1
|
De Cock L, Palubeckaitė I, Bersani F, Faehling T, Pasquali S, Umbaugh S, Meister MT, Danks MR, Manasterski P, Miallot R, Krumbholz M, Roohani S, Heymann D, Cidre-Aranaz F, Wozniak A, Schöffski P, Bovée JVMG, Merlini A, Venneker S. Establishment of patient-derived 3D in vitro models of sarcomas: literature review and guidelines on behalf of the FORTRESS working group. Neoplasia 2025; 65:101171. [PMID: 40324303 DOI: 10.1016/j.neo.2025.101171] [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: 03/21/2025] [Accepted: 04/24/2025] [Indexed: 05/07/2025]
Abstract
Sarcomas are a large family of rare and heterogeneous mesenchymal tumors, which respond poorly to available systemic treatments. Translation of preclinical findings into clinical applications has been slow, limiting improvements in patients' outcomes and ultimately highlighting the need for a better understanding of sarcoma biology to develop more effective, subtype-specific therapies. To this end, reliable preclinical models are crucial, but the development of 3D in vitro sarcoma models has been lagging behind that of epithelial cancers. This is primarily due to the rarity and heterogeneity of sarcomas, and lack of widespread knowledge regarding the optimal growth conditions of these in vitro models. In this review, we provide an overview of currently available sarcoma tumoroid models, together with guidelines and suggestions for model development and characterization, on behalf of the FORTRESS (Forum For Translational Research in Sarcomas) international research working group on 3D sarcoma models.
Collapse
Affiliation(s)
- Lore De Cock
- Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium; Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Ieva Palubeckaitė
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Francesca Bersani
- Department of Oncology, Translational Oncology Laboratory "Paola Gilardi", University of Turin, Turin, Italy
| | - Tobias Faehling
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany; Faculty of Medicine, Heidelberg University, Heidelberg, Germany
| | - Sandro Pasquali
- Molecular Pharmacology, Department of Experimental Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sam Umbaugh
- National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Division of Applied Functional Genomics, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Michael Torsten Meister
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Molly R Danks
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Piotr Manasterski
- Cancer Research UK Edinburgh Centre, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, United Kingdom
| | - Richard Miallot
- Department of Surgical and Interventional Sciences, McGill University, Montreal, QC, Canada; Cancer Research Program, The Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Manuela Krumbholz
- University Hospital Erlangen, Department of Pediatrics Erlangen, Germany
| | - Siyer Roohani
- Charité - Universitätsmedizin Berlin, corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiation Oncology, Berlin, Germany; Berlin Institute of Health at Charité-Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité (Junior) Clinician Scientist Program, Berlin, Germany
| | - Dominique Heymann
- Nantes Université, CNRS, UMR6286, US2B, Institut de Cancérologie de l'Ouest, Saint-Herblain, France; Université of Sheffield, School of Medicine and Population Health, Sheffield, United Kingdom
| | - Florencia Cidre-Aranaz
- Hopp Children's Cancer Center Heidelberg (KiTZ), Heidelberg, Germany; National Center for Tumor Diseases (NCT), NCT Heidelberg, a partnership between DKFZ and Heidelberg University Hospital, Heidelberg, Germany; Division of Translational Pediatric Sarcoma Research, German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Heidelberg, Germany
| | - Agnieszka Wozniak
- Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Patrick Schöffski
- Laboratory of Experimental Oncology, KU Leuven, Leuven Cancer Institute, Leuven, Belgium; Department of General Medical Oncology, University Hospitals Leuven, Leuven Cancer Institute, Leuven, Belgium
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | - Alessandra Merlini
- Department of Oncology, Translational Oncology Laboratory "Paola Gilardi", University of Turin, Turin, Italy; Division of Medical Oncology, San Luigi Gonzaga University Hospital, Orbassano, Turin, Italy
| | - Sanne Venneker
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands.
| |
Collapse
|
2
|
Liu L, Li Y, Li B. Interactions between cancer cells and tumor-associated macrophages in tumor microenvironment. Biochim Biophys Acta Rev Cancer 2025; 1880:189344. [PMID: 40345263 DOI: 10.1016/j.bbcan.2025.189344] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025]
Abstract
Tumor microenvironment (TME) refers to the local environment in which various cancer cells grow, encompassing tumor cells, adjacent non-tumor cells, and associated non-cellular elements, all of which collectively promote cancer occurrence and progression. As a principal immune component in the TME, tumor-associated macrophages (TAMs) exert a considerable influence on cancer behaviors via their interactions with cancer cells. The interactive loops between cancer cells and TAMs, including secretory factors derived from both cancer cells and TAMs, are crucial for the proliferation, stemness, drug resistance, invasion, migration, metastasis, and immune escape of various cancers. Cancer cells release paracrine proteins (HMGB1, AREG etc.), cytokines (IL-6, CCL2 etc.), RNAs (miR-21-5p, circPLEKHM1, LINC01812 etc.), and metabolites (lactic acid, succinate etc.) to regulate the polarization phenotype, mediator secretion and function of TAMs. In turn, mediators (TGF-β, IL-10, IL-6 etc.) from TAMs promote cancer progression. This review summarizes recent advancements in the interactive loops between cancer cells and TAMs in TME. Inhibiting the recruitment and M2 polarization of TAMs, reprogramming TAMs from M2 to M1 phenotype, blocking TAMs-mediated immunosuppression and immune escape, and combining with existing immunotherapy can target TAMs to overcome immunotherapy resistance in various cancers. The new breakthroughs lie in identifying effective targets for drug development, improving the drug delivery system to enhance the drug delivery efficiency, and adopting combined therapy. Interventions targeting secretory factors, cell surface receptors, intracellular signaling pathways, and metabolic modulation in the interactive loops between cancer cells and TAMs are expected to suppress cancer progression and improve therapeutic effects.
Collapse
Affiliation(s)
- Lu Liu
- Department of Oral Anatomy and Physiology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Yafei Li
- Department of Oral Anatomy and Physiology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Bo Li
- Department of Oral Anatomy and Physiology, Jilin Provincial Key Laboratory of Oral Biomedical Engineering, Hospital of Stomatology, Jilin University, Changchun 130021, China.
| |
Collapse
|
3
|
Liu D, Liu L, Zhao X, Zhang X, Chen X, Che X, Wu G. A comprehensive review on targeting diverse immune cells for anticancer therapy: Beyond immune checkpoint inhibitors. Crit Rev Oncol Hematol 2025; 210:104702. [PMID: 40122356 DOI: 10.1016/j.critrevonc.2025.104702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/02/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025] Open
Abstract
Although immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment, primary resistance and acquired resistance continue to limit their efficacy for many patients. To address resistance and enhance the anti-tumor activity within the tumor immune microenvironment (TIME), numerous therapeutic strategies targeting both innate and adaptive immune cells have emerged. These include combination therapies with ICIs, chimeric antigen receptor T-cell (CAR-T), chimeric antigen receptor macrophages (CAR-Ms) or chimeric antigen receptor natural killer cell (CAR-NK) therapy, colony stimulating factor 1 receptor (CSF1R) inhibitors, dendritic cell (DC) vaccines, toll-like receptor (TLR) agonists, cytokine therapies, and chemokine inhibition. These approaches underscore the significant potential of the TIME in cancer treatment. This article provides a comprehensive and up-to-date review of the mechanisms of action of various innate and adaptive immune cells within the TIME, as well as the therapeutic strategies targeting each immune cell type, aiming to deepen the understanding of their therapeutic potential.
Collapse
Affiliation(s)
- Dequan Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Lei Liu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xinming Zhao
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaoman Zhang
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China
| | - Xiaochi Chen
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Xiangyu Che
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| | - Guangzhen Wu
- Department of Urology, the First Affiliated Hospital of Dalian Medical University, Dalian 116011, China.
| |
Collapse
|
4
|
Gorvel L, Panouillot M, Rouvière MS, Billon E, Fattori S, Sonongbua J, Boucherit N, Ben Amara A, Quilichini O, Granjeaud S, Degos C, Nunès JA, Carcopino X, Lambaudie E, Chrétien AS, Sabatier R, Dieu-Nosjean MC, Olive D. Tertiary Lymphoid Structures Are Associated with Enhanced Macrophage Activation and Immune Checkpoint Expression and Predict Outcome in Cervical Cancer. Cancer Immunol Res 2025; 13:712-728. [PMID: 39888676 DOI: 10.1158/2326-6066.cir-24-0979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/23/2024] [Accepted: 01/31/2025] [Indexed: 02/02/2025]
Abstract
Cervical tumors are usually treated using surgery, chemotherapy, and radiotherapy and would benefit from immunotherapies. However, the immune microenvironment in cervical cancer remains poorly described. Tertiary lymphoid structures (TLS) were recently described as markers for better immunotherapy response and overall better prognosis in patients with cancer. We evaluated the cervical tumor immune microenvironment, specifically focusing on TLS, using combined high-throughput phenotyping, soluble factor concentration dosage in the tumor microenvironment, and spatial interaction analyses. We found that TLS presence was associated with a more inflammatory soluble microenvironment, with the presence of B cells as well as more activated macrophages and dendritic cells (DC). Furthermore, this myeloid cell activation was associated with the expression of immune checkpoints, such as PD-L1 and CD40, and the proximity of activated conventional type 2 DCs to CD8+ T cells, indicating better immune interactions and tumor control. Finally, we associated TLS presence, greater B-cell density, and activated DC density with improved progression-free survival, substantiating TLS presence as a potential prognostic marker. Our results provide evidence that TLS presence denotes cell activation and immunotherapy target expression.
Collapse
Affiliation(s)
- Laurent Gorvel
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Marylou Panouillot
- UMRS1135 Sorbonne University, INSERM U1135, Centre of Immunology and Microbial Infections (Cimi), Immune Microenvironment and Immunotherapy Team, Paris, France
| | - Marie-Sarah Rouvière
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Emilien Billon
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Stéphane Fattori
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Jumaporn Sonongbua
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Nicolas Boucherit
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Amira Ben Amara
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Olivia Quilichini
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Samuel Granjeaud
- CRCM Integrative Bioinformatics Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Clara Degos
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Jacques A Nunès
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Xavier Carcopino
- Department of Obstetrics and Gynecology, Hôpital Nord, APHM, Aix-Marseille University (AMU), CNRS, IRD, IMBE UMR 7263, 13397, Marseille, France
| | - Eric Lambaudie
- Department of Surgical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Anne-Sophie Chrétien
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| | - Renaud Sabatier
- Predictive Oncology Laboratory, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Department of Medical Oncology, Institut Paoli-Calmettes, Marseille, France
| | - Marie-Caroline Dieu-Nosjean
- UMRS1135 Sorbonne University, INSERM U1135, Centre of Immunology and Microbial Infections (Cimi), Immune Microenvironment and Immunotherapy Team, Paris, France
| | - Daniel Olive
- Immunity and Cancer Team, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
- Immunomonitoring Platform, Cancer Research Center of Marseille, CRCM, INSERM U1068, CNRS UMR7258, Aix-Marseille University U105, Marseille, France
| |
Collapse
|
5
|
Ma Z, Liu Y, Chen R, Fan H, Kong L, Cao X. A novel perspective on bone tumors: advances in organoid research. Front Pharmacol 2025; 16:1550163. [PMID: 40271075 PMCID: PMC12015983 DOI: 10.3389/fphar.2025.1550163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Bone tumor organoids are three-dimensional cell culture models derived from patient tissues or cells, capable of highly replicating the growth patterns and cell interactions of bone tumors in vitro. Current treatments for bone tumors are hindered by challenges such as drug resistance, recurrence, and metastasis. Organoids enhance the physiological relevance of bone tumor models, thereby improving treatment precision and overcoming the limitations of current therapeutic approaches. Organoid technology has made preliminary applications in bone tumor research, including primary bone tumors, metastatic bone tumors, and bone marrow-derived bone tumors. This review will explore the establishment of bone tumor organoids, summarize their applications and prospects in various bone tumor diseases, and discuss their integration with emerging technologies. Additionally, the limitations and future directions of bone tumor organoid research will be discussed. In the future, bone tumor organoids are expected to promote the further development of precision medicine.
Collapse
Affiliation(s)
- Zebing Ma
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yibing Liu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rui Chen
- Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
| | - Huayu Fan
- Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
| | - Liang Kong
- Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
| | - Xiangyang Cao
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
- Institute of Intelligent Medical and Bioengineering Henan Academy of Traditional Chinese Medicine Sciences, Zhengzhou, Henan, China
- Henan Province Artificial Intelligence Engineering Research Center for Bone Injury Rehabilitation, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
6
|
Hu WT, Li M, Ma PJ, Yang D, Liu XD, Wang Y. A silence catalyst: CCL5-mediated intercellular communication in cancer. Arch Toxicol 2025:10.1007/s00204-025-04036-w. [PMID: 40167774 DOI: 10.1007/s00204-025-04036-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 03/19/2025] [Indexed: 04/02/2025]
Abstract
Chemokine CCL5 (RANTES), as a key mediator of intercellular communication in cancers, and its role in cancer development, metastasis and immune escape has received increasing attention. CCL5 and its receptors are important components of the tumor microenvironment and play a tumor promoting role in different ways by triggering signaling pathways through binding to the primary receptor CCR5. CCL5 was viewed as indispensable "gate keepers" of immunity and inflammation, it remains unclear of CCL5-mediated intercellular communication. Therefore, in this review, we summarize the latest information on the origin, structure, and characterization of CCL5 and role of CCL5 in the tumor microenvironment. It includes CCL5-mediated intercellular communication through exosomes, microvesicles and others in breast, lung, and ovarian cancers. CCL5 has a multifaceted role in cancer and has potential applications as a biomarker for cancer diagnosis and prognosis, which provides theoretical bases and therapeutic targets for the development of new cancer therapeutic strategies.
Collapse
Affiliation(s)
- Wei-Ting Hu
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Ming Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Pei-Jun Ma
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Ding Yang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China
| | - Xiao-Dong Liu
- Department of Pharmacy, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China.
| | - Yun Wang
- Department of Clinical Pharmacology, School of Pharmacy, China Medical University, No.77 Puhe Road, Shenyang North New Area, Shenyang, 110122, Liaoning, China.
| |
Collapse
|
7
|
Godinez C, Campilan B, Schroeder C, Arditi J, Michles MJ, Herrera BC, Gallagher K, Robinson TA, Owens T, Gokaslan ZL, Sullivan P, Martinez-Moreno M. Combinatorial therapies for epigenetic, immunotherapeutic, and genetic targeting of chordoma. J Neurooncol 2025; 172:307-315. [PMID: 39738914 DOI: 10.1007/s11060-024-04920-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 12/16/2024] [Indexed: 01/02/2025]
Abstract
PURPOSE Chordoma, a rare malignancy of the axial skeleton and skull base, presents significant therapeutic challenges due to the high rates of tumor recurrence and resistance. While surgical resection and radiation therapy remain the gold standard of treatment, the lack of additional treatment options necessitates the exploration of novel therapies. Combinatorial therapies hold significant potential in shaping patient prognosis. By targeting the immunotherapeutic, epigenetic, and genetic landscapes of chordoma, these methods enable the more effective and personalized management of the diverse molecular mechanisms driving chordoma growth and resistance. METHODS To elucidate such potential, we conducted a literature review of all published articles on the usage of immunotherapeutic, epigenetic, and genetic approaches for chordoma treatment from 2014 to 2024. RESULTS Eighty-one papers were excluded based on our inclusion criteria. From the remaining thirty-nine publications, we found evidence supporting the efficacy of immune checkpoint inhibitors (ICIs), chimeric antigen receptor (CAR) T-cell therapies, and monoclonal antibodies; the roles of DNA methylation patterns, histone modification pathways, and miRNA regulation; and the contribution of cancer stem-like cells (CSCs) to chordoma progression. CONCLUSION Our findings underscore the importance of a multidirectional approach in chordoma treatment throughout the disease progression to reduce morbidity and improve patient outcomes despite the heterogeneity of chordoma.
Collapse
Affiliation(s)
- Christian Godinez
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Beatrice Campilan
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Christian Schroeder
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Jonathan Arditi
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Madison J Michles
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Benjamín Córdova Herrera
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Kaylee Gallagher
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Tati-Anna Robinson
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Tyler Owens
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Ziya L Gokaslan
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Patricia Sullivan
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA
| | - Margot Martinez-Moreno
- Department of Neurosurgery, Warren Alpert Medical School of Brown University, 222 Richmond St, Providence, RI, 02903, USA.
| |
Collapse
|
8
|
Lou M. Systemic trafficking of macrophages in implant wear debris-induced periprosthetic osteolysis. SLAS Technol 2025; 31:100254. [PMID: 39914493 DOI: 10.1016/j.slast.2025.100254] [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: 11/25/2024] [Revised: 01/15/2025] [Accepted: 02/03/2025] [Indexed: 02/24/2025]
Abstract
Periprosthetic osteolysis (PPOL) is a significant complication post-joint replacement, often instigated by implant wear debris, leading to chronic inflammation and bone resorption. Herein, this review summarizes the immune mechanisms of PPOL, specifically, the processes where macrophages are recruited by implant wear debris, the mechanisms by which macrophages trigger inflammatory cascades, and the role of chemokines that facilitate macrophage migration, including CCL2, CCL3, CCL4, CCL5, CXCL8, CX3CL1, and XCL1. This review highlights novel findings on these processes and suggests that illustrating these mechanisms offers promising avenues for future therapeutic strategies to prevent and treat PPOL, such as the potential use of anti-inflammatory drugs.
Collapse
Affiliation(s)
- Mengyun Lou
- Department of General Practice, Shanghai Sixth People's Hospital, Shanghai 200233, China.
| |
Collapse
|
9
|
Shi B, Du M, Chen Z. Advances in tumor immunotherapy targeting macrophages. Expert Rev Clin Immunol 2025; 21:259-276. [PMID: 39636579 DOI: 10.1080/1744666x.2024.2438721] [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: 04/08/2024] [Revised: 11/03/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
INTRODUCTION In recent years, immunotherapy has shown significant therapeutic potential in patients with advanced tumors. However, only a small number of individuals benefit, mainly due to the tumor microenvironment (TME), which provides conditions for the development of tumors. Macrophages in TME, known as tumor-associated macrophages (TAM), are mainly divided into M1 anti-tumor and M2 pro-tumor phenotypes, which play a regulatory role in various stages of tumorigenesis, promote tumorigenesis and metastasis, and cause immunotherapy resistance. AREAS COVERED This review focuses on research strategies and preclinical/clinical research progress in translating TAM into antitumor phenotype by referring to the PubMed database for five years. These include small molecule chemotherapy drug development, metabolic regulation, gene editing, physical stimulation, nanotechnology-mediated combination therapy strategies, and chimeric antigen receptor-based immunotherapy. EXPERT OPINION It is necessary to explore the surface-specific receptors and cell signaling pathways of TAM further to improve the specificity and targeting of drugs and to strengthen research in the field of probes that can monitor changes in TAM in real time. In addition, the physical stimulation polarization strategy has the advantages of being noninvasive, economical, and stable and will have excellent clinical transformation value in the future.
Collapse
Affiliation(s)
- Binrui Shi
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- Department of Medical imaging, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Meng Du
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
- The Seventh Affiliated Hospital, Hunan Veterans Administration Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
| | - Zhiyi Chen
- Key Laboratory of Medical Imaging Precision Theranostics and Radiation Protection, College of Hunan Province, the Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, Hunan, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| |
Collapse
|
10
|
Gui Y, Wu H, Fan H. Pseudostellaria heterophylla (Miq.) Pax, a traditional folk medicine, ameliorates colorectal cancer by remodeling the tumor immune microenvironment. JOURNAL OF ETHNOPHARMACOLOGY 2025; 342:119362. [PMID: 39828144 DOI: 10.1016/j.jep.2025.119362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 01/08/2025] [Accepted: 01/11/2025] [Indexed: 01/22/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Pseudostellaria heterophylla (Miq.) Pax (PH) is a traditional folk medicine, which is widely used clinically for digestive system tumors such as esophageal, gastric, colorectal, and liver cancers. The anti-tumor effect and mechanism of PH in colorectal cancer (CRC) deserves further study. AIM OF THE STUDY The objective of this study is to examine the effects and the underlying mechanisms of aqueous extract of Pseudostellaria heterophylla (Miq.) Pax (AEPH) in the CRC. MATERIALS AND METHODS The components of AEPH were fully resolved using ultra-high-performance liquid chromatography coupled with quadrupole-orbitrap high-resolution mass spectrometry (UHPLC-Q/Orbitrap HRMS). The effect of AEPH was evaluated in vivo using the MC38 mouse colon cancer model, and its impact on the tumor microenvironment was analyzed by flow cytometry. Bioinformatics analysis, combined with transcriptome sequencing, was utilized to further investigate the signaling pathways of AEPH in CRC cells. RESULTS A mass spectrometry analysis identified 371 compounds in AEPH, each with a comprehensive score exceeding 60. In vivo experiments demonstrated that AEPH suppressed the growth of MC38 tumors without exhibiting obvious toxicity. Mechanistic studies revealed that AEPH inhibited the JNK signaling pathway, reduced Chemokine C-C Motif Chemokine Ligand 5 (CCL5) secreted by CRC cells, hindered the recruitment of M2-like tumor-associated macrophages (TAMs), promoted the infiltration of IFN-γ+ CD8+ T cells, and improved the immunosuppressive microenvironment of CRC. CONCLUSION AEPH contributes to the remodeling of the tumor immune microenvironment primarily through the inhibition of CCL5-mediated recruitment of M2-like TAMs. The findings of this study offer a novel perspective on the potential development of AEPH as a therapeutic agent for CRC.
Collapse
Affiliation(s)
- Yang Gui
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Hui Wu
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Heng Fan
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
| |
Collapse
|
11
|
Turkbey M, Karaguzel D, Uzunkaya AD, Aracagok YD, Karaaslan C. The immune response of upper and lower airway epithelial cells to Aspergillus fumigatus and Candida albicans-derived β-glucan in Th17 type cytokine environment. Arch Microbiol 2025; 207:70. [PMID: 39992431 DOI: 10.1007/s00203-025-04266-7] [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: 10/28/2024] [Revised: 01/24/2025] [Accepted: 02/03/2025] [Indexed: 02/25/2025]
Abstract
The fungal cell wall component β-glucan activates inflammation via the Dectin-1 receptor and IL-17 coordinates the antifungal immunity. However, the molecular crosstalk between IL-17, Dectin-1, and β-glucan in epithelial cells and fungal immunity remains unclear. We investigated the impact of A.fumigatus-derived β-glucan (AFBG) and C.albicans-derived β-glucan (CABG) on Dectin-1 and cytokines in nasal epithelial cells (NECs) and bronchial epithelial cells (BECs) in the presence of IL-17. CABG reduced BEC viability more than AFBG despite similar Dectin-1 expression. IL-17 reduced β-glucan-dependent Dectin-1 expression in NECs but increased it in BECs after 12 h. AFBG synergized with IL-17, enhancing pro-inflammatory cytokines and chemokine expressions. IL-6 and IL-8 production increased in the presence of IL-17. Th17 cytokine influenced the Dectin-1 response to fungal β-glucan in NECs and BECs, impacting the initiation and nature of epithelial cell reactions to AFBG and CABG. Uncovering the molecular mechanisms of fungal β-glucans in the respiratory tract could lead to novel strategies for preventing fungal diseases.
Collapse
Affiliation(s)
- Murat Turkbey
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye
| | - Dilara Karaguzel
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye
| | - Ali Doruk Uzunkaya
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye
| | - Yusuf Doruk Aracagok
- Department of Biology, Biotechnology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye
| | - Cagatay Karaaslan
- Department of Biology, Molecular Biology Section, Faculty of Science, Hacettepe University, 06800, Ankara, Türkiye.
| |
Collapse
|
12
|
Wu Q, Lv J, Li X. Membrane palmitoylated protein MPP1 inhibits immune escape by regulating the USP12/ CCL5 axis in urothelial carcinoma. Int Immunopharmacol 2025; 146:113802. [PMID: 39700963 DOI: 10.1016/j.intimp.2024.113802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/27/2024] [Accepted: 12/04/2024] [Indexed: 12/21/2024]
Abstract
BACKGROUND The response rate to immunotherapy in patients with urothelial carcinoma remains limited. Studies have shown that membrane palmitoylated proteins (MPPs) play key roles in tumor progression. However, the mechanisms by which MPP1 regulates immune escape in urothelial carcinoma are not well understood. METHODS The TCGA and BEST databases were used to analyze the associations between the expression of members of the MPP family and the prognosis or immunotherapy sensitivity of urothelial carcinoma patients. MPP1 was identified due to its significant association with survival and immunotherapy sensitivity. MPP1 expression in urothelial carcinoma tissues and cell lines was examined. An MPP1 overexpression vector was used to transfect urothelial carcinoma cells. The functional assays included proliferation, migration, urothelial carcinoma cell-CD8+ T-cell coculture, CD8+ T-cell chemotaxis, and tumorigenesis in human immune reconstitution NOG mice (HuNOG). Bioinformatics, coimmunoprecipitation (CO-IP), mass spectrometry, quantitative real-time polymerase chain reaction (RT-qPCR), and western blotting were used to validate the activity of the MPP1/USP12/CCL5 cascade. RESULTS Analysis of the BEST data revealed that, compared with other MPP family genes, MPP1 was more strongly associated with urothelial carcinoma prognosis and immunotherapy response. Low MPP1 expression was observed in urothelial carcinoma patients and was positively associated with better survival. MPP1 inhibited the proliferation and migration of urothelial carcinoma cells. Bioinformatics, in vitro coculture assays, and in vivo tumorigenesis experiments demonstrated that MPP1 promotes CCL5 production and CD8+ T-cell chemotaxis in the urothelial carcinoma tumor microenvironment (TME). Mechanistically, bioinformatics, mass spectrometry, co-IP, RT-qPCR, and western blot analyses indicated that MPP1 increases CCL5 expression by binding to and promoting USP12. CONCLUSIONS MPP1 significantly inhibits urothelial carcinoma cell proliferation and immune escape via the MPP1/USP12/CCL5 cascade. MPP1 has the potential to serve as a biomarker for guiding immunotherapy in patients with urothelial carcinoma.
Collapse
Affiliation(s)
- Qikai Wu
- Laboratory of Urology and Andrology, Jiangsu Clinical Medicine Research Institution, Nanjing 210029, China
| | - Jiancheng Lv
- Department of Urology, First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang 310058, China.
| | - Xiaojun Li
- Department of Urology, Taicang Affiliated Hospital of Soochow University, the First People's Hospital of Taicang, Taicang 215400, China.
| |
Collapse
|
13
|
Xie P, Guo L, Yu Q, Zhao Y, Yu M, Wang H, Wu M, Xu W, Xu M, Zhu XD, Xu Y, Xiao YS, Huang C, Zhou J, Fan J, Hung MC, Sun H, Ye QH, Zhang B, Li H. ACE2 Enhances Sensitivity to PD-L1 Blockade by Inhibiting Macrophage-Induced Immunosuppression and Angiogenesis. Cancer Res 2025; 85:299-313. [PMID: 39495239 DOI: 10.1158/0008-5472.can-24-0954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Revised: 08/07/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Anti-PD-L1-based combination immunotherapy has become the first-line treatment for unresectable hepatocellular carcinoma (HCC). However, the objective response rate is lower than 40%, highlighting the need to identify mechanisms of tolerance to immune checkpoint inhibitors and accurate biomarkers of response. In this study, we used next-generation sequencing to analyze HCC samples from 10 patients receiving anti-PD-L1 therapy. Activation of the renin-angiotensin system was elevated in nonresponders compared with responders, and angiotensin-converting enzyme 2 (ACE2) expression was significantly downregulated in nonresponders. ACE2 deficiency promoted HCC development and anti-PD-L1 resistance, whereas ACE2 overexpression inhibited HCC progression in immune-competent mice. Mass cytometry by time of flight revealed that ACE2-deficient murine orthotopic tumor tissues featured elevated M2-like tumor-associated macrophages, displayed a CCR5+PD-L1+ immunosuppressive phenotype, and exhibited high VEGFα expression. ACE2 downregulated tumor-intrinsic chemokine (C-C motif) ligand 5 expression by suppressing NF-κB signaling through the ACE2/angiotensin-(1-7)/Mas receptor axis. The lower chemokine (C-C motif) ligand 5 levels led to reduced activation of the JAK-STAT3 pathway and suppressed PD-L1 and VEGFα expression in macrophages, blocking macrophage infiltration and M2-like polarization. Pharmacologic targeting of CCR5 using maraviroc enhanced the tumor-suppressive effect of anti-PD-L1 therapy. Together, these findings suggest that activation of the ACE2 axis overcomes the immunosuppressive microenvironment of HCC and may serve as an immunotherapeutic target and predictive biomarker of response to PD-L1 blockade. Significance: ACE2 regulates the immune landscape of hepatocellular carcinoma by abrogating M2-like macrophage polarization and sensitizes tumors to anti-PD-L1, suggesting that harnessing the ACE2 axis could be a promising strategy to improve immunotherapy efficacy.
Collapse
Affiliation(s)
- Peiyi Xie
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Lei Guo
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Qiang Yu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Yufei Zhao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Mincheng Yu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Hui Wang
- Department of Molecular Biology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, P.R. China
| | - Mengyuan Wu
- Department of Thoracic Surgery, Zhongshan Hospital, Fudan University, Shanghai, P.R. China
| | - Wenxin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Min Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Yongfeng Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Yong-Sheng Xiao
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Cheng Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology, and Center for Molecular Medicine, China Medical University, Taichung, Taiwan
| | - Huichuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Qing-Hai Ye
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Bo Zhang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
| | - Hui Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, P.R. China
- Shanghai Medical College and Zhongshan Hospital Immunotherapy Technology Translational Research Center, Shanghai, P.R. China
| |
Collapse
|
14
|
Chen Y, Guo Y, Li S, Xu J, Zhao C, Wang J, Yang J, Ning W, Qu Y, Zhang M, Wang S, Zhang H. Tumor-derived IL-6 promotes chordoma invasion by stimulating tumor-associated macrophages M2 polarization and TNFα secretion. Int Immunopharmacol 2024; 143:113315. [PMID: 39393273 DOI: 10.1016/j.intimp.2024.113315] [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: 01/29/2024] [Revised: 09/04/2024] [Accepted: 10/01/2024] [Indexed: 10/13/2024]
Abstract
AIMS Chordoma is a rare and aggressive bone tumor with high-recurrence and lack of effective treatment methods. Tumor associated macrophages (TAMs) are abundant in tumor microenvironment (TME) and polarize toward M2 in chordoma. It has been observed that the high proportion of M2 cells is associated with chordoma rapid progression. However, the mechanism of TAMs polarization and promotion to tumor progression in chordoma is still unclear. The is an urgent need for further research. MATERIALS AND METHODS Flow cytometry and immunohistochemical staining was used to detect the degree of macrophages infiltration in chordoma. A co-culture model of chordoma cells and macrophages was established in vitro to investigate the effects of their interaction on cell function, cytokine secretion, and RNA transcriptome expression. KEY FINDINGS In this study, we found M2 macrophage was predominantly abundant immune cell population in chordoma, and its proportion was associated with the degree of bone destruction. We demonstrated that interleukin 6 (IL-6) derived from chordoma cells could induce TAMs polarization by activating STAT3 phosphorylation, and TAMs could enhance chordoma cells migration and invasion through TNFα/NF-κB pathway. The interaction of chordoma cells and TAMs could promote the bone destruction-related factor Cathepsin B (CTSB) and inhibitory immune checkpoints expression. We also confirmed blocking IL-6/STAT3 pathway could significantly attenuate the M2 polarization of TAMs and decrease the secretion of TNFα. SIGNIFICANCE This study illustrates the dynamics between chordoma cells and TAMs in promoting chordoma invasion and suggests that IL-6/STAT3 pathway is a potential therapeutic target to reduce TAM-induced chordoma invasion.
Collapse
Affiliation(s)
- Yujia Chen
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Yuduo Guo
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Shenglun Li
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Jiacheng Xu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Chao Zhao
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Jun Wang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Jingjing Yang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Weihai Ning
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Yanming Qu
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Mingshan Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China
| | - Shengdian Wang
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, Chinese Academy of Sciences, 100101 Beijing, China.
| | - Hongwei Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing 100093, China.
| |
Collapse
|
15
|
Tang W, Li R, Lai X, Yu X, He R. Prognostic factors and overall survival in pelvic Ewing's sarcoma and chordoma: A comparative SEER database analysis. Heliyon 2024; 10:e37013. [PMID: 39286090 PMCID: PMC11402751 DOI: 10.1016/j.heliyon.2024.e37013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/24/2024] [Accepted: 08/26/2024] [Indexed: 09/19/2024] Open
Abstract
Background This study aimed to develop and validate nomograms to predict overall survival (OS) for pelvic Ewing's sarcoma (EWS) and chordoma, identify prognostic factors, and compare outcomes between the two conditions. Methods We identified patients diagnosed with pelvic EWS or chordoma from the SEER database (2001-2019). Independent risk factors were identified using univariate and multivariate Cox regression analyses, and these factors were used to construct nomograms predicting 3-, 5-, and 10-year OS. Validation methods included AUC, calibration plots, C-index, and decision curve analysis (DCA). Kaplan-Meier curves and log-rank tests compared survival differences between low- and high-risk groups. Results The study included 1175 patients (EWS: 611, chordoma: 564). Both groups were randomly divided into training (70 %) and validation (30 %) cohorts. OS was significantly higher for chordoma. Multivariate analysis showed year of diagnosis, income, stage, and surgery were significant for EWS survival, while age, time to treatment, stage, and surgery were significant for chordoma survival. Validation showed the nomograms had strong predictive performance and clinical utility. Conclusions The nomograms reliably predict overall survival (OS) in pelvic EWS and chordoma, helping to identify high-risk patients early and guide preventive measures. The study also found that survival rates are significantly higher for chordoma, highlighting different prognostic profiles between EWS and chordoma.
Collapse
Affiliation(s)
- Wanyun Tang
- Department of Orthopedics, Zigong First People's Hospital, Zigong, China
| | - Runzhuo Li
- Department of Digestion,The First People's Hospital of Yibin, Yibin, China
| | - Xiaoying Lai
- Department of Orthopedics, Zigong First People's Hospital, Zigong, China
| | - Xiaohan Yu
- Department of General Surgery, Dandong Central Hospital, China Medical University, Dandong, China
| | - Renjian He
- Department of Orthopedics, Zigong First People's Hospital, Zigong, China
| |
Collapse
|
16
|
Chen Y, Zhang H. Immune microenvironment and immunotherapy for chordoma. Front Oncol 2024; 14:1374249. [PMID: 38983929 PMCID: PMC11232415 DOI: 10.3389/fonc.2024.1374249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/11/2024] [Indexed: 07/11/2024] Open
Abstract
Chordoma, as a rare, low-grade malignant tumor that tends to occur in the midline of the body, grows slowly but often severely invades surrounding tissues and bones. Due to the severe invasion and damage to the surrounding tissues, chordoma is difficult to be gross totally resected in surgery, and the progression of the residual tumor is often unavoidable. Besides, the tumor is insensitive to conventional radiotherapy and chemotherapy, thus finding effective treatment methods for chordoma is urgent. Nowadays, immunotherapy has made a series of breakthroughs and shown good therapeutic effects in kinds of tumors, which brings new insights into tumors without effective treatment strategies. With the deepening of research on immunotherapy, some studies focused on the immune microenvironment of chordoma have been published, most of them concentrated on the infiltration of immune cells, the expression of tumor-specific antigen or the immune checkpoint expression. On this basis, a series of immunotherapy studies of chordoma are under way, some of which have shown encouraging results. In this review, we reviewed the research about immune microenvironment and immunotherapy for chordoma, combined with the existing clinical trials data, hoping to clarify the frontiers and limitations of chordoma immune research, and provide reference for follow-up immunotherapy research on chordoma.
Collapse
Affiliation(s)
| | - Hongwei Zhang
- Department of Neurosurgery, Sanbo Brain Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
17
|
Wu H, Li X, Zhang B, Liu P, Qi M, Du Y, Zhang C, Duan W, Chen Z. Single-cell sequencing reveals VEGFR as a potential target for CAR-T cell therapy in chordoma. Br J Cancer 2024; 130:1609-1620. [PMID: 38605247 PMCID: PMC11091087 DOI: 10.1038/s41416-024-02635-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Revised: 02/16/2024] [Accepted: 02/21/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Chordomas are rare osseous neoplasms with a dismal prognosis when they recur. Here we identified cell surface proteins that could potentially serve as novel immunotherapeutic targets in patients with chordoma. METHODS Fourteen chordoma samples from patients attending Xuanwu Hospital Capital Medical University were subjected to single-cell RNA sequencing. Target molecules were identified on chordoma cells and cancer metastasis-related signalling pathways characterised. VEGFR-targeting CAR-T cells and VEGFR CAR-T cells with an additional TGF-β scFv were synthesised and their in vitro antitumor activities were evaluated, including in a primary chordoma organoid model. RESULTS Single-cell transcriptome sequencing identified the chordoma-specific antigen VEGFR and TGF-β as therapeutic targets. VRGFR CAR-T cells and VEGFR/TGF-β scFv CAR-T cells recognised antigen-positive cells and exhibited significant antitumor effects through CAR-T cell activation and cytokine secretion. Furthermore, VEGFR/TGF-β scFv CAR-T cells showed enhanced and sustained cytotoxicity of chordoma cell lines in vitro compared with VRGFR CAR-T cells. CONCLUSIONS This study provides a comprehensive single-cell landscape of human chordoma and highlights its heterogeneity and the role played by TGF-β in chordoma progression. Our findings substantiate the potential of VEGFR as a target for CAR-T cell therapies in chordoma which, together with modulated TGF-β signalling, may augment the efficacy of CAR-T cells.
Collapse
Affiliation(s)
- Huantong Wu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Lab of Spinal Cord, Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China
| | - Xinqiang Li
- Organ Transplantation Center, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Boyan Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Lab of Spinal Cord, Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China
| | - Penghao Liu
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Lab of Spinal Cord, Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China
| | - Maoyang Qi
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Lab of Spinal Cord, Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China
| | - Yueqi Du
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Lab of Spinal Cord, Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China
| | - Can Zhang
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China
- Lab of Spinal Cord, Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China
| | - Wanru Duan
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Lab of Spinal Cord, Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China.
| | - Zan Chen
- Department of Neurosurgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
- Lab of Spinal Cord, Injury and Functional Reconstruction, China International Neuroscience Institute (CHINA-INI), Beijing, China.
| |
Collapse
|
18
|
Yu W, Yao Y, Ye N, Zhao Y, Ye Z, Wei W, Zhang L, Chen J. The myokine CCL5 recruits subcutaneous preadipocytes and promotes intramuscular fat deposition in obese mice. Am J Physiol Cell Physiol 2024; 326:C1320-C1333. [PMID: 38497114 DOI: 10.1152/ajpcell.00591.2023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/19/2024]
Abstract
Intramuscular fat (IMF) refers to the lipid stored in skeletal muscle tissue. The number and size of intramuscular adipocytes are the primary factors that regulate IMF content. Intramuscular adipocytes can be derived from either in situ or ectopic migration. In this study, it was discovered that the regulation of IMF levels is achieved through the chemokine (C-C motif) ligand 5 (CCL5)/chemokine (C-C motif) receptor 5 (CCR5) pathway by modulating adipocyte migration. In coculture experiments, C2C12 myotubes were more effective in promoting the migration of 3T3-L1 preadipocytes than C2C12 myoblasts, along with increasing CCL5. Correspondingly, overexpressing the CCR5, one of the receptors of CCL5, in 3T3-L1 preadipocytes facilitated their migration. Conversely, the application of the CCL5/CCR5 inhibitor, MARAVIROC (MVC), reduced this migration. In vivo, transplanted experiments of subcutaneous adipose tissue (SCAT) from transgenic mice expressing green fluorescent protein (GFP) provided evidence that injecting recombinant CCL5 (rCCL5) into skeletal muscle promotes the migration of subcutaneous adipocytes to the skeletal muscle. The level of CCL5 in skeletal muscle increased with obesity. Blocking the CCL5/CCR5 axis by MVC inhibited IMF deposition, whereas elevated skeletal muscle CCL5 promoted IMF deposition in obese mice. These results establish a link between the IMF and the CCL5/CCR5 pathway, which could have a potential application for modulating IMF through adipocyte migration.NEW & NOTEWORTHY C2C12 myotubes attract 3T3-L1 preadipocyte migration regulated by the chemokine (C-C motif) ligand 5 (CCL5)/ chemokine (C-C motif) receptor 5 (CCR5) axis. High levels of skeletal muscle-specific CCL5 promote the migration of subcutaneous adipocytes to skeletal muscle and induce the intramuscular fat (IMF) content.
Collapse
Affiliation(s)
- Wensai Yu
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Yao Yao
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Nanwei Ye
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Yuelei Zhao
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Zijian Ye
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Wei Wei
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Lifan Zhang
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| | - Jie Chen
- College of Animal Science and TechnologyNanjing Agricultural University, NanjingPeople's Republic of China
| |
Collapse
|
19
|
Ma Y, Yu X, Ye S, Li W, Yang Q, Li YX, Wang Y, Wang YL. Immune-regulatory properties of endovascular extravillous trophoblast cells in human placenta. Placenta 2024; 145:107-116. [DOI: pmid:38128221 doi: 10.1016/j.placenta.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
|
20
|
Ma Y, Yu X, Ye S, Li W, Yang Q, Li YX, Wang Y, Wang YL. Immune-regulatory properties of endovascular extravillous trophoblast cells in human placenta. Placenta 2024; 145:107-116. [PMID: 38128221 DOI: 10.1016/j.placenta.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 11/04/2023] [Accepted: 12/07/2023] [Indexed: 12/23/2023]
Abstract
INTRODUCTION Uterine spiral artery remodeling is the prerequisite for ensuring adequate blood supply to the maternal-fetal interface during human pregnancy. One crucial cellular event in this process involves the extensive replacement of the spiral artery endothelial cells by endovascular extravillous trophoblasts (enEVTs), a subtype of extravillous trophoblasts (EVTs). However, our understanding of the properties of enEVTs remains limited. METHODS Human enEVTs in decidual tissues during early pregnancy was purified using flow sorting by specific makers, NCAM1 and HLA-G. The high-throughput RNA sequencing analysis as well as the cytokine antibody array experiments were carried out to analyze for cell properties. Gene ontology (GO) enrichment, kyoto encyclopedia of genes and genomes (KEGG) enrichment, and gene set enrichment analysis (GSEA) were performed on differentially expressed genes of enEVTs. Immunofluorescent assays were used to verify the analysis results. RESULTS Both enEVTs and interstitial EVTs (iEVTs) exhibited gene expression patterns typifying EVT characteristics. Intriguingly, enEVTs displayed gene expression associated with immune responses, particularly reminiscent of M2 macrophage characteristics. The active secretion of multiple cytokines and chemokines by enEVTs provided partial validation for their expression pattern of immune-regulatory genes. DISCUSSION Our study reveals the immune-regulatory properties of human enEVTs and provides new insights into their functions and mechanisms involved in spiral artery remodeling.
Collapse
Affiliation(s)
- Yeling Ma
- School of Medicine, Shaoxing University, Shaoxing, Zhejiang, 312000, China; State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xin Yu
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China
| | - Shenglong Ye
- Department of Gynecology and Obstetrics, Peking University Third Hospital, Beijing, China
| | - Wenlong Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, China
| | - Qian Yang
- NHC Key Lab of Reproduction Regulation, Shanghai Engineering Research Center of Reproductive Health Drug and Devices, Shanghai Institute for Biomedical and Pharmaceutical Technologies, Shanghai, China
| | - Yu-Xia Li
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yongqing Wang
- Department of Gynecology and Obstetrics, Peking University Third Hospital, Beijing, China.
| | - Yan-Ling Wang
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China; Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
21
|
Shen J, Chen C, Chen Z, Gong P, Lee LS, Schmeusser BN, Zhuang Q, Sun Y, Xue D, He X. CCL5 promotes the proliferation and metastasis of bladder cancer via the JAK2/STAT3 signaling pathway. Transl Androl Urol 2023; 12:1845-1858. [PMID: 38196701 PMCID: PMC10772649 DOI: 10.21037/tau-23-540] [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: 10/24/2023] [Accepted: 12/14/2023] [Indexed: 01/11/2024] Open
Abstract
Background Non-muscle invasive bladder cancer (NMIBC) is one of the most common malignant tumors of the urinary system. There is an urgent need for further studies to elucidate the underlying mechanisms of bladder cancer (BC) progression. It has been observed that C-C chemokine ligand 5 (CCL5) and its receptor C-C chemokine receptor type 5 (CCR5) are expressed abnormally and activated in solid tumors and hematological malignancies, which is gaining increasing attention. However, the underlying mechanism of CCL5 in BC remains unclear. Methods The expression levels of CCL5 were analyzed by real-time polymerase chain reaction (RT-PCR) and western blot. Proliferation analysis of cells was carried out using Cell Counting Kit-8 (CCK-8). The assessment of the migration was conducted using a wound-healing assay. A Matrigel-coated transwell chamber was used to test cell invasiveness. A subcutaneous transplantation tumor model and tail vein injection pulmonary metastasis tumor model were used to evaluate the proliferation and metastasis of BC cell in vivo. Results This study showed that CCL5 promotes proliferative, migratory, and tumor-growing BC cells in vitro and tumor metastasizing BC cells in vivo. Moreover, we found that the tumor-promotive role of CCL5 is dependent on activation of the JAK2/STAT3 signaling pathway. Conclusions CCL5 may play an oncogenic role in BC and may also serve as a potential diagnostic and prognostic biomarker.
Collapse
Affiliation(s)
- Jie Shen
- Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Cheng Chen
- Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Zhen Chen
- Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Pengfeng Gong
- Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lui Shiong Lee
- Department of Urology, Sengkang General Hospital, Singapore, Singapore
| | | | - Qianfeng Zhuang
- Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yangyang Sun
- Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dong Xue
- Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiaozhou He
- Department of Urology, the Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
22
|
de Nigris F, Meo C, Palinski W. Combination of Genomic Landsscape and 3D Culture Functional Assays Bridges Sarcoma Phenotype to Target and Immunotherapy. Cells 2023; 12:2204. [PMID: 37681936 PMCID: PMC10486752 DOI: 10.3390/cells12172204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 08/24/2023] [Accepted: 08/30/2023] [Indexed: 09/09/2023] Open
Abstract
Genomic-based precision medicine has not only improved tumour therapy but has also shown its weaknesses. Genomic profiling and mutation analysis have identified alterations that play a major role in sarcoma pathogenesis and evolution. However, they have not been sufficient in predicting tumour vulnerability and advancing treatment. The relative rarity of sarcomas and the genetic heterogeneity between subtypes also stand in the way of gaining statistically significant results from clinical trials. Personalized three-dimensional tumour models that reflect the specific histologic subtype are emerging as functional assays to test anticancer drugs, complementing genomic screening. Here, we provide an overview of current target therapy for sarcomas and discuss functional assays based on 3D models that, by recapitulating the molecular pathways and tumour microenvironment, may predict patient response to treatments. This approach opens new avenues to improve precision medicine when genomic and pathway alterations are not sufficient to guide the choice of the most promising treatment. Furthermore, we discuss the aspects of the 3D culture assays that need to be improved, such as the standardisation of growth conditions and the definition of in vitro responses that can be used as a cut-off for clinical implementation.
Collapse
Affiliation(s)
- Filomena de Nigris
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Concetta Meo
- Department of Precision Medicine, School of Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy;
| | - Wulf Palinski
- Department of Medicine, University of California San Diego, La Jolla, CA 92037, USA;
| |
Collapse
|