1
|
Faa G, Ziranu P, Pretta A, Cau F, Castagnola M, Spanu D, Saba G, D'Agata AP, Tiwari E, Suri JS, Scartozzi M, Saba L. Cancer-associated fibroblasts (CAFs) and plaque-associated fibroblasts (PAFs): Unraveling the cellular crossroads of atherosclerosis and cancer. Biomed Pharmacother 2025; 188:118145. [PMID: 40373629 DOI: 10.1016/j.biopha.2025.118145] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 05/04/2025] [Accepted: 05/05/2025] [Indexed: 05/17/2025] Open
Abstract
Atherosclerosis is a complex process involving various cells and molecules within the atherosclerotic plaque. Recent evidence suggests that plaque-associated fibroblasts (PAFs), also known as atherosclerosis-associated fibroblasts (AAFs), might play a significant role in the development and progression of the disease. The microenvironment of the atherosclerotic plaque, resembling the tumor microenvironment (TME), includes various cellular populations like plaque-associated macrophages (PAMs), plaque-associated neutrophils (PANs), vascular smooth muscle cells (VSMCs), myeloid-derived suppressor cells (MDSCs), and PAFs. Similar to cancer-associated fibroblasts (CAFs) in tumors, PAFs exhibits a wide range of characteristics and functions. Their interactions with endothelial cells, smooth muscle cells, and other stromal cells, including adventitial fibroblast precursors, significantly influence atherosclerosis progression. Moreover, the ability of PAFs to express various markers such as alpha-SMA, Desmin, VEGF, and GFAP, highlights their diverse origins from different precursor cells, including vascular smooth muscle cells, endothelial cells, glial cells of the enteric nervous system, adventitial fibroblast precursors, as well as resident and circulating fibrocytes. This article explores the molecular interactions between PAFs, cells associated with atherosclerosis, and other stromal cells. It further examines the role of PAFs in the development and progression of atherosclerosis, and compares their features with those of CAFs. The research suggests that studying tumor-associated fibroblasts can help understand fibroblast subpopulations in atherosclerotic plaque. Identifying specific subpopulations could provide new insight into atherosclerosis complexity and lead to the development of innovative drugs for medical intervention.
Collapse
Affiliation(s)
- Gavino Faa
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy; Department of Biology, College of Science and Technology, Temple University, Philadelphia, PA, USA
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy.
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Flaviana Cau
- Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Massimo Castagnola
- Laboratory of Proteomics, Centro Europeo di Ricerca sul Cervello, IRCCS Fondazione Santa Lucia, Rome, Italy
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Alessandra Pia D'Agata
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Ekta Tiwari
- Department of Innovation. Global Biomedical Technologies, Inc., Roseville, CA 95661, USA
| | - Jasjit S Suri
- Department of ECE, Idaho State University, Pocatello, ID, 83209, USA; Department of CE, Graphics Era Deemed to be University, Dehradun 248002, India; University Center for Research & Development, Chandigarh University, Mohali, India; Symbiosis Institute of Technology, Nagpur Campus, Symbiosis International (Deemed University), Pune, INDIA; Stroke Diagnostic and Monitoring Division, AtheroPoint, Roseville, CA 95661, USA
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, Cagliari 09042, Italy
| | - Luca Saba
- Department of Medical Sciences and Public Health, Unit of Radiology, University fo Cagliari, Cagliari, Italy
| |
Collapse
|
2
|
Lee YL, Longmore GD, Pathak A. Distinct roles of protrusions and collagen deformation in collective invasion of cancer cell types. Biophys J 2025; 124:1506-1520. [PMID: 40170350 DOI: 10.1016/j.bpj.2025.03.032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/03/2025] Open
Abstract
The breast tumor microenvironment is composed of heterogeneous cell populations, including normal epithelial cells, cancer-associated fibroblasts (CAFs), and tumor cells that lead collective cell invasion. Both leader tumor cells and CAFs are known to play important roles in tumor invasion across the collagen-rich stromal boundary. However, their individual abilities to utilize their cell-intrinsic protrusions and perform force-based collagen remodeling to collectively invade remain unclear. To compare collective invasion phenotypes of leader-like tumor cells and CAFs, we embedded spheroids composed of 4T1 tumor cells or mouse tumor-derived CAF cell lines within 3D collagen gels and analyzed their invasion and collagen deformation. We found that 4T1s undergo greater invasion while generating lower collagen deformation compared with CAFs. Although force-driven collagen deformations are conventionally associated with higher cellular forces and invasion, here 4T1s specifically rely on actin-based protrusions, while CAFs rely on myosin-based contractility for collective invasion. In denser collagen, both cell types slowed their invasion, and selective pharmacological inhibitions show that Arp2/3 is required but myosin-II is dispensable for 4T1 invasion. Furthermore, depletion of CDH3 from 4T1s and DDR2 from CAFs reduces their ability to distinguish between collagen densities. For effective invasion, both cell types reorient and redistribute magnetically prealigned collagen fibers. With heterogeneous cell populations of cocultured CAFs and 4T1s, higher percentage of CAFs impeded invasion while increasing collagen fiber alignment. Overall, our findings demonstrate distinctive mechanisms of collective invasion adopted by 4T1 tumor cells and CAFs, one relying more on protrusions and the other on force-based collagen deformation. These results suggest that individually targeting cellular protrusions or contractility may not be universally applicable for all cell types or collagen densities, and a better cell-type-dependent approach could enhance effectiveness of cancer therapies.
Collapse
Affiliation(s)
- Ye Lim Lee
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri
| | - Gregory D Longmore
- Department of Medicine (Oncology), Washington University in St. Louis, St. Louis, Missouri; ICCE Institute, Washington University in St. Louis, St. Louis, Missouri
| | - Amit Pathak
- Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, Missouri; Department of Mechanical Engineering and Materials Science, Washington University in St. Louis, St. Louis, Missouri.
| |
Collapse
|
3
|
Liao T, Zeng Y, Xu W, Shi X, Shen C, Du Y, Zhang M, Zhang Y, Li L, Ding P, Hu W, Huang Z, Fung MHM, Ji Q, Wang Y, Li S, Wei W. A spatially resolved transcriptome landscape during thyroid cancer progression. Cell Rep Med 2025; 6:102043. [PMID: 40157360 PMCID: PMC12047530 DOI: 10.1016/j.xcrm.2025.102043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 07/03/2024] [Accepted: 03/05/2025] [Indexed: 04/01/2025]
Abstract
Tumor microenvironment (TME) remodeling plays a pivotal role in thyroid cancer progression, yet its spatial dynamics remain unclear. In this study, we integrate spatial transcriptomics and single-cell RNA sequencing to map the TME architecture across para-tumor thyroid (PT) tissue, papillary thyroid cancer (PTC), locally advanced PTC (LPTC), and anaplastic thyroid carcinoma (ATC). Our integrative analysis reveals extensive molecular and cellular heterogeneity during thyroid cancer progression, enabling the identification of three distinct thyrocyte meta-clusters, including TG+IYG+ subpopulation in PT, HLA-DRB1+HLA-DRA+ subpopulation in early cancerous stages, and APOE+APOC1+ subpopulation in late-stage progression. We reveal stage-specific tumor leading edge remodeling and establish high-confidence cell-cell interactions, such as COL8A1-ITHB1 in PTC, LAMB2-ITGB4 in LPTC, and SERPINE1-PLAUR in ATC. Notably, both SERPINE1 expression level and SERPINE1+ fibroblast abundance correlate with malignant progression and prognosis. These findings provide a spatially resolved framework of TME remodeling, offering insights for thyroid cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Tian Liao
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yu Zeng
- Precision Research Center for Refractory Diseases, Shanghai Jiao Tong University Pioneer Research Institute for Molecular and Cell Therapies, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China; State Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Weibo Xu
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xiao Shi
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Cenkai Shen
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yuxin Du
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Meng Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Yan Zhang
- Department of Pathology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China; Institute of Pathology, Fudan University, Shanghai 200032, China
| | - Ling Li
- Fudan University Shanghai Cancer Center and Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Peipei Ding
- Fudan University Shanghai Cancer Center and Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Weiguo Hu
- Fudan University Shanghai Cancer Center and Institute of Biomedical Sciences, Shanghai Medical College, Fudan University, Shanghai 200032, China; Key Laboratory of Breast Cancer in Shanghai, Fudan University Shanghai Cancer Center, Fudan University, Shanghai 200032, China
| | - Zhiheng Huang
- Endocrine Surgery Division, The University of HongKong-Shenzhen Hospital, Shenzhen, Guangdong 518053, China
| | - Man Him Matrix Fung
- Division of Endocrine Surgery, Department of Surgery, Li Ka Shing Faculty of Medicine, University of Hong Kong Queen Mary Hospital, Hong Kong SAR 999077, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Shengli Li
- Precision Research Center for Refractory Diseases, Shanghai Jiao Tong University Pioneer Research Institute for Molecular and Cell Therapies, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 201620, China; State Key Laboratory of Innovative Immunotherapy, School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai 200240, China.
| | - Wenjun Wei
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
4
|
Khan A, Alzahrani HA, Felemban SG, Algarni AS, Alenezi ABS, Kamal M, Rehman ZU, Asdaq SMB, Ahmed N, Alharbi BM, Alanazi BS, Imran M. Exploring TGF-β signaling in benign prostatic hyperplasia: from cellular senescence to fibrosis and therapeutic implications. Biogerontology 2025; 26:79. [PMID: 40159577 DOI: 10.1007/s10522-025-10226-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/20/2025] [Indexed: 04/02/2025]
Abstract
As men get older, they often develop benign prostatic hyperplasia (BPH), an enlarged prostate that is not cancerous or dangerous. Although the etiology of BPH is unknown, increasing evidence indicates that the TGF-β signaling pathway might be a key player in its pathogenesis. TGF-β is a pleiotropic cytokine involved in proliferation, differentiation, and extracellular matrix re-modeling, which are all dysregulated in BPH. Cellular senescence is primarily initiated by TGF-β--induced, irreversible growth arrest and usually limits the prostate gland's hyperplastic growth. Moreover, senescent cells generate a Senescence-Associated Secretory Phenotype (SASP), which consists of numerous proinflammatory and profibrotic factors that can worsen disease ontogeny. In addition, TGF-β is among the most fibrogenic factors. At the same time, fibrosis involves a massive accumulation of extracellular matrix proteins, which can increase tissue stiffness and a loss of normal organ functions. TGF-β-mediated fibrosis in BPH changes the mechanical properties of the prostate and surrounding tissues to contribute to lower urinary tract symptoms. This review discusses the complicated molecular signaling of TGF-β underlying changes in cellular senescence and fibrosis during BPH concerning its therapeutic potential.
Collapse
Affiliation(s)
- Abida Khan
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia
- Center for Health Research, Northern Border University, Arar, 73213, Saudi Arabia
| | - Hayat Ali Alzahrani
- Medical Laboratory Technology Department, College of Medical Applied Science, Northern Border University, Arar, Saudi Arabia
| | - Shatha Ghazi Felemban
- Medical Laboratory Sciences Department, Fakeeh College for Medical Sciences, 21461, Jeddah, Saudi Arabia
| | - Alanood Saeed Algarni
- Department of Pharmacology and Toxicology, College of Pharmacy, Umm Al-Qura University, Makkah, Saudi Arabia
| | | | - Mehnaz Kamal
- Department of Pharmaceutical Chemistry, College of Pharmacy, Prince Sattam Bin Abdulaziz University, 11942, Al-Kharj, Saudi Arabia
| | - Zia Ur Rehman
- Health Research Centre, Jazan University, P.O. Box 114, 45142, Jazan, Saudi Arabia
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Jazan University, P.O. Box 114, Jazan, 45142, Kingdom of Saudi Arabia
| | | | - Naveed Ahmed
- Department of Assistance Medical Sciences, Applied College, University of Tabuk, 71491, Tabuk, Saudi Arabia
| | - Bashayer Mohammed Alharbi
- Department of Pharmacy, Johns Hopkins Aramco Healthcare, P.O. Box 10352, 31311, Dhahran, Eastern Province, Saudi Arabia
| | - Bander Sharqi Alanazi
- Department of Nursing Administration, Northern Area Armed Forces Hospital, 31991, Hafer AlBaten, Saudi Arabia
| | - Mohd Imran
- Department of Pharmaceutical Chemistry, College of Pharmacy, Northern Border University, 91911, Rafha, Saudi Arabia.
- Center for Health Research, Northern Border University, Arar, 73213, Saudi Arabia.
| |
Collapse
|
5
|
Ramadan DR, Osman HA, Madhy SA, Teleb M, Darwish AI, Abu-Serie MM, Haiba NS, Khattab SN, Khalil HH. A tailored 4G s-triazine-based dendrimer vehicle for quercetin endowed with MMP-2/9 inhibition and VEGF downregulation for targeting breast cancer progression and liver metastasis. RSC Adv 2025; 15:10426-10441. [PMID: 40182507 PMCID: PMC11967334 DOI: 10.1039/d5ra01588j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2025] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
Motivated by our recent research progress on the exploitation of s-triazine dendritic platforms as bioactive carriers for well-known anticancer agents and/or targeting ligands, we set out to synthesize new rationally designed dendrimers endowed with MMP-2/9 inhibition potential for halting both breast and liver cancer progression with reduced off-target side effects. New three and four generation s-triazine based dendrimers were developed to incorporate potential ZBGs (Zinc Binding Groups) and carboxyl terminal groups to facilitate direct conjugation of anti-cancer drugs (quercetin) and/or targeting ligands (lactobionic acid) through a biodegradable ester bond. Compared to free quercetin (QUR), MTT assay revealed that all the quercetin-coupled dendrimers displayed better anticancer potential (IC50 = 12.690-29.316, 4.137-29.090 μM) against MCF-7 and HepG-2 cancer cells, respectively within their safe doses (EC100 = 134.35-78.44 μM). Conjugation of lactobionic acid and PEG boosted the anticancer potency against both treated cells, improved apoptosis and down regulated MMP-9 and VEGF gene expression levels in both treated cancer cells. Generally, the more branched G4 dendrimer conjugates exhibited a superior overall anticancer performance compared to their respective G3 analogues, except for their MMP-9 inhibition where G3 conjugate appeared to be more potent and more selective than its G4 analogue.
Collapse
Affiliation(s)
- Doaa R Ramadan
- Chemistry Department, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| | - Heba A Osman
- Department of Physics and Chemistry, Faculty of Education, Alexandria University Alexandria Egypt
| | - Somaya Aly Madhy
- Chemistry Department, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| | - Mohamed Teleb
- Department of Pharmaceutical Chemistry, Faculty of Pharmacy, Alexandria University Alexandria 21521 Egypt
- Cancer Nanotechnology Research Laboratory (CNRL), Faculty of Pharmacy, Alexandria University Alexandria 21521 Egypt
| | - A I Darwish
- Department of Physics and Chemistry, Faculty of Education, Alexandria University Alexandria Egypt
| | - Marwa M Abu-Serie
- Medical Biotechnology Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications (SRTA-City) Alexandria Egypt
| | - Nesreen S Haiba
- Department of Physics and Chemistry, Faculty of Education, Alexandria University Alexandria Egypt
| | - Sherine N Khattab
- Chemistry Department, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| | - Hosam H Khalil
- Chemistry Department, Faculty of Science, Alexandria University Alexandria 21321 Egypt
| |
Collapse
|
6
|
Ismailov A, Spallone A, Belogurov A, Herbert A, Poptsova M. Molecular biology of the deadliest cancer - glioblastoma: what do we know? Front Immunol 2025; 16:1530305. [PMID: 40191211 PMCID: PMC11968700 DOI: 10.3389/fimmu.2025.1530305] [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: 11/18/2024] [Accepted: 03/07/2025] [Indexed: 04/09/2025] Open
Abstract
Glioblastomas are the most prevalent primary brain tumors and are associated with a dramatically poor prognosis. Despite an intensive treatment approach, including maximal surgical tumor removal followed by radio- and chemotherapy, the median survival for glioblastoma patients has remained around 18 months for decades. Glioblastoma is distinguished by its highly complex mechanisms of immune evasion and pronounced heterogeneity. This variability is apparent both within the tumor itself, which can exhibit multiple phenotypes simultaneously, and in its surrounding microenvironment. Another key feature of glioblastoma is its "cold" microenvironment, characterized by robust immunosuppression. Recent advances in single-cell RNA sequencing have uncovered new promising insights, revealing previously unrecognized aspects of this tumor. In this review, we consolidate current knowledge on glioblastoma cells and its microenvironment, with an emphasis on their biological properties and unique patterns of molecular communication through signaling pathways. The evidence underscores the critical need for personalized poly-immunotherapy and other approaches to overcome the plasticity of glioblastoma stem cells. Analyzing the tumor microenvironment of individual patients using single-cell transcriptomics and implementing a customized immunotherapeutic strategy could potentially improve survival outcomes for those facing this formidable disease.
Collapse
Affiliation(s)
- Aly Ismailov
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
| | - Aldo Spallone
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
- Laboratory of Hormonal Regulation Proteins, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
| | - Alexey Belogurov
- Laboratory of Hormonal Regulation Proteins, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry Russian Academy of Sciences (RAS), Moscow, Russia
- Scientific and Educational Institute of Fundamental Medicine named after V.I. Pokrovsky, Department of Biological Chemistry, Russian University of Medicine, Moscow, Russia
| | - Alan Herbert
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
- Discovery Department, InsideOutBio, Boston, MA, United States
| | - Maria Poptsova
- International Laboratory of Bioinformatics, Institute of Artificial Intelligence and Digital Sciences, Faculty of Computer Science, National Research University Higher School of Economics, Moscow, Russia
| |
Collapse
|
7
|
Yang M, Nebozhyn MV, Schell MJ, Gandhi N, Pflieger L, Loboda A, Pledger WJ, Soundararajan R, Maurin M, Wang H, Silva JR, Alden A, Coppola D, Elliott A, Sledge G, Khushman M, Lou E, Goel S, Yeatman TJ. Identifying distinct prognostic and predictive contributions of tumor epithelium versus tumor microenvironment in colorectal cancer. BMC Cancer 2025; 25:441. [PMID: 40075322 PMCID: PMC11899100 DOI: 10.1186/s12885-025-13829-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 02/27/2025] [Indexed: 03/14/2025] Open
Abstract
BACKGROUND Accumulating evidence has suggested that cancer progression and therapeutic response depend on both tumor epithelium (EPI) and tumor microenvironment (TME). However, the dependency of clinical outcomes on the tumor EPI vs. the TME has neither been clearly defined nor quantified. METHODS We classified 2373 colorectal cancer (CRC) tumors into the consensus molecular subtypes (CMS1-4) and generated the 10-gene TMES and the 10-gene EPIS signatures as the serendipitous derivatives of the most (positively vs. negatively) correlated genes of a highly-prognostic, ~ 500-gene signature we previously identified. Distinct TME vs. EPI cellular features of the signature genes were identified by CIBERSORT deconvolution and validated by scRNASEQ in an independent public dataset. RESULTS The TMES signature was strongly associated with the immune/stromal TME-rich CMS1/CMS4 subtypes that portended worse survival, whereas the EPIS signature was predominantly related to the TME-poor, epithelial CMS2/CMS3 classes that portended better survival. Multivariable Cox regression analysis against 29 TME-related signatures revealed that the TMES signature was the most strikingly impacted by the "Cancer-associated fibroblasts" signature (HR: 10.87 vs. 0.13, both P < 0.0001). Moreover, the TMES score was strongly correlated with EMT, SRC activation and MEK inhibitor resistance in 2373 CRC tumors (Spearman r = 0.727, 0.802, 0.824, respectively), which was validated in two independent CRC datasets (n = 626 and n = 566). By contrast, the EPIS score was the dominant force in associating with longer progression free survival in cetuximab-treated metastatic CRC patients derived from two independent clinical trials (Logrank trend P = 0.0005/n = 80; P = 0.0013/n = 44). This finding was further validated in a large real-world clinico-genomics dataset with EGFR inhibitor therapy, which demonstrated that higher EPIS scores were associated with increased overall survival (EGFRi, Logrank trend P < 0.0001/n = 2343) and time on treatment (cetuximab, P = 0.003/n = 953; panitumumab, P < 0.0001/n = 1307). CONCLUSIONS Here we identified a pair of new, distinct 10-gene signatures (the EPIS vs. the TMES) capable of distinguishing the cellular contribution of the tumor EPI vs. the TME in determining CRC prognosis and therapeutic outcomes. With targeted approaches emerging to address both tumor epithelial cells and the TME, the EPIS vs. TMES signature scores may have a novel biomarker role to permit optimization of CRC therapy by identifying sensitive vs. resistant subpopulations.
Collapse
Affiliation(s)
- Mingli Yang
- Department of Surgery, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA.
| | - Michael V Nebozhyn
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - Michael J Schell
- Department of Biostatistics and Bioinformatics, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Nishant Gandhi
- Medical Affairs, Caris Life Sciences, 4610 S 44th Pl, Phoenix, AZ, 85040, USA
| | - Lance Pflieger
- Phenome Health, 401 Terry Ave N, Seattle, WA, 98109, USA
| | - Andrey Loboda
- Merck Research Laboratories, 33 Avenue Louis Pasteur, Boston, MA, 02115, USA
| | - W Jack Pledger
- Department of Molecular Medicine, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL, 33612, USA
- Tampa General Hospital Cancer Institute, 1 Tampa General Circle, Tampa, FL, 33606, USA
| | - Ramani Soundararajan
- Department of Surgery, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA
| | - Michelle Maurin
- Department of Surgery, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA
| | - Heiman Wang
- Department of Surgery, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA
| | - Jetsen Rodriguez Silva
- Department of Surgery, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA
| | - Ashley Alden
- Department of Surgery, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA
| | - Domenico Coppola
- Department of Pathology, Florida Digestive Health Specialists, 10920 Technology Ter, Lakewood Ranch, FL, 34202, USA
- Department of Pathology, Moffitt Cancer Center & Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Andrew Elliott
- Medical Affairs, Caris Life Sciences, 4610 S 44th Pl, Phoenix, AZ, 85040, USA
| | - George Sledge
- Medical Affairs, Caris Life Sciences, 4610 S 44th Pl, Phoenix, AZ, 85040, USA
| | - Moh'd Khushman
- Division of Medical Oncology, Department of Medicine, Washington University, 4590 Nash Way, St. Louis, MO, 63110, USA
| | - Emil Lou
- Division of Hematology, Oncology and Transplantation, Department of Medicine and Masonic Cancer Center, University of Minnesota, 420 Delaware Street SE, Minneapolis, MN, 55455, USA
| | - Sanjay Goel
- Department of Medical Oncology, Rutgers Cancer Institute of New Jersey, Robert Wood Johnson School of Medicine, 195 Little Albany Street, New Brunswick, NJ, 08903, USA
| | - Timothy J Yeatman
- Department of Surgery, University of South Florida, 560 Channelside Drive, Tampa, FL, 33602, USA.
- Department of Molecular Medicine, University of South Florida, 12901 Bruce B. Downs Boulevard, Tampa, FL, 33612, USA.
- Tampa General Hospital Cancer Institute, 1 Tampa General Circle, Tampa, FL, 33606, USA.
| |
Collapse
|
8
|
Gao Y, Luo C, Yang H, Xie Q, He H, Li J, Miao J. Enhanced efficacy of dual chimeric antigen receptor-T cells targeting programmed death-ligand 1 and cancer-associated fibroblasts in colorectal cancer in vitro. Cytojournal 2025; 22:29. [PMID: 40260068 PMCID: PMC12010817 DOI: 10.25259/cytojournal_245_2024] [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: 12/02/2024] [Accepted: 01/17/2025] [Indexed: 04/23/2025] Open
Abstract
Objective Colorectal cancer (CRC) presents significant treatment challenges, including immune evasion and tumor microenvironment (TME) suppression. Chimeric antigen receptor (CAR) T-cell therapy has shown promise in hematologic malignancies, but its effectiveness against solid tumors is hampered by the detrimental effects of the TME. This article aims to explore the potential of bispecific CAR T cells targeting programmed death-ligand 1 (PD-L1) and cancer-associated fibroblasts (CAFs) in CRC treatment. Material and Methods Dual-targeted CAR-T cells against PD-L1 and CAF were engineered using the GV400 lentiviral vector. Programmed death-1 (PD-1)/nanobody (Nb) and fibroblast activation protein (FAP)/Nb-encoding lentiviral vectors were generated, and CAR T cells were produced through a three-plasmid system in 293T cells. Human peripheral blood mononuclear cells (PBMCs) were separated, transduced with these vectors, and then expanded. Functional characterization of CAR-T cells was performed through enzyme-linked immunosorbent assay (ELISA), Western blot analysis, flow cytometry, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assays, and cell counting kit-8 (CCK-8) assay. Migration and invasion assays were conducted using Transwell chambers to assess the ability of FAP-PD-1/Nb CAR-T cells to migrate toward tumor cells and invade the extracellular matrix. Results We developed dual-targeted CAR-T cells incorporating PD-L1 and CAF Nbs, which continuously secreted PD-1/Nb. Western blot confirmed PD-1/Nb expression in PD-1/Nb and FAP-PD-1/Nb CAR-T cells, with no expression in the untreated (UTD) group (P < 0.01). Flow cytometry showed a significantly higher cluster of differentiation (CD)25 and CD69 expression in FAP-PD-1/Nb CAR-T cells upon stimulation with FAP-positive target cells compared with the other groups (P < 0.01). TUNEL, flow cytometry, and CCK-8 assays revealed that FAP-PD-1/Nb CAR-T cells exhibited superior cytotoxicity and proliferation inhibition against FAP-positive HCT116 cells (P < 0.01). ELISA demonstrated increased interferon-gamma and tumor necrosis factor-alpha levels and reduced interleukin-10 (P < 0.01), suggesting enhanced cytokine modulation and antitumor immunity. Compared with single-target CAR-T cells and UTD, FAP-PD-1/Nb CAR-T cells showed notably enhanced Matrigel penetration and invasion (P < 0.01). Safety tests confirmed minimal cytotoxicity to normal PBMCs, indicating favorable safety. Conclusion This study successfully developed dual-targeted CAR-T cells against PD-L1 and CAF and demonstrated their superior antitumor activity and immunomodulatory effects on CRC treatment. This novel therapeutic strategy was established using CAR T-cell technology for the treatment of CRC.
Collapse
Affiliation(s)
- Yang Gao
- Health Management Center, Zigong Fourth People’s Hospital, Zigong, Sichuan Province, China
| | - CanJing Luo
- Health Management Center, Zigong Fourth People’s Hospital, Zigong, Sichuan Province, China
| | - Hua Yang
- Department of General Surgery, Zigong Fourth People’s Hospital, Zigong, Sichuan Province, China
| | - QiaoJin Xie
- Health Management Center, Zigong Fourth People’s Hospital, Zigong, Sichuan Province, China
| | - HaoJie He
- Health Management Center, Zigong Fourth People’s Hospital, Zigong, Sichuan Province, China
| | - JiaWei Li
- Department of Oncology, Zigong Fourth People’s Hospital, Zigong, Sichuan Province, China
| | - JiDong Miao
- Department of Oncology, Zigong Fourth People’s Hospital, Zigong, Sichuan Province, China
| |
Collapse
|
9
|
Handschin C, Shalhoub H, Mazet A, Guyon C, Dusserre N, Boutet-Robinet E, Oliveira H, Guillermet-Guibert J. Biotechnological advances in 3D modeling of cancer initiation. Examples from pancreatic cancer research and beyond. Biofabrication 2025; 17:022008. [PMID: 40018875 DOI: 10.1088/1758-5090/adb51c] [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: 07/19/2024] [Accepted: 02/12/2025] [Indexed: 03/01/2025]
Abstract
In recent years, biofabrication technologies have garnered significant attention within the scientific community for their potential to create advancedin vitrocancer models. While these technologies have been predominantly applied to model advanced stages of cancer, there exists a pressing need to develop pertinent, reproducible, and sensitive 3D models that mimic cancer initiation lesions within their native tissue microenvironment. Such models hold profound relevance for comprehending the intricacies of cancer initiation, to devise novel strategies for early intervention, and/or to conduct sophisticated toxicology assessments of putative carcinogens. Here, we will explain the pivotal factors that must be faithfully recapitulated when constructing these models, with a specific focus on early pancreatic cancer lesions. By synthesizing the current state of research in this field, we will provide insights into recent advances and breakthroughs. Additionally, we will delineate the key technological and biological challenges that necessitate resolution in future endeavors, thereby paving the way for more accurate and insightfulin vitrocancer initiation models.
Collapse
Affiliation(s)
- C Handschin
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - H Shalhoub
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
| | - A Mazet
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - C Guyon
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - N Dusserre
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - E Boutet-Robinet
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| | - H Oliveira
- Université de Bordeaux, Tissue Bioengineering - BioTis, INSERM U1026, Bordeaux, F-33000, France
- INSERM U1026, ART BioPrint, F-33000 Bordeaux, France
| | - J Guillermet-Guibert
- CRCT, Université de Toulouse, Inserm, CNRS, Centre de Recherches en Cancérologie de Toulouse, 2 av Hubert Curien, Toulouse, France
- Labex Toucan, 2 av Hubert Curien, Toulouse, France
- Toxalim (Research Centre in Food Toxicology), Université de Toulouse, INRAE, ENVT, INP-Purpan, UT3, Toulouse, France
| |
Collapse
|
10
|
Esmaili M, Jafari N, Ahmadzadeh F, Toosi SMV, Abediankenari S. Effect of conditioned medium from miRNA-34a transfected gastric cancer-associated fibroblast on peripheral blood mononuclear cells. BMC Immunol 2025; 26:9. [PMID: 40000950 PMCID: PMC11854116 DOI: 10.1186/s12865-025-00688-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2024] [Accepted: 02/12/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND Cancer-associated fibroblast (CAF) cells play an important role in gastric malignancy. MiRNA dysregulation has been detected in CAF cells, which is related to the tumor progression ability of these cells. Therefore, this study aimed to evaluate the function of miRNA34a in CAF cells in gastric carcinoma. METHOD We transiently transfected miRNA-34a mimic in CAF cells and examined the effect of the overexpressed miRNA on PD-L1 expression using real-time PCR. Next, we evaluated the role of transfected CAF-conditioned medium (CM) on the immune response and viability of gastric cancer cell lines. RESULTS We have shown that miRNA-34a significantly reduced PD-L1 expression in CAF cells (p < 0.05). However, the conditioned medium of transfected cells had no significant effect on the immune response. We also found that CM of miRNA-34a transfected CAF cells significantly suppressed gastric cancer cell line viability relative to the control group (P < 0.05). CONCLUSION We indicated that CM of miRNA-34a transfected CAF can reduce gastric cancer cell line proliferation. Additionally, miRNA-34a in these cells may improve immune response via PD-L1 reduction. Thus, miRNA-34a could be a potential therapeutic agent in gastric cancer treatment. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Mozhgan Esmaili
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Narjes Jafari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | - Fatemeh Ahmadzadeh
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran
| | | | - Saeid Abediankenari
- Immunogenetics Research Center, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| |
Collapse
|
11
|
Yu K, Zhang S, Shen J, Yu M, Su Y, Wang Y, Zhou K, Liu L, Chen X. Integrating Hypoxia Signatures from scRNA-seq and Bulk Transcriptomes for Prognosis Prediction and Precision Therapy in Cervical Squamous Cell Carcinoma and Endocervical Adenocarcinoma. Int J Mol Sci 2025; 26:1362. [PMID: 39941131 PMCID: PMC11818358 DOI: 10.3390/ijms26031362] [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: 12/20/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Hypoxia, a common feature in many malignancies, is particularly prominent in cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC). Investigating the mechanisms underlying hypoxia is essential for understanding the heterogeneity of CESC and developing personalized therapeutic regimens. Firstly, the CESC-specific hypoxia gene sets shared between single-cell RNA sequencing (scRNA-seq) and bulk data were identified through Weighted Gene Correlation Network Analysis (WGCNA)and FindMarkers analyses. A CESC-specific hypoxia-related score (CSHRS) risk model was constructed using the least absolute shrinkage and selection operator (LASSO)and Cox regression analyses based on these genes. The prognostic differences were analyzed in terms of immune infiltration, mutations, and drug resistance. Finally, a nomogram model was constructed by integrating clinicopathological features to facilitate precision treatment for CESC. This study constructed a CSHRS risk model that divides patients into two groups, and this model can comprehensively evaluate the tumor microenvironment characteristics of CESC, provide accurate prognostic predictions, and offer rational treatment options for patients.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lei Liu
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (K.Y.); (S.Z.); (J.S.); (M.Y.); (Y.S.); (Y.W.); (K.Z.)
| | - Xiujie Chen
- Department of Pharmacogenomics, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China; (K.Y.); (S.Z.); (J.S.); (M.Y.); (Y.S.); (Y.W.); (K.Z.)
| |
Collapse
|
12
|
Zhang X, Li P, Gan Y, Xiang S, Gu L, Zhou J, Zhou X, Wu P, Zhang B, Deng D. Driving effect of P16 methylation on telomerase reverse transcriptase-mediated immortalization and transformation of normal human fibroblasts. Chin Med J (Engl) 2025; 138:332-342. [PMID: 38420748 PMCID: PMC11771662 DOI: 10.1097/cm9.0000000000003004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Indexed: 03/02/2024] Open
Abstract
BACKGROUND P16 inactivation is frequently accompanied by telomerase reverse transcriptase ( TERT ) amplification in human cancer genomes. P16 inactivation by DNA methylation often occurs automatically during immortalization of normal cells by TERT . However, direct evidence remains to be obtained to support the causal effect of epigenetic changes, such as P16 methylation, on cancer development. This study aimed to provide experimental evidence that P16 methylation directly drives cancer development. METHODS A zinc finger protein-based P16 -specific DNA methyltransferase (P16-Dnmt) vector containing a "Tet-On" switch was used to induce extensive methylation of P16 CpG islands in normal human fibroblast CCD-18Co cells. Battery assays were used to evaluate cell immortalization and transformation throughout their lifespan. Cell subcloning and DNA barcoding were used to track the diversity of cell evolution. RESULTS Leaking P16-Dnmt expression (without doxycycline-induction) could specifically inactivate P16 expression by DNA methylation. P16 methylation only promoted proliferation and prolonged lifespan but did not induce immortalization of CCD-18Co cells. Notably, cell immortalization, loss of contact inhibition, and anchorage-independent growth were always prevalent in P16-Dnmt&TERT cells, indicating cell transformation. In contrast, almost all TERT cells died in the replicative crisis. Only a few TERT cells recovered from the crisis, in which spontaneous P16 inactivation by DNA methylation occurred. Furthermore, the subclone formation capacity of P16-Dnmt&TERT cells was two-fold that of TERT cells. DNA barcoding analysis showed that the diversity of the P16-Dnmt&TERT cell population was much greater than that of the TERT cell population. CONCLUSION P16 methylation drives TERT -mediated immortalization and transformation of normal human cells that may contribute to cancer development.
Collapse
Affiliation(s)
- Xuehong Zhang
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Paiyun Li
- Division of Etiology, Beijing Cancer Hospital, Beijing 100142, China
- Radiation Oncology Department, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Ying Gan
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Shengyan Xiang
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Liankun Gu
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Jing Zhou
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| | - Xiaorui Zhou
- Department of Biomedical Engineering, Peking University Cancer Hospital and Institute, Beijing 100871, China
| | - Peihuang Wu
- Department of Biomedical Engineering, Peking University Cancer Hospital and Institute, Beijing 100871, China
| | - Baozhen Zhang
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
- Division of Etiology, Beijing Cancer Hospital, Beijing 100142, China
| | - Dajun Deng
- Key Laboratory of Carcinogenesis and Translational Research (MOE/Beijing), Division of Etiology, Peking University Cancer Hospital and Institute, Beijing 100142, China
| |
Collapse
|
13
|
Yagi T, Kagawa S, Nogi S, Taniguchi A, Yoshimoto M, Suemori K, Nagai Y, Fujita S, Kuroda S, Kikuchi S, Kakiuchi Y, Teraishi F, Takagi K, Ohara T, Tazawa H, Fujiwara T. Cancer-associated fibroblasts promote pro-tumor functions of neutrophils in pancreatic cancer via IL-8: potential suppression by pirfenidone. Cancer Immunol Immunother 2025; 74:96. [PMID: 39904796 PMCID: PMC11794937 DOI: 10.1007/s00262-025-03946-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
BACKGROUND The mechanisms by which neutrophils acquire pro-tumor properties remain poorly understood. In pancreatic cancer, cancer-associated fibroblasts (CAFs) may interact with neutrophils, directing them to promote tumor progression. METHODS To validate the association between CAFs and neutrophils, the localization of neutrophils was examined in clinically resected pancreatic cancer specimens. CAFs were produced by culturing in cancer-conditioned media, and the effects of these CAFs on neutrophils were examined. In vitro migration and invasion assays assess the effect of CAF-activated neutrophils on cancer cells. The factors secreted by the activated neutrophils were also explored. Finally, pirfenidone (PFD) was tested to determine whether it could suppress the pro-tumor functions of activated neutrophils. RESULTS In pancreatic cancer specimens, neutrophils tended to co-localize with IL-6-positive CAFs. Neutrophils co-cultured with CAFs increased migratory capacity and prolonged life span. CAF-affected neutrophils enhance the migratory and invasive activities of pancreatic cancer cells. IL-8 is the most upregulated cytokine secreted by the neutrophils. PFD suppresses IL-8 secretion from CAF-stimulated neutrophils and mitigates the malignant traits of pancreatic cancer cells. CONCLUSION CAFs activate neutrophils and enhance the malignant phenotype of pancreatic cancer. The interactions between cancer cells, CAFs, and neutrophils can be disrupted by PFD, highlighting a potential therapeutic approach.
Collapse
Affiliation(s)
- Tomohiko Yagi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Shunsuke Kagawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan.
| | - Shohei Nogi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Atsuki Taniguchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Masashi Yoshimoto
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Kanto Suemori
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Yasuo Nagai
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Shuto Fujita
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Shinji Kuroda
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama, Japan
| | - Satoru Kikuchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Yoshihiko Kakiuchi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama, Japan
| | - Fuminori Teraishi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Minimally Invasive Therapy Center, Okayama University Hospital, Okayama, Japan
| | - Kosei Takagi
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| | - Toshiaki Ohara
- Departments of Pathology and Experimental Medicine, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, Okayama, Japan
| | - Hiroshi Tazawa
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
- Center for Innovative Clinical Medicine, Okayama University Hospital, Okayama, Japan
| | - Toshiyoshi Fujiwara
- Department of Gastroenterological Surgery, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 2-5-1 Shikata-Cho, Kita-Ku, Okayama, 700-8558, Japan
| |
Collapse
|
14
|
Liu Z, Zhang C, Xiao J, He Y, Liang H, Huang J, Cai Z, Yi Z, Chen M, Li Y, Zhang J, liu F, Ren P, Li H, Chen J, Fan B, Hu J, Zu X, Deng D. TBX3 shapes an immunosuppressive microenvironment and induces immunotherapy resistance. Theranostics 2025; 15:1966-1986. [PMID: 39897553 PMCID: PMC11780534 DOI: 10.7150/thno.103175] [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: 09/03/2024] [Accepted: 12/28/2024] [Indexed: 02/04/2025] Open
Abstract
Background: Identifying biomarkers that predict immunotherapy efficacy and discovering new targets for combination therapies are critical elements for improving the prognosis of bladder cancer (BLCA) patients. Methods: Firstly, we explored the expression patterns of TBX3 in normal and pan-cancer tissues and the correlation between TBX3 and the immune microenvironment using data from multiple public databases. Then, we combined various techniques, including bulk RNA sequencing, single-cell RNA sequencing, high-throughput cytokine arrays, functional experiments, ProcartaPlex multiplex immunoassays and TissueFAXS panoramic tissue quantification assays, to demonstrate that TBX3 shapes an immunosuppressive tumor microenvironment (TME) in BLCA. Results: We identified TBX3 as a key factor associated with the immunosuppressive microenvironment in BLCA through a systematic multi-omics analysis. We found that TBX3 is primarily expressed in malignant cells, where TBX3high tumor cells increase the secretion of TGFβ1, which promotes the infiltration of cancer-associated fibroblasts (CAFs), thereby forming an immunosuppressive microenvironment. We further demonstrated that TBX3 enhances TGFβ1 expression by binding to the TGFβ1 promoter, and blocking TGFβ1 counteracts the immunosuppressive effects of TBX3. Moreover, TBX3 reduced the cancer-killing efficiency of CD8+ T cells by decreasing the proportion of GZMB+ CD8+ T cells, and knocking down TBX3 combined with anti-PD-1 treatment increased CD8+ T cell infiltration and reduced CAFs in vivo. We also validated the inverse relationship between TBX3+ malignant cells and CD8+ T cells and the positive relationship with CAFs in tissue microarrays. Lastly, we found that TBX3 predicted immunotherapy efficacy in our real-world immunotherapy cohort and multiple public cohorts. Conclusion: In summary, TBX3 promotes BLCA progression and immunotherapy resistance by inducing an immunosuppressive microenvironment, and targeting TBX3 could enhance the efficacy of immunotherapy for BLCA.
Collapse
Affiliation(s)
- Zhi Liu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Chunyu Zhang
- Department of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jiatong Xiao
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Yunbo He
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Haisu Liang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Jinliang Huang
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Zhiyong Cai
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Zhenglin Yi
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Mingfeng Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Yixiao Li
- Department of Urology, The second people's Hospital of Hunan province, Changsha, China
| | - Jun Zhang
- Department of Imaging, The first people's Hospital of Kaili city, Kaili, China
| | - Fenglian liu
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Peng Ren
- Department of Urology, The Second Affiliated Hospital of Guizhou Medical University, Kaili, China
| | - Huihuang Li
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Jinbo Chen
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Benyi Fan
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Jiao Hu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| | - Xiongbing Zu
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- Department of Urology, The First Affiliated Hospital of Hunan Normal University, Hunan Normal University, Changsha, China
| | - Dingshan Deng
- Department of Urology, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders and FuRong Laboratory, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
15
|
John DS, Tomar DA, Jain DA, Gupta DS. Molecular insights into hybrid tumors: Exploring the heterogeneity in plexiform ameloblastoma. Semin Diagn Pathol 2025; 42:5-12. [PMID: 39304423 DOI: 10.1053/j.semdp.2024.09.002] [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: 08/31/2024] [Accepted: 09/06/2024] [Indexed: 09/22/2024]
Abstract
For many years, odontogenic tumors have been known to present both clinical and histopathological challenges due to their origins in the epithelial, ectomesenchymal, and/or mesenchymal components of tooth-forming tissues. Gaining a comprehensive understanding of both common and rare odontogenic tumors is crucial for their effective study and clinical management. One particularly puzzling tumor is the "plexiform ameloblastoma," a variant of the solid multicystic ameloblastoma. This term describes a distinct pattern of epithelial proliferation within the cystic cavity. Numerous studies have emphasized the variability of the stromal component, further highlighting the enigmatic nature of ameloblastoma. The presence of unique and rare features, such as primitive, mature desmoplastic, hemangiomatous, or ghost cells within the stroma of plexiform ameloblastoma, underscores the differentiation potential of the neoplastic odontogenic epithelium and offers significant insights into the tissue reactions associated with this condition. This case review discusses four instances of plexiform ameloblastoma, illustrating various atypical stromal changes and their influence on patient prognosis. It also provides important criteria for analyzing stromal alterations related to this complex odontogenic tumor.
Collapse
Affiliation(s)
- Dr Sharon John
- Department of Oral Pathology, King George's Medical University, Lucknow, UP 226003, India
| | - Dr Arushi Tomar
- Department of Oral Pathology, King George's Medical University, Lucknow, UP 226003, India
| | - Dr Ayushi Jain
- Department of Oral Pathology, King George's Medical University, Lucknow, UP 226003, India
| | - Dr Shalini Gupta
- Head of Department of Oral Pathology, King George's Medical University, Lucknow, UP 226003, India.
| |
Collapse
|
16
|
Yong Y, Demmler R, Zohud BA, Fang Q, Zhang T, Zhou Y, Petter K, Flierl C, Gass T, Geppert CI, Merkel S, Schellerer VS, Naschberger E, Stürzl M. AMIGO2 characterizes cancer-associated fibroblasts in metastatic colon cancer and induces the release of paracrine active tumorigenic secretomes. J Pathol 2025; 265:14-25. [PMID: 39523830 PMCID: PMC11638658 DOI: 10.1002/path.6363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/24/2024] [Accepted: 10/01/2024] [Indexed: 11/16/2024]
Abstract
Secretomes of cancer-associated fibroblasts (CAFs) in colorectal cancer (CRC) contribute to malignancy. Detailed knowledge is available on the components and functions of CAF secretomes. Little is known about the regulation of CAF secretomes. Here, we searched for receptor-like membrane-bound molecules in CAFs, which may regulate the production and release of tumor-activating secretomes. The adhesion molecule with Ig-like domain 2 (AMIGO2) was significantly upregulated in cultivated CAFs compared to normal tissue-associated fibroblasts (NAFs), and this was confirmed in patient-derived tissues. AMIGO2 expression was low or absent in healthy colon, significantly increased in fibroblasts of primary CRC, and highest in the stromal tissues of CRC-derived liver metastases. AMIGO2 expression in CAFs correlated with a higher T-category, increased lymph node metastasis, progressed tumor stages and was associated with reduced survival in different cohorts of CRC patients. Interestingly, AMIGO2 expression was induced by transforming growth factor-β and higher in female patients, who exhibit a more aggressive disease course. In functional studies, conditioned media of NAFs with experimentally induced AMIGO2 overexpression enhanced proliferation and migration of different CRC tumor cells, while siRNA-mediated inhibition of AMIGO2 in CAFs attenuated these effects. Accordingly, therapeutic inhibition of the receptor-like AMIGO2 protein in CRC CAFs could prevent tumorigenic secretomes in CRC. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Yongsong Yong
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
- Xinghua People's Hospital Affiliated to Yangzhou UniversityTaizhouPR China
| | - Richard Demmler
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Bisan Abdalfatah Zohud
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Qi Fang
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Tong Zhang
- Xinghua People's Hospital Affiliated to Yangzhou UniversityTaizhouPR China
| | - Yonghua Zhou
- Xinghua People's Hospital Affiliated to Yangzhou UniversityTaizhouPR China
| | - Katja Petter
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Christian Flierl
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Tobias Gass
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Carol I Geppert
- Institute of Pathology, Universitätsklinikum ErlangenFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Susanne Merkel
- Department of Surgery, Universitätsklinikum ErlangenFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
| | - Vera S Schellerer
- Department of Surgery, Universitätsklinikum ErlangenFriedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
- Department of Pediatric SurgeryUniversity Medicine GreifswaldGreifswaldGermany
| | - Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
- CCC Erlangen‐EMN: Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)Universitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA)ErlangenGermany
- BZKF: Bavarian Cancer Research Center (BZKF)ErlangenGermany
| | - Michael Stürzl
- Division of Molecular and Experimental Surgery, Department of SurgeryUniversitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
- CCC Erlangen‐EMN: Comprehensive Cancer Center Erlangen‐EMN (CCC ER‐EMN)Universitätsklinikum Erlangen, Friedrich‐Alexander‐Universität (FAU) Erlangen‐NürnbergErlangenGermany
- CCC WERA: Comprehensive Cancer Center Alliance WERA (CCC WERA)ErlangenGermany
- BZKF: Bavarian Cancer Research Center (BZKF)ErlangenGermany
| |
Collapse
|
17
|
Priya, Kumar A, Kumar D. Molecular heterogeneity and MYC dysregulation in triple-negative breast cancer: genomic advances and therapeutic implications. 3 Biotech 2025; 15:33. [PMID: 39777154 PMCID: PMC11700964 DOI: 10.1007/s13205-024-04195-0] [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: 08/25/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is characterized by a diverse range of molecular features that have been extensively studied. MYC plays a critical role in regulating metabolism, differentiation, proliferation, cell growth, and apoptosis. Dysregulation of MYC is associated with poor prognosis and contributes to the development and progression of breast cancer. A particularly intriguing aspect of TNBC is its association with tumors in BRCA1 mutation carriers, especially in younger women. MYC may also contribute to resistance to adjuvant treatments. For TNBC, targeting MYC-regulated pathways in combination with inhibitors of other carcinogenic pathways offers a promising therapeutic approach. Several signaling pathways regulate TNBC, and targeting these pathways could lead to effective therapeutic strategies for breast cancer. Advances in genomic tools, such as CRISPR-Cas9, next-generation sequencing, and whole-exome sequencing, are revolutionizing breast cancer diagnoses. These technologies have significantly enhanced our understanding of MYC oncogenesis, particularly through CRISPR-Cas9 and NGS. Targeting MYC and its partner MAX could provide valuable insights into TNBC. Moreover, the therapeutic potential of targeting MYC-driven signaling mechanisms and their interactions with other oncogenic pathways, including PI3K/AKT/mTOR and Wnt/β-catenin, is increasingly recognized. Next-generation sequencing and CRISPR-Cas9 represent significant breakthroughs in genomic tools that open new opportunities to explore MYC's role in TNBC and facilitate the development of personalized treatment plans. This review discusses the future clinical applications of personalized treatment strategies for patients with TNBC.
Collapse
Affiliation(s)
- Priya
- School of Health Sciences and Technology (SoHST), UPES, Dehradun, Uttarakhand 248007 India
| | - Arun Kumar
- Mahavir Cancer Sansthan and Research Centre, Patna, Bihar 801505 India
| | - Dhruv Kumar
- School of Health Sciences and Technology (SoHST), UPES, Dehradun, Uttarakhand 248007 India
| |
Collapse
|
18
|
Zhao J, Li Y, Huang Y, Su P, Nie F, Yang P, Yang C. Tumor-Derived GDF15 Induces Tumor Associated Fibroblast Transformation From BMSCs and Fibroblasts in Oral Squamous Cell Carcinoma. J Cell Physiol 2025; 240:e31498. [PMID: 39639678 DOI: 10.1002/jcp.31498] [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: 05/23/2024] [Revised: 11/02/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024]
Abstract
Cancer associated fibroblasts (CAFs) are the predominant stromal cell-type in the solid tumor microenvironment, originating from various cell types and playing a crucial role in promoting tumor progression and metastasis The generation of CAFs is influenced by complex factors secreted by tumor cells, with particular emphasis on transforming growth factor-β (TGF-β). However, it remains largely unknown whether growth/differentiation factor-15 (GDF15), as a member of the TGF-β superfamily, exerts similar effects to TGF-β in oral squamous cell carcinoma (OSCC). In this study, we investigated the impact of GDF15 derived from tumor cells on CAF transformation and elucidated the underlying mechanisms. Exogenous GDF15 and OSCC cells induced the transformation of bone marrow mesenchymal stem cells (BMSCs) and human gingival fibroblasts (HGFs) into CAFs, as evidenced by α-smooth muscle actin (α-SMA) as a phenotypic marker and TGF-β, interleukin 6 (IL-6), and vascular endothelial-derived growth factor (VEGF) as functional markers. Conversely, knockdown of GDF15 in OSCC cells reversed CAF transformation. Mechanistically, extracellular signal-regulated kinases 1/2(ERK1/2) pathway was associated with GDF15-mediated promotion of CAF transformation. Furthermore, OSCC-induced CAFs enhanced migration and invasion abilities of OSCC cells; but this pro-cancer effect was abolished upon knockdown of GDF15 in OSCC cells. Subcutaneous coinjection of OSCC cells with BMSCs or HGFs into mice revealed the promoted tumor growth along with increased expression levels of α-SMA and Ki67 compared with alone OSCC cells injection; these effects were attenuated when GDF15 was knocked down in OSCC cells. Collectively, our findings suggest that tumor-derived GDF15 contributes to the progression of OSCC by promoting CAF transformation through activation of the ERK1/2 pathway.
Collapse
Affiliation(s)
- Jingjing Zhao
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Yahui Li
- Department of Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, China
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Shanghai, China
| | - Yingying Huang
- Department of Stomatology, Huashan Hospital, Fudan University, Shanghai, China
| | - Peng Su
- Department of Pathology, Jinan, China
| | - Fujiao Nie
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Pishan Yang
- Department of Periodontology, School and Hospital of Stomatology, Cheeloo College of Medicine, Shandong University & Shandong Key Laboratory of Oral Tissue Regeneration & Shandong Engineering Research Center of Dental Materials and Oral Tissue Regeneration & Shandong Provincial Clinical Research Center for Oral Diseases, Jinan, China
| | - Chengzhe Yang
- Department of Oral and Maxillofacial Surgery, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
19
|
Kim J, Brunetti B, Kumar A, Mangla A, Honda K, Yoshida A. Inhibition of glutaminase elicits senolysis in therapy-induced senescent melanoma cells. Cell Death Dis 2024; 15:902. [PMID: 39695080 DOI: 10.1038/s41419-024-07284-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/21/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024]
Abstract
The cyclin D1-Cyclin-Dependent Kinases 4 and 6 (CDK4/6) complex is crucial for the development of melanoma. We previously demonstrated that targeting CDK4/6 using small molecule inhibitors (CDK4/6i) suppresses BrafV600E melanoma growth in vitro and in vivo through induction of cellular senescence. However, clinical trials investigating CDK4/6i in melanoma have not yielded successful outcomes, underscoring the necessity to enhance the therapeutic efficacy of CDK4/6i. Accumulated research has shown that while senescence initially suppresses cell proliferation, a prolonged state of senescence eventually leads to tumor relapse by altering the tumor microenvironment, suggesting that removal of those senescent cells (in a process referred to as senolysis) is of clinical necessity to facilitate clinical response. We demonstrate that glutaminase 1 (GLS1) expression is specifically upregulated in CDK4/6i-induced senescent BrafV600E melanoma cells. Upregulated GLS1 expression renders BrafV600E melanoma senescent cells vulnerable to GLS1 inhibitor (GLS1i). Furthermore, we demonstrate that this senolytic approach targeting upregulated GLS1 expression is applicable even though those cells developed resistance to the BrafV600E inhibitor vemurafenib, a frequently encountered substantial clinical challenge to treating patients. Thus, this novel senolytic approach may revolutionize current CDK4/6i mediated melanoma treatment if melanoma cells undergo senescence prior to developing resistance to CDK4/6i. Given that we demonstrate that a low dose of vemurafenib induced senescence, which renders BrafV600E melanoma cells susceptible to GLS1i and recent accumulated research shows many cancer cells undergo senescence in response to chemotherapy, radiation, and immunotherapy, this senolytic therapy approach may prove applicable to a wide range of cancer types once senescence and GLS1 expression are induced.
Collapse
Affiliation(s)
- Justin Kim
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Bryce Brunetti
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Ayanesh Kumar
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Ankit Mangla
- Department of Hematology and Oncology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Kord Honda
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA
| | - Akihiro Yoshida
- Department of Dermatology, Case Western Reserve University and University Hospitals Cleveland Medical Center, Cleveland, OH, 44106, USA.
- Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, 44106, USA.
| |
Collapse
|
20
|
James CD, Lewis RL, Witt AJ, Carter C, Rais NM, Wang X, Bristol ML. Fibroblasts regulate the transcriptional signature of human papillomavirus-positive keratinocytes. Tumour Virus Res 2024; 19:200302. [PMID: 39667669 PMCID: PMC11699615 DOI: 10.1016/j.tvr.2024.200302] [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: 09/16/2024] [Revised: 12/05/2024] [Accepted: 12/08/2024] [Indexed: 12/14/2024] Open
Abstract
Persistent human papillomavirus (HPV) infection is necessary but insufficient for viral oncogenesis. Additional contributing co-factors, such as immune evasion and viral integration have been implicated in HPV-induced cancer progression. It is widely accepted that HPV + keratinocytes require co-culture with fibroblasts to maintain viral DNA as episomes. How fibroblasts regulate viral episome maintenance is a critical knowledge gap. Here we present comprehensive RNA sequencing and proteomic analysis demonstrating that coculture with fibroblasts is supportive of the viral life cycle, and is confirmatory of previous observations. Novel observations suggest that errors in "cross-talk" between fibroblasts and infected keratinocytes may regulate HPV integration and drive oncogenic progression. Our co-culture models offer new insights into HPV-related transformation mechanisms.
Collapse
Affiliation(s)
- Claire D James
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, VA, USA
| | - Rachel L Lewis
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, VA, USA
| | - Austin J Witt
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, VA, USA
| | | | - Nabiha M Rais
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, VA, USA
| | - Xu Wang
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, VA, USA
| | - Molly L Bristol
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, VA, USA; VCU Massey Comprehensive Cancer Center, Richmond, VA, USA.
| |
Collapse
|
21
|
Bod L, Shalapour S. B cells spatial organization defines their phenotype and function in cancer "Tell me with whom you consort, and I will tell you who you are" - Goethe. Curr Opin Immunol 2024; 91:102504. [PMID: 39547092 DOI: 10.1016/j.coi.2024.102504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/15/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
The presence of B cells and their subtypes in the tumor environment has been recognized a for very long time. Immunoglobulins specific for more than thousands of tumor-associated antigens were detected in the sera of patients with cancer; however, antibody-mediated cancer cell killing is usually impaired. The role of humoral immune response remained elusive until recently, with new discoveries regarding their contribution in regulating antitumor immunity, particularly during immunotherapy. Humoral immunity has been described to promote or attenuate tumorigenesis and can have opposing effects on therapeutic outcome in different tumor entities. The antagonism effect of B cells depends on their subtypes and immunoglobulin isotypes and is regulated by their spatial distribution and localization. In this short review, we will focus on how the spatial organization of B cells within the tumor microenvironment, tumor-associated lymph nodes, and tertiary lymphoid structures define their fate and function and contribute to the regulation of antitumor immunity.
Collapse
Affiliation(s)
- Lloyd Bod
- Department of Medicine, Krantz Family Center for Cancer Research, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Shabnam Shalapour
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA.
| |
Collapse
|
22
|
Tobe-Nishimoto A, Morita Y, Nishimura J, Kitahira Y, Takayama S, Kishimoto S, Matsumiya-Matsumoto Y, Matsunaga K, Imai T, Uzawa N. Tumor microenvironment dynamics in oral cancer: unveiling the role of inflammatory cytokines in a syngeneic mouse model. Clin Exp Metastasis 2024; 41:891-908. [PMID: 39126553 PMCID: PMC11607012 DOI: 10.1007/s10585-024-10306-1] [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: 02/20/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
The process of cervical lymph node metastasis is dependent on the phenotype of the tumor cells and their interaction with the host microenvironment and immune system; conventional research methods that focus exclusively on tumor cells are limited in their ability to elucidate the metastatic mechanism. In cancer tissues, a specialized environment called the tumor microenvironment (TME) is established around tumor cells, and inflammation in the TME has been reported to be closely associated with the development and progression of many types of cancer and with the response to anticancer therapy. In this study, to elucidate the mechanism of metastasis establishment, including the TME, in the cervical lymph node metastasis of oral cancer, we established a mouse-derived oral squamous cell carcinoma cervical lymph node highly metastatic cell line and generated a syngeneic orthotopic transplantation mouse model. In the established highly metastatic cells, epithelial-mesenchymal transition (EMT) induction was enhanced compared to that in parental cells. In the syngeneic mouse model, lymph node metastasis was observed more frequently in tumors of highly metastatic cells than in parental cells, and Cyclooxygenase-2 (COX-2) expression and lymphatic vessels in primary tumor tissues were increased, suggesting that this model is highly useful. Moreover, in the established highly metastatic cells, EMT induction was enhanced compared to that in the parent cell line, and CCL5 and IL-6 secreted during inflammation further enhanced EMT induction in cancer cells. This suggests the possibility of a synergistic effect between EMT induction and inflammation. This model, which allows for the use of two types of cells with different metastatic and tumor growth potentials, is very useful for oral cancer research involving the interaction between cancer cells and the TME in tumor tissues and for further searching for new therapeutic agents.
Collapse
Affiliation(s)
- Ayano Tobe-Nishimoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Yoshihiro Morita
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan.
| | - Junya Nishimura
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Yukiko Kitahira
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Shun Takayama
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Satoko Kishimoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Yuka Matsumiya-Matsumoto
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Kazuhide Matsunaga
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Tomoaki Imai
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| | - Narikazu Uzawa
- Department of Oral & Maxillofacial Oncology and Surgery, Osaka University Graduate School of Dentistry, 1-8 Yamadaoka, Suita-shi, Osaka, 565-0871, Japan
| |
Collapse
|
23
|
Verocq C, Noël JC, Charry M, Zindy E, Rorive S, Salmon I, Decaestecker C, Catteau X. Inverse correlation between the amounts of lymphocytic infiltrate and stroma in breast carcinoma. Heliyon 2024; 10:e40295. [PMID: 39641033 PMCID: PMC11617241 DOI: 10.1016/j.heliyon.2024.e40295] [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: 08/07/2023] [Revised: 11/07/2024] [Accepted: 11/07/2024] [Indexed: 12/07/2024] Open
Abstract
Background Previous breast carcinoma studies focused on the evaluation of tumour-infiltrating lymphocytes (TILs) or of tumoural stroma via the tumour stroma ratio (TSR). Few studies assessed peritumoural lymphocytes and almost no studies investigated a possible relationship between lymphocytes and stroma. This prompted us to evaluate the amount of tumour cells, intra- and peritumoural lymphocytes, and stroma in breast cancer to support the hypothesis that the stroma may block the infiltration of lymphocytes inside the tumour. Methods We collected a retrospective series of 158 breast cancers (<25 mm). In addition to standard TILs and TSR evaluations, we assessed the percentages of tumour cells, stromal myofibroblasts, intra- and peritumoural lymphocytes on full-section tumours with haematoxylin and eosin and immunohistochemical staining. Results We showed significant negative correlations between the amounts of stroma and both intra- and peritumoural lymphocyte percentages. Considering the estrogen receptor positive invasive breast cancer of no special type cases, we showed that TSR had a positive prognostic value with an optimal threshold of 10 %. Conclusions This study is one of the first to show inverse correlations between tumoural stroma amount and intra- and peritumoural lymphocyte percentages, which supports the hypothesis that tumoural stroma can prevent the recruitment of lymphocytes around and within the tumour.
Collapse
Affiliation(s)
- Camille Verocq
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
| | - Jean-Christophe Noël
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
| | - Manon Charry
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
| | - Egor Zindy
- DIAPath - Center for Microscopy and Molecular Imaging, ULB, Rue Adrienne Bolland 8, 6041, Gosselies, Belgium
| | - Sandrine Rorive
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
| | - Isabelle Salmon
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
- DIAPath - Center for Microscopy and Molecular Imaging, ULB, Rue Adrienne Bolland 8, 6041, Gosselies, Belgium
| | - Christine Decaestecker
- DIAPath - Center for Microscopy and Molecular Imaging, ULB, Rue Adrienne Bolland 8, 6041, Gosselies, Belgium
- Laboratory of Image Synthesis and Analysis, Ecole Polytechnique de Bruxelles, ULB, 50, Avenue F. Roosevelt, 1050, Brussels, Belgium
| | - Xavier Catteau
- Department of Pathology, Erasme University Hospital, Université Libre de Bruxelles, Route de Lennik 808, 1070, Brussels, Belgium
- CurePath (Centre Universitaire inter Régional d’Expertise en anatomie Pathologique Hospitalière, CHU Tivoli - CHIREC), Rue de Borfilet, 12A, 6040, Jumet, Belgium
| |
Collapse
|
24
|
Zhang S, Wang J, Chen Y, Liang W, Liu H, Du R, Sun Y, Hu C, Shang Z. CAFs-derived lactate enhances the cancer stemness through inhibiting the MST1 ubiquitination degradation in OSCC. Cell Biosci 2024; 14:144. [PMID: 39605072 PMCID: PMC11603751 DOI: 10.1186/s13578-024-01329-y] [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: 08/03/2024] [Accepted: 11/22/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND Cancer-associated fibroblasts (CAFs), a predominant stromal cell type in the tumor microenvironment, significantly affect the progression of oral squamous cell carcinoma (OSCC). RESULTS The specific mechanisms through which CAFs influence the cancer stem cell phenotype in OSCC are not fully understood. This study explored the effects of lactic acid produced by CAFs on the cancer stem cells (CSCs) phenotype of OSCC cells. Our results demonstrated that CAFs exhibit increased glycolysis and lactic acid production. Lactic acid treatment enhances CSCs-related markers expression, sphere formation, and clonogenic ability of OSCC cells. RNA sequencing revealed that lactic acid treatment elevates Discs Large Homolog 5 (DLG5) expression and markedly affects the Hippo pathway. Further investigation revealed that DLG5 mediates the effects of lactic acid on the CSCs phenotype. DLG5 knockdown results in elevated expression of E3 ubiquitin ligase Cullin 3, which can promote the ubiquitination and degradation of MST1, but the expression of phosphorylated MST1 remains unchanged. This leads to enhanced binding of phosphorylated MST1 to YAP1, increasing YAP1 phosphorylation and activating the Hippo pathway. CONCLUSION Collectively, our findings suggest that lactic acid from CAFs promotes the CSCs phenotype in OSCC through the DLG5/CUL3/MST1 axis. Therefore, targeting lactic acid exchange between CAFs and tumor cells may provide a novel therapeutic approach to suppress the CSCs phenotype in OSCC.
Collapse
Affiliation(s)
- Shuzhen Zhang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Central Department School & Hospital of Stomatology, Wuhan University, Wuhan, 430022, China
| | - Jingjing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yang Chen
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Oral and Maxillofacial Surgery, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Weilian Liang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
- Day Surgery Center, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Hanzhe Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ruixue Du
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yunqing Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Chuanyu Hu
- Department of Stomatology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China.
- School of Stomatology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Hubei Province Key Laboratory of Oral and Maxillofacial Development and Regeneration, Wuhan, China.
| | - Zhengjun Shang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral and Maxillofacial-Head and Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
25
|
Liu X, Sun M, Zhang F, Zhang J, Xuanyuan T, Liu W. A heterotypic tumor-on-a-chip platform for user-friendly combinatorial chemotherapeutic testing. Anal Chim Acta 2024; 1330:343278. [PMID: 39489960 DOI: 10.1016/j.aca.2024.343278] [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: 07/22/2024] [Revised: 09/09/2024] [Accepted: 09/24/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Three-dimensional (3D) tumor microdevices are promising platform for biomimetic antitumor prediction and high-throughput chemotherapeutic screening and play crucial roles in the exploration of cancer-associated pharmaceutics and therapeutics. Traditional cell manipulation tools (e.g., non-adhesive surfaces and hanging drops) and recent microengineered systems (e.g., microfluidic chips and micropatterned array chips) have progressed in terms of microscale control, substantial tumor production, programmable drug combinations, and throughput analysis. However, establishing a facile 3D tumor microdevice to construct heterotypic tumor-microenvironmental profiles and for throughput, implementable, multi-instrument-compatible analysis of chemotherapies to advance consumer-grade tumor modelling tools is still being explored. RESULTS In this study, we present a facilely operated tumor-on-a-chip platform for massive production of heterotypic 3D tumors and diverse investigations of combinatorial chemotherapy screening. Large quantity of heterotypic tumor generation with high geometric controllability (size difference: 19.6 μm) and operational repeatability (n = 10) was achieved using simple-to-fabricate micropatterned chips. Multiple characteristics of solid tumors, including phenotypic gradients (viability and proliferation) and heterogeneous cellular compositions (multi-cell participation and stroma composition), were reproduced in heterotypic tumors, being more biomimetic than homotypic tumors. We completed the user-friendly analytical evaluation of individual and combinatorial drug therapies, and demonstrated the high applicability of the platform in biomimetic tumor-related large-scale manipulation and on-chip analysis, as well as its high compatibility for off-chip detection. The entire operative process during tumor production and chemotherapy only requires the routine and easy-to-master pipetting manipulation. SIGNIFICANCE The establishment of a biomimetic and easy-to-use 3D tumor platform and the large-scale screening-like evaluation of combinatorial chemotherapies based on the usage of the micropatterned chip was achieved in a user-friendly manner. This advancement has significant application potential in the fields of oncology, drug discovery, and tissue engineering, and is expected to be valuable for developing accessible and generalizable tumor-on-a-chip microsystems for exploring cancer therapies.
Collapse
Affiliation(s)
- Xufang Liu
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Meilin Sun
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Fen Zhang
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China; Department of Bioengineering, School of Food and Bioengineering, Xuzhou University of Technology, Xuzhou, 221018, China.
| | - Jinwei Zhang
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Tingting Xuanyuan
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China
| | - Wenming Liu
- Departments of Biomedical Engineering and Pathology, School of Basic Medical Science, Central South University, Changsha, Hunan, 410013, China.
| |
Collapse
|
26
|
Ronghe R, Tavares AAS. The skeleton: an overlooked regulator of systemic glucose metabolism in cancer? Front Oncol 2024; 14:1481241. [PMID: 39588310 PMCID: PMC11586348 DOI: 10.3389/fonc.2024.1481241] [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: 08/15/2024] [Accepted: 10/22/2024] [Indexed: 11/27/2024] Open
Abstract
Recent discoveries demonstrated the skeleton's role as an endocrine organ regulating whole-body glucose homeostasis. Glucose metabolism is critical for rapid cell proliferation and tumour growth through increasing glucose uptake and fermentation of glucose to lactate despite being in an aerobic environment. This hypothesis paper discusses emerging evidence on how bones can regulate whole-body glucose homeostasis with potential to impact on tumour growth and proliferation. Moreover, it proposes a clinical link between bone glucose metabolism and prognosis of cancer based on recent clinical trial data. Targeting metabolic pathways related with classic glucose metabolism and also bone metabolism, novel methods of cancer therapy and treatment could be developed. This paper objective is to highlight the need for future research on this altered metabolism with potential to change future management of cancer patients.
Collapse
Affiliation(s)
- Rucha Ronghe
- Edinburgh Medical School, The University of Edinburgh, Edinburgh, United Kingdom
| | - Adriana A. S. Tavares
- University/British Heart Foundation Centre for Cardiovascular Science, The University of Edinburgh, Queens Medical Research Institute, Edinburgh, United Kingdom
- Edinburgh Imaging, The University of Edinburgh, Queens Medical Research Institute, Edinburgh, United Kingdom
| |
Collapse
|
27
|
Yan P, Jimenez ER, Li Z, Bui T, Seehawer M, Nishida J, Foidart P, Stevens LE, Xie Y, Gomez MM, Park SY, Long HW, Polyak K. Midkine as a driver of age-related changes and increase in mammary tumorigenesis. Cancer Cell 2024; 42:1936-1954.e9. [PMID: 39366375 PMCID: PMC11560576 DOI: 10.1016/j.ccell.2024.09.002] [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: 01/17/2024] [Revised: 07/30/2024] [Accepted: 09/11/2024] [Indexed: 10/06/2024]
Abstract
Aging is a pivotal risk factor for cancer, yet the underlying mechanisms remain poorly defined. Here, we explore age-related changes in the rat mammary gland by single-cell multiomics. Our findings include increased epithelial proliferation, loss of luminal identity, and decreased naive B and T cells with age. We discover a luminal progenitor population unique to old rats with profiles reflecting precancerous changes and identify midkine (Mdk) as a gene upregulated with age and a regulator of age-related luminal progenitors. Midkine treatment of young rats mimics age-related changes via activating PI3K-AKT-SREBF1 pathway and promotes nitroso-N-methylurea-induced mammary tumorigenesis. Midkine levels increase with age in human blood and mammary epithelium, and higher MDK in normal breast tissue is associated with higher breast cancer risk in younger women. Our findings reveal a link between aging and susceptibility to tumor initiation and identify midkine as a mediator of age-dependent increase in breast tumorigenesis.
Collapse
Affiliation(s)
- Pengze Yan
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Ernesto Rojas Jimenez
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Zheqi Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Triet Bui
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Marco Seehawer
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Jun Nishida
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Pierre Foidart
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Laura E Stevens
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Yingtian Xie
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Miguel Munoz Gomez
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - So Yeon Park
- Department of Pathology, Seoul National University, Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea
| | - Henry W Long
- Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Kornelia Polyak
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Medicine, Harvard Medical School, Boston, MA 02115, USA; Department of Medicine, Brigham and Women's Hospital, Boston, MA 02115, USA; Center for Functional Cancer Epigenetics, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Pathology, Seoul National University, Bundang Hospital, Seoul National University College of Medicine, Seongnam, Republic of Korea; Harvard Stem Cell Institute, Cambridge, MA 02142, USA.
| |
Collapse
|
28
|
Gallo S, Folco CB, Crepaldi T. The MET Oncogene: An Update on Targeting Strategies. Pharmaceuticals (Basel) 2024; 17:1473. [PMID: 39598385 PMCID: PMC11597589 DOI: 10.3390/ph17111473] [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: 10/10/2024] [Revised: 10/28/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The MET receptor, commonly known as HGF (hepatocyte growth factor) receptor, is a focus of extensive scientific research. MET has been linked to embryonic development, tissue regeneration following injury, tumorigenesis, and cancer metastasis. These functions underscore its involvement in numerous cellular processes, including stemness, proliferation, motility, cell dissociation, and survival. However, the enigmatic nature of MET becomes apparent in the context of cancer. When MET remains persistently activated, since its gene undergoes genetic alterations, it initiates a complex signaling cascade setting in motion an aggressive and metastatic program that is characteristic of malignant cells and is known as "invasive growth". The expanding knowledge of MET signaling has opened up numerous opportunities for therapeutic interventions, particularly in the realm of oncology. Targeting MET presents a promising strategy for developing novel anti-cancer treatments. In this review, we provide an updated overview of drugs designed to modulate MET signaling, highlighting MET kinase inhibitors, degraders, anti-MET/HGF monoclonal antibodies, and MET-targeted antibody-drug conjugates. Through this review, we aim to contribute to the ongoing advancement of therapeutic strategies targeting MET signaling.
Collapse
Affiliation(s)
- Simona Gallo
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Consolata Beatrice Folco
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| | - Tiziana Crepaldi
- Department of Oncology, University of Turin, Regione Gonzole 10, 10143 Orbassano, Italy; (S.G.); (C.B.F.)
- Candiolo Cancer Institute, FPO-IRCCS, SP142, Km 3.95, 10060 Candiolo, Italy
| |
Collapse
|
29
|
Panda SK, Robinson N, Desiderio V. Decoding secret role of mesenchymal stem cells in regulating cancer stem cells and drug resistance. Biochim Biophys Acta Rev Cancer 2024; 1879:189205. [PMID: 39481663 DOI: 10.1016/j.bbcan.2024.189205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/23/2024] [Accepted: 10/22/2024] [Indexed: 11/02/2024]
Abstract
Drug resistance caused by the efflux of chemotherapeutic drugs is one of the most challenging obstacles to successful cancer therapy. Several efflux transporters have been identified since the discovery of the P-gp/ABCB1 transporter in 1976. Over the last four decades, researchers have focused on developing efflux transporter inhibitors to overcome drug resistance. However, even with the third-generation inhibitors available, we are still far from effectively inhibiting the efflux transporters. Additionally, Cancer stem cells (CSCs) pose another significant challenge, contributing to cancer recurrence even after successful treatment. The ability of CSCs to enter dormancy and evade detection makes them almost invulnerable to chemotherapeutic drug treatment. In this review, we discuss how Mesenchymal stem cells (MSCs), one of the key components of the Tumor Microenvironment (TME), regulate both the CSCs and efflux transporters. We propose a new approach focusing on MSCs, which can be crucial to successfully address CSCs and efflux transporters.
Collapse
Affiliation(s)
- Sameer Kumar Panda
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy; Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| | - Nirmal Robinson
- Center for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5001, Australia
| | - Vincenzo Desiderio
- Department of Experimental Medicine, University of Campania "Luigi Vanvitelli", Naples 80138, Italy.
| |
Collapse
|
30
|
Abe K, Watabe T, Kaneda-Nakashima K, Shirakami Y, Kadonaga Y, Naka S, Ooe K, Toyoshima A, Giesel F, Usui T, Masunaga N, Mishima C, Tsukabe M, Yoshinami T, Sota Y, Miyake T, Tanei T, Shimoda M, Shimazu K. Evaluation of Targeted Alpha Therapy Using [ 211At]FAPI1 in Triple-Negative Breast Cancer Xenograft Models. Int J Mol Sci 2024; 25:11567. [PMID: 39519118 PMCID: PMC11547022 DOI: 10.3390/ijms252111567] [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: 09/14/2024] [Revised: 10/14/2024] [Accepted: 10/24/2024] [Indexed: 11/16/2024] Open
Abstract
Triple-negative breast cancer (TNBC) presents limited therapeutic options and is associated with poor prognosis. Early detection and the development of novel therapeutic agents are therefore imperative. Fibroblast activation protein (FAP) is a membrane protein expressed on cancer-associated fibroblasts (CAFs) that plays an essential role in TNBC proliferation, migration, and invasion. Consequently, it is hypothesized that the Astatine (211At)-labeled FAP inhibitor (FAPI) selectively exerts anti-tumor effects through alpha-particle emission. In this study, we aimed to assess its theranostic capabilities by integrating [18F]FAPI-74 PET imaging with targeted alpha therapy using [211At]FAPI1 in TNBC models. Mice xenografts were established by transplanting MDA-MB-231 and HT1080 cells (control). As a parallel diagnostic method, [18F]FAPI-74 was administered for PET imaging to validate FAP expression. A single dose of [211At]FAPI1 (1.04 ± 0.10 MBq) was administered to evaluate the therapeutic efficacy. [18F]FAPI-74 exhibited high accumulation in MDA-MB-231 xenografts, and FAP expression was pathologically confirmed via immunostaining. The group that received [211At]FAPI1 (n = 11) demonstrated a significantly enhanced anti-tumor effect compared with the control group (n = 7) (p = 0.002). In conclusion, [18F]FAPI-74 PET imaging was successfully used to diagnose FAP expression, and as [211At]FAPI1 showed promising therapeutic efficacy in TNBC models, it is expected to be a viable therapeutic option.
Collapse
Affiliation(s)
- Kaori Abe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Tadashi Watabe
- Department of Radiology, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
| | - Kazuko Kaneda-Nakashima
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
- Core for Medicine and Science Collaborative Research and Education, Forefront Research Center, Graduate School of Medicine, Osaka University, Suita 560-0043, Japan
| | | | - Yuichiro Kadonaga
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
| | - Sadahiro Naka
- Department of Pharmacy, Osaka University Hospital, Suita 565-0871, Japan
| | - Kazuhiro Ooe
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
| | - Atsushi Toyoshima
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
| | - Frederik Giesel
- Institute for Radiation Sciences, Osaka University, Suita 565-0871, Japan
- Department of Nuclear Medicine, University Hospital Duesseldorf, Medical Faculty, Heinrich-Heine-University, 40225 Duesseldorf, Germany
| | - Takeshi Usui
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Nanae Masunaga
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Chieko Mishima
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Masami Tsukabe
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Tetsuhiro Yoshinami
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Yoshiaki Sota
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Tomohiro Miyake
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Tomonori Tanei
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Masafumi Shimoda
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| | - Kenzo Shimazu
- Department of Breast and Endocrine Surgery, Graduate School of Medicine, Osaka University, Suita 565-0871, Japan; (K.A.)
| |
Collapse
|
31
|
Wang N, Fang Y, Hou Y, Cheng D, Dressler EV, Wang H, Wang J, Wang G, Li Y, Liu H, Xiang R, Yang S, Sun P. Senescent cells promote breast cancer cells motility by secreting GM-CSF and bFGF that activate the JNK signaling pathway. Cell Commun Signal 2024; 22:478. [PMID: 39375718 PMCID: PMC11457416 DOI: 10.1186/s12964-024-01861-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2024] [Accepted: 09/30/2024] [Indexed: 10/09/2024] Open
Abstract
BACKGROUND Cellular senescence can be induced in mammalian tissues by multiple stimuli, including aging, oncogene activation and loss of tumor suppressor genes, and various types of stresses. While senescence is a tumor suppressing mechanism when induced within premalignant or malignant tumor cells, senescent cells can promote cancer development through increased secretion of growth factors, cytokines, chemokines, extracellular matrix, and degradative enzymes, collectively known as senescence-associated secretory phenotype (SASP). Previous studies indicated that senescent cells, through SASP factors, stimulate tumor cell invasion that is a critical step in cancer cell metastasis. METHODS In the current study, we investigated the effect of senescent cells on the motility of breast cancer cells, which is another key step in cancer cell metastasis. We analyzed the motility of breast cancer cells co-cultured with senescent cells in vitro and metastasis of the breast cancer cells co-injected with senescent cells in orthotopic xenograft models. We also delineated the signaling pathway mediating the effect of senescent cells on cancer cell motility. RESULTS Our results indicate that senescent cells stimulated the migration of breast cancer cells through secretion of GM-CSF and bFGF, which in turn induced activation of the JNK pathway in cancer cells. More importantly, senescent cells promoted breast cancer metastasis, with a minimum effect on the primary tumor growth, in orthotopic xenograft mouse models. CONCLUSIONS These results have revealed an additional mechanism by which senescent cells promote tumor cell metastasis and tumor progression, and will potentially lead to identification of novel targets for cancer therapies that suppress metastasis, the major cause of cancer mortality.
Collapse
Affiliation(s)
- Nan Wang
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin, China
- School of Medicine, Nankai University, Tianjin, China
- Department of Cancer Biology, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Yan Fang
- School of Medicine, Nankai University, Tianjin, China
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Yigong Hou
- School of Medicine, Nankai University, Tianjin, China
- Department of Cancer Biology, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Dongmei Cheng
- Department of Cancer Biology, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Emily V Dressler
- Department of Biostatistics and Data Science, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
| | - Hao Wang
- Department of Cancer Biology, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
- Department of General Surgery, Jiangnan University Medical Center, Wuxi, China
| | - Juan Wang
- Department of Cancer Biology, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
- College of Life Sciences, Hebei Agricultural University, Baoding, China
| | - Guanwen Wang
- Department of Cancer Biology, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA
- Breast Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Yilei Li
- Department of Pharmacy, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hong Liu
- The Second Surgical Department of Breast Cancer, Tianjin Medical University Cancer Institute & Hospital, Key Laboratory of Cancer Prevention and Therapy, National Clinical Research Center for Cancer, Tianjin, China
| | - Rong Xiang
- School of Medicine, Nankai University, Tianjin, China
| | - Shuang Yang
- School of Medicine, Nankai University, Tianjin, China.
| | - Peiqing Sun
- Department of Cancer Biology, Wake Forest University School of Medicine, Atrium Health Wake Forest Baptist Comprehensive Cancer Center, Winston-Salem, NC, USA.
| |
Collapse
|
32
|
Soydas T, Yenmis G, Tuncdemir M, Kalkan MT, Sarac EY, Bilir A, Sultuybek GK. Metformin represses the carcinogenesis potentially induced by 50 Hz magnetic fields in aged mouse fibroblasts via inhibition of NF-kB. J Cell Mol Med 2024; 28:e70132. [PMID: 39350724 PMCID: PMC11442989 DOI: 10.1111/jcmm.70132] [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: 08/31/2023] [Revised: 09/10/2024] [Accepted: 09/20/2024] [Indexed: 10/04/2024] Open
Abstract
Aging is a risk factor for various human disorders, including cancer. Current literature advocates that the primary principles of aging depend on the endogenous stress-induced DNA damage caused by reactive oxygen species 50 Hz low-frequency magnetic field was suggested to induce DNA damage and chromosomal instability. NF-kB, activated by DNA damage, is upregulated in age-related cancers and inhibition of NF-kB results in aging-related delayed pathologies. Metformin (Met), an NF-kB inhibitor, significantly reduces both NF-kB activation and expression in aging and cancer. This in vitro study, therefore, was set out to assess the effects of 5mT MF in 50 Hz frequency and Met treatment on the viability and proliferation of aged mouse NIH/3T3 fibroblasts and expression of RELA/p65, matrix metalloproteinases MMP2 and MMP9, and E-cadherin (CDH1) genes. The trypan blue exclusion assay was used to determine cell viability and the BrdU incorporation assay to determine cell proliferation. The MMP-2/9 protein analysis was carried out by immunocytochemistry, NF-kB activity by ELISA and the expressions of targeted genes by qRT-PCR methods. Four doses of Met (500 uM, 1 mM, 2 mM and 10 mM) suppressed both the proliferation and viability of fibroblasts exposed to the MF in a dose-dependent pattern, and the peak inhibition was recorded at the 10 mM dose. Met reduced the expression of NF-kB, and MMP2/9, elevated CDH1 expression and suppressed NF-kB activity. These findings suggest that Met treatment suppresses the carcinogenic potential of 50 Hz MFs in aged mouse fibroblasts, possibly through modulation of NF-kB activation and epithelial-mesenchymal transition modulation.
Collapse
Affiliation(s)
- Tugba Soydas
- Department of Medical Biology, Medical FacultyIstanbul Aydin UniversityIstanbulTurkey
- Department of Medical BiologyIstanbul University‐Cerrahpasa, Cerrahpasa Faculty of MedicineIstanbulTurkey
| | - Guven Yenmis
- Department of Medical BiologyHatay Mustafa Kemal University, Tayfur Sokmen Faculty of MedicineHatayTurkey
| | - Matem Tuncdemir
- Department of Medical BiologyIstanbul University‐Cerrahpasa, Cerrahpasa Faculty of MedicineIstanbulTurkey
| | - Mustafa Tunaya Kalkan
- Department of Medical BiophysicsIstanbul Aydin University, Medical FacultyIstanbulTurkey
| | - Elif Yaprak Sarac
- Department of Molecular Biology‐Genetics and BiotechnologyIstanbul Technical University, Faculty of Science and LettersIstanbulTurkey
| | - Ayhan Bilir
- Department of Histology and EmbryologyAtlas University, Medical FacultyIstanbulTurkey
| | - Gonul Kanigur Sultuybek
- Department of Medical BiologyIstanbul University‐Cerrahpasa, Cerrahpasa Faculty of MedicineIstanbulTurkey
| |
Collapse
|
33
|
James CD, Lewis RL, Witt AJ, Carter C, Rais NM, Wang X, Bristol ML. Fibroblasts Regulate the Transformation Potential of Human Papillomavirus-positive Keratinocytes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.16.613347. [PMID: 39345623 PMCID: PMC11430071 DOI: 10.1101/2024.09.16.613347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
Persistent human papillomavirus (HPV) infection is necessary but insufficient for viral oncogenesis. Additional contributing co-factors, such as immune evasion and viral integration have been implicated in HPV-induced cancer progression. It is widely accepted that HPV+ keratinocytes require co-culture with fibroblasts to maintain viral episome expression, yet the exact mechanisms for this have yet to be elucidated. Here we present comprehensive RNA sequencing and proteomic analysis demonstrating that fibroblasts not only support the viral life cycle, but reduce HPV+ keratinocyte transformation. Our co-culture models offer novel insights into HPV-related transformation mechanisms.
Collapse
Affiliation(s)
- Claire D. James
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, Virginia, USA
| | - Rachel L. Lewis
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, Virginia, USA
| | - Austin J. Witt
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, Virginia, USA
| | | | - Nabiha M. Rais
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, Virginia, USA
| | - Xu Wang
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, Virginia, USA
| | - Molly L. Bristol
- Philips Institute for Oral Health Research, School of Dentistry, Virginia Commonwealth University (VCU), Richmond, Virginia, USA
- VCU Massey Comprehensive Cancer Center, Richmond, Virginia, USA
| |
Collapse
|
34
|
Yadav R, Sharma A, Dahiya D, Bal A, Bhatia A. Comparative morphology of tumour microenvironment in claudin-low and claudin-high breast cancers. Pathol Res Pract 2024; 261:155502. [PMID: 39079385 DOI: 10.1016/j.prp.2024.155502] [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: 04/26/2024] [Revised: 07/15/2024] [Accepted: 07/27/2024] [Indexed: 08/18/2024]
Abstract
BACKGROUND Claudin-low breast cancers (BCs) exhibit more aggressive behaviour compared to claudin-high types. Claudin-low BCs are often characterized by features such as a higher grade, enrichment of stemness characteristics, and a propensity for metastasis. Tumour microenvironment (TME) defined as the intricate network of surrounding cells, blood vessels, and extracellular matrix components influences the behaviour of cancer cells within the breast tissue. Understanding the TME is crucial for comprehending the aggressive characteristics of claudin-low BCs. METHODS In this study, we have studied the morphology of immune and non-immune TME using Haematoxylin and eosin (H&E)-stained slides of 15 claudin-low and 12 claudin-high tissue samples of BC. RESULTS TME of claudin-low BCs was observed to have a significantly higher frequency of retraction clefts (66.6 %; n = 10/15), immature desmoplastic response (40 %; n = 6/15), higher stromal cellularity (60 %; n = 9/15); and fibroblastic proliferation (53.3 %; n = 8/15) with a low prevalence of elastosis (66.6 %; n = 10/15). The immune microenvironment revealed a higher frequency of total (80 %; n = 12/15) as well as stromal (86.67 %; n = 13/15) and intra-tumoural TILs (60 %; n = 9/15) in them. CONCLUSION The above morphology-based study revealed that claudin-low tumours have unique immune and non-immune TME as compared to claudin-high tumours. Future studies exploring the molecular correlates of each of the above morphological features can help in identifying novel therapeutic targets for the treatment of claudin-low BCs.
Collapse
Affiliation(s)
- Reena Yadav
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Aditti Sharma
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Divya Dahiya
- Department of General Surgery, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Amanjit Bal
- Department of Histopathology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| | - Alka Bhatia
- Department of Experimental Medicine and Biotechnology, Postgraduate Institute of Medical Education and Research, Chandigarh, India.
| |
Collapse
|
35
|
Zhang Y, Tang L, Liu H, Cheng Y. The Multiple Functions of HB-EGF in Female Reproduction and Related Cancer: Molecular Mechanisms and Targeting Strategies. Reprod Sci 2024; 31:2588-2603. [PMID: 38424408 DOI: 10.1007/s43032-024-01454-6] [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: 08/16/2023] [Accepted: 01/03/2024] [Indexed: 03/02/2024]
Abstract
Heparin-binding growth factor (HB-EGF) is a member of the epidermal growth factor (EGF) ligand family which has a crucial role in women's health. However, there is a lack of comprehensive review to summarize the significance of HB-EGF. Therefore, this work first described the expression patterns of HB-EGF in the endometrium and ovary of different species and gestational time. Then, the focus was on exploring how it promotes the successful implantation and regulates the process of decidualization and the function of ovarian granulosa cells as an intermediate molecule. Otherwise, we also focused on the clinical and prognostic significance of HB-EGF in female-related cancers (including ovarian cancer, cervical cancer, and endometrial cancer) and breast cancer. Lastly, the article also summarizes the current drugs targeting HB-EGF in the treatment of ovarian cancer and breast cancer. Overall, these studies found that the expression of HB-EGF in the endometrium is spatiotemporal and species-specific. And it mediates the dialogue between the blastocyst and endometrium, promoting synchronous development of the blastocyst and endometrium as an intermediate molecule. HB-EGF may serve as a potentially valuable prognostic clinical indicator in tumors. And the specific inhibitor of HB-EGF (CRM197) has a certain anti-tumor ability, which can exert synergistic anti-tumor effects with conventional chemotherapy drugs. However, it also suggests that more research is needed in the future to elucidate its specific mechanisms and to accommodate clinical studies with a larger sample size to clarify its clinical value.
Collapse
Affiliation(s)
- Yuwei Zhang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Lujia Tang
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China
| | - Hua Liu
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China.
| | - Yanxiang Cheng
- Department of Obstetrics and Gynecology, Renmin Hospital of Wuhan University, 99 Zhang Zhidong Road, Wuhan, Hubei, 430060, People's Republic of China.
| |
Collapse
|
36
|
Hu Q, Remsing Rix LL, Desai B, Miroshnychenko D, Li X, Welsh EA, Fang B, Wright GM, Chaudhary N, Kroeger JL, Doebele RC, Koomen JM, Haura EB, Marusyk A, Rix U. Cancer-associated fibroblasts confer ALK inhibitor resistance in EML4-ALK -driven lung cancer via concurrent integrin and MET signaling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.27.609975. [PMID: 39253447 PMCID: PMC11383036 DOI: 10.1101/2024.08.27.609975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/11/2024]
Abstract
Cancer-associated fibroblasts (CAFs) are associated with tumor progression and modulate drug sensitivity of cancer cells. However, the underlying mechanisms are often incompletely understood and crosstalk between tumor cells and CAFs involves soluble secreted as well as adhesion proteins. Interrogating a panel of non-small cell lung cancer (NSCLC) cell lines driven by EML4-ALK fusions, we observed substantial CAF-mediated drug resistance to clinical ALK tyrosine kinase inhibitors (TKIs). Array-based cytokine profiling of fibroblast-derived conditioned- media identified HGF-MET signaling as a major contributor to CAF-mediated paracrine resistance that can be overcome by MET TKIs. However, 'Cell Type specific labeling using Amino acid Precursors' (CTAP)-based expression and phosphoproteomics in direct coculture also highlighted a critical role for the fibronectin-integrin pathway. Flow cytometry analysis confirmed activation of integrin β1 (ITGB1) in lung cancer cells by CAF coculture. Treatment with pharmacological inhibitors, cancer cell-specific silencing or CRISPR-Cas9-mediated knockout of ITGB1 overcame adhesion protein-mediated resistance. Concurrent targeting of MET and integrin signaling effectively abrogated CAF-mediated resistance of EML4-ALK -driven NSCLC cells to ALK TKIs in vitro . Consistently, combination of the ALK TKI alectinib with the MET TKI capmatinib and/or the integrin inhibitor cilengitide was significantly more efficacious than single agent treatment in suppressing tumor growth using an in vivo EML4-ALK -dependent allograft mouse model of NSCLC. In summary, these findings emphasize the complexity of resistance-associated crosstalk between CAFs and cancer cells, which can involve multiple concurrent signaling pathways, and illustrate how comprehensive elucidation of paracrine and juxtacrine resistance mechanisms can inform on more effective therapeutic approaches.
Collapse
|
37
|
Yao S, Liu X, Feng Y, Li Y, Xiao X, Han Y, Xia S. Unveiling the Role of HGF/c-Met Signaling in Non-Small Cell Lung Cancer Tumor Microenvironment. Int J Mol Sci 2024; 25:9101. [PMID: 39201787 PMCID: PMC11354629 DOI: 10.3390/ijms25169101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 09/03/2024] Open
Abstract
Non-small cell lung cancer (NSCLC) is characterized by several molecular alterations that contribute to its development and progression. These alterations include the epidermal growth factor receptor (EGFR), anaplastic lymphoma kinase (ALK), human epidermal growth factor receptor 2 (HER2), and mesenchymal-epithelial transition factor (c-MET). Among these, the hepatocyte growth factor (HGF)/c-MET signaling pathway plays a crucial role in NSCLC. In spite of this, the involvement of the HGF/c-MET signaling axis in remodeling the tumor microenvironment (TME) remains relatively unexplored. This review explores the biological functions of the HGF/c-MET signaling pathway in both normal and cancerous cells, examining its multifaceted roles in the NSCLC tumor microenvironment, including tumor cell proliferation, migration and invasion, angiogenesis, and immune evasion. Furthermore, we summarize the current progress and clinical applications of MET-targeted therapies in NSCLC and discuss future research directions, such as the development of novel MET inhibitors and the potential of combination immunotherapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Shu Xia
- Department of Oncology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China; (S.Y.); (X.L.); (Y.F.); (Y.L.); (X.X.); (Y.H.)
| |
Collapse
|
38
|
Hosseini TM, Park SJ, Guo T. The Mutational and Microenvironmental Landscape of Cutaneous Squamous Cell Carcinoma: A Review. Cancers (Basel) 2024; 16:2904. [PMID: 39199674 PMCID: PMC11352924 DOI: 10.3390/cancers16162904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/09/2024] [Accepted: 08/19/2024] [Indexed: 09/01/2024] Open
Abstract
Cutaneous squamous cell carcinoma (cSCC) manifests through the complex interactions of UV-induced DNA damage, genetic mutations, and alterations in the tumor microenvironment. A high mutational burden is present in cSCC, as well as both cSCC precursors and normal skin, making driver genes difficult to differentiate. Despite this, several key driver genes have been identified, including TP53, the NOTCH family, CDKN2A, PIK3CA, and EGFR. In addition to mutations, the tumor microenvironment and the manipulation and evasion of the immune system play a critical role in cSCC progression. Novel therapeutic approaches, such as immunotherapy and EGFR inhibitors, have been used to target these dysregulations, and have shown promise in treating advanced cSCC cases, emphasizing the need for targeted interventions considering both genetic and microenvironmental factors for improved patient outcomes.
Collapse
Affiliation(s)
- Tara M. Hosseini
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
| | - Soo J. Park
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Division of Hematology-Oncology, Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Theresa Guo
- Gleiberman Head and Neck Cancer Center, Moores Cancer Center, University of California San Diego, La Jolla, CA 92093, USA
- Department of Otolaryngology-Head & Neck Surgery, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
39
|
Choudhury AR, Nagesh AM, Gupta S, Chaturvedi PK, Kumar N, Sandeep K, Pandey D. MicroRNA signature of stromal-epithelial interactions in prostate and breast cancers. Exp Cell Res 2024; 441:114171. [PMID: 39029573 DOI: 10.1016/j.yexcr.2024.114171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 07/01/2024] [Accepted: 07/17/2024] [Indexed: 07/21/2024]
Abstract
Stromal-epithelial communication is an absolute necessity when it comes to the morphogenesis and pathogenesis of solid tissues, including the prostate and breast. So far, signalling pathways of several growth factors have been investigated. Besides such chemical factors, non-coding RNAs such as miRNAs have recently gained much interest because of their variety and complexity of action. Prostate and breast tissues being highly responsive to steroid hormones such as androgen and estrogen, respectively, it is not surprising that a huge set of available literature critically investigated the interplay between such hormones and miRNAs, especially in carcinogenesis. This review showcases our effort to highlight hormonally-related miRNAs that also somehow perturb the regular stromal-epithelial interactions during carcinogenesis in the prostate and breast. In future, we look forward to exploring how hormonal changes in the tissue microenvironment bring about miRNA-mediated changes in stromal-epithelial interactome in carcinogenesis and cancer progression.
Collapse
Affiliation(s)
- Ankit Roy Choudhury
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India; Department of Biology, Philipps University, Marburg, Germany
| | - A Muni Nagesh
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Surabhi Gupta
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | | | - Neeraj Kumar
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India
| | - Kumar Sandeep
- Department of Preventive Oncology, Dr. Bhim Rao Ambedkar Institute Rotary Cancer Hospital, All India Institute of Medical Sciences, New Delhi, India
| | - Deepak Pandey
- Department of Reproductive Biology, All India Institute of Medical Sciences, New Delhi, India.
| |
Collapse
|
40
|
Guo H, Xu X, Zhang J, Du Y, Yang X, He Z, Zhao L, Liang T, Guo L. The Pivotal Role of Preclinical Animal Models in Anti-Cancer Drug Discovery and Personalized Cancer Therapy Strategies. Pharmaceuticals (Basel) 2024; 17:1048. [PMID: 39204153 PMCID: PMC11357454 DOI: 10.3390/ph17081048] [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: 07/14/2024] [Revised: 08/02/2024] [Accepted: 08/07/2024] [Indexed: 09/03/2024] Open
Abstract
The establishment and utilization of preclinical animal models constitute a pivotal aspect across all facets of cancer research, indispensably contributing to the comprehension of disease initiation and progression mechanisms, as well as facilitating the development of innovative anti-cancer therapeutic approaches. These models have emerged as crucial bridges between basic and clinical research, offering multifaceted support to clinical investigations. This study initially focuses on the importance and benefits of establishing preclinical animal models, discussing the different types of preclinical animal models and recent advancements in cancer research. It then delves into cancer treatment, studying the characteristics of different stages of tumor development and the development of anti-cancer drugs. By integrating tumor hallmarks and preclinical research, we elaborate on the path of anti-cancer drug development and provide guidance on personalized cancer therapy strategies, including synthetic lethality approaches and novel drugs widely adopted in the field. Ultimately, we summarize a strategic framework for selecting preclinical safety experiments, tailored to experimental modalities and preclinical animal species, and present an outlook on the prospects and challenges associated with preclinical animal models. These models undoubtedly offer new avenues for cancer research, encompassing drug development and personalized anti-cancer protocols. Nevertheless, the road ahead continues to be lengthy and fraught with obstacles. Hence, we encourage researchers to persist in harnessing advanced technologies to refine preclinical animal models, thereby empowering these emerging paradigms to positively impact cancer patient outcomes.
Collapse
Affiliation(s)
- Haochuan Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Xinru Xu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Jiaxi Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Yajing Du
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Xinbing Yang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Zhiheng He
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; (Z.H.); (L.Z.)
| | - Linjie Zhao
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; (Z.H.); (L.Z.)
| | - Tingming Liang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, School of Life Science, Nanjing Normal University, Nanjing 210023, China; (H.G.); (X.X.); (J.Z.); (Y.D.); (X.Y.)
| | - Li Guo
- State Key Laboratory of Organic Electronics and Information Displays & Institute of Advanced Materials (IAM), Nanjing University of Posts & Telecommunications, 9 Wenyuan Road, Nanjing 210023, China; (Z.H.); (L.Z.)
| |
Collapse
|
41
|
Koyama Y, Ogawa C, Kurihara C, Hashimoto N, Shinagawa S, Okazaki H, Koyama T, Sugahara K, Katakura A. Pathological examination of factors involved in PD-L1 expression in patients with oral tongue squamous cell carcinoma. Maxillofac Plast Reconstr Surg 2024; 46:31. [PMID: 39115623 PMCID: PMC11310371 DOI: 10.1186/s40902-024-00441-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 08/01/2024] [Indexed: 08/11/2024] Open
Abstract
BACKGROUND Tumor tissues comprise cancer cells and stromal cells, and their interactions form the cancer microenvironment. Therefore, treatments targeting cells other than cancer cells are also actively being developed, and among them, treatment targeting PD-1, an immune checkpoint molecule that is important in tumor immune evasion, has also been indicated for head and neck cancer. PD-L1, a ligand of PD-1, is expressed in both tumor cells and stromal cells, and the scoring system based on the combined positivity rates of both types of cells, the combined positive score (CPS), is used for predicting treatment effect. However, much is unknown regarding the expression of PD-L1. In this study, we histopathologically examined factors controlling the expression of PD-1/PD-L1. This study included 37 patients who underwent resection surgery for tongue squamous cell carcinoma in the Department of Oral and Maxillofacial Surgery at Tokyo Dental College Suidobashi Hospital. The expression levels of PD-L1, α-SMA, and p53 were assessed by immunohistochemical staining. RESULTS Seven participants had CPS ≥ 20, twenty-four participants had 1 ≤ CPS < 20, and six participants had CPS < 1. The overall positivity rate of α-SMA, a marker for cancer-associated fibroblasts (CAFs), was 27% (10/37 participants), and the positivity rates of α-SMA for the three CPS groups were 85.7% (6/7 participants), 16.7% (4/24 participants), and 0% (0/6 participants), respectively. In addition, the overall positivity rate of p53 was 37.8% (14/37 participants), and the positivity rates of p53 for the three CPS groups were 71.4% (5/7 participants), 37.5% (9/24 participants), and 0% (0/6 participants), respectively. CONCLUSIONS The expression of PD-L1 demonstrated an association with α-SMA and p53 positivity. In addition, compared with the expression of p53, the expression of α-SMA demonstrated a higher association with PD-L1 expression in patients with a high CPS. The abovementioned findings suggest that the interactions between CAFs, cancer cells, and immunocompetent cells may regulate the expression of PD-L1.
Collapse
Affiliation(s)
- Yu Koyama
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan
| | - Chiharu Ogawa
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan
- Department of Oral and Maxillofacial Surgery, Okinawa Prefectural Chubu Hospital, Okinawa, Japan, 281 Miyazato, Uruma-Shi, Okinawa, Japan
| | - Chihiro Kurihara
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan
| | - Nao Hashimoto
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan
| | - Shota Shinagawa
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan
| | - Hiroya Okazaki
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan
| | - Takumi Koyama
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan
| | - Keisuke Sugahara
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan.
| | - Akira Katakura
- Department of Oral Pathobiological Science and Surgery, Tokyo Dental College, Tokyo, Japan, 2-9-18 Kanda Misaki-Cho, Chiyoda-Ku, Tokyo, Japan
| |
Collapse
|
42
|
Valdivia-Silva J, Chinney-Herrera A. Chemokine receptors and their ligands in breast cancer: The key roles in progression and metastasis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2024; 388:124-161. [PMID: 39260935 DOI: 10.1016/bs.ircmb.2024.07.002] [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: 09/13/2024]
Abstract
Chemokines and their receptors are a family of chemotactic cytokines with important functions in the immune response in both health and disease. Their known physiological roles such as the regulation of leukocyte trafficking and the development of immune organs generated great interest when it was found that they were also related to the control of early and late inflammatory stages in the tumor microenvironment. In fact, in breast cancer, an imbalance in the synthesis of chemokines and/or in the expression of their receptors was attributed to be involved in the regulation of disease progression, including invasion and metastasis. Research in this area is progressing rapidly and the development of new agents based on chemokine and chemokine receptor antagonists are emerging as attractive alternative strategies. This chapter provides a snapshot of the different functions reported for chemokines and their receptors with respect to the potential to regulate breast cancer progression.
Collapse
Affiliation(s)
- Julio Valdivia-Silva
- Centro de Investigación en Bioingenieria (BIO), Universidad de Ingenieria y Tecnologia-UTEC, Barranco, Lima, Peru.
| | - Alberto Chinney-Herrera
- Facultad de Medicina, Universidad Nacional Autonoma de Mexico-UNAM, Ciudad Universitaria, Coyoacan, Ciudad de Mexico, Mexico
| |
Collapse
|
43
|
Kaufmann J, Haist M, Kur IM, Zimmer S, Hagemann J, Matthias C, Grabbe S, Schmidberger H, Weigert A, Mayer A. Tumor-stroma contact ratio - a novel predictive factor for tumor response to chemoradiotherapy in locally advanced oropharyngeal cancer. Transl Oncol 2024; 46:102019. [PMID: 38833784 PMCID: PMC11190748 DOI: 10.1016/j.tranon.2024.102019] [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: 04/19/2023] [Revised: 05/25/2024] [Accepted: 05/27/2024] [Indexed: 06/06/2024] Open
Abstract
The growth pattern of oropharyngeal squamous cell carcinomas (OPSCC) varies from compact tumor cell aggregates to diffusely infiltrating tumor cell-clusters. The influence of the growth pattern on local tumor control and survival has been studied mainly for surgically treated oral cavity carcinomas on a visual basis. In this study, we used multiplex immunofluorescence staining (mIF) to examine the antigens pan-cytokeratin, p16INK4a, Ki67, CD271, PD-L1, and CD8 in pretherapeutic biopsies from 86 OPSCC. We introduce Tumor-stroma contact ratio (TSC), a novel parameter, to quantify the relationship between tumor cells in contact with the stromal surface and the total number of epithelial tumor cells. mIF tumor cores were analyzed at the single-cell level, and tumor-stromal contact area was quantified using the R package "Spatstat". TSC was correlated with the visually assessed invasion pattern by two independent investigators. Furthermore, TSC was analyzed in relation to clinical parameters and patient survival data to evaluate its potential prognostic significance. Higher TSC correlated with poor response to (chemo-)radiotherapy (r = 0.3, p < 0.01), and shorter overall (OS) and progression-free (PFS) survival (median OS: 13 vs 136 months, p < 0.0001; median PFS: 5 vs 85 months, p < 0.0001). Visual categorization of growth pattern according to established criteria of tumor aggressiveness showed interobserver variability increasing with more nuanced categories (2 categories: k = 0.7, 95 %-CI: 0.55 - 0.85; 4 categories k = 0.48, 95 %-CI: 0.35 - 0.61). In conclusion, TSC is an objective and reproducible computer-based parameter to quantify tumor-stroma contact area. We demonstrate its relevance for the response of oropharyngeal carcinomas to primary (chemo-)radiotherapy.
Collapse
Affiliation(s)
- Justus Kaufmann
- Department of Radiation Oncology and Radiotherapy, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany.
| | - Maximilian Haist
- Department of Dermatology, University Medical Center of the Johannes-Gutenberg-University, 55131 Mainz, Germany; Department of Pathology, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Microbiology & Immunology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Ivan-Maximiliano Kur
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60596 Frankfurt, Germany
| | - Stefanie Zimmer
- Institute of Pathology, University Medical Center of the Johannes-Gutenberg-University, 55131 Mainz, Germany
| | - Jan Hagemann
- Department of Otorhinolaryngology, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany
| | - Christoph Matthias
- Department of Otorhinolaryngology, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes-Gutenberg-University, 55131 Mainz, Germany
| | - Heinz Schmidberger
- Department of Radiation Oncology and Radiotherapy, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, 60596 Frankfurt, Germany
| | - Arnulf Mayer
- Department of Radiation Oncology and Radiotherapy, University Medical Center of the Johannes-Gutenberg-University, Mainz 55131, Germany; Division of Radiation Oncology, Peter MacCallum Cancer Centre, Melbourne, Australia
| |
Collapse
|
44
|
Lin SL, Yang SY, Tsai CH, Fong YC, Chen WL, Liu JF, Lin CY, Tang CH. Nerve growth factor promote VCAM-1-dependent monocyte adhesion and M2 polarization in osteosarcoma microenvironment: Implications for larotrectinib therapy. Int J Biol Sci 2024; 20:4114-4127. [PMID: 39247831 PMCID: PMC11379077 DOI: 10.7150/ijbs.95463] [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: 02/19/2024] [Accepted: 07/09/2024] [Indexed: 09/10/2024] Open
Abstract
Osteosarcoma is the most prevalent form of primary malignant bone tumor, primarily affecting children and adolescents. The nerve growth factors (NGF) referred to as neurotrophins have been associated with cancer-induced bone pain; however, the role of NGF in osteosarcoma has yet to be elucidated. In osteosarcoma samples from the Genomic Data Commons data portal, we detected higher levels of NGF and M2 macrophage markers, but not M1 macrophage markers. In cellular experiments, NGF-stimulated osteosarcoma conditional medium was shown to facilitate macrophage polarization from the M0 to the M2 phenotype. NGF also enhanced VCAM-1-dependent monocyte adhesion within the osteosarcoma microenvironment by down-regulating miR-513c-5p levels through the FAK and c-Src cascades. In in vivo xenograft models, the overexpression of NGF was shown to enhance tumor growth, while the oral administration of the TrK inhibitor larotrectinib markedly antagonized NGF-promoted M2 macrophage expression and tumor progression. These results suggest that larotrectinib could potentially be used as a therapeutic agent aimed at mitigating NGF-mediated osteosarcoma progression.
Collapse
Affiliation(s)
- Syuan-Ling Lin
- Translational Medicine Research Center, China Medical University Hospital, Taichung, Taiwan
| | - Shang-Yu Yang
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
| | - Chun-Hao Tsai
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Chin Fong
- Department of Sports Medicine, College of Health Care, China Medical University, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Hospital, Taichung, Taiwan
- Department of Orthopedic Surgery, China Medical University Beigang Hospital, Yunlin, Taiwan
| | - Wei-Li Chen
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Ju-Fang Liu
- School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taipei City, Taiwan
| | - Chih-Yang Lin
- Translational Medicine Center, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan
| | - Chih-Hsin Tang
- Department of Medical Laboratory Science and Biotechnology, Asia University, Taichung, Taiwan
- Department of Pharmacology, School of Medicine, China Medical University, Taichung, Taiwan
- Chinese Medicine Research Center, China Medical University, Taichung, Taiwan
- Department of Medical Research, China Medical University Hsinchu Hospital, Hsinchu, Taiwan
| |
Collapse
|
45
|
Qi K, Li G, Jiang Y, Tan X, Qiao Q. Stromal cell-expressed malignant gene patterns contribute to the progression of squamous cell carcinomas across different sites. Front Genet 2024; 15:1342306. [PMID: 39071777 PMCID: PMC11272565 DOI: 10.3389/fgene.2024.1342306] [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: 11/24/2023] [Accepted: 06/18/2024] [Indexed: 07/30/2024] Open
Abstract
Background Squamous cell carcinomas (SCCs) across different anatomical locations possess common molecular features. Recent studies showed that stromal cells may contribute to tumor progression and metastasis of SCCs. Limited by current sequencing technology and analysis methods, it has been difficult to combine stroma expression profiles with a large number of clinical information. Methods With the help of transfer learning on the cell line, single-cell, and bulk tumor sequencing data, we identified and validated 2 malignant gene patterns (V1 and V5) expressed by stromal cells of SCCs from head and neck (HNSCC), lung (LUSC), cervix (CESC), esophagus, and breast. Results Pattern V5 reflected a novel malignant feature that explained the mixed signals of HNSCC molecular subtypes. Higher expression of pattern V5 was related to shorter PFI with gender and cancer-type specificity. The other stromal gene pattern V1 was associated with poor PFI in patients after surgery in all the three squamous cancer types (HNSCC p = 0.0055, LUSC p = 0.0292, CESC p = 0.0451). Cancer-associated fibroblasts could induce HNSCC cancer cells to express pattern V1. Adjuvant radiotherapy may weaken the effect of high V1 on recurrence and metastasis, depending on the tumor radiosensitivity. Conclusion Considering the prognostic value of stromal gene patterns and its universality, we suggest that the genetic subtype classification of SCCs may be improved to a new system that integrates both malignant and non-malignant components.
Collapse
Affiliation(s)
- Kaiyan Qi
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| | - Guangqi Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Yuanjun Jiang
- Department of Urology, The First Hospital of China Medical University, Shenyang, China
| | - Xuexin Tan
- Department of Oral Maxillofacial-Head and Neck Surgery, School and Hospital of Stomatology, China Medical University, Shenyang, China
| | - Qiao Qiao
- Department of Radiation Oncology, The First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
46
|
Yang S, Zhang D, Sun Q, Nie H, Zhang Y, Wang X, Huang Y, Sun Y. Single-Cell and Spatial Transcriptome Profiling Identifies the Transcription Factor BHLHE40 as a Driver of EMT in Metastatic Colorectal Cancer. Cancer Res 2024; 84:2202-2217. [PMID: 38657117 DOI: 10.1158/0008-5472.can-23-3264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 02/14/2024] [Accepted: 04/18/2024] [Indexed: 04/26/2024]
Abstract
Colorectal cancer is one of the most common malignant tumors in humans, with liver metastasis being the primary cause of mortality. The epithelial-mesenchymal transition (EMT) process endows cancer cells with enhanced metastatic potential. To elucidate the cellular mechanisms driving EMT in colorectal cancer, we analyzed single-cell RNA sequencing data from 11 nonmetastatic primary tumors (TnM) and 11 metastatic primary tumors (TM) from colorectal cancer patients. Compared with the TnM group, the TM samples showed elevated numbers of malignant epithelial cell and cancer-associated fibroblast (CAF) subsets that displayed enrichments of EMT, angiogenesis, and TGFβ signaling pathways. One specific TM-enriched subgroup of malignant epithelial cells underwent EMT to transdifferentiate into CXCL1+ CAFs that subsequently differentiated into SFRP2+ CAFs, which was validated by spatial transcriptomic and pseudotime trajectory analyses. Furthermore, cell-cell communication analysis identified BHLHE40 as a probable key transcription factor driving EMT that was associated with poor prognosis. Finally, in vitro and in vivo experiments functionally substantiated that BHLHE40 promoted the proliferation, invasion, migration, EMT, and liver metastasis of colorectal cancer cells. In summary, this study identified BHLHE40 as a key transcription factor regulating EMT that promotes liver metastasis in colorectal cancer. Significance: Integrated analysis of single-cell RNA sequencing and spatial transcriptomics in metastatic colorectal cancer provides insights into the mechanisms underlying EMT and cancer-associated fibroblast differentiation, which could help improve patient diagnosis and treatment.
Collapse
Affiliation(s)
- Sheng Yang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Dongsheng Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Qingyang Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Hongxu Nie
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Yue Zhang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Xiaowei Wang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Yuanjian Huang
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| | - Yueming Sun
- Department of General Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
- The First School of Clinical Medicine, Nanjing Medical University, Nanjing, China
- The Colorectal Institute of Nanjing Medical University, Nanjing, China
- Jiangsu Province Engineering Research Center of Colorectal Cancer Precision Medicine and Translational Medicine, Nanjing, China
| |
Collapse
|
47
|
Steffens S, Kayser C, Roesner A, Rawluk J, Schmid S, Gkika E, Kayser G. Low densities of immune cells indicate unfavourable overall survival in patients suffering from squamous cell carcinoma of the lung. Sci Rep 2024; 14:14250. [PMID: 38902361 PMCID: PMC11190142 DOI: 10.1038/s41598-024-64956-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: 11/09/2023] [Accepted: 06/14/2024] [Indexed: 06/22/2024] Open
Abstract
Carcinogenesis and tumor proliferation are characterized by a complex interaction of cancer cells with the tumor microenvironment. In particular, a tumor-promoting effect can be assumed for the stroma and its fibroblasts. An influence of the immune system on non small cell lung cancer (NSCLC) is now also suspected. In our study, we examined 309 sections of squamous cell carcinoma (SCC), a subtype of NSCLC. We determined the cell densities and areas of the different tissues in SCC using the software QuPath. Spearman rank correlation showed a significant positive correlation between the different tumor cell densities and stromal cell densities, and between tumor cell densities and immune cell densities. Overall survival curves by the Kaplan-Meier method revealed a prominent negative curve in cases of low immune cell density. Based on our results, we can assume a positive influence of the tumor microenvironment, especially the stromal cells, on tumor proliferation in SCC. We have also revealed that low density of immune cells is prognostically unfavorable.
Collapse
Affiliation(s)
- Simone Steffens
- Institute of Pathology Naehrig Mattern Kayser, Bötzinger Strasse 60, Freiburg, Germany.
- Institute of Surgical Pathology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Breisacher Strasse 115a, Freiburg, Germany.
| | - Claudia Kayser
- Institute for Dermatopathology Laaf, Sasbacher Strasse 10, Freiburg, Germany
| | - Anuschka Roesner
- Dental Clinic Zahnzentrum Roesner & Kollegen, Englerstraße 4a, Offenburg, Germany
| | - Justyna Rawluk
- Department of Hematology and Oncology, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, Freiburg, Germany
| | - Severin Schmid
- Department of Thoracic Surgery, University Medical Center Freiburg, Faculty of Medicine, University of Freiburg, Hugstetter Strasse 55, Freiburg, Germany
| | - Eleni Gkika
- Department of Radiation Oncology, University Medical Center Bonn, Faculty of Medicine, University of Bonn, Venusberg-Campus 1, Bonn, Germany
| | - Gian Kayser
- Institute of Pathology Naehrig Mattern Kayser, Bötzinger Strasse 60, Freiburg, Germany
| |
Collapse
|
48
|
Ryu KB, Seo JA, Lee K, Choi J, Yoo G, Ha JH, Ahn MR. Drug-Resistance Biomarkers in Patient-Derived Colorectal Cancer Organoid and Fibroblast Co-Culture System. Curr Issues Mol Biol 2024; 46:5794-5811. [PMID: 38921017 PMCID: PMC11202770 DOI: 10.3390/cimb46060346] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/27/2024] Open
Abstract
Colorectal cancer, the third most commonly occurring tumor worldwide, poses challenges owing to its high mortality rate and persistent drug resistance in metastatic cases. We investigated the tumor microenvironment, emphasizing the role of cancer-associated fibroblasts in the progression and chemoresistance of colorectal cancer. We used an indirect co-culture system comprising colorectal cancer organoids and cancer-associated fibroblasts to simulate the tumor microenvironment. Immunofluorescence staining validated the characteristics of both organoids and fibroblasts, showing high expression of epithelial cell markers (EPCAM), colon cancer markers (CK20), proliferation markers (KI67), and fibroblast markers (VIM, SMA). Transcriptome profiling was conducted after treatment with anticancer drugs, such as 5-fluorouracil and oxaliplatin, to identify chemoresistance-related genes. Changes in gene expression in the co-cultured colorectal cancer organoids following anticancer drug treatment, compared to monocultured organoids, particularly in pathways related to interferon-alpha/beta signaling and major histocompatibility complex class II protein complex assembly, were identified. These two gene groups potentially mediate drug resistance associated with JAK/STAT signaling. The interaction between colorectal cancer organoids and fibroblasts crucially modulates the expression of genes related to drug resistance. These findings suggest that the interaction between colorectal cancer organoids and fibroblasts significantly influences gene expression related to drug resistance, highlighting potential biomarkers and therapeutic targets for overcoming chemoresistance. Enhanced understanding of the interactions between cancer cells and their microenvironment can lead to advancements in personalized medical research..
Collapse
Affiliation(s)
| | | | | | | | | | - Ji-hye Ha
- Clinical Research Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (K.-B.R.)
| | - Mee Ryung Ahn
- Clinical Research Division, National Institute of Food and Drug Safety Evaluation, Ministry of Food and Drug Safety, Cheongju 28159, Chungcheongbuk-do, Republic of Korea; (K.-B.R.)
| |
Collapse
|
49
|
Thakur A, Rana M, Mishra A, Kaur C, Pan CH, Nepali K. Recent advances and future directions on small molecule VEGFR inhibitors in oncological conditions. Eur J Med Chem 2024; 272:116472. [PMID: 38728867 DOI: 10.1016/j.ejmech.2024.116472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 05/12/2024]
Abstract
"A journey of mixed emotions" is a quote that best describes the progress chart of vascular endothelial growth factor receptor (VEGFR) inhibitors as cancer therapeutics in the last decade. Exhilarated with the Food and Drug Administration (FDA) approvals of numerous VEGFR inhibitors coupled with the annoyance of encountering the complications associated with their use, drug discovery enthusiasts are on their toes with an unswerving determination to enhance the rate of translation of VEGFR inhibitors from preclinical to clinical stage. The recently crafted armory of VEGFR inhibitors is a testament to their growing dominance over other antiangiogenic therapies for cancer treatment. This review perspicuously underscores the earnest attempts of the researchers to extract the antiproliferative potential of VEGFR inhibitors through the design of mechanistically diverse structural assemblages. Moreover, this review encompasses sections on structural/molecular properties and physiological functions of VEGFR, FDA-approved VEGFR inhibitors, and hurdles restricting the activity range/clinical applicability of VEGFR targeting antitumor agents. In addition, tactics to overcome the limitations of VEGFR inhibitors are discussed. A clear-cut viewpoint transmitted through this compilation can provide practical directions to push the cart of VEGFR inhibitors to advanced-stage clinical investigations in diverse malignancies.
Collapse
Affiliation(s)
- Amandeep Thakur
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Mandeep Rana
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Anshul Mishra
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan
| | - Charanjit Kaur
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, India
| | - Chun-Hsu Pan
- Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan
| | - Kunal Nepali
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei 110031, Taiwan; Ph.D. Program in Drug Discovery and Development Industry, College of Pharmacy, Taipei Medical University, Taiwan.
| |
Collapse
|
50
|
Carmi YK, Agbarya A, Khamaisi H, Farah R, Shechtman Y, Korobochka R, Gopas J, Mahajna J. Ovarian cancer ascites confers platinum chemoresistance to ovarian cancer cells. Transl Oncol 2024; 44:101939. [PMID: 38489872 PMCID: PMC10955424 DOI: 10.1016/j.tranon.2024.101939] [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: 09/21/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 03/17/2024] Open
Abstract
Ovarian cancer (OC), the second most common form of gynecologic malignancy, has a poor prognosis and is often discovered in the late stages. Platinum-based chemotherapy is the first line of therapy. Nevertheless, treatment OC has proven challenging due to toxicity and the development of acquired resistance to therapy. Tumor microenvironment (TME) has been associated with platinum chemoresistance. Malignant ascites has been used as OC tumor microenvironment and its ability to induce platinum chemoresistance has been investigated. Our results suggest that exposure to OC ascites induces platinum chemoresistance in 11 of 13 cases (85 %) on OC cells. In contrast, 75 % of cirrhotic ascites (3 of 4) failed to confer platinum chemoresistance to OC cells. Cytokine array analysis revealed that IL -6 and to a lesser extent HGF were enriched in OC ascites, whereas IL -22 was enriched in cirrhotic ascites. Pharmaceutical inhibitors targeting the IL -6/ JAK pathway were mildly effective in overcoming platinum chemoresistance induced by malignant ascites. In contrast, crizotinib, an HGF/c- MET inhibitor, and 2-hydroxyestradiol (2HE2) were effective in restoring platinum chemosensitivity to OC. Our results demonstrate the importance of OC ascites in supporting platinum chemoresistance and the potential of combination therapy to restore chemosensitivity of OC cells.
Collapse
Affiliation(s)
- Yifat Koren Carmi
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel; Shraga Segal Department of Microbiology, Immunology and Genetics, and Department of Oncology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Abed Agbarya
- Oncology Department, Bnai Zion MC, Haifa, Israel
| | - Hazem Khamaisi
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel
| | - Raymond Farah
- Department of Internal Medicine, Ziv Medical Center, Safed, Israel
| | | | | | - Jacob Gopas
- Shraga Segal Department of Microbiology, Immunology and Genetics, and Department of Oncology, Soroka University Medical Center, Ben Gurion University of the Negev, Beer Sheva, Israel
| | - Jamal Mahajna
- Department of Nutrition and Natural Products, Migal - Galilee Research Institute, Kiryat Shmona, Israel; Department of Biotechnology, Tel-Hai College, Kiryat Shmona, Israel.
| |
Collapse
|