51
|
Li G, Chen W, Jiang K, Huang J, Zhong J, Liu X, Wei T, Gong R, Li Z, Zhu J, Shi H, Lei J. Exosome-mediated Delivery of miR-519e-5p Promotes Malignant Tumor Phenotype and CD8+ T-Cell Exhaustion in Metastatic PTC. J Clin Endocrinol Metab 2024; 109:1601-1617. [PMID: 38078691 DOI: 10.1210/clinem/dgad725] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Indexed: 05/18/2024]
Abstract
CONTEXT Distant metastases are the primary cause of therapy failure and mortality in patients with papillary thyroid carcinomas (PTCs). However, the underlying mechanism responsible for the initiation of tumor cell dissemination and metastasis in PTCs has rarely been investigated. OBJECTIVE The aim of this study was to investigate effects and underlying molecular mechanisms of circulating exosomal microRNAs (miRNAs) in distant metastatic PTCs. METHODS The most relevant circulating exosomal miRNA to distant metastatic PTCs were verified between distant metastatic PTCs and nondistant metastatic PTCs by miRNA microarray, quantitative real-time polymerase chain reaction (qRT-PCR) assays and receiver operating characteristic (ROC) curves. The parental and recipient cells of that circulating exosomal miRNA were then explored. In vitro and in vivo experiments were further performed to elucidate the function and potential mechanisms of circulating exosomal miRNAs that contribute to the development of distant metastases. RESULTS We determined that PTC-derived exosomal miR-519e-5p was significantly upregulated in the circulatory system in distant metastatic PTCs. Further tests demonstrated that PTC cells can acquire a more malignant phenotype via hnRNPA2B1-mediated sorting of tumor suppressor miR-519e-5p into exosomes to activate Wnt signaling pathway via upregulating PLAGL2. Furthermore, miR-519e-5p included in PTC-derived exosomes can be transferred to recipient CD8+ T cells and aid in tumor immune escape in distant organs through inhibiting Notch signaling pathway by downregulating NOTCH2. CONCLUSION Our findings highlight the dual role of PTC-derived exosomal miR-519e-5p in distant metastasis, which may improve our understanding of exosome-mediated distant metastatic mechanisms.
Collapse
Affiliation(s)
- Genpeng Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenjie Chen
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ke Jiang
- Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, 510060, China
| | - Jing Huang
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jinjing Zhong
- Department of Pathology, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Xiaowei Liu
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Tao Wei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Rixiang Gong
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Zhihui Li
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jingqiang Zhu
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Hubing Shi
- State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Jianyong Lei
- Division of Thyroid Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| |
Collapse
|
52
|
Li X, Yu D, Chen X, Huang Z, Zhao Y. A strategy for oral delivery of FGF21 for mitigating inflammation and multi-organ damage in sepsis. Int J Pharm 2024; 656:124115. [PMID: 38614430 DOI: 10.1016/j.ijpharm.2024.124115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 04/01/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024]
Abstract
Fibroblast growth factor 21 (FGF21) shows great therapeutic potential in metabolic, neurodegenerative and inflammatory diseases. However, current FGF21 administration predominantly relies on injection rather than oral ingestion due to its limited stability and activity post-gastrointestinal transit, thereby hindering its clinical utility. Milk-derived exosomes (mEx) have emerged as a promising vehicle for oral drug delivery due to their ability to maintain structural integrity in the gastrointestinal milieu. To address the challenge associated with oral delivery of FGF21, we encapsulated FGF21 within mEx (mEx@FGF21) to protect its activity post-oral administration. Additionally, we modified the surface of mEx@FGF21 by introducing transferrin (TF) to enhance intestinal absorption and transport, designated TF-mEx@FGF21. In vitro results demonstrated that the surface modification of TF promoted FGF21 internalization by intestinal epithelial cells. Orally administered TF-mEx@FGF21 showed promising therapeutic effects in septic mice. This study represents a practicable strategy for advancing the clinical application of oral FGF21 delivery.
Collapse
Affiliation(s)
- Xinze Li
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Department of Emergency, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, China
| | - Dedong Yu
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Xuanhe Chen
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China
| | - Zhiwei Huang
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju 61186, Republic of Korea.
| | - Yingzheng Zhao
- Department of Pharmaceutics, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou 325035, China; Cixi Biomedical Research Institute, Wenzhou Medical University, Ningbo 315300, China.
| |
Collapse
|
53
|
Sun W, Zhao P, Hu S, Zhao Z, Liu B, Yang X, Yang J, Fu Z, Li S, Yu W. NUFIP1-engineered exosomes derived from hUMSCs regulate apoptosis and neurological injury induced by propofol in newborn rats. Neurotoxicology 2024; 102:81-95. [PMID: 38599287 DOI: 10.1016/j.neuro.2024.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 04/05/2024] [Accepted: 04/07/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Propofol can increase neurotoxicity in infants but the precise mechanism is still unknown. Our previous study revealed that nuclear FMR1 interacting protein 1 (NUFIP1), a specific ribophagy receptor, can alleviate T cell apoptosis in sepsis. Yet, the effect of NUFIP1-engineered exosomes elicited from human umbilical cord blood mesenchymal stem cells (hUMSCs) on nerve injury induced by propofol remains unclear. This study intended to investigate the effect of NUFIP1-engineered exosomes on propofol-induced nerve damage in neonatal rats. METHODS Firstly, NUFIP1-engineered exosomes were extracted from hUMSCs serum and their identification was conducted using transmission electron microscopy (TEM), Flow NanoAnalyzer, quantitative real-time polymerase chain reaction (qRT-PCR), and western blot (WB). Subsequently, the optimal exposure duration and concentration of propofol induced apoptosis were determined in SH-SY5Y cell line using WB. Following this, we co-cultured the NUFIP1-engineered exosomes in the knockdown group (NUFIP1-KD) and overexpression group (NUFIP1-OE) with SH-SY5Y cells and assessed their effects on the apoptosis of SH-SY5Y cells using terminal-deoxynucleotidyl transferase mediated nick end labeling (TUNEL) assay, Hoechst 33258 staining, WB, and flow cytometry, respectively. Finally, NUFIP1-engineered exosomes were intraperitoneally injected into neonatal rats, and their effects on the learning and memory ability of neonatal rats were observed through the righting reflex and Morris water maze (MWM) test. Hippocampi were extracted from different groups for hematoxylin-eosin (HE) staining, immunohistochemistry, immunofluorescence, and WB to observe their effects on apoptosis in neonatal rats. RESULTS TEM, Flow NanoAnalyzer, qRT-PCR, and WB analyses confirmed that the exosomes extracted from hUMSCs serum exhibited the expected morphology, diameter, surface markers, and expression of target genes. This confirmed the successful construction of NUFIP1-KD and NUFIP1-OE-engineered exosomes. Optimal exposure duration and concentration of propofol were determined to be 24 hours and 100 µg/ml, respectively. Co-culture of NUFIP1 engineered exosomes and SH-SY5Y cells resulted in significant up-regulation of pro-apoptotic proteins Bax and c-Caspase-3 in the KD group, while anti-apoptotic protein Bcl-2 was significantly decreased. The OE group showed the opposite trend. TUNEL apoptosis assay, Hoechst 33258 staining, and flow cytometry yielded consistent results. Animal experiments demonstrated that intraperitoneal injection of NUFIP1-KD engineered exosomes prolonged the righting reflex recovery time of newborn rats, and MWM tests revealed a significant diminution in the time and number of newborn rats entering the platform. HE staining, immunohistochemistry, immunofluorescence, and WB results also indicated a significant enhancement in apoptosis in this group. Conversely, the experimental results of neonatal rats in the OE group revealed a certain degree of anti-apoptotic effect. CONCLUSIONS NUFIP1-engineered exosomes from hUMSCs have the potential to regulate nerve cell apoptosis and mitigate neurological injury induced by propofol in neonatal rats. Targeting NUFIP1 may hold great significance in ameliorating propofol-induced nerve injury.
Collapse
Affiliation(s)
- Wen Sun
- The First Central Clinical School, Tianjin Medical University, Tianjin, China; Department of Anesthesiology, the Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Pengyue Zhao
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Shidong Hu
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Zhenting Zhao
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Boyan Liu
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Xingpeng Yang
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Jiaqi Yang
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Ze Fu
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Songyan Li
- Department of General Surgery, First Medical Center of the Chinese PLA General Hospital, Beijing, China.
| | - Wenli Yu
- The First Central Clinical School, Tianjin Medical University, Tianjin, China; Department of Anesthesiology, Tianjin First Central Hospital, Tianjin, China.
| |
Collapse
|
54
|
Xu C, Xu P, Zhang J, He S, Hua T, Huang A. Exosomal noncoding RNAs in gynecological cancers: implications for therapy resistance and biomarkers. Front Oncol 2024; 14:1349474. [PMID: 38737906 PMCID: PMC11082286 DOI: 10.3389/fonc.2024.1349474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2023] [Accepted: 04/15/2024] [Indexed: 05/14/2024] Open
Abstract
Gynecologic cancers, including ovarian cancer (OC), cervical cancer (CC), and endometrial cancer (EC), pose a serious threat to women's health and quality of life due to their high incidence and lethality. Therapeutic resistance in tumors refers to reduced sensitivity of tumor cells to therapeutic drugs or radiation, which compromises the efficacy of treatment or renders it ineffective. Therapeutic resistance significantly contributes to treatment failure in gynecologic tumors, although the specific molecular mechanisms remain unclear. Exosomes are nanoscale vesicles released and received by distinct kinds of cells. Exosomes contain proteins, lipids, and RNAs closely linked to their origins and functions. Recent studies have demonstrated that exosomal ncRNAs may be involved in intercellular communication and can modulate the progression of tumorigenesis, aggravation and metastasis, tumor microenvironment (TME), and drug resistance. Besides, exosomal ncRNAs also have the potential to become significant diagnostic and prognostic biomarkers in various of diseases. In this paper, we reviewed the biological roles and mechanisms of exosomal ncRNAs in the drug resistance of gynecologic tumors, as well as explored the potential of exosomal ncRNAs acting as the liquid biopsy molecular markers in gynecologic cancers.
Collapse
Affiliation(s)
| | | | | | | | | | - Aiwu Huang
- Department of Gynecology and Obstetrics , Hangzhou Lin'an Traditional Chinese Medicine Hospital, Hangzhou, Zhejiang, China
| |
Collapse
|
55
|
Xue X, Wang X, Pang M, Yu L, Qian J, Li X, Tian M, Lu C, Xiao C, Liu Y. An exosomal strategy for targeting cancer-associated fibroblasts mediated tumors desmoplastic microenvironments. J Nanobiotechnology 2024; 22:196. [PMID: 38644492 PMCID: PMC11032607 DOI: 10.1186/s12951-024-02452-1] [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: 01/25/2024] [Accepted: 04/01/2024] [Indexed: 04/23/2024] Open
Abstract
Tumors desmoplastic microenvironments are characterized by abundant stromal cells and extracellular matrix (ECM) deposition. Cancer-associated fibroblasts (CAFs), as the most abundant of all stromal cells, play significant role in mediating microenvironments, which not only remodel ECM to establish unique pathological barriers to hinder drug delivery in desmoplastic tumors, but also talk with immune cells and cancer cells to promote immunosuppression and cancer stem cells-mediated drug resistance. Thus, CAFs mediated desmoplastic microenvironments will be emerging as promising strategy to treat desmoplastic tumors. However, due to the complexity of microenvironments and the heterogeneity of CAFs in such tumors, an effective deliver system should be fully considered when designing the strategy of targeting CAFs mediated microenvironments. Engineered exosomes own powerful intercellular communication, cargoes delivery, penetration and targeted property of desired sites, which endow them with powerful theranostic potential in desmoplastic tumors. Here, we illustrate the significance of CAFs in tumors desmoplastic microenvironments and the theranostic potential of engineered exosomes targeting CAFs mediated desmoplastic microenvironments in next generation personalized nano-drugs development.
Collapse
Affiliation(s)
- Xiaoxia Xue
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiangpeng Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Mingshi Pang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Liuchunyang Yu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Jinxiu Qian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Xiaoyu Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Meng Tian
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Cheng Lu
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, 100700, China.
| | - Cheng Xiao
- Institute of Clinical Medicine, China-Japan Friendship Hospital, Beijing, 100029, China.
| | - Yuanyan Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 100029, China.
| |
Collapse
|
56
|
Rathi D, Rossi C, Pospíšil P, Ramalingam Manoharan R, Talarico L, Magnani A, Prasad A. NOX2 and NOX4 expression in monocytes and macrophages-extracellular vesicles in signalling and therapeutics. Front Cell Dev Biol 2024; 12:1342227. [PMID: 38690564 PMCID: PMC11058225 DOI: 10.3389/fcell.2024.1342227] [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: 11/21/2023] [Accepted: 03/25/2024] [Indexed: 05/02/2024] Open
Abstract
Extracellular vesicles (EVs) are a type of cytoplasmic vesicles secreted by a variety of cells. EVs originating from cells have been known to participate in cell communication, antigen presentation, immune cell activation, tolerance induction, etc. These EVs can also carry the active form of Nicotinamide Adenine Dinucleotide Phosphate Oxidase Hydrogen (NADPH) oxidase, which is very essential for the production of reactive oxygen species (ROS) and that can then modulate processes such as cell regeneration. The aim of this study is to characterize the EVs isolated from U-937 and THP-1 cells, identify the NADPH oxidase (NOX) isoforms, and to determine whether EVs can modulate NOX4 and NOX2 in monocytes and macrophages. In our study, isolated EVs of U-937 were characterized using dynamic light scattering (DLS) spectroscopy and immunoblotting. The results showed that the exogenous addition of differentiation agents (either phorbol 12-myristate 13-acetate (PMA) or ascorbic acid) or the supplementation of EVs used in the study did not cause any stress leading to alterations in cell proliferation and viability. In cells co-cultured with EVs for 72 h, strong suppression of NOX4 and NOX2 is evident when monocytes transform into macrophagic cells. We also observed lower levels of oxidative stress measured using immunoblotting and electron paramagnetic resonance spectroscopy under the EVs co-cultured condition, which also indicates that EVs might contribute significantly by acting as an antioxidant source, which agrees with previous studies that hypothesized the role of EVs in therapeutics. Therefore, our results provide evidence for NOX regulation by EVs in addition to its role as an antioxidant cargo.
Collapse
Affiliation(s)
- Deepak Rathi
- Department of Biophysics, Faculty of Science, Palacký University, Olomouc, Czechia
| | - Claudio Rossi
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Center for Colloid and Surface Science (CSGI), Florence, Italy
| | - Pavel Pospíšil
- Department of Biophysics, Faculty of Science, Palacký University, Olomouc, Czechia
| | | | - Luigi Talarico
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Center for Colloid and Surface Science (CSGI), Florence, Italy
| | - Agnese Magnani
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Siena, Italy
- Center for Colloid and Surface Science (CSGI), Florence, Italy
| | - Ankush Prasad
- Department of Biophysics, Faculty of Science, Palacký University, Olomouc, Czechia
| |
Collapse
|
57
|
Ramachandran A, Dhar R, Devi A. Stem Cell-Derived Exosomes: An Advanced Horizon to Cancer Regenerative Medicine. ACS APPLIED BIO MATERIALS 2024; 7:2128-2139. [PMID: 38568170 DOI: 10.1021/acsabm.4c00089] [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: 04/16/2024]
Abstract
Cancer research has made significant progress in recent years, and extracellular vesicles (EVs) based cancer investigation reveals several facts about cancer. Exosomes are a subpopulation of EVs. In the present decade, exosomes is mostly highlighted for cancer theranostic research. Tumor cell derived exosomes (TEXs) promote cancer but there are multiple sources of exosomes that can be used as cancer therapeutic agents (plant exosomes, stem cell-derived exosomes, modified or synthetic exosomes). Stem cells based regenerative medicine faces numerous challenges, such as promote tumor development, cellular reprogramming etc., and therefore addressing these complications becomes essential. Stem cell-derived exosomes serves as an answer to these problems and offers a better solution. Global research indicates that stem cell-derived exosomes also play a dual role in the cellular system by either inhibiting or promoting cancer. Modified exosomes which are genetically engineered exosomes or surface modified exosomes to increase the efficacy of the therapeutic properties can also be considered to target the above concerns. However, the difficulties associated with the exosomes include variations in exosomes heterogenity, isolation protocols, large scale production, etc., and these have to be managed effectively. In this review, we explore exosomes biogenesis, multiple stem cell-derived exosome sources, drug delivery, modified stem cells exosomes, clinical trial of stem cells exosomes, and the related challenges in this domain and future orientation. This article may encourage researchers to explore stem cell-derived exosomes and develop an effective and affordable cancer therapeutic solution.
Collapse
Affiliation(s)
- Aparna Ramachandran
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Rajib Dhar
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Arikketh Devi
- Cancer and Stem Cell Biology Laboratory, Department of Genetic Engineering, SRM Institute of Science and Technology, Kattankulathur, Tamil Nadu 603203, India
| |
Collapse
|
58
|
Ma X, Yang R, Li H, Zhang X, Zhang X, Li X. Role of exosomes in the communication and treatment between OSCC and normal cells. Heliyon 2024; 10:e28148. [PMID: 38560136 PMCID: PMC10981056 DOI: 10.1016/j.heliyon.2024.e28148] [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: 11/15/2023] [Revised: 02/06/2024] [Accepted: 03/13/2024] [Indexed: 04/04/2024] Open
Abstract
Oral squamous cell carcinoma (OSCC) is a prevalent cancer that needs new therapeutic targets due to the poor postoperative prognosis in patients. Exosomes are currently one of important research areas owing to their unique properties. Exosomes are capable of acting as drug transporters, as well as facilitating interactions between OSCC and normal cells. Exosomes can be detected in body fluids such as blood, urine, cerebrospinal fluid, and bile. When exosomes are released from donor cells, they can carry various bioactive molecules to recipient cells, where these molecules participate in biological processes. This review highlights the mechanisms of exosome transfer between normal and OSCC cells. Exosomes isolated from donor OSCC cells can carry circular RNAs (circRNAs), long non-coding RNAs (lncRNAs), and microRNAs (miRNAs) and play a role in signaling processes in the recipient OSCC cells, human umbilical vein endothelial cells, and macrophages. Exosomes secreted by carcinoma-associated fibroblasts, macrophages, and stem cells can also enter the recipient OSCC cells and modulate signaling events in these cells. Exosomes isolated from OSCC plasma, serum, and saliva are also associated with OSCC prognosis. Furthermore, while exosomes were shown to be associated with chemotherapy resistance in OSCC, they can also be used for drug delivery during OSCC treatment. In this paper, we reviewed the molecular mechanisms and functions of exosomes from different cell sources in OSCC cells, providing a basis for diagnosis and prognosis prediction in OSCC patients, and offering guidance for the design of molecular targets carried by exosomes in OSCC.
Collapse
Affiliation(s)
- Xingyue Ma
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Hebei Medical University, Key Laboratory of Stomatology and Clinical Research Centre for Oral Diseases, Hebei Province, Shijiazhuang, 050017, China
| | - Ruisi Yang
- Hebei Medical University, Hebei Province, Shijiazhuang, 050017, China
| | - Haiyang Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Hebei Medical University, Key Laboratory of Stomatology and Clinical Research Centre for Oral Diseases, Hebei Province, Shijiazhuang, 050017, China
| | - Xiaoyan Zhang
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Hebei Medical University, Key Laboratory of Stomatology and Clinical Research Centre for Oral Diseases, Hebei Province, Shijiazhuang, 050017, China
| | - Xiao Zhang
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Hebei Medical University, Key Laboratory of Stomatology and Clinical Research Centre for Oral Diseases, Hebei Province, Shijiazhuang, 050017, China
| | - Xiangjun Li
- Department of Oral and Maxillofacial Surgery, School of Stomatology, Hebei Medical University, Key Laboratory of Stomatology and Clinical Research Centre for Oral Diseases, Hebei Province, Shijiazhuang, 050017, China
| |
Collapse
|
59
|
Niu Z, Wu J, Zhao Q, Zhang J, Zhang P, Yang Y. CAR-based immunotherapy for breast cancer: peculiarities, ongoing investigations, and future strategies. Front Immunol 2024; 15:1385571. [PMID: 38680498 PMCID: PMC11045891 DOI: 10.3389/fimmu.2024.1385571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 03/27/2024] [Indexed: 05/01/2024] Open
Abstract
Surgery, chemotherapy, and endocrine therapy have improved the overall survival and postoperative recurrence rates of Luminal A, Luminal B, and HER2-positive breast cancers but treatment modalities for triple-negative breast cancer (TNBC) with poor prognosis remain limited. The effective application of the rapidly developing chimeric antigen receptor (CAR)-T cell therapy in hematological tumors provides new ideas for the treatment of breast cancer. Choosing suitable and specific targets is crucial for applying CAR-T therapy for breast cancer treatment. In this paper, we summarize CAR-T therapy's effective targets and potential targets in different subtypes based on the existing research progress, especially for TNBC. CAR-based immunotherapy has resulted in advancements in the treatment of breast cancer. CAR-macrophages, CAR-NK cells, and CAR-mesenchymal stem cells (MSCs) may be more effective and safer for treating solid tumors, such as breast cancer. However, the tumor microenvironment (TME) of breast tumors and the side effects of CAR-T therapy pose challenges to CAR-based immunotherapy. CAR-T cells and CAR-NK cells-derived exosomes are advantageous in tumor therapy. Exosomes carrying CAR for breast cancer immunotherapy are of immense research value and may provide a treatment modality with good treatment effects. In this review, we provide an overview of the development and challenges of CAR-based immunotherapy in treating different subtypes of breast cancer and discuss the progress of CAR-expressing exosomes for breast cancer treatment. We elaborate on the development of CAR-T cells in TNBC therapy and the prospects of using CAR-macrophages, CAR-NK cells, and CAR-MSCs for treating breast cancer.
Collapse
Affiliation(s)
- Zhipu Niu
- Clinical Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Jingyuan Wu
- Clinical Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Qiancheng Zhao
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Jinyu Zhang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun, China
| | - Pengyu Zhang
- Clinical Medicine, China-Japan Union Hospital of Jilin University, Changchun, China
| | - Yiming Yang
- Department of Cell Biology and Medical Genetics, College of Basic Medical Sciences, Jilin University, Changchun, China
| |
Collapse
|
60
|
Choi W, Park DJ, Eliceiri BP. Defining tropism and activity of natural and engineered extracellular vesicles. Front Immunol 2024; 15:1363185. [PMID: 38660297 PMCID: PMC11039936 DOI: 10.3389/fimmu.2024.1363185] [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: 12/30/2023] [Accepted: 03/25/2024] [Indexed: 04/26/2024] Open
Abstract
Extracellular vesicles (EVs) have important roles as mediators of cell-to-cell communication, with physiological functions demonstrated in various in vivo models. Despite advances in our understanding of the biological function of EVs and their potential for use as therapeutics, there are limitations to the clinical approaches for which EVs would be effective. A primary determinant of the biodistribution of EVs is the profile of proteins and other factors on the surface of EVs that define the tropism of EVs in vivo. For example, proteins displayed on the surface of EVs can vary in composition by cell source of the EVs and the microenvironment into which EVs are delivered. In addition, interactions between EVs and recipient cells that determine uptake and endosomal escape in recipient cells affect overall systemic biodistribution. In this review, we discuss the contribution of the EV donor cell and the role of the microenvironment in determining EV tropism and thereby determining the uptake and biological activity of EVs.
Collapse
Affiliation(s)
- Wooil Choi
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Dong Jun Park
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
| | - Brian P. Eliceiri
- Department of Surgery, University of California San Diego, La Jolla, CA, United States
- Department of Dermatology, University of California San Diego, La Jolla, CA, United States
| |
Collapse
|
61
|
Saleh RO, Hjazi A, Bansal P, Ahmad I, Kaur H, Ali SHJ, Deorari M, Abosaoda MK, Hamzah HF, Mohammed BA. Mysterious interactions between macrophage-derived exosomes and tumors; what do we know? Pathol Res Pract 2024; 256:155261. [PMID: 38518733 DOI: 10.1016/j.prp.2024.155261] [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: 02/07/2024] [Revised: 03/05/2024] [Accepted: 03/08/2024] [Indexed: 03/24/2024]
Abstract
Through their ability to modify the tumor microenvironment and cancer cells, macrophages play a crucial role in the promotion of tumorigenesis, development of tumors and metastasis, and chemotherapy resistance. A growing body of research has indicated that exosomes may be essential for coordinating the communication between cancer cells and macrophages. One type of extracellular vehicle called an exosome is utilized for delivering a variety of molecules, such as proteins, lipids, and nucleic acids, to specific cells in order to produce pleiotropic effects. Exosomes derived from macrophages exhibit heterogeneity across various cancer types and function paradoxically, suppressing tumor growth while stimulating it, primarily through post-transcriptional control and protein phosphorylation regulation in the receiving cells. Exosomes released by various macrophage phenotypes offer a variety of therapeutic alternatives in the interim. We outlined the most recent developments in this article, including our understanding of the roles that mechanisms and macrophage-derived exosomal biogenesis play in mediating the progression of cancer and their possible therapeutic uses.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Department of Medical Laboratory Techniques, Al-Maarif University College, Al-Anbar, Iraq.
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia.
| | - Pooja Bansal
- Department of Biotechnology and Genetics, Jain (Deemed-to-be) University, Bengaluru, Karnataka 560069, India; Department of Allied Healthcare and Sciences, Vivekananda Global University, Jaipur, Rajasthan 303012, India.
| | - Irfan Ahmad
- Department of Clinical Laboratory Sciences, College of Applied Medical Science, King Khalid University, Abha, Saudi Arabia.
| | - Harpreet Kaur
- School of Basic & Applied Sciences, Shobhit University, Gangoh, Uttar Pradesh 247341, India; Department of Health & Allied Sciences, Arka Jain University, Jamshedpur, Jharkhand 831001, India.
| | - Saad Hayif Jasim Ali
- Department of medical laboratory, College of Health and Medical Technololgy, Al-Ayen University, Thi-Qar, Iraq.
| | - Mahamedha Deorari
- Uttaranchal Institute of Pharmaceutical Sciences, Uttaranchal University, Dehradun, India.
| | - Munther Kadhim Abosaoda
- College of pharmacy, the Islamic University, Najaf, Iraq; College of pharmacy, the Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq; College of pharmacy, the Islamic University of Babylon, Al Diwaniyah, Iraq.
| | - Hamza Fadhel Hamzah
- Department of Medical Laboratories Technology, AL-Nisour University College, Baghdad, Iraq.
| | | |
Collapse
|
62
|
Cao J, Lv G, Wei F. Engineering exosomes to reshape the immune microenvironment in breast cancer: Molecular insights and therapeutic opportunities. Clin Transl Med 2024; 14:e1645. [PMID: 38572668 PMCID: PMC10993163 DOI: 10.1002/ctm2.1645] [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: 11/28/2023] [Revised: 02/19/2024] [Accepted: 03/17/2024] [Indexed: 04/05/2024] Open
Abstract
BACKGROUND Breast cancer remains a global health challenge, necessitating innovative therapeutic approaches. Immunomodulation and immunotherapy have emerged as promising strategies for breast cancer treatment. Engineered exosomes are the sort of exosomes modified with surface decoration and internal therapeutic molecules. Through suitable modifications, engineered exosomes exhibit the capability to overcome the limitations associated with traditional therapeutic approaches. This ability opens up novel avenues for the development of more effective, personalized, and minimally invasive interventions. MAIN BODY In this comprehensive review, we explore the molecular insights and therapeutic potential of engineered exosomes in breast cancer. We discuss the strategies employed for exosome engineering and delve into their molecular mechanisms in reshaping the immune microenvironment of breast cancer. CONCLUSIONS By elucidating the contribution of engineered exosomes to breast cancer immunomodulation, this review underscores the transformative potential of this emerging field for improving breast cancer therapy. HIGHLIGHTS Surface modification of exosomes can improve the targeting specificity. The engineered exosome-loaded immunomodulatory cargo regulates the tumour immune microenvironment. Engineered exosomes are involved in the immune regulation of breast cancer.
Collapse
Affiliation(s)
- Jilong Cao
- Party Affairs and Administration Officethe Fourth Affiliated Hospital of China Medical UniversityShenyangP. R. China
| | - Gang Lv
- Department of Thyroid and Breast SurgeryChaohu Hospital of Anhui Medical UniversityChaohuP. R. China
| | - Fang Wei
- Department of General Surgerythe Fourth Affiliated Hospital of China Medical UniversityShenyangP. R. China
| |
Collapse
|
63
|
Ren X, Xu R, Xu C, Su J. Harnessing exosomes for targeted therapy: strategy and application. BIOMATERIALS TRANSLATIONAL 2024; 5:46-58. [PMID: 39220669 PMCID: PMC11362351 DOI: 10.12336/biomatertransl.2024.01.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 01/22/2024] [Accepted: 02/06/2024] [Indexed: 09/04/2024]
Abstract
Exosomes, nanoscopic extracellular vesicles produced by cells, are pivotal in mediating intracellular communication by transporting nucleic acids, proteins, lipids, and other bioactive molecules, thereby influencing physiological and pathological states. Their endogenous origin and inherent diversity confer distinct advantages over synthetic vehicles like liposomes and nanoparticles in diagnostic and therapeutic applications. Despite their potential, the clinical utility of exosomes is hampered by challenges such as limited storage stability, yield, purity, and targeting efficiency. This review focuses on exosomes as targeted therapeutic agents, examining their biogenesis, classification, isolation, and characterisation, while also addressing the current limitations in yield, purity, and targeting. We delve into the literature to propose optimisation strategies that can enhance their therapeutic efficacy and accelerate the translation of exosome-based therapies into clinical practice.
Collapse
Affiliation(s)
- Xiaoxiang Ren
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| | - Ruixue Xu
- Department of Microbial Pathogenesis, Yale University School of Medicine, New Haven, CT, USA
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong Special Administrative Region, China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, China
- Department of Orthopedic, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, China
| |
Collapse
|
64
|
Si C, Gao J, Ma X. Engineered exosomes in emerging cell-free therapy. Front Oncol 2024; 14:1382398. [PMID: 38595822 PMCID: PMC11003191 DOI: 10.3389/fonc.2024.1382398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024] Open
Abstract
The discovery and use of exosomes ushered in a new era of cell-free therapy. Exosomes are a subgroup of extracellular vesicles that show great potential in disease treatment. Engineered exosomes. with their improved functions have attracted intense interests of their application in translational medicine research. However, the technology of engineering exosomes still faces many challenges which have been the great limitation for their clinical application. This review summarizes the current status of research on engineered exosomes and the difficulties encountered in recent years, with a view to providing new approaches and ideas for future exosome modification and new drug development.
Collapse
Affiliation(s)
| | - Jianen Gao
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xu Ma
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
65
|
Huang Z, Liu X, Guo Q, Zhou Y, Shi L, Cai Q, Tang S, Ouyang Q, Zheng J. Extracellular vesicle-mediated communication between CD8 + cytotoxic T cells and tumor cells. Front Immunol 2024; 15:1376962. [PMID: 38562940 PMCID: PMC10982391 DOI: 10.3389/fimmu.2024.1376962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Tumors pose a significant global public health challenge, resulting in numerous fatalities annually. CD8+ T cells play a crucial role in combating tumors; however, their effectiveness is compromised by the tumor itself and the tumor microenvironment (TME), resulting in reduced efficacy of immunotherapy. In this dynamic interplay, extracellular vesicles (EVs) have emerged as pivotal mediators, facilitating direct and indirect communication between tumors and CD8+ T cells. In this article, we provide an overview of how tumor-derived EVs directly regulate CD8+ T cell function by carrying bioactive molecules they carry internally and on their surface. Simultaneously, these EVs modulate the TME, indirectly influencing the efficiency of CD8+ T cell responses. Furthermore, EVs derived from CD8+ T cells exhibit a dual role: they promote tumor immune evasion while also enhancing antitumor activity. Finally, we briefly discuss current prevailing approaches that utilize functionalized EVs based on tumor-targeted therapy and tumor immunotherapy. These approaches aim to present novel perspectives for EV-based tumor treatment strategies, demonstrating potential for advancements in the field.
Collapse
Affiliation(s)
- Zeyu Huang
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xuehui Liu
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Qinghao Guo
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yihang Zhou
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Linlin Shi
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qingjin Cai
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shupei Tang
- Department of Shigatse Branch, Xinqiao Hospital, Third Military Medical University, Shigatse, China
| | - Qin Ouyang
- Department of Medicinal Chemistry, College of Pharmacy, Third Military Medical University, Chongqing, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
66
|
Dasari N, Guntuku GS, Pindiprolu SKSS. Targeting triple negative breast cancer stem cells using nanocarriers. DISCOVER NANO 2024; 19:41. [PMID: 38453756 PMCID: PMC10920615 DOI: 10.1186/s11671-024-03985-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
Breast cancer is a complex and heterogeneous disease, encompassing various subtypes characterized by distinct molecular features, clinical behaviors, and treatment responses. Categorization of subtypes is based on the presence or absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2), leading to subtypes such as luminal A, luminal B, HER2-positive, and triple-negative breast cancer (TNBC). TNBC, comprising around 20% of all breast cancers, lacks expression of ER, PR, and HER2 receptors, rendering it unresponsive to targeted therapies and presenting significant challenges in treatment. TNBC is associated with aggressive behavior, high rates of recurrence, and resistance to chemotherapy. Tumor initiation, progression, and treatment resistance in TNBC are attributed to breast cancer stem cells (BCSCs), which possess self-renewal, differentiation, and tumorigenic potential. Surface markers, self-renewal pathways (Notch, Wnt, Hedgehog signaling), apoptotic protein (Bcl-2), angiogenesis inhibition (VEGF inhibitors), and immune modulation (cytokines, immune checkpoint inhibitors) are among the key targets discussed in this review. However, targeting the BCSC subpopulation in TNBC presents challenges, including off-target effects, low solubility, and bioavailability of anti-BCSC agents. Nanoparticle-based therapies offer a promising approach to target various molecular pathways and cellular processes implicated in survival of BSCS in TNBC. In this review, we explore various nanocarrier-based approaches for targeting BCSCs in TNBC, aiming to overcome these challenges and improve treatment outcomes for TNBC patients. These nanoparticle-based therapeutic strategies hold promise for addressing the therapeutic gap in TNBC treatment by delivering targeted therapies to BCSCs while minimizing systemic toxicity and enhancing treatment efficacy.
Collapse
Affiliation(s)
- Nagasen Dasari
- Andhra University College of Pharmaceutical Sciences, Andhra University, Vishakhapatnam, Andhra Pradesh, India.
- Aditya Pharmacy College, Surampalem, Andhra Pradesh, India.
- Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh, India.
| | - Girija Sankar Guntuku
- Andhra University College of Pharmaceutical Sciences, Andhra University, Vishakhapatnam, Andhra Pradesh, India
| | - Sai Kiran S S Pindiprolu
- Aditya Pharmacy College, Surampalem, Andhra Pradesh, India
- Jawaharlal Nehru Technological University, Kakinada, Andhra Pradesh, India
| |
Collapse
|
67
|
Ray R, Chowdhury SG, Karmakar P. A vivid outline demonstrating the benefits of exosome-mediated drug delivery in CNS-associated disease environments. Arch Biochem Biophys 2024; 753:109906. [PMID: 38272158 DOI: 10.1016/j.abb.2024.109906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 01/22/2024] [Accepted: 01/22/2024] [Indexed: 01/27/2024]
Abstract
The efficacy of drug delivery mechanisms has been improvised with time for different therapeutic purposes. In most cases, nano-sized delivery systems have been modeled over decades for the on-target applicability of the drugs. The use of synthetic drug delivery materials has been a common practice, although research has now focussed more on using natural vehicles, to avoid the side effects of synthetic delivery systems and easy acceptance by the body. Exosome is such a natural nano-sized vehicle that exceeds the efficiency of many natural vehicles, for being immune-friendly, due to its origin. Unlike, other natural drug delivery systems, exosomes are originated within the body's cells, and from there, they happen to travel through the extracellular matrices into neighboring cells. This capacity of exosomes has made them an efficient drug delivery system over recent years and now a large number of researches have been carried out to develop exosomes as natural drug delivery vehicles. Several experimental strategies have been practiced in this regard which have shown that exosomes are exclusively capable of carrying drugs and they can also be used in targeted delivery, for which they efficiently can reach and release the drug at their target cells for consecutive effects. One of the most interesting features of exosomes is they can cross the blood-brain barrier (BBB) in the body and hence, for the disease where other delivery vehicles are incapable of reaching the destination of the drug, exosomes can overcome the hurdle. This review particularly, focuses on the different aspects of using exosomes as a potential nano-sized drug delivery system for some of the severe diseases associated with the central nervous system of the human body.
Collapse
Affiliation(s)
- Rachayeeta Ray
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, 700032, India
| | | | - Parimal Karmakar
- Department of Life Science and Biotechnology, Jadavpur University, Kolkata, 700032, India.
| |
Collapse
|
68
|
Sun Z, Sun Z, Liu J, Gao X, Jiao L, Zhao Q, Chu Y, Wang X, Deng G, Cai L. Engineered Extracellular Vesicles Expressing Siglec-10 Camouflaged AIE Photosensitizer to Reprogram Macrophages to Active M1 Phenotype and Present Tumor-Associated Antigens for Photodynamic Immunotherapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307147. [PMID: 37941517 DOI: 10.1002/smll.202307147] [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: 09/12/2023] [Revised: 10/15/2023] [Indexed: 11/10/2023]
Abstract
Cancer immunotherapy has attracted considerable attention due to its advantages of persistence, targeting, and ability to kill tumor cells. However, the efficacy of tumor immunotherapy in practical applications is limited by tumor heterogeneity and complex tumor immunosuppressive microenvironments in which abundant of M2 macrophages and immune checkpoints (ICs) are present. Herein, two type-I aggregation-induced emission (AIE)-active photosensitizers with various reactive oxygen species (ROS)-generating efficiencies are designed and synthesized. Engineered extracellular vesicles (EVs) that express ICs Siglec-10 are first obtained from 4T1 tumor cells. The engineered EVs are then fused with the AIE photosensitizer-loaded lipidic nanosystem to form SEx@Fc-NPs. The ROS generated by the inner type-I AIE photosensitizer of the SEx@Fc-NPs through photodynamic therapy (PDT) can convert M2 macrophages into M1 macrophages to improve tumor immunosuppressive microenvironment. The outer EV-antigens that carry 4T1 tumor-associated antigens directly stimulate dendritic cells maturation to activate different types of tumor-specific T cells in overcoming tumor heterogeneity. In addition, blocking Siglec-10 reversed macrophage exhaustion for enhanced antitumor ability. This study presents that a combination of PDT, immune checkpoints, and EV-antigens can greatly improve the efficiency of tumor immunotherapy and is expected to serve as an emerging strategy to improve tumor immunosuppressive microenvironment and overcome immune escape.
Collapse
Affiliation(s)
- Zhihong Sun
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
- Queen Mary School, Nanchang University, Nanchang, 330031, P. R. China
| | - Zhuokai Sun
- Queen Mary School, Nanchang University, Nanchang, 330031, P. R. China
| | - Jie Liu
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Xiaohan Gao
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Liping Jiao
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Qi Zhao
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Yongli Chu
- The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, 264000, P. R. China
| | - Xiaozhong Wang
- The Second Affiliated Hospital of Nanchang University, Nanchang, 330031, P. R. China
- School of Public Health, Nanchang University, Nanchang, 330031, P. R. China
| | - Guanjun Deng
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, P. R. China
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS-HK Joint Lab for Biomaterials, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
- Sino-Euro Center of Biomedicine and Health, Shenzhen, 518024, P. R. China
| |
Collapse
|
69
|
Khoushab S, Aghmiuni MH, Esfandiari N, Sarvandani MRR, Rashidi M, Taheriazam A, Entezari M, Hashemi M. Unlocking the potential of exosomes in cancer research: A paradigm shift in diagnosis, treatment, and prevention. Pathol Res Pract 2024; 255:155214. [PMID: 38430814 DOI: 10.1016/j.prp.2024.155214] [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: 12/16/2023] [Revised: 02/11/2024] [Accepted: 02/15/2024] [Indexed: 03/05/2024]
Abstract
Exosomes, which are tiny particles released by cells, have the ability to transport various molecules, including proteins, lipids, and genetic material containing non-coding RNAs (ncRNAs). They are associated with processes like cancer metastasis, immunity, and tissue repair. Clinical trials have shown exosomes to be effective in treating cancer, inflammation, and chronic diseases. Mesenchymal stem cells (MSCs) and dendritic cells (DCs) are common sources of exosome production. Exosomes have therapeutic potential due to their ability to deliver cargo, modulate the immune system, and promote tissue regeneration. Bioengineered exosomes could revolutionize disease treatment. However, more research is needed to understand exosomes in tumor growth and develop new therapies. This paper provides an overview of exosome research, focusing on cancer and exosome-based therapies including chemotherapy, radiotherapy, and vaccines. It explores exosomes as a drug delivery system for cancer therapy, highlighting their advantages. The article discusses using exosomes for various therapeutic agents, including drugs, antigens, and RNAs. It also examines challenges with engineered exosomes. Analyzing exosomes for clinical purposes faces limitations in sensitivity, specificity, and purification. On the other hand, Nanotechnology offers solutions to overcome these challenges and unlock exosome potential in healthcare. Overall, the article emphasizes the potential of exosomes for personalized and targeted cancer therapy, while acknowledging the need for further research.
Collapse
Affiliation(s)
- Saloomeh Khoushab
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mina Hobabi Aghmiuni
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Negin Esfandiari
- Department of Epidemiology, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | | | - Mohsen Rashidi
- The Health of Plant and Livestock Products Research Center, Mazandaran University of Medical Sciences, Sari, Iran; Department Pharmacology, Faculty of Medicine, Mazandaran University of Medical Sciences, Sari, Iran.
| | - Afshin Taheriazam
- Department of Orthopedics, Faculty of Medicine, Tehran medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
70
|
Chak PT, Kam NW, Choi TH, Dai W, Kwong DLW. Unfolding the Complexity of Exosome-Cellular Interactions on Tumour Immunity and Their Clinical Prospects in Nasopharyngeal Carcinoma. Cancers (Basel) 2024; 16:919. [PMID: 38473281 DOI: 10.3390/cancers16050919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 02/19/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Nasopharyngeal carcinoma (NPC) is an epithelial malignancy situated in the posterolateral nasopharynx. NPC poses grave concerns in Southeast Asia due to its late diagnosis. Together with resistance to standard treatment combining chemo- and radiotherapy, NPC presents high metastatic rates and common recurrence. Despite advancements in immune-checkpoint inhibitors (ICIs) and cytotoxic-T-lymphocytes (CTLs)-based cellular therapy, the exhaustive T cell profile and other signs of immunosuppression within the NPC tumour microenvironment (TME) remain as concerns to immunotherapy response. Exosomes, extracellular vesicles of 30-150 nm in diameter, are increasingly studied and linked to tumourigenesis in oncology. These bilipid-membrane-bound vesicles are packaged with a variety of signalling molecules, mediating cell-cell communications. Within the TME, exosomes can originate from tumour, immune, or stromal cells. Although there are studies on tumour-derived exosomes (TEX) in NPC and their effects on tumour processes like angiogenesis, metastasis, therapeutic resistance, there is a lack of research on their involvement in immune evasion. In this review, we aim to enhance the comprehension of how NPC TEX contribute to cellular immunosuppression. Furthermore, considering the detectability of TEX in bodily fluids, we will also discuss the potential development of TEX-related biomarkers for liquid biopsy in NPC as this could facilitate early diagnosis and prognostication of the disease.
Collapse
Affiliation(s)
- Paak-Ting Chak
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Ngar-Woon Kam
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Laboratory for Synthetic Chemistry and Chemical Biology Limited, Hong Kong Science Park, New Territories, Hong Kong 999077, China
| | - Tsz-Ho Choi
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
| | - Wei Dai
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| | - Dora Lai-Wan Kwong
- Department of Clinical Oncology, Centre of Cancer Medicine, School of Clinical Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong 999077, China
- Clinical Oncology Center, The University of Hong Kong-Shenzhen Hospital, Shenzhen 518053, China
| |
Collapse
|
71
|
Song Y, Hu J, Ma C, Liu H, Li Z, Yang Y. Macrophage-Derived Exosomes as Advanced Therapeutics for Inflammation: Current Progress and Future Perspectives. Int J Nanomedicine 2024; 19:1597-1627. [PMID: 38406601 PMCID: PMC10888065 DOI: 10.2147/ijn.s449388] [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: 11/22/2023] [Accepted: 02/10/2024] [Indexed: 02/27/2024] Open
Abstract
The development of numerous diseases is significantly influenced by inflammation. Macrophage-derived exosomes (M-Exos) play a role in controlling inflammatory reactions in various conditions, including chronic inflammatory pain, hypertension, and diabetes. However, the specific targets and roles of M-Exos in regulating inflammation in diseases remain largely unknown. This review summarizes current knowledge on M-Exos biogenesis and provides updated information on M-Exos' biological function in inflammation modulation. Furthermore, this review highlights the functionalization and engineering strategies of M-Exos, while providing an overview of cutting-edge approaches to engineering M-Exos and advancements in their application as therapeutics for inflammation modulation. Finally, multiple engineering strategies and mechanisms are presented in this review along with their perspectives and challenges, and the potential contribution that M-Exos may have in diseases through the modulation of inflammation is discussed.
Collapse
Affiliation(s)
- Yanjuan Song
- Graduate School, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
| | - Jing Hu
- Wuhan Children’s Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, Hubei Province, People’s Republic of China
| | - Chunlian Ma
- Fitness Monitoring and Chronic Disease Intervention Research Center, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- College of Sports Medicine, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- Hubei Key Laboratory of Exercise Training and Monitoring, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
| | - Hua Liu
- Fitness Monitoring and Chronic Disease Intervention Research Center, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- College of Sports Medicine, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- Hubei Key Laboratory of Exercise Training and Monitoring, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
| | - Zhanghua Li
- Department of Orthopedics, Wuhan Third Hospital, Tongren Hospital of Wuhan University, Wuhan, Hubei Province, People’s Republic of China
| | - Yi Yang
- Fitness Monitoring and Chronic Disease Intervention Research Center, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- College of Sports Medicine, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
- Hubei Key Laboratory of Exercise Training and Monitoring, Wuhan Sports University, Wuhan, Hubei Province, People’s Republic of China
| |
Collapse
|
72
|
Xu Y, Han J, Zhang X, Zhang X, Song J, Gao Z, Qian H, Jin J, Liang Z. Exosomal circRNAs in gastrointestinal cancer: Role in occurrence, development, diagnosis and clinical application (Review). Oncol Rep 2024; 51:19. [PMID: 38099408 PMCID: PMC10777447 DOI: 10.3892/or.2023.8678] [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: 07/24/2023] [Accepted: 10/27/2023] [Indexed: 12/18/2023] Open
Abstract
Gastrointestinal cancer is frequently detected at an advanced stage and has an undesirable prognosis due to the absence of efficient and precise biomarkers and therapeutic targets. Exosomes are small, living‑cell‑derived vesicles that serve a critical role in facilitating intercellular communication by transporting molecules from donor cells to receiver cells. circular RNAs (circRNAs) are mis‑expressed in a variety of diseases, including gastrointestinal cancer, and are promising as diagnostic biomarkers and tumor therapeutic targets for gastrointestinal cancer. The main features of exosomes and circRNAs are discussed in the present review, along with research on the biological function of exosomal circRNAs in the development and progression of gastrointestinal cancer. It also assesses the advantages and disadvantages of implementing these findings in clinical applications.
Collapse
Affiliation(s)
- Yumeng Xu
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213017, P.R. China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jiayi Han
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xuan Zhang
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Xinyi Zhang
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213017, P.R. China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jiajia Song
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213017, P.R. China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Zihan Gao
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213017, P.R. China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Hui Qian
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213017, P.R. China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| | - Jianhua Jin
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213017, P.R. China
| | - Zhaofeng Liang
- Wujin Institute of Molecular Diagnostics and Precision Cancer Medicine of Jiangsu University, Wujin Hospital Affiliated with Jiangsu University, Changzhou, Jiangsu 213017, P.R. China
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu 212013, P.R. China
| |
Collapse
|
73
|
Essola JM, Zhang M, Yang H, Li F, Xia B, Mavoungou JF, Hussain A, Huang Y. Exosome regulation of immune response mechanism: Pros and cons in immunotherapy. Bioact Mater 2024; 32:124-146. [PMID: 37927901 PMCID: PMC10622742 DOI: 10.1016/j.bioactmat.2023.09.018] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 11/07/2023] Open
Abstract
Due to its multiple features, including the ability to orchestrate remote communication between different tissues, the exosomes are the extracellular vesicles arousing the highest interest in the scientific community. Their size, established as an average of 30-150 nm, allows them to be easily uptaken by most cells. According to the type of cells-derived exosomes, they may carry specific biomolecular cargoes used to reprogram the cells they are interacting with. In certain circumstances, exosomes stimulate the immune response by facilitating or amplifying the release of foreign antigens-killing cells, inflammatory factors, or antibodies (immune activation). Meanwhile, in other cases, they are efficiently used by malignant elements such as cancer cells to mislead the immune recognition mechanism, carrying and transferring their cancerous cargoes to distant healthy cells, thus contributing to antigenic invasion (immune suppression). Exosome dichotomic patterns upon immune system regulation present broad advantages in immunotherapy. Its perfect comprehension, from its early biogenesis to its specific interaction with recipient cells, will promote a significant enhancement of immunotherapy employing molecular biology, nanomedicine, and nanotechnology.
Collapse
Affiliation(s)
- Julien Milon Essola
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
- University of Chinese Academy of Sciences. Beijing 100049, PR China
| | - Mengjie Zhang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Haiyin Yang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
| | - Bozhang Xia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, No. 11, First North Road, Zhongguancun, Beijing, 100190, PR China
- University of Chinese Academy of Sciences. Beijing 100049, PR China
| | - Jacques François Mavoungou
- Université Internationale de Libreville, Libreville, 20411, Gabon
- Central and West African Virus Epidemiology, Libreville, 2263, Gabon
- Département de phytotechnologies, Institut National Supérieur d’Agronomie et de Biotechnologie, Université des Sciences et Techniques de Masuku, Franceville, 901, Gabon
- Institut de Recherches Agronomiques et Forestiers, Centre National de la Recherche Scientifique et du développement Technologique, Libreville, 16182, Gabon
| | - Abid Hussain
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Yuanyu Huang
- School of Life Science, Advanced Research Institute of Multidisciplinary Science, School of Medical Technology, Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecule Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
- Rigerna Therapeutics Co. Ltd., China
| |
Collapse
|
74
|
Yang Q, Li S, Ou H, Zhang Y, Zhu G, Li S, Lei L. Exosome-based delivery strategies for tumor therapy: an update on modification, loading, and clinical application. J Nanobiotechnology 2024; 22:41. [PMID: 38281957 PMCID: PMC10823703 DOI: 10.1186/s12951-024-02298-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/02/2024] [Indexed: 01/30/2024] Open
Abstract
Malignancy is a major public health problem and among the leading lethal diseases worldwide. Although the current tumor treatment methods have therapeutic effect to a certain extent, they still have some shortcomings such as poor water solubility, short half-life, local and systemic toxicity. Therefore, how to deliver therapeutic agent so as to realize safe and effective anti-tumor therapy become a problem urgently to be solved in this field. As a medium of information exchange and material transport between cells, exosomes are considered to be a promising drug delivery carrier due to their nano-size, good biocompatibility, natural targeting, and easy modification. In this review, we summarize recent advances in the isolation, identification, drug loading, and modification of exosomes as drug carriers for tumor therapy alongside their application in tumor therapy. Basic knowledge of exosomes, such as their biogenesis, sources, and characterization methods, is also introduced herein. In addition, challenges related to the use of exosomes as drug delivery vehicles are discussed, along with future trends. This review provides a scientific basis for the application of exosome delivery systems in oncological therapy.
Collapse
Affiliation(s)
- Qian Yang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shisheng Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Haibo Ou
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Yuming Zhang
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Gangcai Zhu
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China
| | - Shaohong Li
- Department of Otorhinolaryngology Head and Neck Surgery, The Second Xiangya Hospital, Central South University, Changsha, 410011, Hunan, China.
| | - Lanjie Lei
- Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, Zhejiang, China.
| |
Collapse
|
75
|
Peng C, Xu Y, Wu J, Wu D, Zhou L, Xia X. TME-Related Biomimetic Strategies Against Cancer. Int J Nanomedicine 2024; 19:109-135. [PMID: 38192633 PMCID: PMC10773252 DOI: 10.2147/ijn.s441135] [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: 09/20/2023] [Accepted: 12/21/2023] [Indexed: 01/10/2024] Open
Abstract
The tumor microenvironment (TME) plays an important role in various stages of tumor generation, metastasis, and evasion of immune monitoring and treatment. TME targeted therapy is based on TME components, related pathways or active molecules as therapeutic targets. Therefore, TME targeted therapy based on environmental differences between TME and normal cells has been widely studied. Biomimetic nanocarriers with low clearance, low immunogenicity, and high targeting have enormous potential in tumor treatment. This review introduces the composition and characteristics of TME, including cancer‑associated fibroblasts (CAFs), extracellular matrix (ECM), tumor blood vessels, non-tumor cells, and the latest research progress of biomimetic nanoparticles (NPs) based on TME. It also discusses the opportunities and challenges of clinical transformation of biomimetic nanoparticles.
Collapse
Affiliation(s)
- Cheng Peng
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Yilin Xu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Jing Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Donghai Wu
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Lili Zhou
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| | - Xinhua Xia
- School of Pharmacy, Hunan University of Chinese Medicine, Changsha, People’s Republic of China
| |
Collapse
|
76
|
Tiwari P, Shukla RP, Yadav K, Singh N, Marwaha D, Gautam S, Bakshi AK, Rai N, Kumar A, Sharma D, Mishra PR. Dacarbazine-primed carbon quantum dots coated with breast cancer cell-derived exosomes for improved breast cancer therapy. J Control Release 2024; 365:43-59. [PMID: 37935257 DOI: 10.1016/j.jconrel.2023.11.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
Imprecise targeting of chemotherapeutic drugs often leads to severe toxicity during breast cancer therapy. To address this issue, we have devised a strategy to load dacarbazine (DC) into fucose-based carbon quantum dots (CQDs), which are subsequently coated with exosomes (Ex-DC@CQDs) derived from breast cancer cells. Nanoparticle tracking analysis and western blotting revealed that Ex-DC@CQDs retained the structural and functional characteristics of exosomes. We found that exosomes facilitated the transport of DC@CQDs to cancer cells via heparan sulfate proteoglycan (HSPG) receptors, followed by an augmented depolarization of the mitochondrial membrane potential, ROS generation, and induction of apoptosis leading to cell death. In vivo imaging and pharmacokinetic studies demonstrated enhanced antitumor targeting and efficacy compared to free DC which we attribute to an improved pharmacokinetic profile, a greater tumor accumulation via exosome-mediated- HSPG receptor-driven cell uptake, and sustained release of the Ex-DC@CQDs. Our findings may pave the way for the further development of biologically sourced nanocarriers for breast cancer targeting.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Jawaharlal Nehru University, New Delhi, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Neha Singh
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Avijit Kumar Bakshi
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Nikhil Rai
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ankit Kumar
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Deepak Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
77
|
Keyvani V, Ghale-Noie ZN, Mollazadeh S, Mahmoudian RA, Ghorbani E, Naderi H, Khazaei M, Hassanian SM, Ferns GA, Avan A, Anvari K. Recent Progress in the Application of Exosome Analysis in Ovarian Cancer Management. Curr Cancer Drug Targets 2024; 24:920-929. [PMID: 38284712 DOI: 10.2174/0115680096281906231213055422] [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: 09/12/2023] [Revised: 10/22/2023] [Accepted: 10/25/2023] [Indexed: 01/30/2024]
Abstract
Exosomes are very small (nano-sized) vesicles participating in tumor development by involvement in intercellular communication mediated by transferring biocomponents. Exosomes appear to play vital roles in various cancer development, such as ovarian cancer, a common malignancy in women. Several hallmarks of ovarian cancer are reported to be affected by the exosomemediated cellular cross-talk, including modulating peritoneal dissemination and chemoresistance. Since the expression of some biomolecules, such as miRNAs and mRNA, is changed in ovarian cancer, these exo-biomolecules can be applied as prognostic, diagnostic, and therapeutic biomarkers. Also, the selective loading of specific chemotherapeutic agents into exosomes highlights these biocarries as potential delivery devices. Exosomes could be artificially provided and engineered to better target the site of interest in ovarian cancer. In the present review, we summarize the notable achievement of exosome application in ovarian cancer management to gain applicable transitional insight against this cancer.
Collapse
Affiliation(s)
- Vahideh Keyvani
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
- Medical Genetics Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Zari Naderi Ghale-Noie
- Department of Medical Genetics and Molecular Medicine, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samaneh Mollazadeh
- Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Reihaneh Alsadat Mahmoudian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Elnaz Ghorbani
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- College of Medicine, University of Warith Al-Anbiyaa, Karbala, Iraq
| | - Hamid Naderi
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Majid Khazaei
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Seyed Mahdi Hassanian
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Gordon A Ferns
- Brighton & Sussex Medical School, Division of Medical Education, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Amir Avan
- Metabolic Syndrome Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
- Faculty of Health, School of Biomedical Sciences, Queensland University of Technology (QUT), Brisbane, 4059, Australia
- College of Medicine and Health Sciences, National University of Science and Technology, Sultanate of Oman
| | - Kazem Anvari
- Cancer Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
78
|
Sahoo RK, Tripathi SK, Biswal S, Panda M, Mathapati SS, Biswal BK. Transforming native exosomes to engineered drug vehicles: A smart solution to modern cancer theranostics. Biotechnol J 2024; 19:e2300370. [PMID: 38375578 DOI: 10.1002/biot.202300370] [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: 07/27/2023] [Revised: 12/13/2023] [Accepted: 12/22/2023] [Indexed: 02/21/2024]
Abstract
Exosomes have been the hidden treasure of the cell in terms of cellular interactions, transportation and therapy. The native exosomes (NEx) secreted by the parent cells hold promising aspects in cancer diagnosis and therapy. NEx has low immunogenicity, high biocompatibility, low toxicity and high stability which enables them to be an ideal prognostic biomarker in cancer diagnosis. However, due to heterogeneity, NEx lacks specificity and accuracy to be used as therapeutic drug delivery vehicle in cancer therapy. Transforming these NEx with their innate structure and multiple receptors to engineered exosomes (EEx) can provide better opportunities in the field of cancer theranostics. The surface of the NEx exhibits numeric receptors which can be modified to pave the direction of its therapeutic drug delivery in cancer therapy. Through surface membrane, EEx can be modified with increased drug loading potentiality and higher target specificity to act as a therapeutic nanocarrier for drug delivery. This review provides insights into promising aspects of NEx as a prognostic biomarker and drug delivery tool along with its need for the transformation to EEx in cancer theranostics. We have also highlighted different methods associated with NEx transformations, their nano-bio interaction with recipient cells and major challenges of EEx for clinical application in cancer theranostics.
Collapse
Affiliation(s)
- Rajeev Kumar Sahoo
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Surya Kant Tripathi
- Lineberger Comprehensive Cancer Centre, School of Medicine, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Stuti Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Munmun Panda
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| | - Santosh S Mathapati
- Translational Health Science and Technology Institute Faridabad, Faridabad, Haryana, India
| | - Bijesh Kumar Biswal
- Cancer Drug Resistance Laboratory, Department of Life Science, National Institute of Technology Rourkela, Rourkela, Odisha, India
| |
Collapse
|
79
|
Zhou X, Jia Y, Mao C, Liu S. Small extracellular vesicles: Non-negligible vesicles in tumor progression, diagnosis, and therapy. Cancer Lett 2024; 580:216481. [PMID: 37972701 DOI: 10.1016/j.canlet.2023.216481] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/26/2023] [Accepted: 11/04/2023] [Indexed: 11/19/2023]
Abstract
Small extracellular vesicles (sEVs) such as exosomes are nanoscale membranous particles (<200 nm) that have emerged as crucial targets for liquid biopsy and as promising drug delivery vehicles. They play a significant role in tumor progression as intercellular messengers. They can serve as biomarkers for tumor diagnosis and as drug carriers for cancer treatment. This article reviews recent studies on sEVs in oncology and explores their potential as biomarkers and drug delivery vehicles. Following tumorigenesis, sEVs in the tumor microenvironment (TME) and circulatory system undergo modifications to regulate various events in the TME, including angiogenesis, epithelial-mesenchymal transition (EMT), and tumor immunity, with either pro- or anti-tumor effects. sEVs have been investigated for use as diagnostic and prognostic biomarkers for a variety of tumors, including lung cancer, melanoma, breast cancer, prostate cancer, and hepatocellular carcinoma. sEVs can be used for cancer therapy by packaging drugs or proteins into them through pre- and post-isolation modification techniques. The clinical trials of sEVs as biomarkers and drug carriers are also summarized. Finally, the challenges in the use of sEVs are described and the possible approaches to tackling them are suggested. Overall, sEVs will advance the precision cancer medicine and has shown great potential in clinical applications.
Collapse
Affiliation(s)
- Xinru Zhou
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Yin Jia
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China; School of Materials Science & Engineering, Zhejiang University, Hangzhou, Zhejiang, China.
| | - Shanrong Liu
- Department of Laboratory Diagnostics, Changhai Hospital, Navy Military Medical University, Shanghai, China.
| |
Collapse
|
80
|
Al-Jipouri A, Eritja À, Bozic M. Unraveling the Multifaceted Roles of Extracellular Vesicles: Insights into Biology, Pharmacology, and Pharmaceutical Applications for Drug Delivery. Int J Mol Sci 2023; 25:485. [PMID: 38203656 PMCID: PMC10779093 DOI: 10.3390/ijms25010485] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Revised: 12/19/2023] [Accepted: 12/25/2023] [Indexed: 01/12/2024] Open
Abstract
Extracellular vesicles (EVs) are nanoparticles released from various cell types that have emerged as powerful new therapeutic option for a variety of diseases. EVs are involved in the transmission of biological signals between cells and in the regulation of a variety of biological processes, highlighting them as potential novel targets/platforms for therapeutics intervention and/or delivery. Therefore, it is necessary to investigate new aspects of EVs' biogenesis, biodistribution, metabolism, and excretion as well as safety/compatibility of both unmodified and engineered EVs upon administration in different pharmaceutical dosage forms and delivery systems. In this review, we summarize the current knowledge of essential physiological and pathological roles of EVs in different organs and organ systems. We provide an overview regarding application of EVs as therapeutic targets, therapeutics, and drug delivery platforms. We also explore various approaches implemented over the years to improve the dosage of specific EV products for different administration routes.
Collapse
Affiliation(s)
- Ali Al-Jipouri
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
| | - Àuria Eritja
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| | - Milica Bozic
- Institute for Transfusion Medicine, University Hospital Essen, University of Duisburg-Essen, D-45147 Essen, Germany;
- Vascular and Renal Translational Research Group, Biomedical Research Institute of Lleida Dr. Pifarré Foundation (IRBLLEIDA), 25196 Lleida, Spain;
| |
Collapse
|
81
|
Tian J, Han Z, Song D, Peng Y, Xiong M, Chen Z, Duan S, Zhang L. Engineered Exosome for Drug Delivery: Recent Development and Clinical Applications. Int J Nanomedicine 2023; 18:7923-7940. [PMID: 38152837 PMCID: PMC10752020 DOI: 10.2147/ijn.s444582] [Citation(s) in RCA: 17] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 12/16/2023] [Indexed: 12/29/2023] Open
Abstract
Exosomes are nano-sized membrane vesicles that transfer bioactive molecules between cells and modulate various biological processes under physiological and pathological conditions. By applying bioengineering technologies, exosomes can be modified to express specific markers or carry therapeutic cargo and emerge as novel platforms for the treatment of cancer, neurological, cardiovascular, immune, and infectious diseases. However, there are many challenges and uncertainties in the clinical translation of exosomes. This review aims to provide an overview of the recent advances and challenges in the translation of engineered exosomes, with a special focus on the methods and strategies for loading drugs into exosomes, the pros and cons of different loading methods, and the optimization of exosome production based on the drugs to be encapsulated. Moreover, we also summarize the current clinical applications and prospects of engineered exosomes, as well as the potential risks and limitations that need to be addressed in exosome engineering, including the standardization of exosome preparation and engineering protocols, the quality and quantity of exosomes, the control of drug release, and the immunogenicity and cytotoxicity of exosomes. Overall, engineered exosomes represent an exciting frontier in nanomedicine, but they still face challenges in large-scale production, the maintenance of storage stability, and clinical translation. With continuous advances in this field, exosome-based drug formulation could offer great promise for the targeted treatment of human diseases.
Collapse
Affiliation(s)
- Jiaqi Tian
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
| | - Zhengpu Han
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
- School of Public Health, Weifang Medical University, Weifang, People’s Republic of China
| | - Dandan Song
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
| | - Yanjie Peng
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
| | - Min Xiong
- School of Public Health, North China University of Science and Technology, Tangshan, People’s Republic of China
| | - Zhen Chen
- School of Public Health, Weifang Medical University, Weifang, People’s Republic of China
| | - Shuyin Duan
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, People’s Republic of China
| | - Lin Zhang
- Clinical Medical Research Center for Women and Children Diseases, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, People’s Republic of China
- Key Laboratory of Birth Defect Prevention and Genetic Medicine of Shandong Health Commission, Jinan, People’s Republic of China
| |
Collapse
|
82
|
Wysor SK, Marcus RK. Quantitative Recoveries of Exosomes and Monoclonal Antibodies from Chinese Hamster Ovary Cell Cultures by Use of a Single, Integrated Two-Dimensional Liquid Chromatography Method. Anal Chem 2023; 95:17886-17893. [PMID: 37995145 PMCID: PMC11095952 DOI: 10.1021/acs.analchem.3c04044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2023]
Abstract
Cultured cell lines are very commonly used for the mass production of therapeutic proteins, such as monoclonal antibodies (mAbs). In particular, Chinese hamster ovary (CHO) cell lines are widely employed due to their high tolerance to variations in experimental conditions and their ability to grow in suspension or serum free media. CHO cell lines are known for their ability to produce high titers of biotherapeutic products such as immunoglobulin G (IgG). An emergent alternative means of treating diseases, such as cancer, is the use of gene therapies, wherein genetic cargo is "packaged" in nanosized vesicular structures, referred to as "vectors". One particularly attractive vector option is extracellular vesicles (EVs), of which exosomes are of greatest interest. While exosomes can be harvested from virtually any human body fluid, bovine milk, or even plants, their production in cell cultures is an attractive commercial approach. In fact, the same CHO cell types employed for mAb production also produce exosomes as a natural byproduct. Here, we describe a single integrated 2D liquid chromatography (2DLC) method for the quantitative recovery of both exosomes and antibodies from a singular sample aliquot. At the heart of the method is the use of polyester capillary-channeled polymer (C-CP) fibers as the first dimension column, wherein exosomes/EVs are captured from the supernatant sample and subsequently determined by multiangle light scattering (MALS), while the mAbs are captured, eluted, and quantified using a protein A-modified C-CP fiber column in the second dimension, all in a 10 min workflow. These efforts demonstrate the versatility of the C-CP fiber phases with the capacity to harvest both forms of therapeutics from a single bioreactor, suggesting an appreciable potential impact in the field of biotherapeutics production.
Collapse
Affiliation(s)
- Sarah K Wysor
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, South Carolina 29634-0973, United States
| | - R Kenneth Marcus
- Department of Chemistry, Biosystems Research Complex, Clemson University, Clemson, South Carolina 29634-0973, United States
| |
Collapse
|
83
|
Gong L, Tian L, Cui K, Chen Y, Liu B, Li D, Feng Y, Yao S, Yin Y, Wu Z, Huang Z. An off-the-shelf small extracellular vesicle nanomedicine for tumor targeting therapy. J Control Release 2023; 364:672-686. [PMID: 37967724 DOI: 10.1016/j.jconrel.2023.11.013] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/02/2023] [Accepted: 11/08/2023] [Indexed: 11/17/2023]
Abstract
Small extracellular vesicles (sEVs) have shown excellent prospects as drug delivery systems for cancer therapy. However, the inherent non-targeting and short blood circulation characteristics severely restrict their practical applications as a delivery system. In addition, post-encapsulating drugs into sEVs also remains challenging. Here, we constructed an engineered cell line that secreted multifunctional sEVs (termed NBsEV204) with 7D12 (an anti-EGFR nanobody) and hCD47 decorations on their surface, as well as high levels of miR-204-5p encapsulation. NBsEV204 exhibited extended blood circulation and improved macrophage-mediated phagocytosis of tumor cells by blocking CD47 signaling. Importantly, NBsEV204 specifically targeted EGFR+ tumor cells and showed robust tumor-suppressive effects both in vitro and in vivo. Overall, this study provides a convenient and feasible method to produce off-the-shelf anticancer sEV nanomedicine, which exhibits tremendous potential for clinical translation.
Collapse
Affiliation(s)
- Liang Gong
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 214062 Wuxi, People's Republic of China; Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, People's Republic of China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Lu Tian
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 214062 Wuxi, People's Republic of China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Kaisa Cui
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 214062 Wuxi, People's Republic of China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Ying Chen
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 214062 Wuxi, People's Republic of China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Bingxin Liu
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 214062 Wuxi, People's Republic of China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Dan Li
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Yuyang Feng
- Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Surui Yao
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 214062 Wuxi, People's Republic of China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Yuan Yin
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 214062 Wuxi, People's Republic of China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Zhimeng Wu
- Key Laboratory of Carbohydrate Chemistry & Biotechnology, Ministry of Education, School of Biotechnology, Jiangnan University, 214122 Wuxi, People's Republic of China.
| | - Zhaohui Huang
- Wuxi Cancer Institute, Affiliated Hospital of Jiangnan University, 214062 Wuxi, People's Republic of China; Laboratory of Cancer Epigenetics, Wuxi School of Medicine, Jiangnan University, 214122 Wuxi, People's Republic of China.
| |
Collapse
|
84
|
Guo L, Yang J, Wang H, Yi Y. Multistage Self-Assembled Nanomaterials for Cancer Immunotherapy. Molecules 2023; 28:7750. [PMID: 38067480 PMCID: PMC10707962 DOI: 10.3390/molecules28237750] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 11/18/2023] [Accepted: 11/22/2023] [Indexed: 12/18/2023] Open
Abstract
Advances in nanotechnology have brought innovations to cancer therapy. Nanoparticle-based anticancer drugs have achieved great success from bench to bedside. However, insufficient therapy efficacy due to various physiological barriers in the body remains a key challenge. To overcome these biological barriers and improve the therapeutic efficacy of cancers, multistage self-assembled nanomaterials with advantages of stimuli-responsiveness, programmable delivery, and immune modulations provide great opportunities. In this review, we describe the typical biological barriers for nanomedicines, discuss the recent achievements of multistage self-assembled nanomaterials for stimuli-responsive drug delivery, highlighting the programmable delivery nanomaterials, in situ transformable self-assembled nanomaterials, and immune-reprogramming nanomaterials. Ultimately, we perspective the future opportunities and challenges of multistage self-assembled nanomaterials for cancer immunotherapy.
Collapse
Affiliation(s)
- Lamei Guo
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Jinjun Yang
- Tianjin Key Laboratory of Hazardous Waste Safety Disposal and Recycling Technology, School of Environmental Science and Safety Engineering, Tianjin University of Technology, 391 Binshui Xidao, Xiqing District, Tianjin 300384, China; (L.G.); (J.Y.)
| | - Hao Wang
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| | - Yu Yi
- CAS Center for Excellence in Nanoscience, CAS Key Laboratory for Biological Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology (NCNST), No. 11 Beiyitiao, Zhongguancun, Beijing 100190, China;
| |
Collapse
|
85
|
Huang L, Wu E, Liao J, Wei Z, Wang J, Chen Z. Research Advances of Engineered Exosomes as Drug Delivery Carrier. ACS OMEGA 2023; 8:43374-43387. [PMID: 38027310 PMCID: PMC10666244 DOI: 10.1021/acsomega.3c04479] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Revised: 09/05/2023] [Accepted: 10/27/2023] [Indexed: 12/01/2023]
Abstract
Exosomes are nanoscale vesicles secreted by living cells that have similar membrane composition to parental cells and carry a variety of proteins, lipids, and nucleic acids. Therefore, exosomes have certain biological activities and play an important role in intercellular communication. On the basis of its potential as a carrier for drug delivery systems, exosomes have been engineered to compensate for the shortage of natural exosomes through various engineering strategies for improving drug delivery efficiency, enhancing targeting to tissues and organs, and extending the circulating half-life of exosomes. This review focuses on the engineered exosomes loading drugs through different strategies, discussions on exosome surface modification strategies, and summarizes the advantages and disadvantages of different strategies. In addition, this review provides an overview of the recent applications of engineered exosomes in a number of refractory and relapsable diseases. This review has the potential to provide a reference for further research and development of engineered exosomes.
Collapse
Affiliation(s)
- Lianghui Huang
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Enguang Wu
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Jiawei Liao
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Zongyi Wei
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Jin Wang
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| | - Zhenhua Chen
- Jiangxi Province Key Laboratory of
Drug Design and Evaluation, School of Pharmacy, Jiangxi Science and Technology Normal University, Nanchang 330013, P. R. China
| |
Collapse
|
86
|
Li Y, Peng Q, Wang L. EphA2 as a phase separation protein associated with ferroptosis and immune cell infiltration in colorectal cancer. Aging (Albany NY) 2023; 15:12952-12965. [PMID: 37980165 DOI: 10.18632/aging.205212] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 10/03/2023] [Indexed: 11/20/2023]
Abstract
Colorectal cancer is one of the most common malignant tumors in the digestive system, and its high incidence and metastasis rate make it a terrible killer that threatens human health. In-depth exploration of the targets affecting the progression of colorectal cancer cells and the development of specific targeted drugs for them are of great significance for the prognosis of colorectal cancer patients. Erythropoietin-producing hepatocellular A2 (EphA2) is a member of the Eph subfamily with tyrosine kinase activity, plays a key role in the regulation of signaling pathways related to the malignant phenotype of various tumor cells, but its specific regulatory mechanism in colorectal cancer needs to be further clarified. Here, we found that EphA2 was abnormally highly expressed in colorectal cancer and that patients with colorectal cancer with high EphA2 expression had a worse prognosis. We also found that EphA2 can form liquid-liquid phase separation condensates on cell membrane, which can be disrupted by ALW-II-41-27, an inhibitor of EphA2. In addition, we found that EphA2 expression in colorectal cancer was positively correlated with the expression of ferroptosis-related genes and the infiltration of multiple immune cells. These findings suggest that EphA2 is a novel membrane protein with phase separation ability and is associated with ferroptosis and immune cell infiltration, which further suggests that malignant progression of colorectal cancer may be inhibited by suppressing the phase separation ability of EphA2.
Collapse
Affiliation(s)
- Yanling Li
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Qiu Peng
- Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Lujuan Wang
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan, China
| |
Collapse
|
87
|
Ratajczak K, Grel H, Olejnik P, Jakiela S, Stobiecka M. Current progress, strategy, and prospects of PD-1/PDL-1 immune checkpoint biosensing platforms for cancer diagnostics, therapy monitoring, and drug screening. Biosens Bioelectron 2023; 240:115644. [PMID: 37660460 DOI: 10.1016/j.bios.2023.115644] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 08/22/2023] [Accepted: 08/26/2023] [Indexed: 09/05/2023]
Abstract
Recent technological advancements in testing and monitoring instrumentation have greatly contributed to the progress in cancer treatment by surgical, chemotherapeutic and radiotherapeutic interventions. However, the mortality rate still remains high, calling for the development of new treatment strategies with higher efficacy. Extensive efforts driven in this direction have included broadening of early cancer screening and applying innovative theranostic nanotechnologies. They have been supported by platforms introduced to enable the detection and monitoring of cancer biomarkers, inhibitors, and other agents, able to slow down cancer progression and prevent metastasis. Despite of the well-recognized principles of the immune checkpoint blockade, the efficacy of immunotherapy achieved so far does not meet the well-founded expectations. For a successful cancer treatment, highly sensitive, robust, and inexpensive multiplex biosensors have to be designed to aid in the biomarkers monitoring and in the development of new inhibitors. In this review, we provide an overview of the efforts undertaken to aid in the development and monitoring of anticancer immunotherapy, based on the programmed cell-death immune checkpoint (PD-1/PDL-1) blockade, by designing biosensors for the detection of relevant cancer biomarkers and their inhibitors screening. This review also emphasizes alternative targets made by exosomes carrying PD-L1 overexpressed in cancer cells and passed into the excreted exosomes. Evaluated are also novel targeted drug delivery nanocarriers, providing simultaneous biosensing, thereby contributing to the emerging immune checkpoint cancer therapy. On the basis of the current trends and the emerging technologies, future perspectives of cancer diagnostics and treatment monitoring using biosensing platforms are projected.
Collapse
Affiliation(s)
- Katarzyna Ratajczak
- Department of Physics and Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland
| | - Hubert Grel
- Department of Physics and Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland
| | - Piotr Olejnik
- Department of Physics and Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland
| | - Slawomir Jakiela
- Department of Physics and Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland.
| | - Magdalena Stobiecka
- Department of Physics and Biophysics, Warsaw University of Life Sciences (SGGW), 159 Nowoursynowska Street, 02776, Warsaw, Poland.
| |
Collapse
|
88
|
Hu J, Zhu J, Chai J, Zhao Y, Luan J, Wang Y. Application of exosomes as nanocarriers in cancer therapy. J Mater Chem B 2023; 11:10595-10612. [PMID: 37927220 DOI: 10.1039/d3tb01991h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Cancer remains the most common lethal disease in the world. Although the treatment choices for cancer are still limited, significant progress has been made over the past few years. By improving targeted drug therapy, drug delivery systems promoted the therapeutic effects of anti-cancer medications. Exosome is a kind of natural nanoscale delivery system with natural substance transport properties, good biocompatibility, and high tumor targeting, which shows great potential in drug carriers, thereby providing novel strategies for cancer therapy. In this review, we present the formation, distribution, and characteristics of exosomes. Besides, extraction and isolation techniques are discussed. We focus on the recent progress and application of exosomes in cancer therapy in four aspects: exosome-mediated gene therapy, chemotherapy, photothermal therapy, and combination therapy. The current challenges and future developments of exosome-mediated cancer therapy are also discussed. Finally, the latest advances in the application of exosomes as drug delivery carriers in cancer therapy are summarized, which provide practical value and guidance for the development of cancer therapy.
Collapse
Affiliation(s)
- Jiawei Hu
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Junfei Zhu
- China-Japan Friendship Hospital, No. 2 Sakura East Street, Chaoyang District, Beijing, China
| | - Jingjing Chai
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Yudie Zhao
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Jiajie Luan
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| | - Yan Wang
- Department of Pharmacy, The First Affiliated Hospital of Wannan Medical College, Yijishan Hospital of Wannan Medical College, Wuhu, China.
| |
Collapse
|
89
|
Deng M, Wu S, Huang P, Liu Y, Li C, Zheng J. Engineered exosomes-based theranostic strategy for tumor metastasis and recurrence. Asian J Pharm Sci 2023; 18:100870. [PMID: 38161784 PMCID: PMC10755545 DOI: 10.1016/j.ajps.2023.100870] [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: 09/02/2023] [Revised: 10/10/2023] [Accepted: 10/15/2023] [Indexed: 01/03/2024] Open
Abstract
Metastasis-associated processes are the predominant instigator of fatalities linked to cancer, wherein the pivotal role of circulating tumor cells lies in the resurgence of malignant growth. In recent epochs, exosomes, constituents of the extracellular vesicle cohort, have garnered attention within the field of tumor theranostics owing to their inherent attributes encompassing biocompatibility, modifiability, payload capacity, stability, and therapeutic suitability. Nonetheless, the rudimentary functionalities and limited efficacy of unmodified exosomes curtail their prospective utility. In an effort to surmount these shortcomings, intricate methodologies amalgamating nanotechnology with genetic manipulation, chemotherapy, immunotherapy, and optical intervention present themselves as enhanced avenues to surveil and intercede in tumor metastasis and relapse. This review delves into the manifold techniques currently employed to engineer exosomes, with a specific focus on elucidating the interplay between exosomes and the metastatic cascade, alongside the implementation of tailored exosomes in abating tumor metastasis and recurrence. This review not only advances comprehension of the evolving landscape within this domain but also steers the trajectory of forthcoming investigations.
Collapse
Affiliation(s)
- Min Deng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Shuang Wu
- Medical Research Institute, Southwest University, Chongqing 400716, China
| | - Peizheng Huang
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| | - Yun Liu
- Department of Radiology, Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China
| | - Chong Li
- Medical Research Institute, Southwest University, Chongqing 400716, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China
| | - Ji Zheng
- Department of Urology, Urologic Surgery Center, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing 400037, China
| |
Collapse
|
90
|
Sun M, Yang J, Fan Y, Zhang Y, Sun J, Hu M, Sun K, Zhang J. Beyond Extracellular Vesicles: Hybrid Membrane Nanovesicles as Emerging Advanced Tools for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303617. [PMID: 37749882 PMCID: PMC10646251 DOI: 10.1002/advs.202303617] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2023] [Revised: 08/02/2023] [Indexed: 09/27/2023]
Abstract
Extracellular vesicles (EVs), involved in essential physiological and pathological processes of the organism, have emerged as powerful tools for disease treatment owing to their unique natural biological characteristics and artificially acquired advantages. However, the limited targeting ability, insufficient production yield, and low drug-loading capability of natural simplex EVs have greatly hindered their development in clinical translation. Therefore, the establishment of multifunctional hybrid membrane nanovesicles (HMNVs) with favorable adaptability and flexibility has become the key to expanding the practical application of EVs. This timely review summarizes the current progress of HMNVs for biomedical applications. Different HMNVs preparation strategies including physical, chemical, and chimera approaches are first discussed. This review then individually describes the diverse types of HMNVs based on homologous or heterologous cell membrane substances, a fusion of cell membrane and liposome, as well as a fusion of cell membrane and bacterial membrane. Subsequently, a specific emphasis is placed on the highlight of biological applications of the HMNVs toward various diseases with representative examples. Finally, ongoing challenges and prospects of the currently developed HMNVs in clinical translational applications are briefly presented. This review will not only stimulate broad interest among researchers from diverse disciplines but also provide valuable insights for the development of promising nanoplatforms in precision medicine.
Collapse
Affiliation(s)
- Meng Sun
- Key Laboratory of Molecular Medicine and BiotherapySchool of Life SciencesBeijing Institute of TechnologyBeijing100081P. R. China
| | - Jiani Yang
- Key Laboratory of Molecular Medicine and BiotherapySchool of Life SciencesBeijing Institute of TechnologyBeijing100081P. R. China
| | - Yueyun Fan
- Key Laboratory of Molecular Medicine and BiotherapySchool of Life SciencesBeijing Institute of TechnologyBeijing100081P. R. China
| | - Yinfeng Zhang
- International Medical CenterBeijing Friendship HospitalCapital Medical UniversityBeijing100050P. R. China
| | - Jian Sun
- Department of Hepatobiliary SurgeryJinan University First Affiliated HospitalGuangzhou510630P. R. China
| | - Min Hu
- Department of Hepatobiliary SurgeryJinan University First Affiliated HospitalGuangzhou510630P. R. China
| | - Ke Sun
- Department of Urinary surgeryThe First Affiliated Hospital of Zhengzhou UniversityZhengzhouHenan450052China
| | - Jinfeng Zhang
- Key Laboratory of Molecular Medicine and BiotherapySchool of Life SciencesBeijing Institute of TechnologyBeijing100081P. R. China
| |
Collapse
|
91
|
Cao M, Diao N, Cai X, Chen X, Xiao Y, Guo C, Chen D, Zhang X. Plant exosome nanovesicles (PENs): green delivery platforms. MATERIALS HORIZONS 2023; 10:3879-3894. [PMID: 37671650 DOI: 10.1039/d3mh01030a] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/07/2023]
Abstract
Natural plants have been attracting increasing attention in biomedical research due to their numerous benefits. Plant exosome-derived vesicles, some of the plant's components, are small nanoscale vesicles secreted by plant cells. These vesicles are rich in bioactive substances and play significant roles in intercellular communication, information transfer, and maintaining homeostasis in organisms. They also hold promise for treating diseases, and their vesicular structures make them suitable carriers for drug delivery, with large-scale production feasible. Therefore, this paper aims to provide an overview of nanovesicles from different plant sources and their extraction methods. We also outline the biological activities of nanovesicles, including their anti-inflammatory, anti-viral, and anti-tumor properties, and systematically introduce their applications in drug delivery. These applications include transdermal delivery, targeted drug delivery, gene delivery, and their potential use in the modern food industry. This review provides new ideas and methods for future research on plant exosomes, including their empowerment by artificial intelligence and gene editing, as well as their potential application in the biomedicine, food, and agriculture industries.
Collapse
Affiliation(s)
- Min Cao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai 264005, P. R. China.
| | - Ningning Diao
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai 264005, P. R. China.
| | - Xiaolu Cai
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xing Chen
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| | - Yi Xiao
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| | - Chunjing Guo
- College of Marine Life Science, Ocean University of China, 5# Yushan 10 Road, Qingdao 266003, P. R. China.
| | - Daquan Chen
- Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs, School of Pharmacy, Yantai University, Yantai 264005, P. R. China.
| | - Xingcai Zhang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
92
|
Cheng Z, Shang J, Wang H, Yu L, Yuan Z, Zhang Y, Du Y, Tian J. Molecular imaging-guided extracellular vesicle-based drug delivery for precise cancer management: Current status and future perspectives. J Control Release 2023; 362:97-120. [PMID: 37625599 DOI: 10.1016/j.jconrel.2023.08.040] [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: 05/18/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
Extracellular vesicles (EVs), the mediators of intercellular communication, have attracted the attention of researchers for the important roles they play in cancer treatment. Compared with other inorganic nano-materials, EVs possess the advantages of higher biocompatibility, better physiochemical stability, easier surface modification, and excellent biosafety. They can be used as an advanced drug delivery system with an improved therapeutic index for various therapeutic agents. Engineered EV-based imaging and therapeutic agents (engineered EVs) have emerged as useful tools in targeted cancer diagnosis and therapy. Non-invasive tracing of engineered EVs contributes to a better evaluation of their functions in cancer progression, in vivo dynamic biodistribution, therapeutic response, and drug-loading efficiency. Recent advances in real-time molecular imaging (MI), and innovative EV labeling strategies have led to the development of novel tools that can evaluate the pharmacokinetics of engineered EVs in cancer management, which may accelerate further clinical translation of novel EV-based drug delivery platforms. Herein, we review the latest advances in EVs, their characteristics, and current examples of EV-based targeted drug delivery for cancer. Then, we discuss the prominent applications of MI for tracing both natural and engineered EVs. Finally, we discuss the current challenges and considerations of EVs in targeted cancer treatment and the limitations of different MI modalities. In the coming decades, EV-based therapeutic applications for cancer with improved drug loading and targeting abilities will be developed, and better anti-cancer effects of drug delivery nanoplatform will be achieved.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jihuan Shang
- School of Clinical Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Huarong Wang
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China
| | - Leyi Yu
- Beijing Haidian Hospital, Beijing 100080, China
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China.
| | - Yinlong Zhang
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100080, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, China; Science and Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
93
|
Luo M, Luan X, Jiang G, Yang L, Yan K, Li S, Xiang W, Zhou J. The Dual Effects of Exosomes on Glioma: A Comprehensive Review. J Cancer 2023; 14:2707-2719. [PMID: 37779868 PMCID: PMC10539397 DOI: 10.7150/jca.86996] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/21/2023] [Indexed: 10/03/2023] Open
Abstract
Glioma is a frequently occurring type of cancer that affects the central nervous system. Despite the availability of standardized treatment options including surgical resection, concurrent radiotherapy, and adjuvant temozolomide (TMZ) therapy, the prognosis for glioma patients is often unfavorable. Exosomes act as vehicles for intercellular communication, contributing to tissue repair, immune modulation, and the transfer of metabolic cargo to recipient cells. However, the transmission of abnormal substances can also contribute to pathologic states such as cancer, metabolic diseases, and neurodegenerative disorders. The field of exosome research in oncology has seen significant advancements, with exosomes identified as dynamic modulators of tumor cell proliferation, migration, and invasion, as well as angiogenesis and drug resistance. Exosomes have negligible cytotoxicity, low immunogenicity, and small size, rendering them an ideal therapeutic candidate for glioma. This comprehensive review discusses the dual effects of exosomes in glioma, with an emphasis on their role in facilitating drug resistance. Furthermore, the clinical applications and current limitations of exosomes in glioma therapy are also discussed in detail.
Collapse
Affiliation(s)
- Maowen Luo
- Southwest Medical University, Luzhou 646000, China
| | - Xingzhao Luan
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Department of Neurosurgery, the Affiliated Hospital of PanZhiHua University, PanZhiHua 617000, China
| | - Gen Jiang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Luxia Yang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Kekun Yan
- Department of Neurosurgery, the Affiliated Hospital of PanZhiHua University, PanZhiHua 617000, China
| | - Shenjie Li
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Sichuan Clinical Research Center for Neurosurgery, Luzhou 646000, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China
| | - Wei Xiang
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Sichuan Clinical Research Center for Neurosurgery, Luzhou 646000, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China
| | - Jie Zhou
- Department of Neurosurgery, the Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
- Sichuan Clinical Research Center for Neurosurgery, Luzhou 646000, China
- Academician (Expert) Workstation of Sichuan Province, Luzhou 646000, China
| |
Collapse
|
94
|
Landry J, Shows K, Jagdeesh A, Shah A, Pokhriyal M, Yakovlev V. Regulatory miRNAs in cancer cell recovery from therapy exposure and its implications as a novel therapeutic strategy for preventing disease recurrence. Enzymes 2023; 53:113-196. [PMID: 37748835 DOI: 10.1016/bs.enz.2023.07.007] [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] [Indexed: 09/27/2023]
Abstract
The desired outcome of cancer therapies is the eradication of disease. This can be achieved when therapy exposure leads to therapy-induced cancer cell death as the dominant outcome. Theoretically, a permanent therapy-induced growth arrest could also contribute to a complete response, which has the potential to lead to remission. However, preclinical models have shown that therapy-induced growth arrest is not always durable, as recovering cancer cell populations can contribute to the recurrence of cancer. Significant research efforts have been expended to develop strategies focusing on the prevention of recurrence. Recovery of cells from therapy exposure can occur as a result of several cell stress adaptations. These include cytoprotective autophagy, cellular quiescence, a reversable form of senescence, and the suppression of apoptosis and necroptosis. It is well documented that microRNAs regulate the response of cancer cells to anti-cancer therapies, making targeting microRNAs therapeutically a viable strategy to sensitization and the prevention of recovery. We propose that the use of microRNA-targeting therapies in prolonged sequence, that is, a significant period after initial therapy exposure, could reduce toxicity from the standard combination strategy, and could exploit new epigenetic states essential for cancer cells to recover from therapy exposure. In a step toward supporting this strategy, we survey the available scientific literature to identify microRNAs which could be targeted in sequence to eliminate residual cancer cell populations that were arrested as a result of therapy exposure. It is our hope that by successfully identifying microRNAs which could be targeted in sequence we can prevent disease recurrence.
Collapse
Affiliation(s)
- Joseph Landry
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States.
| | - Kathryn Shows
- Department of Biology, Virginia State University, Petersburg, VA, United States
| | - Akash Jagdeesh
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Aashka Shah
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Mihir Pokhriyal
- Department of Human and Molecular Genetics, VCU Institute of Molecular Medicine, Massey Cancer Center, Virginia Commonwealth University School of Medicine, Richmond, VA, United States
| | - Vasily Yakovlev
- Department of Radiation Oncology, Virginia Commonwealth University, Richmond, VA, United States.
| |
Collapse
|
95
|
Thakur N, Quazi S, Naik B, Jha SK, Singh P. New insights into molecular signaling pathways and current advancements in prostate cancer diagnostics & therapeutics. Front Oncol 2023; 13:1193736. [PMID: 37664036 PMCID: PMC10469924 DOI: 10.3389/fonc.2023.1193736] [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: 03/25/2023] [Accepted: 07/18/2023] [Indexed: 09/05/2023] Open
Abstract
Prostate adenocarcinoma accounts for more than 20% of deaths among males due to cancer. It is the fifth-leading cancer diagnosed in males across the globe. The mortality rate is quite high due to prostate cancer. Despite the fact that advancements in diagnostics and therapeutics have been made, there is a lack of effective drugs. Metabolic pathways are altered due to the triggering of androgen receptor (AR) signaling pathways, and elevated levels of dihydrotestosterone are produced due to defects in AR signaling that accelerate the growth of prostate cancer cells. Further, PI3K/AKT/mTOR pathways interact with AR signaling pathway and act as precursors to promote prostate cancer. Prostate cancer therapy has been classified into luminal A, luminal B, and basal subtypes. Therapeutic drugs inhibiting dihydrotestosterone and PI3K have shown to give promising results to combat prostate cancer. Many second-generation Androgen receptor signaling antagonists are given either as single agent or with the combination of other drugs. In order to develop a cure for metastasized prostate cancer cells, Androgen deprivation therapy (ADT) is applied by using surgical or chemical methods. In many cases, Prostatectomy or local radiotherapy are used to control metastasized prostate cancer. However, it has been observed that after 1.5 years to 2 years of Prostatectomy or castration, there is reoccurrence of prostate cancer and high incidence of castration resistant prostate cancer is seen in population undergone ADT. It has been observed that Androgen derivation therapy combined with drugs like abiraterone acetate or docetaxel improve overall survival rate in metastatic hormone sensitive prostate cancer (mHSPC) patients. Scientific investigations have revealed that drugs inhibiting poly ADP Ribose polymerase (PARP) are showing promising results in clinical trials in the prostate cancer population with mCRPC and DNA repair abnormalities. Recently, RISUG adv (reversible inhibition of sperm under guidance) has shown significant results against prostate cancer cell lines and MTT assay has validated substantial effects of this drug against PC3 cell lines. Current review paper highlights the advancements in prostate cancer therapeutics and new drug molecules against prostate cancer. It will provide detailed insights on the signaling pathways which need to be targeted to combat metastasized prostate cancer and castration resistant prostate cancer.
Collapse
Affiliation(s)
- Neha Thakur
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| | - Sameer Quazi
- Department of Chemistry, Akshara First Grade College, Bengaluru, India
- GenLab Biosolutions Private Limited, Bangalore, Karnataka, India
- Department of Biomedical Sciences, School of Life Sciences, Anglia Ruskin University, Cambridge, United Kingdom
- School of Health Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, United Kingdom
- Solution Chemistry of Advanced Materials and Technologies (SCAMT) Institute, ITMO University, St. Petersburg, Russia
| | - Bindu Naik
- Department of Food Science and Technology, Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Saurabh Kumar Jha
- Department of Biotechnology, School of Engineering and Technology, Sharda University, Greater Noida, India
- Department of Biotechnology Engineering and Food Technology, Chandigarh University, Mohali, India
- Department of Biotechnology, School of Applied & Life Sciences (SALS), Uttaranchal University, Dehradun, India
| | - Pallavi Singh
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, India
| |
Collapse
|
96
|
Kadriya A, Falah M. Nanoscale Phytosomes as an Emerging Modality for Cancer Therapy. Cells 2023; 12:1999. [PMID: 37566078 PMCID: PMC10417745 DOI: 10.3390/cells12151999] [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: 06/27/2023] [Revised: 07/31/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023] Open
Abstract
Extracellular vesicle (EV) research has expanded substantially over the years. EVs have been identified in all living organisms and are produced and released as a means of intercellular communication or as a defense mechanism. Recently, nano-scaled vesicles were successfully isolated from edible plant sources. Plant-derived EVs, referred to here as phytosomes, are of a size reported to range between 30 nm and 120 nm in diameter, similar to small mammalian extracellular vesicles, and carry various bioactive molecules such as mRNA, proteins, miRNA and lipids. Due to the availability of many plants, phytosomes can be easily isolated on a large scale. The methods developed for EV isolation from mammalian cells have been successfully applied for isolation and purification of phytosomes. The therapeutic effects of phytosomes on different disease models, such as inflammation and autoimmune disease, have been reported, and a handful of studies have suggested their therapeutic effects on cancer diseases. Overall, the research on phytosomes is still in its infancy and requires more exploration. This review will narrate the anti-cancer activity and characteristics of phytosomes derived from edible plants as well as describe studies which have utilized phytosomes as drug delivery vehicles for cancer with the ultimate objective of significantly reducing the adverse effects associated with conventional therapeutic approaches.
Collapse
Affiliation(s)
- Ahmad Kadriya
- Medical Research Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
| | - Mizied Falah
- Medical Research Institute, The Holy Family Hospital Nazareth, Nazareth 1641100, Israel;
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed 1311502, Israel
| |
Collapse
|
97
|
Dong X, Wu W, Pan P, Zhang XZ. Engineered Living Materials for Advanced Diseases Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023:e2304963. [PMID: 37436776 DOI: 10.1002/adma.202304963] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/25/2023] [Revised: 07/04/2023] [Accepted: 07/11/2023] [Indexed: 07/13/2023]
Abstract
Natural living materials serving as biotherapeutics exhibit great potential for treating various diseases owing to their immunoactivity, tissue targeting, and other biological activities. In this review, the recent developments in engineered living materials, including mammalian cells, bacteria, viruses, fungi, microalgae, plants, and their active derivatives that are used for treating various diseases are summarized. Further, the future perspectives and challenges of such engineered living material-based biotherapeutics are discussed to provide considerations for future advances in biomedical applications.
Collapse
Affiliation(s)
- Xue Dong
- Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, P. R. China
| | - Wei Wu
- Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, 400037, P. R. China
| | - Pei Pan
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| | - Xian-Zheng Zhang
- Institute for Advanced Studies, Wuhan University, Wuhan, 430072, P. R. China
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, P. R. China
| |
Collapse
|
98
|
Lu Y, Zhang M, Zhou J, Liu X, Wang L, Hu X, Mao Y, Gan R, Chen Z. Extracellular vesicles in renal cell carcinoma: challenges and opportunities coexist. Front Immunol 2023; 14:1212101. [PMID: 37469514 PMCID: PMC10352798 DOI: 10.3389/fimmu.2023.1212101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 06/19/2023] [Indexed: 07/21/2023] Open
Abstract
Renal cell carcinoma (RCC) represents an extremely challenging disease in terms of both diagnosis and treatment. It poses a significant threat to human health, with incidence rates increasing at a yearly rate of roughly 2%. Extracellular vesicles (EVs) are lipid-based bilayer structures of membranes that are essential for intercellular interaction and have been linked to the advancement of RCC. This review provides an overview of recent studies on the role of EVs in RCC progression, including involvement in the interaction of tumor cells with M2 macrophages, mediating the generation of immune tolerance, and assuming the role of communication messengers in the tumor microenvironment leading to disease progression. Finally, the " troika " of EVs in RCC therapy is presented, including engineered sEVs' or EVs tumor vaccines, mesenchymal stem cell EVs therapy, and reduction of tumor-derived EVs secretion. In this context, we highlight the limitations and challenges of EV-based research and the prospects for future developments in this field. Overall, this review provides a comprehensive summary of the role of EVs in RCC and their potential as a viable pathway for the future treatment of this complex disease.
Collapse
Affiliation(s)
- Yukang Lu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Mengting Zhang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Jiajun Zhou
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xiulan Liu
- Department of Medical School, Kunming University of Science and Technology, Kunming, China
| | - Lanfeng Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Xinyi Hu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Yiping Mao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Rongfa Gan
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Zhiping Chen
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
99
|
Sun Y, Li M, Zhang X, Xu D, Wu J, Gu X, Khan A, Shen H, Li Z. A simple and available measurement of onco-sEV dsDNA to protein ratio as a potential tumor marker. BMC Cancer 2023; 23:614. [PMID: 37400751 DOI: 10.1186/s12885-023-10886-3] [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: 01/16/2023] [Accepted: 04/25/2023] [Indexed: 07/05/2023] Open
Abstract
BACKGROUND Small extracellular vesicles (sEVs) have great potential as new biomarkers in liquid biopsy. However, due to the limitations of sEVs extraction and component analysis procedures, further clinical applications of sEVs are hampered. Carcinoembryonic antigen (CEA) is a commonly used broad-spectrum tumor marker that is strongly expressed in a variety of malignancies. RESULTS In this study, CEA+ sEVs were directly separated from serum using immunomagnetic beads, and the nucleic acid to protein ultraviolet absorption ratio (NPr) of CEA+ sEVs was determined. It was found that the NPr of CEA+ sEVs in tumor group was higher than that of healthy group. We further analyzed the sEV-derived nucleic acid components using fluorescent staining and found that the concentration ratio of double-stranded DNA to protein (dsDPr) in CEA+ sEVs was also significantly different between the two groups, with a sensitivity of 100% and a specificity of 41.67% for the diagnosis of pan-cancer. The AUC of dsDPr combined with NPr was 0.87 and the ACU of dsDPr combined with CA242 could reach 0.94, showing good diagnostic performance for pan-cancer. CONCLUSIONS This study demonstrates that the dsDPr of CEA+ sEVs can effectively distinguish sEVs derived from tumor patients and healthy individuals, which can be employed as a simple and cost-effective non-invasive screening technology to assist tumor diagnosis.
Collapse
Affiliation(s)
- Yifan Sun
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Miao Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Xiaoshan Zhang
- College of Life Science, Yangtze University, Jingzhou, China
| | - Dongjie Xu
- College of Life Science, Yangtze University, Jingzhou, China
| | - Jie Wu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China
| | - Xinrui Gu
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Adeel Khan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, National Demonstration Center for Experimental Biomedical Engineering Education (Southeast University, Southeast University, Nanjing, China
| | - Han Shen
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China.
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| | - Zhiyang Li
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Nanjing, China.
- Department of Laboratory Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China.
| |
Collapse
|
100
|
Su P, Wu Y, Xie F, Zheng Q, Chen L, Liu Z, Meng X, Zhou F, Zhang L. A Review of Extracellular Vesicles in COVID-19 Diagnosis, Treatment, and Prevention. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2206095. [PMID: 37144543 PMCID: PMC10323633 DOI: 10.1002/advs.202206095] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/19/2022] [Revised: 04/15/2023] [Indexed: 05/06/2023]
Abstract
The 2019 novel coronavirus disease (COVID-19) pandemic caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) is ongoing, and has necessitated scientific efforts in disease diagnosis, treatment, and prevention. Interestingly, extracellular vesicles (EVs) have been crucial in these developments. EVs are a collection of various nanovesicles which are delimited by a lipid bilayer. They are enriched in proteins, nucleic acids, lipids, and metabolites, and naturally released from different cells. Their natural material transport properties, inherent long-term recycling ability, excellent biocompatibility, editable targeting, and inheritance of parental cell properties make EVs one of the most promising next-generation drug delivery nanocarriers and active biologics. During the COVID-19 pandemic, many efforts have been made to exploit the payload of natural EVs for the treatment of COVID-19. Furthermore, strategies that use engineered EVs to manufacture vaccines and neutralization traps have produced excellent efficacy in animal experiments and clinical trials. Here, the recent literature on the application of EVs in COVID-19 diagnosis, treatment, damage repair, and prevention is reviewed. And the therapeutic value, application strategies, safety, and biotoxicity in the production and clinical applications of EV agents for COVID-19 treatment, as well as inspiration for using EVs to block and eliminate novel viruses are discussed.
Collapse
Affiliation(s)
- Peng Su
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Yuchen Wu
- Department of Clinical MedicineThe First School of MedicineWenzhou Medical UniversityWenzhouZhejiang325035P. R. China
| | - Feng Xie
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Qinghui Zheng
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
| | - Long Chen
- Center for Translational MedicineThe Affiliated Zhangjiagang Hospital of Soochow UniversityZhangjiagangJiangsu215600China
| | - Zhuang Liu
- Institute of Functional Nano and Soft Materials (FUNSOM)Jiangsu Key Laboratory for Carbon‐Based Functional Materials and DevicesSoochow UniversitySuzhouJiangsu215123China
| | - Xuli Meng
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
| | - Fangfang Zhou
- Institutes of Biology and Medical ScienceSoochow UniversitySuzhou215123P. R. China
| | - Long Zhang
- Department of Breast SurgeryZhejiang Provincial People's HospitalHangzhou310014P. R. China
- MOE Laboratory of Biosystems Homeostasis & Protection and Innovation Center for Cell Signaling NetworkLife Sciences InstituteZhejiang UniversityHangzhou310058P. R. China
- Cancer CenterZhejiang UniversityHangzhouZhejiang310058P. R. China
| |
Collapse
|