1
|
Zhang Y, Han C, Li K, Liu T, Cai H, Xu D, Cui W. Single circulating tumor cell sequencing based on improved high-porosity membranes and nanoporous microchambers. Biosens Bioelectron 2025; 276:117263. [PMID: 39978235 DOI: 10.1016/j.bios.2025.117263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/10/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
Circulating tumor cells (CTCs) are crucial for understanding tumor heterogeneity and progression. Despite extensive research over the years, most studies have focused on CTCs counting, with fewer efforts directed toward single-cell sequencing (SCS) of CTCs. In this study, we developed two novel nanodevices---a high-porosity ultrathin filter membrane and a nanowell chip--- to isolate single CTCs. Automated scanning and single-cell picking systems were employed to locate and isolate individual CTCs, enabling the establishment of an efficient and automated workflow for single CTC sequencing using filter-based systems. We conducted an in-depth comparison to evaluate the effects of different filter membranes and cell adhesion types on genomic integrity, cell viability, sequencing coverage, and depth. The results showed that the high-porosity filter membrane outperformed other photolithographic filters for SCS of CTCs. Validation using NCI-H358 cell lines and patient-derived CTCs demonstrated that this workflow could accurately and comprehensively detect gene mutations, amplifications, and copy number variations (CNVs). CNV profiles of CTCs from patients with the same tumor type were highly consistent, while intra-patient CTCs revealed significant heterogeneity. Furthermore, we identified and overcame challenges related to cell adhesion to the filter membrane and the impact of cell viability on sequencing outcomes during CTC enrichment. This workflow offers new insights into the development of CTC-based approaches for exploring tumor progression, heterogeneity, and mechanisms of drug resistance.
Collapse
Affiliation(s)
- Yujuan Zhang
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Han
- Anfang Biotech Co., Ltd., Guangzhou Life Science Center, Bio-Island, Guangzhou, China
| | - Kexin Li
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tiejun Liu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongqing Cai
- Department of Neurosurgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Danfei Xu
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Cui
- Department of Clinical Laboratory, State Key Laboratory of Molecular Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
2
|
Mumme HL, Huang C, Ohlstrom D, Bakhtiari M, Raikar SS, DeRyckere D, Qayed M, Castellino SM, Wechsler DS, Porter CC, Graham DK, Bhasin SS, Bhasin M. Identification of leukemia-enriched signature through the development of a comprehensive pediatric single-cell atlas. Nat Commun 2025; 16:4114. [PMID: 40316535 PMCID: PMC12048633 DOI: 10.1038/s41467-025-59362-5] [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: 04/30/2024] [Accepted: 04/21/2025] [Indexed: 05/04/2025] Open
Abstract
Single-cell transcriptome profiling enables unparalleled characterization of the heterogeneous microenvironment of pediatric leukemias. To facilitate comparative analyses and generate pediatric leukemia signatures, we collect, process, and annotate single-cell data comprising over 540,000 cells from 159 different pediatric acute leukemia (myeloid, lymphoid, mixed phenotype lineages) and healthy bone marrow (BM) samples, profiled in our lab and curated from publicly available studies. The analysis identifies a leukemia-enriched signature of nine genes with over-expression in leukemic blast compared to healthy BM cells. This signature is also consistently over-expressed in leukemia samples compared to normal BM in bulk RNA-seq datasets (over 2000 samples). Outcome-based analysis on diagnosis samples using measurable residual disease (MRD) status depicts a significant association of oncogene-induced senescence and g-protein activation pathways with MRD positivity. MRD positivity across pediatric leukemias is also correlated with significant depletion of CD8+ and CD4+ naïve T-cells and M1-macrophages at diagnosis. To enable easy access to this comprehensive pediatric leukemia single-cell atlas, we develop the Pediatric Single-cell Cancer Atlas (PedSCAtlas, https://bhasinlab.bmi.emory.edu/PediatricSCAtlas/ ). The atlas allows for quick exploration of single-cell data based on genes, cell type composition, and clinical outcomes to understand the cellular landscape of pediatric leukemias.
Collapse
Affiliation(s)
- Hope L Mumme
- Department of Biomedical Informatics, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Chenbin Huang
- Department of Biomedical Informatics, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Denis Ohlstrom
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Mojtaba Bakhtiari
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Sunil S Raikar
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Deborah DeRyckere
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Muna Qayed
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Sharon M Castellino
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Daniel S Wechsler
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Christopher C Porter
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Douglas K Graham
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Swati S Bhasin
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA
| | - Manoj Bhasin
- Department of Biomedical Informatics, Emory University, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
- Coulter Department of Biomedical Engineering, Georgia Institute of Technology, 313 Ferst Dr NW, Atlanta, GA, 30332, USA.
- Department of Pediatrics, Emory University School of Medicine, 2015 Uppergate Dr, Atlanta, GA, 30322, USA.
- Aflac Cancer and Blood Disorders Center, Children Healthcare of Atlanta, 2220 N Druid Hills Rd NE, Atlanta, GA, 30329, USA.
| |
Collapse
|
3
|
Yang C, Li Y, Wang Z, Shan H, Zhang G, Meng X, Wang G, Hou Z, Zhao X, Zhang X, Liu A, Bing Y, Lei G, Jin Y, Luo J, Guo L, Yin Y. Identification of a cancer stem cell-like subpopulation that promotes HCC metastasis. JHEP Rep 2025; 7:101302. [PMID: 40242316 PMCID: PMC11999271 DOI: 10.1016/j.jhepr.2024.101302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Revised: 12/05/2024] [Accepted: 12/10/2024] [Indexed: 04/18/2025] Open
Abstract
Background & Aims Cancer stem cells (CSCs) are well-established drivers of tumorigenesis, but their role in regulating tumor metastasis remains poorly understood. Here, we report the identification and characterization of a cluster of metastasis-promoting CSC-like cells in hepatocellular carcinoma (HCC). Methods CSC-like cells in HCC were identified through the analysis of single cell RNA-sequencing data from 19 HCC samples. The stemness and invasive characteristics of these cells were evaluated using bioinformatical analyses of nine clinical cohorts and experimental validations. Spatial transcriptomics sequencing of 12 HCC samples revealed the cellular interactions between the CSC-like cells and tumor microenvironments, which were validated through gene co-expression analyses and immunohistochemistry. Finally, signaling pathway blockade was used to assess the potential clinical application of CSC-like cells. Results Through comprehensive analyses of single cell RNA-sequencing data from 19 patients with HCC and spatial transcriptomics data from 12 patients with HCC, a metastasis-promoting CSC-like subpopulation was identified. These CSC-like cells expressed high levels of epithelial-mesenchymal transition genes and were associated with poor prognosis of HCC. Histologically, CSC-like cells were enriched in highly aggressive tumors, especially in intrahepatic disseminated foci, where they interacted with immune cells. Functionally, CSC-like cells induced macrophage M2 polarization and T cell exhaustion through the ICAM1 signaling pathway, forming immunosuppressive microenvironments. Downregulation of ICAM1 expression in CSC-like cells suppressed macrophage M2-polarization and T cell exhaustion, thereby reversing antitumor immune effects. Conclusions Our study identified a metastasis-promoting CSC subpopulation, providing a potential perspective for CSC-targeted therapies in HCC. Impact and implications The heterogeneity of CSCs in HCC has been identified, yet the identification and characterization of metastasis-promoting CSC subpopulations remain unexplored. Here, we identified a CSC-like tumor cell subpopulation that promotes HCC metastasis by increasing cell invasiveness and suppressing antitumor immune responses via the ICAM1 signaling pathway. Our study uncovers novel mechanisms of HCC metastasis from the perspective of CSCs, and proposes potential tumor therapeutic strategies by inhibiting cellular interactions between CSC-like cells and immune cells.
Collapse
Affiliation(s)
- Chunyuan Yang
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yang Li
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Zhaohai Wang
- Faculty of Hepato-Pancreato-Biliary Surgery, Chinese PLA General Hospital, Beijing 100853, China
| | - Hui Shan
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Guangze Zhang
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xiangyan Meng
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Guangxi Wang
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Zhiyuan Hou
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xuyang Zhao
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Xin Zhang
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Anhang Liu
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yuntao Bing
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Guanglin Lei
- Senior Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital, 100039 Beijing, China
| | - Yan Jin
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Jianyuan Luo
- Department of Medical Genetics, School of Basic Medical Sciences Peking University, Beijing 100191, China
| | - Limei Guo
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
| | - Yuxin Yin
- Institute of Systems Biomedicine, Department of Pathology, Center of Basic Medical Research, Institute of Medical Innovation and Research, School of Basic Medical Sciences, Peking-Tsinghua Center for Life Sciences, Peking University Third Hospital, Peking University, Beijing 100191, China
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| |
Collapse
|
4
|
Xu Q, Wang Y, Dou S, Xu Y, Xu Z, Xu H, Zhang Y, Xia Y, Xue Y, Li H, Ma X, Zhang K, Wang H, Ma F, Wang Q, Li B, Wang W. High-Viability Circulating Tumor Cells Sorting From Whole Blood at Single Cell Level Using Laser-Induced Forward Transfer-Assisted Microfiltration. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2414195. [PMID: 39868845 PMCID: PMC12079465 DOI: 10.1002/advs.202414195] [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: 11/02/2024] [Revised: 01/20/2025] [Indexed: 01/28/2025]
Abstract
The efficient isolation and molecular analysis of circulating tumor cells (CTCs) from whole blood at single-cell level are crucial for understanding tumor metastasis and developing personalized treatments. The viability of isolated cells is the key prerequisite for the downstream molecular analysis, especially for RNA sequencing. This study develops a laser-induced forward transfer -assisted microfiltration system (LIFT-AMFS) for high-viability CTC enrichment and retrieval from whole blood. The LIFT-compatible double-stepped microfilter (DSMF), central to this system, comprises two micropore layers: the lower layer's smaller micropores facilitate size-based cell separation, and the upper layer's larger micropores enable liquid encapsulating captured cells. By optimizing the design of the DSMFs, the system has a capture efficiency of 88% at the processing throughput of up to 15.0 mL min-1 during the microfilter-based size screening stage, with a single-cell yield of over 95% during the retrieval stage. The retrieved single cells, with high viability, are qualified for ex vivo culture and direct RNA sequencing. The cDNA yield from isolated CTCs surpassed 4.5 ng, sufficient for library construction. All single-cell sequencing data exhibited Q30 scores above 95.92%. The LIFT-AMFS shows promise in cellular and biomedical research.
Collapse
Affiliation(s)
- Qingmei Xu
- School of Integrated CircuitsPeking UniversityBeijing100871China
- Department of Electrical EngineeringTaiyuan Institute of TechnologyTaiyuan030008China
| | - Yuntong Wang
- Changchun Institute of OpticsFine Mechanics and PhysicsChinese Academy of SciencesChangchun130033China
- University of Chinese Academy of SciencesBeijing100049China
| | - Songtao Dou
- School of Integrated CircuitsPeking UniversityBeijing100871China
| | - Yang Xu
- Cancer Translational Medicine Research CenterThe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
- Department of Respiratory MedicineThe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
| | - Zhenhe Xu
- State Key Laboratory of Bioreactor EngineeringShanghai Frontiers Science Center of Optogenetic Techniques for Cell MetabolismEast China University of Science and TechnologyShanghai200237China
| | - Han Xu
- School of Integrated CircuitsPeking UniversityBeijing100871China
| | - Yi Zhang
- School of Integrated CircuitsPeking UniversityBeijing100871China
| | - Yanming Xia
- Guangzhou National LaboratoryGuangzhou510320China
| | - Ying Xue
- Hooke LaboratoryChangchun130033China
| | - Hang Li
- Hooke LaboratoryChangchun130033China
| | - Xiao Ma
- Hangzhou Branemagic Medical Technology Co. Ltd.Hangzhou310021China
| | | | - Huan Wang
- Changchun Institute of OpticsFine Mechanics and PhysicsChinese Academy of SciencesChangchun130033China
- University of Chinese Academy of SciencesBeijing100049China
| | - Fengzhou Ma
- School of Integrated CircuitsPeking UniversityBeijing100871China
- Hangzhou Branemagic Medical Technology Co. Ltd.Hangzhou310021China
| | - Qi Wang
- Cancer Translational Medicine Research CenterThe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
- Department of Respiratory MedicineThe Second Affiliated Hospital of Dalian Medical UniversityDalian116027China
| | - Bei Li
- Changchun Institute of OpticsFine Mechanics and PhysicsChinese Academy of SciencesChangchun130033China
- University of Chinese Academy of SciencesBeijing100049China
| | - Wei Wang
- School of Integrated CircuitsPeking UniversityBeijing100871China
- National Key Laboratory of Advanced Micro and Nano Manufacture TechnologyBeijing100871China
- Beijing Advanced Innovation Center for Integrated CircuitsBeijing100871China
| |
Collapse
|
5
|
Zhang G, Zhang L, Feng Q, Ma P, Zheng C, Wang L, Xue Q, Li Y. Outcomes of Intraoperative Radiotherapy for Locally Advanced Adenocarcinoma of the Esophagogastric Junction After Neoadjuvant Therapy: A Single-Arm, Phase 1 Trial From the Chinese National Cancer Center. Ann Surg Oncol 2025; 32:3138-3146. [PMID: 39648241 DOI: 10.1245/s10434-024-16620-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: 08/20/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024]
Abstract
BACKGROUND The role of intraoperative radiotherapy (IORT) for adenocarcinoma of the esophagogastric junction (AEG) remains uncertain. Therefore, a prospective phase 1 trial was conducted to assess the safety and feasibility of IORT for locally advanced AEG. METHODS The study enrolled patients with AEG at stages II-IVA from January 2019 to September 2019. Eligible patients received esophagectomy and a single fraction of electron beam radiotherapy. The primary endpoint of the study was a safety profile for IORT. Additionally, survival outcomes and the locoregional recurrence rate (LRR) were compared between the non-IORT and IORT cohorts using propensity score-matching. RESULTS For 15 (93.8 %) of the 16 patients in the study, R0 resection was successfully achieved, with only one patient undergoing R1 resection. A total postoperative complication morbidity rate of 43.8 % (7/16) was observed, with major complications (Clavien-Dindo classification ≥3) in 12.5 % of the cases (2/16). Total treatment-related adverse events were reported for seven patients (43.8 %, 7/16). After matching, a lower LRR was observed in the IORT group than in the non-IORT group (0 % [0/12] vs 33.3 % [4/12]; p = 0.028). However, the two groups did not differ significantly in 3-year progression-free survival (PFS: IORT [50.9 %] vs non-IORT [53.4 %]; p = 0.93) or 3-year overall survival (OS: IORT [58.3 %] vs IORT [72.9 %]; p = 0.23). CONCLUSIONS The current study demonstrated favorable feasibility and safety of IORT for locally advanced AEG. Although IORT is beneficial for improving local control, it may not prolong PFS or OS for patients with locally advanced AEG. A phase 2 trial is warranted for further validation of these outcomes.
Collapse
Affiliation(s)
- Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Long Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qinfu Feng
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Pan Ma
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Chao Zheng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lide Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Yong Li
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
6
|
Wang PX, Zhong YC, Duan B, Cheng JW, Sun YF, Zheng WJ, Zhou KQ, Xu Y, Peng HX, Jin WX, Li HM, Sun XJ, Guo W, Zhou J, Liu Q, Fan J, Yang XR. Exploring morphological heterogeneity of circulating tumor cells: machine learning-based approach for cell identification and prognostic implications. Sci Bull (Beijing) 2025:S2095-9273(25)00433-5. [PMID: 40348669 DOI: 10.1016/j.scib.2025.04.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 12/28/2024] [Accepted: 03/31/2025] [Indexed: 05/14/2025]
Affiliation(s)
- Peng-Xiang Wang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Yu-Chen Zhong
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Bin Duan
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China; Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Jian-Wen Cheng
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Yun-Fan Sun
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Wen-Jing Zheng
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Kai-Qian Zhou
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Yang Xu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | | | | | - Hai-Min Li
- Dunwill Med-Tech, Shanghai 200032, China
| | | | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Zhou
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Qi Liu
- Translational Medical Center for Stem Cell Therapy and Institute for Regenerative Medicine, Shanghai East Hospital, Bioinformatics Department, School of Life Sciences and Technology, Tongji University, Shanghai 200092, China.
| | - Jia Fan
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.
| | - Xin-Rong Yang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.
| |
Collapse
|
7
|
Tang R, Luo S, Liu H, Sun Y, Liu M, Li L, Ren H, Angele MK, Börner N, Yu K, Guo Z, Yin G, Luo H. Circulating Tumor Microenvironment in Metastasis. Cancer Res 2025; 85:1354-1367. [PMID: 39992721 PMCID: PMC11997552 DOI: 10.1158/0008-5472.can-24-1241] [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: 04/15/2024] [Revised: 11/12/2024] [Accepted: 02/19/2025] [Indexed: 02/26/2025]
Abstract
Activation of invasion and metastasis is a central hallmark of cancer, contributing to the primary cause of death for patients with cancer. In the multistep metastatic process, cancer cells must infiltrate the circulation, survive, arrest at capillary beds, extravasate, and form metastatic clones in distant organs. However, only a small proportion of circulating tumor cells (CTC) successfully form metastases, with transit of CTCs in the circulation being the rate-limiting step. The fate of CTCs is influenced by the circulating tumor microenvironment (cTME), which encompasses factors affecting their biological behaviors in the circulation. This liquid and flowing microenvironment differs significantly from the primary TME or the premetastatic niche. This review summarizes the latest advancements in identifying the biophysical cues, key components, and biological roles of the cTME, highlighting the network among biophysical attributes, blood cells, and nonblood factors in cancer metastasis. In addition to the potential of the cTME as a therapeutic target for inhibiting metastasis, the cTME could also represent as a biomarker for predicting patient outcomes and developing strategies for treating cancer.
Collapse
Affiliation(s)
- Rui Tang
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Shujuan Luo
- Department of Obstetrics, Women and Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Hui Liu
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Yan Sun
- Department of Cell Biology and Medical Genetics, Basic Medical School, Chongqing Medical University, Chongqing, China
| | - Manran Liu
- Key Laboratory of Laboratory Medical Diagnostics, Chinese Ministry of Education, Chongqing Medical University, Chongqing, China
| | - Lu Li
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haoyu Ren
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Martin K. Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Nikolaus Börner
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich Munich, Germany
| | - Keda Yu
- Department of Breast Surgery, Precision Cancer Medicine Center, Fudan University Shanghai Cancer Center, Shanghai, P.R. China
| | - Zufeng Guo
- Center for Novel Target and Therapeutic Intervention, Institute of Life Sciences, Chongqing Medical University, Chongqing, China
| | - Guobing Yin
- Department of Thyroid and Breast Surgery, The Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Haojun Luo
- Department of Thyroid and Breast Surgery, Renji Hospital, School of Medicine, Chongqing University, Chongqing, China
| |
Collapse
|
8
|
Xu C, Du D, Han Z, Si H, Li W, Li L, Tang B. Separation and Analysis of Rare Tumor Cells in Various Body Fluids Based on Microfluidic Technology for Clinical Applications. Anal Chem 2025; 97:7567-7588. [PMID: 40186540 DOI: 10.1021/acs.analchem.4c06925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/07/2025]
Affiliation(s)
- Chang Xu
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Dexin Du
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Zhaojun Han
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Haibin Si
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Wei Li
- Department of Hematology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan 250012, P. R. China
| | - Lu Li
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
| | - Bo Tang
- College of Chemistry, Chemical Engineering and Materials Science, Collaborative Innovation Center of Functionalized Probes for Chemical Imaging, Key Laboratory of Molecular and Nano Probes, Ministry of Education, Shandong Normal University, Jinan 250014, P. R. China
- Laoshan Laboratory, Qingdao 266237, P. R. China
| |
Collapse
|
9
|
Masmoudi D, Villalba M, Alix-Panabières C. Natural killer cells: the immune frontline against circulating tumor cells. J Exp Clin Cancer Res 2025; 44:118. [PMID: 40211394 PMCID: PMC11983744 DOI: 10.1186/s13046-025-03375-x] [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: 01/10/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Natural killer (NK) play a key role in controlling tumor dissemination by mediating cytotoxicity towards cancer cells without the need of education. These cells are pivotal in eliminating circulating tumor cells (CTCs) from the bloodstream, thus limiting cancer spread and metastasis. However, aggressive CTCs can evade NK cell surveillance, facilitating tumor growth at distant sites. In this review, we first discuss the biology of NK cells, focusing on their functions within the tumor microenvironment (TME), the lymphatic system, and circulation. We then examine the immune evasion mechanisms employed by cancer cells to inhibit NK cell activity, including the upregulation of inhibitory receptors. Finally, we explore the clinical implications of monitoring circulating biomarkers, such as NK cells and CTCs, for therapeutic decision-making and emphasize the need to enhance NK cell-based therapies by overcoming immune escape mechanisms.
Collapse
Affiliation(s)
- Doryan Masmoudi
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France
| | - Martin Villalba
- IRMB, Univ Montpellier, INSERM, CHU Montpellier, CNRS, Montpellier, France
| | - Catherine Alix-Panabières
- Laboratory of Rare Circulating Human Cells, University Medical Center of Montpellier, Montpellier, France.
- CREEC/CANECEV, MIVEGEC (CREES), University of Montpellier, CNRS, Montpellier, IRD, France.
- European Liquid Biopsy Society (ELBS), Hamburg, Germany.
- LCCRH, Site Unique de Biologie (SUB), 641, Avenue du Doyen Gaston Giraud, Montpellier, 34093, France.
| |
Collapse
|
10
|
Yadav P, Rajendrasozhan S, Lajimi RH, Patel RR, Heymann D, Prasad NR. Circulating tumor cell markers for early detection and drug resistance assessment through liquid biopsy. Front Oncol 2025; 15:1494723. [PMID: 40260304 PMCID: PMC12009936 DOI: 10.3389/fonc.2025.1494723] [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: 09/11/2024] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Circulating tumor cells (CTCs) are cancerous cells that extravasate from the primary tumor or metastatic foci and travel through the bloodstream to distant organs. CTCs provide crucial insights into cancer metastasis, the evolution of tumor genotypes during treatment, and the development of chemo- and/or radio-resistance during disease progression. The process of Epithelial-to-mesenchymal transition (EMT) plays a key role in CTCs formation, as this process enhances cell's migration properties and is often associated with increased invasiveness thereby leading to chemotherapy resistance. During the EMT process, tumor cells lose epithelial markers like EpCAM and acquire mesenchymal markers such as vimentin driven by transcription factors like Snail and Twist. CTCs are typically identified using specific cell surface markers, which vary depending on the cancer type. Common markers include EpCAM, used for epithelial cancers; CD44 and CD24, which are associated with cancer stem cells; and cytokeratins, such as CK8 and CK18. Other markers like HER2/neu and vimentin can also be used to target CTCs in specific cancer types and stages. Commonly, immune-based isolation techniques are being implemented for the isolation and enrichment of CTCs. This review emphasizes the clinical relevance of CTCs, particularly in understanding drug resistance mechanisms, and underscores the importance of EMT-derived CTCs in multidrug resistance (MDR). Moreover, the review also discusses CTCs-specific surface markers that are crucial for their isolation and enrichment. Ultimately, the EMT-specific markers found in CTCs could provide significant information to halt the disease progression and enable personalized therapies.
Collapse
Affiliation(s)
- Priya Yadav
- Department of Biochemistry and Biotechnology, Annamalai University, Chidambaram, Tamil Nadu, India
| | - Saravanan Rajendrasozhan
- Department of Chemistry, College of Science, University of Ha’il, Ha’il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha’il, Ha’il, Saudi Arabia
| | - Ramzi Hadj Lajimi
- Department of Chemistry, College of Science, University of Ha’il, Ha’il, Saudi Arabia
- Medical and Diagnostic Research Centre, University of Ha’il, Ha’il, Saudi Arabia
| | - Raja Ramadevi Patel
- Medical and Diagnostic Research Centre, University of Ha’il, Ha’il, Saudi Arabia
- Department of Biology, College of Science, University of Ha’il, Ha’il, Saudi Arabia
| | - Dominique Heymann
- Nantes Université, CNRS, US2B, UMR 6286, Nantes, France
- Institut de Cancérologie de l’Ouest, Tumor Heterogeneity and Precision Medecine Laboratory, Saint-Herblain, France
- Medical School, University of Sheffield, Sheffield, United Kingdom
| | - N. Rajendra Prasad
- Department of Biochemistry and Biotechnology, Annamalai University, Chidambaram, Tamil Nadu, India
| |
Collapse
|
11
|
Hou Y, Lin J, Yao H, Wu Z, Lin Y, Lin J. Linking Metastatic Behavior and Metabolic Heterogeneity of Circulating Tumor Cells at Single-Cell Level Using an Integrative Microfluidic System. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413978. [PMID: 39960842 PMCID: PMC11984876 DOI: 10.1002/advs.202413978] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/28/2024] [Indexed: 04/12/2025]
Abstract
Circulating tumor cells (CTCs) are pivotal biomarkers in tumor metastasis, however, the underlying molecular mechanism of CTCs behavioral heterogeneity during metastasis remains unexplored. Here, an integrative workflow is developed to link behavior characteristics to metabolic profiling within individual CTCs, which simulates the metastatic process on a microfluidic system and combined with single-cell mass spectrometry (MS) detection. Spheroid-derived HCT116 cells are tracked and extracted via a temporary vascular system, revealing various arrest patterns under biomimetic vascular shear flow. Downstream MS analysis characterizes 17 cellular metabolites and associates metabolic profiles with de-adhesion behaviors of the same CTCs, identifying a potential high-metastatic subpopulation with enhanced arrest ability and evaluating critical metabolites involved in metastasis pathways. Additionally, the metastasis-inhibiting effect of anti-tumor drug 5-fluorouracil by reducing high-metastatic cells in spheroids is elucidated. This approach offers a valuable opportunity to dissect the interplay of the metastatic behavior and metabolic profiles of CTCs and foster insights into the molecular mechanisms underlying behavioral phenotypes in the tumor metastasis process.
Collapse
Affiliation(s)
- Ying Hou
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Jiaxu Lin
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Hongren Yao
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Zengnan Wu
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Yongning Lin
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| | - Jin‐Ming Lin
- Department of ChemistryBeijing Key Laboratory of Microanalytical Methods and InstrumentationKey Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology (Ministry of Education)Tsinghua UniversityBeijing100084China
| |
Collapse
|
12
|
Xu Z, Huang W, Zou X, Liu S. Integrated machine learning constructed a circadian-rhythm-related model to assess clinical outcomes and therapeutic advantages in hepatocellular carcinoma. Transl Cancer Res 2025; 14:1799-1823. [PMID: 40224982 PMCID: PMC11985180 DOI: 10.21037/tcr-24-1155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 01/18/2025] [Indexed: 04/15/2025]
Abstract
Background Circadian rhythm (CR) coordinates a variety of internal biological processes with the external daily cycles of light and dark. However, the implications of CR-related regulator in hepatocellular carcinoma (HCC) are quite obscure. Here, we aimed to identify pivotal CR-related markers in HCC for predicting survival and treatment outcomes. Methods The prognostic value of CR regulators in HCC was analyzed. Multi-step machine learning feature selection approaches were employed to establish a model. Thereafter, we evaluated its capacity of clinical prediction and treatment guidance. Results First, we depicted the prognostic stratification value of CR regulators in HCC. Two CR-related phenotypes were identified, revealing a distinct clinical outcome, biological pathways and drug sensitivity. Subsequently, via four topological approaches and differentially expressed genes (DEGs) from real-world cohorts, we screened out CRY2 as the pivotal CR regulator with significant prognostic value in HCC. We performed the relevant basic assay validation for CRY2. Overexpression of CRY2 inhibited the proliferation and migration abilities of Huh7 and Hep3B cells. Moreover, three machine learning algorithms [random forest (RF), extreme gradient boosting (XGBoost) and least absolute shrinkage and selection operator (LASSO)] were implemented to construct a risk-scoring model named CR predictor, which exhibited clinical benefits and therapeutic advantages for HCC. An online nomogram based on CR predictor was developed for predicting individualized survival (https://lihc.shinyapps.io/CR_predictor/). Finally, Mendelian randomization (MR) was performed. Among model genes in CR predictor, PPARGC1A revealed a significant causal effect on HCC. Conclusions We proposed a CR-related risk classifier in HCC, to predict patients' overall survival (OS) and therapeutic response. Targeting CR could be a promising treatment modality against HCC.
Collapse
Affiliation(s)
- Ziyuan Xu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Wei Huang
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xi Zou
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Shenlin Liu
- Department of Oncology, Affiliated Hospital of Nanjing University of Chinese Medicine, Nanjing, China
- No. 1 Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
13
|
Ma L, Luan Y, Lu L. Analyze the Diversity and Function of Immune Cells in the Tumor Microenvironment From the Perspective of Single-Cell RNA Sequencing. Cancer Med 2025; 14:e70622. [PMID: 40062730 PMCID: PMC11891933 DOI: 10.1002/cam4.70622] [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: 08/29/2024] [Revised: 12/14/2024] [Accepted: 01/09/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND Cancer development is closely associated with complex alterations in the tumor microenvironment (TME). Among these, immune cells within the TME play a huge role in personalized tumor diagnosis and treatment. OBJECTIVES This review aims to summarize the diversity of immune cells in the TME, their impact on patient prognosis and treatment response, and the contributions of single-cell RNA sequencing (scRNA-seq) in understanding their functional heterogeneity. METHODS We analyzed recent studies utilizing scRNA-seq to investigate immune cell populations in the TME, focusing on their interactions and regulatory mechanisms. RESULTS ScRNA-seq reveals the functional heterogeneity of immune cells, enhances our understanding of their role in tumor antibody responses, and facilitates the construction of immune cell interaction networks. These insights provide guidance for the development of cancer immunotherapies and personalized treatment approaches. CONCLUSION Applying scRNA-seq to immune cell analysis in the TME offers a novel pathway for personalized cancer treatment. Despite its promise, several challenges remain, highlighting the need for further advancements to fully integrate scRNA-seq into clinical applications.
Collapse
Affiliation(s)
- Lujuan Ma
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Yu Luan
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| | - Lin Lu
- Department of Medical Oncology, Guangzhou First People's Hospital, School of MedicineSouth China University of TechnologyGuangzhouGuangdongChina
| |
Collapse
|
14
|
Sun Y, Li T, Ding L, Wang J, Chen C, Liu T, Liu Y, Li Q, Wang C, Huo R, Wang H, Tian T, Zhang C, Pan B, Zhou J, Fan J, Yang X, Yang W, Wang B, Guo W. Platelet-mediated circulating tumor cell evasion from natural killer cell killing through immune checkpoint CD155-TIGIT. Hepatology 2025; 81:791-807. [PMID: 38779918 DOI: 10.1097/hep.0000000000000934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND AIMS Circulating tumor cells (CTCs) are precursors of cancer metastasis. However, how CTCs evade immunosurveillance during hematogenous dissemination remains unclear. APPROACH AND RESULTS We identified CTC-platelet adhesions by single-cell RNA sequencing and multiplex immunofluorescence of blood samples from multiple cancer types. Clinically, CTC-platelet aggregates were associated with significantly shorter progression-free survival and overall survival in patients with HCC. In vitro, ex vivo, and in vivo assays demonstrated direct platelet adhesions gifted cancer cells with an evasive ability from NK cell killing by upregulating inhibitory checkpoint CD155 (PVR cell adhesion molecule), therefore facilitating distant metastasis. Mechanistically, CD155 was transcriptionally regulated by the FAK/JNK/c-Jun cascade in a platelet contact-dependent manner. Further competition assays and cytotoxicity experiments revealed that CD155 on CTCs inhibited NK-cell cytotoxicity only by engaging with immune receptor TIGIT, but not CD96 and DNAM1, another 2 receptors for CD155. Interrupting the CD155-TIGIT interactions with a TIGIT antibody restored NK-cell immunosurveillance on CTCs and markedly attenuated tumor metastasis. CONCLUSIONS Our results demonstrated CTC evasion from NK-cell-mediated innate immunosurveillance mainly through immune checkpoint CD155-TIGIT, potentially offering an immunotherapeutic strategy for eradicating CTCs.
Collapse
Affiliation(s)
- Yunfan Sun
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Tong Li
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Ding
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiyan Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Chen
- Department of Cell Biology, Shanghai Dunwill Medical Technology Company, Shanghai, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Liu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian Li
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuyu Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ran Huo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tongtong Tian
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Baishen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xinrong Yang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
- Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
15
|
Xu K, Zhang H, Dai H, Mao W. Machine learning and multi-omics characterization of SLC2A1 as a prognostic factor in hepatocellular carcinoma: SLC2A1 is a prognostic factor in HCC. Gene 2025; 938:149178. [PMID: 39681148 DOI: 10.1016/j.gene.2024.149178] [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: 07/31/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by high incidence, significant mortality, and marked heterogeneity, making accurate molecular subtyping essential for effective treatment. Using multi-omics data from HCC patients, we applied diverse clustering algorithms to identify three HCC subtypes (HSs) with distinct prognostic characteristics. Among these, HS1 emerged as an immune-compromised subtype associated with the poorest prognosis. Additionally, we developed a novel, robust, and highly accurate machine learning-guided prognostic signature (MLPS) by integrating multiple machine learning algorithms and their combinations. Our study also identified SLC2A1, the core gene of MLPS, as being highly expressed during advanced stages of tumor progression. Knockdown experiments demonstrated that reducing SLC2A1 expression significantly suppressed the malignant behavior of HCC cells. Furthermore, SLC2A1 expression was linked to responsiveness to dasatinib and vincristine, suggesting potential therapeutic relevance. MLPS and SLC2A1 offer promising tools for individualized prognosis prediction and targeted therapy in HCC, providing new opportunities to improve patient outcomes.
Collapse
Affiliation(s)
- Kangjie Xu
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China; Binhai County People's Hospital, Jiangsu Province, Yancheng 224000, PR China
| | - Houliang Zhang
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China
| | - Hua Dai
- Yangzhou University Clinical Medical College, Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Jiangsu Province, Yangzhou 225009, PR China.
| | - Weipu Mao
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China; Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
16
|
Park J, Lee YT, Agopian VG, Liu JS, Koltsova EK, You S, Zhu Y, Tseng HR, Yang JD. Liquid biopsy in hepatocellular carcinoma: Challenges, advances, and clinical implications. Clin Mol Hepatol 2025; 31:S255-S284. [PMID: 39604328 PMCID: PMC11925447 DOI: 10.3350/cmh.2024.0541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/15/2024] [Accepted: 11/25/2024] [Indexed: 11/29/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive primary liver malignancy often diagnosed at an advanced stage, resulting in a poor prognosis. Accurate risk stratification and early detection of HCC are critical unmet needs for improving outcomes. Several blood-based biomarkers and imaging tests are available for early detection, prediction, and monitoring of HCC. However, serum protein biomarkers such as alpha-fetoprotein have shown relatively low sensitivity, leading to inaccurate performance. Imaging studies also face limitations related to suboptimal accuracy, high cost, and limited implementation. Recently, liquid biopsy techniques have gained attention for addressing these unmet needs. Liquid biopsy is non-invasive and provides more objective readouts, requiring less reliance on healthcare professional's skills compared to imaging. Circulating tumor cells, cell-free DNA, and extracellular vesicles are targeted in liquid biopsies as novel biomarkers for HCC. Despite their potential, there are debates regarding the role of these novel biomarkers in the HCC care continuum. This review article aims to discuss the technical challenges, recent technical advancements, advantages and disadvantages of these liquid biopsies, as well as their current clinical application and future directions of liquid biopsy in HCC.
Collapse
Affiliation(s)
- Jaeho Park
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yi-Te Lee
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Vatche G. Agopian
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- Department of Surgery, University of California Los Angeles, Los Angeles, CA, USA
| | - Jessica S Liu
- Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL, USA
| | - Ekaterina K. Koltsova
- Smidt Heart Institute, Department of Medicine, Department of Biomedical Sciences, 8700 Beverly Blvd, Los Angeles, CA, USA
| | - Sungyong You
- Department of Urology and Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Yazhen Zhu
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
- Department of Pathology and Laboratory Medicine, University of California, Los Angeles, CA, USA
| | - Hsian-Rong Tseng
- Jonsson Comprehensive Cancer Center, University of California Los Angeles, Los Angeles, CA, USA
- California NanoSystems Institute, Crump Institute for Molecular Imaging, Department of Molecular and Medical Pharmacology, University of California, Los Angeles, CA, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
17
|
Yan J, Jiang Z, Zhang S, Yu Q, Lu Y, Miao R, Tang Z, Fan J, Wu L, Duda DG, Zhou J, Yang X. Spatial‒temporal heterogeneities of liver cancer and the discovery of the invasive zone. Clin Transl Med 2025; 15:e70224. [PMID: 39924620 PMCID: PMC11807767 DOI: 10.1002/ctm2.70224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Accepted: 01/19/2025] [Indexed: 02/11/2025] Open
Abstract
Solid tumours are intricate and highly heterogeneous ecosystems, which grow in and invade normal organs. Their progression is mediated by cancer cells' interaction with different cell types, such as immune cells, stromal cells and endothelial cells, and with the extracellular matrix. Owing to its high incidence, aggressive growth and resistance to local and systemic treatments, liver cancer has particularly high mortality rates worldwide. In recent decades, spatial heterogeneity has garnered significant attention as an unfavourable biological characteristic of the tumour microenvironment, prompting extensive research into its role in liver tumour development. Advances in spatial omics have facilitated the detailed spatial analysis of cell types, states and cell‒cell interactions, allowing a thorough understanding of the spatial and temporal heterogeneities of tumour microenvironment and informing the development of novel therapeutic approaches. This review illustrates the latest discovery of the invasive zone, and systematically introduced specific macroscopic spatial heterogeneities, pathological spatial heterogeneities and tumour microenvironment heterogeneities of liver cancer.
Collapse
Affiliation(s)
- Jiayan Yan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhifeng Jiang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Shiyu Zhang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Qichao Yu
- College of Life SciencesUniversity of Chinese Academy of SciencesBeijingChina
- BGI‐ShenzhenBeishan Industrial ZoneShenzhenChina
| | - Yijun Lu
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Runze Miao
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
- Zhongshan‐BGI Precision Medical CenterZhongshan HospitalFudan UniversityShanghaiChina
| | - Zhaoyou Tang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Jia Fan
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Liang Wu
- BGI‐ShenzhenBeishan Industrial ZoneShenzhenChina
| | - Dan G. Duda
- Steele Laboratories for Tumor BiologyDepartment of Radiation OncologyMassachusetts General Hospital and Harvard Medical SchoolBostonMassachusettsUSA
| | - Jian Zhou
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| | - Xinrong Yang
- Department of Liver Surgery & TransplantationLiver Cancer InstituteZhongshan HospitalFudan UniversityShanghaiChina
- Key Laboratory of Carcinogenesis and Cancer InvasionMinistry of EducationShanghaiChina
| |
Collapse
|
18
|
Zhang Z, Geng C, Song M, Liang H, Zhou K, Liu Y, Wu J, Huang X, Zhou J, Fan J, Peng DH, Zhang L, Cang Y, Sun Y. Loss of SGK1 supports metastatic colonization in hepatocellular carcinoma by promoting resistance to T cell-mediated immunity. J Hepatol 2025:S0168-8278(25)00064-9. [PMID: 39892819 DOI: 10.1016/j.jhep.2025.01.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 01/10/2025] [Accepted: 01/15/2025] [Indexed: 02/04/2025]
Abstract
BACKGROUND & AIMS Immune evasion by tumor cells is a principal obstacle to effectively targeting metastasis in hepatocellular carcinoma (HCC). However, the specific molecular mechanisms facilitating immune escape during metastatic seeding are not fully elucidated. METHODS Utilizing in vivo CRISPR library screening in murine HCC metastasis models under conditions of both intact and depleted T-cell immunity, we identified genes critical to tumor immune evasion during metastatic colonization and investigated intrinsic mechanisms using several experimental approaches. RESULTS Our screens identified Sgk1 as an essential suppressor of metastatic colonization under T-cell immunosurveillance. Sgk1-deficient tumor cells displayed significantly enhanced metastatic capacity in the presence of CD8+ T cells, underscoring the role of Sgk1 in regulating immune escape. Clinical analyses corroborated these findings, showing markedly lower SGK1 expression in circulating tumor cells and metastatic lesions relative to matched primary tumors in patients with HCC, with low SGK1 expression associating with compromised T-cell function and poorer clinical outcomes. Mechanistically, Sgk1 inactivation in tumor cells attenuated CD8+ T cell-mediated, RIPK1-dependent necroptosis - a cell death pathway essential for cytotoxic T cell-mediated restriction of metastasis. Loss of Sgk1 consequently enabled tumor cells to circumvent T cell-induced cytotoxicity, thereby promoting metastatic colonization. Furthermore, the outgrowth of Sgk1-deficient metastatic cells induced a microenvironmental shift toward terminal T-cell exhaustion, establishing conditions conducive to sustained immune evasion. CONCLUSIONS These findings establish SGK1 as a crucial regulator of immune-mediated control over metastatic growth in HCC. SGK1 expression in metastatic lesions may serve as a predictive biomarker for response to immune checkpoint inhibitors, presenting new avenues for therapeutic intervention to overcome immune resistance in metastatic HCC. IMPACT AND IMPLICATIONS Despite metastasis being a common occurrence and lethal determinant in cancers, the mechanism underlying tumor immune evasion during metastatic seeding is unclear. Our study reveals that loss of Sgk1 confers metastatic tumor cells with a survival advantage by abrogating CD8+ T cell-induced RIPK1-dependent necroptosis. Growth of Sgk1-silenced metastasis led to infiltration of terminally exhausted CD8+ T cells, which could be reversed by immune checkpoint inhibitors administered at an early stage of metastatic seeding. These findings provide valuable insights into potential therapeutic strategies targeting resistance to T-cell immunity in cancer metastasis.
Collapse
Affiliation(s)
- Zefan Zhang
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Chenlu Geng
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China; Life Sciences Institute, Zhejiang University, Hangzhou, Zhejiang 310058, China
| | - Minfang Song
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Hengbin Liang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Kaiqian Zhou
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Yang Liu
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Jing Wu
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Xingxu Huang
- Research Center for Life Sciences Computing, Zhejiang Lab, Hangzhou, Zhejiang 311121, China
| | - Jian Zhou
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - Jia Fan
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China
| | - David H Peng
- Dunwill Med-Tech Co., Ltd, Shanghai 201210, China.
| | - Liye Zhang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Yong Cang
- School of Life Science and Technology, ShanghaiTech University, Shanghai 201210, China.
| | - Yunfan Sun
- Department of Hepatobiliary Surgery and Liver Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai 200032, China.
| |
Collapse
|
19
|
Gautam D, Clarke EM, Roweth HG, Smith MR, Battinelli EM. Platelets and circulating (tumor) cells: partners in promoting metastatic cancer. Curr Opin Hematol 2025; 32:52-60. [PMID: 39508182 DOI: 10.1097/moh.0000000000000852] [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: 11/08/2024]
Abstract
PURPOSE OF REVIEW Despite being discovered decades ago, metastasis remains a formidable challenge in cancer treatment. During the intermediate phase of metastasis, tumor cells detach from primary tumor or metastatic sites and travel through the bloodstream and lymphatic system to distant tissues. These tumor cells in the circulation are known as circulating tumor cells (CTCs), and a higher number of CTCs has been linked to poor prognoses in various cancers. The blood is an inhospitable environment for any foreign cells, including CTCs, as they face numerous challenges, such as the shear stress within blood vessels and their interactions with blood and immune cells. However, the exact mechanisms by which CTCs survive the hostile conditions of the bloodstream remain enigmatic. Platelets have been studied for their interactions with tumor cells, promoting their survival, growth, and metastasis. This review explores the latest clinical methods for enumerating CTCs, recent findings on platelet-CTC crosstalk, and current research on antiplatelet therapy as a potential strategy to inhibit metastasis, offering new therapeutic insights. RECENT FINDINGS Laboratory and clinical data have provided insights into the role of platelets in promoting CTC survival, while clinical advancements in CTC enumeration offer improved prognostic tools. SUMMARY CTCs play a critical role in metastasis, and their interactions with platelets aid their survival in the hostile environment of the bloodstream. Understanding this crosstalk offers insights into potential therapeutic strategies, including antiplatelet therapy, to inhibit metastasis and improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Deepa Gautam
- Division of Hematology, Department of Medicine; Brigham and Women's Hospital
- Harvard Medical School, Boston, Massachusetts, USA
| | - Emily M Clarke
- Division of Hematology, Department of Medicine; Brigham and Women's Hospital
| | - Harvey G Roweth
- Division of Hematology, Department of Medicine; Brigham and Women's Hospital
- Harvard Medical School, Boston, Massachusetts, USA
| | - Margaret R Smith
- Division of Hematology, Department of Medicine; Brigham and Women's Hospital
- Harvard Medical School, Boston, Massachusetts, USA
| | - Elisabeth M Battinelli
- Division of Hematology, Department of Medicine; Brigham and Women's Hospital
- Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
20
|
Li H, Li J, Zhang Z, Yang Q, Du H, Dong Q, Guo Z, Yao J, Li S, Li D, Pang N, Li C, Zhang W, Zhou L. Digital Quantitative Detection for Heterogeneous Protein and mRNA Expression Patterns in Circulating Tumor Cells. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410120. [PMID: 39556692 PMCID: PMC11727120 DOI: 10.1002/advs.202410120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/21/2024] [Indexed: 11/20/2024]
Abstract
Hepatocellular carcinoma (HCC) circulating tumor cells (CTCs) exhibit significant phenotypic heterogeneity and diverse gene expression profiles due to epithelial-mesenchymal transition (EMT). However, current detection methods lack the capacity for simultaneous quantification of multidimensional biomarkers, impeding a comprehensive understanding of tumor biology and dynamic changes. Here, the CTC Digital Simultaneous Cross-dimensional Output and Unified Tracking (d-SCOUT) technology is introduced, which enables simultaneous quantification and detailed interpretation of HCC transcriptional and phenotypic biomarkers. Based on self-developed multi-real-time digital PCR (MRT-dPCR) and algorithms, d-SCOUT allows for the unified quantification of Asialoglycoprotein Receptor (ASGPR), Glypican-3 (GPC-3), and Epithelial Cell Adhesion Molecule (EpCAM) proteins, as well as Programmed Death Ligand 1 (PD-L1), GPC-3, and EpCAM mRNA in HCC CTCs, with good sensitivity (LOD of 3.2 CTCs per mL of blood) and reproducibility (mean %CV = 1.80-6.05%). In a study of 99 clinical samples, molecular signatures derived from HCC CTCs demonstrated strong diagnostic potential (AUC = 0.950, sensitivity = 90.6%, specificity = 87.5%). Importantly, by integrating machine learning, d-SCOUT allows clustering of CTC characteristics at the mRNA and protein levels, mapping normalized heterogeneous 2D molecular profiles to assess HCC metastatic risk. Dynamic digital tracking of eight HCC patients undergoing different treatments visually illustrated the therapeutic effects, validating this technology's capability to quantify the treatment efficacy. CTC d-SCOUT enhances understanding of tumor biology and HCC management.
Collapse
Affiliation(s)
- Hao Li
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
- School of Biomedical Engineering (Suzhou)Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230026China
| | - Jinze Li
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
| | - Zhiqi Zhang
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
| | - Qi Yang
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
| | - Hong Du
- The Second Affiliated Hospital of Soochow UniversitySuzhou215000China
| | - Qiongzhu Dong
- Department of General SurgeryHuashan Hospital & Cancer Metastasis InstituteFudan UniversityShanghai200040China
| | - Zhen Guo
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
- School of Biomedical Engineering (Suzhou)Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230026China
| | - Jia Yao
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
| | - Shuli Li
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
| | - Dongshu Li
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
- School of Biomedical Engineering (Suzhou)Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230026China
| | - Nannan Pang
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
| | - Chuanyu Li
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
- School of Biomedical Engineering (Suzhou)Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230026China
| | - Wei Zhang
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
- School of Biomedical Engineering (Suzhou)Division of Life Sciences and MedicineUniversity of Science and Technology of ChinaHefei230026China
| | - Lianqun Zhou
- Suzhou Institute of Biomedical Engineering and TechnologyChinese Academy of ScienceSuzhou215163China
| |
Collapse
|
21
|
Zhou Z, Cai S, Zhou X, Zhao W, Sun J, Zhou Z, Yang Z, Li W, Wang Z, Zou H, Fu H, Wang X, Khoo BL, Yang M. Circulating Tumor Cells Culture: Methods, Challenges, and Clinical Applications. SMALL METHODS 2024:e2401026. [PMID: 39726345 DOI: 10.1002/smtd.202401026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Revised: 11/10/2024] [Indexed: 12/28/2024]
Abstract
Circulating tumor cells (CTCs) play a pivotal role in cancer metastasis and hold considerable potential for clinical diagnosis, therapeutic monitoring, and prognostic evaluation. Nevertheless, the limited quantity of CTCs in liquid biopsy samples poses challenges for comprehensive downstream analysis. In vitro culture of CTCs can effectively address the issue of insufficient CTC numbers. Furthermore, research based on CTC cell lines serves as a valuable complement to traditional cancer cell line-based research. While numerous reports exist on CTC in vitro culture and even the establishment of CTC cell lines, the methods used vary, leading to disparate culture outcomes. This review presents the developmental history and current status of CTC in vitro culture research. Additionally, the culture strategies applied in different methods and analyzed the impact of various steps on culture outcomes are compared. Overall, the review indicates that while the short-term culture of CTCs is relatively straightforward, long-term culture success has been achieved for various specific cancer types but still faces challenges. Further optimization of efficient and widely applicable culture strategies is needed. Additionally, ongoing applications of CTC in vitro culture are summarized, highlighting the potential of expanded CTCs for drug susceptibility testing and as therapeutic tools in personalized treatment.
Collapse
Affiliation(s)
- Zhengdong Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Songhua Cai
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, 518116, China
| | - Xiaoyu Zhou
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| | - Wei Zhao
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Jiayu Sun
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zhihang Zhou
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zihan Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Wenxiu Li
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Zhe Wang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Heng Zou
- Cellomics (Shenzhen) Limited, Shenzhen, 518118, China
| | - Huayang Fu
- Cellomics (Shenzhen) Limited, Shenzhen, 518118, China
| | - Xicheng Wang
- The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, 510080, China
| | - Bee Luan Khoo
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Mengsu Yang
- Department of Precision Diagnostic and Therapeutic Technology, City University of Hong Kong Shenzhen Futian Research Institute, Shenzhen, 518057, China
- Department of Biomedical Sciences, Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR, 999077, China
- Key Laboratory of Biochip Technology, Biotech and Health Centre, Shenzhen Research Institute of City University of Hong Kong, Shenzhen, 518057, China
| |
Collapse
|
22
|
Solhi R, Pourhamzeh M, Zarrabi A, Hassan M, Mirzaei H, Vosough M. Novel biomarkers for monitoring and management of hepatocellular carcinoma. Cancer Cell Int 2024; 24:428. [PMID: 39719624 DOI: 10.1186/s12935-024-03600-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 12/05/2024] [Indexed: 12/26/2024] Open
Abstract
Due to current challenges in the early detection, less than 40% of individuals diagnosed with hepatocellular carcinoma (HCC) are viable candidates for surgical intervention. Therefore, validating and launching of a novel precise diagnostic approach is essential for early diagnosis. Based on developing evidence using circulating tumor cells and their derivatives, circulating miRNAs, and extracellular vesicles (EVs), liquid biopsy may offer a reliable platform for the HCC's early diagnosis. Each liquid biopsy analyte may provide significant areas for diagnosis, prognostic assessment, and treatment monitoring of HCC patients depending on its kind, sensitivity, and specificity. The current review addresses potential clinical applications, current research, and future developments for liquid biopsy in HCC management.
Collapse
Affiliation(s)
- Roya Solhi
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Pourhamzeh
- Departments of Pathology and Medicine, UC San Diego, La Jolla, CA, USA
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul, 34396, Turkey
| | - Moustapha Hassan
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden
| | - Hamed Mirzaei
- Research Center for Biochemistry and Nutrition in Metabolic Diseases, Institute for Basic Sciences, Kashan University of Medical Sciences, Kashan, Islamic Republic of Iran.
| | - Massoud Vosough
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
- Experimental Cancer Medicine, Institution for Laboratory Medicine, Karolinska Institute, Stockholm, Sweden.
| |
Collapse
|
23
|
Huang T, Li F, Wang Y, Gu J, Lu L. Tumor-infiltrating regulatory T cell: A promising therapeutic target in tumor microenvironment. Chin Med J (Engl) 2024; 137:2996-3009. [PMID: 39679474 PMCID: PMC11706582 DOI: 10.1097/cm9.0000000000003450] [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: 07/31/2024] [Indexed: 12/17/2024] Open
Abstract
ABSTRACT Regulatory T cell (Tregs) predominantly maintain the immune balance and prevent autoimmunity via their immunosuppressive functions. However, tumor-infiltrating Tregs (TI-Tregs) may mediate tumor immune tolerance in complex tumor microenvironments, resulting in poor prognosis. Distinguishing specific TI-Treg subpopulations from peripheral Tregs and intratumoral conventional T cells (Tconvs) has recently emerged as an important topic in antitumor therapy. In this review, we summarize novel therapeutic approaches targeting both the metabolic pathways and hallmarks of TI-Tregs in preclinical and clinical studies. Although the phenotypic and functional diversity of TI-Tregs remains unclear, our review provides new insights into TI-Treg-based therapies and facilitates precision medicine for tumor treatment.
Collapse
Affiliation(s)
- Tianning Huang
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
| | - Fan Li
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
| | - Yiming Wang
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Jian Gu
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
| | - Ling Lu
- Department of Plastic and Cosmetic Surgery of the Affiliated Friendship Plastic Surgery Hospital & Hepatobiliary Center of the First Affiliated Hospital, Nanjing Medical University, Nanjing, Jiangsu 210029, China
- Research Unit of Liver Transplantation and Transplant Immunology, Chinese Academy of Medical Sciences, Nanjing, Jiangsu 210029, China
- Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221011, China
| |
Collapse
|
24
|
Li W, He H, Wang H, Wen W. Dynamics of liver cancer cellular taxa revealed through single-cell RNA sequencing: Advances and challenges. Cancer Lett 2024; 611:217394. [PMID: 39689824 DOI: 10.1016/j.canlet.2024.217394] [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: 08/20/2024] [Revised: 10/13/2024] [Accepted: 12/14/2024] [Indexed: 12/19/2024]
Abstract
Liver cancer is a leading cause of death worldwide, representing a substantial public health challenge. The advent of single-cell RNA sequencing has significantly advanced our understanding of cellular dynamics from the onset of liver cancer to therapeutic intervention. This technology has unveiled profound insights into cancer heterogeneity and the tumor microenvironment (TME), enabling the identification of key molecular drivers and phenotypic landscapes of liver cancer at a single-cell resolution. This review highlights recent advancements in mapping functional cell subsets, phenotypic alterations, and the diversity of the TME. These insights are pivotal for advancing targeted therapies and developing prognostic tools. Moreover, this review covers the ongoing challenges and advances from tumor initiation to progression, offering a detailed perspective on advancing personalized treatment.
Collapse
Affiliation(s)
- Wenxin Li
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Clinical Laboratory Medicine, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China
| | - Huisi He
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Oncology, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China
| | - Hongyang Wang
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; The Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, 200438, China.
| | - Wen Wen
- Third Affiliated Hospital of Naval Medical University (Second Military Medical University), National Center for Liver Cancer, Shanghai, 200438, China; Department of Clinical Laboratory Medicine, Third Affiliated Hospital of Naval Medical University (Second Military Medical University), Shanghai, 200438, China; The Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai, 200438, China.
| |
Collapse
|
25
|
Chen J, Jiang Y, Hou M, Liu C, Liu E, Zong Y, Wang X, Meng Z, Gu M, Su Y, Wang H, Fu J. Nuclear translocation of plasma membrane protein ADCY7 potentiates T cell-mediated antitumour immunity in HCC. Gut 2024; 74:128-140. [PMID: 39349007 DOI: 10.1136/gutjnl-2024-332902] [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: 05/21/2024] [Accepted: 08/31/2024] [Indexed: 10/02/2024]
Abstract
BACKGROUND The potency of T cell-mediated responses is a determinant of immunotherapy effectiveness in treating malignancies; however, the clinical efficacy of T-cell therapies has been limited in hepatocellular carcinoma (HCC) owing to the extensive immunosuppressive microenvironment. OBJECTIVE Here, we aimed to investigate the key genes contributing to immune escape in HCC and raise a new therapeutic strategy for remoulding the HCC microenvironment. DESIGN The genome-wide in vivo clustered regularly interspaced short palindromic repeats (CRISPR) screen library was conducted to identify the key genes associated with immune tolerance. Single-cell RNA-seq (scRNA-seq), flow cytometry, HCC mouse models, chromatin immunoprecipitation and coimmunoprecipitation were used to explore the function and mechanism of adenylate cyclase 7 (ADCY7) in HCC immune surveillance. RESULTS Here, a genome-wide in vivo CRISPR screen identified a novel immune modulator-ADCY7. The transmembrane protein ADCY7 undergoes subcellular translocation via caveolae-mediated endocytosis and then translocates to the nucleus with the help of leucine-rich repeat-containing protein 59 (LRRC59) and karyopherin subunit beta 1 (KPNB1). In the nucleus, it functions as a transcription cofactor of CCAAT/enhancer binding protein alpha (CEBPA) to induce CCL5 transcription, thereby increasing CD8+ T cell infiltration to restrain HCC progression. Furthermore, ADCY7 can be secreted as exosomes and enter neighbouring tumour cells to promote CCL5 induction. Exosomes with high ADCY7 levels promote intratumoural infiltration of CD8+ T cells and suppress HCC tumour growth. CONCLUSION We delineate the unconventional function and subcellular location of ADCY7, highlighting its pivotal role in T cell-mediated immunity in HCC and its potential as a promising treatment target.
Collapse
Affiliation(s)
- Jianan Chen
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Youhai Jiang
- Cancer Research Center, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology, Anhui, China
| | - Minghui Hou
- Research Center for Organoids, Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Chunliang Liu
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Erdong Liu
- Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, China
| | - Yali Zong
- Institute of Metabolism & Integrative Biology, Fudan University, Shanghai, China
| | - Xiang Wang
- Second Department of Biliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhengyuan Meng
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Mingye Gu
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Su
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hongyang Wang
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Jing Fu
- International Cooperation Laboratory on Signal Transduction, National Center for Liver Cancer, Ministry of Education Key Laboratory on Signaling Regulation and Targeting Therapy of Liver Cancer, Shanghai Key Laboratory of Hepato-biliary Tumor Biology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| |
Collapse
|
26
|
Rab SO, Roopashree R, Altalbawy FMA, Kumar MR, Chahar M, Singh M, Kubaev A, Alamir HTA, Mohammed F, Kadhim AJ, Alhadrawi M. Phytochemicals and Their Nanoformulations for Targeting Hepatocellular Carcinoma: Exploring Potential and Targeting Strategies. Cell Biochem Funct 2024; 42:e70013. [PMID: 39521962 DOI: 10.1002/cbf.70013] [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: 08/23/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/16/2024]
Abstract
Hepatocellular carcinoma (HCC) continues to pose a global health concern, necessitating the exploration of innovative therapeutic approaches. In the recent decade, targeting tumor stroma consisting of extracellular matrix (ECM), immune cells, vascular system, hypoxia, and also suppressive mechanisms in HCC has attracted interest in repressing tumor growth and metastasis. Phytochemicals have attained considerable attention because of their manifold biological effects and high capacity for anticancer activities. These chemical agents have shown the capability to modulate different cells and secretions within the stroma of malignancies. In recent years, the development of nanoformulations has further enhanced the therapeutic potential of phytochemicals by improving their solubility, bioavailability, and targeted delivery to tumor tissues. This review aims to provide an encyclopedic overview of the potential of phytochemicals and their nanoformulations as promising therapeutic strategies for targeting HCC. The review initially highlights the broad array of phytochemicals exhibiting potent anticancer properties, including flavonoids, alkaloids, terpenoids, and phenolic compounds, among others. Then, the nanoformulations and modification of these agents will be reviewed. Finally, we will review the latest experiments that have examined the modulation of HCC using adjuvant phytochemicals and their nanoformulations.
Collapse
Affiliation(s)
- Safia Obaidur Rab
- Central Labs, King Khalid University, AlQura'a, Abha, Saudi Arabia
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia
| | - M Ravi Kumar
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering & Technology, NIMS University Rajasthan, Jaipur, Rajasthan, India
| | - Manmeet Singh
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Aziz Kubaev
- Department of Maxillofacial Surgery, Samarkand State Medical University, Samarkand, Uzbekistan
| | | | - Faraj Mohammed
- Department of Pharmacy, Al-Manara College for Medical Sciences, Amarah, Maysan, Iraq
| | - Abed J Kadhim
- Department of Medical Engineering, Al-Nisour University College, Baghdad, Iraq
| | - Merwa Alhadrawi
- College of Technical Engineering, The Islamic University, Najaf, Iraq
- College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
27
|
Han Y, Song L, Lv L, Fan C, Ding H. Unraveling the Heterogeneity of Tumor Immune Microenvironment in Hepatocellular Carcinoma by SingleCell RNA Sequencing and its Implications for Prognosis and Therapeutic Response. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2024; 35:876-888. [PMID: 39641225 PMCID: PMC11639598 DOI: 10.5152/tjg.2024.24118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 07/20/2024] [Indexed: 12/07/2024]
Abstract
Tumor immune microenvironment (TIME) has become a new hotspot in cancer research over the past few years. Tumor immune microenvironment of hepatocellular carcinoma (HCC) is especially intriguing as HCC is reported to be highly heterogeneous by previous genomic and cytological studies. It is also closely related to patient prognosis and therapeutic outcome. The recently emerged single-cell RNA sequencing (scRNAseq) technique provides a new tool for TIME study, and current studies have made great advances in defining the roles of TIME in HCC pathogenesis and therapy. Current evidence suggests that heterogeneity is a key player influencing therapeutic response, drug resistance, and prognosis. However, our understanding is limited on the roles of TIME heterogeneity in HCC development, prognosis, and therapeutic response, especially in the era of immunotherapy. This review aims to unravel the heterogeneity of TIME in HCC by scRNAseq, with specific focuses on the cellular, transcriptional, and marker perspectives of TIME heterogeneity in HCC, as well as assessing prognostic and therapeutic response by heterogeneity markers. By summarizing current discoveries regarding TIME heterogeneity, we hope to provide clues on the crucial roles of various cellular components in the development and progression of HCC. We also hope to identify potential markers and therapeutic targets for prognosis assessment and personalized treatment to improve patient outcomes. Combined therapies from multiple dimensions regarding heterogeneity may provide new opportunities to treat HCC more effectively.
Collapse
Affiliation(s)
- Ying Han
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lele Song
- Department of Radiotherapy, the Eighth Medical Center of the Chinese PLA General Hospital, Beijing, China
- Genetron Health (Beijing) Co. Ltd., Beijing, China
| | - Lingna Lv
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Chunlei Fan
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Huiguo Ding
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Ji B, Wang X, Wang X, Xu L, Peng S. scDCA: deciphering the dominant cell communication assembly of downstream functional events from single-cell RNA-seq data. Brief Bioinform 2024; 26:bbae663. [PMID: 39694816 DOI: 10.1093/bib/bbae663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 11/24/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
Cell-cell communications (CCCs) involve signaling from multiple sender cells that collectively impact downstream functional processes in receiver cells. Currently, computational methods are lacking for quantifying the contribution of pairwise combinations of cell types to specific functional processes in receiver cells (e.g. target gene expression or cell states). This limitation has impeded understanding the underlying mechanisms of cancer progression and identifying potential therapeutic targets. Here, we proposed a deep learning-based method, scDCA, to decipher the dominant cell communication assembly (DCA) that have a higher impact on a particular functional event in receiver cells from single-cell RNA-seq data. Specifically, scDCA employed a multi-view graph convolution network to reconstruct the CCCs landscape at single-cell resolution, and then identified DCA by interpreting the model with the attention mechanism. Taking the samples from advanced renal cell carcinoma as a case study, the scDCA was successfully applied and validated in revealing the DCA affecting the crucial gene expression in immune cells. The scDCA was also applied and validated in revealing the DCA responsible for the variation of 14 typical functional states of malignant cells. Furthermore, the scDCA was applied and validated to explore the alteration of CCCs under clinical intervention by comparing the DCA for certain cytotoxic factors between patients with and without immunotherapy. In summary, scDCA provides a valuable and practical tool for deciphering the cell type combinations with the most dominant impact on a specific functional process of receiver cells, which is of great significance for precise cancer treatment. Our data and code are free available at a public GitHub repository: https://github.com/pengsl-lab/scDCA.git.
Collapse
Affiliation(s)
- Boya Ji
- College of Computer Science and Electronic Engineering, Hunan University, Yuelu, 410006 Changsha, China
| | - Xiaoqi Wang
- College of Computer Science and Electronic Engineering, Hunan University, Yuelu, 410006 Changsha, China
| | - Xiang Wang
- The Second Xiangya Hospital, Central South University, Yuelu, 410006 Changsha, China
| | - Liwen Xu
- College of Computer Science and Electronic Engineering, Hunan University, Yuelu, 410006 Changsha, China
| | - Shaoliang Peng
- College of Computer Science and Electronic Engineering, Hunan University, Yuelu, 410006 Changsha, China
| |
Collapse
|
29
|
Dompé C, Chojnowska A, Ramlau R, Nowicki M, Alix-Panabières C, Budna-Tukan J. Unveiling the dynamics of circulating tumor cells in colorectal cancer: from biology to clinical applications. Front Cell Dev Biol 2024; 12:1498032. [PMID: 39539964 PMCID: PMC11557528 DOI: 10.3389/fcell.2024.1498032] [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: 09/19/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
This review delves into the pivotal role of circulating tumor cells (CTCs) in colorectal cancer (CRC) metastasis, focusing on their biological properties, interactions with the immune system, advanced detection techniques, and clinical implications. We explored how metastasis-competent CTCs evade immune surveillance and proliferate, utilizing cutting-edge detection and isolation technologies, such as microfluidic devices and immunological assays, to enhance sensitivity and specificity. The review highlights the significant impact of CTC interactions with immune cells on tumor progression and patient outcomes. It discusses the application of these findings in clinical settings, including non-invasive liquid biopsies for early diagnosis, prognosis, and treatment monitoring. Despite advancements, challenges remain, such as the need for standardized methods to consistently capture and analyze CTCs. Addressing these challenges through further molecular and cellular research on CTCs could lead to improved interventions and outcomes for CRC patients, underscoring the importance of unraveling the complex dynamics of CTCs in cancer progression.
Collapse
Affiliation(s)
- Claudia Dompé
- Department of Immunology, Poznan University of Medical Sciences, Poznan, Poland
- Doctoral School, Poznan University of Medical Sciences, Poznan, Poland
| | | | - Rodryg Ramlau
- Department of Oncology, Poznan University of Medical Sciences, Poznan, Poland
| | - Michal Nowicki
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
| | - Catherine Alix-Panabières
- Laboratory of Rare Human Circulating Cells and Liquid Biopsy (LCCRH), University Medical Centre of Montpellier, Montpellier, France
- Centre de Recherche en Ecologie et Evolution du Cancer, Maladies Infectieuses et Vecteurs: Ecologie, Génétique, Evolution et Contrôle, University of Montpellier, Centre National de la Recherche Scientifique, Institut de Recherche Pour le Dévelopement, Montpellier, France
- European Liquid Biopsy Society (ELBS), Hamburg, Germany
| | - Joanna Budna-Tukan
- Department of Histology and Embryology, Poznan University of Medical Sciences, Poznan, Poland
- Department of Anatomy and Histology, Collegium Medicum, University of Zielona Gora, Zielona Gora, Poland
| |
Collapse
|
30
|
Liu Z, Chen J, Ren Y, Liu S, Ba Y, Zuo A, Luo P, Cheng Q, Xu H, Han X. Multi-stage mechanisms of tumor metastasis and therapeutic strategies. Signal Transduct Target Ther 2024; 9:270. [PMID: 39389953 PMCID: PMC11467208 DOI: 10.1038/s41392-024-01955-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Revised: 07/18/2024] [Accepted: 08/24/2024] [Indexed: 10/12/2024] Open
Abstract
The cascade of metastasis in tumor cells, exhibiting organ-specific tendencies, may occur at numerous phases of the disease and progress under intense evolutionary pressures. Organ-specific metastasis relies on the formation of pre-metastatic niche (PMN), with diverse cell types and complex cell interactions contributing to this concept, adding a new dimension to the traditional metastasis cascade. Prior to metastatic dissemination, as orchestrators of PMN formation, primary tumor-derived extracellular vesicles prepare a fertile microenvironment for the settlement and colonization of circulating tumor cells at distant secondary sites, significantly impacting cancer progression and outcomes. Obviously, solely intervening in cancer metastatic sites passively after macrometastasis is often insufficient. Early prediction of metastasis and holistic, macro-level control represent the future directions in cancer therapy. This review emphasizes the dynamic and intricate systematic alterations that occur as cancer progresses, illustrates the immunological landscape of organ-specific PMN creation, and deepens understanding of treatment modalities pertinent to metastasis, thereby identifying some prognostic and predictive biomarkers favorable to early predict the occurrence of metastasis and design appropriate treatment combinations.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jingqi Chen
- Department of Clinical Medicine, Zhengzhou University, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Shutong Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuhao Ba
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Anning Zuo
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, Zhengzhou, Henan, China.
| |
Collapse
|
31
|
Cheng X, Cao Y, Liu X, Li Y, Li Q, Gao D, Yu Q. Single-cell and spatial omics unravel the spatiotemporal biology of tumour border invasion and haematogenous metastasis. Clin Transl Med 2024; 14:e70036. [PMID: 39350478 PMCID: PMC11442492 DOI: 10.1002/ctm2.70036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 08/14/2024] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
Solid tumours exhibit a well-defined architecture, comprising a differentiated core and a dynamic border that interfaces with the surrounding tissue. This border, characterised by distinct cellular morphology and molecular composition, serves as a critical determinant of the tumour's invasive behaviour. Notably, the invasive border of the primary tumour represents the principal site for intravasation of metastatic cells. These cells, known as circulating tumour cells (CTCs), function as 'seeds' for distant dissemination and display remarkable heterogeneity. Advancements in spatial sequencing technology are progressively unveiling the spatial biological features of tumours. However, systematic investigations specifically targeting the characteristics of the tumour border remain scarce. In this comprehensive review, we illuminate key biological insights along the tumour body-border-haematogenous metastasis axis over the past five years. We delineate the distinctive landscape of tumour invasion boundaries and delve into the intricate heterogeneity and phenotype of CTCs, which orchestrate haematogenous metastasis. These insights have the potential to explain the basis of tumour invasion and distant metastasis, offering new perspectives for the development of more complex and precise clinical interventions and treatments.
Collapse
Affiliation(s)
- Xifu Cheng
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Pathogen Biology and ImmunologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yuke Cao
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Xiangyi Liu
- Queen Mary SchoolJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Yuanheng Li
- Queen Mary SchoolJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qing Li
- Department of Oncologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Dian Gao
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
- Department of Pathogen Biology and ImmunologySchool of Basic Medical SciencesJiangxi Medical CollegeNanchang UniversityNanchangChina
| | - Qiongfang Yu
- Department of Gastroenterology and Hepatologythe Second Affiliated HospitalJiangxi Medical CollegeNanchang UniversityNanchangChina
| |
Collapse
|
32
|
Balaji N, Kukal S, Bhat A, Pradhan N, Minocha S, Kumar S. A quartet of cancer stem cell niches in hepatocellular carcinoma. Cytokine Growth Factor Rev 2024; 79:39-51. [PMID: 39217065 DOI: 10.1016/j.cytogfr.2024.08.007] [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: 07/03/2024] [Revised: 08/20/2024] [Accepted: 08/20/2024] [Indexed: 09/04/2024]
Abstract
Hepatocellular Carcinoma (HCC), the most prevalent type of primary liver cancer, is known for its aggressive behavior and poor prognosis. The Cancer Stem Cell theory, which postulates the presence of a small population of self-renewing cells called Cancer Stem Cells (CSCs), provides insights into various clinical and molecular features of HCC such as tumor heterogeneity, metabolic adaptability, therapy resistance, and recurrence. These CSCs are nurtured in the tumor microenvironment (TME), where a mix of internal and external factors creates a tumor-supportive niche that is continuously evolving both spatially and temporally, thus enhancing the tumor's complexity. This review details the origins of hepatic CSCs (HCSCs) and the factors influencing their stem-like qualities. It highlights the reciprocal crosstalk between HCSCs and the TME (hypoxic, vascular, invasive, and immune niches), exploring the signaling pathways involved and how these interactions control the malignant traits of CSCs. Additionally, it discusses potential therapeutic approaches targeting the HCSC niche and their possible uses in clinical practice.
Collapse
Affiliation(s)
- Neha Balaji
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India
| | - Samiksha Kukal
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India
| | - Anjali Bhat
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India
| | - Nikita Pradhan
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India
| | - Shilpi Minocha
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India.
| | - Saran Kumar
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, 110016, India.
| |
Collapse
|
33
|
Guan Y, Liu X, Tian J, Yang G, Xu F, Guo N, Guo L, Wan Z, Huang Z, Gao M, Chong T. CCL5 promotes the epithelial-mesenchymal transition of circulating tumor cells in renal cancer. J Transl Med 2024; 22:817. [PMID: 39227943 PMCID: PMC11370314 DOI: 10.1186/s12967-024-05297-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 05/12/2024] [Indexed: 09/05/2024] Open
Abstract
BACKGROUND Circulating tumor cells (CTCs) are pivotal in tumor metastasis across cancers, yet their specific role in renal cancer remains unclear. METHODS This study investigated C-C motif chemokine ligand 5 (CCL5)'s tumorigenic impact on renal cancer cells and CTCs using bioinformatics, in vivo, and in vitro experiments. It also assessed renal cancer patients' CTCs prognostic value through Lasso regression and Kaplan-Meier survival curves. RESULTS Bioinformatics analysis revealed differential genes focusing on cellular adhesion and migration between CTCs and tumor cells. CCL5 exhibited high expression in various CTCs, correlating with poor prognosis in renal cancer. In 786-O-CTCs, CCL5 enhanced malignancy, while in renal cell carcinoma cell line CAKI-2 and 786-O, it promoted epithelial-mesenchymal transition (EMT) via smad2/3, influencing cellular characteristics. The nude mouse model suggested CCL5 increased CTCs and intensified EMT, enhancing lung metastasis. Clinical results shown varying prognostic values for different EMT-typed CTCs, with mesenchymal CTCs having the highest value. CONCLUSIONS In summary, CCL5 promoted EMT in renal cancer cells and CTCs through smad2/3, enhancing the malignant phenotype and facilitating lung metastasis. Mesenchymal-type CTC-related factors can construct a risk model for renal cancer patients, allowing personalized treatment based on metastatic risk prediction.
Collapse
Affiliation(s)
- Yibing Guan
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
- Department of Urology, The First Affiliated Hospital, Zhengzhou University, No 1 Jianshe East Road, Zhengzhou, 450052, He Nan, China
| | - Xueyi Liu
- School of Chemistry, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Juanhua Tian
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Guang Yang
- Henan Key Lab Reprod & Genet, The First Affiliated Hospital, Zhengzhou University, No 1 Jianshe East Road, Zhengzhou, 450052, He Nan, China
| | - Fangshi Xu
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Ni Guo
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Lingyu Guo
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Ziyan Wan
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Zhixin Huang
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Mei Gao
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China
| | - Tie Chong
- Department of Urology, School of Medicine, The Second Affiliated Hospital, Xi'an Jiaotong University, No 157 Xiwu Road, Xi'an, 710004, Shaan Xi, China.
| |
Collapse
|
34
|
Gu X, Wei S, Lv X. Circulating tumor cells: from new biological insights to clinical practice. Signal Transduct Target Ther 2024; 9:226. [PMID: 39218931 PMCID: PMC11366768 DOI: 10.1038/s41392-024-01938-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/31/2024] [Accepted: 07/29/2024] [Indexed: 09/04/2024] Open
Abstract
The primary reason for high mortality rates among cancer patients is metastasis, where tumor cells migrate through the bloodstream from the original site to other parts of the body. Recent advancements in technology have significantly enhanced our comprehension of the mechanisms behind the bloodborne spread of circulating tumor cells (CTCs). One critical process, DNA methylation, regulates gene expression and chromosome stability, thus maintaining dynamic equilibrium in the body. Global hypomethylation and locus-specific hypermethylation are examples of changes in DNA methylation patterns that are pivotal to carcinogenesis. This comprehensive review first provides an overview of the various processes that contribute to the formation of CTCs, including epithelial-mesenchymal transition (EMT), immune surveillance, and colonization. We then conduct an in-depth analysis of how modifications in DNA methylation within CTCs impact each of these critical stages during CTC dissemination. Furthermore, we explored potential clinical implications of changes in DNA methylation in CTCs for patients with cancer. By understanding these epigenetic modifications, we can gain insights into the metastatic process and identify new biomarkers for early detection, prognosis, and targeted therapies. This review aims to bridge the gap between basic research and clinical application, highlighting the significance of DNA methylation in the context of cancer metastasis and offering new avenues for improving patient outcomes.
Collapse
Affiliation(s)
- Xuyu Gu
- Department of Oncology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Shiyou Wei
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China
| | - Xin Lv
- Department of Anesthesiology, Shanghai Pulmonary Hospital, School of Medicine, Tongji University, Shanghai, China.
| |
Collapse
|
35
|
Gan L, Wang W, Jiang J, Tian K, Liu W, Cao Z. Dual role of Nrf2 signaling in hepatocellular carcinoma: promoting development, immune evasion, and therapeutic challenges. Front Immunol 2024; 15:1429836. [PMID: 39286246 PMCID: PMC11402828 DOI: 10.3389/fimmu.2024.1429836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Accepted: 08/12/2024] [Indexed: 09/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the predominant form of liver cancer and ranks as the third leading cause of cancer-related mortality globally. The liver performs a wide range of tasks and is the primary organ responsible for metabolizing harmful substances and foreign compounds. Oxidative stress has a crucial role in growth and improvement of hepatocellular carcinoma (HCC). Nuclear factor erythroid 2 (1)-related factor 2 (Nrf2) is an element that regulates transcription located in the cytoplasm. It controls the balance of redox reactions by stimulating the expression of many genes that depend on antioxidant response elements. Nrf2 has contrasting functions in the normal, healthy liver and HCC. In the normal liver, Nrf2 provides advantageous benefits, while in HCC it promotes harmful effects that support the growth and survival of HCC. Continuous activation of Nrf2 has been detected in HCC and promotes its advancement and aggressiveness. In addition, Activation of Nrf2 may lead to immune evasion, weakening the immune cells' ability to attack tumors and thereby promoting tumor development. Furthermore, chemoresistance in HCC, which is considered a form of stress response to chemotherapy medications, significantly impedes the effectiveness of HCC treatment. Stress management is typically accomplished by activating specific signal pathways and chemical variables. One important element in the creation of chemoresistance in HCC is nuclear factor-E2-related factor 2 (Nrf2). Nrf2 is a transcription factor that regulates the activation and production of a group of genes that encode proteins responsible for protecting cells from damage. This occurs through the Nrf2/ARE pathway, which is a crucial mechanism for combating oxidative stress within cells.
Collapse
Affiliation(s)
- Lin Gan
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Jinxiu Jiang
- Department of Gastroenterology, The First Affiliated Hospital of Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Ke Tian
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Wei Liu
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| | - Zhumin Cao
- Department of Hepatobiliary Surgery, The Seventh People’s Hospital of Chongqing, Chongqing, China
| |
Collapse
|
36
|
Zhou S, Xu H, Duan Y, Tang Q, Huang H, Bi F. Survival mechanisms of circulating tumor cells and their implications for cancer treatment. Cancer Metastasis Rev 2024; 43:941-957. [PMID: 38436892 DOI: 10.1007/s10555-024-10178-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Accepted: 02/26/2024] [Indexed: 03/05/2024]
Abstract
Metastasis remains the principal trigger for relapse and mortality across diverse cancer types. Circulating tumor cells (CTCs), which originate from the primary tumor or its metastatic sites, traverse the vascular system, serving as precursors in cancer recurrence and metastasis. Nevertheless, before CTCs can establish themselves in the distant parenchyma, they must overcome significant challenges present within the circulatory system, including hydrodynamic shear stress (HSS), oxidative damage, anoikis, and immune surveillance. Recently, there has been a growing body of compelling evidence suggesting that a specific subset of CTCs can persist within the bloodstream, but the precise mechanisms of their survival remain largely elusive. This review aims to present an outline of the survival challenges encountered by CTCs and to summarize the recent advancements in understanding the underlying survival mechanisms, suggesting their implications for cancer treatment.
Collapse
Affiliation(s)
- Shuang Zhou
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huanji Xu
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Yichun Duan
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Qiulin Tang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Huixi Huang
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China
| | - Feng Bi
- Division of Abdominal Cancer, Department of Medical Oncology, Cancer Center and Laboratory of Molecular Targeted Therapy in Oncology, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, China.
| |
Collapse
|
37
|
Liu B, Hu S, Wang X. Applications of single-cell technologies in drug discovery for tumor treatment. iScience 2024; 27:110486. [PMID: 39171294 PMCID: PMC11338156 DOI: 10.1016/j.isci.2024.110486] [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] [Indexed: 08/23/2024] Open
Abstract
Single-cell technologies have been known as advanced and powerful tools to study tumor biological systems at the single-cell resolution and are playing increasingly critical roles in multiple stages of drug discovery and development. Specifically, single-cell technologies can promote the discovery of drug targets, help high-throughput screening at single-cell level, and contribute to pharmacokinetic studies of anti-tumor drugs. Emerging single-cell analysis technologies have been developed to further integrating multidimensional single-cell molecular features, expanding the scale of single-cell data, profiling phenotypic impact of genes in single cell, and providing full-length coverage single-cell sequencing. In this review, we systematically summarized the applications of single-cell technologies in various sections of drug discovery for tumor treatment, including target identification, high-throughput drug screening, and pharmacokinetic evaluation and highlighted emerging single-cell technologies in providing in-depth understanding of tumor biology. Single-cell-technology-based drug discovery is expected to further optimize therapeutic strategies and improve clinical outcomes of tumor patients.
Collapse
Affiliation(s)
- Bingyu Liu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
| | - Shunfeng Hu
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Shandong University, Jinan, Shandong 250021, China
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China
- Taishan Scholars Program of Shandong Province, Jinan, Shandong 250021, China
| |
Collapse
|
38
|
Jiang H. Latest Research Progress of Liquid Biopsy in Tumor-A Narrative Review. Cancer Manag Res 2024; 16:1031-1042. [PMID: 39165347 PMCID: PMC11335005 DOI: 10.2147/cmar.s479338] [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: 05/21/2024] [Accepted: 08/14/2024] [Indexed: 08/22/2024] Open
Abstract
Human life expectancy is significantly impacted by cancer, with liquid biopsy emerging as an advantageous method for cancer detection because of its noninvasive nature, high accuracy, ease of sampling, and cost-effectiveness compared with conventional tissue biopsy techniques. Liquid biopsy shows promise in early cancer detection, real-time monitoring, and personalized treatment for various cancers, including lung, cervical, and prostate cancers, and offers innovative approaches for cancer diagnosis and management. By utilizing circulating tumor DNA, circulating tumor cells, and exosomes as biomarkers, liquid biopsy enables the tracking of cancer progression. Various techniques commonly used in life sciences research, such as polymerase chain reaction (PCR), next-generation sequencing (NGS), and droplet digital PCR, are employed to assess cancer progression on the basis of different indicators. This review examines the latest advancements in liquid biopsy markers-circulating tumor DNA (ctDNA), circulating tumor cells (CTCs), and exosomes-for cancer diagnosis over the past three years, with a focus on their detection methodologies and clinical applications. It encapsulates the pivotal aims of liquid biopsy, including early detection, therapy response prediction, treatment monitoring, prognostication, and its relevance in minimal residual disease, while also addressing the challenges facing routine clinical adoption. By combining the latest research advancements and practical clinical experiences, this work focuses on discussing the clinical significance of DNA methylation biomarkers and their applications in tumor screening, auxiliary diagnosis, companion diagnosis, and recurrence monitoring. These discussions may help enhance the application of liquid biopsy throughout the entire process of tumor diagnosis and treatment, thereby providing patients with more precise and effective treatment plans.
Collapse
Affiliation(s)
- Hua Jiang
- Department of Urology, The Fifth Affiliated Hospital of Zunyi Medical University (Zhuhai Sixth People’s Hospital), Zhuhai, People’s Republic of China
| |
Collapse
|
39
|
Safri F, Nguyen R, Zerehpooshnesfchi S, George J, Qiao L. Heterogeneity of hepatocellular carcinoma: from mechanisms to clinical implications. Cancer Gene Ther 2024; 31:1105-1112. [PMID: 38499648 PMCID: PMC11327108 DOI: 10.1038/s41417-024-00764-w] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 03/06/2024] [Accepted: 03/08/2024] [Indexed: 03/20/2024]
Abstract
Hepatocellular Carcinoma (HCC) is one of the most common types of primary liver cancer. Current treatment options have limited efficacy against this malignancy, primarily owing to difficulties in early detection and the inherent resistance to existing drugs. Tumor heterogeneity is a pivotal factor contributing significantly to treatment resistance and recurrent manifestations of HCC. Intratumoral heterogeneity is an important aspect of the spectrum of complex tumor heterogeneity and contributes to late diagnosis and treatment failure. Therefore, it is crucial to thoroughly understand the molecular mechanisms of how tumor heterogeneity develops. This review aims to summarize the possible molecular dimensions of tumor heterogeneity with an emphasis on intratumoral heterogeneity, evaluate its profound impact on the diagnosis and therapeutic strategies for HCC, and explore the suitability of appropriate pre-clinical models that can be used to best study tumor heterogeneity; thus, opening new avenues for cancer treatment.
Collapse
Affiliation(s)
- Fatema Safri
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Romario Nguyen
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Shadi Zerehpooshnesfchi
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia
| | - Jacob George
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
| | - Liang Qiao
- Storr Liver Centre, The Westmead Institute for Medical Research, The University of Sydney, Westmead, NSW, 2145, Australia.
| |
Collapse
|
40
|
Chen HC, Ma Y, Cheng J, Chen YC. Advances in Single-Cell Techniques for Linking Phenotypes to Genotypes. CANCER HETEROGENEITY AND PLASTICITY 2024; 1:0004. [PMID: 39156821 PMCID: PMC11328949 DOI: 10.47248/chp2401010004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/20/2024]
Abstract
Single-cell analysis has become an essential tool in modern biological research, providing unprecedented insights into cellular behavior and heterogeneity. By examining individual cells, this approach surpasses conventional population-based methods, revealing critical variations in cellular states, responses to environmental cues, and molecular signatures. In the context of cancer, with its diverse cell populations, single-cell analysis is critical for investigating tumor evolution, metastasis, and therapy resistance. Understanding the phenotype-genotype relationship at the single-cell level is crucial for deciphering the molecular mechanisms driving tumor development and progression. This review highlights innovative strategies for selective cell isolation based on desired phenotypes, including robotic aspiration, laser detachment, microraft arrays, optical traps, and droplet-based microfluidic systems. These advanced tools facilitate high-throughput single-cell phenotypic analysis and sorting, enabling the identification and characterization of specific cell subsets, thereby advancing therapeutic innovations in cancer and other diseases.
Collapse
Affiliation(s)
- Hsiao-Chun Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| | - Yushu Ma
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| | - Jinxiong Cheng
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
| | - Yu-Chih Chen
- UPMC Hillman Cancer Center, University of Pittsburgh, 5115 Centre Ave, Pittsburgh, PA 15232, USA
- Department of Computational and Systems Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
- Department of Bioengineering, Swanson School of Engineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15260, USA
- CMU-Pitt Ph.D. Program in Computational Biology, University of Pittsburgh, 3420 Forbes Avenue, Pittsburgh, PA 15260, USA
| |
Collapse
|
41
|
Peeters F, Cappuyns S, Piqué-Gili M, Phillips G, Verslype C, Lambrechts D, Dekervel J. Applications of single-cell multi-omics in liver cancer. JHEP Rep 2024; 6:101094. [PMID: 39022385 PMCID: PMC11252522 DOI: 10.1016/j.jhepr.2024.101094] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 03/18/2024] [Accepted: 03/27/2024] [Indexed: 07/20/2024] Open
Abstract
Primary liver cancer, more specifically hepatocellular carcinoma (HCC), remains a significant global health problem associated with increasing incidence and mortality. Clinical, biological, and molecular heterogeneity are well-known hallmarks of cancer and HCC is considered one of the most heterogeneous tumour types, displaying substantial inter-patient, intertumoural and intratumoural variability. This heterogeneity plays a pivotal role in hepatocarcinogenesis, metastasis, relapse and drug response or resistance. Unimodal single-cell sequencing techniques have already revolutionised our understanding of the different layers of molecular hierarchy in the tumour microenvironment of HCC. By highlighting the cellular heterogeneity and the intricate interactions among cancer, immune and stromal cells before and during treatment, these techniques have contributed to a deeper comprehension of tumour clonality, hematogenous spreading and the mechanisms of action of immune checkpoint inhibitors. However, major questions remain to be elucidated, with the identification of biomarkers predicting response or resistance to immunotherapy-based regimens representing an important unmet clinical need. Although the application of single-cell multi-omics in liver cancer research has been limited thus far, a revolution of individualised care for patients with HCC will only be possible by integrating various unimodal methods into multi-omics methodologies at the single-cell resolution. In this review, we will highlight the different established single-cell sequencing techniques and explore their biological and clinical impact on liver cancer research, while casting a glance at the future role of multi-omics in this dynamic and rapidly evolving field.
Collapse
Affiliation(s)
- Frederik Peeters
- Digestive Oncology, Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Centre for Cancer Biology, Leuven, Belgium
| | - Sarah Cappuyns
- Digestive Oncology, Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Centre for Cancer Biology, Leuven, Belgium
| | - Marta Piqué-Gili
- Liver Cancer Translational Research Laboratory, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gino Phillips
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Centre for Cancer Biology, Leuven, Belgium
| | - Chris Verslype
- Digestive Oncology, Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium
- VIB Centre for Cancer Biology, Leuven, Belgium
| | - Jeroen Dekervel
- Digestive Oncology, Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| |
Collapse
|
42
|
He J, Duan P, Liu Y, Feng T, Wang S, Lin X, Xie J, Liu X. Unveiling the Impact of Hemodynamics on Endothelial Inflammation-Mediated Hepatocellular Carcinoma Metastasis Using a Biomimetic Vascular Flow Model. Adv Healthc Mater 2024; 13:e2304439. [PMID: 38486060 DOI: 10.1002/adhm.202304439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Revised: 02/28/2024] [Indexed: 03/23/2024]
Abstract
Hepatocellular carcinoma (HCC) hematogenous dissemination is a leading cause of HCC-related deaths. The inflammatory facilitates this process by promoting the adhesion and invasion of tumor cells in the circulatory system. But the contribution of hemodynamics to this process remains poorly understood due to the lack of a suitable vascular flow model for investigation. This study develops a vascular flow model to examine the impact of hemodynamics on endothelial inflammation-mediated HCC metastasis. This work finds the increasing shear stress will reduce the recruitment of HCC cells by disturbing adhesion forces between endothelium and HCC cells. However, this reduction will be restored by the inflammation. When applying high FSS (4-6 dyn cm-2) to the inflammatory endothelium, there will be a 4.8-fold increase in HCC cell adhesions compared to normal condition. Nevertheless, the increase fold of cell adhesions is inapparent, around 1.5-fold, with low and medium FSS. This effect can be attributed to the FSS-induced upregulation of ICAM-1 and VCAM-1 of the inflammatory endothelium, which serve to strengthen cell binding forces. These findings indicate that hemodynamics plays a key role in HCC metastasis during endothelial inflammation by regulating the expression of adhesion-related factors.
Collapse
Affiliation(s)
- Jia He
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Peiyan Duan
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Yi Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Tang Feng
- Department of Biotherapy, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Shuo Wang
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xinyi Lin
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Jing Xie
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Xiaoheng Liu
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
43
|
Ji Y, An Q, Wen X, Xu Z, Xia Z, Xia Z, Hu Q, Lei S. Liver cancer from the perspective of single-cell sequencing: a review combined with bibliometric analysis. J Cancer Res Clin Oncol 2024; 150:316. [PMID: 38910204 PMCID: PMC11194221 DOI: 10.1007/s00432-024-05855-7] [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: 03/16/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
BACKGROUND Liver cancer (LC) is a prevalent malignancy and a leading cause of cancer-related mortality worldwide. Extensive research has been conducted to enhance patient outcomes and develop effective prevention strategies, ranging from molecular mechanisms to clinical interventions. Single-cell sequencing, as a novel bioanalysis technology, has significantly contributed to the understanding of the global cognition and dynamic changes in liver cancer. However, there is a lack of bibliometric analysis in this specific research area. Therefore, the objective of this study is to provide a comprehensive overview of the knowledge structure and research hotspots in the field of single-cell sequencing in liver cancer research through the use of bibliometrics. METHOD Publications related to the application of single-cell sequencing technology to liver cancer research as of December 31, 2023, were searched on the web of science core collection (WoSCC) database. VOSviewers, CiteSpace, and R package "bibliometrix" were used to conduct this bibliometric analysis. RESULTS A total of 331 publications from 34 countries, primarily led by China and the United States, were included in this study. The research focuses on the application of single cell sequencing technology to liver cancer, and the number of related publications has been increasing year by year. The main research institutions involved in this field are Fudan University, Sun Yat-Sen University, and the Chinese Academy of Sciences. Frontiers in Immunology and Nature Communications is the most popular journal in this field, while Cell is the most frequently co-cited journal. These publications are authored by 2799 individuals, with Fan Jia and Zhou Jian having the most published papers, and Llovet Jm being the most frequently co-cited author. The use of single cell sequencing to explore the immune microenvironment of liver cancer, as well as its implications in immunotherapy and chemotherapy, remains the central focus of this field. The emerging research hotspots are characterized by keywords such as 'Gene-Expression', 'Prognosis', 'Tumor Heterogeneity', 'Immunoregulation', and 'Tumor Immune Microenvironment'. CONCLUSION This is the first bibliometric study that comprehensively summarizes the research trends and developments on the application of single cell sequencing in liver cancer. The study identifies recent research frontiers and hot directions, providing a valuable reference for researchers exploring the landscape of liver cancer, understanding the composition of the immune microenvironment, and utilizing single-cell sequencing technology to guide and enhance the prognosis of liver cancer patients.
Collapse
Affiliation(s)
- Yanwei Ji
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qi An
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Xinyu Wen
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zhou Xu
- The Second Clinical Medical College, Jiangxi Medical College, Nanchang University, Jiangxi, Nanchang, China
| | - Zhengyuan Xia
- Department of Anesthesiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, China
- Faculty of Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, Macao, China
| | - Zhongyuan Xia
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Shaoqing Lei
- Department of Anesthesiology, Renmin Hospital of Wuhan University, Wuhan, China.
| |
Collapse
|
44
|
Miao Y, Li Z, Feng J, Lei X, Shan J, Qian C, Li J. The Role of CD4 +T Cells in Nonalcoholic Steatohepatitis and Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:6895. [PMID: 39000005 PMCID: PMC11240980 DOI: 10.3390/ijms25136895] [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: 05/06/2024] [Revised: 06/14/2024] [Accepted: 06/19/2024] [Indexed: 07/14/2024] Open
Abstract
Hepatocellular carcinoma (HCC) has become the fourth leading cause of cancer-related deaths worldwide; annually, approximately 830,000 deaths related to liver cancer are diagnosed globally. Since early-stage HCC is clinically asymptomatic, traditional treatment modalities, including surgical ablation, are usually not applicable or result in recurrence. Immunotherapy, particularly immune checkpoint blockade (ICB), provides new hope for cancer therapy; however, immune evasion mechanisms counteract its efficiency. In addition to viral exposure and alcohol addiction, nonalcoholic steatohepatitis (NASH) has become a major cause of HCC. Owing to NASH-related aberrant T cell activation causing tissue damage that leads to impaired immune surveillance, NASH-associated HCC patients respond much less efficiently to ICB treatment than do patients with other etiologies. In addition, abnormal inflammation contributes to NASH progression and NASH-HCC transition, as well as to HCC immune evasion. Therefore, uncovering the detailed mechanism governing how NASH-associated immune cells contribute to NASH progression would benefit HCC prevention and improve HCC immunotherapy efficiency. In the following review, we focused our attention on summarizing the current knowledge of the role of CD4+T cells in NASH and HCC progression, and discuss potential therapeutic strategies involving the targeting of CD4+T cells for the treatment of NASH and HCC.
Collapse
Affiliation(s)
- Yadi Miao
- School of Medicine, Chongqing University, Chongqing 400030, China
| | - Ziyong Li
- School of Medicine, Chongqing University, Chongqing 400030, China
| | - Juan Feng
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Xia Lei
- School of Medicine, Chongqing University, Chongqing 400030, China
| | - Juanjuan Shan
- School of Medicine, Chongqing University, Chongqing 400030, China
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Cheng Qian
- School of Medicine, Chongqing University, Chongqing 400030, China
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| | - Jiatao Li
- School of Medicine, Chongqing University, Chongqing 400030, China
- Center for Precision Medicine of Cancer, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing 400030, China
| |
Collapse
|
45
|
Wang X, Bai L, Kong L, Guo Z. Advances in circulating tumor cells for early detection, prognosis and metastasis reduction in lung cancer. Front Oncol 2024; 14:1411731. [PMID: 38974237 PMCID: PMC11224453 DOI: 10.3389/fonc.2024.1411731] [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: 04/03/2024] [Accepted: 06/07/2024] [Indexed: 07/09/2024] Open
Abstract
Globally, lung cancer stands as the leading type of cancer in terms of incidence and is the major source of mortality attributed to cancer. We have outlined the molecular biomarkers for lung cancer that are available clinically. Circulating tumor cells (CTCs) spread from the original location, circulate in the bloodstream, extravasate, and metastasize, forming secondary tumors by invading and establishing a favorable environment. CTC analysis is considered a common liquid biopsy method for lung cancer. We have enumerated both in vivo and ex vivo techniques for CTC separation and enrichment, examined the advantages and limitations of these methods, and also discussed the detection of CTCs in other bodily fluids. We have evaluated the value of CTCs, as well as CTCs in conjunction with other biomarkers, for their utility in the early detection and prognostic assessment of patients with lung cancer. CTCs engage with diverse cells of the metastatic process, interfering with the interaction between CTCs and various cells in metastasis, potentially halting metastasis and enhancing patient prognosis.
Collapse
Affiliation(s)
- Xiaochen Wang
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Lu Bai
- Department of Pathology and Pathophysiology, Inner Mongolia Medical University, Hohhot, Inner Mongolia, China
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Linghui Kong
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| | - Zhijuan Guo
- Department of Pathology, Cancer Hospital Affiliated to Inner Mongolia Medical University / Peking University Cancer Hospital Inner Mongolia Hospital, Hohhot, Inner Mongolia, China
| |
Collapse
|
46
|
Zhang YW, Gvozdenovic A, Aceto N. A Molecular Voyage: Multiomics Insights into Circulating Tumor Cells. Cancer Discov 2024; 14:920-933. [PMID: 38581442 DOI: 10.1158/2159-8290.cd-24-0218] [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: 02/14/2024] [Revised: 03/08/2024] [Accepted: 03/13/2024] [Indexed: 04/08/2024]
Abstract
Circulating tumor cells (CTCs) play a pivotal role in metastasis, the leading cause of cancer-associated death. Recent improvements of CTC isolation tools, coupled with a steady development of multiomics technologies at single-cell resolution, have enabled an extensive exploration of CTC biology, unlocking insights into their molecular profiles. A detailed molecular portrait requires CTC interrogation across various levels encompassing genomic, epigenetic, transcriptomic, proteomic and metabolic features. Here, we review how state-of-the-art multiomics applied to CTCs are shedding light on how cancer spreads. Further, we highlight the potential implications of CTC profiling for clinical applications aimed at enhancing cancer diagnosis and treatment. SIGNIFICANCE Exploring the complexity of cancer progression through cutting-edge multiomics studies holds the promise of uncovering novel aspects of cancer biology and identifying therapeutic vulnerabilities to suppress metastasis.
Collapse
Affiliation(s)
- Yu Wei Zhang
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Ana Gvozdenovic
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| | - Nicola Aceto
- Department of Biology, Institute of Molecular Health Sciences, Swiss Federal Institute of Technology Zurich (ETH Zurich), Zurich, Switzerland
| |
Collapse
|
47
|
Tan T, Hu H, Zhang W, Cui J, Lu Z, Li X, Song J. Novel immune classification based on machine learning of pathological images predicts early recurrence of hepatocellular carcinoma. Front Oncol 2024; 14:1391486. [PMID: 38826785 PMCID: PMC11140080 DOI: 10.3389/fonc.2024.1391486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 05/06/2024] [Indexed: 06/04/2024] Open
Abstract
Introduction Immune infiltration within the tumor microenvironment (TME) plays a significant role in the onset and progression of hepatocellular carcinoma (HCC). Machine learning applied to pathological images offers a practical means to explore the TME at the cellular level. Our former research employed a transfer learning procedure to adapt a convolutional neural network (CNN) model for cell recognition, which could recognize tumor cells, lymphocytes, and stromal cells autonomously and accurately within the images. This study introduces a novel immune classification system based on the modified CNN model. Method Patients with HCC from both Beijing Hospital and The Cancer Genome Atlas (TCGA) database were included in this study. Additionally, least absolute shrinkage and selection operator (LASSO) analyses, along with logistic regression, were utilized to develop a prognostic model. We proposed an immune classification based on the percentage of lymphocytes, with a threshold set at the median lymphocyte percentage. Result Patients were categorized into high or low infiltration subtypes based on whether their lymphocyte percentages were above or below the median, respectively. Patients with different immune infiltration subtypes exhibited varying clinical features and distinct TME characteristics. The low-infiltration subtype showed a higher incidence of hypertension and fatty liver, more advanced tumor stages, downregulated immune-related genes, and higher infiltration of immunosuppressive cells. A reliable prognostic model for predicting early recurrence of HCC based on clinical features and immune classification was established. The area under the curve (AUC) of the receiver operating characteristic (ROC) curves was 0.918 and 0.814 for the training and test sets, respectively. Discussion In conclusion, we proposed a novel immune classification system based on cell information extracted from pathological slices, provides a novel tool for prognostic evaluation in HCC.
Collapse
Affiliation(s)
- Tianhua Tan
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Huijuan Hu
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Wei Zhang
- Department of Pathology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Ju Cui
- The Key Laboratory of Geriatrics, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhenhua Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Gastrointestinal Cancer Center, Ward I, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xuefei Li
- Key Laboratory of Quantitative Synthetic Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, Guangdong, China
| | - Jinghai Song
- Department of General Surgery, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
48
|
Singh A, Liu H, El-Shennawy L. Multi-omic features and clustering phenotypes of circulating tumor cells associated with metastasis and clinical outcomes. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 392:67-100. [PMID: 40287221 DOI: 10.1016/bs.ircmb.2024.03.009] [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: 04/29/2025]
Abstract
Metastasis is a lethal disease of cancer, spreading from primary tumors to the bloodstream as circulating tumor cells (CTCs), which disseminate to distant organs at low efficiency for secondary tumor regeneration, thereby contributing to unfavorable patient outcomes. The detection of dynamic CTC alterations can be indicative of cancer progression (residual cancer, aggressiveness, therapy resistance) or regression (therapy response), serving as biomarkers for diagnoses and prognoses. CTC heterogeneity is impacted by both intrinsic oncogenic changes and extrinsic microenvironmental factors (e.g. the immune system and circadian rhythm), altering the genomic/genetic, epigenomic/epigenetic, proteomic, post-translational, and metabolomic landscapes. In addition to homeostatic dynamics, regenerative stemness, and metabolic plasticity, a newly discovered feature of CTCs that influences metastatic outcomes is its intercellular clustering. While the dogma suggests that CTCs play solo as single cells in the circulation, CTCs can orchestrate with other CTCs or white blood cells to form homotypic or heterotypic multi-cellular clusters, with 20-100 times enhanced metastatic potential than single CTCs. CTC clusters promote cell survival and stemness through DNA hypomethylation and signaling pathways activated by clustering-driving proteins (CD44, CD81, ICAM1, Podocalyxin, etc). Heterotypic CTC clusters may protect CTCs from immune cell attacks if not being cleared by cytotoxic immune cells. This chapter mainly focused on CTC biology related to multi-omic features and metastatic outcomes. We speculate that CTCs could guide therapeutic targeting and be targeted specifically by anti-CTC therapeutics to reduce or eliminate cancer and cancer metastasis.
Collapse
Affiliation(s)
- Anmol Singh
- Department of Pharmacology, Northwestern University, Chicago, IL, United States
| | - Huiping Liu
- Department of Pharmacology, Northwestern University, Chicago, IL, United States; Hematology & Oncology Division, Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL, United States; Lurie Comprehensive Cancer Center, Feinberg School of Medicine, Northwestern University, Chicago, IL, United States.
| | - Lamiaa El-Shennawy
- Department of Pharmacology, Northwestern University, Chicago, IL, United States.
| |
Collapse
|
49
|
Lan Z, Zou K, Cui H, Zhao Y, Yu G. Porphyromonas gingivalis suppresses oral squamous cell carcinoma progression by inhibiting MUC1 expression and remodeling the tumor microenvironment. Mol Oncol 2024; 18:1174-1188. [PMID: 37666495 PMCID: PMC11076995 DOI: 10.1002/1878-0261.13517] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Revised: 06/07/2023] [Accepted: 09/01/2023] [Indexed: 09/06/2023] Open
Abstract
Bacteria are the causative agents of various infectious diseases; however, the anti-tumor effect of some bacterial species has attracted the attention of many scientists. The human oral cavity is inhabited by abundant and diverse bacterial communities and some of these bacterial communities could play a role in tumor suppression. Therefore, it is crucial to find oral bacterial species that show anti-tumor activity on oral cancers. In the present study, we found that a high abundance of Porphyromonas gingivalis, an anaerobic periodontal pathogen, in the tumor microenvironment (TME) was positively associated with the longer survival of patients with oral squamous cell carcinoma (OSCC). An in vitro assay confirmed that P. gingivalis accelerated the death of OSCC cells by inducing cell cycle arrest at the G2/M phase, thus exerting its anti-tumor effect. We also found that P. gingivalis significantly decreased tumor growth in a 4-nitroquinoline-1-oxide-induced in situ OSCC mouse model. The transcriptomics data demonstrated that P. gingivalis suppressed the biosynthesis of mucin O-glycan and other O-glycans, as well as the expression of chemokines. Validation experiments further confirmed the downregulation of mucin-1 (MUC1) and C-X-C motif chemokine 17 (CXCL17) expression by P. gingivalis treatment. Flow cytometry analysis showed that P. gingivalis successfully reversed the immunosuppressive TME, thereby suppressing OSCC growth. In summary, the findings of the present study indicated that the rational use of P. gingivalis could serve as a promising therapeutic strategy for OSCC.
Collapse
Affiliation(s)
- Zhou Lan
- Stomatological Hospital, School of StomatologySouthern Medical UniversityGuangzhouChina
| | - Ke‐Long Zou
- Stomatological Hospital, School of StomatologySouthern Medical UniversityGuangzhouChina
| | - Hao Cui
- Stomatological Hospital, School of StomatologySouthern Medical UniversityGuangzhouChina
| | - Yu‐Yue Zhao
- Stomatological Hospital, School of StomatologySouthern Medical UniversityGuangzhouChina
| | - Guang‐Tao Yu
- Stomatological Hospital, School of StomatologySouthern Medical UniversityGuangzhouChina
| |
Collapse
|
50
|
Du Q, An Q, Zhang J, Liu C, Hu Q. Unravelling immune microenvironment features underlying tumor progression in the single-cell era. Cancer Cell Int 2024; 24:143. [PMID: 38649887 PMCID: PMC11036673 DOI: 10.1186/s12935-024-03335-z] [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: 11/06/2023] [Accepted: 04/18/2024] [Indexed: 04/25/2024] Open
Abstract
The relationship between the immune cell and tumor occurrence and progression remains unclear. Profiling alterations in the tumor immune microenvironment (TIME) at high resolution is crucial to identify factors influencing cancer progression and enhance the effectiveness of immunotherapy. However, traditional sequencing methods, including bulk RNA sequencing, exhibit varying degrees of masking the cellular heterogeneity and immunophenotypic changes observed in early and late-stage tumors. Single-cell RNA sequencing (scRNA-seq) has provided significant and precise TIME landscapes. Consequently, this review has highlighted TIME cellular and molecular changes in tumorigenesis and progression elucidated through recent scRNA-seq studies. Specifically, we have summarized the cellular heterogeneity of TIME at different stages, including early, late, and metastatic stages. Moreover, we have outlined the related variations that may promote tumor occurrence and metastasis in the single-cell era. The widespread applications of scRNA-seq in TIME will comprehensively redefine the understanding of tumor biology and furnish more effective immunotherapy strategies.
Collapse
Affiliation(s)
- Qilian Du
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Qi An
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Jiajun Zhang
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China
| | - Chao Liu
- Department of Radiation Oncology, Peking University First Hospital, Beijing, 100034, China.
| | - Qinyong Hu
- Department of Oncology, Renmin Hospital of Wuhan University, Wuhan, 430060, China.
| |
Collapse
|