1
|
Huang P, Chen Y, Shi Y, Zhong C, Lin H, Yu X, Chen K, Huang Z, Zhang L, Fang S, Lu J, Chen J. Phosphoribosyl transferase domain containing 1: A prognostic biomarker in testicular germ cell tumors. MOLECULAR THERAPY. ONCOLOGY 2025; 33:200958. [PMID: 40241724 PMCID: PMC12001118 DOI: 10.1016/j.omton.2025.200958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 01/12/2025] [Accepted: 02/24/2025] [Indexed: 04/18/2025]
Abstract
Due to the heterogeneity and complex classification of testicular germ cell tumors (TGCTs), prognostic evaluation and therapeutic targets remain unclear. Therefore, identifying a novel biomarker to comprehensively assess TGCT prognosis and immunotherapy response is crucial. We collected data from 457 TGCT patient samples and 12 normal testicular samples across six cohorts. Differential expression analysis combined with univariate Cox regression identified prognostic markers for TGCT. Multivariate Cox regression and survival analysis further evaluated the prognostic value of phosphoribosyl transferase domain containing 1 (PRTFDC1). Immunohistochemistry on tissue microarrays validated PRTFDC1's predictive value in clinical samples. We then investigated the relationship between PRTFDC1 and somatic mutations, copy number variations, immune cell infiltration, and immunotherapy response. Through these analyses, we identified PRTFDC1 as an independent risk factor indicating poor prognosis in TGCT. Immunohistochemistry demonstrated high PRTFDC1 expression in TGCT tissues. Gene set enrichment analysis revealed that PRTFDC1 suppresses immune-related pathways. Immune infiltration showed that high PRTFDC1 expression is associated with low CD8+ T cell infiltration. Immunotherapy response analysis indicated that low PRTFDC1 expression predicts better immunotherapy response and favorable prognosis. In conclusion, this study elucidates the biological and clinical significance of PRTFDC1, suggesting it as an effective and reliable biomarker for predicting TGCT prognosis and immunotherapy response.
Collapse
Affiliation(s)
- Peisheng Huang
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Yihao Chen
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Yongcheng Shi
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Chuanfan Zhong
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, China
| | - Huawei Lin
- The Second Clinical College of Guangzhou Medical University, Guangzhou, Guangdong 510180, China
| | - Xiaoxue Yu
- The Second Clinical College of Guangzhou Medical University, Guangzhou, Guangdong 510180, China
| | - Kai Chen
- Department of Pathology, Guangzhou Medical University Affiliated Women and Children′s Medical Center, Guangzhou 510623, China
| | - Zhuoya Huang
- Department of Pathology, Huizhou Central People’s Hospital, No. 41, Eling North Road, Huizhou, Guangdong 516001, China
| | - Le Zhang
- Institute for Integrative Genome Biology, University of California, Riverside, CA 92507, USA
| | - Shumin Fang
- Science Research Center, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| | - Jianming Lu
- Department of Andrology, Guangzhou First People’s Hospital, Guangzhou Medical University, Guangzhou 510180, China
| | - Jiahong Chen
- The First Clinical Medical College, Guangdong Medical University, Zhanjiang, Guangdong 524023, China
- Department of Urology, Huizhou Central People’s Hospital, Huizhou, Guangdong 516001, China
| |
Collapse
|
2
|
Gupta G, Afzal M, Goyal A, PadmaPriya G, Srivastava M, Chennakesavulu K, Mohanty B, Rekha A, Mazumder A, Goyal K, Ali H, Shahwan M. Beta-2 microglobulin in lymphoma. Clin Chim Acta 2025; 576:120418. [PMID: 40490112 DOI: 10.1016/j.cca.2025.120418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2025] [Revised: 06/05/2025] [Accepted: 06/05/2025] [Indexed: 06/11/2025]
Abstract
Lymphomas are heterogeneous hematologic malignancies characterised by the abnormal proliferation of lymphocytes. β2-Microglobulin (β2M) functions both as a structural subunit of primary histocompatibility complex class I (MHC I) and as a circulating biomarker with established diagnostic and prognostic significance. Serum β2M > 2.5 mg/L is elevated in 60 % of mantle cell lymphoma and in > 50 % of advanced-stage diffuse large B-cell lymphoma, correlating with higher tumor burden, International Prognostic Index scores, and inferior survival; cerebrospinal fluid β2M enhances central nervous system lymphoma diagnosis with 97 % sensitivity and specificity. Mechanistically, β2M stabilizes MHC I to enable CD8+ T-cell antigen presentation and, when shed, activates JAK/STAT and NF-κB pathways that drive tumor proliferation and immune evasion. Preclinical strategies targeting these β2M-driven signals such as anti-β2M antibodies combined with proteasome inhibitors demonstrate enhanced cytotoxicity in resistant models. Advanced three-dimensional scaffold culture platforms preserve β2M-tumour-immune interactions, allowing for the investigation of matrix stiffness effects on signalling. Emerging mechanotherapy approaches leverage extracellular matrix rigidity to modulate β2M-related pathways and sensitize lymphoma cells to therapy. The remaining challenges include assay standardization, cohort variability, and lack of prospective validation of β2M-based indices. Future efforts should focus on harmonising β2M measurement methods and integrating mechanistic insights into refined risk stratification and therapeutic strategies.
Collapse
Affiliation(s)
- Gaurav Gupta
- Centre for Research Impact & Outcome, Chitkara College of Pharmacy, Chitkara University, Rajpura, Punjab 140401, India; Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates.
| | - Muhammad Afzal
- Department of Pharmaceutical Sciences, Pharmacy Program, Batterjee Medical College, P.O. Box 6231, Jeddah 21442, Saudi Arabia
| | - Ahsas Goyal
- Institute of Pharmaceutical Research, GLA University, Mathura, Uttar Pradesh, India
| | - G PadmaPriya
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Manish Srivastava
- Department of Endocrinology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Kattela Chennakesavulu
- Department of CHEMISTRY, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Biswaranjan Mohanty
- Department of Nephrology IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha 751003, India
| | - A Rekha
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307 Punjab, India
| | - Avijit Mazumder
- Noida Institute of Engineering and Technology(Pharmacy Institute), 19 Knowledge Park 2, Greater Noida, India
| | - Kavita Goyal
- Department of Biotechnology, Graphic Era (Deemed to be University), Clement Town, Dehradun 248002, India
| | - Haider Ali
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Moyad Shahwan
- Centre of Medical and Bio-allied Health Sciences Research, Ajman University, Ajman, United Arab Emirates
| |
Collapse
|
3
|
Cai G, Rodgers NC, Liu AP. Unjamming Transition as a Paradigm for Biomechanical Control of Cancer Metastasis. Cytoskeleton (Hoboken) 2025; 82:388-403. [PMID: 39633605 PMCID: PMC12137693 DOI: 10.1002/cm.21963] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/27/2024] [Accepted: 11/18/2024] [Indexed: 12/07/2024]
Abstract
Tumor metastasis is a complex phenomenon that poses significant challenges to current cancer therapeutics. While the biochemical signaling involved in promoting motile phenotypes is well understood, the role of biomechanical interactions has recently begun to be incorporated into models of tumor cell migration. Specifically, we propose the unjamming transition, adapted from physical paradigms describing the behavior of granular materials, to better discern the transition toward an invasive phenotype. In this review, we introduce the jamming transition broadly and narrow our discussion to the different modes of 3D tumor cell migration that arise. Then we discuss the mechanical interactions between tumor cells and their neighbors, along with the interactions between tumor cells and the surrounding extracellular matrix. We center our discussion on the interactions that induce a motile state or unjamming transition in these contexts. By considering the interplay between biochemical and biomechanical signaling in tumor cell migration, we can advance our understanding of biomechanical control in cancer metastasis.
Collapse
Affiliation(s)
- Grace Cai
- Applied Physics ProgramUniversity of MichiganAnn ArborMichiganUSA
| | - Nicole C. Rodgers
- Department of Mechanical EngineeringUniversity of MichiganAnn ArborMichiganUSA
| | - Allen P. Liu
- Applied Physics ProgramUniversity of MichiganAnn ArborMichiganUSA
- Department of Mechanical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of Biomedical EngineeringUniversity of MichiganAnn ArborMichiganUSA
- Department of BiophysicsUniversity of MichiganAnn ArborMichiganUSA
| |
Collapse
|
4
|
Saeed Issa B, Adhab AH, Salih Mahdi M, Kyada A, Ganesan S, Bhanot D, Naidu KS, Kaur S, Mansoor AS, Radi UK, Saadoun Abd N, Kariem M. Decoding the complex web: cellular and molecular interactions in the lung tumour microenvironment. J Drug Target 2025; 33:666-690. [PMID: 39707828 DOI: 10.1080/1061186x.2024.2445772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2024] [Revised: 12/10/2024] [Accepted: 12/15/2024] [Indexed: 12/23/2024]
Abstract
The lung tumour microenvironment (TME) or stroma is a dynamic space of numerous cells and their released molecules. This complicated web regulates tumour progression and resistance to different modalities. Lung cancer cells in conjunction with their stroma liberate a wide range of factors that dampen antitumor attacks by innate immunity cells like natural killer (NK) cells and also adaptive responses by effector T cells. These factors include numerous growth factors, exosomes and epigenetic regulators, and also anti-inflammatory cytokines. Understanding the intricate interactions between tumour cells and various elements within the lung TME, such as immune and stromal cells can help provide novel strategies for better management and treatment of lung malignancies. The current article discusses the complex network of cells and signalling molecules, which mediate communications in lung TME. By elucidating these multifaceted interactions, we aim to provide insights into potential therapeutic targets and strategies for lung cancer treatment.
Collapse
Affiliation(s)
| | | | | | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, Gujarat, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Deepak Bhanot
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sharnjeet Kaur
- Department of Applied Sciences, Chandigarh Engineering College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | | | - Usama Kadem Radi
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | - Nasr Saadoun Abd
- Medical Technical College, Al-Farahidi University, Baghdad, Iraq
| | - Muthena Kariem
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
5
|
Ou F, Pan Y, Chen Q, Zeng L, Wei K, Liu D, Guo Q, Zhou L, Yang J. Integrating machine learning and multi-omics analysis to unveil key programmed cell death patterns and immunotherapy targets in kidney renal clear cell carcinoma. Sci Rep 2025; 15:18403. [PMID: 40419510 DOI: 10.1038/s41598-025-00759-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/30/2025] [Indexed: 05/28/2025] Open
Abstract
Kidney renal clear cell carcinoma (KIRC), a cancer characterized by substantial immune infiltration, exhibits limited sensitivity to conventional radiochemotherapy. Although immunotherapy has shown efficacy in some patients, its applicability is not universally effective. Studies have indicated that programmed cell death (PCD) can modulate the activity of immune cells and participate in the regulation of antitumor immune responses. However, systematic research on how various PCD patterns in KIRC affect the responsiveness to immunotherapy is lacking and requires in-depth investigation. We utilized a combination of 101 machine learning algorithms to analyze the TCGA-KIRC cohort and the GSE22541 KIRC patients, screening for cell death patterns closely associated with prognosis from 18 potential modes. Integrating multi-omics analysis, including immune cell infiltration, phenotyping, functional analysis, immune checkpoint exploration, and gene set enrichment analysis (GSEA), we explored the relationship between key cell death patterns and patients' responses to immunotherapy. Finally, potential drug targets were identified through drug sensitivity screening and molecular docking techniques. Our sophisticated risk assessment model successfully identified two PCD patterns, Anoikis and lysosome-dependent cell death (LDCD), closely associated with the prognosis of KIRC patients, with the high-risk group exhibiting poor outcomes. Immune cell analysis revealed upregulated expression of T follicular helper (Tfh) cells in both PCD patterns. Analysis of immune checkpoints disclosed enhanced expression of human leukocyte antigen E (HLA-E) across both patterns. Frequent mutations in the TTN and MUC16 genes were observed in the Anoikis pattern, whereas in the LDCD pattern, although the high-risk group had a higher mutation rate, there was no significant difference in tumor mutational burden. GSEA analysis indicated significant enrichment of the primary immunodeficiency pathway in the Anoikis high-risk group and significant enrichment of the spliceosomal tri-snrnp complex assembly pathway in the LDCD high-risk group. Drug sensitivity analysis showed notable sensitivity to SB505124 in both PCD patterns. HMOX1 and PIK3CG were identified as common genes in the two key PCD patterns, and molecular docking analysis confirmed stable binding affinity between Carnosol and HMOX1, and between PROTAC and PIK3CG. Our study identifies Anoikis and LDCD as prognostic PCD patterns in KIRC, with key immune cells, genetic mutations, and drug sensitivity profiles. HMOX1 and PIK3CG are common genes with stable binding to Carnosol and PROTAC, respectively, while SB505124 shows significant sensitivity to both PCD modes, suggesting potential therapeutic targets.
Collapse
Affiliation(s)
- Fanyan Ou
- Department of Clinical Pathology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yi Pan
- Department of Urology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qiuli Chen
- Department of Clinical Pathology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lixiong Zeng
- Department of Clinical Pathology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Kanglai Wei
- Department of Clinical Pathology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Delin Liu
- Department of Clinical Pathology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qian Guo
- Department of General Practice, the Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Liquan Zhou
- Department of Urology, the Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China.
| | - Jie Yang
- Clinical Medical Research Center, the Second Affiliated Hospital of Guangxi Medical University, Nanning, 530007, Guangxi, China.
| |
Collapse
|
6
|
Bakrim S, Fessikh ME, Elhrech H, Omari NE, Amanullah M, Ming LC, Moshawih S, Bouyahya A. Targeting inflammation in cancer therapy: from mechanistic insights to emerging therapeutic approaches. J Transl Med 2025; 23:588. [PMID: 40420174 DOI: 10.1186/s12967-025-06583-3] [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: 03/08/2025] [Accepted: 05/07/2025] [Indexed: 05/28/2025] Open
Abstract
Inflammation is a complex and finely tuned component of the host defense mechanism, responding sensitively to a range of physical, chemical, and biological stressors. Current research is advancing our grasp of both cellular and molecular mechanisms that initiate and regulate interactions within inflammatory pathways. Substantial evidence now indicates a profound link between inflammation, innate immunity, and cancer. Dysregulation of inflammatory pathways is known to be a pivotal factor in the induction, growth, and metastasis of tumors through multiple mechanistic pathways. Basically, the tumor microenvironment (TME), characterized by dynamic interplay between cancerous cells and surrounding inflammatory and stromal cells, plays a central role in these processes. Increasingly, controlled acute inflammation is being explored as a promising therapeutic tool in certain types of cancer. However, inflammatory cells in the TME exhibit remarkable plasticity, with shifting phenotypic and functional roles that facilitate cancer cell survival, proliferation, and migration, especially under chronic inflammatory conditions. Additionally, signaling molecules associated with the innate immune system, like chemokines, are co-opted by malignant cells to support invasion, migration, and metastasis. These findings underscore the need for deeper insights into the mechanisms connecting inflammation to cancer pathology, which could pave the way for innovative diagnostic approaches and targeted anti-inflammatory therapies to counter tumor development. The current review underlines the critical involvement of inflammation in cancer development, examining the connection between the immune system, key inflammatory mediators, biomarkers, and their associated pathways in cancer. We also discuss the impact of inflammation-targeted therapies on anticancer signaling pathways. Furthermore, we review major anti-inflammatory drugs with potential applications in oncology, assessing how inflammation is modulated in cancer management. Lastly, we outline an overview of ongoing discoveries in the field, highlighting both the challenges and the therapeutic promise of targeting inflammation in cancer therapy.
Collapse
Affiliation(s)
- Saad Bakrim
- Geo-Bio-Environment Engineering and Innovation Laboratory, Molecular Engineering, Biotechnology and Innovation Team, Polydisciplinary Faculty of Taroudant, Ibn Zohr University, Agadir, 80000, Morocco
| | - Meriem El Fessikh
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Hamza Elhrech
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco
| | - Nasreddine El Omari
- High Institute of Nursing Professions and Health Techniques of Tetouan, Tetouan, Morocco
| | - Mohammed Amanullah
- Department of clinical Biochemistry, College of Medicine, King Khalid University, Abha, Kingdom of Saudi Arabia
| | - Long Chiau Ming
- Datta Meghe College of Pharmacy, Datta Meghe Institute of Higher Education and Research (deemed to be University), Sawangi (M), Wardha, India
- Faculty of Medical and Life Sciences, Sunway University, Sunway City, Malaysia
| | - Said Moshawih
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Abdelhakim Bouyahya
- Laboratory of Human Pathologies Biology, Faculty of Sciences, Mohammed V University in Rabat, Rabat, Morocco.
- Department of Pharmaceutical Sciences, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan.
| |
Collapse
|
7
|
Shah S, D'Souza GGM. Modeling Tumor Microenvironment Complexity In Vitro: Spheroids as Physiologically Relevant Tumor Models and Strategies for Their Analysis. Cells 2025; 14:732. [PMID: 40422235 DOI: 10.3390/cells14100732] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2025] [Revised: 05/11/2025] [Accepted: 05/14/2025] [Indexed: 05/28/2025] Open
Abstract
Drug delivery to solid tumors is challenged by multiple physiological barriers arising from the tumor microenvironment, including dense extracellular matrix, cellular heterogeneity, hypoxic gradients, and elevated interstitial fluid pressure. These features hinder the uniform distribution and accumulation of therapeutics, reducing treatment efficacy. Despite their widespread use, conventional two-dimensional monolayer cultures fail to reproduce these complexities, contributing to the poor translational predictability of many preclinical candidates. Three-dimensional multicellular tumor spheroids have emerged as more representative in vitro models that capture essential features of tumor architecture, stromal interactions, and microenvironmental resistance mechanisms. Spheroids exhibit spatially organized regions of proliferation, quiescence, and hypoxia, and can incorporate non-tumor cells to mimic tumor-stroma crosstalk. Advances in spheroid analysis now enable detailed evaluation of drug penetration, cellular migration, cytotoxic response, and molecular gradients using techniques such as optical and confocal imaging, large-particle flow cytometry, biochemical viability assays, and microfluidic integration. By combining physiological relevance with analytical accessibility, spheroid models support mechanistic studies of drug transport and efficacy under tumor-like conditions. Their adoption into routine preclinical workflows has the potential to improve translational accuracy while reducing reliance on animal models.
Collapse
Affiliation(s)
- Shrey Shah
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
- Atom Bioworks Inc., Cary, NC 27513, USA
| | - Gerard G M D'Souza
- Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, Boston, MA 02115, USA
| |
Collapse
|
8
|
Mavaei M, Farokhi S, Yousefi MH, Fakouri A, Shadab A, Abdolmohammadi MH, Fallahian F, Afkhami H. Exploring two tumor treatment strategies: effectiveness of ribosome inactivating proteins and mesenchymal stem cells/MSC derived extracellular vesicles in cancer treatment. Front Oncol 2025; 15:1533065. [PMID: 40444089 PMCID: PMC12120475 DOI: 10.3389/fonc.2025.1533065] [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: 11/23/2024] [Accepted: 04/15/2025] [Indexed: 06/02/2025] Open
Abstract
Cancer is a complex and heterogeneous disease that often requires multifaceted treatment strategies to achieve optimal therapeutic outcomes. Given the limitations of single-agent therapies, particularly in the face of intricate biological signaling networks and treatment resistance, there is a growing need for combinatory approaches. This article presents a novel hypothesis: the simultaneous use of ribosome-inactivating proteins (RIPs) and mesenchymal stem cells (MSCs) or MSC-derived extracellular vesicles (EVs) in cancer treatment. RIPs, with their potent cytotoxic properties, can target tumor cells effectively, while MSCs, known for their tumor-homing abilities and regenerative potential, can serve as delivery vehicles, potentially enhancing the targeting precision and reducing the systemic toxicity of RIPs. This hypothesis explores the synergistic potential of combining these two therapeutic modalities, leveraging the advantages of both techniques to create a more effective cancer treatment strategy. By combining RIPs' ability to inhibit protein synthesis with MSCs or MSC-derived EVs' capability to modulate the tumor microenvironment and deliver therapeutic agents. This approach offers a promising avenue for overcoming cancer's inherent complexity. However, challenges remain, such as optimizing dosing protocols, addressing safety concerns, and ensuring efficient drug delivery. Future research and clinical trials are necessary to validate this combination as a viable cancer therapy.
Collapse
Affiliation(s)
- Maryamosadat Mavaei
- Student Research Committee, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Mohammad Hasan Yousefi
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Arshia Fakouri
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Alireza Shadab
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Iran University of Medical Sciences, Deputy of Health, Tehran, Iran
| | | | - Faranak Fallahian
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Hamed Afkhami
- Student Research Committee, Qom University of Medical Sciences, Qom, Iran
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
9
|
Murray NP. Immunomodulation and Immunotherapy for Patients with Prostate Cancer: An Up-to-Date Review. Biomedicines 2025; 13:1179. [PMID: 40427006 PMCID: PMC12109314 DOI: 10.3390/biomedicines13051179] [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: 02/27/2025] [Revised: 05/07/2025] [Accepted: 05/08/2025] [Indexed: 05/29/2025] Open
Abstract
Immunotherapy alone or in combination with chemotherapy or radiotherapy is the frontline treatment for melanoma and lung cancer. However, its role in prostate cancer is usually as a fourth-line treatment. It is usually employed in patients with metastasis, after androgen blockade and chemotherapy. This article reviews the immunosuppressive effects of prostate cancer and possible uses of various types of immunotherapies. It also considers when would be the optimal time to employ this type of therapy.
Collapse
Affiliation(s)
- Nigel P. Murray
- Faculty of Medicine, Universidad Finis Terrae, Santiago 7501015, Chile;
- Department of Medicine, Hospital de Carabineros de Chile, Santiago 7770199, Chile
| |
Collapse
|
10
|
Xiao Q, Liu Y, Li T, Wang C, He S, Zhai L, Yang Z, Zhang X, Wu Y, Liu Y. Viral oncogenesis in cancer: from mechanisms to therapeutics. Signal Transduct Target Ther 2025; 10:151. [PMID: 40350456 PMCID: PMC12066790 DOI: 10.1038/s41392-025-02197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 05/14/2025] Open
Abstract
The year 2024 marks the 60th anniversary of the discovery of the Epstein-Barr virus (EBV), the first virus confirmed to cause human cancer. Viral infections significantly contribute to the global cancer burden, with seven known Group 1 oncogenic viruses, including hepatitis B virus (HBV), human papillomavirus (HPV), EBV, Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), and human immunodeficiency virus (HIV). These oncogenic viruses induce cellular transformation and cancer development by altering various biological processes within host cells, particularly under immunosuppression or co-carcinogenic exposures. These viruses are primarily associated with hepatocellular carcinoma, gastric cancer, cervical cancer, nasopharyngeal carcinoma, Kaposi sarcoma, lymphoma, and adult T-cell leukemia/lymphoma. Understanding the mechanisms of viral oncogenesis is crucial for identifying and characterizing the early biological processes of virus-related cancers, providing new targets and strategies for treatment or prevention. This review first outlines the global epidemiology of virus-related tumors, milestone events in research, and the process by which oncogenic viruses infect target cells. It then focuses on the molecular mechanisms by which these viruses induce tumors directly or indirectly, including the regulation of oncogenes or tumor suppressor genes, induction of genomic instability, disruption of regular life cycle of cells, immune suppression, chronic inflammation, and inducing angiogenesis. Finally, current therapeutic strategies for virus-related tumors and recent advances in preclinical and clinical research are discussed.
Collapse
Affiliation(s)
- Qing Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Tingting Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Chaoyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Sanxiu He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Liuyue Zhai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zailin Yang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yongzhong Wu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
11
|
Ghafoor S, Garcia E, Jay DJ, Persad S. Molecular Mechanisms Regulating Epithelial Mesenchymal Transition (EMT) to Promote Cancer Progression. Int J Mol Sci 2025; 26:4364. [PMID: 40362600 PMCID: PMC12072817 DOI: 10.3390/ijms26094364] [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: 03/05/2025] [Revised: 04/27/2025] [Accepted: 04/29/2025] [Indexed: 05/15/2025] Open
Abstract
The process of epithelial-mesenchymal transition (EMT) is crucial in various physiological/pathological circumstances such as development, wound healing, stem cell behavior, and cancer progression. It involves the conversion of epithelial cells into a mesenchymal phenotype, which causes the cells to become highly motile. This reprogramming is initiated and controlled by various signaling pathways and governed by several key transcription factors, including Snail 1, Snail 2 (Slug), TWIST 1, TWIST2, ZEB1, ZEB2, PRRX1, GOOSECOID, E47, FOXC2, SOX4, SOX9, HAND1, and HAND2. The intracellular signaling pathways are activated/inactivated by signals received from the extracellular environment and the transcription factors are carefully regulated at the transcriptional, translational, and post-translational levels to maintain tight regulatory control of EMT. One of the most important pathways involved in this process is the transforming growth factor-β (TGFβ) family signaling pathway. This review will discuss the role of EMT in promoting epithelial cancer progression and the convergence/interplay of multiple signaling pathways and transcription factors that regulate this phenomenon.
Collapse
Affiliation(s)
| | | | | | - Sujata Persad
- Department of Pediatrics, Faculty of Medicine and Dentistry, 3020R Katz Group Centre for Pharmacy and Health Research, University of Alberta, Edmonton, AB T6G 2E1, Canada; (S.G.); (E.G.); (D.J.J.)
| |
Collapse
|
12
|
Liu Z, Mao X, Xie Y, Yan Y, Wang X, Mi J, Yuan H, Zhang J, Huang C, Chen J, Jili M, Huang S, Zhang Q, Wang F, Mo Z, Yang R. Single-cell RNA sequencing reveals a fibroblast gene signature that promotes T-cell infiltration in muscle-invasive bladder cancer. Commun Biol 2025; 8:696. [PMID: 40319103 PMCID: PMC12049545 DOI: 10.1038/s42003-025-08094-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 04/16/2025] [Indexed: 05/07/2025] Open
Abstract
Muscle-invasive bladder cancer (MIBC) is characterized by a complex tumor microenvironment (TME) that drives aggressive progression and treatment resistance. Previous studies have highlighted the roles of cancer-associated fibroblasts (CAFs) and exhausted T (Tex) cells in MIBC, but their interactive mechanisms remain poorly understood. Here, single-cell RNA sequencing of 19 tissue samples from 12 patients-7 MIBC, 3 non-muscle-invasive bladder cancer (NMIBC), and 9 normal tissue samples-identified 13 transcriptionally distinct fibroblast clusters and 10 functionally heterogeneous T-cell subsets. Two interferon (IFN)-responsive fibroblast populations, F-ISG15 (inflammatory CAFs) and F-POSTN (myofibroblastic CAFs), were shown to predominate in the MIBC TME. In vivo experiments demonstrated that IFN-γ secreted by Tex cells polarizes CAFs to secrete CXCL12, which recruits CXCR4-expressing T cells via the CXCL12-CXCR4 chemotactic axis. Spatial analysis revealed a bidirectional loop: Tex-derived IFN-γ sustains CAF activation, whereas CAF-secreted CXCL12 amplifies Tex infiltration. Clinically, activated CAF signatures correlate with advanced disease stages and reduced patient survival in MIBC. These findings establish CXCL12 and IFN signaling as critical therapeutic targets, offering new strategies to disrupt immunosuppressive TME crosstalk and improve outcomes for MIBC patients.
Collapse
Affiliation(s)
- Zige Liu
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Xingning Mao
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Collaborative Innovation Centre of Regenerative Medicine and Medical BioResource Development and Application Coconstructed by the Province and Ministry, Guangxi Medical University, Nanning, Guangxi, China
| | - Yuli Xie
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Yunkun Yan
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Xiang Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Junhao Mi
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Hao Yuan
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Jiange Zhang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Second Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Caisheng Huang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Nanning Second People's Hospital, The Third Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianxin Chen
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Mujia Jili
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China
| | - Shengzhu Huang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Qingyun Zhang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
- Department of Urology, The Affiliated Tumor Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Fubo Wang
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China
| | - Zengnan Mo
- Institute of Urology and Nephrology, the First Affiliated Hospital of Guangxi Medical University, Guangxi Medical University, Nanning, Guangxi, China.
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China.
| | - Rirong Yang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Collaborative Innovation Center for Genomic and Personalized Medicine, University Engineering Research Center of Digital Medicine and Healthcare, Guangxi Medical University, Nanning, Guangxi, China.
- Department of Immunology, School of Basic Medical Sciences, Guangxi Medical University, Nanning, Guangxi, China.
| |
Collapse
|
13
|
Frantzi M, Tymoszuk P, Salcher S, Gomez-Gomez E, Morillo AC, Melchior F, Blanca A, Mischak H, Vlahou A, Pircher A, Heidegger I. Prediction of Prostate Cancer Biochemical Recurrence After Radical Prostatectomy by Collagen Models Using Multiomic Profiles. Eur Urol Oncol 2025:S2588-9311(25)00094-X. [PMID: 40312179 DOI: 10.1016/j.euo.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/28/2025] [Accepted: 03/28/2025] [Indexed: 05/03/2025]
Abstract
BACKGROUND AND OBJECTIVE The interplay between prostate cancer and the tumor microenvironment is well documented and of primary importance in disease evolution. Herein, we investigated the prognostic value of tissue and urinary collagen-related molecular signatures in predicting biochemical recurrence (BCR) after radical prostatectomy (RP). METHODS A comprehensive analysis of 55 collagen-related features was conducted using transcriptomic datasets (n = 1393), with further validation at the proteomic level (n = 69). Additionally, a distinct cohort (n = 73) underwent a urine-based peptidomic analysis, culminating in the validation of a urine-derived prognostic model. Independent prognostic significance was assessed using Cox proportional hazards modeling, while the model's predictive performance was benchmarked against established clinical metrics. KEY FINDINGS AND LIMITATIONS An expression analysis of 55 collagen-related transcripts identified 11 transcripts significantly associated with BCR (C-index: 0.55-0.72, p < 0.002). Multivariable models incorporating these transcripts enhanced prognostic accuracy, surpassing clinical variables (C-index: 0.66-0.89, p < 0.002). Proteomic validation confirmed five key collagen proteins, while a urine-based collagen model (C-index: 0.73, 95% confidence interval: 0.62-0.85) demonstrated a strong prognostic potential, although limited by small patient numbers. Additionally, tissue collagen-based models predicted overall survival with a significant prognostic value (C-index: 0.59-0.70, p < 0.01). CONCLUSIONS AND CLINICAL IMPLICATIONS Collagen-based molecular signatures in both tissue and urine emerge as robust prognostic biomarkers for predicting BCR following RP.
Collapse
Affiliation(s)
- Maria Frantzi
- Department of Biomarker Research, Mosaiques Diagnostics GmbH, Hannover, Germany
| | | | - Stefan Salcher
- Department of Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Enrique Gomez-Gomez
- Urology Department, Reina Sofía University Hospital, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), University of Cordoba (UCO), Cordoba, Spain
| | - Ana C Morillo
- Urology Department, Reina Sofía University Hospital, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), University of Cordoba (UCO), Cordoba, Spain
| | - Felix Melchior
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria
| | - Ana Blanca
- Urology Department, Reina Sofía University Hospital, Maimonides Institute of Biomedical Research of Cordoba (IMIBIC), University of Cordoba (UCO), Cordoba, Spain
| | - Harald Mischak
- Department of Biomarker Research, Mosaiques Diagnostics GmbH, Hannover, Germany
| | - Antonia Vlahou
- Systems Biology Center, Biomedical Research Foundation, Academy of Athens, Athens, Greece
| | - Andreas Pircher
- Department of Internal Medicine V, Hematology and Oncology, Medical University of Innsbruck, Innsbruck, Austria
| | - Isabel Heidegger
- Department of Urology, Medical University of Innsbruck, Innsbruck, Austria.
| |
Collapse
|
14
|
Pan Z, Liu Y, Li H, Qiu H, Zhang P, Li Z, Wang X, Tian Y, Feng Z, Zhu S, Wang X. The role and mechanism of aerobic glycolysis in nasopharyngeal carcinoma. PeerJ 2025; 13:e19213. [PMID: 40191756 PMCID: PMC11971989 DOI: 10.7717/peerj.19213] [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: 06/20/2024] [Accepted: 03/05/2025] [Indexed: 04/09/2025] Open
Abstract
This review delves into the pivotal role and intricate mechanisms of aerobic glycolysis in nasopharyngeal carcinoma (NPC). NPC, a malignancy originating from the nasopharyngeal epithelium, displays distinct geographical and clinical features. The article emphasizes the significance of aerobic glycolysis, a pivotal metabolic alteration in cancer cells, in NPC progression. Key enzymes such as hexokinase 2, lactate dehydrogenase A, phosphofructokinase 1, and pyruvate kinase M2 are discussed for their regulatory functions in NPC glycolysis through signaling pathways like PI3K/Akt and mTOR. Further, the article explores how oncogenic signaling pathways and transcription factors like c-Myc and HIF-1α modulate aerobic glycolysis, thereby affecting NPC's proliferation, invasion, metastasis, angiogenesis, and immune evasion. By elucidating these mechanisms, the review aims to advance research and clinical practice in NPC, informing the development of targeted therapeutic strategies that enhance treatment precision and reduce side effects. Overall, this review offers a broad understanding of the multifaceted role of aerobic glycolysis in NPC and its potential impact on therapeutic outcomes.
Collapse
Affiliation(s)
- Zhiyong Pan
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuyi Liu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Hui Li
- Department of Ophthalmology, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Huisi Qiu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Pingmei Zhang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhiying Li
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xinyu Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Yuxiao Tian
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Zhengfu Feng
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Song Zhu
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| | - Xin Wang
- Department of Radiotherapy, Affiliated Qingyuan Hospital, Guangzhou Medical University, Qingyuan, Guangdong, China
| |
Collapse
|
15
|
Zhang Y, Long J, Xu J, Zhong P, Wang B. Single-cell RNA sequencing reveals ECM remodeling-tumor stiffness-FAK as a key driver of vestibular schwannoma progression. Prog Neurobiol 2025; 247:102730. [PMID: 39988022 DOI: 10.1016/j.pneurobio.2025.102730] [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: 06/23/2024] [Revised: 12/17/2024] [Accepted: 02/18/2025] [Indexed: 02/25/2025]
Abstract
Vestibular schwannoma (VS), characterized by the absence of merlin expression, is the most prevalent benign tumor located at the cerebellopontine angle, lacking approved pharmaceutical interventions except for off-label utilization of bevacizumab. The role of Tumor stiffness-Focal adhesion kinase (FAK) activation in fueling tumor progression is well-established, with merlin deficiency serving as a biomarker for tumor sensitivity to FAK inhibitors. In this context, we investigated whether Tumor stiffness-FAK contributes to VS progression. Single-cell RNA sequencing revealed associations between VS progression and gene sets related to "Response to mechanical stimulus" and "Neurotrophin signaling pathway". Histological studies indicated a potential involvement of neurotrophins in early stages of VS tumorigenesis, while enhanced Extracellular matrix (ECM) remodeling-Tumor stiffness-FAK signaling accompanies later stages of VS progression. In vitro experiments demonstrated that elevated matrix stiffness induces cytoskeletal remodeling, cell proliferation, and metalloproteinase expression in VS cells by activating FAK. Conversely, FAK inhibition diminishes these effects. Collectively, this study suggests that ECM remodeling-Tumor stiffness contributes to VS progression via FAK activation, positioning FAK as a promising therapeutic target in treating VS.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Jianfei Long
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Jian Xu
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China
| | - Ping Zhong
- Department of Neurosurgery, Huashan Hospital, Fudan University, Shanghai 200032, China.
| | - Bin Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai 200032, China.
| |
Collapse
|
16
|
Niland S, Eble JA. Decoding the MMP14 integrin link: Key player in the secretome landscape. Matrix Biol 2025; 136:36-51. [PMID: 39828138 DOI: 10.1016/j.matbio.2025.01.004] [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: 08/21/2024] [Revised: 01/16/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Rapid progress has been made in the exciting field of secretome research in health and disease. The tumor secretome, which is a significant proportion of the tumor proteome, is secreted into the extracellular space to promote intercellular communication and thus tumor progression. Among the many molecules of the secretome, integrins and matrix metalloproteinase 14 (MMP14) stand out as the interplay of adhesion and proteolysis drives invasion. Integrins serve as mechanosensors that mediate the contact of cells with the scaffold of the extracellular matrix and are significantly involved in the precise positioning and activity control of the membrane-bound collagenase MMP14. As a secretome proteinase, MMP14 influences and modifies the secretome itself. While integrins and MT-MMPs are membrane bound, but can be released and are therefore border crossers between the cell surface and the secretome, the extracellular matrix is not constitutively cell-bound, but its binding to integrins and other cell receptors is a stringently regulated process. To understand the mutual interactions in detail, we first summarize the structure and function of MMP14 and how it is regulated at the enzymatic and cellular level. In particular, the mutual interactions between integrins and MMP14 include the proteolytic cleavage of integrins themselves by MMP14. We then review the biochemical, cell biological and physiological effects of MMP14 on the composition and associated functions in the tumor secretome when either bound to the cell membrane, or located on extracellular microvesicles, or as a proteolytically shed non-membrane-bound ectodomain. Novel methods of proteomics, including the analysis of extravesicular vesicles, and new methods for the quantification of MMP14 will provide new research and diagnostic tools. The proteolytic modification of the tumor secretome, especially by MMP14, may bring an additional aspect to tumor secretome studies and will have an impact on the diagnosis and most likely also on the therapy of cancer patients.
Collapse
Affiliation(s)
- Stephan Niland
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany
| | - Johannes A Eble
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster, Germany.
| |
Collapse
|
17
|
Shi Z, Hu C, Li Q, Sun C. Cancer-Associated Fibroblasts as the "Architect" of the Lung Cancer Immune Microenvironment: Multidimensional Roles and Synergistic Regulation with Radiotherapy. Int J Mol Sci 2025; 26:3234. [PMID: 40244052 PMCID: PMC11989671 DOI: 10.3390/ijms26073234] [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: 02/19/2025] [Revised: 03/20/2025] [Accepted: 03/25/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer-associated fibroblasts (CAFs), as the "architect" of the immune microenvironment in lung cancer, play a multidimensional role in tumor progression and immune regulation. In this review, we summarize the heterogeneity of the origin and the molecular phenotype of CAFs in lung cancer, and explore the complex interactions between CAFs and multiple components of the tumor microenvironment, including the regulatory relationships with innate immune cells (e.g., tumor-associated macrophages, tumor-associated neutrophils), adaptive immune cells (e.g., T cells), and extracellular matrix (ECM). CAFs significantly influence tumor progression and immunomodulation through the secretion of cytokines, remodeling of the ECM, and the regulation of immune cell function significantly affects the immune escape and treatment resistance of tumors. In addition, this review also deeply explored the synergistic regulatory relationship between CAF and radiotherapy, revealing the key role of CAF in radiotherapy-induced remodeling of the immune microenvironment, which provides a new perspective for optimizing the comprehensive treatment strategy of lung cancer. By comprehensively analyzing the multidimensional roles of CAF and its interaction with radiotherapy, this review aims to provide a theoretical basis for the precise regulation of the immune microenvironment and clinical treatment of lung cancer.
Collapse
Affiliation(s)
- Zheng Shi
- School of Biopharmaceutical and Engineering, Lanzhou Jiaotong University, Lanzhou 730070, China
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (C.H.); (Q.L.); (C.S.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Cuilan Hu
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (C.H.); (Q.L.); (C.S.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Qiang Li
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (C.H.); (Q.L.); (C.S.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, Lanzhou 730000, China; (C.H.); (Q.L.); (C.S.)
- University of Chinese Academy of Sciences, Beijing 101408, China
| |
Collapse
|
18
|
Lo Cicero A, Campora S, Lo Buglio G, Cinà P, Lo Pinto M, Scilabra SD, Ghersi G. Enhancing therapeutic efficacy through degradation of endogenous extracellular matrix in primary breast tumor spheroids. FEBS J 2025. [PMID: 40098313 DOI: 10.1111/febs.70069] [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: 07/18/2024] [Revised: 11/08/2024] [Accepted: 03/10/2025] [Indexed: 03/19/2025]
Abstract
Solid tumors have a complex extracellular matrix (ECM) that significantly affects tumor behavior and response to therapy. Understanding the ECM's role is crucial for advancing cancer research and treatment. This study established an in vitro model using primary cells isolated from a rat breast tumor to generate three-dimensional spheroids. Monolayer cells and spheroid cultures exhibited different protein expression patterns, with primary tumor spheroids presenting an increased level of ECM-related proteins and a more complex extracellular environment. Furthermore, spheroids produce endogenous collagen type I matrix, which is the main component of the tumoral ECM. This matrix is arranged predominantly around the 3D structure, mimicking the conditions of solid tumors. Treatments with recombinant collagenases class II (acting on the linear collagen region) and class I (acting on the 3D-helix region) completely degrade collagen within the spheroid structure. Collagenase pretreatment enhances the accessibility of the anticancer drug doxorubicin to penetrate the core of spheroids and sensitize them to doxorubicin-induced cytotoxicity. Our findings highlight the importance of overcoming drug resistance in breast cancer by targeting the ECM and proposing a novel strategy for improving therapeutic outcomes in solid tumors. By employing a three-dimensional spheroid model, with an endogenous ECM, we can offer more relevant insights into tumor biology and treatment responses.
Collapse
Affiliation(s)
- Alessandra Lo Cicero
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
| | - Simona Campora
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Department of Biomedical Engineering Bioscience Center of the University of Cincinnati, OH, USA
| | - Gabriele Lo Buglio
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Department of Pharmacy, University of Copenhagen, Denmark
| | | | - Margot Lo Pinto
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Simone Dario Scilabra
- Proteomics Group of Ri.MED Foundation, Research Department IRCCS ISMETT (Istituto Mediterraneo per i Trapianti e Terapie ad Alta Specializzazione), Palermo, Italy
| | - Giulio Ghersi
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Italy
- Abiel Srl, Palermo, Italy
| |
Collapse
|
19
|
Clemens C, Gehring R, Riedl P, Pompe T. Matrix deformation and mechanotransduction as markers of breast cancer cell phenotype alteration at matrix interfaces. Biomater Sci 2025; 13:1578-1589. [PMID: 39960148 DOI: 10.1039/d4bm01589d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2025]
Abstract
The dissemination of metastatic cells from the primary tumor into the surrounding tissue is a key event in the progression of cancer. This process involves the migration of cells across defined tissue interfaces that separate the dense tumor tissue from the adjacent healthy tissue. Prior research showed that cell transmigration across collagen I matrix interfaces induces a switch towards a more aggressive phenotype including a change in directionality of migration and chemosensitivity correlated to increased DNA damage during transmigration. Hence, mechanical forces acting at the nucleus during transmigration are hypothesized to trigger phenotype switching. Here, we present results from a particle image velocimetry (PIV) based live cell analysis of breast cancer cell transmigration across sharp matrix interfaces constituted of two collagen type I networks with different pore sizes. We found strong and highly localized collagen network deformation caused by cellular forces at the moment of crossing interfaces from dense into open matrices. Additionally, an increased contractility of transmigrated cells was determined for cells with the switch phenotype. Moreover, studies on mechanotransductive signaling at the nucleus, emerin translocation and YAP activation, indicated a misregulation of these signals for transmigrated cells with altered phenotype. These findings show that matrix interfaces between networks of different pore sizes mechanically challenge invasive breast cancer cells during transmigration by a strong asymmetry of contracting forces, impeding nuclear mechanotransduction pathways, with a subsequent trigger of more aggressive phenotypes.
Collapse
Affiliation(s)
- Cornelia Clemens
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Rosa Gehring
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Philipp Riedl
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| | - Tilo Pompe
- Institute of Biochemistry, Leipzig University, Johannisallee 21-23, 04103, Leipzig, Germany.
| |
Collapse
|
20
|
Liu Z, Zhang X, Ben T, Li M, Jin Y, Wang T, Song Y. Focal adhesion in the tumour metastasis: from molecular mechanisms to therapeutic targets. Biomark Res 2025; 13:38. [PMID: 40045379 PMCID: PMC11884212 DOI: 10.1186/s40364-025-00745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/11/2025] [Indexed: 03/09/2025] Open
Abstract
The tumour microenvironment is the "hotbed" of tumour cells, providing abundant extracellular support for growth and metastasis. However, the tumour microenvironment is not static and is constantly remodelled by a variety of cellular components, including tumour cells, through mechanical, biological and chemical means to promote metastasis. Focal adhesion plays an important role in cell-extracellular matrix adhesion. An in-depth exploration of the role of focal adhesion in tumour metastasis, especially their contribution at the biomechanical level, is an important direction of current research. In this review, we first summarize the assembly of focal adhesions and explore their kinetics in tumour cells. Then, we describe in detail the role of focal adhesion in various stages of tumour metastasis, especially its key functions in cell migration, invasion, and matrix remodelling. Finally, we describe the anti-tumour strategies targeting focal adhesion and the current progress in the development of some inhibitors against focal adhesion proteins. In this paper, we summarize for the first time that focal adhesion play a positive feedback role in pro-tumour metastatic matrix remodelling by summarizing the five processes of focal adhesion assembly in a multidimensional way. It is beneficial for researchers to have a deeper understanding of the role of focal adhesion in the biological behaviour of tumour metastasis and the potential of focal adhesion as a therapeutic target, providing new ideas for the prevention and treatment of metastases.
Collapse
Affiliation(s)
- Zonghao Liu
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Xiaofang Zhang
- Department of Medical Oncology, The First Hospital of China Medical University, Shenyang, Liaoning, 110001, China
| | - Tianru Ben
- The First Clinical College, China Medical University, Shenyang, Liaoning Province, 110122, P. R. China
| | - Mo Li
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Yi Jin
- Department of Breast Surgery, Liaoning Cancer Hospital and Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China
| | - Tianlu Wang
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Cancer Hospital of Dalian University of Technology, Shenyang, Liaoning Province, 110042, People's Republic of China.
- Faculty of Medicine, Dalian University of Technology, Dalian, Liaoning Province, 116024, P. R. China.
| | - Yingqiu Song
- Department of Radiotherapy, Cancer Hospital of China Medical University, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
- Department of Radiotherapy, Liaoning Cancer Hospital & Institute, No.44 Xiaoheyan Road, Dadong District, Shenyang, Liaoning Province, 110042, P. R. China.
| |
Collapse
|
21
|
Ge Y, Jiang L, Dong Q, Xu Y, Yam JWP, Zhong X. Exosome-mediated Crosstalk in the Tumor Immune Microenvironment: Critical Drivers of Hepatocellular Carcinoma Progression. J Clin Transl Hepatol 2025; 13:143-161. [PMID: 39917466 PMCID: PMC11797817 DOI: 10.14218/jcth.2024.00302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 02/09/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a significant global health issue, ranking as the sixth most prevalent malignancy and the fourth leading cause of cancer-related mortality worldwide. Despite advancements in therapeutic strategies, mortality rates for HCC remain high. The tumor immune microenvironment (TIME) plays a vital role in HCC progression by influencing tumor cell survival and growth. Recent studies highlight the essential role of exosomes in mediating intercellular communication within the TIME, particularly in interactions among tumor cells, immune cells, and fibroblasts. These interactions drive critical aspects of tumor development, including immune escape, angiogenesis, drug resistance, and metastasis. A detailed understanding of the molecular mechanisms by which exosomes modulate the TIME is essential for developing targeted therapies. This review systematically evaluated the roles and regulatory mechanisms of exosomes within the TIME of HCC, examining the impact of both HCC-derived and non-HCC-derived exosomes on various cellular components within the TIME. It emphasized their regulatory effects on cell phenotypes and functions, as well as their roles in HCC progression. The review also explored the potential applications of exosome-based immunotherapies, offering new insights into improving therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Yifei Ge
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lixue Jiang
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Qingfu Dong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yi Xu
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
- Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-targeting Theranostics, Guangxi Key Laboratory of Bio-targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Fuzhou, Fujian, China
- Fujian Provincial Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Xiangyu Zhong
- Department of Hepatopancreatobiliary Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| |
Collapse
|
22
|
Maiques O, Sallan MC, Laddach R, Pandya P, Varela A, Crosas-Molist E, Barcelo J, Courbot O, Liu Y, Graziani V, Arafat Y, Sewell J, Rodriguez-Hernandez I, Fanshawe B, Jung-Garcia Y, Imbert PR, Grasset EM, Albrengues J, Santacana M, Macià A, Tarragona J, Matias-Guiu X, Marti RM, Tsoka S, Gaggioli C, Orgaz JL, Fruhwirth GO, Wallberg F, Betteridge K, Reyes-Aldasoro CC, Haider S, Braun A, Karagiannis SN, Elosegui-Artola A, Sanz-Moreno V. Matrix mechano-sensing at the invasive front induces a cytoskeletal and transcriptional memory supporting metastasis. Nat Commun 2025; 16:1394. [PMID: 39952917 PMCID: PMC11829002 DOI: 10.1038/s41467-025-56299-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 01/13/2025] [Indexed: 02/17/2025] Open
Abstract
The extracellular matrix (ECM) controls tumour dissemination. We characterise ECM organization in human and mouse tumours, identifying three regions: tumour body, proximal invasive front and distal invasive front. Invasive areas show increased matrix density, fibre thickness, length, and alignment, with unique radial fibre orientation at the distal invasive front correlating with amoeboid invasive features. Using patient samples and murine models, we find that metastases recapitulate ECM features of the primary tumour. Ex vivo culture of murine cancer cells isolated from the different tumour regions reveals a spatial cytoskeletal and transcriptional memory. Several in vitro models recapitulate the in vivo ECM organisation showing that increased matrix induces 3D confinement supporting Rho-ROCK-Myosin II activity, while radial orientation enhances directional invasion. Spatial transcriptomics identifies a mechano-inflammatory program associated with worse prognosis across multiple tumour types. These findings provide mechanistic insights into how ECM organization shapes local invasion and distant metastasis.
Collapse
Affiliation(s)
- Oscar Maiques
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
- Cancer Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, UK
| | - Marta C Sallan
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Roman Laddach
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, SE1 9RT, London, UK
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Bush House, London, WC2B 4BG, UK
| | - Pahini Pandya
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Adrian Varela
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Eva Crosas-Molist
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Jaume Barcelo
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Olivia Courbot
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, NW1 1AT, UK
| | - Yanbo Liu
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Vittoria Graziani
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Youssef Arafat
- Department of Computer Science, City St George's, University of London, London, UK
| | - Joanne Sewell
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Irene Rodriguez-Hernandez
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
| | - Bruce Fanshawe
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
| | - Yaiza Jung-Garcia
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Paul Rc Imbert
- CMR Advanced Bio-imaging Facility, Centre for Microvascular Research, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Eloise M Grasset
- University Cote d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Jean Albrengues
- University Cote d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Maria Santacana
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida, CIBERONC, Lleida, 25198, Spain
| | - Anna Macià
- Oncologic Pathology Group, IRBLleida, Departments of Experimental Medicine and Basic Medical Sciences, University of Lleida, Lleida, 25198, Spain
| | - Jordi Tarragona
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida, CIBERONC, Lleida, 25198, Spain
| | - Xavier Matias-Guiu
- Department of Pathology and Molecular Genetics, Hospital Universitari Arnau de Vilanova, University of Lleida, IRBLleida, CIBERONC, Lleida, 25198, Spain
- Oncologic Pathology Group, IRBLleida, Departments of Experimental Medicine and Basic Medical Sciences, University of Lleida, Lleida, 25198, Spain
- Department of Pathology, Hospital Universitari de Bellvitge University of Barcelona, IDIBELL, CIBERONC, L'Hospitalet-, Barcelona, 08907, Spain
| | - Rosa M Marti
- Department of Dermatology, Hospital Universitari Arnau de Vilanova, CIBERONC, University of Lleida, CIBERONC, IRB Lleida, Lleida, 25198, Spain
| | - Sophia Tsoka
- Department of Informatics, Faculty of Natural and Mathematical Sciences, King's College London, Bush House, London, WC2B 4BG, UK
| | - Cedric Gaggioli
- University Cote d'Azur, CNRS UMR7284, INSERM U1081, Institute for Research on Cancer and Aging, Nice (IRCAN), Nice, France
| | - Jose L Orgaz
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK
- Instituto de Investigaciones Biomédicas Sols-Morreale (IIBM), CSIC-UAM, 28029, Madrid, Spain
| | - Gilbert O Fruhwirth
- Comprehensive Cancer Centre, School of Cancer and Pharmaceutical Sciences, King's College London, London, SE1 1UL, UK
| | - Fredrik Wallberg
- Quell Therapeutics, Translation & Innovation Hub, 84 Wood Ln, London, W12 0BZ, UK
- Light Microscopy Facility, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Kai Betteridge
- Light Microscopy Facility, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Constantino Carlos Reyes-Aldasoro
- Department of Computer Science, City St George's, University of London, London, UK
- Integrated Pathology Unit, Division of Molecular Pathology, The Institute of Cancer Research, Sutton, UK
| | - Syed Haider
- Breast Cancer Research Bioinformatics Group, Chester Beatty Laboratories, London, SW3 6JB, UK
| | - Andrejs Braun
- Centre for Haemato-Oncology, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK
| | - Sophia N Karagiannis
- St. John's Institute of Dermatology, School of Basic and Medical Biosciences, King's College London, SE1 9RT, London, UK
- Breast Cancer Now Research Unit, School of Cancer & Pharmaceutical Sciences, King's College London, Guy's Cancer Centre, London, SE1 9RT, UK
| | | | - Victoria Sanz-Moreno
- Cytoskeleton and metastasis Team, The Breast Cancer Now Toby Robins Research Centre Division of Breast Cancer Research, The Institute of Cancer Research, Chester Beatty Laboratories, London, SW3 6JB, UK.
- Centre for Tumour Microenvironment, Barts Cancer Institute, Queen Mary University of London, John Vane Science Building, Charterhouse Square, London, EC1M 6BQ, UK.
- Randall Division of Cell and Molecular Biophysics, King's College London, New Hunt's House, Guy's Campus, London, SE1 1UL, UK.
| |
Collapse
|
23
|
Shah S, Osuala KO, Brock EJ, Ji K, Sloane BF, Mattingly RR. Three-Dimensional Models: Biomimetic Tools That Recapitulate Breast Tissue Architecture and Microenvironment to Study Ductal Carcinoma In Situ Transition to Invasive Ductal Breast Cancer. Cells 2025; 14:220. [PMID: 39937011 PMCID: PMC11817749 DOI: 10.3390/cells14030220] [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: 12/24/2024] [Revised: 01/30/2025] [Accepted: 01/31/2025] [Indexed: 02/13/2025] Open
Abstract
Diagnosis of ductal carcinoma in situ (DCIS) presents a challenge as we cannot yet distinguish between those lesions that remain dormant from cases that may progress to invasive ductal breast cancer (IDC) and require therapeutic intervention. Our overall interest is to develop biomimetic three-dimensional (3D) models that more accurately recapitulate the structure and characteristics of pre-invasive breast cancer in order to study the underlying mechanisms driving malignant progression. These models allow us to mimic the microenvironment to investigate many aspects of mammary cell biology, including the role of the extracellular matrix (ECM), the interaction between carcinoma-associated fibroblasts (CAFs) and epithelial cells, and the dynamics of cytoskeletal reorganization. In this review article, we outline the significance of 3D culture models as reliable pre-clinical tools that mimic the in vivo tumor microenvironment and facilitate the study of DCIS lesions as they progress to invasive breast cancer. We also discuss the role of CAFs and other stromal cells in DCIS transition as well as the clinical significance of emerging technologies like tumor-on-chip and co-culture models.
Collapse
Affiliation(s)
- Seema Shah
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
| | | | - Ethan J. Brock
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
| | - Kyungmin Ji
- Department of Neurology, Henry Ford Health, Detroit, MI 48202, USA
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Bonnie F. Sloane
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48201, USA; (S.S.); (E.J.B.)
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Raymond R. Mattingly
- Department of Pharmacology, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Department of Pharmacology and Toxicology, Brody School of Medicine, East Carolina University, Greenville, NC 27834, USA
| |
Collapse
|
24
|
Klabukov I, Kabakov AE, Yakimova A, Baranovskii D, Sosin D, Atiakshin D, Ignatyuk M, Yatsenko E, Rybachuk V, Evstratova E, Eygel D, Kudlay D, Stepanenko V, Shegay P, Kaprin AD. Tumor-Associated Extracellular Matrix Obstacles for CAR-T Cell Therapy: Approaches to Overcoming. Curr Oncol 2025; 32:79. [PMID: 39996879 PMCID: PMC11854105 DOI: 10.3390/curroncol32020079] [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: 12/29/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy yields good results in the treatment of various hematologic malignancies. However, the efficacy of CAR-T cell therapy against solid tumors has proven to be limited, primarily because the tumor-associated extracellular matrix (ECM) creates an intractable barrier for the cytotoxic CAR-T cells that are supposed to kill cancer cells. This review unravels the multifaceted role of the tumor-associated ECM in impeding CAR-T cell infiltration, survival, and functions within solid tumors. We analyze the situations when intratumoral ECM limits the efficacy of CAR-T cell therapy by being a purely physical barrier that complicates lymphocyte penetration/migration and also acts as an immunosuppressive factor that impairs the antitumor activities of CAR-T cells. In addition, we highlight promising approaches such as engineering CAR-T cells with improved capabilities to penetrate and migrate into/through the intratumoral ECM, combination therapies aimed at attenuating the high density and immunosuppressive potential of the intratumoral ECM, and others that enable overcoming ECM-related obstacles. A detailed overview of the data of relevant studies not only helps to better understand the interactions between CAR-T cells and the intratumoral ECM but also outlines potential ways to more effectively use CAR-T cell therapy against solid tumors.
Collapse
Affiliation(s)
- Ilya Klabukov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
- GMP-Laboratory for Advanced Therapy Medicinal Products, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
- Obninsk Institute for Nuclear Power Engineering of the National Research Nuclear University MEPhI, Studgorodok 1, 249039 Obninsk, Russia
| | - Alexander E. Kabakov
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Anna Yakimova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Denis Baranovskii
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
- GMP-Laboratory for Advanced Therapy Medicinal Products, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklay St. 6, 117198 Moscow, Russia
- University Hospital Basel, Basel University, 4001 Basel, Switzerland
| | - Dmitry Sosin
- Centre for Strategic Planning and Management of Biomedical Health Risks of the Federal Medical Biological Agency, 119121 Moscow, Russia
| | - Dmitry Atiakshin
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Michael Ignatyuk
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Elena Yatsenko
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Victoria Rybachuk
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
| | - Ekaterina Evstratova
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Daria Eygel
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- A. Tsyb Medical Radiological Research Center—Branch of the National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Zhukova St. 10, 249036 Obninsk, Russia
| | - Dmitry Kudlay
- Immunology Department, Institute of Immunology FMBA of Russia, 115552 Moscow, Russia
- Department of Pharmacognosy and Industrial Pharmacy, Faculty of Fundamental Medicine, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Vasiliy Stepanenko
- Institute of Pharmacy, Sechenov First Moscow State Medical University (Sechenov University), 119435 Moscow, Russia
| | - Peter Shegay
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Centre of the Ministry of Health of the Russian Federation, Koroleva St. 4, 249036 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Patrice Lumumba Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
25
|
Xia Y, Huang C, Zhong M, Zhong H, Ruan R, Xiong J, Yao Y, Zhou J, Deng J. Targeting HGF/c-MET signaling to regulate the tumor microenvironment: Implications for counteracting tumor immune evasion. Cell Commun Signal 2025; 23:46. [PMID: 39856684 PMCID: PMC11762533 DOI: 10.1186/s12964-025-02033-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The hepatocyte growth factor (HGF) along with its receptor (c-MET) are crucial in preserving standard cellular physiological activities, and imbalances in the c-MET signaling pathway can lead to the development and advancement of tumors. It has been extensively demonstrated that immune checkpoint inhibitors (ICIs) can result in prolonged remission in certain patients. Nevertheless, numerous preclinical studies have shown that MET imbalance hinders the effectiveness of anti-PD-1/PD-L1 treatments through various mechanisms. Consequently, clarifying the link between the c-MET signaling pathway and the tumor microenvironment (TME), as well as uncovering the effects of anti-MET treatment on ICI therapy, is crucial for enhancing the outlook for tumor patients. In this review, we examine the impact of abnormal activation of the HGF/c-MET signaling pathway on the control of the TME and the processes governing PD-L1 expression in cancer cells. The review thoroughly examines both clinical and practical evidence regarding the use of c-MET inhibitors alongside PD-1/PD-L1 inhibitors, emphasizing that focusing on c-MET with immunotherapy enhances the effectiveness of treating MET tumors exhibiting elevated PD-L1 expression.
Collapse
Affiliation(s)
- Yang Xia
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Chunye Huang
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Min Zhong
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Hongguang Zhong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Ruiwen Ruan
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Jianping Xiong
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China
| | - Yangyang Yao
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jing Zhou
- Department of Pulmonary and Critical Care Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| | - Jun Deng
- Department of Oncology, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
- Jiangxi Key Laboratory for Individual Cancer Therapy, 17 Yongwaizheng Street, Nanchang, Jiangxi Province, 330006, China.
| |
Collapse
|
26
|
Lian H, Zhang J, Hou S, Ma S, Yu J, Zhao W, Zhao D, Zhang Z. Immunotherapy of osteosarcoma based on immune microenvironment modulation. Front Immunol 2025; 15:1498060. [PMID: 39916962 PMCID: PMC11799554 DOI: 10.3389/fimmu.2024.1498060] [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: 09/19/2024] [Accepted: 12/30/2024] [Indexed: 02/09/2025] Open
Abstract
Osteosarcoma is a highly malignant tumor with unsatisfactory therapeutic outcomes achieved by chemotherapy, radiotherapy, and surgery. As an emerging oncological treatment, immunotherapy has shown potential in the clinical management of many tumors but has a poor response rate in osteosarcoma. The immunosuppressive microenvironment in osteosarcoma is the main reason for the ineffectiveness of immunotherapy, in which the low immune response rate of immune effector cells and the high activation of immunosuppressive cells contribute to this outcome. Therefore, modulating the function of the immune microenvironment in osteosarcoma is expected to remodel the immunosuppressive microenvironment of osteosarcoma and enhance the efficacy of immunotherapy. This article reviews the role of immune cells in the progression of osteosarcoma, describes the corresponding regulatory tools for the characteristics of different cells to enhance the efficacy of osteosarcoma immunotherapy, and concludes the prospects and future challenges of osteosarcoma immunotherapy.
Collapse
Affiliation(s)
- Heping Lian
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
- Bone and Soft Tissue Tumours Research Centre of Yunnan Province, Department of Orthopaedics, The Third Affiliated Hospital of Kunming Medical University (Yunnan Cancer Hospital, Yunnan Cancer Center), Kunming, China
| | - Jiakui Zhang
- Department of Surgical Oncology, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuna Hou
- Department of General Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Shuang Ma
- Nursing Department, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Jiachen Yu
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Wei Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Duoyi Zhao
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhiyu Zhang
- Department of Orthopedics, The Fourth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
27
|
Tadgell B, García-Astrain C, Henriksen-Lacey M, Martín VF, Obelleiro-Liz M, Liz-Marzán LM. Monitoring of the Local Extracellular Environment Using Chiral Gold Nanoparticles. J Am Chem Soc 2025; 147:2860-2870. [PMID: 39779303 DOI: 10.1021/jacs.4c16686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
In three-dimensional (3D)-printed tissue models, sensitive, noninvasive techniques are required to detect in situ changes in hydrogel structure caused by cellular remodeling. We demonstrate herein that circular dichroism (CD) spectroscopy provides a reliable method for detecting hydrogel structural variations. We probe directly the plasmonic optical activity of chiral gold nanorods (c-AuNRs) embedded within the hydrogel matrix, in response to variations in the local environment. Unlike extinction spectroscopy, we found that CD features such as zero-point crossings do not get masked by the strong scattering due to the porous hydrogel structure and are therefore sensitive to changes in the refractive index of the medium, reversible swelling and deswelling of the hydrogel, and other interactions between the hydrogel and the c-AuNR surface. By culturing metastatic breast cancer cells within the 3D hydrogel nanocomposite, we demonstrate that CD can noninvasively probe the restructuring of the hydrogel. This study highlights the potential of CD spectroscopy in combination with c-AuNRs to investigate complex changes in biological or polymeric systems.
Collapse
Affiliation(s)
- Ben Tadgell
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
| | - Clara García-Astrain
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
| | - Malou Henriksen-Lacey
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián 20014, Spain
| | - Víctor F Martín
- Signal Theory and Communications Department, King Juan Carlos University, Madrid 28032, Spain
| | - Manuel Obelleiro-Liz
- Departamento Tecnología de los Computadores y de las Comunicaciones, Universidad de Extremadura, Cáceres 10003, Spain
| | - Luis M Liz-Marzán
- CIC biomaGUNE, Basque Research and Technology Alliance (BRTA), Donostia-San Sebastián 20014, Spain
- Centro de Investigación Biomédica en Red, Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Donostia-San Sebastián 20014, Spain
- Ikerbasque, Basque Foundation for Science, Bilbao 48009, Spain
| |
Collapse
|
28
|
Cáceres-Calle D, Torre-Cea I, Marcos-Zazo L, Carrera-Aguado I, Guerra-Paes E, Berlana-Galán P, Muñoz-Félix JM, Sánchez-Juanes F. Integrins as Key Mediators of Metastasis. Int J Mol Sci 2025; 26:904. [PMID: 39940673 PMCID: PMC11816423 DOI: 10.3390/ijms26030904] [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: 12/17/2024] [Revised: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 02/16/2025] Open
Abstract
Metastasis is currently becoming a major clinical concern, due to its potential to cause therapeutic resistance. Its development involves a series of phases that describe the metastatic cascade: preparation of the pre-metastatic niche, epithelial-mesenchymal transition, dissemination, latency and colonization of the new tissue. In the last few years, new therapeutic targets, such as integrins, are arising to face this disease. Integrins are transmembrane proteins found in every cell that have a key role in the metastatic cascade. They intervene in adhesion and intracellular signaling dependent on the extracellular matrix and cytokines found in the microenvironment. In this case, integrins can initiate the epithelial-mesenchymal transition, guide the formation of the pre-metastatic niche and increase tumor migration and survival. Integrins also take part in the tumor vascularization process necessary to sustain metastasis. This fact emphasizes the importance of inhibitory therapies capable of interfering with the function of integrins in metastasis.
Collapse
Affiliation(s)
- Daniel Cáceres-Calle
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Irene Torre-Cea
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Laura Marcos-Zazo
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Iván Carrera-Aguado
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Elena Guerra-Paes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Patricia Berlana-Galán
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - José M. Muñoz-Félix
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| | - Fernando Sánchez-Juanes
- Departamento de Bioquímica y Biología Molecular, Universidad de Salamanca, 37007 Salamanca, Spain; (D.C.-C.); (I.T.-C.); (L.M.-Z.); (I.C.-A.); (E.G.-P.); (P.B.-G.)
- Instituto de Investigación Biomédica de Salamanca (IBSAL), 37007 Salamanca, Spain
| |
Collapse
|
29
|
Zhou C, Zhong R, Zhang L, Yang R, Luo Y, Lei H, Li L, Cao J, Yuan Z, Tan X, Xie M, Qu H, He Z. Exploring the mechanism of rosmarinic acid in the treatment of lung adenocarcinoma based on bioinformatics methods and experimental validation. Discov Oncol 2025; 16:47. [PMID: 39812944 PMCID: PMC11735722 DOI: 10.1007/s12672-025-01784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
OBJECTIVE Rosmarinic acid (RosA) is a natural polyphenol compound that has been shown to be effective in the treatment of inflammatory disease and a variety of malignant tumors. However, its specific mechanism for the treatment of lung adenocarcinoma (LUAD) has not been fully elucidated. Therefore, this study aims to clarify the mechanism of RosA in the treatment of LUAD by integrating bioinformatics, network pharmacology and in vivo experiments, and to explore the potential of the active ingredients of traditional Chinese medicine in treating LUAD. METHODS Firstly, the network pharmacology was used to screen the RosA targets, and LUAD-related differential expressed genes (DEGs) were acquired from the GEO database. The intersection of LUAD regulated by RosA (RDEGs) was obtained through the Venn diagram. Secondly, GO and KEGG enrichment analysis of RDEGs were performed, and protein-protein interaction networks (PPIs) were constructed to identify and visualize hub RDEGs. Then, molecular docking between hub RDEGs and RosA was performed, and further evaluation was carried out by using bioinformatics for the predictive value of the hub RDEGs. Finally, the mechanism of RosA in the treatment of LUAD was verified by establishing a xenograft model of NSCLC in nude mouse. RESULTS Bioinformatics and other analysis showed that, compared with the control group, the expressions of MMP-1, MMP-9, IGFBP3 and PLAU in LUAD tissues were significantly up-regulated, and the expressions of PPARG and FABP4 were significantly down-regulated, and these hub RDEGs had potential predictive value for LUAD. In vivo experimental results showed that RosA could inhibit the growth of transplanted tumors in nude mice bearing tumors of lung cancer cells, reduce the positive expression of Ki67 in lung tumor tissue, and hinder the proliferation of lung tumor cells. Upregulated expression of PPARG and FABP4 by activating the PPAR signaling pathway increases the level of ROS in lung tumor tissues and promotes apoptosis of lung tumor cells. In addition, RosA can also reduce the expression of MMP-9 and IGFBP3, inhibit the migration and invasion of lung tumor tissue cells. CONCLUSIONS This study demonstrated that RosA could induce apoptosis by regulating the PPAR signaling pathway and the expression of MMP-9, inhibit the proliferation, migration and invasion of lung cancer cells, thereby exerting anti-LUAD effects. This study provides new insight into the potential mechanism of RosA in treating LUAD and provides a new therapeutic avenue for treatment of LUAD.
Collapse
Affiliation(s)
- Chaowang Zhou
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Ruqian Zhong
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
| | - Lei Zhang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Renyi Yang
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
| | - Yuxin Luo
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Huijun Lei
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Liang Li
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Jianzhong Cao
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Provincial Key Laboratory of Traditional Chinese Medicine Diagnostics, Changsha, 410208, Hunan, China
| | - Zhiying Yuan
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China
| | - Xiaoning Tan
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Yuelu District, Changsha, 410006, Hunan, China
| | - Mengzhou Xie
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China.
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China.
| | - Haoyu Qu
- Hunan University of Chinese Medicine, 300 Xueshi Road, Yuelu District, Changsha, 410208, Hunan, China.
- Hunan Engineering Technology Research Center for Medicinal and Functional Food, Changsha, 410208, Hunan, China.
| | - Zuomei He
- Hunan Provincial Hospital of Integrated Traditional Chinese and Western Medicine, No. 58, Yuelu District, Changsha, 410006, Hunan, China.
| |
Collapse
|
30
|
Lei KF, Bai KC, Pai PC. Study of cell migration trajectory on two-dimensional continuous stiffness gradient surface edited by grayscale photopolymerization. Talanta 2025; 281:126899. [PMID: 39298803 DOI: 10.1016/j.talanta.2024.126899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/06/2024] [Accepted: 09/15/2024] [Indexed: 09/22/2024]
Abstract
In native tissues, cells encounter a diverse range of stiffness, which can significantly affect their behavior and function. The ability of cells to sense and respond to these mechanical cues is essential for various physiological processes, including cell migration. Cell migration is a complex process influenced by multiple factors, with substrate stiffness emerging as a critical determinant. This study developed a technique to edit the stiffness of polyacrylamide (PAA) hydrogel substrates by adjusting the grayscale level of a photomask during photopolymerization. By analyzing cell morphologies on the hydrogel, we confirmed the development of a single PAA hydrogel substrate with continuous stiffness gradients. This method was used to explore the correlation between substrate stiffness and cell migration dynamics. The study found that cells typically migrated from softer to stiffer surfaces. When the cells initially located on stiffer surfaces, they were able to travel longer distances. Additionally, a continuous 2D stiffness gradient surface was fabricated to explore how cells migrate on smoother versus steeper stiffness gradients. The results showed that cells tended to migrate more readily on smoother stiffness gradient surfaces compared to steeper ones. This study provides valuable insights into cell migration dynamics on substrates with varying stiffness gradients. The results underscore the importance of the mechanical environment in cancer cell migration and offer promising directions for developing interventions to prevent cancer spread.
Collapse
Affiliation(s)
- Kin Fong Lei
- Department of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan; Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan; Department of Electrical & Electronic Engineering, Yonsei University, Seoul, 03722, South Korea.
| | - Kuo-Cheng Bai
- Department of Biomedical Engineering, Chang Gung University, Taoyuan, 33302, Taiwan
| | - Ping-Ching Pai
- Department of Radiation Oncology, Chang Gung Memorial Hospital, Linkou, Taoyuan, 33305, Taiwan
| |
Collapse
|
31
|
Zhang X, Al‐Danakh A, Zhu X, Feng D, Yang L, Wu H, Li Y, Wang S, Chen Q, Yang D. Insights into the mechanisms, regulation, and therapeutic implications of extracellular matrix stiffness in cancer. Bioeng Transl Med 2025; 10:e10698. [PMID: 39801760 PMCID: PMC11711218 DOI: 10.1002/btm2.10698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2024] [Revised: 06/19/2024] [Accepted: 06/29/2024] [Indexed: 01/03/2025] Open
Abstract
The tumor microenvironment (TME) is critical for cancer initiation, growth, metastasis, and therapeutic resistance. The extracellular matrix (ECM) is a significant tumor component that serves various functions, including mechanical support, TME regulation, and signal molecule generation. The quantity and cross-linking status of ECM components are crucial factors in tumor development, as they determine tissue stiffness and the interaction between stiff TME and cancer cells, resulting in aberrant mechanotransduction, proliferation, migration, invasion, angiogenesis, immune evasion, and treatment resistance. Therefore, broad knowledge of ECM dysregulation in the TME might aid in developing innovative cancer therapies. This review summarized the available information on major ECM components, their functions, factors that increase and decrease matrix stiffness, and related signaling pathways that interplay between cancer cells and the ECM in TME. Moreover, mechanotransduction alters during tumorogenesis, and current drug therapy based on ECM as targets, as well as future efforts in ECM and cancer, are also discussed.
Collapse
Affiliation(s)
- Ximo Zhang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Abdullah Al‐Danakh
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Xinqing Zhu
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Dan Feng
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Linlin Yang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Haotian Wu
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
| | - Yingying Li
- Department of Discipline ConstructionDalian Medical UniversityDalianChina
| | - Shujing Wang
- Department of Biochemistry and Molecular Biology, Institute of GlycobiologyDalian Medical UniversityDalianChina
| | - Qiwei Chen
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Zhongda Hospital, Medical School Advanced Institute Life HealthSoutheast UniversityNanjingChina
| | - Deyong Yang
- Department of UrologyFirst Affiliated Hospital of Dalian Medical UniversityDalianChina
- Department of SurgeryHealinghands ClinicDalianChina
| |
Collapse
|
32
|
Ahmad I, Altameemi KKA, Hani MM, Ali AM, Shareef HK, Hassan ZF, Alubiady MHS, Al-Abdeen SHZ, Shakier HG, Redhee AH. Shifting cold to hot tumors by nanoparticle-loaded drugs and products. Clin Transl Oncol 2025; 27:42-69. [PMID: 38922537 DOI: 10.1007/s12094-024-03577-3] [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: 05/28/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024]
Abstract
Cold tumors lack antitumor immunity and are resistant to therapy, representing a major challenge in cancer medicine. Because of the immunosuppressive spirit of the tumor microenvironment (TME), this form of tumor has a low response to immunotherapy, radiotherapy, and also chemotherapy. Cold tumors have low infiltration of immune cells and a high expression of co-inhibitory molecules, such as immune checkpoints and immunosuppressive molecules. Therefore, targeting TME and remodeling immunity in cold tumors can improve the chance of tumor repression after therapy. However, tumor stroma prevents the infiltration of inflammatory cells and hinders the penetration of diverse molecules and drugs. Nanoparticles are an intriguing tool for the delivery of immune modulatory agents and shifting cold to hot tumors. In this review article, we discuss the mechanisms underlying the ability of nanoparticles loaded with different drugs and products to modulate TME and enhance immune cell infiltration. We also focus on newest progresses in the design and development of nanoparticle-based strategies for changing cold to hot tumors. These include the use of nanoparticles for targeted delivery of immunomodulatory agents, such as cytokines, small molecules, and checkpoint inhibitors, and for co-delivery of chemotherapy drugs and immunomodulatory agents. Furthermore, we discuss the potential of nanoparticles for enhancing the efficacy of cancer vaccines and cell therapy for overcoming resistance to treatment.
Collapse
Affiliation(s)
- Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | | | - Mohaned Mohammed Hani
- Department of Medical Instrumentation Engineering Techniques, Imam Ja'afar Al-Sadiq University, Al Muthanna, Iraq
| | - Afaq Mahdi Ali
- Department of Pharmaceutics, Al-Turath University College, Baghdad, Iraq
| | - Hasanain Khaleel Shareef
- Department of Medical Biotechnology, College of Science, Al-Mustaqbal University, Hilla, Iraq
- Biology Department, College of Science for Women, University of Babylon, Hilla, Iraq
| | | | | | | | | | - Ahmed Huseen Redhee
- Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
33
|
Muhammad FA, Altalbawy FMA, Mandaliya V, Saraswat SK, Rekha MM, Aulakh D, Chahar M, Mahdi MS, Jaber MA, Alhadrawi M. Targeting breast tumor extracellular matrix and stroma utilizing nanoparticles. Clin Transl Oncol 2024:10.1007/s12094-024-03793-x. [PMID: 39692807 DOI: 10.1007/s12094-024-03793-x] [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: 09/30/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Breast cancer is a complicated malignancy and is known as the most common cancer in women. Considerable experiments have been devoted to explore the basic impacts of the tumor stroma, particularly the extracellular matrix (ECM) and stromal components, on tumor growth and resistance to treatment. ECM is made up of an intricate system of proteins, glycosaminoglycans, and proteoglycans, and maintains structural support and controls key signaling pathways involved in breast tumors. ECM can block different drugs such as chemotherapy and immunotherapy drugs from entering the tumor stroma. Furthermore, the stromal elements, such as cancer-associated fibroblasts (CAFs), immune cells, and blood vessels, have crucial impacts on tumor development and therapeutic resistance. Recently, promising outcomes have been achieved in using nanotechnology for delivering drugs to tumor stroma and crossing ECM in breast malignancies. Nanoparticles have various benefits for targeting the breast tumor stroma, such as improved permeability and retention, extended circulation time, and the ability to actively target the area. This review covers the latest developments in nanoparticle therapies that focus on breast tumor ECM and stroma. We will explore different approaches using nanoparticles to target the delivery of anticancer drugs like chemotherapy, small molecule drugs, various antitumor products, and other specific synthetic therapeutic agents to the breast tumor stroma. Furthermore, we will investigate the utilization of nanoparticles in altering the stromal elements, such as reprogramming CAFs and immune cells, and also remodeling ECM.
Collapse
Affiliation(s)
| | - Farag M A Altalbawy
- Department of Chemistry, University College of Duba, University of Tabuk, Tabuk, Saudi Arabia.
- National Institute of Laser Enhanced Sciences (NILES), University of Cairo, Giza, 12613, Egypt.
| | - Viralkumar Mandaliya
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | | | - M M Rekha
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Damanjeet Aulakh
- Centre for Research Impact and Outcome, Chitkara University Institute of Engineering and Technology Chitkara University, Rajpura, Punjab, 140401, India
| | - Mamata Chahar
- Department of Chemistry, NIMS Institute of Engineering and Technology, NIMS University Rajasthan, Jaipur, India
| | | | | | - Merwa Alhadrawi
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University, Najaf, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Refrigeration and air Conditioning Techniques, College of Technical Engineering, The Islamic University of Babylon, Babylon, Iraq
| |
Collapse
|
34
|
Lv K, He T. Cancer-associated fibroblasts: heterogeneity, tumorigenicity and therapeutic targets. MOLECULAR BIOMEDICINE 2024; 5:70. [PMID: 39680287 PMCID: PMC11649616 DOI: 10.1186/s43556-024-00233-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/04/2024] [Accepted: 11/19/2024] [Indexed: 12/17/2024] Open
Abstract
Cancer, characterized by its immune evasion, active metabolism, and heightened proliferation, comprises both stroma and cells. Although the research has always focused on parenchymal cells, the non-parenchymal components must not be overlooked. Targeting cancer parenchymal cells has proven to be a formidable challenge, yielding limited success on a broad scale. The tumor microenvironment(TME), a critical niche for cancer cell survival, presents a novel way for cancer treatment. Cancer-associated fibroblast (CAF), as a main component of TME, is a dynamically evolving, dual-functioning stromal cell. Furthermore, their biological activities span the entire spectrum of tumor development, metastasis, drug resistance, and prognosis. A thorough understanding of CAFs functions and therapeutic advances holds significant clinical implications. In this review, we underscore the heterogeneity of CAFs by elaborating on their origins, types and function. Most importantly, by elucidating the direct or indirect crosstalk between CAFs and immune cells, the extracellular matrix, and cancer cells, we emphasize the tumorigenicity of CAFs in cancer. Finally, we highlight the challenges encountered in the exploration of CAFs and list targeted therapies for CAF, which have implications for clinical treatment.
Collapse
Affiliation(s)
- Keke Lv
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China
| | - Tianlin He
- Department of Hepatopanreatobiliary Surgery, Changhai Hospital, 168 Changhai Road, Yangpu District, Shanghai, 200433, China.
| |
Collapse
|
35
|
Fan G, Zhang C. FNDC1 Facilitates Proliferation, Migration, and Invasion of Breast Cancer Cells Through Modulating Wnt/β-Catenin Pathway. Biochem Genet 2024:10.1007/s10528-024-10994-0. [PMID: 39671143 DOI: 10.1007/s10528-024-10994-0] [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: 10/17/2024] [Accepted: 12/03/2024] [Indexed: 12/14/2024]
Abstract
Breast cancer is the most common malignant cancer and the leading fatal cancer in women around the world. Fibronectin type III domain-containing protein 1 (FNDC1) has been demonstrated to play crucial roles in various tumors. However, the function of FNDC1 in breast cancer remains to be addressed. Increased FNDC1 expression was found in breast cancer that is associated with individual cancer stages and lymph node metastasis through UALCAN analysis. Quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot assays indicated that FNDC1 expression was up-regulated in breast cancer cells. The results of Cell Counting Kit-8 and colony formation assays indicated that FNDC1 promoted the proliferation of breast cancer cells. Moreover, FNDC1 knockdown suppressed xenograft tumor growth and inhibited the levels of FNDC1 and marker of proliferation Ki-67. Transwell assay demonstrated that FNDC1 promoted the migration and invasion of breast cancer cells. Importantly, mechanism analysis implied that FNDC1 promoted the Wnt/β-catenin signaling pathway. Notably, Wnt/β-catenin activation with LiCl significantly enhanced the proliferation and epithelial-mesenchymal transformation (EMT) inhibition effect of silencing FNDC1, whereas Wnt/β-catenin inhibition with XAV-939 significantly weakened the proliferation and EMT promotion effect of FNDC1. Analysis of β-catenin expression in the nucleus and cytoplasm showed that FNDC1 promoted β-catenin nuclear translocation. These data suggested that FNDC1 exerts its oncogene function through modulating Wnt/β-catenin signaling pathway. In conclusion, FNDC1 promotes cell proliferation, migration, invasion, and EMT through modulating Wnt/β-catenin signaling pathway in breast cancer, providing a new idea for the development of breast cancer therapeutic targets.
Collapse
Affiliation(s)
- Guocai Fan
- Department of Breast Surgery, The People's Hospital of Suichang County, Lishui, 323300, China
| | - Chen Zhang
- Department of Gynecology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), No. 54, Youdian Road, Shangcheng District, Hangzhou, 310006, Zhejiang, China.
| |
Collapse
|
36
|
Shen P, Ma Z, Xu X, Li W, Li Y. Dental pulp stem cells promote malignant transformation of oral epithelial cells through mitochondrial transfer. Med Mol Morphol 2024; 57:306-319. [PMID: 39122902 DOI: 10.1007/s00795-024-00403-1] [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: 05/05/2024] [Accepted: 08/01/2024] [Indexed: 08/12/2024]
Abstract
Oral epithelial dysplasia includes a range of clinical oral mucosal diseases with potentially malignant traits. Dental pulp stem cells (DPSCs) are potential candidates for cell-based therapies targeting various diseases. However, the effect of DPSCs on the progression of oral mucosal precancerous lesions remains unclear. Animal experiments were conducted to assess the effect of human DPSCs (hDPSCs). We measured the proliferation, motility and mitochondrial respiratory function of the human dysplastic oral keratinocyte (DOK) cells cocultured with hDPSCs. Mitochondrial transfer experiments were performed to determine the role mitochondria from hDPSCs in the malignant transformation of DOK cells. hDPSCs injection accelerated carcinogenesis in 4NQO-induced oral epithelial dysplasia in mice. Coculture with hDPSCs increased the proliferation, migration, invasion and mitochondrial respiratory function of DOK cells. Mitochondria from hDPSCs could be transferred to DOK cells, and activated mTOR signaling pathway in DOK cells. Our study demonstrates that hDPSCs activate the mTOR signaling pathway through mitochondrial transfer, promoting the malignant transformation of oral precancerous epithelial lesions.
Collapse
Affiliation(s)
- Peiqi Shen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Zeyi Ma
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Xiaoqing Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Weiyu Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China
| | - Yaoyin Li
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-Sen University, Guangzhou, Guangdong, 510055, People's Republic of China.
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, Guangdong, China.
| |
Collapse
|
37
|
Wenta T, Nastaly P, Lipinska B, Manninen A. Remodeling of the extracellular matrix by serine proteases as a prerequisite for cancer initiation and progression. Matrix Biol 2024; 134:197-219. [PMID: 39500383 DOI: 10.1016/j.matbio.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 10/16/2024] [Accepted: 10/31/2024] [Indexed: 11/25/2024]
Abstract
The extracellular matrix (ECM) serves as a physical scaffold for tissues that is composed of structural proteins such as laminins, collagens, proteoglycans and fibronectin, forming a three dimensional network, and a wide variety of other matrix proteins with ECM-remodeling and signaling functions. The activity of ECM-associated signaling proteins is tightly regulated. Thus, the ECM serves as a reservoir for water and growth regulatory signals. The ECM architecture is dynamically modulated by multiple serine proteases that process both structural and signaling proteins to regulate physiological processes such as organogenesis and tissue homeostasis but they also contribute to pathological events, especially cancer progression. Here, we review the current literature regarding the role of ECM remodeling by serine proteases (KLKs, uPA, furin, HtrAs, granzymes, matriptase, hepsin) in tumorigenesis.
Collapse
Affiliation(s)
- Tomasz Wenta
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland.
| | - Paulina Nastaly
- Laboratory of Translational Oncology, Intercollegiate Faculty of Biotechnology, University of Gdansk and Medical University of Gdansk, Gdansk, Poland
| | - Barbara Lipinska
- Department of General and Medical Biochemistry, Faculty of Biology, University of Gdansk, Poland
| | - Aki Manninen
- Disease Networks Research Unit, Faculty of Biochemistry and Molecular Medicine & Biocenter Oulu, University of Oulu, Oulu, Finland.
| |
Collapse
|
38
|
Qi B, Zhang H, Zhu J, Wang M, Ma C, Genin GM, Lu TJ, Liu S. Estimates of natural frequencies for nuclear vibration, and an assessment of the feasibility of selective ultrasound ablation of cancer cells. J Mech Behav Biomed Mater 2024; 160:106778. [PMID: 39413547 DOI: 10.1016/j.jmbbm.2024.106778] [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: 05/21/2024] [Revised: 10/07/2024] [Accepted: 10/11/2024] [Indexed: 10/18/2024]
Abstract
Selective ablation of cancer cells by ultrasound would be transformative for cancer therapy, but has not yet been possible. A key challenge is that cancerous and non-cancerous cells typically have similar acoustic impedance and are thus indistinguishable as materials in their responses to ultrasound. However, in certain cancers, cytoskeletal and nuclear lamin structures differ between healthy and malignant cells, opening the possibility of exploiting structural differences that manifest as different vibrational responses. To assess the possibility that the nuclei of certain cancerous cells might vibrate at different frequencies, we measured sizes and effective indentation moduli of a range of cancerous and non-cancerous cells from several cell lines and regions of the brain, and estimated the natural frequencies for nuclear vibration. Results suggest a potential difference in natural frequency for nuclear vibration between certain cancerous and non-cancerous cells, on the order of tens of kHz. This gap is potentially sufficient for selective ablation and motivates future experimentation on these specific cell types.
Collapse
Affiliation(s)
- Bing Qi
- State Key Laboratory of Mechanics and Control for Aerospace Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, PR China
| | - Hao Zhang
- State Key Laboratory of Mechanics and Control for Aerospace Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, PR China
| | - Junhao Zhu
- Department of Neurosurgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Ming Wang
- MOE Key Laboratory of Biomedical Information Engineering, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, PR China; Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, PR China
| | - Chiyuan Ma
- Department of Neurosurgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002, PR China
| | - Guy M Genin
- National Science Foundation Science and Technology Center for Engineering Mechanobiology, Washington University, St. Louis, MO, 63130, USA
| | - Tian Jian Lu
- State Key Laboratory of Mechanics and Control for Aerospace Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, PR China.
| | - Shaobao Liu
- State Key Laboratory of Mechanics and Control for Aerospace Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, PR China; MIIT Key Laboratory of Multifunctional Lightweight Materials and Structures, Nanjing University of Aeronautics and Astronautics, Nanjing, 210016, PR China.
| |
Collapse
|
39
|
Wang H, Wang T, Yan S, Tang J, Zhang Y, Wang L, Xu H, Tu C. Crosstalk of pyroptosis and cytokine in the tumor microenvironment: from mechanisms to clinical implication. Mol Cancer 2024; 23:268. [PMID: 39614288 PMCID: PMC11607834 DOI: 10.1186/s12943-024-02183-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 11/22/2024] [Indexed: 12/01/2024] Open
Abstract
In the realm of cancer research, the tumor microenvironment (TME) plays a crucial role in tumor initiation and progression, shaped by complex interactions between cancer cells and surrounding non-cancerous cells. Cytokines, as essential immunomodulatory agents, are secreted by various cellular constituents within the TME, including immune cells, cancer-associated fibroblasts, and cancer cells themselves. These cytokines facilitate intricate communication networks that significantly influence tumor initiation, progression, metastasis, and immune suppression. Pyroptosis contributes to TME remodeling by promoting the release of pro-inflammatory cytokines and sustaining chronic inflammation, impacting processes such as immune escape and angiogenesis. However, challenges remain due to the complex interplay among cytokines, pyroptosis, and the TME, along with the dual effects of pyroptosis on cancer progression and therapy-related complications like cytokine release syndrome. Unraveling these complexities could facilitate strategies that balance inflammatory responses while minimizing tissue damage during therapy. This review delves into the complex crosstalk between cytokines, pyroptosis, and the TME, elucidating their contribution to tumor progression and metastasis. By synthesizing emerging therapeutic targets and innovative technologies concerning TME, this review aims to provide novel insights that could enhance treatment outcomes for cancer patients.
Collapse
Affiliation(s)
- Hua Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Tao Wang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Shuxiang Yan
- Department of Nephrology, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Jinxin Tang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Yibo Zhang
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China
| | - Liming Wang
- School of Biomedical Sciences, Hunan University, Changsha, Hunan, 410011, China.
| | - Haodong Xu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Center for Precision Health, McWilliams School of Biomedical Informatics, The University of Texas Health Science Center at Houston, Houston, TX, 77030, USA.
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, 410011, China.
- Shenzhen Research Institute of Central South University, Guangdong, 518063, China.
- Hunan Engineering Research Center of AI Medical Equipment, The Second Xiangya Hospital of Central, South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
40
|
Roszkowska M. Multilevel Mechanisms of Cancer Drug Resistance. Int J Mol Sci 2024; 25:12402. [PMID: 39596466 PMCID: PMC11594576 DOI: 10.3390/ijms252212402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Cancer drug resistance represents one of the most significant challenges in oncology and manifests through multiple interconnected molecular and cellular mechanisms. Objective: To provide a comprehensive analysis of multilevel processes driving treatment resistance by integrating recent advances in understanding genetic, epigenetic, and microenvironmental factors. This is a systematic review of the recent literature focusing on the mechanisms of cancer drug resistance, including genomic studies, clinical trials, and experimental research. Key findings include the following: (1) Up to 63% of somatic mutations can be heterogeneous within individual tumors, contributing to resistance development; (2) cancer stem cells demonstrate enhanced DNA repair capacity and altered metabolic profiles; (3) the tumor microenvironment, including cancer-associated fibroblasts and immune cell populations, plays a crucial role in promoting resistance; and (4) selective pressure from radiotherapy drives the emergence of radioresistant phenotypes through multiple adaptive mechanisms. Understanding the complex interplay between various resistance mechanisms is essential for developing effective treatment strategies. Future therapeutic approaches should focus on combination strategies that target multiple resistance pathways simultaneously, guided by specific biomarkers.
Collapse
Affiliation(s)
- Malgorzata Roszkowska
- Department of Clinical Neuropsychology, Collegium Medicum, Nicolaus Copernicus University, 85-067 Bydgoszcz, Poland
| |
Collapse
|
41
|
Dalpati N, Rai SK, Dash SP, Kumar P, Singh D, Sarangi PP. Integrins α5β1 and αvβ3 Differentially Participate in the Recruitment and Reprogramming of Tumor-associated Macrophages in the In Vitro and In Vivo Models of Breast Tumor. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:1553-1568. [PMID: 39330703 DOI: 10.4049/jimmunol.2400180] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Accepted: 09/10/2024] [Indexed: 09/28/2024]
Abstract
Tumor-associated macrophages (TAMs) drive the protumorigenic responses and facilitate tumor progression via matrix remodeling, angiogenesis, and immunosuppression by interacting with extracellular matrix proteins via integrins. However, the expression dynamics of integrin and its correlation with TAM functional programming in the tumors remain unexplored. In this study, we examined surface integrins' role in TAM recruitment and phenotypic programming in a 4T1-induced murine breast tumor model. Our findings show that integrin α5β1 is upregulated in CD11b+Ly6Chi monocytes in the bone marrow and blood by day 10 after tumor induction. Subsequent analysis revealed elevated integrin α5β1 expression on tumor-infiltrating monocytes (Ly6ChiMHC class II [MHCII]low) and M1 TAMs (F4/80+Ly6ClowMHCIIhi), whereas integrin αvβ3 was predominantly expressed on M2 TAMs (F4/80+Ly6ClowMHCIIlow), correlating with higher CD206 and MERTK expression. Gene profiling of cells sorted from murine tumors showed that CD11b+Ly6G-F4/80+α5+ TAMs had elevated inflammatory genes (IL-6, TNF-α, and STAT1/2), whereas CD11b+Ly6G-F4/80+αv+ TAMs exhibited a protumorigenic phenotype (IL-10, Arg1, TGF-β, and STAT3/6). In vitro studies demonstrated that blocking integrin α5 and αv during macrophage differentiation from human peripheral blood monocytes reduced cell spreading and expression of CD206 and CD163 in the presence of specific matrix proteins, fibronectin, and vitronectin. Furthermore, RNA sequencing data analysis (GEO dataset: GSE195857) from bone marrow-derived monocytes and TAMs in 4T1 mammary tumors revealed differential integrin α5 and αv expression and their association with FAK and SRC kinase. In line with this, FAK inhibition during TAM polarization reduced SRC, STAT1, and STAT6 phosphorylation. In conclusion, these findings underscore the crucial role of integrins in TAM recruitment, polarization, and reprogramming in tumors.
Collapse
Affiliation(s)
- Nibedita Dalpati
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shubham Kumar Rai
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Shiba Prasad Dash
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Puneet Kumar
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Divya Singh
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| | - Pranita P Sarangi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee, Uttarakhand, India
| |
Collapse
|
42
|
Han Q, Yang F, Chen M, Zhang M, Wang L, Wang H, Liu J, Cao Z. Coating Dormant Collagenase-Producing Bacteria with Metal-Anesthetic Networks for Precision Tumor Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2407402. [PMID: 39291426 PMCID: PMC11558152 DOI: 10.1002/advs.202407402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/09/2024] [Indexed: 09/19/2024]
Abstract
Tumor malignancy highly depends on the stiffness of tumor matrix, which mainly consists of collagen. Despite the destruction of tumor matrix is conducive to tumor therapy, it causes the risk of tumor metastasis. Here, metal-anesthetic network-coated dormant collagenase-producing Clostridium is constructed to simultaneously destruct tumor matrix and inhibit tumor metastasis. By metal-phenolic complexation and π-π stacking interactions, a Fe3+-propofol network is formed on bacterial surface. Coated dormant Clostridium can selectively germinate and rapidly proliferate in tumor sites due to the ability of carried Fe3+ ions to promote bacterial multiplication. Intratumoral colonization of Clostridium produces sufficient collagenases to degrade tumor collagen mesh and the loaded propofol restrains tumor metastasis by inhibiting tumor cell migration and invasion. Meanwhile, the delivered Fe3+ ions are reduced to the Fe2+ form by intracellular glutathione, thereby inducing potent Fenton reaction to trigger lipid peroxidation and ultimate ferroptosis of tumor cells. In addition to a satisfactory safety, a single intratumoral injection of coated dormant Clostridium not only effectively retards the growth of established large primary tumors, but also significantly suppresses distal lung metastasis in two different orthotopic tumor models. This work proposes a strategy to develop advanced therapeutics for malignant tumor treatment and metastasis prevention.
Collapse
Affiliation(s)
- Qiuju Han
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Fengmin Yang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| | - Mian Chen
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Mengmeng Zhang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| | - Lu Wang
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| | - Hongxia Wang
- Department of Medical OncologyFudan University Shanghai Cancer CenterDepartment of OncologyShanghai Medical CollegeFudan UniversityShanghai200032China
| | - Jinyao Liu
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
- School of Chemistry and Chemical EngineeringShanghai Jiao Tong UniversityShanghai200240China
| | - Zhenping Cao
- Shanghai Key Laboratory for Nucleic Acid Chemistry and NanomedicineInstitute of Molecular MedicineState Key Laboratory of Systems Medicine for CancerShanghai Cancer InstituteRenji HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghai200127China
| |
Collapse
|
43
|
Hammad R, Kandeel EZ, Lambert C, Sack U, Kujumdshiev S, Kamhawy A, Abo-Elkheir OI, Abd El Hakam FEZ, Mashaal A, Ramadan M, Zidan AAA, Hamdy NM. Leukemic B cells expression of CD200 and Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1, CD305) in Chronic Lymphocytic Leukemia patients in relation to Treg frequency. Pathol Res Pract 2024; 263:155669. [PMID: 39471525 DOI: 10.1016/j.prp.2024.155669] [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: 08/09/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 11/01/2024]
Abstract
BACKGROUND Chronic lymphocytic leukemia (CLL) is characterized by a wide range of tumor-induced immune alterations. Regulatory T cells (Treg) play a central role in these immune responses. CD200 and Leukocyte-associated immunoglobulin-like receptor-1 (LAIR-1, CD305) are inhibitory markers said to be involved in Treg immune response. We aimed to analyze the expression of CD200 and LAIR-1 on leukemic cells and assess their interactions with the Treg frequency to elucidate their role in the CLL course. SUBJECTS AND METHODS This study was conducted on 70 participants: 50 newly diagnosed CLL cases classified according to Rai staging system into group 1 (n = 25) patients with stages 0, I, and II, and group 2 (n = 25) of advanced patients with stages III and IV. In addition to control group (n = 20) of healthy adults. Flow cytometry was used to investigate Treg frequency in bone marrow (BM) proportional to CD4+ T cell and to assess leukemic cell expression of CD200 and LAIR-1. Also, in-silico database analysis was performed to identify study markers interactions for future personalized target therapy. RESULTS Comparison between CLL groups 1 and 2 revealed increased leukemic cell percentage expressing LAIR-1 (p = 0.021) in group 1. Group 2 showed significant increase in frequency of Treg in BM and leukemic cells expressing CD200. There was a strong positive correlation between frequency of Treg and leukemic cells expressing CD200 (r = 0.669, p = 0.000). On the other hand, there was a negative correlation between frequency of Treg and leukemic cell expressing LAIR-1 (r = -0.342, p = 0.015). ROC curve analysis revealed that increased frequency of leukemic cells expressing CD200 yielded sensitivity (SN) and specificity (SP) of 96 % and 84 %, respectively in detecting CLL progression, with an AUC of 0.965. Leukemic cell percentages expressing LAIR-1 yielded a lower SN (75 %), SP (72 %), with an AUC of 0.688. CONCLUSION Treg frequency in BM was significantly increased in CLL advanced stages according to Rai classification. Leukemic cells CD200 and LAIR-1 expression were differently associated with Treg frequency. Increased CD200 expressions on leukemic cells can be considered a sensitive and specific biomarker in detecting CLL progression. As demonstrated by the in-silico research, CD200 blockade targeting may offer therapeutic benefits for CLL treatment through Treg suppression.
Collapse
MESH Headings
- Humans
- Receptors, Immunologic/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/metabolism
- Male
- Female
- Aged
- Middle Aged
- Antigens, CD/metabolism
- Adult
- Aged, 80 and over
- B-Lymphocytes/immunology
- B-Lymphocytes/metabolism
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/metabolism
Collapse
Affiliation(s)
- Reham Hammad
- Clinical Pathology Department, Faculty of Medicine (for Girls), Al-Azhar University, Nasr City, Cairo 11754, Egypt
| | - Eman Z Kandeel
- Clinical Pathology Department, National Cancer Institute, Cairo University, Cairo 11976, Egypt
| | - Claude Lambert
- Cytometry unit, Immunology Laboratory, Saint-Etienne University Hospital, France; LCOMS/ENOSIS, University of Lorraine - Metz; International Federation of Clinical Chemistry and Laboratory medicine Flow cytometry working group (IFCC FC-WG)
| | - Ulrich Sack
- Institute for Clinical Immunology, Leipzig University Hospital, Germany
| | - Sandy Kujumdshiev
- German University of Applied Sciences for Health and Sports, Berlin, Germany
| | - Arwa Kamhawy
- Medical Microbiology and Immunology Department, Faculty of Medicine for Girls, Al-Azhar University, Nasr City, Cairo 11754, Egypt
| | - Omaima I Abo-Elkheir
- Community Medicine and Public Health, Faculty of Medicine, Al-Azhar University, Nasr City, Cairo 11754, Egypt
| | | | - Alya Mashaal
- Immunology, Zoology & Entomology Department, Faculty of Science for Girls (Cairo), Al-Azhar University, Nasr City, Cairo 11754, Egypt
| | - Mohammed Ramadan
- Clinical Pathology Department, Faculty of Medicine (for Boys), Al-Azhar University (Assiut), Egypt
| | - Abdel-Aziz A Zidan
- Immunology Division; Zoology Department; Faculty of Science; Damanhour University, Egypt
| | - Nadia M Hamdy
- Biochemistry Department, Faculty of Pharmacy, Ain Shams University, Abbasia, Cairo 11566, Egypt.
| |
Collapse
|
44
|
Zhou L, Zhang W, Hu X, Wang D, Tang D. Metabolic Reprogramming of Cancer-Associated Fibroblast in the Tumor Microenvironment: From Basics to Clinic. Clin Med Insights Oncol 2024; 18:11795549241287058. [PMID: 39450056 PMCID: PMC11500237 DOI: 10.1177/11795549241287058] [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: 03/19/2024] [Accepted: 09/09/2024] [Indexed: 10/26/2024] Open
Abstract
Metabolic reprogramming occurs when tumor cells replenish themselves with nutrients required for growth to meet their metabolic needs. Cancer-associated fibroblasts (CAFs) are activated fibroblasts involved in building the c (TME) to promote tumor progression and metastasis. Metabolic reprogramming of CAFs can interact with cancer cells to generate metabolic crosstalk. Furthermore, CAF metabolic reprogramming has great potential as a new field of tumor treatment. This review summarizes the role of CAFs in TME and the mechanisms by which metabolic reprogramming of CAFs causes cancer progression and metastasis, demonstrating the great potential of CAF metabolic reprogramming in cancer chemotherapy and immunotherapy treatment. Furthermore, we provide an outlook for future CAF metabolic reprogramming for cancer treatment.
Collapse
Affiliation(s)
- Lujia Zhou
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Wenjie Zhang
- Department of Clinical Medicine, School of Medicine, Chongqing University, Chongqing, China
| | - Xiaoxue Hu
- Department of Clinical Medicine, Medical College, Yangzhou University, Yangzhou, China
| | - Daorong Wang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
- Department of General Surgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
- Department of General Surgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Yangzhou, China
- Department of General Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou, China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, China
| | - Dong Tang
- Department of General Surgery, Institute of General Surgery, Northern Jiangsu People’s Hospital Affiliated to Yangzhou University, Yangzhou University, Yangzhou, China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Yangzhou, China
- Department of General Surgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
- Department of General Surgery, The Yangzhou School of Clinical Medicine of Dalian Medical University, Yangzhou, China
- Department of General Surgery, The Yangzhou School of Clinical Medicine of Nanjing Medical University, Yangzhou, China
- Department of General Surgery, Northern Jiangsu People’s Hospital, Clinical Teaching Hospital of Medical School, Nanjing University, Yangzhou, China
| |
Collapse
|
45
|
Voutouri C, Englezos D, Zamboglou C, Strouthos I, Papanastasiou G, Stylianopoulos T. A convolutional attention model for predicting response to chemo-immunotherapy from ultrasound elastography in mouse tumor models. COMMUNICATIONS MEDICINE 2024; 4:203. [PMID: 39420199 PMCID: PMC11487255 DOI: 10.1038/s43856-024-00634-4] [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: 10/05/2023] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
BACKGROUND In the era of personalized cancer treatment, understanding the intrinsic heterogeneity of tumors is crucial. Despite some patients responding favorably to a particular treatment, others may not benefit, leading to the varied efficacy observed in standard therapies. This study focuses on the prediction of tumor response to chemo-immunotherapy, exploring the potential of tumor mechanics and medical imaging as predictive biomarkers. We have extensively studied "desmoplastic" tumors, characterized by a dense and very stiff stroma, which presents a substantial challenge for treatment. The increased stiffness of such tumors can be restored through pharmacological intervention with mechanotherapeutics. METHODS We developed a deep learning methodology based on shear wave elastography (SWE) images, which involved a convolutional neural network (CNN) model enhanced with attention modules. The model was developed and evaluated as a predictive biomarker in the setting of detecting responsive, stable, and non-responsive tumors to chemotherapy, immunotherapy, or the combination, following mechanotherapeutics administration. A dataset of 1365 SWE images was obtained from 630 tumors from our previous experiments and used to train and successfully evaluate our methodology. SWE in combination with deep learning models, has demonstrated promising results in disease diagnosis and tumor classification but their potential for predicting tumor response prior to therapy is not yet fully realized. RESULTS We present strong evidence that integrating SWE-derived biomarkers with automatic tumor segmentation algorithms enables accurate tumor detection and prediction of therapeutic outcomes. CONCLUSIONS This approach can enhance personalized cancer treatment by providing non-invasive, reliable predictions of therapeutic outcomes.
Collapse
Affiliation(s)
- Chrysovalantis Voutouri
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| | - Demetris Englezos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus
| | - Constantinos Zamboglou
- Department of Radiation Oncology, University of Freiburg - Medical Center, Freiburg, Germany
- German Cancer Consortium (DKTK), Partner Site Freiburg, Freiburg, Germany
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Iosif Strouthos
- Department of Radiation Oncology, German Oncology Center, European University Cyprus, Limassol, Cyprus
| | - Giorgos Papanastasiou
- Archimedes Unit, Athena Research Centre, Athens, Greece
- School of Computer Science and Electronic Engineering University of Essex, Wivenhoe Park, UK
| | - Triantafyllos Stylianopoulos
- Cancer Biophysics Laboratory, Department of Mechanical and Manufacturing Engineering, University of Cyprus, Nicosia, Cyprus.
| |
Collapse
|
46
|
Wang J, Zhang Y, Chen H, Wu Y, Liu J, Che H, Zhang Y, Zhu X. Motor-Cargo Structured Nanotractors for Augmented NIR Phototherapy via Gas-Boosted Tumor Penetration and Respiration-Impaired Mitochondrial Dysfunction. Adv Healthc Mater 2024:e2402063. [PMID: 39380347 DOI: 10.1002/adhm.202402063] [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: 06/05/2024] [Revised: 09/26/2024] [Indexed: 10/10/2024]
Abstract
Tumor microenvironment, characterized by dense extracellular matrix and severe hypoxia, has caused pronounced resistance to photodynamic therapy (PDT). Herein, it has designed an artificial nitric oxide (NO) nanotractor with a unique "motor-cargo" structure, where a photoswitching upconversion nanoparticle (UCNP) core serves as the optical engine to harvest NIR light and asymmetrically coated mesoporous silica (SiO2) shell acts as a cargo unit to load nitric oxide (NO) fuel molecule (RBS, Roussin's black salt) and PDT photosensitizer (ZnPc, zinc phthalocyanine). Upon illumination by 980 nm light, the UCNP emits blue light to excite RBS salt and release NO gas. On one hand, NO is used as the driving force to propel the particle with a high speed of ≈194 µm s-1 that generates significant rupture stress (over 0.95 kPa) on cell membrane to promote cellular endocytosis and intratumoral penetration. On the other hand, NO enables to alleviate tumor hypoxia by inhibiting cellular respiration as an oxygen conserver. When the excitation is subsequently switched to 808 nm light, the UCNP emits red light, triggering ZnPc to produce large amount of reactive oxygen species for PDT treatment. This study explores Janus-typed nanostructures for cell-particle interaction and gas-assisted phototherapy, opening avenues for versatile bioapplications.
Collapse
Affiliation(s)
- Jing Wang
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yi Zhang
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, 510632, China
| | - Huadong Chen
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yihan Wu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Jinliang Liu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Hailong Che
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| | - Yong Zhang
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong SAR, 999077, China
| | - Xiaohui Zhu
- School of Environmental and Chemical Engineering, Shanghai University, Shanghai, 200444, China
| |
Collapse
|
47
|
Klabukov I, Smirnova A, Yakimova A, Kabakov AE, Atiakshin D, Petrenko D, Shestakova VA, Sulina Y, Yatsenko E, Stepanenko VN, Ignatyuk M, Evstratova E, Krasheninnikov M, Sosin D, Baranovskii D, Ivanov S, Shegay P, Kaprin AD. Oncomatrix: Molecular Composition and Biomechanical Properties of the Extracellular Matrix in Human Tumors. JOURNAL OF MOLECULAR PATHOLOGY 2024; 5:437-453. [DOI: 10.3390/jmp5040029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025] Open
Abstract
The extracellular matrix is an organized three-dimensional network of protein-based molecules and other macromolecules that provide structural and biochemical support to tissues. Depending on its biochemical and structural properties, the extracellular matrix influences cell adhesion and signal transduction and, in general, can influence cell differentiation and proliferation through specific mechanisms of chemical and mechanical sensing. The development of body tissues during ontogenesis is accompanied by changes not only in cells but also in the composition and properties of the extracellular matrix. Similarly, tumor development in carcinogenesis is accompanied by a continuous change in the properties of the extracellular matrix of tumor cells, called ‘oncomatrix’, as the tumor matures, from the development of the primary focus to the stage of metastasis. In this paper, the characteristics of the composition and properties of the extracellular matrix of tumor tissues are considered, as well as changes to the composition and properties of the matrix during the evolution of the tumor and metastasis. The extracellular matrix patterns of tumor tissues can be used as biomarkers of oncological diseases as well as potential targets for promising anti-tumor therapies.
Collapse
Affiliation(s)
- Ilya Klabukov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Anna Smirnova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Anna Yakimova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Alexander E. Kabakov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Dmitri Atiakshin
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Daria Petrenko
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Victoria A. Shestakova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Obninsk Institute of Nuclear Power Engineering of the National Research Nuclear University MEPhI, 249034 Obninsk, Russia
| | - Yana Sulina
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Elena Yatsenko
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Vasiliy N. Stepanenko
- Department of Obstetrics and Gynecology, Sechenov First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia
| | - Michael Ignatyuk
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Ekaterina Evstratova
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Michael Krasheninnikov
- Scientific and Educational Resource Center for Cellular Technologies, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Dmitry Sosin
- Center for Strategic Planning and Management of Medical and Biological Health Risks of the FMBA of Russia, 119121 Moscow, Russia
| | - Denis Baranovskii
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| | - Sergey Ivanov
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Peter Shegay
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
| | - Andrey D. Kaprin
- National Medical Research Radiological Center of the Ministry of Health of Russian Federation, 249036 Obninsk, Russia
- Scientific and Educational Resource Center for Innovative Technologies of Immunophenotyping, Digital Spatial Profiling and Ultrastructural Analysis, Peoples’ Friendship University of Russia (RUDN University), 117198 Moscow, Russia
| |
Collapse
|
48
|
Stewart DC, Brisson BK, Dekky B, Berger AC, Yen W, Mauldin EA, Loebel C, Gillette D, Assenmacher CA, Quincey C, Stefanovski D, Cristofanilli M, Cukierman E, Burdick JA, Borges VF, Volk SW. Prognostic and therapeutic implications of tumor-restrictive type III collagen in the breast cancer microenvironment. NPJ Breast Cancer 2024; 10:86. [PMID: 39358397 PMCID: PMC11447064 DOI: 10.1038/s41523-024-00690-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Accepted: 09/03/2024] [Indexed: 10/04/2024] Open
Abstract
Collagen plays a critical role in regulating breast cancer progression and therapeutic resistance. An improved understanding of both the features and drivers of tumor-permissive and -restrictive collagen matrices are critical to improve prognostication and develop more effective therapeutic strategies. In this study, using a combination of in vitro, in vivo and bioinformatic experiments, we show that type III collagen (Col3) plays a tumor-restrictive role in human breast cancer. We demonstrate that Col3-deficient, human fibroblasts produce tumor-permissive collagen matrices that drive cell proliferation and suppress apoptosis in non-invasive and invasive breast cancer cell lines. In human triple-negative breast cancer biopsy samples, we demonstrate elevated deposition of Col3 relative to type I collagen (Col1) in non-invasive compared to invasive regions. Similarly, bioinformatics analysis of over 1000 breast cancer patient biopsies from The Cancer Genome Atlas BRCA cohort revealed that patients with higher Col3:Col1 bulk tumor expression had improved overall, disease-free, and progression-free survival relative to those with higher Col1:Col3 expression. Using an established 3D culture model, we show that Col3 increases spheroid formation and induces the formation of lumen-like structures that resemble non-neoplastic mammary acini. Finally, our in vivo study shows co-injection of murine breast cancer cells (4T1) with rhCol3-supplemented hydrogels limits tumor growth and decreases pulmonary metastatic burden compared to controls. Taken together, these data collectively support a tumor-suppressive role for Col3 in human breast cancer and suggest that strategies that increase Col3 may provide a safe and effective therapeutic modality to limit recurrence in breast cancer patients.
Collapse
Affiliation(s)
- Daniel C Stewart
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Becky K Brisson
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Bassil Dekky
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ashton C Berger
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - William Yen
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Elizabeth A Mauldin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Claudia Loebel
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- Department of Materials Science & Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Deborah Gillette
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Charles-Antoine Assenmacher
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Corisa Quincey
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Darko Stefanovski
- Department of Clinical Studies-New Bolton Center, School of Veterinary Medicine, University of Pennsylvania, Kennett Square, PA, USA
| | - Massimo Cristofanilli
- Department of Medicine, Division of Hematology-Oncology, Weill Cornell Medicine, New York, NY, USA
| | - Edna Cukierman
- Cancer Signaling and Microenvironment Program, The Martin and Concetta Greenberg Pancreatic Cancer Institute, Fox Chase Cancer Center, Temple University Lewis Katz School of Medicine, Philadelphia, PA, USA
| | - Jason A Burdick
- Department of Bioengineering, School of Engineering and Applied Science, University of Pennsylvania, Philadelphia, PA, USA
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, USA
| | - Virginia F Borges
- Department of Medicine, Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
- University of Colorado Cancer Center, Aurora, CO, USA
- Young Women's Breast Cancer Translational Program, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Susan W Volk
- Department of Clinical Sciences and Advanced Medicine, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
49
|
Wang Y, Jia J, Wang F, Fang Y, Yang Y, Zhou Q, Yuan W, Gu X, Hu J, Yang S. Pre-metastatic niche: formation, characteristics and therapeutic implication. Signal Transduct Target Ther 2024; 9:236. [PMID: 39317708 PMCID: PMC11422510 DOI: 10.1038/s41392-024-01937-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 06/29/2024] [Accepted: 07/23/2024] [Indexed: 09/26/2024] Open
Abstract
Distant metastasis is a primary cause of mortality and contributes to poor surgical outcomes in cancer patients. Before the development of organ-specific metastasis, the formation of a pre-metastatic niche is pivotal in promoting the spread of cancer cells. This review delves into the intricate landscape of the pre-metastatic niche, focusing on the roles of tumor-derived secreted factors, extracellular vesicles, and circulating tumor cells in shaping the metastatic niche. The discussion encompasses cellular elements such as macrophages, neutrophils, bone marrow-derived suppressive cells, and T/B cells, in addition to molecular factors like secreted substances from tumors and extracellular vesicles, within the framework of pre-metastatic niche formation. Insights into the temporal mechanisms of pre-metastatic niche formation such as epithelial-mesenchymal transition, immunosuppression, extracellular matrix remodeling, metabolic reprogramming, vascular permeability and angiogenesis are provided. Furthermore, the landscape of pre-metastatic niche in different metastatic organs like lymph nodes, lungs, liver, brain, and bones is elucidated. Therapeutic approaches targeting the cellular and molecular components of pre-metastatic niche, as well as interventions targeting signaling pathways such as the TGF-β, VEGF, and MET pathways, are highlighted. This review aims to enhance our understanding of pre-metastatic niche dynamics and provide insights for developing effective therapeutic strategies to combat tumor metastasis.
Collapse
Affiliation(s)
- Yuhang Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Jiachi Jia
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Fuqi Wang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Yingshuai Fang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Yabing Yang
- College of Medicine, Zhengzhou University, Zhengzhou, 450001, China
| | - Quanbo Zhou
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Weitang Yuan
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China
| | - Xiaoming Gu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Junhong Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| | - Shuaixi Yang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, 1 East Jianshe Road, Zhengzhou, 450000, China.
| |
Collapse
|
50
|
Li S, Feng T, Yuan H, Li Q, Zhao G, Li K. DEAD-box RNA helicases in the multistep process of tumor metastasis. Mol Biol Rep 2024; 51:1006. [PMID: 39306810 DOI: 10.1007/s11033-024-09912-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 09/03/2024] [Indexed: 01/04/2025]
Abstract
RNA helicases constitute a large family of proteins that share a catalytic core with high structural similarity. DEAD-box (DDX) proteins belong to the largest RNA helicase subfamily, and DDX members have been implicated in all facets of RNA metabolism, from transcription to translation, miRNA maturation, and RNA delay and degradation. Interestingly, an increasing number of studies have suggested a relationship between DDX proteins and cancer initiation and progression. The expression levels of many DDX proteins are elevated in a majority of cancers, and recent studies have demonstrated that some DDX proteins have a potent positive effect on promoting the metastasis of malignant cells. Metastasis is a complex, multistep cascade process that includes local invasion, intravasation and survival in the circulation, arrest at a distant organ site, extravasation and metastatic colonization; here, we review this process and present the suggested functions and mechanisms of DDX family proteins in particular steps of the invasion‒metastasis cascade.
Collapse
Affiliation(s)
- Shan Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Tianyu Feng
- Department of Laboratory Medicine, Clinical Laboratory Medicine Research Center of West China Hospital, Sichuan Clinical Research Center for Laboratory Medicine, West China Hospital, Sichuan University, Chengdu City, People's Republic of China
| | - Hang Yuan
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Qin Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China
| | - Gang Zhao
- Division of Abdominal Tumor, Department of Medical Oncology, Cancer Center and State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China.
| | - Kai Li
- Cancer Center and Lab of Experimental Oncology, State Key Laboratory of Biotherapy, and Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, No.37 Guoxue Alley, Wuhou District, Chengdu City, Sichuan Province, People's Republic of China.
| |
Collapse
|