prognostic model for HCC and identified novel subgroups at the single-cell level, potentially assisting in the assessment of prognostic risk for HCC patients and facilitating personalized drug therapy.
Collapse
Affiliation(s)
- Shizhou Li
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Yan Lin
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Xing Gao
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Dandan Zeng
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Weijie Cen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Yuejiao Su
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Jingting Su
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Can Zeng
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Zhenbo Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Haoyu Zeng
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Shilin Huang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Minchao Tang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Xiaoqing Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Min Luo
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Zhihu Huang
- Department of Clinical Laboratory, Minzu Hospital Guangxi Zhuang Autonomous Region, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, Guangxi, ;China
| | - Rong Liang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Jiazhou Ye
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| |
Collapse
|
50
|
Gong J, Lu J, Zhang W, Huang W, Li J, Yang Z, Meng F, Sun H, Zhao L. A CT-based subregional radiomics nomogram for predicting local recurrence-free survival in esophageal squamous cell cancer patients treated by definitive chemoradiotherapy: a multicenter study. J Transl Med 2024; 22:1108. [PMID: 39639328 PMCID: PMC11619118 DOI: 10.1186/s12967-024-05897-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: 08/27/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND To develop and validate an online individualized model for predicting local recurrence-free survival (LRFS) in esophageal squamous cell carcinoma (ESCC) treated by definitive chemoradiotherapy (dCRT). METHODS ESCC patients from three hospitals were randomly stratified into the training set (715) and the internal testing set (179), and patients from the other hospital as the external testing set (120). The important radiomic features extracted from contrast-enhanced computed tomography (CECT)-based subregions clustered from the whole volume of tumor and peritumor were selected and used to construct the subregion-based radiomic signature by using COX proportional hazards model, which was compared with the tumor-based radiomic signature. The clinical model and the radiomics model combing the clinical factors and the radiomic signature were further constructed and compared, which were validated in two testing sets. RESULTS The subresion-based radiomic signature showed better prognostic performance than the tumor-based radiomic signature (training: 0.642 vs. 0.621, internal testing: 0.657 vs. 0.638, external testing: 0.636 vs. 0.612). Although the tumor-based radiomic signature, the subregion-based radiomic signature, the tumor-based radiomics model, and the subregion-based radiomics model had better performance compared to the clinical model, only the subregion-based radiomics model showed a significant advantage (p < 0.05; training: 0.666 vs. 0.616, internal testing: 0.689 vs. 0.649, external testing: 0.642 vs. 0.604). The clinical model and the subregion-based radiomics model were visualized as the nomograms, which are available online and could interactively calculate LRFS probability. CONCLUSIONS We established and validated a CECT-based online radiomics nomogram for predicting LRFS in ESCC received dCRT, which outperformed the clinical model and might serve as a powerful tool to facilitate individualized treatment. TRIAL REGISTRATION This retrospective study was approved by the ethics committee (KY20222145-C-1).
Collapse
Affiliation(s)
- Jie Gong
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, China
| | - Jianchao Lu
- Department of Radiation Oncology, Sichuan Cancer Center, School of Medicine, Sichuan Cancer Hospital and Institution, University of Electronic Science and Technology of China, Chengdu, China
| | - Wencheng Zhang
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Wei Huang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, China
| | - Jie Li
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, China
| | - Zhi Yang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, China
| | - Fan Meng
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, China
| | - Hongfei Sun
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, China
| | - Lina Zhao
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, 127 West Changle Road, Xi'an, China.
| |
Collapse
|