201
|
Alptekin A, Parvin M, Chowdhury HI, Rashid MH, Arbab AS. Engineered exosomes for studies in tumor immunology. Immunol Rev 2022; 312:76-102. [PMID: 35808839 DOI: 10.1111/imr.13107] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 05/27/2022] [Indexed: 12/14/2022]
Abstract
Exosomes are a type of extracellular vesicle (EV) with diameters of 30-150 nm secreted by most of the cells into the extracellular spaces and can alter the microenvironment through cell-to-cell interactions by fusion with the plasma membrane and subsequent endocytosis and release of the cargo. Because of their biocompatibility, low toxicity and immunogenicity, permeability (even through the blood-brain barrier (BBB)), stability in biological fluids, and ability to accumulate in the lesions with higher specificity, investigators have started making designer's exosomes or engineered exosomes to carry biologically active protein on the surface or inside the exosomes as well as using exosomes to carry drugs, micro RNA, and other products to the site of interest. In this review, we have discussed biogenesis, markers, and contents of various exosomes including exosomes of immune cells. We have also discussed the current methods of making engineered and designer's exosomes as well as the use of engineered exosomes targeting different immune cells in the tumors, stroke, as well as at peripheral blood. Genetic engineering and customizing exosomes create an unlimited opportunity to use in diagnosis and treatment. Very little use has been discovered, and we are far away to reach its limits.
Collapse
Affiliation(s)
- Ahmet Alptekin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | - Mahrima Parvin
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| | | | | | - Ali S Arbab
- Georgia Cancer Center, Augusta University, Augusta, Georgia, USA
| |
Collapse
|
202
|
Mukherjee A, Bisht B, Dutta S, Paul MK. Current advances in the use of exosomes, liposomes, and bioengineered hybrid nanovesicles in cancer detection and therapy. Acta Pharmacol Sin 2022; 43:2759-2776. [PMID: 35379933 PMCID: PMC9622806 DOI: 10.1038/s41401-022-00902-w] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 03/15/2022] [Indexed: 12/17/2022]
Abstract
Three major approaches of cancer therapy can be enunciated as delivery of biotherapeutics, tumor image analysis, and immunotherapy. Liposomes, artificial fat bubbles, are long known for their capacity to encapsulate a diverse range of bioactive molecules and release the payload in a sustained, stimuli-responsive manner. They have already been widely explored as a delivery vehicle for therapeutic drugs as well as imaging agents. They are also extensively being used in cancer immunotherapy. On the other hand, exosomes are naturally occurring nanosized extracellular vesicles that serve an important role in cell-cell communication. Importantly, the exosomes also have proven their capability to carry an array of active pharmaceuticals and diagnostic molecules to the tumor cells. Exosomes, being enriched with tumor antigens, have numerous immunomodulatory effects. Much to our intrigue, in recent times, efforts have been directed toward developing smart, bioengineered, exosome-liposome hybrid nanovesicles, which are augmented by the benefits of both vesicular systems. This review attempts to summarize the contemporary developments in the use of exosome and liposome toward cancer diagnosis, therapy, as a vehicle for drug delivery, diagnostic carrier for tumor imaging, and cancer immunotherapy. We shall also briefly reflect upon the recent advancements of the exosome-liposome hybrids in cancer therapy. Finally, we put forward future directions for the use of exosome/liposome and/or hybrid nanocarriers for accurate diagnosis and personalized therapies for cancers.
Collapse
Affiliation(s)
| | - Bharti Bisht
- Division of Thoracic Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Suman Dutta
- International Institute of Innovation and Technology, New Town, Kolkata, 700156, India
| | - Manash K Paul
- Division of Pulmonary and Critical Care Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
203
|
Rajput A, Varshney A, Bajaj R, Pokharkar V. Exosomes as New Generation Vehicles for Drug Delivery: Biomedical Applications and Future Perspectives. Molecules 2022; 27:7289. [PMID: 36364116 PMCID: PMC9658823 DOI: 10.3390/molecules27217289] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/20/2022] [Accepted: 10/21/2022] [Indexed: 11/30/2022] Open
Abstract
Currently, particular interest among the scientific community is focused on exploring the use of exosomes for several pharmaceutical and biomedical applications. This is due to the identification of the role of exosomes as an excellent intercellular communicator by delivering the requisite cargo comprising of functional proteins, metabolites and nucleic acids. Exosomes are the smallest extracellular vesicles (EV) with sizes ranging from 30-100 nm and are derived from endosomes. Exosomes have similar surface morphology to cells and act as a signal transduction channel between cells. They encompass different biomolecules, such as proteins, nucleic acids and lipids, thus rendering them naturally as an attractive drug delivery vehicle. Like the other advanced drug delivery systems, such as polymeric nanoparticles and liposomes to encapsulate drug substances, exosomes also gained much attention in enhancing therapeutic activity. Exosomes present many advantages, such as compatibility with living tissues, low toxicity, extended blood circulation, capability to pass contents from one cell to another, non-immunogenic and special targeting of various cells, making them an excellent therapeutic carrier. Exosome-based molecules for drug delivery are still in the early stages of research and clinical trials. The problems and clinical transition issues related to exosome-based drugs need to be overcome using advanced tools for better understanding and systemic evaluation of exosomes. In this current review, we summarize the most up-to-date knowledge about the complex biological journey of exosomes from biogenesis and secretion, isolation techniques, characterization, loading methods, pharmaceutical and therapeutic applications, challenges and future perspectives of exosomes.
Collapse
Affiliation(s)
| | | | | | - Varsha Pokharkar
- Department of Pharmaceutics, Poona College of Pharmacy, Bharti Vidyapeeth Deemed University, Erandwane, Pune 411038, Maharashtra, India
| |
Collapse
|
204
|
Zhou Y, Bréchard S. Neutrophil Extracellular Vesicles: A Delicate Balance between Pro-Inflammatory Responses and Anti-Inflammatory Therapies. Cells 2022; 11:cells11203318. [PMID: 36291183 PMCID: PMC9600967 DOI: 10.3390/cells11203318] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/18/2022] [Accepted: 10/20/2022] [Indexed: 11/16/2022] Open
Abstract
Extracellular vesicles (EVs) are released in the extracellular environment during cell activation or apoptosis. Working as signal transducers, EVs are important mediators of intercellular communication through the convoying of proteins, nucleic acids, lipids, and metabolites. Neutrophil extracellular vesicles (nEVs) contain molecules acting as key modulators of inflammation and immune responses. Due to their potential as therapeutic tools, studies about nEVs have been increasing in recent years. However, our knowledge about nEVs is still in its infancy. In this review, we summarize the current understanding of the role of nEVs in the framework of neutrophil inflammation functions and disease development. The therapeutic potential of nEVs as clinical treatment strategies is deeply discussed. Moreover, the promising research landscape of nEVs in the near future is also examined.
Collapse
|
205
|
Komuro H, Aminova S, Lauro K, Harada M. Advances of engineered extracellular vesicles-based therapeutics strategy. SCIENCE AND TECHNOLOGY OF ADVANCED MATERIALS 2022; 23:655-681. [PMID: 36277506 PMCID: PMC9586594 DOI: 10.1080/14686996.2022.2133342] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 09/26/2022] [Accepted: 09/28/2022] [Indexed: 05/09/2023]
Abstract
Extracellular vesicles (EVs) are a heterogeneous population of lipid bilayer membrane-bound vesicles which encapsulate bioactive molecules, such as nucleic acids, proteins, and lipids. They mediate intercellular communication through transporting internally packaged molecules, making them attractive therapeutics carriers. Over the last decades, a significant amount of research has implied the potential of EVs servings as drug delivery vehicles for nuclear acids, proteins, and small molecular drugs. However, several challenges remain unresolved before the clinical application of EV-based therapeutics, including lack of specificity, stability, biodistribution, storage, large-scale manufacturing, and the comprehensive analysis of EV composition. Technical development is essential to overcome these issues and enhance the pre-clinical therapeutic effects. In this review, we summarize the current advancements in EV engineering which demonstrate their therapeutic potential.
Collapse
Affiliation(s)
- Hiroaki Komuro
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Shakhlo Aminova
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Katherine Lauro
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA
| |
Collapse
|
206
|
Hussen BM, Faraj GSH, Rasul MF, Hidayat HJ, Salihi A, Baniahmad A, Taheri M, Ghafouri-Frad S. Strategies to overcome the main challenges of the use of exosomes as drug carrier for cancer therapy. Cancer Cell Int 2022; 22:323. [PMID: 36258195 PMCID: PMC9580186 DOI: 10.1186/s12935-022-02743-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/05/2022] [Indexed: 11/10/2022] Open
Abstract
Exosomes are naturally occurring nanosized particles that aid intercellular communication by transmitting biological information between cells. Exosomes have therapeutic efficacy that can transfer their contents between cells as natural carriers. In addition, the exosomal contents delivered to the recipient pathological cells significantly inhibit cancer progression. However, exosome-based tumor treatments are inadequately precise or successful, and various challenges should be adequately overcome. Here, we discuss the significant challenges that exosomes face as drug carriers used for therapeutic targets and strategies for overcoming these challenges in order to promote this new incoming drug carrier further and improve future clinical outcomes. We also present techniques for overcoming these challenges.
Collapse
Affiliation(s)
- Bashdar Mahmud Hussen
- Department of Pharmacognosy, College of Pharmacy, Hawler Medical University, Erbil, Kurdistan Region, Iraq.,Center of Research and Strategic Studies, Lebanese French University, Erbil, Iraq
| | - Goran Sedeeq Hama Faraj
- College of Medicine, Department of Medical Laboratory Sciences, Komar University of Science and Technology, Sulaymaniyah, Iraq
| | - Mohammad Fatih Rasul
- Department of Pharmaceutical Basic Science, Faculty of Pharmacy, Tishk International University, Erbil, Kurdistan Region, Iraq
| | - Hazha Jamal Hidayat
- Department of Biology, College of Education, Salahaddin University, Erbil, Kurdistan Region, Iraq
| | - Abbas Salihi
- Department of Biology, College of Science, Salahaddin University, Erbil, Kurdistan Region, Iraq
| | - Aria Baniahmad
- Institute of Human Genetics, Jena University Hospital, Jena, Germany
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany. .,Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Soudeh Ghafouri-Frad
- Department of Medical Genetics,, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
207
|
Yao Y, Jiang Y, Song J, Wang R, Li Z, Yang L, Wu W, Zhang L, Peng Q. Exosomes as Potential Functional Nanomaterials for Tissue Engineering. Adv Healthc Mater 2022:e2201989. [PMID: 36253093 DOI: 10.1002/adhm.202201989] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 09/14/2022] [Indexed: 11/10/2022]
Abstract
Exosomes are cell-derived extracellular vesicles of 40-160 nm diameter, which carry numerous biomolecules and transmit information between cells. They are used as functional nanomaterials with great potential in biomedical areas, such as active agents and delivery systems for advanced drug delivery and disease therapy. In recent years, potential applications of exosomes in tissue engineering have attracted significant attention, and some critical progress has been made. This review gives a complete picture of exosomes and their applications in the regeneration of various tissues, such as the central nervous systems, kidney, bone, cartilage, heart, and endodontium. Approaches employed for modifying exosomes to equip them with excellent targeting capacity are summarized. Furthermore, current concerns and future outlook of exosomes in tissue engineering are discussed.
Collapse
Affiliation(s)
- Yang Yao
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Yuhuan Jiang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Jialu Song
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Ruojing Wang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Zhaoping Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Lei Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Weimin Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Luyue Zhang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| | - Qiang Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, No.14, Block 3, Renmin Road South, Chengdu, 610041, P. R. China
| |
Collapse
|
208
|
Yin Y, Han X, Li C, Sun T, Li K, Liu X, Liu M. The status of industrialization and development of exosomes as a drug delivery system: A review. Front Pharmacol 2022; 13:961127. [PMID: 36304147 PMCID: PMC9592696 DOI: 10.3389/fphar.2022.961127] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2022] [Accepted: 09/27/2022] [Indexed: 12/04/2022] Open
Abstract
Exosomes, as natural biomolecular carriers produced by cells, have the potential and advantage of delivering drugs to target organs or cells in vivo. The steps to improve exosomes as a drug delivery system can be divided into three steps:large-scale preparation of exosomes, loading of drugs and targeted delivery of exosomes. Based on the existing production process and technology, there is still much room for improvement. This review highlights the research progress in three aspects and proposes new technologies and innovative approaches to improve the efficiency of exosome delivery.
Collapse
Affiliation(s)
- Yi Yin
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xing Han
- School of Basic Medical Science, Central South University, Changsha, Hunan, China
| | - Cheng Li
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Tonghui Sun
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Kailin Li
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Xionghao Liu
- Center for Medical Genetics and Hunan Key Laboratory of Medical Genetics, School of Life Sciences, Central South University, Hunan, China
| | - Mujun Liu
- Department of Cell Biology, School of Life Sciences, Central South University, Changsha, Hunan, China
- *Correspondence: Mujun Liu,
| |
Collapse
|
209
|
Mir SA, Hamid L, Bader GN, Shoaib A, Rahamathulla M, Alshahrani MY, Alam P, Shakeel F. Role of Nanotechnology in Overcoming the Multidrug Resistance in Cancer Therapy: A Review. Molecules 2022; 27:6608. [PMID: 36235145 PMCID: PMC9571152 DOI: 10.3390/molecules27196608] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 09/29/2022] [Accepted: 10/02/2022] [Indexed: 11/06/2022] Open
Abstract
Cancer is one of the leading causes of morbidity and mortality around the globe and is likely to become the major cause of global death in the coming years. As per World Health Organization (WHO) report, every year there are over 10 and 9 million new cases and deaths from this disease. Chemotherapy, radiotherapy, and surgery are the three basic approaches to treating cancer. These approaches are aiming at eradicating all cancer cells with minimum off-target effects on other cell types. Most drugs have serious adverse effects due to the lack of target selectivity. On the other hand, resistance to already available drugs has emerged as a major obstacle in cancer chemotherapy, allowing cancer to proliferate irrespective of the chemotherapeutic agent. Consequently, it leads to multidrug resistance (MDR), a growing concern in the scientific community. To overcome this problem, in recent years, nanotechnology-based drug therapies have been explored and have shown great promise in overcoming resistance, with most nano-based drugs being explored at the clinical level. Through this review, we try to explain various mechanisms involved in multidrug resistance in cancer and the role nanotechnology has played in overcoming or reversing this resistance.
Collapse
Affiliation(s)
- Suhail Ahmad Mir
- Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar 190006, India
| | - Laraibah Hamid
- Department of Zoology, University of Kashmir, Hazratbal, Srinagar 190006, India
| | - Ghulam Nabi Bader
- Department of Pharmaceutical Sciences, University of Kashmir, Hazratbal, Srinagar 190006, India
| | - Ambreen Shoaib
- Department of Pharmacy Practice, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Mohamed Rahamathulla
- Department of Pharmaceutics, College of Pharmacy, King Khalid University, Abha 61421, Saudi Arabia
| | - Mohammad Y. Alshahrani
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha 61421, Saudi Arabia
| | - Prawez Alam
- Department of Pharmacognosy, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Faiyaz Shakeel
- Department of Pharmaceutics, College of Pharmacy, King Saud University, Riyadh 11451, Saudi Arabia
| |
Collapse
|
210
|
Ezhilarasan D, Lakshmi T, Mallineni SK. Nano-based targeted drug delivery for lung cancer: therapeutic avenues and challenges. Nanomedicine (Lond) 2022; 17:1855-1869. [PMID: 35311343 DOI: 10.2217/nnm-2021-0364] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2021] [Accepted: 03/04/2022] [Indexed: 12/24/2022] Open
Abstract
Most anticancer drugs often fail in clinical trials due to poor solubility, poor bioavailability, lack of targeted delivery and several off-target effects. Polymeric nanoparticles such as poly(lactide), poly(lactic-co-glycolic acid), ALB-loading paclitaxel (Abraxane® ABI-007), lomustine-loaded chitosan, gelatin (decorated with EGF receptor-targeted biotinylated EGF) and so on offer controlled and sustained drug-release properties, biocompatibility and promising anticancer effects. EGF, folic acid, transferrin, sigma and urokinase plasminogen activator receptors-targeting nano preparations improve bioavailability and accumulate drugs on the lung tumor cell surface. However, route of administration, size, pharmacokinetic properties, immune clearance and so on hamper nanomedicines' clinical uses. This review focuses on the benefits, avenues and challenges of nanoparticle-based drug-delivery systems for lung cancer treatment.
Collapse
Affiliation(s)
- Devaraj Ezhilarasan
- Department of Pharmacology, Gold Lab, Saveetha Dental College, Saveetha Institute of Medical & Technical Sciences (SIMATS), Chennai, Tamil Nadu, 600077, India
| | - Thangavelu Lakshmi
- Department of Pharmacology, Gold Lab, Saveetha Dental College, Saveetha Institute of Medical & Technical Sciences (SIMATS), Chennai, Tamil Nadu, 600077, India
| | - Sreekanth Kumar Mallineni
- Department of Preventive Dental Sciences, College of Dentistry, Majmaah University, Almajmaah, 11952, Saudi Arabia
| |
Collapse
|
211
|
Song H, Chen X, Hao Y, Wang J, Xie Q, Wang X. Nanoengineering facilitating the target mission: targeted extracellular vesicles delivery systems design. J Nanobiotechnology 2022; 20:431. [PMID: 36175866 PMCID: PMC9524104 DOI: 10.1186/s12951-022-01638-9] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2022] [Accepted: 09/04/2022] [Indexed: 11/10/2022] Open
Abstract
Precision medicine has put forward the proposition of "precision targeting" for modern drug delivery systems. Inspired by techniques from biology, pharmaceutical sciences, and nanoengineering, numerous targeted drug delivery systems have been developed in recent decades. But the large-scale applications of these systems are limited due to unsatisfactory targeting efficiency, cytotoxicity, easy removability, and instability. As such, the natural endogenous cargo delivery vehicle-extracellular vesicles (EVs)-have sparked significant interest for its unique inherent targeting properties, biocompatibility, transmembrane ability, and circulatory stability. The membranes of EVs are enriched for receptors or ligands that interact with target cells, which endows them with inherent targeting mission. However, most of the natural therapeutic EVs face the fate of being cleared by macrophages, resulting in off-target. Therefore, the specificity of natural EVs delivery systems urgently needs to be further improved. In this review, we comprehensively summarize the inherent homing mechanisms of EVs and the effects of the donor cell source and administration route on targeting specificity. We then go over nanoengineering techniques that modify EVs for improving specific targeting, such as source cell alteration and modification of EVs surface. We also highlight the auxiliary strategies to enhance specificity by changing the external environment, such as magnetic and photothermal. Furthermore, contemporary issues such as the lack of a gold standard for assessing targeting efficiency are discussed. This review will provide new insights into the development of precision medicine delivery systems.
Collapse
Affiliation(s)
- Haoyue Song
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xiaohang Chen
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Yujia Hao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jia Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Qingpeng Xie
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xing Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China. .,Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China.
| |
Collapse
|
212
|
Qian K, Fu W, Li T, Zhao J, Lei C, Hu S. The roles of small extracellular vesicles in cancer and immune regulation and translational potential in cancer therapy. J Exp Clin Cancer Res 2022; 41:286. [PMID: 36167539 PMCID: PMC9513874 DOI: 10.1186/s13046-022-02492-1] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/08/2022] [Indexed: 11/23/2022] Open
Abstract
Extracellular vesicles (EVs) facilitate the extracellular transfer of proteins, lipids, and nucleic acids and mediate intercellular communication among multiple cells in the tumour environment. Small extracellular vesicles (sEVs) are defined as EVs range in diameter from approximately 50 to 150 nm. Tumour-derived sEVs (TDsEVs) and immune cell-derived sEVs have significant immunological activities and participate in cancer progression and immune responses. Cancer-specific molecules have been identified on TDsEVs and can function as biomarkers for cancer diagnosis and prognosis, as well as allergens for TDsEVs-based vaccination. Various monocytes, including but not limited to dendritic cells (DCs), B cells, T cells, natural killer (NK) cells, macrophages, and myeloid-derived suppressor cells (MDSCs), secrete sEVs that regulate immune responses in the complex immune network with either protumour or antitumour effects. After engineered modification, sEVs from immune cells and other donor cells can provide improved targeting and biological effects. Combined with their naïve characteristics, these engineered sEVs hold great potential as drug carriers. When used in a variety of cancer therapies, they can adjunctly enhance the safety and antitumor efficacy of multiple therapeutics. In summary, both naïve sEVs in the tumour environment and engineered sEVs with effector cargoes are regarded as showing promising potential for use in cancer diagnostics and therapeutics.
Collapse
|
213
|
Gao X, Gao B, Li S. Extracellular vesicles: A new diagnostic biomarker and targeted drug in osteosarcoma. Front Immunol 2022; 13:1002742. [PMID: 36211364 PMCID: PMC9539319 DOI: 10.3389/fimmu.2022.1002742] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Accepted: 09/12/2022] [Indexed: 11/25/2022] Open
Abstract
Osteosarcoma (OS) is a primary bone cancer that is highly prevalent among adolescents and adults below the age of 20 years. The prognostic outcome of metastatic OS or relapse is extremely poor; thus, developing new diagnostic and therapeutic strategies for treating OS is necessary. Extracellular vesicles (EVs) ranging from 30–150 nm in diameter are commonly produced in different cells and are found in various types of body fluids. EVs are rich in biologically active components like proteins, lipids, and nucleic acids. They also strongly affect pathophysiological processes by modulating the intercellular signaling pathways and the exchange of biomolecules. Many studies have found that EVs influence the occurrence, development, and metastasis of osteosarcoma. The regulation of inflammatory communication pathways by EVs affects OS and other bone-related pathological conditions, such as osteoarthritis and rheumatoid arthritis. In this study, we reviewed the latest findings related to diagnosis, prognosis prediction, and the development of treatment strategies for OS from the perspective of EVs.
Collapse
Affiliation(s)
- Xiaozhuo Gao
- Department of Pathology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
| | - Bo Gao
- Department of Pathology, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
| | - Shenglong Li
- Department of Bone and Soft Tissue Tumor Surgery, Liaoning Cancer Hospital & Institute, Cancer Hospital of Dalian University of Technology, Cancer Hospital of China Medical University, Shenyang, China
- *Correspondence: Shenglong Li, ;
| |
Collapse
|
214
|
Gomes ER, Carvalho AT, Barbosa TC, Ferreira LL, Calado HDR, Sabino AP, Oliveira MC. Fusion of Tumor-Derived Exosomes with Long-Circulating and pH-Sensitive Liposomes Loaded with Doxorubicin for the Treatment of Breast Cancer. AAPS PharmSciTech 2022; 23:255. [DOI: 10.1208/s12249-022-02349-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 06/28/2022] [Indexed: 12/15/2022] Open
|
215
|
AS1411 aptamer-functionalized exosomes in the targeted delivery of doxorubicin in fighting colorectal cancer. Biomed Pharmacother 2022; 155:113690. [PMID: 36099793 DOI: 10.1016/j.biopha.2022.113690] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/06/2022] [Accepted: 09/07/2022] [Indexed: 11/21/2022] Open
Abstract
Severe side effects of chemotherapy agents on vital organs are the major causes of cancer-related mortality, not merely cancer disease. Encapsulating chemotherapeutic molecules in nanocarriers is a justifiable solution in decreasing the risk of their side effects and boosting the efficiency of treatment. The present study has developed the doxorubicin (DOX)-loaded AS1411 (anti-nucleolin) aptamer surface-functionalized exosome (DOX-Apt-Exo) to treat colorectal cancer in both in-vitro and in-vivo experimental models. HEK293-derived exosomes were loaded with DOX through the incubation method with a nearly 13% encapsulation efficiency. Afterwards, the 5-terminal carboxyl group of AS1411-aptamer was converted into amine-reactive NHS esters with EDC/NHS amide coupling chemistry before being conjugated to the amine groups on the exosome surface. DLS and TEM estimated the designed formulation (DOX-Apt-Exo) size of about 200 nm. Aptamer-binding affinity and cellular uptake of DOX-Apt-Exo by nucleolin-overexpressing cancer cells were depicted through fluorescence microscopy. Comparing the in-vitro cytotoxicity impact of DOX-loaded exosomes, either targeted or non-targeted by MTT assay, clearly verified a high effectiveness of ligand-receptor mediated target therapy. Subsequently, in-vivo experiments which were conducted on four groups of ectopic mouse models of colon cancer (5 in each group) demonstrated the tumor growth suppression through professional long-term accumulation and retention of DOX-Apt-Exo at the tumor site by ligand-receptor interaction. The results suggested that AS1411 aptamer-functionalized exosomes can be recommended as a safe and effective system to site-specific drug delivery in possible clinical applications of colon cancer.
Collapse
|
216
|
Ahmadi M, Hassanpour M, Rezaie J. Engineered extracellular vesicles: A novel platform for cancer combination therapy and cancer immunotherapy. Life Sci 2022; 308:120935. [PMID: 36075472 DOI: 10.1016/j.lfs.2022.120935] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 08/25/2022] [Accepted: 09/03/2022] [Indexed: 12/19/2022]
Abstract
Extracellular vesicles (EVs), phospholipid membrane-bound vesicles, produced by most cells, contribute to cell-cell communication. They transfer several proteins, lipids, and nucleic acids between cells both locally and systemically. Owing to the biocompatibility and immune activity of EVs, therapeutic approaches using these vesicles as drug delivery systems are being developed. Different methods are used to design more effective engineered EVs, which can serve as smart tools in cancer therapy and immunotherapy. Recent progress in the field of targeted-cancer therapy has led to the gradual use of engineered EVs in combinational therapy to combat heterogeneous tumor cells and multifaceted tumor microenvironments. The high plasticity, loading ability, and genetic manipulation capability of engineered EVs have made them the ideal platforms to realize numerous combinations of cancer therapy approaches. From the combination therapy view, engineered EVs can co-deliver chemotherapy with various therapeutic agents to target tumor cells effectively, further taking part in immunotherapy-related cancer combination therapy. However, a greater number of studies were done in pre-clinical platforms and the clinical translation of these studies needs further scrutiny because some challenges are associated with the application of engineered EVs. Given the many therapeutic potentials of engineered EVs, this review discusses their function in various cancer combination therapy and immunotherapy-related cancer combination therapy. In addition, this review describes the opportunities and challenges associated with the clinical application of engineered EVs.
Collapse
Affiliation(s)
- Mahdi Ahmadi
- Drug Applied Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mehdi Hassanpour
- Department of Biochemistry and Clinical Laboratories, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran; Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Jafar Rezaie
- Solid Tumor Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran.
| |
Collapse
|
217
|
Extracellular vesicles as an emerging drug delivery system for cancer treatment: Current strategies and recent advances. Biomed Pharmacother 2022; 153:113480. [DOI: 10.1016/j.biopha.2022.113480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/19/2022] Open
|
218
|
Aarsund M, Segers FM, Wu Y, Inngjerdingen M. Comparison of characteristics and tumor targeting properties of extracellular vesicles derived from primary NK cells or NK-cell lines stimulated with IL-15 or IL-12/15/18. Cancer Immunol Immunother 2022; 71:2227-2238. [PMID: 35119498 PMCID: PMC9374793 DOI: 10.1007/s00262-022-03161-0] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 01/19/2022] [Indexed: 12/17/2022]
Abstract
NK cell-based therapies have shown promise for hematological cancer forms, but their use against solid tumors is hampered by their poor ability to infiltrate the tumor. NK cells release extracellular vesicles (EVs) containing cytolytic proteins, indicating that NK-cell derived EVs may have therapeutic potential. In this study, we compared the tumor-targeting potential of EVs derived from either primary NK cells or the NK cell lines NK-92 and KHYG-1 cultured in IL-15 alone or in combination with IL-12 and IL-18. Primary NK cells were also stimulated through the activating receptor CD16. Tumor cell apoptosis was measured using a panel of human colon, melanoma, glioblastoma, prostate, breast, and ovarian tumor cell line spheroids. NK cells or NK-92 cells stimulated with IL-12, IL-15, and IL-18 generated EVs with higher efficiency than EVs from resting cells, although similar amounts of EVs were produced under both conditions. Proteomic analysis indicated similar distribution of cytolytic proteins in EVs from primary NK cells and NK-92, but lower levels in KHYG-1 EVs that translated into poor capacity for KHYG-1 EVs at targeting tumor cell lines. Further, we show that CD16-stimulated NK cells release low amounts of EVs devoid of cytolytic proteins. Importantly, EVs from cytokine-stimulated NK cells penetrate into the spheroid core, and tumor spheroid susceptibility to NK-cell derived EVs was linked to differential expression of the NKG2D ligands MICA/B, which was blocked with an anti-NKG2D antibody. We conclude that EVs from activated primary NK cells or NK-92 cells has the best potential to infiltrate and target solid tumors.
Collapse
Affiliation(s)
- Miriam Aarsund
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Filip M Segers
- Department of Pharmacology, Clinic of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Yunjie Wu
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway
| | - Marit Inngjerdingen
- Department of Pharmacology, Institute of Clinical Medicine, University of Oslo, Oslo, Norway.
| |
Collapse
|
219
|
Dai X, Ye Y, He F. Emerging innovations on exosome-based onco-therapeutics. Front Immunol 2022; 13:865245. [PMID: 36119094 PMCID: PMC9473149 DOI: 10.3389/fimmu.2022.865245] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Accepted: 07/29/2022] [Indexed: 01/30/2023] Open
Abstract
Exosomes, nano-sized extracellular vesicles for intercellular communications, are gaining rapid momentum as a novel strategy for the diagnosis and therapeutics of a spectrum of diseases including cancers. Secreted by various cell sources, exosomes pertain numerous functionalities from their parental cells and have enhanced stability that enable them with many features favorable for clinical use and commercialization. This paper focuses on the possible roles of exosomes in cancer therapeutics and reviews current exosome-based innovations toward enhanced cancer management and challenges that limit their clinical translation. Importantly, this paper casts insights on how cold atmospheric plasma, an emerging anticancer strategy, may aid in innovations on exosome-based onco-therapeutics toward improved control over cancers.
Collapse
Affiliation(s)
- Xiaofeng Dai
- Wuxi School of Medicine, Jiangnan University, Wuxi, China
- CAPsoul Medical Biotechnology Company, Ltd., Beijing, China
- *Correspondence: Fule He, ; Yongju Ye,
| | - Yongju Ye
- Department of Gynaecology, Lishui Hospital of Traditional Chinese Medicine, Lishui, China
- *Correspondence: Fule He, ; Yongju Ye,
| | - Fule He
- Department of Gynaecology, Lishui Hospital of Traditional Chinese Medicine, Lishui, China
- *Correspondence: Fule He, ; Yongju Ye,
| |
Collapse
|
220
|
Takakura Y, Takahashi Y. Strategies for persistent retention of macromolecules and nanoparticles in the blood circulation. J Control Release 2022; 350:486-493. [PMID: 36029894 DOI: 10.1016/j.jconrel.2022.05.063] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 05/23/2022] [Indexed: 12/22/2022]
Abstract
The enhanced permeability and retention (EPR) effect has been the gold standard in developing drug delivery systems for passive tumor targeting. Although the importance of this concept remains unchanged, some controversies have arisen. In this review, various strategies for tumor targeting using macromolecules and nanoparticles based on the EPR effect are discussed from the viewpoint of pharmacokinetics. Overall, such strategies seek to retain therapeutic material in the blood circulation, which is a key factor for successful targeting. Strategies using macromolecules, including antibody-drug conjugates, serum albumin-based delivery systems, PEGylated recombinant proteins, and stealth liposomes as well as nanoparticle-based strategies such as those based on lipid nanoparticles, and polymeric micelles, have been discussed. The feasibility of small extracellular vesicles, a new class of nanosized delivery carriers, is also discussed.
Collapse
Affiliation(s)
- Yoshinobu Takakura
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan.
| | - Yuki Takahashi
- Department of Biopharmaceutics and Drug Metabolism, Graduate School of Pharmaceutical Sciences, Kyoto University, 46-29 Yoshidashimoadachi-cho, Sakyo-ku, Kyoto 606-8501, Japan
| |
Collapse
|
221
|
Wan Z, Gan X, Mei R, Du J, Fan W, Wei M, Yang G, Qin W, Zhu Z, Liu L. ROS triggered local delivery of stealth exosomes to tumors for enhanced chemo/photodynamic therapy. J Nanobiotechnology 2022; 20:385. [PMID: 35999549 PMCID: PMC9400243 DOI: 10.1186/s12951-022-01591-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Accepted: 08/10/2022] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Exosomes are recognized as effective platforms for targeted delivery for their high physicochemical stability and biocompatibility. However, most of the exosomes are inevitably and rapidly cleared by mononuclear phagocytic system (MPS) during cancer therapy. How to engineer exosome to enhance the delivery efficiency is being intensively explored. In this study, we have constructed mPEG2000-TK-CP05 decorated exosomes as effective delivery platforms to achieve enhanced photodynamic/chemical cancer therapy. RESULTS Exosomes were coated with CP05-TK-mPEG2000, in which CP05 is a peptide with high affinity to exosomal CD63 and TK could be cleaved by ROS. The resulted exosomes, namely stealth Exo, were electroporated to load RB (photosensitizer Rose Bengal) and Dox (Doxorubicin). We verified that the Stealth Exo@RB (Stealth Exo additionally loaded with RB) could escape MPS while accumulate in the tumor region efficiently in the xenograft model when laser irradiation conducted locally. Additionally, we revealed that the Stealth Exo serves as an efficient platform for Dox delivery. Dox, together with the RB mediated photodynamic therapy induce tumor cell damage synergistically in the tumor region. Moreover, the proposed switchable stealth exosomes minimized the dose of toxic Dox and thus allowed robust tumor immune response. CONCLUSIONS Our results indicated that the proposed Stealth Exo greatly improves both the accessibility and efficiency of drug delivery, with minimal chemical or genetic engineering. The proposed Stealth Exo serve as a promising and powerful drug delivery nanoplatform in cancer treatment.
Collapse
Affiliation(s)
- Zhuo Wan
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an, 710032, China
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an, 710032, China
| | - Xueqi Gan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, Chengdu, China
| | - Ruiyan Mei
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an, 710032, China
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an, 710032, China
| | - Jianbin Du
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Wen Fan
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an, 710032, China
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an, 710032, China
| | - Mengying Wei
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Guodong Yang
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Weiwei Qin
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an, 710032, China.
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an, 710032, China.
| | - Zhuoli Zhu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Sichuan, 610041, Chengdu, China.
| | - Li Liu
- Department of Hematology, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Shaanxi Department of National Clinical Research Center for Hematological Diseases, Xi'an, 710032, China.
- Clinical Medical Research Center for Hematological Diseases of Shaanxi Province, Xi'an, 710032, China.
| |
Collapse
|
222
|
Pollalis D, Kim D, Nair GKG, Kang C, Nanda AV, Lee SY. Intraocular RGD-Engineered Exosomes and Active Targeting of Choroidal Neovascularization (CNV). Cells 2022; 11:2573. [PMID: 36010651 PMCID: PMC9406786 DOI: 10.3390/cells11162573] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2022] [Revised: 08/02/2022] [Accepted: 08/12/2022] [Indexed: 11/16/2022] Open
Abstract
PURPOSE To assess the transretinal penetration of intravitreally injected retinal multicell-derived exosomes and to develop exosome-based active targeting of choroidal neovascularization (CNV) by bioengineering with ASL, which is composed of a membrane Anchor (BODIPY), Spacer (PEG), and targeting Ligands (cyclic RGD peptide). METHODS Retinal multicell-derived exosomes were recovered from a whole mouse retina using differential ultracentrifugation. Their size, number, and morphology were characterized using nanoparticle tracking analysis (NTA) and transmission electron microscopy (TEM). Exosome markers were confirmed using an exosome detection antibody array. Intravitreal injection of fluorescent (PKH-26)-labeled or engineered ASL exosomes (1 × 106 exosomes/μL) were given to the wild-type mouse or laser-induced CNV mouse model. Retinal uptake of exosomes was assessed by in vivo retinal imaging microscopy and histological staining with DAPI, GSA, and anti-integrin αv for retinal sections or choroid/RPE flat mounts. Active targeting of CNV was assessed by comparing retinal uptake between areas with and without CNV and by colocalization analysis of ASL exosomes with integrin αv within CNV. Staining with anti-F4/80, anti-ICAM-1, and anti-GFAP antibodies on retinal sections were performed to identify intracellular uptake of exosomes and immediate reactive retinal gliosis after exosome treatment. RESULTS An average of 2.1 × 109 particles/mL with a peak size of 140 nm exosomes were recovered. Rapid retinal penetration of intravitreally injected exosomes was confirmed by retinal imaging microscopy at 3 and 24 h post-injection. Intravitreally delivered PKH-26-labeled exosomes reached inner and outer retinal layers including IPL, INL, OPL, and ONL at 1 and 7 days post-injection. Intravitreally injected ASL exosomes were predominantly delivered to the area of CNV including ONL, RPE, and choroid in laser-induced CNV mouse models with 89.5% of colocalization with integrin αv. Part of exosomes was also taken intracellularly to vascular endothelial cells and macrophages. After intravitreal injection, neither naive exosomes nor ASL exosomes induced immediate reactive gliosis. CONCLUSIONS Intravitreally delivered retinal multicell-derived exosomes have good retinal penetration, and ASL modification of exosomes actively targets CNV with no immediate reactive gliosis. ASL exosomes have a great potential to serve as an intraocular drug delivery vehicle, allowing an active targeting strategy.
Collapse
Affiliation(s)
- Dimitrios Pollalis
- USC Roski Eye Institute, USC Ginsburg Institute for Biomedical Therapeutics, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Dongin Kim
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Gopa Kumar Gopinadhan Nair
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Changsun Kang
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Arjun V. Nanda
- College of Medicine, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| | - Sun Young Lee
- USC Roski Eye Institute, USC Ginsburg Institute for Biomedical Therapeutics, Department of Ophthalmology, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
- Department of Ophthalmology, Dean McGee Eye Institute, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
- Department of Physiology, University of Oklahoma Health Science Center, Oklahoma City, OK 73104, USA
| |
Collapse
|
223
|
Słomka A, Wang B, Mocan T, Horhat A, Willms AG, Schmidt-Wolf IGH, Strassburg CP, Gonzalez-Carmona MA, Lukacs-Kornek V, Kornek MT. Extracellular Vesicles and Circulating Tumour Cells - complementary liquid biopsies or standalone concepts? Theranostics 2022; 12:5836-5855. [PMID: 35966579 PMCID: PMC9373826 DOI: 10.7150/thno.73400] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2022] [Accepted: 07/06/2022] [Indexed: 12/11/2022] Open
Abstract
Liquid biopsies do promise a lot, but are they keeping it? In the past decade, additional novel biomarkers qualified to be called like that, of which, some took necessary hurdles resulting in FDA approval and clinical use. Some others are since a while around, well known and were once regarded to be a game changer in cancer diagnosis or cancer screening. But, during their clinical use limitations were observed from statistical significance and questions raised regarding their robustness, that eventually led to be dropped from associated clinical guidelines for certain applications including cancer diagnosis. The purpose of this review isn't to give a broad overview of all current liquid biopsy as biomarkers, weight them and promise a brighter future in cancer prevention, but rather to take a deeper look on two of those who do qualify to be called liquid biopsies now or then. These two are probably of greatest interest conceptually and methodically, and likely have the highest chances to be in clinical use soon, with a portfolio extension over their original conceptual usage. We aim to dig deeper beyond cancer diagnosis or cancer screening. Actually, we aim to review in depth extracellular vesicles (EVs) and compare with circulating tumour cells (CTCs). The latter methodology is partially FDA approved and in clinical use. We will lay out similarities as taking advantage of surface antigens on EVs and CTCs in case of characterization and quantification. But drawing readers' attention to downstream application based on capture/isolation methodology and simply on their overall nature, here apparently being living material eventually recoverable as CTCs are vs. dead material with transient effects on recipient cell as in case of EVs. All this we try to bring in perspective, compare and conclude towards which future direction we are aiming for, or should aim for. Do we announce a winner between CTCs vs EVs? No, but we provide good reasons to intensify research on them.
Collapse
Affiliation(s)
- Artur Słomka
- Department of Pathophysiology, Nicolaus Copernicus University in Toruń, Ludwik Rydygier Collegium Medicum in Bydgoszcz, 85-067 Bydgoszcz, Poland
| | - Bingduo Wang
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany.,Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Tudor Mocan
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Adelina Horhat
- Octavian Fodor Institute for Gastroenterology and Hepatology, Iuliu Haţieganu, University of Medicine and Pharmacy, 400162 Cluj-Napoca, Romania
| | - Arnulf G Willms
- Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany.,Department of General, Visceral and Vascular Surgery, German Armed Forces Hospital Hamburg, 22049 Hamburg, Germany
| | - Ingo G H Schmidt-Wolf
- Department of Integrated Oncology, Center for Integrated Oncology (CIO), University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Christian P Strassburg
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Maria A Gonzalez-Carmona
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Veronika Lukacs-Kornek
- Institute of Molecular Medicine & Experimental Immunology, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| | - Miroslaw T Kornek
- Department of Internal Medicine I, University Hospital Bonn of the Rheinische Friedrich-Wilhelms-University, 53127 Bonn, Germany
| |
Collapse
|
224
|
Fang Z, Ding Y, Xue Z, Li P, Li J, Li F. Roles of exosomes as drug delivery systems in cancer immunotherapy: a mini-review. Discov Oncol 2022; 13:74. [PMID: 35962862 PMCID: PMC9375799 DOI: 10.1007/s12672-022-00539-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 08/08/2022] [Indexed: 11/04/2022] Open
Abstract
Exosomes can be released by a variety of cells and participate in intercellular communication in many physiological processes in the body. They can be used as carriers of cancer therapeutic drugs and have natural delivery capabilities. Some biologically active substances on exosomes, such as major histocompatibility complex (MHC), have been shown to be involved in exosome-mediated anticancer immune responses and have important regulatory effects on the immune system. Exosome-based drug delivery systems hold great promise in future cancer immunotherapy. However, there are still substantial challenges to be overcome in the clinical application of exosomes as drug carriers. This article reviews the biological characteristics of exosome drug delivery systems and their potential applications and challenges in cancer immunotherapy.
Collapse
Affiliation(s)
- Zhen Fang
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Yixuan Ding
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Zhigang Xue
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China
| | - Peijuan Li
- Dalian Medical University, Dalian, Liaoning, China.
| | - Jia Li
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital of Capital Medical University, Beijing, 100053, China.
| |
Collapse
|
225
|
Khan FH, Reza MJ, Shao YF, Perwez A, Zahra H, Dowlati A, Abbas A. Role of exosomes in lung cancer: A comprehensive insight from immunomodulation to theragnostic applications. Biochim Biophys Acta Rev Cancer 2022; 1877:188776. [PMID: 35961620 DOI: 10.1016/j.bbcan.2022.188776] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Revised: 07/18/2022] [Accepted: 08/02/2022] [Indexed: 12/18/2022]
Abstract
Exosomes are 30 to 150 nm-diameter lipid bilayer-enclosed extracellular vesicles that enable cell-to-cell communication through secretion and uptake. The exosomal cargoes contain RNA, lipids, proteins, and metabolites which can be delivered to recipient cells in vivo. In a healthy lung, exosomes facilitate interaction between adaptive and innate immunity and help maintain normal lung physiology. However, tumor-derived exosomes in lung cancer (LC) can, on the other hand, restrict immune cell proliferation, cause apoptosis in activated CD8+ T effector cells, reduce natural killer cell activity, obstruct monocyte differentiation, and promote proliferation of myeloid-derived suppressor and regulatory T cells. In addition, exosomes in the tumor microenvironment may also play a critical role in cancer progression and the development of drug resistance. In this review, we aim to comprehensively examine the current updates on the role of exosomes in lung carcinogenesis and their potential application as a diagnostic, prognostic, and therapeutic tool in lung cancer.
Collapse
Affiliation(s)
- Faizan Haider Khan
- Discipline of Pathology, Lambe Institute for Translational Research, School of Medicine, National University of Ireland Galway, Galway, Ireland
| | - Malik Johid Reza
- College of Pharmacy, University of Nebraska Medical Center, Omaha, NE 68131, USA
| | - Yusra Fatima Shao
- Department of Oncology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | - Ahmad Perwez
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Honey Zahra
- Department of Anatomy, King George's Medical University, Lucknow, UP 226003, India
| | - Afshin Dowlati
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; University Hospitals Seidman Cancer Center, Cleveland, OH 44106, USA; Developmental Therapeutics Program, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44116, USA.
| | - Ata Abbas
- Division of Hematology and Oncology, Department of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA; Developmental Therapeutics Program, Case Comprehensive Cancer Center, Case Western Reserve University School of Medicine, Cleveland, OH 44116, USA.
| |
Collapse
|
226
|
Dutta A, Paul S. Advancement in exosome-based cancer therapeutics: A new era in cancer treatment. FRONTIERS IN NANOTECHNOLOGY 2022. [DOI: 10.3389/fnano.2022.939197] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
In the modern era of rapid development and advancement in cancer therapeutics and management, there is a growing awareness in the application of exosomes as a potential tool to target cancer cells. Exosomes are cell-derived nano-vesicles that modulate intercellular communications and transport. Due to their ideal native structure and characteristics, exosomes have emerged as a promising nanocarrier for clinical use. Nevertheless, their medical application is coupled with some intrinsic restrictions which hinder their widespread use. In order to make exosomes more effective, they are engineered at the cellular level to develop designer exosomes. The focus of this review is to summarize the various exosome bio-engineering approaches aimed at the development of designer exosomes and their application in cancer treatment.
Collapse
|
227
|
Cheng J, Sun Y, Ma Y, Ao Y, Hu X, Meng Q. Engineering of MSC-Derived Exosomes: A Promising Cell-Free Therapy for Osteoarthritis. MEMBRANES 2022; 12:membranes12080739. [PMID: 36005656 PMCID: PMC9413347 DOI: 10.3390/membranes12080739] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 02/06/2023]
Abstract
Osteoarthritis (OA) is characterized by progressive cartilage degeneration with increasing prevalence and unsatisfactory treatment efficacy. Exosomes derived from mesenchymal stem cells play an important role in alleviating OA by promoting cartilage regeneration, inhibiting synovial inflammation and mediating subchondral bone remodeling without the risk of immune rejection and tumorigenesis. However, low yield, weak activity, inefficient targeting ability and unpredictable side effects of natural exosomes have limited their clinical application. At present, various approaches have been applied in exosome engineering to regulate their production and function, such as pretreatment of parental cells, drug loading, genetic engineering and surface modification. Biomaterials have also been proved to facilitate efficient delivery of exosomes and enhance treatment effectiveness. Here, we summarize the current understanding of the biogenesis, isolation and characterization of natural exosomes, and focus on the large-scale production and preparation of engineered exosomes, as well as their therapeutic potential in OA, thus providing novel insights into exploring advanced MSC-derived exosome-based cell-free therapy for the treatment of OA.
Collapse
Affiliation(s)
- Jin Cheng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Yixin Sun
- Peking Unversity First Hospital, Peking University Health Science Center, Beijing 100034, China;
| | - Yong Ma
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Yingfang Ao
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
| | - Xiaoqing Hu
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
- Correspondence: (X.H.); (Q.M.); Tel.: +86-010-8226-5680 (Q.M.)
| | - Qingyang Meng
- Department of Sports Medicine, Peking University Third Hospital, Institute of Sports Medicine of Peking University, Beijing Key Laboratory of Sports Injuries, Beijing 100191, China; (J.C.); (Y.M.); (Y.A.)
- Correspondence: (X.H.); (Q.M.); Tel.: +86-010-8226-5680 (Q.M.)
| |
Collapse
|
228
|
Flores de los Rios PA, Casañas Pimentel RG, San Martín Martínez E. Nanodrugs against cancer: biological considerations in its redesign. INT J POLYM MATER PO 2022. [DOI: 10.1080/00914037.2022.2097680] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Affiliation(s)
- P. A. Flores de los Rios
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada Legaria 694, Irrigación, Ciudad de México, México
| | - R. G. Casañas Pimentel
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada Legaria 694, Irrigación, Ciudad de México, México
| | - E. San Martín Martínez
- Instituto Politécnico Nacional, Centro de Investigación en Ciencia Aplicada y Tecnología Avanzada Legaria 694, Irrigación, Ciudad de México, México
| |
Collapse
|
229
|
The Exosome Journey: From Biogenesis to Regulation and Function in Cancers. JOURNAL OF ONCOLOGY 2022; 2022:9356807. [PMID: 35898929 PMCID: PMC9313905 DOI: 10.1155/2022/9356807] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/25/2022] [Revised: 05/01/2022] [Accepted: 06/20/2022] [Indexed: 12/26/2022]
Abstract
Exosomes are a type of small endosomal-derived vesicles ranging from 30 to 150 nm, which can serve as functional mediators in cell-to-cell communication and various physiological and pathological processes. In recent years, exosomes have emerged as crucial mediators of intracellular communication among tumor cells, immune cells, and stromal cells, which can shuttle bioactive molecules, such as proteins, lipids, RNA, and DNA. Exosomes exhibit the high bioavailability, biological stability, targeting specificity, low toxicity, and immune characteristics, suggesting their potentials in the diagnosis and treatment of cancers. They can be applied as an effective tool in the diagnostics, therapeutics, and drug delivery in cancers. This review summarizes the regulation and functions of exosomes in various cancers to augment our understanding of exosomes, which paves the way for parallel advancements in the therapeutic approach of cancers. In this review, we also discuss the challenges and prospects for clinical application of exosome-based diagnostics and therapeutics for cancers.
Collapse
|
230
|
Bie N, Yong T, Wei Z, Gan L, Yang X. Extracellular vesicles for improved tumor accumulation and penetration. Adv Drug Deliv Rev 2022; 188:114450. [PMID: 35841955 DOI: 10.1016/j.addr.2022.114450] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/26/2022] [Accepted: 07/06/2022] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs), including microparticles and exosomes, have emerged as potential tools for tumor targeting delivery during the past years. Recently, mass of strategies are applied to assist EVs to accumulate and penetrate into deep tumor sites. In this review, EVs from different cells with unique innate characters and engineered approaches (e.g. chemical engineering, genetical engineering and biomimetic engineering) as drug delivery systems to enhance tumor accumulation and penetration are summarized. Meanwhile, efficient biological function modulation (e.g. extracellular matrix degradation, mechanical property regulation and transcytosis) is introduced to facilitate tumor accumulation and penetration of EVs. Finally, the prospects and challenges on further clinical applications of EVs are discussed.
Collapse
Affiliation(s)
- Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China; Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Huazhong University of Science and Technology, Wuhan 430074, China.
| |
Collapse
|
231
|
Paskeh MDA, Entezari M, Mirzaei S, Zabolian A, Saleki H, Naghdi MJ, Sabet S, Khoshbakht MA, Hashemi M, Hushmandi K, Sethi G, Zarrabi A, Kumar AP, Tan SC, Papadakis M, Alexiou A, Islam MA, Mostafavi E, Ashrafizadeh M. Emerging role of exosomes in cancer progression and tumor microenvironment remodeling. J Hematol Oncol 2022; 15:83. [PMID: 35765040 PMCID: PMC9238168 DOI: 10.1186/s13045-022-01305-4] [Citation(s) in RCA: 313] [Impact Index Per Article: 104.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2022] [Accepted: 06/13/2022] [Indexed: 12/14/2022] Open
Abstract
Cancer is one of the leading causes of death worldwide, and the factors responsible for its progression need to be elucidated. Exosomes are structures with an average size of 100 nm that can transport proteins, lipids, and nucleic acids. This review focuses on the role of exosomes in cancer progression and therapy. We discuss how exosomes are able to modulate components of the tumor microenvironment and influence proliferation and migration rates of cancer cells. We also highlight that, depending on their cargo, exosomes can suppress or promote tumor cell progression and can enhance or reduce cancer cell response to radio- and chemo-therapies. In addition, we describe how exosomes can trigger chronic inflammation and lead to immune evasion and tumor progression by focusing on their ability to transfer non-coding RNAs between cells and modulate other molecular signaling pathways such as PTEN and PI3K/Akt in cancer. Subsequently, we discuss the use of exosomes as carriers of anti-tumor agents and genetic tools to control cancer progression. We then discuss the role of tumor-derived exosomes in carcinogenesis. Finally, we devote a section to the study of exosomes as diagnostic and prognostic tools in clinical courses that is important for the treatment of cancer patients. This review provides a comprehensive understanding of the role of exosomes in cancer therapy, focusing on their therapeutic value in cancer progression and remodeling of the tumor microenvironment.
Collapse
Affiliation(s)
- Mahshid Deldar Abad Paskeh
- 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
| | - 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
| | - Sepideh Mirzaei
- Department of Biology, Faculty of Science, Islamic Azad University, Science and Research Branch, Tehran, Iran
| | - Amirhossein Zabolian
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Hossein Saleki
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohamad Javad Naghdi
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Sina Sabet
- Young Researchers and Elite Club, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Mohammad Amin Khoshbakht
- Young Researchers and Elite Club, 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
| | - Kiavash Hushmandi
- Division of Epidemiology, Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Iran
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, 34396, Istanbul, Turkey
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Shing Cheng Tan
- UKM Medical Molecular Biology Institute, Universiti Kebangsaan Malaysia, Kuala Lumpur, Malaysia.
| | - Marios Papadakis
- Department of Surgery II, University Hospital Witten-Herdecke, University of Witten-Herdecke, Heusnerstrasse 40, 42283, Wuppertal, Germany.
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, Australia
- AFNP Med Austria, Vienna, Austria
| | - Md Asiful Islam
- Department of Haematology, School of Medical Sciences, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
- Institute of Metabolism and Systems Research, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ebrahim Mostafavi
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Milad Ashrafizadeh
- Faculty of Engineering and Natural Sciences, Sabanci University, Orta Mahalle, Üniversite Caddesi No. 27, Orhanlı, Tuzla, Istanbul, Turkey.
| |
Collapse
|
232
|
Ye J, Liu X. Macrophage-Derived Small Extracellular Vesicles in Multiple Diseases: Biogenesis, Function, and Therapeutic Applications. Front Cell Dev Biol 2022; 10:913110. [PMID: 35832790 PMCID: PMC9271994 DOI: 10.3389/fcell.2022.913110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 05/20/2022] [Indexed: 12/24/2022] Open
Abstract
Macrophages (Mφs), as immune cells, play a pivotal role against pathogens and many diseases, such as cancer, inflammation, cardiovascular diseases, orthopedic diseases, and metabolic disorders. In recent years, an increasing number of studies have shown that small extracellular vesicles (sEVs) derived from Mφs (M-sEVs) play important roles in these diseases, suggesting that Mφs carry out their physiological functions through sEVs. This paper reviews the mechanisms underlying M-sEVs production via different forms of polarization and their biological functions in multiple diseases. In addition, the prospects of M-sEVs in disease diagnosis and treatment are described.
Collapse
Affiliation(s)
- Jingyao Ye
- Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xuehong Liu
- The Third School of Clinical Medicine of Guangzhou University of Chinese Medicine, Guangzhou, China
- *Correspondence: Xuehong Liu,
| |
Collapse
|
233
|
Liu H, Huang Y, Huang M, Huang Z, Wang Q, Qing L, Li L, Xu S, Jia B. Current Status, Opportunities, and Challenges of Exosomes in Oral Cancer Diagnosis and Treatment. Int J Nanomedicine 2022; 17:2679-2705. [PMID: 35733418 PMCID: PMC9208818 DOI: 10.2147/ijn.s365594] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/01/2022] [Indexed: 12/13/2022] Open
Abstract
Oral cancer is one of the most common cancers in the world, with more than 300,000 cases diagnosed each year, of which oral squamous cell carcinoma accounts for more than 90%, with a 5-year survival rate of only 40–60%, and poor prognosis. Exploring new strategies for the early diagnosis and treatment of oral cancer is key to improving the survival rate. Exosomes are nanoscale lipid bilayer membrane vesicles that are secreted by almost all cell types. During the development of oral cancer, exosomes can transport their contents (DNA, RNA, proteins, etc) to target cells and promote or inhibit the proliferation, invasion, and metastasis of oral cancer cells by influencing the host immune response, drug-resistant metastasis, and tumour angiogenesis. Therefore, exosomes have great potential and advantages as biomarkers for oral cancer diagnosis, and as drug delivery vehicles or targets for oral cancer therapy. In this review, we first describe the biogenesis, biological functions, and isolation methods of exosomes, followed by their relationship with oral cancer. Here, we focused on the potential of exosomes as oral cancer biomarkers, drug carriers, and therapeutic targets. Finally, we provide an insightful discussion of the opportunities and challenges of exosome application in oral cancer diagnosis and treatment, intending to offer new ideas for the clinical management of oral cancer.
Collapse
Affiliation(s)
- Hongyu Liu
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Yisheng Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Mingshu Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Zhijie Huang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Qin Wang
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Ling Qing
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Li Li
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Shuaimei Xu
- Department of Endodontics, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| | - Bo Jia
- Department of Oral Surgery, Stomatological Hospital, Southern Medical University, Guangzhou, People's Republic of China
| |
Collapse
|
234
|
Pompili S, Vetuschi A, Sferra R, Cappariello A. Extracellular Vesicles and Resistance to Anticancer Drugs: A Tumor Skeleton Key for Unhinging Chemotherapies. Front Oncol 2022; 12:933675. [PMID: 35814444 PMCID: PMC9259994 DOI: 10.3389/fonc.2022.933675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 05/20/2022] [Indexed: 11/13/2022] Open
Abstract
Although surgical procedures and clinical care allow reaching high success in fighting most tumors, cancer is still a formidable foe. Recurrence and metastatization dampen the patients’ overall survival after the first diagnosis; nevertheless, the large knowledge of the molecular bases drives these aspects. Chemoresistance is tightly linked to these features and is mainly responsible for the failure of cancer eradication, leaving patients without a crucial medical strategy. Many pathways have been elucidated to trigger insensitiveness to drugs, generally associated with the promotion of tumor growth, aggressiveness, and metastatisation. The main mechanisms reported are the expression of transporter proteins, the induction or mutations of oncogenes and transcription factors, the alteration in genomic or mitochondrial DNA, the triggering of autophagy or epithelial-to-mesenchymal transition, the acquisition of a stem phenotype, and the activation of tumor microenvironment cells. Extracellular vesicles (EVs) can directly transfer or epigenetically induce to a target cell the molecular machinery responsible for the acquisition of resistance to drugs. In this review, we resume the main body of knowledge supporting the crucial role of EVs in the context of chemoresistance, with a particular emphasis on the mechanisms related to some of the main drugs used to fight cancer.
Collapse
|
235
|
Zhao Y, Haney MJ, Fallon JK, Rodriguez M, Swain CJ, Arzt CJ, Smith PC, Loop MS, Harrison EB, El-Hage N, Batrakova EV. Using Extracellular Vesicles Released by GDNF-Transfected Macrophages for Therapy of Parkinson Disease. Cells 2022; 11:1933. [PMID: 35741061 PMCID: PMC9222008 DOI: 10.3390/cells11121933] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 06/09/2022] [Accepted: 06/11/2022] [Indexed: 02/04/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nanoparticles that facilitate transport of proteins, lipids, and genetic material, playing important roles in intracellular communication. They have remarkable potential as non-toxic and non-immunogenic nanocarriers for drug delivery to unreachable organs and tissues, in particular, the central nervous system (CNS). Herein, we developed a novel platform based on macrophage-derived EVs to treat Parkinson disease (PD). Specifically, we evaluated the therapeutic potential of EVs secreted by autologous macrophages that were transfected ex vivo to express glial-cell-line-derived neurotrophic factor (GDNF). EV-GDNF were collected from conditioned media of GDNF-transfected macrophages and characterized for GDNF content, size, charge, and expression of EV-specific proteins. The data revealed that, along with the encoded neurotrophic factor, EVs released by pre-transfected macrophages carry GDNF-encoding DNA. Four-month-old transgenic Parkin Q311(X)A mice were treated with EV-GDNF via intranasal administration, and the effect of this therapeutic intervention on locomotor functions was assessed over a year. Significant improvements in mobility, increases in neuronal survival, and decreases in neuroinflammation were found in PD mice treated with EV-GDNF. No offsite toxicity caused by EV-GDNF administration was detected. Overall, an EV-based approach can provide a versatile and potent therapeutic intervention for PD.
Collapse
Affiliation(s)
- Yuling Zhao
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.Z.); (M.J.H.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Matthew J. Haney
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.Z.); (M.J.H.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - John K. Fallon
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Myosotys Rodriguez
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (N.E.-H.)
| | - Carson J. Swain
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Camryn J. Arzt
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Philip C. Smith
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Matthew Shane Loop
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Emily B. Harrison
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| | - Nazira El-Hage
- Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA; (M.R.); (N.E.-H.)
| | - Elena V. Batrakova
- Center for Nanotechnology in Drug Delivery, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (Y.Z.); (M.J.H.)
- Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, NC 27599, USA; (J.K.F.); (C.J.S.); (C.J.A.); (P.C.S.); (M.S.L.); (E.B.H.)
| |
Collapse
|
236
|
Zhang X, Wu Y, Cheng Q, Bai L, Huang S, Gao J. Extracellular Vesicles in Cardiovascular Diseases: Diagnosis and Therapy. Front Cell Dev Biol 2022; 10:875376. [PMID: 35721498 PMCID: PMC9198246 DOI: 10.3389/fcell.2022.875376] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Accepted: 05/13/2022] [Indexed: 12/20/2022] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of global mortality. Therapy of CVDs is still a great challenge since many advanced therapies have been developed. Multiple cell types produce nano-sized extracellular vesicles (EVs), including cardiovascular system-related cells and stem cells. Compelling evidence reveals that EVs are associated with the pathophysiological processes of CVDs. Recently researches focus on the clinical transformation in EVs-based diagnosis, prognosis, therapies, and drug delivery systems. In this review, we firstly discuss the current knowledge about the biophysical properties and biological components of EVs. Secondly, we will focus on the functions of EVs on CVDs, and outline the latest advances of EVs as prognostic and diagnostic biomarkers, and therapeutic agents. Finally, we will introduce the specific application of EVs as a novel drug delivery system and its application in CVDs therapy. Specific attention will be paid to summarize the perspectives, challenges, and applications on EVs’ clinical and industrial transformation.
Collapse
Affiliation(s)
- Xiaojing Zhang
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
- *Correspondence: Xiaojing Zhang, ; Jun Gao,
| | - Yuping Wu
- Department of Scientific Research, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Qifa Cheng
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
| | - Liyang Bai
- Department of Clinical Medicine, The Third Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Shuqiang Huang
- Department of Clinical Medicine, The Sixth Clinical School of Guangzhou Medical University, Guangzhou, China
| | - Jun Gao
- Department of Pharmacy, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People’s Hospital, Qingyuan, China
- *Correspondence: Xiaojing Zhang, ; Jun Gao,
| |
Collapse
|
237
|
Jokar S, Marques IA, Khazaei S, Martins-Marques T, Girao H, Laranjo M, Botelho MF. The Footprint of Exosomes in the Radiation-Induced Bystander Effects. Bioengineering (Basel) 2022; 9:bioengineering9060243. [PMID: 35735486 PMCID: PMC9220715 DOI: 10.3390/bioengineering9060243] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 05/07/2022] [Accepted: 05/26/2022] [Indexed: 12/12/2022] Open
Abstract
Radiation therapy is widely used as the primary treatment option for several cancer types. However, radiation therapy is a nonspecific method and associated with significant challenges such as radioresistance and non-targeted effects. The radiation-induced non-targeted effects on nonirradiated cells nearby are known as bystander effects, while effects far from the ionising radiation-exposed cells are known as abscopal effects. These effects are presented as a consequence of intercellular communications. Therefore, a better understanding of the involved intercellular signals may bring promising new strategies for radiation risk assessment and potential targets for developing novel radiotherapy strategies. Recent studies indicate that radiation-derived extracellular vesicles, particularly exosomes, play a vital role in intercellular communications and may result in radioresistance and non-targeted effects. This review describes exosome biology, intercellular interactions, and response to different environmental stressors and diseases, and focuses on their role as functional mediators in inducing radiation-induced bystander effect (RIBE).
Collapse
Affiliation(s)
- Safura Jokar
- Department of Nuclear Pharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran P94V+927, Iran;
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (I.A.M.); (M.L.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (T.M.-M.); (H.G.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
| | - Inês A. Marques
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (I.A.M.); (M.L.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (T.M.-M.); (H.G.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Centre of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Faculty of Pharmacy, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Saeedeh Khazaei
- Department of Pharmaceutical Biomaterials, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran P94V+927, Iran;
| | - Tania Martins-Marques
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (T.M.-M.); (H.G.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Henrique Girao
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (T.M.-M.); (H.G.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Mafalda Laranjo
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (I.A.M.); (M.L.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (T.M.-M.); (H.G.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Centre of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
| | - Maria Filomena Botelho
- Institute of Biophysics, Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (I.A.M.); (M.L.)
- Coimbra Institute for Clinical and Biomedical Research (iCBR), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal; (T.M.-M.); (H.G.)
- Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, 3000-548 Coimbra, Portugal
- Centre of Investigation in Environment, Genetics and Oncobiology (CIMAGO), Faculty of Medicine, University of Coimbra, 3000-548 Coimbra, Portugal
- Clinical and Academic Centre of Coimbra (CACC), 3004-561 Coimbra, Portugal
- Correspondence:
| |
Collapse
|
238
|
Barone A, d’Avanzo N, Cristiano MC, Paolino D, Fresta M. Macrophage-Derived Extracellular Vesicles: A Promising Tool for Personalized Cancer Therapy. Biomedicines 2022; 10:1252. [PMID: 35740274 PMCID: PMC9220135 DOI: 10.3390/biomedicines10061252] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2022] [Revised: 05/23/2022] [Accepted: 05/25/2022] [Indexed: 12/12/2022] Open
Abstract
The incidence of cancer is increasing dramatically, affecting all ages of the population and reaching an ever higher worldwide mortality rate. The lack of therapies' efficacy is due to several factors such as a delay in diagnosis, tumor regrowth after surgical resection and the occurrence of multidrug resistance (MDR). Tumor-associated immune cells and the tumor microenvironment (TME) deeply affect the tumor's progression, leading to several physicochemical changes compared to physiological conditions. In this scenario, macrophages play a crucial role, participating both in tumor suppression or progression based on the polarization of onco-suppressive M1 or pro-oncogenic M2 phenotypes. Moreover, much evidence supports the pivotal role of macrophage-derived extracellular vesicles (EVs) as mediators in TME, because of their ability to shuttle the cell-cell and organ-cell communications, by delivering nucleic acids and proteins. EVs are lipid-based nanosystems with a broad size range distribution, which reflect a similar composition of native parent cells, thus providing a natural selectivity towards target sites. In this review, we discuss the impact of macrophage-derived EVs in the cancer's fate as well as their potential implications for the development of personalized anticancer nanomedicine.
Collapse
Affiliation(s)
- Antonella Barone
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy; (A.B.); (M.C.C.)
| | - Nicola d’Avanzo
- Department of Pharmacy, University “G. d’Annunzio” of Chieti-Pescara, Via dei Vestini n.31, 66100 Chieti, Italy;
| | - Maria Chiara Cristiano
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy; (A.B.); (M.C.C.)
| | - Donatella Paolino
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy; (A.B.); (M.C.C.)
| | - Massimo Fresta
- Department of Health Science, University “Magna Græcia” of Catanzaro Campus Universitario-Germaneto, Viale Europa, 88100 Catanzaro, Italy;
| |
Collapse
|
239
|
Yang L, Huang S, Zhang Z, Liu Z, Zhang L. Roles and Applications of Red Blood Cell-Derived Extracellular Vesicles in Health and Diseases. Int J Mol Sci 2022; 23:ijms23115927. [PMID: 35682606 PMCID: PMC9180222 DOI: 10.3390/ijms23115927] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 12/10/2022] Open
Abstract
Red blood cell-derived extracellular vesicles (RBCEVs) are vesicles naturally produced by red blood cells and play multiple roles such as acting as cell-to-cell communication messengers in both normal physiological and diseased states. RBCEVs are highly promising delivery vehicles for therapeutic agents such as biomolecules and nucleic acids as they are easy to source, safe, and versatile. RBCEVs autonomously target the liver and pass the blood-brain barrier into the brain, which is highly valuable for the treatment of liver and brain diseases. RBCEVs can be modified by various functional units, including various functional molecules and nanoparticles, to improve their active targeting capabilities for tumors or other sites. Moreover, the RBCEV level is significantly shifted in many diseased states; hence, they can also serve as important biomarkers for disease diagnoses. It is clear that RBCEVs have considerable potential in multiple medical applications. In this review, we briefly introduce the biological roles of RBCEVs, presented interesting advances in RBCEV applications, and discuss several challenges that need to be addressed for their clinical translation.
Collapse
Affiliation(s)
- Lan Yang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (L.Y.); (S.H.); (Z.Z.)
| | - Shiqi Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (L.Y.); (S.H.); (Z.Z.)
| | - Zhirong Zhang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China; (L.Y.); (S.H.); (Z.Z.)
| | - Zhenmi Liu
- Med-X Center for Materials, West China School of Public Health, Sichuan University, Chengdu 610041, China;
| | - Ling Zhang
- Med-X Center for Materials, College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
- Correspondence:
| |
Collapse
|
240
|
Xu Y, Fourniols T, Labrak Y, Préat V, Beloqui A, des Rieux A. Surface Modification of Lipid-Based Nanoparticles. ACS NANO 2022; 16:7168-7196. [PMID: 35446546 DOI: 10.1021/acsnano.2c02347] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
There is a growing interest in the development of lipid-based nanocarriers for multiple purposes, including the recent increase of these nanocarriers as vaccine components during the COVID-19 pandemic. The number of studies that involve the surface modification of nanocarriers to improve their performance (increase the delivery of a therapeutic to its target site with less off-site accumulation) is enormous. The present review aims to provide an overview of various methods associated with lipid nanoparticle grafting, including techniques used to separate grafted nanoparticles from unbound ligands or to characterize grafted nanoparticles. We also provide a critical perspective on the usefulness and true impact of these modifications on overcoming different biological barriers, with our prediction on what to expect in the near future in this field.
Collapse
Affiliation(s)
- Yining Xu
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Thibaut Fourniols
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Yasmine Labrak
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
- Bioanalysis and Pharmacology of Bioactive Lipids, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 72 B1.72.01, 1200 Brussels, Belgium
| | - Véronique Préat
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Ana Beloqui
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| | - Anne des Rieux
- Advanced Drug Delivery and Biomaterials, UCLouvain, Université Catholique de Louvain, Louvain Drug Research Institute, Avenue Mounier, 73 B1.73.12, 1200 Brussels, Belgium
| |
Collapse
|
241
|
Chen M, Li Y, Ma N, Zang J. Mesenchymal stem cell‑derived exosomes loaded with 5‑Fu against cholangiocarcinoma in vitro. Mol Med Rep 2022; 25:213. [PMID: 35543159 PMCID: PMC9133964 DOI: 10.3892/mmr.2022.12729] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Accepted: 04/19/2022] [Indexed: 11/06/2022] Open
Abstract
Cholangiocarcinoma (CCA) is an intractable malignant tumour with a high degree of malignancy that is asymptomatic in the early stages. Exosomes have been shown in numerous studies in recent years to be effective delivery vehicles for chemotherapy drugs to suppress tumour proliferation and growth in vivo and in vitro. In order to explore the inhibition of 5-fluorouracil (5-Fu)-loaded exosomes on CCA growth, the present study used human bone marrow mesenchymal stem cell-derived exosomes, as well as incubation and sonication methods for 5-Fu loading into exosomes, to treat CCA in vitro. The findings demonstrated that exosomes isolated from mesenchymal stem cells have typical exosome characteristics. Both the incubation and sonication methods successfully loaded 5-Fu into the exosomes (5-Fu-Exos), with the sonication method having a higher loading efficiency than the incubation method. When compared to the free 5-Fu group, the 5-Fu-Exos group significantly inhibited the viability of CCA cells (P<0.01), indicating that 5-Fu-Exos can be an effective chemotherapy drug for CCA treatment.
Collapse
Affiliation(s)
- Mingzheng Chen
- Department of Medicine, Graduate School, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Yangyang Li
- Department of Medicine, Graduate School, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Ningfu Ma
- Department of Medicine, Graduate School, Dalian Medical University, Dalian, Liaoning 116044, P.R. China
| | - Jinfeng Zang
- Department of Hepatobiliary Surgery, Taizhou People's Hospital, The Fifth Affiliated Hospital of The Medical School of Nantong University, Taizhou, Jiangsu 225300, P.R. China
| |
Collapse
|
242
|
The anti-angiogenic effect of atorvastatin loaded exosomes on glioblastoma tumor cells: An in vitro 3D culture model. Microvasc Res 2022; 143:104385. [DOI: 10.1016/j.mvr.2022.104385] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 05/03/2022] [Accepted: 05/18/2022] [Indexed: 01/10/2023]
|
243
|
Pirisinu M, Pham TC, Zhang DX, Hong TN, Nguyen LT, Le MT. Extracellular vesicles as natural therapeutic agents and innate drug delivery systems for cancer treatment: Recent advances, current obstacles, and challenges for clinical translation. Semin Cancer Biol 2022; 80:340-355. [PMID: 32977006 DOI: 10.1016/j.semcancer.2020.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 08/11/2020] [Accepted: 08/12/2020] [Indexed: 12/13/2022]
Abstract
As cancer poses a significant threat to the well-being of humans on a global scale, many researchers have embarked on the search for effective anticancer therapeutic agents. In recent years, many drugs have been shown to have extraordinary anticancer effects. However, in a lot of cases the treatment is accompanied by undesirable side effects due to some intrinsic properties linked to the therapeutic agents, such as poor targeting selectivity and short half-life in the circulation. In this regard, extracellular vesicles (EVs), a diverse family of natural cell-derived vesicles, steal the show as potential anticancer immunotherapy or delivery vectors of anticancer agents since they are an innate mechanism of intercellular communication. Here, we describe some of the most hotly-debated issues regarding the use of EVs as anticancer therapeutics. First, we review the biology of EVs providing the most up-to-date definition of EVs as well as highlighting their circulation kinetics and homing properties. Next, we share our views on popular methods reported for EV isolation, characterization, and functional analysis. Pioneering and innovative reports along with emerging challenges in the field of EV imaging and EV drug loading strategies are then discussed. Finally, we examine in detail the therapeutic application of EVs in cancer treatment, including their role in cancer immunotherapy and as natural delivery systems for anticancer agents including natural compounds such as paclitaxel and doxorubicin. We consider standardised protocols and proper analytical approaches to be crucial in improving the reproducibility and rigor in EV research and ensuring the successful translation of EVs as anticancer therapeutics.
Collapse
Affiliation(s)
- Marco Pirisinu
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR of China, China
| | - Tin Chanh Pham
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR of China, China
| | - Daniel Xin Zhang
- Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR of China, China; Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA
| | - Tran Nguyen Hong
- Department of Pharmacology and Biochemistry, Vietnam Institute of Medicinal Materials, Hanoi, Vietnam
| | - Lap Thi Nguyen
- Department of Biochemistry, Hanoi University of Pharmacy, Hanoi, Vietnam
| | - Minh Tn Le
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore; Department of Biomedical Sciences, Jockey Club College of Veterinary Medicine and Life Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR of China, China.
| |
Collapse
|
244
|
Wang T, Fu Y, Sun S, Huang C, Yi Y, Wang J, Deng Y, Wu M. Exosome-based drug delivery systems in cancer therapy. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.05.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
245
|
Liu H, Geng Z, Su J. Engineered mammalian and bacterial extracellular vesicles as promising nanocarriers for targeted therapy. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2022; 3:63-86. [PMID: 39698442 PMCID: PMC11648430 DOI: 10.20517/evcna.2022.04] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/21/2022] [Accepted: 04/07/2022] [Indexed: 12/20/2024]
Abstract
Extracellular vesicles (EVs), which are nanocarriers with phospholipid bilayer structures released by most cells, play a key role in regulating physiological and pathological processes. EVs have been investigated due to their loading capacity, low toxicity, immunogenicity, and biofunctions. Although EVs have shown good potential as therapeutic vehicles, natural EVs have a poor targeting ability, which substantially reduces the therapeutic effect. Through the addition of a targeting unit into the membrane surface of EVs or inside EVs by engineering technology, the therapeutic agent can accumulate in specific cells and tissues. Here, we focus on mammalian EVs (MEVs) and bacterial EVs (BEVs), which are the two most common types of EVs in the biomedical field. In this review, we describe engineered MEVs and BEVs as promising nanocarriers for targeted therapy and summarize the biogenesis, isolation, and characterization of MEVs and BEVs. We then describe engineering techniques for enhancement of the targeting ability of EVs. Moreover, we focus on the applications of engineered MEVs and BEVs in targeted therapy, including the treatment of cancer and brain and bone disease. We believe that this review will help improve the understanding of engineered MEVs and BEVs, thereby promoting their application and clinical translation.
Collapse
Affiliation(s)
| | | | - Jiacan Su
- Correspondence to: Prof. Jiacan Su, Institute of Translational Medicine, Shanghai University, Shanghai 200444, China. E-mail:
| |
Collapse
|
246
|
Zhang Q, Ding J, Wang Y, He L, Xue F. Tumor microenvironment manipulates chemoresistance in ovarian cancer (Review). Oncol Rep 2022; 47:102. [PMID: 35362546 DOI: 10.3892/or.2022.8313] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/22/2022] [Indexed: 11/05/2022] Open
Abstract
Ovarian cancer (OC) is the leading cause of mortality among the various types of gynecological cancer, and >75% of the cases are diagnosed at a late stage. Although platinum‑based chemotherapy is able to help the majority of patients to achieve remission, the disease frequently recurs and acquires chemoresistance, resulting in high mortality rates. The complexity of OC therapy is not solely governed by the intrinsic characteristics of the OC cells (OCCs) themselves, but is also largely dependent on the dynamic communication between OCCs and various components of their surrounding microenvironment. The present review attempts to describe the mutual interplay between OCCs and their surrounding microenvironment. Tumor‑associated macrophages (TAMs) and cancer‑associated fibroblasts (CAFs) are the most abundant stromal cell types in OC. Soluble factors derived from CAFs steadily nourish both the OCCs and TAMs, facilitating their proliferation and immune evasion. ATP binding cassette transporters facilitate the extrusion of cytotoxic molecules, eventually promoting cell survival and multidrug resistance. Extracellular vesicles fulfill their role as genetic exchange vectors, transferring cargo from the donor cells to the recipient cells and propagating oncogenic signaling. A greater understanding of the vital roles of the tumor microenvironment will allow researchers to be open to the prospect of developing therapeutic approaches for combating OC chemoresistance.
Collapse
Affiliation(s)
- Qiaoling Zhang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Jiashan Ding
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Yingmei Wang
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| | - Linsheng He
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi 330006, P.R. China
| | - Fengxia Xue
- Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin 300052, P.R. China
| |
Collapse
|
247
|
Zeng W, Wen Z, Chen H, Duan Y. Exosomes as Carriers for Drug Delivery in Cancer Therapy. Pharm Res 2022; 40:873-887. [PMID: 35352281 DOI: 10.1007/s11095-022-03224-y] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Accepted: 03/03/2022] [Indexed: 12/17/2022]
Abstract
Exosomes are extracellular vesicles secreted by cells with a particle size of 30-150 nm in diameter. Exosomes can be used as natural drug carriers. The treatment of cancer with drug-loaded exosomes is an area of high interest. This review introduces the composition, function, isolation and characterization of exosomes, and briefly describes the selection of exosome donor cells and methods for drug loading. Through studies on therapies with drug-loaded exosomes in gastric cancer, lung cancer, brain cancer and other cancers, the advantages and disadvantages of drug-loaded exosomes have been analyzed.
Collapse
Affiliation(s)
- Weiping Zeng
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou, 510006, People's Republic of China
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Zhengbo Wen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China
| | - Honglin Chen
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, People's Republic of China.
| | - Yuyou Duan
- Laboratory of Stem Cells and Translational Medicine, Institutes for Life Sciences, School of Medicine, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou, 510006, People's Republic of China.
- Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou, 510006, People's Republic of China.
| |
Collapse
|
248
|
Allegra A, Petrarca C, Di Gioacchino M, Casciaro M, Musolino C, Gangemi S. Exosome-Mediated Therapeutic Strategies for Management of Solid and Hematological Malignancies. Cells 2022; 11:cells11071128. [PMID: 35406692 PMCID: PMC8997895 DOI: 10.3390/cells11071128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/23/2022] [Accepted: 03/24/2022] [Indexed: 12/24/2022] Open
Abstract
Exosomes are small membrane vesicles of endocytic origin containing cytokines, RNAs, growth factors, proteins, lipids, and metabolites. They have been identified as fundamental intercellular communication controllers in several diseases and an enormous volume of data confirmed that exosomes could either sustain or inhibit tumor onset and diffusion in diverse solid and hematological malignancies by paracrine signaling. Thus, exosomes might constitute a promising cell-free tumor treatment alternative. This review focuses on the effects of exosomes in the treatment of tumors, by discussing the most recent and promising data from in vitro and experimental in vivo studies and the few existing clinical trials. Exosomes are extremely promising as transporters of drugs, antagomir, genes, and other therapeutic substances that can be integrated into their core via different procedures. Moreover, exosomes can augment or inhibit non-coding RNAs, change the metabolism of cancer cells, and modify the function of immunologic effectors thus modifying the tumor microenvironment transforming it from pro-tumor to antitumor milieu. Here, we report the development of currently realized exosome modifiers that offer indications for the forthcoming elaboration of other more effective methods capable of enhancing the activity of the exosomes.
Collapse
Affiliation(s)
- Alessandro Allegra
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
- Correspondence: (A.A.); (M.D.G.)
| | - Claudia Petrarca
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Department of Medicine and Aging Sciences, G. D’Annunzio University, 66100 Chieti, Italy
| | - Mario Di Gioacchino
- Center for Advanced Studies and Technology, G. D’Annunzio University, 66100 Chieti, Italy;
- Institute for Clinical Immunotherapy and Advanced Biological Treatments, 65100 Pescara, Italy
- Correspondence: (A.A.); (M.D.G.)
| | - Marco Casciaro
- Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| | - Caterina Musolino
- Division of Hematology, Department of Human Pathology in Adulthood and Childhood “Gaetano Barresi”, University of Messina, 98125 Messina, Italy;
| | - Sebastiano Gangemi
- Unit of Allergy and Clinical Immunology, Department of Clinical and Experimental Medicine, School of Allergy and Clinical Immunology, University of Messina, 98125 Messina, Italy; (M.C.); (S.G.)
| |
Collapse
|
249
|
Chemically Modified Extracellular Vesicles and Applications in Radiolabeling and Drug Delivery. Pharmaceutics 2022; 14:pharmaceutics14030653. [PMID: 35336027 PMCID: PMC8955996 DOI: 10.3390/pharmaceutics14030653] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Revised: 02/28/2022] [Accepted: 03/11/2022] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) have been exploited as bio-inspired drug delivery systems (DDS) in the biomedical field. EVs have more advantages than synthetic nanoparticles: they are naturally equipped to cross extra- and intra-cellular barriers. Furthermore, they can deliver functional biomolecules from one cell to another even far away in the body. These advantages, along with obtained promising in vivo results, clearly evidenced the potential of EVs in drug delivery. Nevertheless, due to the difficulties of finding a chemical approach that is coherent with EVs’ rational clinical therapeutic use, those in the drug delivery community are expecting more from EVs’ use. Therefore, this review gathered knowledge of the current chemical approaches dealing with the conjugation of EVs for drugs and radiotracers.
Collapse
|
250
|
Li Y, Bao Q, Yang S, Yang M, Mao C. Bionanoparticles in cancer imaging, diagnosis, and treatment. VIEW 2022. [DOI: 10.1002/viw.20200027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Affiliation(s)
- Yan Li
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou Zhejiang China
| | - Qing Bao
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang China
| | - Shuxu Yang
- Department of Neurosurgery Sir Run Run Shaw Hospital School of Medicine Zhejiang University Hangzhou Zhejiang China
| | - Mingying Yang
- Institute of Applied Bioresource Research College of Animal Science Zhejiang University Hangzhou Zhejiang China
| | - Chuanbin Mao
- School of Materials Science and Engineering Zhejiang University Hangzhou Zhejiang China
- Department of Chemistry and Biochemistry Stephenson Life Science Research Center University of Oklahoma Norman Oklahoma USA
| |
Collapse
|