1
|
Ge W, Mu Z, Yang S, Zeng Y, Deng Y, Lin Y, Xie P, Li G. Biosensor-based methods for exosome detection with applications to disease diagnosis. Biosens Bioelectron 2025; 279:117362. [PMID: 40157151 DOI: 10.1016/j.bios.2025.117362] [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: 12/18/2024] [Revised: 02/09/2025] [Accepted: 03/09/2025] [Indexed: 04/01/2025]
Abstract
Exosomes are nanoscale extracellular vesicles (EVs) secreted by most eukaryotic cells and can be found in nearly all human body fluids. Increasing evidence has revealed their pivotal roles in intercellular communication, and their active participation in myriad physiological and pathological activities. Exosomes' functions rely on their contents that are closely correlated with the biological characteristics of parental cells, which may provide a rich resource of molecular information for accurate and detailed diagnosis of a diverse array of diseases, such as differential diagnosis of Alzheimer's disease, early detection and subtyping of various tumors. As a category of sensitive detection devices, biosensors can fully reveal the molecular information and convert them into actionable clinical information. In this review, recent advances in biosensor-based methods for the detection of exosomes are summarized. We have described the fabrication of various biosensors based on the analysis of exosomal proteins, RNAs or glycans for accurate diagnosis, with respect to their elaborate recognition designs, signal amplification strategies, sensing properties, as well as their application potential. The challenges along with corresponding technologies in the future development and clinical translation of these biosensors are also discussed.
Collapse
Affiliation(s)
- Weikang Ge
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Zheying Mu
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Shiao Yang
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yujing Zeng
- Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China
| | - Ying Deng
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China
| | - Yifan Lin
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China
| | - Ping Xie
- Department of Ophthalmology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, People's Republic of China.
| | - Genxi Li
- State Key Laboratory of Analytical Chemistry for Life Science, School of Life Sciences, Nanjing University, Nanjing 210023, People's Republic of China; Center for Molecular Recognition and Biosensing, School of Life Sciences, Shanghai University, Shanghai 200444, People's Republic of China.
| |
Collapse
|
2
|
Arabi S, Fadaee M, Kazemi T, Rahmani M. Advancements in colorectal cancer immunotherapy: from CAR-T cells to exosome-based therapies. J Drug Target 2025; 33:749-760. [PMID: 39754507 DOI: 10.1080/1061186x.2024.2449482] [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: 09/27/2024] [Revised: 12/03/2024] [Accepted: 12/30/2024] [Indexed: 01/06/2025]
Abstract
Colorectal cancer (CRC) continues to be a major worldwide health issue, with elevated death rates linked to late stages of the illness. Immunotherapy has made significant progress in developing effective techniques to improve the immune system's capacity to identify and eradicate cancerous cells. This study examines the most recent advancements in CAR-T cell treatment and exosome-based immunotherapy for CRC. CAR-T cell therapy, although effective in treating blood cancers, encounters obstacles when used against solid tumours such as CRC. These obstacles include the presence of an immunosuppressive tumour microenvironment and a scarcity of tumour-specific antigens. Nevertheless, novel strategies like dual-receptor CAR-T cells and combination therapy involving cytokines have demonstrated promise in surmounting these obstacles. Exosome-based immunotherapy is a promising approach for targeted delivery of therapeutic drugs to tumour cells, with high specificity and minimal off-target effects. However, there are still obstacles to overcome in the field, such as resistance to treatment, adverse effects associated with the immune system, and the necessity for more individualised methods. The current research is focused on enhancing these therapies, enhancing the results for patients, and ultimately incorporating these innovative immunotherapeutic approaches into the standard treatment protocols for CRC.
Collapse
Affiliation(s)
- Sepideh Arabi
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Manouchehr Fadaee
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Mohammadreza Rahmani
- Department of Immunology, Faculty of Medicine, Kurdistan University of Medical Sciences, Sanandaj, Iran
| |
Collapse
|
3
|
Chen Y, Yin W, Liu Z, Lu G, Zhang X, Yang J, Huang Y, Hu X, Chen C, Shang R, Hu W, Wang J, Shen HM, Hu J, Luo G, He W. Exosomes derived from fibroblasts enhance skin wound angiogenesis by regulating HIF-1α/VEGF/VEGFR pathway. BURNS & TRAUMA 2025; 13:tkae071. [PMID: 40433567 PMCID: PMC12107542 DOI: 10.1093/burnst/tkae071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/05/2024] [Revised: 10/19/2024] [Accepted: 10/30/2024] [Indexed: 05/29/2025]
Abstract
Background Angiogenesis is vital for tissue repair but insufficient in chronic wounds due to paradoxical growth factor overexpression yet reduced neovascularization. Therapeutics physiologically promoting revascularization remain lacking. This study aims to investigate the molecular mechanisms underlying fibroblast-derived exosome-mediated angiogenesis during wound repair. Methods To assess the effects of fibroblasts derived exosomes on wound healing and angiogenesis, a full-thickness mouse skin injury model was established, followed by pharmacological inhibition of exosome secretion. The number and state of blood vessels in wounds were assessed by immunofluorescence, immunohistochemistry, hematoxylin-eosin staining, and laser Doppler imaging system. The high-throughput miRNA sequencing was carried out to detect the miRNA profiles of fibroblast-derived exosomes. The roles of candidate miRNAs, their target genes, and relevant pathways were predicted by bioinformatic online software. The knockdown and overexpression of candidate miRNAs, co-culture system, matrigel assay, pharmacological blockade, cell migration, EdU incorporation assay, and cell apoptosis were employed to investigate their contribution to angiogenesis mediated by fibroblast-derived exosomes. The expression of vascular endothelial growth factor A (VEGFA), vascular endothelial growth factor receptor 2 (VEGFR2), hypoxia-inducible factor 1α (HIF-1α), von Hippel-Lindau (VHL), and proline hydroxylases 2 was detected by western blot, co-immunoprecipitation, immunofluorescence, real-time quantitative polymerase chain reaction, flow cytometry, and immunohistochemistry. Furthermore, a full-thickness mouse skin injury model based on type I diabetes mellitus induced by streptozotocin was established for estimating the effect of fibroblast-derived exosomes on chronic wound healing. Results Pharmacological inhibition of exosome biogenesis markedly reduces neovascularization and delays murine cutaneous wound closure. Topical administration of fibroblast-secreted exosomes rescues these defects. Mechanistically, exosomal microRNA-24-3p suppresses VHL E3 ubiquitin ligase levels in endothelial cells to stabilize hypoxia-inducible factor-1α and heighten vascular endothelial growth factor signaling. MicroRNA-24-3p-deficient exosomes exhibit attenuated pro-angiogenic effects. Strikingly, topical application of exosomes derived from fibroblasts onto chronic wounds in diabetic mice improves neovascularization and healing dynamics. Conclusions Overall, we demonstrate central roles for exosomal miR-24-3p in stimulating endothelial HIF-VEGF signaling by inhibiting VHL-mediated degradation. The findings establish fibroblast-derived exosomes as promising acellular therapeutic candidates to treat vascular insufficiency underlying recalcitrant wounds.
Collapse
Affiliation(s)
- Yunxia Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Wenjing Yin
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Zhihui Liu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Guang Lu
- Zhongshan School of Medicine, Sun Yat-sen University, Zhongshan second road, Yuexiu district, Guangzhou 510062, China
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore
| | - Xiaorong Zhang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Jiacai Yang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Yong Huang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Xiaohong Hu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Cheng Chen
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Ruoyu Shang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Wengang Hu
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Jue Wang
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Han-Ming Shen
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, 21 Lower Kent Ridge Road, Singapore 119077, Singapore
- Faculty of Health Sciences, Ministry of Education Frontiers Science Center for Precision Oncology, University of Macau, Macao Taipa University Avenue, Macau 999078, China
| | - Jun Hu
- Department of Neurology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Gaoxing Luo
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| | - Weifeng He
- State Key Laboratory of Trauma, Burn and Combined Injury, Institute of Burn Research, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
- Chongqing Key Lab for Wound Repair and Tissue Regeneration, Southwest Hospital, Third Military Medical University (Army Military Medical University), Gaotanyan street, Shapingba district, Chongqing 400038, China
| |
Collapse
|
4
|
Aakel N, Mohammed R, Fathima A, Kerzabi R, Abdallah A, Ibrahim WN. Role of Exosome in Solid Cancer Progression and Its Potential Therapeutics in Cancer Treatment. Cancer Med 2025; 14:e70941. [PMID: 40344389 PMCID: PMC12063069 DOI: 10.1002/cam4.70941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/14/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Exosomes are extracellular vesicles ranging from 40 to 100 nm in diameter that mediate intercellular communication by transferring proteins, lipids, nucleic acids, and other metabolites. In the context of cancer, exosomes influence the tumor microenvironment by carrying regulatory RNAs such as miRNA, circRNA, and lncRNA. They originate from various cells, including adipocytes, fibroblasts, and hepatocellular carcinoma (HCC) cells, and can either promote or inhibit cancer progression through pathways like MAPK and PI3K-Akt. AIM This review aims to explore the role of exosomes in the progression of solid cancers, emphasizing their self-induced activation mechanisms and how they modulate tumor behavior. METHODOLOGY A comprehensive review of recent literature was conducted, focusing on studies that investigated the biological functions of exosomes in solid tumor progression, including their molecular cargo, cellular origin, and involvement in signaling pathways. RESULTS Findings from multiple studies indicate that cancer-derived exosomes contribute to tumor proliferation, metastasis, and therapy resistance by enhancing communication within the tumor microenvironment. These vesicles activate oncogenic pathways and can serve as biomarkers or therapeutic targets due to their role in disease modulation. CONCLUSION Exosomes play a pivotal role in solid cancer progression and offer significant potential in advancing our understanding of tumor biology. Their capacity to influence key signaling pathways and facilitate intercellular communication makes them promising candidates for novel diagnostic and therapeutic strategies.
Collapse
Affiliation(s)
- Nada Aakel
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Rawdhah Mohammed
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Assela Fathima
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Rabia Kerzabi
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Atiyeh Abdallah
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| | - Wisam Nabeel Ibrahim
- Department of Biomedical ScienceCollege of Health Sciences, QU Health, Qatar UniversityDohaQatar
| |
Collapse
|
5
|
Tiwari PK, Chaudhary AA, Gupta S, Chouhan M, Singh HN, Rustagi S, Khan SUD, Kumar S. Extracellular vesicles in triple-negative breast cancer: current updates, challenges and future prospects. Front Mol Biosci 2025; 12:1561464. [PMID: 40297849 PMCID: PMC12034555 DOI: 10.3389/fmolb.2025.1561464] [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: 01/15/2025] [Accepted: 02/25/2025] [Indexed: 04/30/2025] Open
Abstract
Breast cancer (BC) remains a complex and widespread problem, affecting millions of women worldwide, Among the various subtypes of BC, triple-negative breast cancer (TNBC) is particularly challenging, representing approximately 20% of all BC cases, and the survival rate of TNBC patients is generally worse than other subtypes of BC. TNBC is a heterogeneous disease characterized by lack of expression of three receptors: estrogen (ER), progesterone (PR), and human epidermal growth factor receptor 2 (HER2), resulting conventional hormonal therapies are ineffective for its management. Despite various therapeutic approaches have been explored, but no definitive solution has been found yet for TNBC. Current treatments options are chemotherapy, immunotherapy, radiotherapy and surgery, although, these therapies have some limitations, such as the development of resistance to anti-cancer drugs, and off-target toxicity, which remain primary obstacles and significant challenges for TNBC. Several findings have shown that EVs exhibit significant therapeutic promise in many diseases, and a similar important role has been observed in various types of tumor. Studies suggest that EVs may offer a potential solution for the management of TNBC. This review highlights the multifaceted roles of EVs in TNBC, emphasizing their involvement in disease progression, diagnosis and therapeutic approach, as well as their potential as biomarkers and drug delivery.
Collapse
Affiliation(s)
- Prashant Kumar Tiwari
- Biological and Bio-Computational Lab, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Anis Ahmad Chaudhary
- Department of Biology, College of Science, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Saurabh Gupta
- Department of Biotechnology, GLA University, Mathura, Uttar Pradesh, India
| | - Mandeep Chouhan
- Biological and Bio-Computational Lab, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| | - Himanshu Narayan Singh
- Department of Systems Biology, Columbia University Irving Medical Center, New York, NY, United States
| | - Sarvesh Rustagi
- Department of Food Technology, School of Applied and Life science, Uttaranchal University, Dehradun, Uttarakhand, India
| | - Salah-Ud-Din Khan
- Department of Biochemistry, College of Medicine, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Sanjay Kumar
- Biological and Bio-Computational Lab, Department of Life Sciences, School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, India
| |
Collapse
|
6
|
Massaro C, Sgueglia G, Muro A, Pieragostino D, Lanuti P, Cufaro MC, Giorgio C, D'Agostino E, Torre LD, Baglio SR, Pirozzi M, De Simone M, Altucci L, Dell'Aversana C. Vorinostat impairs the cancer-driving potential of leukemia-secreted extracellular vesicles. J Transl Med 2025; 23:421. [PMID: 40211278 PMCID: PMC11987450 DOI: 10.1186/s12967-025-06361-1] [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: 01/16/2025] [Accepted: 03/10/2025] [Indexed: 04/12/2025] Open
Abstract
BACKGROUND Leukemia-secreted extracellular vesicles (EVs) carry biologically active cargo that promotes cancer-supportive mechanisms, including aberrant proliferative signaling, immune escape, and drug resistance. However, how antineoplastic drugs affect EV secretion and cargo sorting remains underexplored. METHODS Leukemia-secreted extracellular vesicles (EVs) were isolated by Differential UltraCentrifugation, and their miRNome and proteomic profiling cargo were analyzed following treatment with SAHA (Vorinostat) in Acute Myeloid Leukemia (AML) and Chronic Myeloid Leukemia (CML). The epigenetic modulation of leukemia-secreted EVs content on interesting key target molecules was validated, and their differential functional impact on cellular viability, cell cycle progression, apoptosis, and tumorigenicity was assessed. RESULTS SAHA significantly alters the cargo of Leukemia-derived EVs, including miR-194-5p and its target BCLAF1 (mRNA and protein), key regulators of Leukemia cell survival and differentiation. SAHA upregulates miR-194-5p expression while selective loading BCLAF1 into EVs, reducing the miRNA levels in the same compartment. Additionally, SAHA alters miRNA profile and proteomic composition associated with leukemic EVs, altering their tumor-supportive potential, with differential effects observed between AML and CML. Furthermore, in silico predictions suggest that these modified EVs may influence cell sensitivity to antineoplastic agents, suggesting a dual role for SAHA in impairing oncogenic signaling while enhancing therapeutic responsiveness. CONCLUSIONS In conclusion, the capacity of SAHA to modulate secretion and molecular composition of Leukemia-secreted EVs, alongside its direct cytotoxic effects, underscores its potential in combination therapies aimed to overcoming refractory phenotype by targeting EV-mediated communication.
Collapse
MESH Headings
- Extracellular Vesicles/drug effects
- Extracellular Vesicles/metabolism
- Extracellular Vesicles/pathology
- Vorinostat/pharmacology
- Vorinostat/therapeutic use
- Carcinogenesis/drug effects
- Carcinogenesis/genetics
- Carcinogenesis/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Cell Survival/drug effects
- Cell Survival/genetics
- Cell Differentiation/drug effects
- Cell Differentiation/genetics
- Gene Expression Regulation, Leukemic/drug effects
- Gene Expression Regulation, Leukemic/genetics
- Proteomics
- Proteome/analysis
- Proteome/drug effects
- Cell Line, Tumor
- Humans
- MicroRNAs/genetics
- MicroRNAs/metabolism
- Tumor Suppressor Proteins/genetics
- Tumor Suppressor Proteins/metabolism
- Computer Simulation
- Drug Resistance, Neoplasm/drug effects
- Drug Resistance, Neoplasm/genetics
- Antineoplastic Agents/pharmacology
- Antineoplastic Agents/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Epigenesis, Genetic/drug effects
Collapse
Affiliation(s)
- Crescenzo Massaro
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Giulia Sgueglia
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Annamaria Muro
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Damiana Pieragostino
- Department of Innovative Technologies in Medicine & Dentistry, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Paola Lanuti
- Department of Medicine and Aging Sciences, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Maria Concetta Cufaro
- Department of Innovative Technologies in Medicine & Dentistry, Center for Advanced Studies and Technology (CAST), University "G. d'Annunzio" of Chieti-Pescara, 66100, Chieti, Italy
| | - Cristina Giorgio
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
- Research & Early Development (R&D), Dompé Farmaceutici S.p.A, Via De Amicis, 80131, Naples, Italy
| | - Erika D'Agostino
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Laura Della Torre
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy
| | - Serena Rubina Baglio
- Department of Pathology, Cancer Center Amsterdam, Amsterdam UMC, Vrije Universiteit Amsterdam, 1081HV, Amsterdam, The Netherlands
| | - Marinella Pirozzi
- Istituto degli Endotipi in Oncologia, Metabolismo e Immunologia "G. Salvatore" (IEOMI)-National Research Council (CNR), 80131, Naples, Italy
| | - Mariacarla De Simone
- Stem Cell Transplantation Unit, Division Hematology, Cardarelli Hospital, 80131, Naples, Italy
| | - Lucia Altucci
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy.
- Istituto degli Endotipi in Oncologia, Metabolismo e Immunologia "G. Salvatore" (IEOMI)-National Research Council (CNR), 80131, Naples, Italy.
- BIOGEM, 83031, Ariano Irpino, Italy.
- Medical Epigenetics Program, Vanvitelli Hospital, Naples, Italy.
| | - Carmela Dell'Aversana
- Department of Precision Medicine, University of Campania "Luigi Vanvitelli", 80138, Naples, Italy.
- Istituto degli Endotipi in Oncologia, Metabolismo e Immunologia "G. Salvatore" (IEOMI)-National Research Council (CNR), 80131, Naples, Italy.
- Department of Medicine and Surgery, LUM University, Casamassima, BA, Italy.
| |
Collapse
|
7
|
Wu Y, Wang C, Guo Y, Zhang Y, Zhang X, Wang P, Yue W, Zhu X, Liu Z, Zhang Y, Guo H, Han L, Li M. Small extracellular vesicle-based one-step high-throughput microfluidic platform for epithelial ovarian cancer diagnosis. J Nanobiotechnology 2025; 23:278. [PMID: 40189497 PMCID: PMC11974170 DOI: 10.1186/s12951-025-03348-4] [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: 12/16/2024] [Accepted: 03/23/2025] [Indexed: 04/09/2025] Open
Abstract
BACKGROUND Ovarian cancer (OC) is diagnosed at advanced stages, resulting in limited treatment options for patients. While early detection of OC has been investigated, the invasiveness of approaches, high sample requirements, or false-positive rates undermined its benefits. Here, we present a "one-step" high-throughput microfluidic platform for epithelial ovarian cancer (EOC) detection that integrates small extracellular vesicle (sEV) capture, in situ lysis, and protein biomarker detection. RESULTS We identified 1,818 differentially expressed proteins (DEPs) through proteomic analysis of sEVs from patients' serum, combined with cell lines. Through multi-step screening of DEPs, we identified EOC biomarkers to customize the microfluidic platform. We used the microfluidic platform to test the expression of EOC biomarkers with 2 µL of serum from 209 participants in a prospective cohort. Based on the test results, an EOC detection model (P9) was constructed, which achieved a sensitivity of 92.3% (95% CI, 75.9-97.9%) for stage I, 90.0% (95% CI, 69.9-97.2%) for stage II at a specificity of 98.8% (95% CI, 93.6-99.8%) in the training set. The specificities reached 98.8% (95% CI, 93.6-99.8%) in the training set and 100.0% (95% CI, 91.6-100.0%) in the validation set of a held-out group of 105 participants. A model combining the P9 and patient's CA125 value exhibited 100.0% (95% CI, 95.6-100%) specificity in both training and validation, without compromising sensitivity. CONCLUSIONS We developed a non-invasive high-throughput microfluidic platform for EOC sEV-derived biomarker detection. It significantly reduced false positives and sample volume. Given its convenience and low cost, this platform could advance OC early detection to benefit of women.
Collapse
Affiliation(s)
- Yu Wu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Chao Wang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Yuhan Guo
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Yunhong Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Xue Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China
| | - Pan Wang
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Wei Yue
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Xin Zhu
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China
- National Clinical Research Center for Obstetrics and Gynecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China
| | - Zhaofei Liu
- Department of Radiation Medicine, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, 100191, China
| | - Yu Zhang
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China.
- School of Integrated Circuits, Shandong University, Jinan, 250100, China.
| | - Hongyan Guo
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China.
| | - Lin Han
- Institute of Marine Science and Technology, Shandong University, Qingdao, 266237, China.
- School of Integrated Circuits, Shandong University, Jinan, 250100, China.
| | - Mo Li
- State Key Laboratory of Female Fertility Promotion, Center for Reproductive Medicine, Department of Obstetrics and Gynecology, Peking University Third Hospital, Beijing, 100191, China.
- National Clinical Research Center for Obstetrics and Gynecology, Third Hospital, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, 100871, China.
- Key Laboratory of Assisted Reproduction (Peking University), Ministry of Education, Beijing, 100191, China.
- Beijing Key Laboratory of Reproductive Endocrinology and Assisted Reproductive Technology (Peking University Third Hospital), Beijing, 100191, China.
| |
Collapse
|
8
|
Zhang Z, Zhang L, Huang Y, Wang Z, Ren Z. A Planar-Gate Graphene Field-Effect Transistor Integrated Portable Platform for Rapid Detection of Colon Cancer-Derived Exosomes. BIOSENSORS 2025; 15:207. [PMID: 40277521 PMCID: PMC12025066 DOI: 10.3390/bios15040207] [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: 02/10/2025] [Revised: 03/13/2025] [Accepted: 03/19/2025] [Indexed: 04/26/2025]
Abstract
Early diagnosis of diseases would significantly increase the survival rate of cancer patients. However, current screening methods are complex and costly, making them unsuitable for rapid health diagnosis in daily life. Here, we develop a portable platform based on a planar-gate graphene field-effect transistor functionalized with polydopamine self-assembled film (PDA-GFET), capable of identifying colon cancer through the detection of EpCAM protein, which is expressed on colon cancer-derived exosomes, in clinical samples within 10 min. The PDA self-assembled film on the graphene and gate surface enhances the biosensor's functionalization area while suppressing non-specific adsorption, thereby achieving detection limits as low as 112 particles/mL. In addition, the PDA-GFET-based detection platform was used to identify EpCAM protein in real clinical samples from healthy individuals and colon cancer patients within 10 min, and the two showed significant differences (p < 0.001). Results indicate that the proposed PDA-GFET-based detection platform is expected to be a potential tool for the early diagnosis of colon cancer.
Collapse
Affiliation(s)
- Zaiyu Zhang
- Key Laboratory of High-Efficiency and Clean Mechanical Manufacture, Ministry of Education, Department of Mechanical Engineering, Shandong University, Jinan 250100, China; (Z.Z.)
| | - Luyang Zhang
- Key Laboratory of High-Efficiency and Clean Mechanical Manufacture, Ministry of Education, Department of Mechanical Engineering, Shandong University, Jinan 250100, China; (Z.Z.)
| | - Yuting Huang
- Department of Clinical Laboratory, Qilu Hospital of Shandong University, Jinan 250012, China
- Shandong Engineering Research Center of Biomarker and Artificial Intelligence Application, Jinan 250012, China
| | - Ziran Wang
- Key Laboratory of High-Efficiency and Clean Mechanical Manufacture, Ministry of Education, Department of Mechanical Engineering, Shandong University, Jinan 250100, China; (Z.Z.)
| | - Zhongjing Ren
- Key Laboratory of High-Efficiency and Clean Mechanical Manufacture, Ministry of Education, Department of Mechanical Engineering, Shandong University, Jinan 250100, China; (Z.Z.)
| |
Collapse
|
9
|
Chi H, Shi L, Gan S, Fan G, Dong Y. Innovative Applications of Nanopore Technology in Tumor Screening: An Exosome-Centric Approach. BIOSENSORS 2025; 15:199. [PMID: 40277513 PMCID: PMC12024935 DOI: 10.3390/bios15040199] [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: 01/14/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 04/26/2025]
Abstract
Cancer remains one of the leading causes of death worldwide. Its complex pathogenesis and metastasis pose significant challenges for early diagnosis, underscoring the urgent need for innovative and non-invasive tumor screening methods. Exosomes, small extracellular vesicles that reflect the physiological and pathological states of their parent cells, are uniquely suited for cancer liquid biopsy due to their molecular cargo, including RNA, DNA, and proteins. However, traditional methods for exosome isolation and detection are often limited by inadequate sensitivity, specificity, and efficiency. Nanopore technology, characterized by high sensitivity and single-molecule resolution, offers powerful tools for exosome analysis. This review highlights its diverse applications in tumor screening, such as magnetic nanopores for high-throughput sorting, electrochemical sensing for real-time detection, nanomaterial-based assemblies for efficient capture, and plasmon resonance for ultrasensitive analysis. These advancements have enabled precise exosome detection and demonstrated promising potential in the early diagnosis of breast, pancreatic, and prostate cancers, while also supporting personalized treatment strategies. Additionally, this review summarizes commercialized products for exosome-based cancer diagnostics and examines the technical and translational challenges in clinical applications. Finally, it discusses the future prospects of nanopore technology in advancing liquid biopsy toward clinical implementation. The continued progress of nanopore technology not only accelerates exosome-based precision medicine but also represents a significant step forward in next-generation liquid biopsy and tumor screening.
Collapse
Affiliation(s)
- Heng Chi
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
| | - Liuxin Shi
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
| | | | | | - Yuliang Dong
- BGI Research, Shenzhen 518083, China; (H.C.); (L.S.)
- BGI Research, Hangzhou 310030, China;
| |
Collapse
|
10
|
Gao Y, Xie J, Yang Z, Li M, Yuan H, Li R. Functional tumor-derived exosomes in NSCLC progression and clinical implications. Front Pharmacol 2025; 16:1485661. [PMID: 40176898 PMCID: PMC11962733 DOI: 10.3389/fphar.2025.1485661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 02/27/2025] [Indexed: 04/05/2025] Open
Abstract
Non-small cell lung cancer (NSCLC) accounts for approximately 85% of all lung cancer cases and remains one of the leading causes of cancer-related mortality worldwide. The high mortality rate is primarily driven by delayed diagnosis, rapid metastasis, and frequent recurrence. Tumor-derived exosomes (TEXs) have emerged as critical mediators in NSCLC progression, offering valuable insights into the tumor microenvironment. Exosomes are small membrane vesicles that facilitate intercellular communication and transport bioactive molecules, including proteins, RNAs, and DNAs, thereby reflecting the genetic complexity of tumors. These exosomes play a key role in promoting tumor metastasis, epithelial-mesenchymal transition (EMT), neovascularization, drug resistance, and immune evasion, all of which are pivotal in the development of NSCLC. This review explores the diverse roles of TEXs in NSCLC progression, focusing on their involvement in pre-metastatic niche formation, tissue metastasis, and immune modulation. Specifically, we discuss the roles of exosome-associated RNAs and proteins in NSCLC, and their contribute to tumor growth and metastasis. Furthermore, we explore the potential of TEXs as biomarkers for NSCLC, emphasizing their application in diagnosis, prognosis, and prediction of resistance to targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Yuxin Gao
- Department of Abdominal Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Chengdu, China
- Information Technology Center, West China Sanya Hospital of Sichuan University, Sanya, China
| | - Zhenya Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, China
| | - Mengxi Li
- College of pharmacy, Chengdu Medical College, Chengdu, China
| | - Hongfan Yuan
- Department of Oncology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
11
|
Madhan S, Dhar R, Devi A. Clinical Impact of Exosome Chemistry in Cancer. ACS APPLIED BIO MATERIALS 2025; 8:1862-1876. [PMID: 39936581 DOI: 10.1021/acsabm.4c01920] [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] [Indexed: 02/13/2025]
Abstract
As we progress into the 21st century, cancer stands as one of the most dreaded diseases. With approximately one in every four individuals facing a lifetime risk of developing cancer, cancer remains one of the most serious health challenges worldwide. Its multifaceted nature makes it an arduous and tricky problem to diagnose and treat. Over the years, researchers have explored plenty of approaches and avenues to improve cancer management. One notable strategy includes the study of extracellular vesicles (EVs) as potential biomarkers and therapeutics. Among these EVs, exosomes have emerged as particularly promising candidates due to their unique characteristic properties and functions. They are small membrane-bound vesicles secreted by cells carrying a cargo of biomolecules such as proteins, nucleic acids, and lipids. These vesicles play crucial roles in intercellular communication, facilitating the transfer of biological information between cell-to-cell communication. Exosomes transport cargoes such as DNA, RNA, proteins, and lipids involved in cellular reprogramming and promoting cancer. In this review, we explore the molecular composition of exosomes, significance of exosomes chemistry in cancer development, and its theranostic application as well as exosomes research complications and solutions.
Collapse
Affiliation(s)
- Shrishti Madhan
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu-603203, India
| |
Collapse
|
12
|
Cheng W, Pan H, Chen J, He M, Wang Z, Xiang Y. Enzyme-Free and Triple Sensitivity Amplification for Electrochemical Detection of Exosomal microRNA. Anal Chem 2025; 97:5244-5250. [PMID: 40017114 DOI: 10.1021/acs.analchem.4c06879] [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: 03/01/2025]
Abstract
Exosomal miRNAs, particularly miRNA-21, are promising cancer biomarkers. Current enzyme-dependent detection methods face challenges, such as environmental limitations and high costs. In contrast, enzyme-independent sensors are highly desirable for on-site, miniaturized, and cost-effective miRNA detection. To address these limitations, we developed a nonenzymatic electrochemical sensor featuring a triple-signal amplification system for ultrasensitive detection of miRNA-21. This sensor utilizes cascade toehold-mediated strand displacement reactions to activate molecular machines triggered by target miRNA, generating biotinylated-and-thiol-modified double-stranded DNA for stable immobilization on a gold electrode. Preprepared biotinylated tetrahedron DNA (TDNA)-mediated hybridization chain reaction probes are then linked to the electrode via streptavidin-biotin binding. This amplification process allows for significant DNA duplex immobilization, with electroactive [Ru(NH3)6]3+ (RuHex) adsorbed onto them, producing a robust electrochemical signal. This approach enables accurate detection of miRNA-21 at concentrations as low as 0.43 fM, with a linear range from 1 fM to 1 nM. Clinical testing demonstrates its potential for cancer diagnostics.
Collapse
Affiliation(s)
- Wenting Cheng
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Han Pan
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Jinhua Chen
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Miao He
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
| | - Zhongyun Wang
- Department of Anesthesiology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Yang Xiang
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing 210023, P. R. China
- State Key Laboratory of Natural and Biomimetic Drugs, Peking University, Beijing 100191, P. R. China
| |
Collapse
|
13
|
Chen Y, Zhang M, Qi Y, Lin Y, Liu S, Deng C, Jiang S, Sun N. Efficient extraction via titanium organic frameworks facilitates in-depth profiling of urinary exosome metabolite fingerprints. Anal Bioanal Chem 2025; 417:1543-1555. [PMID: 39853354 DOI: 10.1007/s00216-025-05741-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/03/2025] [Accepted: 01/07/2025] [Indexed: 01/26/2025]
Abstract
Urinary exosome metabolite analysis has demonstrated notable advantages in uncovering disease status, yet its potential in decoding the intricacies of clear cell renal cell carcinoma (ccRCC) remains untapped. To address this, a core-shell magnetic titanium organic framework was designed to capture urinary exosomes and assist laser desorption/ionization mass spectrometry (LDI MS) to decipher the exosomal metabolic profile of ccRCC, with high sensitivity, throughput, and speed. A total of 492 urinary exosome metabolite fingerprints (UEMFs) from 176 samples were extracted for exploring the differences between ccRCC and healthy individuals. Leveraging machine learning algorithms, the exosomal metabolic profile was disclosed, achieving accurate differentiation and prediction of ccRCC patients versus healthy individuals, with an accuracy exceeding 97.3%. Furthermore, an optimized algorithm panel comprising five key features demonstrated consistent and high diagnosing accuracy rates of over 94.0% both in the training and blind test sets for ccRCC, underscoring the remarkable effectiveness and superiority of this strategy in ccRCC detection. This study not only refines the LDI MS method for metabolite analysis in urinary exosomes but also introduces a promising technical approach for unraveling the mysteries of ccRCC.
Collapse
Affiliation(s)
- Yijie Chen
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Man Zhang
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Yu Qi
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch Fudan University, Shanghai, 200032, China
| | - Yiwen Lin
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Shasha Liu
- Department of Chemistry, Fudan University, Shanghai, 200433, China
| | - Chunhui Deng
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
- School of Chemistry and Chemical Engineering, Nanchang University, Nanchang, 330031, China.
| | - Shuai Jiang
- Department of Urology, Zhongshan Hospital, Zhongshan Hospital Wusong Branch Fudan University, Shanghai, 200032, China.
| | - Nianrong Sun
- Department of Gastroenterology and Hepatology, Zhongshan Hospital, Department of Chemistry, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
14
|
Li J, Zhao L, Li L, Wang X, Gao Y, Gao Y, Wang J. Urine exosomal lncRNAs as novel biomarkers for early diagnosis of bladder cancer based on microarray differential expression profiling. Int J Biol Markers 2025; 40:24-34. [PMID: 39943913 DOI: 10.1177/03936155251317551] [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] [Indexed: 03/15/2025]
Abstract
PurposeWe aimed to exploit a urine exosomal long non-coding RNAs (lncRNAs) fingerprint to facilitate the early diagnosis of bladder cancer.MethodsMicroarray differential expression profiling of lncRNAs was for the first time employed in urine exosomes from 10 non-muscle-invasive bladder cancer (NMIBC) patients and 10 healthy controls to screen out candidate exosomal lncRNA biomarkers, which were then verified by quantitative real-time polymerase chain reaction in three independent phases including bladder cancer cells, culture fluid and 200 NMIBC participants. Logistic regression was performed to construct a diagnostic model-the diagnostic potency of which was assessed.ResultsThe profile of three exosome-derived lncRNAs (CCDC148-AS1, XLOC_006419, and RP5-1148A21.3) was screened and further verified to be notably over-expressed in NMIBC patients and bladder cancer cell lines, and exhibited area under the receiver-operating characteristic curve values of 0.873, 0.825, and 0.834, respectively, in training, validation, and double-blind validation phases. The profile was superior to urinary cytology in discriminating NMIBC from healthy controls (P < 0.0001). A significant correlation existed between a higher level of CCDC148-AS1 and a higher tumor grade (P < 0.001), and up-regulated CCDC148-AS1 as well as XLOC_006419 were statistically related with tumor node metastasis stage (P = 0.004 and P = 0.031, respectively). These three identified lncRNAs were confirmed to originate from bladder cancer cells and be packaged within exosomes, thus staying sufficiently stable in urine.ConclusionsTumor-originated urine exosomal lncRNAs, as fingerprint in NMIBC, exhibited satisfying clinical significance in early diagnosis of bladder cancer.
Collapse
Affiliation(s)
- Jun Li
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Liming Zhao
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Luning Li
- Department of Gastroenterology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Xiaohua Wang
- Department of General Internal Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Yisheng Gao
- Department of Urology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Yongli Gao
- Department of Oncology, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| | - Jinfeng Wang
- Department of Nuclear Medicine, Linyi People's Hospital, Shandong Second Medical University, Linyi, Shandong Province 276003, China
| |
Collapse
|
15
|
Chen J, Hu S, Liu J, Jiang H, Wang S, Yang Z. Exosomes: a double-edged sword in cancer immunotherapy. MedComm (Beijing) 2025; 6:e70095. [PMID: 39968497 PMCID: PMC11831209 DOI: 10.1002/mco2.70095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 01/08/2025] [Accepted: 01/10/2025] [Indexed: 02/20/2025] Open
Abstract
Over the past few decades, immunotherapy has emerged as a powerful strategy to overcome the limitations of conventional cancer treatments. The use of extracellular vesicles, particularly exosomes, which carry cargoes capable of modulating the immune response, has been extensively explored as a potential therapeutic approach in cancer immunotherapy. Exosomes can deliver their cargo to target cells, thereby influencing their phenotype and immunomodulatory functions. They exhibit either immunosuppressive or immune-activating characteristics, depending on their internal contents. These exosomes originate from diverse cell sources, and their internal contents can vary, suggesting that there may be a delicate balance between immune suppression and stimulation when utilizing them for immunotherapy. Therefore, a thorough understanding of the molecular mechanisms underlying the role of exosomes in cancer progression is essential. This review focuses on the molecular mechanisms driving exosome function and their impact on the tumor microenvironment (TME), highlighting the intricate balance between immune suppression and activation that must be navigated in exosome-based therapies. Additionally, it underscores the challenges and ongoing efforts to optimize exosome-based immunotherapies, thereby making a significant contribution to the advancement of cancer immunotherapy research.
Collapse
Affiliation(s)
- Jiayi Chen
- School of Life SciencesJilin UniversityChangchunChina
| | - Siyuan Hu
- School of Life SciencesJilin UniversityChangchunChina
| | - Jiayi Liu
- School of Life SciencesJilin UniversityChangchunChina
| | - Hao Jiang
- School of Life SciencesJilin UniversityChangchunChina
| | - Simiao Wang
- School of Life SciencesJilin UniversityChangchunChina
| | - Zhaogang Yang
- School of Life SciencesJilin UniversityChangchunChina
| |
Collapse
|
16
|
Spinelli S, Tripodi D, Corti N, Zocchi E, Bruschi M, Leoni V, Dominici R. Roles, Functions, and Pathological Implications of Exosomes in the Central Nervous System. Int J Mol Sci 2025; 26:1345. [PMID: 39941112 PMCID: PMC11818369 DOI: 10.3390/ijms26031345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/20/2025] [Accepted: 01/31/2025] [Indexed: 02/16/2025] Open
Abstract
Exosomes are a subset of extracellular vesicles (EVs) secreted by nearly all cell types and have emerged as a novel mechanism for intercellular communication within the central nervous system (CNS). These vesicles facilitate the transport of proteins, nucleic acids, lipids, and metabolites between neurons and glial cells, playing a pivotal role in CNS development and the maintenance of homeostasis. Current evidence indicates that exosomes from CNS cells may function as either inhibitors or enhancers in the onset and progression of neurological disorders. Furthermore, exosomes have been found to transport disease-related molecules across the blood-brain barrier, enabling their detection in peripheral blood. This distinctive property positions exosomes as promising diagnostic biomarkers for neurological conditions. Additionally, a growing body of research suggests that exosomes derived from mesenchymal stem cells exhibit reparative effects in the context of neurological disorders. This review provides a concise overview of the functions of exosomes in both physiological and pathological states, with particular emphasis on their emerging roles as potential diagnostic biomarkers and therapeutic agents in the treatment of neurological diseases.
Collapse
Affiliation(s)
- Sonia Spinelli
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (S.S.); (M.B.)
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (N.C.); (R.D.)
| | - Domenico Tripodi
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (N.C.); (R.D.)
| | - Nicole Corti
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (N.C.); (R.D.)
| | - Elena Zocchi
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy;
| | - Maurizio Bruschi
- Laboratory of Molecular Nephrology, IRCCS Istituto Giannina Gaslini, 16147 Genoa, Italy; (S.S.); (M.B.)
- Department of Experimental Medicine (DIMES), University of Genoa, 16132 Genoa, Italy;
| | - Valerio Leoni
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (N.C.); (R.D.)
- Department of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Roberto Dominici
- Laboratory of Clinical Pathology and Toxicology, Hospital Pio XI of Desio, ASST-Brianza, 20832 Desio, Italy; (D.T.); (N.C.); (R.D.)
| |
Collapse
|
17
|
Chu L, Sun Y, Tang X, Duan X, Zhao Y, Xia H, Xu L, Zhang P, Sun K, Yang G, Wang A. The Tumor-Derived Exosomes Enhanced Bevacizumab across the Blood-Brain Barrier for Antiangiogenesis Therapy against Glioblastoma. Mol Pharm 2025; 22:972-983. [PMID: 39895311 DOI: 10.1021/acs.molpharmaceut.4c01227] [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] [Indexed: 02/04/2025]
Abstract
Antibody therapy has become a mature cancer treatment strategy, but only one antibody drug, bevacizumab (BEV) has been approved to treat glioblastoma (GBM). The natural blood-brain barrier (BBB) significantly limits the penetration of therapeutic antibodies into the brain. In this study, an antibody delivery platform based on exosomes (EXOs) has been developed, which can cross the BBB and effectively enter the brain tissue to deliver BEV for safe and effective GBM therapy. In vitro experiments have shown that EXO-BEV could efficiently penetrate the BBB and significantly inhibit the migration of endothelial cells. Biodistribution studies in vivo have revealed that EXO serves as an effective carrier for transporting a higher concentration of BEV across the BBB into the brain. Furthermore, in vivo antiglioma experiments have illustrated that the introduction of EXO-BEV into the brain can improve the degeneration of pathological tissues, increase the apoptosis of tumor cells, and significantly extend the survival time of the model animals. All of the results suggested that EXO-BEV could cross the BBB, thereby enhancing the apoptosis of tumor cells and mitigating angiogenesis in GBM. In conclusion, this innovative platform for antibody delivery emerges as a highly promising therapeutic strategy for the clinical treatment of GBM and other neurological disorders.
Collapse
Affiliation(s)
- Liuxiang Chu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
- Yantai Laishan Fourth People's Hospital, Yantai, Shandong 264036, PR China
| | - Yuchen Sun
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| | - Xiaohu Tang
- State Key Laboratory of Long-Acting and Targeting Drug Delivery System, Shandong Luye Pharmaceutical Co., Ltd, Yantai, Shandong 264003, PR China
| | - Xinliu Duan
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| | - Yanyan Zhao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| | - Hangyu Xia
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| | - Lixiao Xu
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| | - Peng Zhang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| | - Kaoxiang Sun
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| | - Gangqiang Yang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| | - Aiping Wang
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Key Laboratory of Molecular Pharmacology and Drug Evaluation, Ministry of Education, School of Pharmacy, Yantai University, Yantai, Shandong 264005, PR China
| |
Collapse
|
18
|
Topiwala IS, Ramachandran A, A MS, Sengupta R, Dhar R, Devi A. Exosomes and tumor virus interlink: A complex side of cancer. Pathol Res Pract 2025; 266:155747. [PMID: 39647256 DOI: 10.1016/j.prp.2024.155747] [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: 09/21/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/10/2024]
Abstract
Extracellular Vesicles (EVs) based cancer research reveals several complicated sides of cancer. EVs are classified as several subpopulations such as microvesicles, apoptotic bodies, and exosomes. In cancer, exosomes play a significant role as a cellular messenger in tumor development and progression. Tumor-derived exosomes (TEXs) are also a theranostic tool for cancer. Tumor virus-infected cell-derived EVs promote cancer development. Exosomes (a subpopulation of EVs) play a significant role in converting noninfecting cells to infected cells. It transports several biological active cargo (DNA, RNA, protein, and virions) towards the noninfected cells. This cellular transport enhances infection rates via reprogramming of noninfected cells. In this review, we explore tumor viruses, exosomes and tumor viruses interlink, the theranostic landscape of exosomes in tumor virus-associated cancer and the future orientation of exosomes-based virus oncology.
Collapse
Affiliation(s)
- Ibrahim S Topiwala
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India
| | - Aparna Ramachandran
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India
| | - Meghana Shakthi A
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India
| | - Ranjini Sengupta
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Chengalpattu District, Tamil Nadu 603203, India.
| |
Collapse
|
19
|
Guo Y, Wang H, Liu S, Zhang X, Zhu X, Huang L, Zhong W, Guan L, Chen Y, Xiao M, Ou L, Yang J, Chen X, Huang AC, Mitchell T, Amaravadi R, Karakousis G, Miura J, Schuchter L, Flowers A, Zheng Q, Mou H, Gimotty P, Herlyn M, Guo W, Xu X. Engineered extracellular vesicles with DR5 agonistic scFvs simultaneously target tumor and immunosuppressive stromal cells. SCIENCE ADVANCES 2025; 11:eadp9009. [PMID: 39813334 PMCID: PMC11734719 DOI: 10.1126/sciadv.adp9009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
Small extracellular vesicles (sEVs) are nanosized vesicles. Death receptor 5 (DR5) mediates extrinsic apoptosis. We engineer DR5 agonistic single-chain variable fragment (scFv) expression on the surface of sEVs derived from natural killer cells. PDGFR transmembrane domain delivers DR5-scFvs to the surface of sEVs. DR5-scFv sEVs rapidly induce apoptosis of different types of DR5+ cancer cells, myeloid-derived suppressor cells (MDSCs), and cancer-associated fibroblasts (CAFs). DR5-scFv sEVs migrate specifically to DR5+ tumors in vitro and in vivo. Systemic delivery of DR5-scFv sEVs significantly inhibits the growth of DR5+ melanoma, liver cancer, and breast cancer and prolongs mouse life span without significant toxicity. DR5-scFv sEVs are significantly more efficacious than DR5 antibodies in vivo. In organotypic patient-derived melanoma slice cultures, DR5-scFv sEVs effectively inhibit melanoma cells and MDSCs and activate CD8+ T cells. Our studies demonstrate that DR5-scFv sEVs can inhibit tumor growth by targeting tumor cells and immunosuppressive stromal cells in the TME.
Collapse
Affiliation(s)
- Yeye Guo
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 41000, China
| | - Huaishan Wang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaogang Zhang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xingyue Zhu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lili Huang
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Wenqun Zhong
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lei Guan
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Yeqing Chen
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Min Xiao
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Lingling Ou
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jingbo Yang
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiang Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha 41000, China
| | - Alexander C. Huang
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tara Mitchell
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ravi Amaravadi
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Giorgos Karakousis
- Department of Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - John Miura
- Department of Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Lynn Schuchter
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ahron Flowers
- Department of Medicine and Abramson Cancer Center, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Qiuxian Zheng
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Haiwei Mou
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Phyllis Gimotty
- Department of Biostatistics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program, The Wistar Institute, Philadelphia, PA 19104, USA
| | - Wei Guo
- Department of Biology, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
20
|
Huang M, Ji J, Xu X, Jin D, Wu T, Lin R, Huang Y, Qian J, Tan Z, Jiang F, Hu X, Xu W, Xiao M. Known and unknown: Exosome secretion in tumor microenvironment needs more exploration. Genes Dis 2025; 12:101175. [PMID: 39524543 PMCID: PMC11550746 DOI: 10.1016/j.gendis.2023.101175] [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: 05/17/2023] [Revised: 09/06/2023] [Accepted: 10/10/2023] [Indexed: 11/16/2024] Open
Abstract
Exosomes, extracellular vesicles originating from endosomes, were discovered in the late 1980s and their function in intercellular communication has since garnered considerable interest. Exosomes are lipid bilayer-coated vesicles that range in size from 30 to 150 nm and appear as sacs under the electron microscope. Exosome secretion is crucial for cell-to-cell contact in both normal physiology and the development and spread of tumors. Furthermore, cancer cells can secrete more exosomes than normal cells. Scientists believe that intercellular communication in the complex tissue environment of the human body is an important reason for cancer cell invasion and metastasis. For example, some particles containing regulatory molecules are secreted in the tumor microenvironment, including exosomes. Then the contents of exosomes can be released by donor cells into the environment and interact with recipient cells to promote the migration and invasion of tumor cells. Therefore, in this review, we summarized the biogenesis of exosome, as well as exosome cargo and related roles. More importantly, this review introduces and discusses the factors that have been reported to affect exosome secretion in tumors and highlights the important role of exosomes in tumors.
Collapse
Affiliation(s)
- Mengxiang Huang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jie Ji
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xuebing Xu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Dandan Jin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Tong Wu
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Renjie Lin
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Yuxuan Huang
- Clinical Medicine, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Jiawen Qian
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Zhonghua Tan
- Department of Nuclear Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Feng Jiang
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| | - Xiaogang Hu
- Department of Respiratory Medicine, Rudong County People's Hospital, Nantong, Jiangsu 226400, China
| | - Weisong Xu
- Department of Gastroenterology, Affiliated Nantong Rehabilitation Hospital of Nantong University, Nantong, Jiangsu 226001, China
| | - Mingbing Xiao
- Department of Gastroenterology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, Jiangsu 226001, China
| |
Collapse
|
21
|
Singer J, Knezic N, Gohring G, Fite O, Christiansen J, Huard J. Synovial mesenchymal stem cells. ORTHOBIOLOGICS 2025:141-154. [DOI: 10.1016/b978-0-12-822902-6.00005-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
22
|
Haghbin M, Sotoodeh Jahromi A, Hashemi Tayer A, Ghasemi Nejad Z. The Potential Clinical Relevance of Procoagulant Microparticles as Biomarkers of Blood Coagulation in Breast Cancer: A Systematic Review. Asian Pac J Cancer Prev 2025; 26:23-32. [PMID: 39873982 PMCID: PMC12082420 DOI: 10.31557/apjcp.2025.26.1.23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 01/11/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND Breast cancer (BC) is a global challenge that affects a large portion of individuals, especially women. It has been suggested that microparticles (MPs) can be used as a diagnostic, prognostic, or therapeutic biomarker in various diseases. Moreover, MPs are known to elevate in cancer cases. Platelet-derived MPs (PMPs) play a crucial role in the metastasis of BC, warranting specific focus. This study aimed to explore the involvement of procoagulant MPs in BC. METHODS This systematic review was carried out using the Preferred Reporting Items for Systematic reviews, and Meta-Analyses (PRISMA). Terms defined as MESH keywords were searched PubMed/MEDLINE, Embase, Web of Science, and Cochrane Library searched in from 2011 to March 2024. Experimental and quasi-experimental studies were assessed by the CONSORT checklist. RESULTS Eventually, 15 studies were included. 426 participants were studied in the included articles. The potential clinical relevance of MPs as biomarkers in BC was indicated. Also, the role of MPs in immune modulation and multidrug resistance was approved. PMPs were found to enhance malignant features, including migration and invasion. Moreover, there were lower levels of MPs before neo-adjuvant chemotherapy, suggesting a potential impact of chemotherapy on MPs levels. The study highlights the remarkable capacity of multidrug-resistant BC-derived MPs to alter the phenotype and functionality of immune cells. CONCLUSIONS The findings underscore the intricate interplay between MPs and cellular signaling pathways, shedding light on their potential as diagnostic biomarkers, and therapeutic targets in cancer. Specifically, the association between MPs levels and disease severity, as evidenced by their correlation with tissue-based biomarkers, tumor grading, and distant metastasis, highlights their clinical relevance in prognostication and risk stratification.
Collapse
Affiliation(s)
| | | | - Akbar Hashemi Tayer
- Research Center for Noncommunicable Disease, Jahrom University of Medical Sciences, Jahrom, Iran.
| | | |
Collapse
|
23
|
Huang C, Li J, Xie Z, Hu X, Huang Y. Relationship between exosomes and cancer: formation, diagnosis, and treatment. Int J Biol Sci 2025; 21:40-62. [PMID: 39744442 PMCID: PMC11667803 DOI: 10.7150/ijbs.95763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/02/2024] [Indexed: 01/11/2025] Open
Abstract
Exosomes are a member of extracellular vesicles. However, their biological characteristics differ from those of other vesicles, and recently, their powerful functions as information molecules, biomarkers, and carriers have been demonstrated. Malignancies are the leading cause of high morbidity and mortality worldwide. The cure rate of malignancies can be improved by improving early screening rates and therapy. Moreover, a close correlation between exosomes and malignancies has been observed. An in-depth study of exosomes can provide new methods for diagnosing and treating tumors. Therefore, this study aimed to review, sort, and summarize such achievements, and present ideas and opinions on the application of exosomes in tumor treatment.
Collapse
Affiliation(s)
- Chen Huang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Jiajin Li
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Zichuan Xie
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Xiangjun Hu
- Sichuan university, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Yan Huang
- Health Management Center, General Practice Medical Center, West China Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Respiratory Health and Multimorbidity, China
- Research Laboratory for Prediction and Evaluation of Chronic Diseases in the Elderly, National Clinical Research Center for Geriatric Diseases, China
- General Practice Research Institute, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
Lahouty M, Fadaee M, Shanehbandi D, Kazemi T. Exosome-driven nano-immunotherapy: revolutionizing colorectal cancer treatment. Mol Biol Rep 2024; 52:83. [PMID: 39724304 DOI: 10.1007/s11033-024-10157-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 12/06/2024] [Indexed: 12/28/2024]
Abstract
Colorectal cancer (CRC) ranks as the third most common cancer worldwide and remains a major cause of cancer-related deaths, necessitating the development of innovative therapeutic approaches beyond conventional treatment modalities. Conventional therapies, such as radiation, chemotherapy, and surgery, are hindered by challenges like imprecise targeting, substantial toxicity, and the development of resistance. Exosome-driven nano-immunotherapy has emerged as a groundbreaking approach that leverages the natural properties of exosomes-cell-derived vesicles known for their role in intercellular communication-to deliver therapeutic agents with high precision and specificity. This approach utilizes the natural ability of exosomes to serve as natural nanocarriers for various biomolecules, such as proteins, nucleic acids, and lipids, enabling precise drug delivery and immune modulation. Exosomes offer distinct advantages compared to traditional drug delivery systems, including their biocompatibility, capability to traverse biological barriers, and suitability for personalized medicine approaches. We evaluate the effectiveness of exosome-based therapies in comparison to traditional approaches, emphasizing their ability to achieve precise delivery, minimize systemic toxicity, and enhance treatment results. Despite their promise, several challenges remain, including the standardization of exosome isolation and production, optimization of cargo loading techniques, and ensuring safety and efficacy in clinical applications. By overcoming these obstacles and leveraging the distinctive characteristics of exosomes, exosome-driven nano-immunotherapy presents a promising avenue for more efficient therapeutic interventions.
Collapse
Affiliation(s)
- Masoud Lahouty
- Department of Microbiology and Virology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Manouchehr Fadaee
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Student Research Committee, Tabriz University of Medical Science, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Immunology Research Center, Tabriz University of Medical Science, Tabriz, Iran.
| |
Collapse
|
25
|
Li X, Han H, Yang K, Li S, Ma L, Yang Z, Zhao YX. Crosstalk between thyroid CSCs and immune cells: basic principles and clinical implications. Front Immunol 2024; 15:1476427. [PMID: 39776907 PMCID: PMC11703838 DOI: 10.3389/fimmu.2024.1476427] [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: 08/05/2024] [Accepted: 12/04/2024] [Indexed: 01/11/2025] Open
Abstract
Thyroid cancer has become the most common endocrine malignancy. Although the majority of differentiated thyroid cancers have a favorable prognosis, advanced thyroid cancers, iodine-refractory thyroid cancers, and highly malignant undifferentiated carcinomas still face a serious challenge of poor prognosis and even death. Cancer stem cells are recognized as one of the central drivers of tumor evolution, recurrence and treatment resistance. A fresh viewpoint on the oncological aspects of thyroid cancer, including proliferation, invasion, recurrence, metastasis, and therapeutic resistance, has been made possible by the recent thorough understanding of the defining and developing features as well as the plasticity of thyroid cancer stem cells (TCSCs). The above characteristics of TCSCs are complicated and regulated by cell-intrinsic mechanisms (including activation of key stem signaling pathways, somatic cell dedifferentiation, etc.) and cell-extrinsic mechanisms. The complex communication between TCSCs and the infiltrating immune cell populations in the tumor microenvironment (TME) is a paradigm for cell-extrinsic regulators. This review introduces the current advances in the studies of TCSCs, including the origin of TCSCs, the intrinsic signaling pathways regulating the stemness of TCSCs, and emerging biomarkers; We further highlight the underlying principles of bidirectional crosstalk between TCSCs and immune cell populations driving thyroid cancer progression, recurrence, or metastasis, including the specific mechanisms by which immune cells maintain the stemness and other properties of TCSCs and how TCSCs reshape the immune microenvironmental landscape to create an immune evasive and pro-tumorigenic ecological niche. Finally, we outline promising strategies and challenges for targeting key programs in the TCSCs-immune cell crosstalk process to treat thyroid cancer.
Collapse
Affiliation(s)
- Xiaoxiao Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Hengtong Han
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Kaili Yang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Shouhua Li
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, China
| | - Libin Ma
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Ze Yang
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| | - Yong-xun Zhao
- The Seventh Department of General Surgery, Department of Thyroid Surgery, The First Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
26
|
Lin X, Zhu J, Shen J, Zhang Y, Zhu J. Advances in exosome plasmonic sensing: Device integration strategies and AI-aided diagnosis. Biosens Bioelectron 2024; 266:116718. [PMID: 39216205 DOI: 10.1016/j.bios.2024.116718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 08/11/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Exosomes, as next-generation biomarkers, has great potential in tracking cancer progression. They face many detection limitations in cancer diagnosis. Plasmonic biosensors have attracted considerable attention at the forefront of exosome detection, due to their label-free, real-time, and high-sensitivity features. Their advantages in multiplex immunoassays of minimal liquid samples establish the leading position in various diagnostic studies. This review delineates the application principles of plasmonic sensing technologies, highlighting the importance of exosomes-based spectrum and image signals in disease diagnostics. It also introduces advancements in miniaturizing plasmonic biosensing platforms of exosomes, which can facilitate point-of-care testing for future healthcare. Nowadays, inspired by the surge of artificial intelligence (AI) for science and technology, more and more AI algorithms are being adopted to process the exosome spectrum and image data from plasmonic detection. Using representative algorithms of machine learning has become a mainstream trend in plasmonic biosensing research for exosome liquid biopsy. Typically, these algorithms process complex exosome datasets efficiently and establish powerful predictive models for precise diagnosis. This review further discusses critical strategies of AI algorithm selection in exosome-based diagnosis. Particularly, we categorize the AI algorithms into the interpretable and uninterpretable groups for exosome plasmonic detection applications. The interpretable AI enhances the transparency and reliability of diagnosis by elucidating the decision-making process, while the uninterpretable AI provides high diagnostic accuracy with robust data processing by a "black-box" working mode. We believe that AI will continue to promote significant progress of exosome plasmonic detection and mobile healthcare in the near future.
Collapse
Affiliation(s)
- Xiangyujie Lin
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Jiaheng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China
| | - Jiaqing Shen
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China
| | - Youyu Zhang
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| | - Jinfeng Zhu
- Institute of Electromagnetics and Acoustics and Key Laboratory of Electromagnetic Wave Science and Detection Technology, Xiamen University, Xiamen, 361005, China; Shenzhen Research Institute of Xiamen University, Shenzhen, 518057, China.
| |
Collapse
|
27
|
Huang H, Wang Y, Gan J, Bu H, Li C, Li Y, Yang Y, Liu D. FRK exerts oncogenic effects by targeting NQO2 via exosomal miR-9-3p to regulate mitochondrial function. FASEB J 2024; 38:e70221. [PMID: 39625368 DOI: 10.1096/fj.202400951r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 10/29/2024] [Accepted: 11/19/2024] [Indexed: 03/17/2025]
Abstract
The role of Fyn-related kinase (FRK) in promoting tumor progression and metabolism in non-small cell lung cancer (NSCLC) has been demonstrated in our earlier study, and here we further explored whether exosome could serve as a key player in the relevant regulatory mechanism. Exosomes were isolated from the culture medium of FRK-knockout (KO) cells and co-cultured with NSCLC cells, showing negative effects on cell proliferation, metastasis, metabolism, and mitochondrial function. Exosomal miR-9-3p was selected as an intermediate messenger through microRNA (miRNA) sequencing. The miR-9-3p inhibitor and exosome inhibitor GW4869 were then applied in the co-culture system to verify the contribution of exosomal miR-9-3p to cell malignant phenotype and mitochondrial function. Moreover, NQO2, a potential target of exosomal miR-9-3p, was also suggested to be involved in the regulation of mitochondrial functional status. In brief, this study revealed a novel molecular mechanism by which FRK promotes the malignant progression of NSCLC by targeting NQO2 via exosomal miR-9-3p to strengthen mitochondrial function.
Collapse
Affiliation(s)
- Hong Huang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Institute of Clinical Pathology, Key Laboratory of Transplantation Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yan Wang
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jiadi Gan
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Bu
- Institute of Clinical Pathology, Key Laboratory of Transplantation Engineering and Immunology, Ministry of Health, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Cong Li
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yi Li
- Research Core Facility of West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yongfeng Yang
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Liu
- Department of Respiratory and Critical Care Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
28
|
Giannopoulos-Dimitriou A, Saiti A, Malousi A, Anagnostopoulos AK, Vatsellas G, Al-Maghrabi PM, Müllertz A, Fatouros DG, Vizirianakis IS. Molecular Profiling of A549 Cell-Derived Exosomes: Proteomic, miRNA, and Interactome Analysis for Identifying Potential Key Regulators in Lung Cancer. Cancers (Basel) 2024; 16:4123. [PMID: 39766023 PMCID: PMC11674491 DOI: 10.3390/cancers16244123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 11/28/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
BACKGROUND/OBJECTIVES Exosomes, nano-sized extracellular vesicles released by all cells, play a key role in intercellular communication and carry tumorigenic properties that impact surrounding or distant cells. The complexity of the exosomal molecular interactome and its effects on recipient cells still remain unclear. This study aims to decipher the molecular profile and interactome of lung adenocarcinoma A549 cell-derived exosomes using multi-omics and bioinformatics approaches. METHODS We performed comprehensive morphological and physicochemical characterization of exosomes isolated from cell culture supernatant of A549 cells in vitro, using DLS, cryo-TEM, Western blot, and flow cytometry. Proteomic and miRNA high-throughput profiling, coupled with bioinformatics network analysis, were applied to elucidate the exosome molecular cargo. A comparative miRNA analysis was also conducted with exosomes derived from normal lung fibroblast MRC-5 cells. RESULTS Exosomes exhibited an average size of ~40 nm and disk-shaped lipid bilayer structures, with tetraspanins CD9 and CD63 validated as exosomal markers. Proteomic analysis identified 68 proteins, primarily linked to the extracellular matrix organization and metabolic processes. miRNA sequencing revealed 72 miRNAs, notably hsa-miR-619-5p, hsa-miR-122-5p, hsa-miR-9901, hsa-miR-7704, and hsa-miR-151a-3p, which are involved in regulating metabolic processes, gene expression, and tumorigenic pathways. Th integration of proteomic and miRNA data through a proteogenomics approach identified dually affected genes including ERBB2, CD44, and APOE, impacted by both exosomal miRNA targeting and protein interactions through synergistic or antagonistic interactions. Differential analysis revealed a distinct miRNA profile in A549 exosomes, associated with cancer-related biological processes, compared to MRC-5 exosomes; notably, hsa-miR-619-5p emerged as a promising candidate for future clinical biomarker studies. The network analysis also revealed genes targeted by multiple upregulated tumor-associated miRNAs in potential exosome-recipient cells. CONCLUSIONS This integrative study provides insights into the molecular interactome of lung adenocarcinoma A549 cell-derived exosomes, providing a foundation for future research on exosomal cargo and its role in tumor cell communication, growth, and progression.
Collapse
Affiliation(s)
| | - Aikaterini Saiti
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.G.-D.); (A.S.)
| | - Andigoni Malousi
- Laboratory of Biological Chemistry, Medical School, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Athanasios K. Anagnostopoulos
- Proteomics Research Unit, Center of Basic Research II, Biomedical Research Foundation of the Academy of Athens, 11527 Athens, Greece;
| | - Giannis Vatsellas
- Greek Genome Center, Biomedical Research Foundation Academy of Athens, 11527 Athens, Greece;
| | - Passant M. Al-Maghrabi
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Anette Müllertz
- Department of Pharmacy, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Dimitrios G. Fatouros
- Laboratory of Pharmaceutical Technology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece;
| | - Ioannis S. Vizirianakis
- Laboratory of Pharmacology, School of Pharmacy, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece; (A.G.-D.); (A.S.)
- Department of Health Sciences, School of Life and Health Sciences, University of Nicosia, Nicosia 2417, Cyprus
| |
Collapse
|
29
|
Dilsiz N. A comprehensive review on recent advances in exosome isolation and characterization: Toward clinical applications. Transl Oncol 2024; 50:102121. [PMID: 39278189 PMCID: PMC11418158 DOI: 10.1016/j.tranon.2024.102121] [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: 05/04/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024] Open
Abstract
Exosomes are small, round vesicles in the 30 and 120 nm diameter range released by all living cell types. Exosomes play many essential functions in intercellular communication and tissue crosstalk in the human body. They can potentially be used as strong biomarkers and therapeutic agents for early diagnosis, therapy response, and prognosis of different diseases. The main requirements for exosomal large-scale clinical practice application are rapid, easy, high-yield, high purity, characterization, safety, low cost, and therapeutic efficacy. Depending on the sample types, environmental insults, and exosome quantity, exosomes can be isolated from various sources, including body fluids, solid tissues, and cell culture medium using different procedures. This study comprehensively analyzed the current research progress in exosome isolation and characterization strategies along with their advantages and disadvantages. The provided information will make it easier to select exosome separation methods based on the types of biological samples available, and it will facilitate the use of exosomes in translational and clinical research, particularly in cancer. Lay abstract Exosomes have recently received much attention due to their potential to function as biomarkers and novel therapeutic agents for early diagnosis, therapeutic response, and prognosis in various diseases. This review summarizes many approaches for isolating and characterizing exosomes, focusing on developing technologies, and provides an in-depth comparison and analysis of each method, including its principles, advantages, and limitations.
Collapse
Affiliation(s)
- Nihat Dilsiz
- Experimental Medicine Application and Research Center (EMARC) Validebag Research Park, University of Health Sciences, Istanbul, Turkey.
| |
Collapse
|
30
|
Liu Y, Hinnant B, Chen S, Tao H, Huang Z, Qian M, Zhou M, Han Z, Han ZC, Zhang J, Li Z. Hyaluronic acid-modified extracellular vesicles for targeted doxorubicin delivery in hepatocellular carcinoma. Exp Cell Res 2024; 443:114332. [PMID: 39551341 DOI: 10.1016/j.yexcr.2024.114332] [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: 10/06/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
Hepatocellular carcinoma (HCC), a prevalent and deadly cancer, poses a significant challenge with current treatments due to limitations such as poor stability, off-target effects, and severe side effects. Extracellular vesicles (EVs), derived from tumor cells, have the remarkable ability to home back to their cells of origin and can serve as Trojan horses for drug delivery. CD44, a cell surface glycoprotein, promotes cancer stem cell-like properties and is linked to poor prognosis and resistance to chemotherapy in HCC. Therefore, targeting CD44-expressing HCC cells is of interest in the development of novel therapeutic strategies for the treatment of HCC. In this study, we developed tumor cell-derived EVs (TEVs) functionalized with hyaluronic acid (HA) to serve as natural carriers for the precise delivery of doxorubicin (Dox), which specifically targets HCC cells expressing CD44. Our results demonstrated that HA-engineered EVs (HA-EVs) significantly enhanced Dox accumulation within HCC cells. In a mouse model, HA-EVs effectively delivered Dox to tumors, suppressing their growth and progression while minimizing systemic toxicity. This study demonstrates the potential of HA-functionalized EVs as a novel and targeted therapeutic platform for HCC, offering a valuable strategy for improving drug delivery and patient outcomes. This study presents a promising strategy to advance targeted chemotherapy for HCC and address the challenges associated with conventional treatments. Engineered HA-functionalized EVs offer a tailored and efficient approach to increase drug delivery precision, underscoring their potential as a novel therapeutic platform in the realm of HCC treatment.
Collapse
Affiliation(s)
- Yue Liu
- School of Medicine, Nankai University, China; The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, Tianjin, China
| | - Benjamin Hinnant
- Division of Biological Sciences, University of California San Diego, USA
| | - Shang Chen
- School of Medicine, Nankai University, China
| | - Hongyan Tao
- School of Medicine, Nankai University, China
| | - Ziyu Huang
- School of Medicine, Nankai University, China
| | - Meng Qian
- The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, Tianjin, China
| | - Manqian Zhou
- Department of Radiation Oncology, Tianjin Union Medical Center, Nankai University, Tianjin, China
| | - Zhibo Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China; Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health Biotech Co., Beijing, China
| | - Zhong-Chao Han
- Tianjin Key Laboratory of Engineering Technologies for Cell Pharmaceutical, National Engineering Research Center of Cell Products, AmCellGene Co., Ltd., Tianjin, China; Beijing Engineering Laboratory of Perinatal Stem Cells, Beijing Institute of Health and Stem Cells, Health Biotech Co., Beijing, China
| | - Jun Zhang
- Department of Anesthesiology and Pain Medical Center, Tianjin Union Medical Center, Nankai University, Tianjin, China.
| | - Zongjin Li
- School of Medicine, Nankai University, China; The Key Laboratory of Bioactive Materials, Ministry of Education, Nankai University, the College of Life Sciences, Tianjin, China; National Key Laboratory of Kidney Diseases, Chinese PLA General Hospital, Beijing, China; Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Zhengzhou, China.
| |
Collapse
|
31
|
Ismail M, Fadul MM, Taha R, Siddig O, Elhafiz M, Yousef BA, Jiang Z, Zhang L, Sun L. Dynamic role of exosomal long non-coding RNA in liver diseases: pathogenesis and diagnostic aspects. Hepatol Int 2024; 18:1715-1730. [PMID: 39306594 DOI: 10.1007/s12072-024-10722-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 08/15/2024] [Indexed: 12/11/2024]
Abstract
BACKGROUND Liver disease has emerged as a significant health concern, characterized by high rates of morbidity and mortality. Circulating exosomes have garnered attention as important mediators of intercellular communication, harboring protein and stable mRNAs, microRNAs, and long non-coding RNAs (lncRNA). This review highlights the involvement of exosomal lncRNA in the pathogenesis and diagnosis of various liver diseases. Notably, exosomal lncRNAs exhibit therapeutic potential as targets for conditions including hepatic carcinoma, hepatic fibrosis, and hepatic viral infections. METHOD An online screening process was employed to identify studies investigating the association between exosomal lncRNA and various liver diseases. RESULT Our study revealed a diverse array of lncRNAs carried by exosomes, including H19, Linc-ROR, VLDLR, MALAT1, DANCR, HEIH, ENSG00000248932.1, ENST00000457302.2, ZSCAN16-AS1, and others, exhibiting varied levels across different liver diseases compared to normal liver tissue. These exosomal-derived lncRNAs are increasingly recognized as pivotal biomarkers for diagnosing and prognosticating liver diseases, supported by emerging evidence. However, the precise mechanisms underlying the involvement of certain exosomal lncRNAs remain incompletely understood. Furthermore, the combined analysis of serum exosomes using ENSG00000258332.1, LINC00635, and serum AFP may serve as novel and valuable biomarker for HCC. Clinically, exosomal ATB expression is upregulated in HCC, while exosomal HEIH and RP11-513I15.6 have shown potential for distinguishing HCC related to HCV infection. CONCLUSION The lack of reliable biomarkers for liver diseases, coupled with the high specificity and sensitivity of exosomal lncRNA and its non-invasive detection, promotes exploring their role in pathogenesis and biomarker for diagnosis, prognosis, and response to treatment liver diseases.
Collapse
Affiliation(s)
- Mohammed Ismail
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
- Department of Pharmacology, Faculty of Medicine and Health Science, Dongola University, Dongola, Sudan
| | - Missaa M Fadul
- Department of Pharmacology, Faculty of Medicine and Health Science, Dongola University, Dongola, Sudan
| | - Reham Taha
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Orwa Siddig
- Department of Pharmaceutical Analysis, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Muhanad Elhafiz
- Department of Pharmacology, Faculty of Pharmacy, Omdurman Islamic University, Khartoum, Sudan
| | - Bashir A Yousef
- Department of Pharmacology, Faculty of Pharmacy, University of Khartoum, Khartoum, Sudan
| | - Zhenzhou Jiang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China
| | - Luyong Zhang
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
- Centre for Drug Research and Development, Guangdong Pharmaceutical University, Guangzhou, 510006, China.
| | - Lixin Sun
- Jiangsu Center for Pharmacodynamics Research and Evaluation, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
32
|
Li X, Wu Y, Jin Y. Exosomal LncRNAs and CircRNAs in lung cancer: Emerging regulators and potential therapeutic targets. Noncoding RNA Res 2024; 9:1069-1079. [PMID: 39022675 PMCID: PMC11254510 DOI: 10.1016/j.ncrna.2024.06.010] [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: 03/28/2024] [Revised: 06/11/2024] [Accepted: 06/11/2024] [Indexed: 07/20/2024] Open
Abstract
Lung cancer remains one of the most prevalent and lethal malignancies globally, characterized by high incidence and mortality rates among all cancers. The delayed diagnosis of lung cancer at intermediate to advanced stages frequently leads to suboptimal treatment outcomes. To improve the management of this disease, it is imperative to identify new, highly sensitive prognostic and diagnostic biomarkers. Exosomes, extracellular vesicles with a lipid-bilayer structure and a size range of 30-150 nm, are pivotal in intercellular communication and play significant roles in lung cancer progression. Non-coding RNAs (ncRNAs), including long non-coding RNAs (lncRNAs) and circular RNAs (circRNAs), are highly prevalent within exosomes and play a crucial role in various pathophysiological processes mediated by these extracellular vesicles. Beyond their established functions in miRNA and protein sequestration, these ncRNAs are involved in regulating translation and interactions within exosomes. Numerous studies have highlighted the importance of exosomal lncRNAs and circRNAs in influencing epithelial-mesenchymal transition (EMT), angiogenesis, proliferation, invasion, migration, and metastasis in lung cancer. Due to their unique functional characteristics, these molecules are promising therapeutic targets and biomarkers for diagnosis and prognosis. This review provides a succinct summary of the formation of exosomal lncRNAs and circRNAs, clarifies their biological roles, and thoroughly explains the mechanisms by which they participate in the progression of lung cancer. Finally, we discuss the potential clinical applications and challenges associated with exosomal lncRNAs and circRNAs in lung cancer.
Collapse
Affiliation(s)
- Xia Li
- Center of Molecular Diagnostic, Northern Jiangsu People's Hospital of Jiangsu Province, Yangzhou, 225001, China
| | - Yunbing Wu
- Department of Medicine Laboratory, Northern Jiangsu People's Hospital of Jiangsu Province, Yangzhou, 225001, China
| | - Yue Jin
- Center of Molecular Diagnostic, Northern Jiangsu People's Hospital of Jiangsu Province, Yangzhou, 225001, China
| |
Collapse
|
33
|
Liang C, Wang M, Huang Y, Yam JWP, Zhang X, Zhang X. Recent Advances of Small Extracellular Vesicles for the Regulation and Function of Cancer-Associated Fibroblasts. Int J Mol Sci 2024; 25:12548. [PMID: 39684264 DOI: 10.3390/ijms252312548] [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: 10/16/2024] [Revised: 11/12/2024] [Accepted: 11/20/2024] [Indexed: 12/18/2024] Open
Abstract
Cancer-associated fibroblasts (CAFs) are a heterogeneous cell population in the tumor microenvironment (TME) that critically affect cancer progression. Small extracellular vesicles (sEVs) act as information messengers by transmitting a wide spectrum of biological molecules, including proteins, nucleic acids, and metabolites, from donor cells to recipient cells. Previous studies have demonstrated that CAFs play important roles in tumor progression by regulating tumor cell proliferation, metastasis, therapeutic resistance, and metabolism via sEVs. In turn, tumor-derived sEVs can also regulate the activation and phenotype switch of CAFs. The dynamic crosstalk between CAFs and cancer cells via sEVs could ultimately determine cancer progression. In this review, we summarized the recent advance of the biological roles and underlying mechanisms of sEVs in mediating CAF-tumor cell interaction and its impact on cancer progression. We also reviewed the clinical applications of tumor- and CAF-derived sEVs, which could identify novel potential targets and biomarkers for cancer diagnosis, therapy, and prognosis.
Collapse
Affiliation(s)
- Chengdong Liang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Maoye Wang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Yongli Huang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Judy Wai Ping Yam
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Xu Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
| | - Xiaoxin Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang 212013, China
- Department of Pathology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| |
Collapse
|
34
|
Yao Y, Qian R, Gao H, Dai Y, Shi Y, An P, Xin B, Liu Z, Zhang N, Wan Y, He Y, Hu X. LSD1 deficiency in breast cancer cells promotes the formation of pre-metastatic niches. NPJ Precis Oncol 2024; 8:260. [PMID: 39528717 PMCID: PMC11555121 DOI: 10.1038/s41698-024-00751-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Lysine-specific demethylase 1 (LSD1), a histone demethylating enzyme, plays a crucial role in cancer metastasis. Studies show LSD1 knockout promotes breast cancer lung metastasis, but it's unknown if it alters the lung microenvironment for metastasis. In this study, we investigated the effects of exosomes from LSD1-knockdown (LSD1 KD) breast cancer cells on pre-metastatic niche formation. Injecting exosomes from LSD1 KD cells in mice resulted in a substantial increase in lung colonization by breast cancer cells, while treatment with exosomes derived from LSD1 KD cells decreased the expression of the ZO-1 and occludin, leading to increased vascular permeability. The LSD1 KD reduced the expression of circDOCK1, which augmented the levels of miR-1270 in exosomes. And miR-1270 inhibited ZO-1 expression in human endothelial cells, which enhanced their permeability. Our study uncovered a novel mechanism in which the LSD1 promotes the formation of pre-metastatic niches via the regulation of exosomal miRNA.
Collapse
Affiliation(s)
- Yutong Yao
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Rui Qian
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Hanwei Gao
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Yonghao Dai
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Yueru Shi
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Peipei An
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Benkai Xin
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Ziyu Liu
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Nan Zhang
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Youzhong Wan
- Cancer Biology Laboratory, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Yuquan He
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China
| | - Xin Hu
- Department of Cardiology, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, Jilin, China.
| |
Collapse
|
35
|
Wang Y, Gao W, Feng B, Shen H, Chen X, Yu S. Surface protein analysis of breast cancer exosomes using visualized strategy on centrifugal disk chip. Int J Biol Macromol 2024; 280:135651. [PMID: 39278429 DOI: 10.1016/j.ijbiomac.2024.135651] [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: 07/30/2024] [Revised: 09/12/2024] [Accepted: 09/12/2024] [Indexed: 09/18/2024]
Abstract
Breast cancer, the most common cancer among women worldwide, lacks specific tumor markers for accurate diagnosis. Recent advances have highlighted tumor-derived exosomes as a promising non-invasive biomarker for cancer detection. Continuous monitoring of surface protein markers on exosomes in the blood could offer valuable insights for breast cancer diagnosis. However, integrating the isolation and detection of exosomes from whole blood is bulky, time-consuming, and requires professional operations. To address this difficulty, we developed a method of integrated centrifugal disk chip (CD chip) exosome enrichment directly from whole blood followed by a colorimetric visualization strategy for multiplex analysis. The disc consists of multi-chambers and multi-microchannels with immediate smartphone-enabled processing of colorimetric results. The combination of CEA + CA125 + EGFR on-chip detection could significantly differentiate the different stages of cancer in tumor-bearing mice and successfully distinguish between breast cancer patients and healthy individuals. Crucially, small volumes (100 μL) of blood samples were adequate. In addition, the chip was simple and fast, with results within 10 min, which provides immediate exosomal information through consecutive blood sampling, which could potentially result in a more timely and well-informed clinical breast cancer diagnosis.
Collapse
Affiliation(s)
- Yanlin Wang
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Wenjing Gao
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Bin Feng
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Hao Shen
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China
| | - Xueqin Chen
- Department of Traditional Medicine, The First Affiliated Hospital of Ningbo University, Ningbo, Zhejiang 315211, China
| | - Shaoning Yu
- Institute of Mass Spectrometry, School of Material Science and Chemical Engineering, Ningbo University, Ningbo, Zhejiang 315211, China.
| |
Collapse
|
36
|
Long C, Shi H, Li J, Chen L, Lv M, Tai W, Wang H, Xu Y. The diagnostic accuracy of urine-derived exosomes for bladder cancer: a systematic review and meta-analysis. World J Surg Oncol 2024; 22:285. [PMID: 39472962 PMCID: PMC11520875 DOI: 10.1186/s12957-024-03569-1] [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: 08/08/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024] Open
Abstract
INTRODUCTION Urine-derived exosomes could potentially be biomarkers for bladder cancer (BC) diagnosis. This study aimed to systematically evaluate the diagnostic worth of urine-derived exosomes in BC patients through a meta-analysis of diverse studies. METHODS A systematic search was carried out in PubMed, Web of Science, Embase, Cochrane, and CNKI databases to obtain the literature concerning the diagnosis of BC via urine-derived exosomes. A literature retrieval strategy was devised to pick articles and extract needed data from the literature. QUADS-2 was used to evaluate the quality of the included literatures, and the aggregated diagnostic effect was assessed by calculating the area under the aggregated SROC curve. All statistical analyses and plots were conducted with STATA 14.0 and RevMan5.3. RESULTS A total of 678 articles were retrieved by means of the search strategy of the online database. Through screening, 21 articles were obtained, involving 3348 participants and 77 studies. The meta-analysis of the results indicated that urinary exosomes had a combined sensitivity of 0.75, a specificity of 0.77, and a combined AUC of 0.83 for the diagnosis of BC, suggesting that urine-derived exosomes have a relatively satisfactory diagnostic effect in the detection of BC. Among the subgroups classified by biomarker, long non-coding RNAs (lncRNAs) had the highest comprehensive sensitivity (SEN = 0.78), and miRNAs had the highest comprehensive specificity (SPN = 0.81). In other subgroup analyses, the biomarker panel for multiple exosomes combined diagnosis demonstrated the best diagnostic efficacy, with a combined the area under the curve ( AUC) of 0.87. CONCLUSIONS As a novel biomarker, urine-derived exosomes have significant diagnostic prospects in the diagnosis of BC. Nevertheless, their application in clinical settings still demands a considerable number of clinical trials to confirm their clinical feasibility and practicability.
Collapse
Affiliation(s)
- Chunyue Long
- Department of Clinical Laboratory, the Second Affiliated Hospital of Kunming Medical University, Kunming , Yunnan, 650000, China
| | - Hongjin Shi
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming , Yunnan, 650000, China
| | - Jinyu Li
- Department of Clinical Laboratory, the Second Affiliated Hospital of Kunming Medical University, Kunming , Yunnan, 650000, China
| | - Lijian Chen
- Department of Clinical Laboratory, Fuqing People's Hospital, Fuqing , Fujian, 350300, China
| | - Mei Lv
- Department of Clinical Laboratory, the Second Affiliated Hospital of Kunming Medical University, Kunming , Yunnan, 650000, China
| | - Wenlin Tai
- Department of Clinical Laboratory, the Second Affiliated Hospital of Kunming Medical University, Kunming , Yunnan, 650000, China
| | - Haifeng Wang
- Department of Urology, the Second Affiliated Hospital of Kunming Medical University, Kunming , Yunnan, 650000, China.
| | - Yiheng Xu
- Department of Clinical Laboratory, the Second Affiliated Hospital of Kunming Medical University, Kunming , Yunnan, 650000, China.
| |
Collapse
|
37
|
Ye J, Li D, Jie Y, Luo H, Zhang W, Qiu C. Exosome-based nanoparticles and cancer immunotherapy. Biomed Pharmacother 2024; 179:117296. [PMID: 39167842 DOI: 10.1016/j.biopha.2024.117296] [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: 06/20/2024] [Revised: 08/06/2024] [Accepted: 08/09/2024] [Indexed: 08/23/2024] Open
Abstract
Over the past decades, cancer immunotherapy has encountered challenges such as immunogenicity, inefficiency, and cytotoxicity. Consequently, exosome-based cancer immunotherapy has gained rapid traction as a promising alternative. Exosomes, a type of extracellular vesicles (EVs) ranging from 50 to 150 nm, are self-originating and exhibit fewer side effects compared to traditional therapies. Exosome-based immunotherapy encompasses three significant areas: cancer vaccination, co-inhibitory checkpoints, and adoptive T-cell therapy. Each of these fields leverages the inherent advantages of exosomes, demonstrating substantial potential for individualized tumor therapy and precision medicine. This review aims to elucidate the reasons behind the promise of exosome-based nanoparticles as cancer therapies by examining their characteristics and summarizing the latest research advancements in cancer immunotherapy.
Collapse
Affiliation(s)
- Jiarong Ye
- Department of General Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang University, Jiangxi Province, 330000 China.
| | - Danni Li
- Second Clinical Medical School, Nanchang University, Jiangxi Province 330000, China
| | - Yiting Jie
- Second Clinical Medical School, Nanchang University, Jiangxi Province 330000, China
| | - Hongliang Luo
- Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, China
| | - Wenjun Zhang
- Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, Jiangxi Province 330000, China
| | - Cheng Qiu
- Gastrointestinal Surgery, Pingxiang People's Hospital, Jiangxi Province 330000, China.
| |
Collapse
|
38
|
Hejazian SM, Rahbar Saadat Y, Hosseiniyan Khatibi SM, Farnood F, Farzamikia N, Hejazian SS, Batoumchi S, Shoja MM, Zununi Vahed S, Ardalan M. Circular RNAs as novel biomarkers in glomerular diseases. Arch Physiol Biochem 2024; 130:568-580. [PMID: 37194131 DOI: 10.1080/13813455.2023.2212328] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 05/03/2023] [Accepted: 05/03/2023] [Indexed: 05/18/2023]
Abstract
Circular RNAs (circRNAs) regulate gene expression and biological procedures by controlling target genes or downstream pathways by sponging their related miRNA (s). Three types of circRNAs have been identified; exonic circRNAs (ecircRNAs), intronic RNAs (ciRNAs), and exon-intron circRNAs (ElciRNAs). It is clarified that altered levels of circRNAs have dynamic pathological and physiological functions in kidney diseases. Evidence suggests that circRNAs can be considered novel diagnostic biomarkers and therapeutic targets for renal diseases. Glomerulonephritis (GN) is a general term used to refer to a wide range of glomerular diseases. GN is an important cause of chronic kidney diseases. Here, we review the biogenesis of circRNAs, and their molecular and physiological functions in the kidney. Moreover, the dysregulated expression of circRNAs and their biological functions are discussed in primary and secondary glomerulonephritis. Moreover, diagnostic and therapeutic values of circRNAs in distinguishing or treating different types of GN are highlighted.
Collapse
Affiliation(s)
| | | | | | - Farahnoosh Farnood
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Negin Farzamikia
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Seyyed Sina Hejazian
- Neurosciences Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Batoumchi
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammadali M Shoja
- College of Allopathic Medicine, Nova Southeastern University, Fort Lauderdale, FL, USA
| | | | | |
Collapse
|
39
|
Torres Quintas S, Canha-Borges A, Oliveira MJ, Sarmento B, Castro F. Special Issue: Nanotherapeutics in Women's Health Emerging Nanotechnologies for Triple-Negative Breast Cancer Treatment. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2300666. [PMID: 36978237 DOI: 10.1002/smll.202300666] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Revised: 03/03/2023] [Indexed: 06/18/2023]
Abstract
Breast cancer appears as the major cause of cancer-related deaths in women, with more than 2 260 000 cases reported worldwide in 2020, resulting in 684 996 deaths. Triple-negative breast cancer (TNBC), characterized by the absence of estrogen, progesterone, and human epidermal growth factor type 2 receptors, represents ≈20% of all breast cancers. TNBC has a highly aggressive clinical course and is more prevalent in younger women. The standard therapy for advanced TNBC is chemotherapy, but responses are often short-lived, with high rate of relapse. The lack of therapeutic targets and the limited therapeutic options confer to individuals suffering from TNBC the poorest prognosis among breast cancer patients, remaining a major clinical challenge. In recent years, advances in cancer nanomedicine provided innovative therapeutic options, as nanoformulations play an important role in overcoming the shortcomings left by conventional therapies: payload degradation and its low solubility, stability, and circulating half-life, and difficulties regarding biodistribution due to physiological and biological barriers. In this integrative review, the recent advances in the nanomedicine field for TNBC treatment, including the novel nanoparticle-, exosome-, and hybrid-based therapeutic formulations are summarized and their drawbacks and challenges are discussed for future clinical applications.
Collapse
Affiliation(s)
- Sofia Torres Quintas
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ana Canha-Borges
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Maria José Oliveira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Rua Jorge de Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- IUCS-CESPU - Instituto de Investigação e Formação Avançada em Ciências e Tecnologias da Saúde, Rua Central de Gandra 1317, 4585-116, Gandra, Portugal
| | - Flávia Castro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
| |
Collapse
|
40
|
Bitiņa-Barlote Ē, Plonis J, Andrejeva M, Vjaters E, Gardovskis J, Daneberga Z, Miklaševičs E, Nakazawa-Miklaševiča M. Profiles of urinal exosomal miRNAs derived from bladder cancer. Cent European J Urol 2024; 77:361-374. [PMID: 40115475 PMCID: PMC11921953 DOI: 10.5173/ceju.2023.279.r1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Accepted: 06/21/2024] [Indexed: 03/23/2025] Open
Abstract
Introduction Exosomes contain nucleic acids and proteins inside of them. These are suggested as cell-cell communication materials and it is considered that they can modulate the status of other cells. Material and methods To understand the bladder cancer (BC) related exosomal microRNAs (miRNAs), we compared the 752 urine exosomal miRNAs in healthy control (n = 7), low grade (LG) BC (n = 6) and high grade (HG) BC (n = 6) by RT-qPCR. Results The differential expressing (DE) urine exosomal miRNAs (2 > fold regulation) were 96 and 78 in LG and HG, respectively. Our exosomal miRNAs profiles cover many miRNAs which have been reported in BC patients' tissues and other biofluids. Most DE exosomal miRNAs were up-regulated in the profiles. Seven up-regulated exosomal miRNAs in the LG group (miR-28-5p, miR-16-5p, miR-28-3p, miR-24-3p, miR-25-3p, miR-19b-3p and miR10b-5p) and 3 miRNAs in the HG group (miR-150-5p, miR-28-5p and miR28-3p) were found as directly TP53 targeting. Twenty-two and 18 PTEN targeting miRNAs were observed in up-regulated miRNAs of LG and HG. The target genes of these exosomal miRNAs and their interaction network predicted that the TP53 is the strongest hub gene in both BC groups exosomal miRNA networks. Several DE miRNAs were found that could potentially be used as biomarkers for the diagnosis of BC. Conclusions Profiles of urinal exosomal miRNAs derived from BC manifested potentially epigenetic regulation of the TP53 and PTEN genes as compared to other oncogenes and tumour suppressors.
Collapse
Affiliation(s)
- Ērika Bitiņa-Barlote
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
- Pauls Stradiņš Clinical University Hospital, Centre of Urology, Riga, Latvia
| | - Juris Plonis
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
- Pauls Stradiņš Clinical University Hospital, Centre of Urology, Riga, Latvia
| | - Margarita Andrejeva
- Center of Oncology, Pauls Stradiņš Clinical University Hospital, Riga, Latvia
| | - Egils Vjaters
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
- Pauls Stradiņš Clinical University Hospital, Centre of Urology, Riga, Latvia
| | - Jānis Gardovskis
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
| | - Zanda Daneberga
- Institute of Oncology and Molecular Genetics, Rīga Stradiņš University, Latvia
| | - Edvīns Miklaševičs
- Department of Biology and Microbiology, Rīga Stradiņš University, Latvia
| | | |
Collapse
|
41
|
Kang HJ, Yun S, Shin SH, Youn DH, Son GH, Lee JJ, Hong JY. Tuberculous Pleural Effusion-Derived Exosomal miR-130b-3p and miR-423-5p Promote the Proliferation of Lung Cancer Cells via Cyclin D1. Int J Mol Sci 2024; 25:10119. [PMID: 39337604 PMCID: PMC11431986 DOI: 10.3390/ijms251810119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 09/08/2024] [Accepted: 09/16/2024] [Indexed: 09/30/2024] Open
Abstract
Epidemiologic studies have shown an association between tuberculosis and lung cancer. The altered tumor microenvironment after tuberculosis infection appears to contribute to cancer progression. Pleural effusions are enriched in exosomes, which act as mediators of intercellular communication. We hypothesized that tuberculous pleural effusion (TPE)-derived exosomes mediate intercellular communication. Then, we examined the interaction between TPE-derived exosomes and cancer cells. Exosomal miRNA profiling of TPE was performed using a microRNA array. An in vitro lung cancer cell experiment and an in vivo mouse xenograft tumor model were used to evaluate the effects of the selected exosomal microRNAs. TPE-derived exosome treatment enhanced the growth of A549 cells both in vitro and in a nude mouse xenograft model. Neighboring cancer cells were observed to take up TPE-derived exosomes, which promoted cancer cell invasion. Exosome-mediated transfer of the selected microRNAs, including miR-130b-3p and miR-423-5p, to A549 lung cancer cells activated cyclin D1 signaling and increased the expression of phosphorylated p65, a cyclin D1 transcription factor. Inhibitors of miR-130b and miR-423-5p suppressed the promotion of lung cancer by TPE-derived exosomes and reduced the expression of p65 and cyclin D1. These results suggest that TPE-derived exosomal miRNAs can serve as a novel therapeutic target in tuberculous fibrosis-induced lung cancer.
Collapse
Affiliation(s)
- Hyun-Jung Kang
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (H.-J.K.); (S.Y.); (S.-H.S.); (D.H.Y.); (G.-H.S.)
| | - Sangho Yun
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (H.-J.K.); (S.Y.); (S.-H.S.); (D.H.Y.); (G.-H.S.)
| | - Seung-Ho Shin
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (H.-J.K.); (S.Y.); (S.-H.S.); (D.H.Y.); (G.-H.S.)
| | - Dong Hyuk Youn
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (H.-J.K.); (S.Y.); (S.-H.S.); (D.H.Y.); (G.-H.S.)
| | - Ga-Hyun Son
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (H.-J.K.); (S.Y.); (S.-H.S.); (D.H.Y.); (G.-H.S.)
| | - Jae Jun Lee
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (H.-J.K.); (S.Y.); (S.-H.S.); (D.H.Y.); (G.-H.S.)
| | - Ji Young Hong
- Institute of New Frontier Research Team, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea; (H.-J.K.); (S.Y.); (S.-H.S.); (D.H.Y.); (G.-H.S.)
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Chuncheon Sacred Heart Hospital, Hallym University Medical Center, Chuncheon 24252, Republic of Korea
| |
Collapse
|
42
|
Wang B, Mao J, Wang L, Zhao Y, Wang B, Yang H. Exosome-mediated transfer of lncRNA RP3-340B19.3 promotes the progression of breast cancer by sponging miR-4510/MORC4 axis. Cancer Cell Int 2024; 24:312. [PMID: 39256868 PMCID: PMC11389435 DOI: 10.1186/s12935-024-03490-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Accepted: 08/24/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND This study aims to explore the molecular mechanism of lncRNA RP3-340B19.3 on breast cancer cell proliferation and metastasis and clinical significance of lncRNA RP3-340B19.3 for breast cancer. METHODS The subcellular localization of lncRNA RP3-340B19.3 was identified using RNA fluorescence in situ hybridization (FISH). The expression of lncRNA RP3-340B19.3 in breast cancer cells, breast cancer tissues, as well as the serum and serum exosomes of breast cancer patients, was measured through quantitative RT-PCR. In the in vitro setting, we conducted experiments to observe the effects of RP3-340B19.3 on both cell migration and proliferation. This was achieved through the utilization of transwell migration assays as well as clone formation assays. Meanwhile, transwell migration assays and clone formation assays were used to observe the effects of MDA-MB-231-exosomes enriched in RP3-340B19.3 on breast cancer microenvironment cells MCF7 and BMMSCs. Additionally, western blotting techniques were used to assess the expression levels of proteins associated with essential cellular processes such as proliferation, apoptosis, and metastasis. In vivo, the impact of RP3-340B19.3 knockdown on tumour weight and volume was observed within a nude mice model. We aimed to delve into the intricate molecular mechanisms involving RP3-340B19.3 by using bioinformatics analysis, dual luciferase reporter gene experiments and western blotting. Moreover, the potential correlations between RP3-340B19.3 expression and various clinical pathological characteristics were analyzed. RESULTS Our investigation revealed that RP3-340B19.3 was expressed in both the cytoplasm and nucleus, with a noteworthy increase in breast cancer cells. Notably, we found that RP3-340B19.3 exerted a promoting influence on the proliferation and migration of breast cancer cells, both in vitro and in vivo. MDA-MB-231-exosomes enriched in RP3-340B19.3 promoted the proliferation and migration of MCF7 and BMMSCs in vitro. Mechanistically, RP3-340B19.3 demonstrated the capability to modulate the expression of MORC4 by forming a complex with miR-4510. This interaction subsequently triggered the activation of the NF-κB and Wnt-β-catenin signaling pathways. Furthermore, our study highlighted the potential diagnostic utility of RP3-340B19.3. We discovered its presence in the serum and exosomes of breast cancer patients, showing promising efficacy as a diagnostic marker. Notably, the diagnostic potential of RP3-340B19.3 was particularly significant in relation to distinguishing between different pathological types of breast cancer and correlating with tumour diameter. CONCLUSION Our findings establish that RP3-340B19.3 plays a pivotal role in driving the proliferation and metastasis of breast cancer. Additionally, exosomes enriched in RP3-340B19.3 could influence MCF7 and BMMSCs in tumour microenvironment, promoting the progression of breast cancer. This discovery positions RP3-340B19.3 as a prospective novel candidate for a tumour marker, offering substantial potential in the realms of breast cancer diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Bo Wang
- Department of Oncology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jiahui Mao
- Department of Central Laboratory, The Affiliated Hospital of Jiangsu University, Zhenjiang, China
| | - Linxia Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuexin Zhao
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Bingying Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Huan Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
43
|
Zhongyu X, Wei X, Hongmei Z, Xiaodong G, Xiaojing Y, Yuanpei L, Li Z, Zhenmin F, Jianda X. Review of pre-metastatic niches induced by osteosarcoma-derived extracellular vesicles in lung metastasis: A potential opportunity for diagnosis and intervention. Biomed Pharmacother 2024; 178:117203. [PMID: 39067163 DOI: 10.1016/j.biopha.2024.117203] [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: 05/30/2024] [Revised: 07/17/2024] [Accepted: 07/22/2024] [Indexed: 07/30/2024] Open
Abstract
Osteosarcoma (OS) has a high propensity for lung metastasis, which is the leading cause of OS-related death and treatment failure. Intercellular communication between OS cells and distant lung host cells is required for the successful lung metastasis of OS cells to the lung. Before OS cells infiltrate the lung, in situ OS cells secrete extracellular vesicles (EVs) that act as mediators of cell-to-cell communication. In recent years, EVs have been confirmed to act as bridges and key drivers between in situ tumors and metastatic lesions by regulating the formation of a pre-metastatic niche (PMN), defined as a microenvironment suitable for disseminated tumor cell engraftment and colonization, in distant target organs. This review summarizes the current knowledge about the underlying mechanisms of PMN formation induced by OS-derived EVs and the potential roles of EVs as targets or drug carriers in regulating PMN formation in the lung. We also provide an overview of their potential EV-based therapeutic strategies for hindering PMN formation in the context of OS lung metastasis.
Collapse
Affiliation(s)
- Xia Zhongyu
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China
| | - Xu Wei
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, Jiangsu 224051, China
| | - Zhang Hongmei
- School of Mechanical Engineering, Jiangsu University, Zhenjiang 212013, China
| | - Ge Xiaodong
- College of Marine and Bioengineering, Yancheng Institute of Technology, Yancheng, Jiangsu 224051, China
| | - Yan Xiaojing
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China
| | - Lian Yuanpei
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China
| | - Zhu Li
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China
| | - Fan Zhenmin
- School of Mechanical Engineering, Jiangsu University of Technology, Changzhou Jiangsu, China.
| | - Xu Jianda
- Department of Orthopaedics, Changzhou hospital affiliated to Nanjing University of Chinese Medicine, 25 North Heping Road, Changzhou, Jiangsu Province 213003, China.
| |
Collapse
|
44
|
Liu Y, Lu X, Sun S, Yu H, Li H. The therapeutic use of exosomes in children with adenoid hypertrophy accompanied by otitis media with effusion (AHOME): a protocol study. BMC Pediatr 2024; 24:521. [PMID: 39134977 PMCID: PMC11318249 DOI: 10.1186/s12887-024-04999-2] [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: 07/28/2023] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
BACKGROUND The adenoids act as a reservoir of bacterial pathogens and immune molecules, and they are significantly involved in children with otitis media with effusion (OME). As an essential carrier of intercellular substance transfer and signal transduction, exosomes with different biological functions can be secreted by various types of cells. There remains significant uncertainty regarding the clinical relevance of exosomes to OME, especially in its pathophysiologic development. In this study, we will seek to determine the biological functions of exosomes in children with adenoid hypertrophy accompanied by OME (AHOME). METHODS The diagnostic criteria for OME in children aged 4-10 years include a disease duration of at least 3 months, type B or C acoustic immittance, and varying degrees of conductive hearing loss. Adenoidal hypertrophy is diagnosed when nasal endoscopy shows at least 60% adenoidal occlusion in the nostrils or when nasopharyngeal lateral X-ray shows A/N > 0.6. Children who meet the indications for adenoidectomy surgery undergo adenoidectomy. Peripheral blood, nasopharyngeal swab, and adenoid tissue will be collected from patients, and the exosomes will be isolated from the samples. Following the initial collection, patients will undergo adenoidectomy and peripheral blood and nasopharyngeal swabs will be collected again after 3 months. EXPECTED RESULTS This study aims to identify differences in exosomes from preoperative adenoid tissue and peripheral blood samples between children with AHOME and those with adenoid hypertrophy alone. Additionally, it seeks to determine changes in microbial diversity in adenoid tissue between these groups. CONCLUSIONS The findings are expected to provide new insights into the diagnosis and treatment of OME, to identify novel biomarkers, and to enhance our understanding of the pathophysiology of OME, potentially leading to the development of innovative diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Yixuan Liu
- Department of ENT Institute and Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine Research, Demin Han's Academician Workstation, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200032, People's Republic of China
| | - Xiaoling Lu
- Department of ENT Institute and Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine Research, Demin Han's Academician Workstation, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200032, People's Republic of China
| | - Shan Sun
- Department of ENT Institute and Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine Research, Demin Han's Academician Workstation, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200032, People's Republic of China.
| | - Huiqian Yu
- Department of ENT Institute and Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine Research, Demin Han's Academician Workstation, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200032, People's Republic of China.
| | - Huawei Li
- Department of ENT Institute and Otorhinolaryngology, NHC Key Laboratory of Hearing Medicine Research, Demin Han's Academician Workstation, Eye & ENT Hospital, Fudan University, 83 Fenyang Road, Shanghai, 200032, People's Republic of China.
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, People's Republic of China.
- The Institutes of Brain Science and the Collaborative Innovation Center for Brain Science, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
45
|
Liao H, Zhang C, Wang F, Jin F, Zhao Q, Wang X, Wang S, Gao J. Tumor-derived extracellular vesicle proteins as new biomarkers and targets in precision oncology. J Mol Med (Berl) 2024; 102:961-971. [PMID: 38814362 PMCID: PMC11269371 DOI: 10.1007/s00109-024-02452-6] [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: 11/15/2023] [Revised: 04/09/2024] [Accepted: 04/25/2024] [Indexed: 05/31/2024]
Abstract
Extracellular vesicles (EVs) are important carriers of signaling molecules, such as nucleic acids, proteins, and lipids, and have become a focus of increasing interest due to their numerous physiological and pathological functions. For a long time, most studies on EV components focused on noncoding RNAs; however, in recent years, extracellular vesicle proteins (EVPs) have been found to play important roles in diagnosis, treatment, and drug resistance and thus have been considered favorable biomarkers and therapeutic targets for various tumors. In this review, we describe the general protocols of research on EVPs and summarize their multifaceted roles in precision medicine applications, including cancer diagnosis, dynamic monitoring of therapeutic efficacy, drug resistance research, tumor microenvironment interaction research, and anticancer drug delivery.
Collapse
Affiliation(s)
- Haiyan Liao
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Cheng Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Fen Wang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Feng Jin
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China
| | - Qiqi Zhao
- Chi Biotech Co., Ltd., Shenzhen, China
| | | | - Shubin Wang
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
| | - Jing Gao
- Department of Oncology, Shenzhen Key Laboratory of Gastrointestinal Cancer Translational Research, Cancer Institute, Peking University Shenzhen Hospital, Shenzhen-Peking University-Hong Kong University of Science and Technology Medical Center, Shenzhen, China.
| |
Collapse
|
46
|
Yu J, Yu C, Jiang K, Yang G, Yang S, Tan S, Li T, Liang H, He Q, Wei F, Li Y, Cheng J, Wang F. Unveiling potential: urinary exosomal mRNAs as non-invasive biomarkers for early prostate cancer diagnosis. BMC Urol 2024; 24:163. [PMID: 39090720 PMCID: PMC11292860 DOI: 10.1186/s12894-024-01540-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 07/09/2024] [Indexed: 08/04/2024] Open
Abstract
BACKGROUND This study investigated the use of urinary exosomal mRNA as a potential biomarker for the early detection of prostate cancer (PCa). METHODS Next-generation sequencing was utilized to analyze exosomal RNA from 10 individuals with confirmed PCa and 10 individuals without cancer. Subsequent validation through qRT-PCR in a larger sample of 43 PCa patients and 92 healthy controls revealed distinct mRNA signatures associated with PCa. RESULTS Notably, mRNAs for RAB5B, WWP1, HIST2H2BF, ZFY, MARK2, PASK, RBM10, and NRSN2 showed promise as diagnostic markers, with AUC values between 0.799 and 0.906 and significance p values. Combining RAB5B and WWP1 in an exoRNA diagnostic model outperformed traditional PSA tests, achieving an AUC of 0.923, 81.4% sensitivity, and 89.1% specificity. CONCLUSIONS These findings highlight the potential of urinary exosomal mRNA profiling, particularly focusing on RAB5B and WWP1, as a valuable strategy for improving the early detection of PCa.
Collapse
Affiliation(s)
- Jiayin Yu
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Chifei Yu
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, No.71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China
| | - Kangxian Jiang
- Department of Urology, The Second Affiliated Hospital of Fujian Medical University, No. 34 Zhongshan North Road, Quanzhou, Fujian, 362000, P.R. China
| | - Guanglin Yang
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, No.71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China
| | - Shubo Yang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Shuting Tan
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Tingting Li
- Department of Urology, Affiliated Tumor Hospital of Guangxi Medical University, No.71 Hedi Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China
| | - Haiqi Liang
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Qihuan He
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Faye Wei
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Yujian Li
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China
| | - Jiwen Cheng
- Department of Urology, The First Affiliated Hospital of Guangxi Medical University, No.6 Shuangyong Road, Qingxiu, Nanning, Guangxi, 530021, P.R. China.
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, No.22 Shuangyong Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China.
| | - Fubo Wang
- Center for Genomic and Personalized Medicine, Guangxi Key Laboratory for Genomic and Personalized Medicine, Guangxi Medical University, No.22 Shuangyong Road, Qingxiu District, Nanning, Guangxi, 530021, P.R. China.
| |
Collapse
|
47
|
Chen X, Li R, Yin YH, Liu X, Zhou XJ, Qu YQ. Pan-cancer prognosis, immune infiltration, and drug resistance characterization of lung squamous cell carcinoma tumor microenvironment-related genes. Biochem Biophys Rep 2024; 38:101722. [PMID: 38711549 PMCID: PMC11070325 DOI: 10.1016/j.bbrep.2024.101722] [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: 11/16/2023] [Revised: 04/21/2024] [Accepted: 04/24/2024] [Indexed: 05/08/2024] Open
Abstract
Background The tumor microenvironment (TME) plays an important role in cancer development; however, its implications in lung squamous cell carcinoma (LUSC) and pan-cancer have been poorly understood. Methods In this study, The Cancer Genome Atlas (TCGA) and Estimation of Stromal and Immune cells in Malignant Tumor tissue using Expression Data (ESTIMATE) datasets were applied to identify differentially expressed genes. Additionally, online public databases were utilized for in-depth bioinformatics analysis of pan-cancer datasets to investigate the prognostic implications of TME-related genes further. Results Our study demonstrated a significant association between stromal scores, immune scores, and specific clinical characteristics in LUSC patients. C3AR1, CSF1R, CCL2, CCR1, and CD14 were identified as prognostic genes related to the TME. All TME-related prognostic genes demonstrated varying degrees of correlation with immune infiltration subtypes and tumor cell stemness. Moreover, our study revealed that TME-related prognostic genes, particularly C3AR1 and CCR1, might contribute to drug resistance in cancer cells. Conclusions The identified TME-related prognostic genes, particularly C3AR1 and CCR1, have potential implications for understanding and targeting drug resistance mechanisms in cancer cells.
Collapse
Affiliation(s)
- Xiao Chen
- Department of Respiratory Medicine, Tai'an City Central Hospital, Tai'an, China
| | - Rui Li
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yun-Hong Yin
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xiao Liu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Xi-Jia Zhou
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| | - Yi-Qing Qu
- Department of Pulmonary and Critical Care Medicine, Qilu Hospital of Shandong University, Jinan, China
| |
Collapse
|
48
|
Wang H, Guo Y, Jiang Y, Ge Y, Wang H, Shi D, Zhang G, Zhao J, Kang Y, Wang L. Exosome-loaded biomaterials for tendon/ligament repair. BIOMATERIALS TRANSLATIONAL 2024; 5:129-143. [PMID: 39351162 PMCID: PMC11438604 DOI: 10.12336/biomatertransl.2024.02.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 04/30/2024] [Accepted: 06/18/2024] [Indexed: 10/04/2024]
Abstract
Exosomes, a specialised type of extracellular vesicle, have attracted significant attention in the realm of tendon/ligament repair as a potential biologic therapeutic tool. While the competence of key substances responsible for the delivery function was gradually elucidated, series of shortcomings exemplified by the limited stability still need to be improved. Therefore, how to take maximum advantage of the biological characteristics of exosomes is of great importance. Recently, the comprehensive exploration and application of biomedical engineering has improved the availability of exosomes and revealed the future direction of exosomes combined with biomaterials. This review delves into the present application of biomaterials such as nanomaterials, hydrogels, and electrospun scaffolds, serving as the carriers of exosomes in tendon/ligament repair. By pinpointing and exploring their strengths and limitations, it offers valuable insights, paving the way the future direction of biomaterials in the application of exosomes in tendon/ligament repair in this field.
Collapse
Affiliation(s)
- Haohan Wang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yonglin Guo
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yiwen Jiang
- School of Materials Science and Engineering, East China University of Science and Technology, Shanghai, China
| | - Yingyu Ge
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hanyi Wang
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dingyi Shi
- Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Guoyang Zhang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jinzhong Zhao
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yuhao Kang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Liren Wang
- Department of Orthopaedics, Shanghai Sixth People’s Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
49
|
Li M, Tai Q, Shen S, Gao M, Zhang X. Biomimetic Exosome-Sheathed Magnetic Mesoporous Anchor with Modification of Glucose Oxidase for Synergistic Targeting and Starving Tumor Cells. ACS APPLIED MATERIALS & INTERFACES 2024; 16:29634-29644. [PMID: 38822821 DOI: 10.1021/acsami.4c02337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2024]
Abstract
Efficient protection and precise delivery of biomolecules are of critical importance in the intervention and therapy of various diseases. Although diverse specific marker-functionalized drug carriers have been developed rapidly, current approaches still encounter substantial challenges, including strong immunogenicity, limited target availability, and potential side effects. Herein, we developed a biomimetic exosome-sheathed magnetic mesoporous anchor modified with glucose oxidase (MNPs@mSiO2-GOx@EM) to address these challenges and achieve synergistic targeting and starving of tumor cells. The MNPs@mSiO2-GOx@EM anchor integrated the unique characteristics of different components. An external decoration of exosome membrane (EM) with high biocompatibility contributed to increased phagocytosis prevention, prolonged circulation, and enhanced recognition and cellular uptake of loaded particles. An internal coated magnetic mesoporous core with rapid responsiveness by the magnetic field guidance and large surface area facilitated the enrichment of nanoparticles at the specific site and provided enough space for modification of glucose oxidase (GOx). The inclusion of GOx in the middle layer accelerated the energy-depletion process within cells, ultimately leading to the starvation and death of target cells with minimal side effects. With these merits, in vitro study manifested that our nanoplatform not only demonstrated an excellent targeting capability of 94.37% ± 1.3% toward homotypic cells but also revealed a remarkably high catalytical ability and cytotoxicity on tumor cells. Assisted by the magnetic guidance, the utilization of our anchor obviously inhibits the tumor growth in vivo. Together, our study is promising to serve as a versatile method for the highly efficient delivery of various target biomolecules to intended locations due to the fungibility of exosome membranes and provide a potential route for the recognition and starvation of tumor cells.
Collapse
Affiliation(s)
- Mengran Li
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Qunfei Tai
- Department of Chemistry, Fudan University, Shanghai 200433, China
| | - Shun Shen
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Mingxia Gao
- Department of Chemistry, Fudan University, Shanghai 200433, China
- Pharmacy Department, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Xiangmin Zhang
- Department of Chemistry, Fudan University, Shanghai 200433, China
| |
Collapse
|
50
|
Ghosh S, Mahajan AA, Dey A, Rajendran RL, Chowdhury A, Sen S, Paul S, Majhi S, Hong CM, Gangadaran P, Ahn BC, Krishnan A. Exosomes in Bone Cancer: Unveiling their Vital Role in Diagnosis, Prognosis, and Therapeutic Advancements. J Cancer 2024; 15:4128-4142. [PMID: 38947401 PMCID: PMC11212077 DOI: 10.7150/jca.95709] [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: 02/26/2024] [Accepted: 05/19/2024] [Indexed: 07/02/2024] Open
Abstract
Bone cancer among adolescents and children exhibits varying survival outcomes based on disease state. While localized bone cancer cases have a survival rate exceeding 70%, metastatic, refractory, and recurrent forms are associated with significantly poorer prognoses. Initially believed to be mere vehicles for cellular waste disposal, exosomes are now recognized as extracellular vesicles facilitating intercellular communication. These vesicles influence cellular behaviors by transporting various biomolecules, such as proteins, DNA, RNA, and lipids, among cells. The role of exosomes in regulating the progression of bone cancer is increasingly evident, impacting critical processes like tumorigenesis, proliferation, metastasis, angiogenesis, immune evasion, and drug resistance. Current research underscores the substantial potential of exosomes in promoting the progression and development of bone cancer. This review delves into the complex process of exosome biogenesis, the variety of cell-derived exosome sources, and their applications in drug delivery and therapeutics. It also examines ongoing clinical trials focused on exosome cargo levels and discusses the challenges and future directions in exosome research. Unlike costly and invasive traditional diagnostic methods, exosomal biomarkers offer a non-invasive, cost-effective, and readily accessible routine screening through simple fluid collection that aims to inspire researchers to investigate the potential of exosomes for cancer theragnostic. Through comprehensive exploration of these areas, the review seeks to enhance understanding and foster innovative solutions to cancer biology in the near future.
Collapse
Affiliation(s)
- Subhrojyoti Ghosh
- Department of Biotechnology, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India
| | - Atharva Anand Mahajan
- Advanced Centre for Treatment, Research, and Education in Cancer, Tata Memorial Centre, Mumbai, Maharashtra 410210, India
| | - Anuvab Dey
- Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, North Guwahati, Assam 781039, India
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
| | - Ankita Chowdhury
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi 110016, India
| | - Sushmita Sen
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi 110016, India
| | - Subhobrata Paul
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi 110016, India
| | - Sourav Majhi
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology, Delhi 110016, India
| | - Chae Moon Hong
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
| | - Prakash Gangadaran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Byeong-Cheol Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu 41944, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu 41944, Republic of Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu 41944, Korea
| | - Anand Krishnan
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein, 9300, South Africa
| |
Collapse
|