1
|
Simon L, Constanzo J, Terraza-Aguirre C, Ibn Elfekih Z, Berthelot J, Benkhaled BT, Haute T, Pednekar K, Clark K, Emerson SJ, Atis S, Benedetti C, Langlois S, Marquant A, Prakash J, Wang A, Devoisselle JM, Montier T, Djouad F, Pouget JP, Lapinte V, Morille M. Surface modification of extracellular vesicles with polyoxazolines to enhance their plasma stability and tumor accumulation. Biomaterials 2025; 313:122748. [PMID: 39180918 DOI: 10.1016/j.biomaterials.2024.122748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 07/23/2024] [Accepted: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Extracellular vesicles (EVs) are future promising therapeutics, but their instability in vivo after administration remains an important barrier to their further development. Many groups evaluated EV surface modification strategies to add a targeting group with the aim of controlling EV biodistribution. Conversely, fewer groups focused on their stabilization to obtain "stealth" allogenic EVs. Modulating their stabilization and biodistribution is an essential prerequisite for their development as nano-therapeutics. Here, we explored polyoxazolines with lipid anchors association to the EV membrane (POxylation as an alternative to PEGylation) to stabilize EVs in plasma and control their biodistribution, while preserving their native properties. We found that this modification maintained and seemed to potentiate the immunomodulatory properties of EVs derived from mesenchymal stem/stromal cells (MSC). Using a radiolabeling protocol to track EVs at a therapeutically relevant concentration in vivo, we demonstrated that POxylation is a promising option to stabilize EVs in plasma because it increased EV half-life by 6 fold at 6 h post-injection. Moreover, EV accumulation in tumors was higher after POxylation than after PEGylation.
Collapse
Affiliation(s)
- L Simon
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Constanzo
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | | | - Z Ibn Elfekih
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Berthelot
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - B T Benkhaled
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Haute
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France
| | - K Pednekar
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - K Clark
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S J Emerson
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - S Atis
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - C Benedetti
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - S Langlois
- Montpellier Ressources Imagerie, BioCampus, University of Montpellier, CNRS, INSERM, Montpellier, France
| | - A Marquant
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - J Prakash
- Department of Advanced Organ Bioengineering and Therapeutics, Engineered Therapeutics Section, Technical Medical Centre, University of Twente, 7500 AE, Enschede, the Netherlands
| | - A Wang
- Center for Surgical Bioengineering, Deparment of Surgery, University of California Davis School of Medicine, Sacramento, CA, USA; Institute for Pediatric Regenerative Medicine, Shriners Children's Northern California, Sacramento, CA, USA
| | - J M Devoisselle
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - T Montier
- Univ Brest, INSERM, EFS, UMR 1078, GGB, F-29200, Brest, France; CHU de Brest, Service de Génétique Médicale et de Biologie de La Reproduction, Centre de Référence des Maladies Rares Maladies Neuromusculaires, 29200, Brest, France
| | - F Djouad
- IRMB, University of Montpellier, INSERM, 34295, Montpellier, France; Clinical Immunology and Osteoarticular Disease Therapeutic Unit, Department of Rheumatology, CHU Montpellier, 34095, Montpellier, France
| | - J P Pouget
- Institut de Recherche en Cancérologie de Montpellier (IRCM), INSERM U1194, Université de Montpellier, Institut Régional Du Cancer de Montpellier (ICM), Montpellier, France
| | - V Lapinte
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France
| | - Marie Morille
- ICGM, Montpellier University, CNRS, ENSCM, Montpellier, France; Institut Universitaire de France (IUF), Paris, France.
| |
Collapse
|
2
|
Bobbili MR, Görgens A, Yan Y, Vogt S, Gupta D, Corso G, Barbaria S, Patrioli C, Weilner S, Pultar M, Jacak J, Hackl M, Schosserer M, Grillari R, Kjems J, Andaloussi SEL, Grillari J. Snorkel-tag based affinity chromatography for recombinant extracellular vesicle purification. J Extracell Vesicles 2024; 13:e12523. [PMID: 39400515 PMCID: PMC11472238 DOI: 10.1002/jev2.12523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/12/2024] [Indexed: 10/15/2024] Open
Abstract
Extracellular vesicles (EVs) are lipid nanoparticles and play an important role in cell-cell communications, making them potential therapeutic agents and allowing to engineer for targeted drug delivery. The expanding applications of EVs in next generation medicine is still limited by existing tools for scaling standardized EV production, single EV tracing and analytics, and thus provide only a snapshot of tissue-specific EV cargo information. Here, we present the Snorkel-tag, for which we have genetically fused the EV surface marker protein CD81, to a series of tags with an additional transmembrane domain to be displayed on the EV surface, resembling a snorkel. This system enables the affinity purification of EVs from complex matrices in a non-destructive form while maintaining EV characteristics in terms of surface protein profiles, associated miRNA patterns and uptake into a model cell line. Therefore, we consider the Snorkel-tag to be a widely applicable tool in EV research, allowing for efficient preparation of EV standards and reference materials, or dissecting EVs with different surface markers when fusing to other tetraspanins in vitro or in vivo.
Collapse
Affiliation(s)
- Madhusudhan Reddy Bobbili
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue Regeneration
| | - André Görgens
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
- Institute for Transfusion Medicine, University Hospital EssenUniversity of Duisburg‐EssenEssenGermany
| | - Yan Yan
- Department of Molecular Biology and Genetics, Centre for Cellular Signal Patterns (CellPat), Interdisciplinary Nanoscience Centre (iNANO)Aarhus UniversityAarhus CDenmark
- Omiics ApSAarhus NDenmark
| | - Stefan Vogt
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Dhanu Gupta
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Institute of Developmental and Regenerative MedicineUniversity of Oxford, IMS‐Tetsuya Nakamura Building, Old Road Campus, Roosevelt Dr, HeadingtonOxfordUnited Kingdom
- Department of PaediatricsUniversity of Oxford, South Parks RoadOxfordUnited Kingdom
| | - Giulia Corso
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Evercyte GmbHViennaAustria
| | - Samir Barbaria
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Carolina Patrioli
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | - Sylvia Weilner
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
| | | | - Jaroslaw Jacak
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- School of Medical Engineering and Applied Social ScienceUniversity of Applied Sciences Upper AustriaLinzAustria
| | - Matthias Hackl
- Austrian Cluster for Tissue Regeneration
- TAmiRNATAmiRNA GmbHViennaAustria
| | - Markus Schosserer
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Austrian Cluster for Tissue Regeneration
- Institute of Medical GeneticsCenter for Pathobiochemistry and GeneticsMedical University of ViennaViennaAustria
| | - Regina Grillari
- Austrian Cluster for Tissue Regeneration
- Evercyte GmbHViennaAustria
| | - Jørgen Kjems
- Department of Molecular Biology and Genetics, Centre for Cellular Signal Patterns (CellPat), Interdisciplinary Nanoscience Centre (iNANO)Aarhus UniversityAarhus CDenmark
| | - Samir EL Andaloussi
- Department of Laboratory Medicine, Division of Biomolecular and Cellular MedicineKarolinska InstitutetStockholmSweden
- Department of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST)Karolinska University Hospital Huddinge and Karolinska Comprehensive Cancer CenterStockholmSweden
| | - Johannes Grillari
- Institute of Molecular Biotechnology, Department of BiotechnologyBOKU UniversityViennaAustria
- Ludwig Boltzmann Institute for TraumatologyThe Research Center in Cooperation with AUVAViennaAustria
- Austrian Cluster for Tissue Regeneration
| |
Collapse
|
3
|
Rosenkrans ZT, Thickens AS, Kink JA, Aluicio-Sarduy E, Engle JW, Hematti P, Hernandez R. Investigating the In Vivo Biodistribution of Extracellular Vesicles Isolated from Various Human Cell Sources Using Positron Emission Tomography. Mol Pharm 2024; 21:4324-4335. [PMID: 39164886 DOI: 10.1021/acs.molpharmaceut.4c00298] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/22/2024]
Abstract
Positron emission tomography (PET) is a powerful tool for investigating the in vivo behavior of drug delivery systems. We aimed to assess the biodistribution of extracellular vesicles (EVs), nanosized vesicles secreted by cells isolated from various human cell sources using PET. EVs were isolated from mesenchymal stromal cells (MSCs) (MSC EVs), human macrophages (Mϕ EVs), and a melanoma cell line (A375 EVs) by centrifugation and were conjugated with deferoxamine for radiolabeling with Zr-89. PET using conjugated and radiolabeled EVs evaluated their in vivo biodistribution and tissue tropisms. Our study also investigated differences in mouse models, utilizing immunocompetent and immunocompromised mice and an A375 xenograft tumor model. Lastly, we investigated the impact of different labeling techniques on the observed EV biodistribution, including covalent surface modification and membrane incorporation. PET showed that all tested EVs exhibited extended in vivo circulation and generally low uptake in the liver, spleen, and lungs. However, Mϕ EVs showed high liver uptake, potentially attributable to the intrinsic tissue tropism of these EVs from the surface protein composition. MSC EV biodistribution differed between immunocompetent and immunodeficient mice, with increased spleen uptake observed in the latter. PET using A375 xenografts demonstrated efficient tumor uptake of EVs, but no preferential tissue-specific tropism of A375 EVs was found. Biodistribution differences between labeling techniques showed that surface-conjugated EVs had preferential blood circulation and low liver, spleen, and lung uptake compared to membrane integration. This study demonstrates the potential of EVs as effective drug carriers for various diseases, highlights the importance of selecting appropriate cell sources for EV-based drug delivery, and suggests that EV tropism can be harnessed to optimize therapeutic efficacy. Our findings indicate that the cellular source of EVs, labeling technique, and animal model can influence the observed biodistribution.
Collapse
Affiliation(s)
- Zachary T Rosenkrans
- Departments of Medical Physics and Radiology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Anna S Thickens
- Departments of Medical Physics and Radiology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, Madison, Wisconsin 53705, United States
| | - John A Kink
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 600 Highland Avenue, Madison, Wisconsin 53792, United States
| | - Eduardo Aluicio-Sarduy
- Departments of Medical Physics and Radiology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
| | - Jonathan W Engle
- Departments of Medical Physics and Radiology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 600 Highland Avenue, Madison, Wisconsin 53792, United States
| | - Peiman Hematti
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, Madison, Wisconsin 53705, United States
- University of Wisconsin Carbone Cancer Center, University of Wisconsin-Madison, 600 Highland Avenue, Madison, Wisconsin 53792, United States
- Division of Hematology and Oncology, Medical College of Wisconsin, 9200 W. Wisconsin Avenue, Milwaukee, Wisconsin 53226, United States
| | - Reinier Hernandez
- Departments of Medical Physics and Radiology, University of Wisconsin-Madison, 1111 Highland Avenue, Madison, Wisconsin 53705, United States
- Department of Medicine, University of Wisconsin-Madison School of Medicine and Public Health, 1685 Highland Avenue, Madison, Wisconsin 53705, United States
| |
Collapse
|
4
|
Gangadaran P, Khan F, Rajendran RL, Onkar A, Goenka A, Ahn BC. Unveiling Invisible Extracellular Vesicles: Cutting-Edge Technologies for Their in Vivo Visualization. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e2009. [PMID: 39439198 DOI: 10.1002/wnan.2009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 09/11/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024]
Abstract
Extracellular vesicles (EVs), nanosized lipid bilayer vesicles released by nearly all types of cells, play pivotal roles as intercellular signaling mediators with diverse biological activities. Their adaptability has attracted interest in exploring their role as disease biomarker theranostics. However, the in vivo biodistribution and pharmacokinetic profiles of EVs, particularly following administration into living subjects, remain unclear. Thus, in vivo imaging is vital to enhance our understanding of the homing and retention patterns, blood and tissue half-life, and excretion pathways of exogenous EVs, thereby advancing real-time monitoring within biological systems and their therapeutic applications. This review examines state-of-the-art methods including EV labeling with various agents, including optical imaging, magnetic resonance imaging, and nuclear imaging. The strengths and weaknesses of each technique are comprehensively explored, emphasizing their clinical translation. Despite the potential of EVs as cancer theranostics, achieving a thorough understanding of their in vivo behavior is challenging. This review highlights the urgency of addressing current questions in the biology and therapeutic applications of EVs. It underscores the need for continued research to unravel the complexities surrounding EVs and their potential clinical implications. By identifying these challenges, this review contributes to ongoing efforts to optimize EV imaging techniques for clinical use. Ultimately, bridging the gap between research advancements and clinical applications will facilitate the integration of EV-based theranostics, marking a crucial step toward harnessing the full potential of EVs in medical practice.
Collapse
Affiliation(s)
- Prakash Gangadaran
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Fatima Khan
- Department of Cancer Biology, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| | - Ramya Lakshmi Rajendran
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, Republic of Korea
| | - Akanksha Onkar
- Department of Laboratory Medicine, University of California San Francisco, San Francisco, California, USA
| | - Anshika Goenka
- Department of Hematology and Medical Oncology, Winship Cancer Institute, Emory University, Atlanta, Georgia, USA
| | - Byeong-Cheol Ahn
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Science, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, Republic of Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, Republic of Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, Republic of Korea
| |
Collapse
|
5
|
Mazahir F, Yadav AK. Recent progress in engineered extracellular vesicles and their biomedical applications. Life Sci 2024; 350:122747. [PMID: 38797364 DOI: 10.1016/j.lfs.2024.122747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2024] [Revised: 05/14/2024] [Accepted: 05/23/2024] [Indexed: 05/29/2024]
Abstract
AIMS To present the recent update on the isolation, engineering techniques for extracellular vesicles, limitations associated with different isolation techniques, different biomedical applications, and challenges of engineered extracellular vesicles for the benefit of researchers from academic, industry, etc. MATERIALS AND METHODS: Peer-reviewed articles from most recognized journals were collected, and presented information was analyzed to discuss collection, chemical, electroporation, cellular, and membrane surface engineering to design extracellular vesicles for various therapeutic applications. In addition, we present the applications and limitations of techniques for the collection of extracellular vesicles. KEY FINDINGS There is a need for isolation techniques with the gold standard. However, advanced extracellular vesicle isolation techniques showed improved recovery, and purity of extracellular vesicles. Tumor therapy is a major part of the therapy section that illustrates the role of engineered extracellular vesicles in synergetic therapy such as phototherapy, theragnostic, and delivery of genetic materials. In addition, extracellular vesicles have shown their potential in the treatment of retinal disorders, neurodegenerative disease, tuberculosis, osteoporosis, inflammatory bowel disease, vaccine production, and wound healing. SIGNIFICANCE Engineered extracellular vesicles can deliver cargo to the specific cells, elicit an immune response and could be used for the development of the vaccines in the future. However, the progress is at the initial stage. Overall, this review will provide a comprehensive understanding and could serve as a reference for researchers in the clinical translation of engineered extracellular vesicles in different biomedical fields.
Collapse
Affiliation(s)
- Farhan Mazahir
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Raebareli, A Transit Campus, Bijnor-Sisendi Road, Bijnor, Lucknow-226002, India
| | - Awesh K Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research Raebareli, A Transit Campus, Bijnor-Sisendi Road, Bijnor, Lucknow-226002, India.
| |
Collapse
|
6
|
Yan Q, Liu H, Sun S, Yang Y, Fan D, Yang Y, Zhao Y, Song Z, Chen Y, Zhu R, Zhang Z. Adipose-derived stem cell exosomes loaded with icariin alleviates rheumatoid arthritis by modulating macrophage polarization in rats. J Nanobiotechnology 2024; 22:423. [PMID: 39026367 PMCID: PMC11256651 DOI: 10.1186/s12951-024-02711-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Accepted: 07/07/2024] [Indexed: 07/20/2024] Open
Abstract
Rheumatoid arthritis (RA) is a chronic autoimmune disease marked by synovitis and cartilage destruction. The active compound, icariin (ICA), derived from the herb Epimedium, exhibits potent anti-inflammatory properties. However, its clinical utility is limited by its water insolubility, poor permeability, and low bioavailability. To address these challenges, we developed a multifunctional drug delivery system-adipose-derived stem cells-exosomes (ADSCs-EXO)-ICA to target active macrophages in synovial tissue and modulate macrophage polarization from M1 to M2. High-performance liquid chromatography analysis confirmed a 92.4 ± 0.008% loading efficiency for ADSCs-EXO-ICA. In vitro studies utilizing cellular immunofluorescence (IF) and flow cytometry demonstrated significant inhibition of M1 macrophage proliferation by ADSCs-EXO-ICA. Enzyme-linked immunosorbent assay, cellular transcriptomics, and real-time quantitative PCR indicated that ADSCs-EXO-ICA promotes an M1-to-M2 phenotypic transition by reducing glycolysis through the inhibition of the ERK/HIF-1α/GLUT1 pathway. In vivo, ADSCs-EXO-ICA effectively accumulated in the joints. Pharmacodynamic assessments revealed that ADSCs-EXO-ICA decreased cytokine levels and mitigated arthritis symptoms in collagen-induced arthritis (CIA) rats. Histological analysis and micro computed tomography confirmed that ADSCs-EXO-ICA markedly ameliorated synovitis and preserved cartilage. Further in vivo studies indicated that ADSCs-EXO-ICA suppresses arthritis by promoting an M1-to-M2 switch and suppressing glycolysis. Western blotting supported the therapeutic efficacy of ADSCs-EXO-ICA in RA, confirming its role in modulating macrophage function through energy metabolism regulation. Thus, this study not only introduces a drug delivery system that significantly enhances the anti-RA efficacy of ADSCs-EXO-ICA but also elucidates its mechanism of action in macrophage function inhibition.
Collapse
Affiliation(s)
- Qiqi Yan
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Haixia Liu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Shiyue Sun
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yongsheng Yang
- Institute of Acupuncture and Moxibustion, China Academy of Chinese Medical Sciences, Beijing, China
| | - DanPing Fan
- Institute of Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yuqin Yang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yukun Zhao
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhiqian Song
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yanjing Chen
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ruyuan Zhu
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Zhiguo Zhang
- Institute of Basic Theory for Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
7
|
Wardhani K, Levina A, Grau GER, Lay PA. Fluorescent, phosphorescent, magnetic resonance contrast and radioactive tracer labelling of extracellular vesicles. Chem Soc Rev 2024; 53:6779-6829. [PMID: 38828885 DOI: 10.1039/d2cs00238h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/05/2024]
Abstract
This review focusses on the significance of fluorescent, phosphorescent labelling and tracking of extracellular vesicles (EVs) for unravelling their biology, pathophysiology, and potential diagnostic and therapeutic uses. Various labeling strategies, such as lipid membrane, surface protein, luminal, nucleic acid, radionuclide, quantum dot labels, and metal complex-based stains, are evaluated for visualizing and characterizing EVs. Direct labelling with fluorescent lipophilic dyes is simple but generally lacks specificity, while surface protein labelling offers selectivity but may affect EV-cell interactions. Luminal and nucleic acid labelling strategies have their own advantages and challenges. Each labelling approach has strengths and weaknesses, which require a suitable probe and technique based on research goals, but new tetranuclear polypyridylruthenium(II) complexes as phosphorescent probes have strong phosphorescence, selective staining, and stability. Future research should prioritize the design of novel fluorescent probes and labelling platforms that can significantly enhance the efficiency, accuracy, and specificity of EV labeling, while preserving their composition and functionality. It is crucial to reduce false positive signals and explore the potential of multimodal imaging techniques to gain comprehensive insights into EVs.
Collapse
Affiliation(s)
- Kartika Wardhani
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Biochemistry and Biotechnology (B-TEK) Group, Bioscience Division, Los Alamos National Laboratory, Los Alamos, New Mexico, 87545, USA
| | - Aviva Levina
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
| | - Georges E R Grau
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Vascular Immunology Unit, School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, 2006, Australia
| | - Peter A Lay
- School of Chemistry, The University of Sydney, Sydney, New South Wales, 2006, Australia.
- Sydney Nano, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Cancer Network, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Marie Bashir Institute, The University of Sydney, Sydney, New South Wales, 2006, Australia
- Sydney Analytical, The University of Sydney, Sydney, New South Wales, 2006, Australia
| |
Collapse
|
8
|
Liam-Or R, Faruqu FN, Walters A, Han S, Xu L, Wang JTW, Oberlaender J, Sanchez-Fueyo A, Lombardi G, Dazzi F, Mailaender V, Al-Jamal KT. Cellular uptake and in vivo distribution of mesenchymal-stem-cell-derived extracellular vesicles are protein corona dependent. NATURE NANOTECHNOLOGY 2024; 19:846-855. [PMID: 38366223 PMCID: PMC11186763 DOI: 10.1038/s41565-023-01585-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Accepted: 11/27/2023] [Indexed: 02/18/2024]
Abstract
Extracellular vesicles (EVs) derived from mesenchymal stem cells are promising nanotherapeutics in liver diseases due to their regenerative and immunomodulatory properties. Nevertheless, a concern has been raised regarding the rapid clearance of exogenous EVs by phagocytic cells. Here we explore the impact of protein corona on EVs derived from two culturing conditions in which specific proteins acquired from media were simultaneously adsorbed on the EV surface. Additionally, by incubating EVs with serum, simulating protein corona formation upon systemic delivery, further resolved protein corona-EV complex patterns were investigated. Our findings reveal the potential influences of corona composition on EVs under in vitro conditions and their in vivo kinetics. Our data suggest that bound albumin creates an EV signature that can retarget EVs from hepatic macrophages. This results in markedly improved cellular uptake by hepatocytes, liver sinusoidal endothelial cells and hepatic stellate cells. This phenomenon can be applied as a camouflage strategy by precoating EVs with albumin to fabricate the albumin-enriched protein corona-EV complex, enhancing non-phagocytic uptake in the liver. This work addresses a critical challenge facing intravenously administered EVs for liver therapy by tailoring the protein corona-EV complex for liver cell targeting and immune evasion.
Collapse
Affiliation(s)
- Revadee Liam-Or
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Farid N Faruqu
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
- Pharmacology Department, Faculty of Medicine, University of Malaya, Kuala Lumpur, Malaysia
| | - Adam Walters
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Shunping Han
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Lizhou Xu
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Julie Tzu-Wen Wang
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, UK
| | - Jennifer Oberlaender
- Max Planck Institute for Polymer Research, Mainz, Germany
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Alberto Sanchez-Fueyo
- Institute of Liver Studies, King's College London University and King's College Hospital, London, UK
| | - Giovanna Lombardi
- Peter Gorer Department of Immunobiology, School of Immunology and Microbial Sciences, King's College London, London, UK
| | - Francesco Dazzi
- Comprehensive Cancer Centre, King's College London, London, UK
| | - Volker Mailaender
- Max Planck Institute for Polymer Research, Mainz, Germany
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Khuloud T Al-Jamal
- Institute of Pharmaceutical Science, Faculty of Life Sciences & Medicine, King's College London, London, UK.
| |
Collapse
|
9
|
Shi Y, Wang S, Wang K, Yang R, Liu D, Liao H, Qi Y, Qiu K, Hu Y, Wen H, Xu K. Relieving Macrophage Dysfunction by Inhibiting SREBP2 Activity: A Hypoxic Mesenchymal Stem Cells-Derived Exosomes Loaded Multifunctional Hydrogel for Accelerated Diabetic Wound Healing. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309276. [PMID: 38247194 DOI: 10.1002/smll.202309276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 12/18/2023] [Indexed: 01/23/2024]
Abstract
Macrophage dysfunction is one of the primary factors leading to the delayed healing of diabetic wounds. Hypoxic bone marrow mesenchymal stem cells-derived exosomes (hyBMSC-Exos) have been shown to play an active role in regulating cellular function through the carried microRNAs. However, the administration of hyBMSC-Exos alone in diabetic wounds usually brings little effect, because the exosomes are inherently unstable and have a short retention time at the wounds. In this study, a multifunctional hydrogel based on gallic acid (GA) conjugated chitosan (Chi-GA) and partially oxidized hyaluronic acid (OHA) is prepared for sustained release of hyBMSC-Exos. The hydrogel not only exhibits needs-satisfying physicochemical properties, but also displays outstanding biological performances such as low hemolysis rate, strong antibacterial capacity, great antioxidant ability, and excellent biocompatibility. It has the ability to boost the stability of hyBMSC-Exos, leading to a continuous and gradual release of the exosomes at wound locations, ultimately enhancing the exosomes' uptake efficiency by target cells. Most importantly, hyBMSC-Exos loaded hydrogel shows an excellent ability to promote diabetic wound healing by regulating macrophage polarization toward M2 phenotype. This may be because exosomal miR-4645-5p and antioxidant property of the hydrogel synergistically inhibit SREBP2 activity in macrophages. This study presents a productive approach for managing diabetic wounds.
Collapse
Affiliation(s)
- Yan Shi
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Shang Wang
- Chongqing Key Laboratory of Traditional Chinese Medicine for Prevention and Cure of Metabolic Diseases, College of Traditional Chinese Medicine, Chongqing Medical University, Chongqing, 400016, P. R. China
| | - Kai Wang
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, P. R. China
| | - Ronghua Yang
- Department of Burn and Plastic Surgery, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, Guangdong, 510650, P. R. China
| | - Dewu Liu
- Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Huaiwei Liao
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yuhan Qi
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Keqing Qiu
- Dermatological Department, The Second Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Yanghong Hu
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, 330006, P. R. China
| | - Huicai Wen
- Department of Plastic, Medical Center of Burn Plastic and Wound Repair, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, 330006, P. R. China
| | - Kui Xu
- Key Laboratory of Xin'an Medicine, Ministry of Education, Anhui University of Chinese Medicine, Hefei, Anhui, 230038, P. R. China
| |
Collapse
|
10
|
Han R, He H, Lu Y, Lu H, Shen S, Wu W. Oral targeted drug delivery to post-gastrointestinal sites. J Control Release 2024; 370:256-276. [PMID: 38679163 DOI: 10.1016/j.jconrel.2024.04.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/21/2024] [Accepted: 04/25/2024] [Indexed: 05/01/2024]
Abstract
As an essential branch of targeted drug delivery, oral targeted delivery is attracting growing attention in recent years. In addition to site-specific delivery for the treatment of locoregional diseases in the gastrointestinal tract (GIT), oral targeted delivery to remote sites beyond the GIT emerges as a cutting-edge research topic. This review aims to provide an overview of the fundamental concepts and most recent advances in this field. Owing to the physiological barriers existing in the GIT, carrier systems should be transported across the enteric epithelia to target remote sites. Recently, pioneer investigations have validated the transport of intact micro- or nanocarriers across gastrointestinal barriers and subsequently to various distal organs and tissues. The microfold (M) cell pathway is the leading mechanism underlying the oral absorption of particulates, but the contribution of the transcellular and paracellular pathways should not be neglected either. In addition to well-acknowledged physicochemical and biological factors, the formation of a protein corona may also influence the biological fate of carrier systems. Although in an early stage of conceptualization, oral targeted delivery to remote diseases has demonstrated promising potential for the treatment of inflammation, tumors, and diseases inflicting the lymphatic and mononuclear phagocytosis systems.
Collapse
Affiliation(s)
- Rongze Han
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Haisheng He
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Yi Lu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China; Fudan Zhangjiang Institute, Shanghai 201203, China
| | - Huiping Lu
- Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China
| | - Shun Shen
- Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China.
| | - Wei Wu
- Key Laboratory of Smart Drug Delivery of MOE, School of Pharmacy, Fudan University, Shanghai 201203, China; Pharmacy Department and Center for Medical Research and Innovation, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai 201399, China; Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai 200443, China; Fudan Zhangjiang Institute, Shanghai 201203, China.
| |
Collapse
|
11
|
Yao C, Zhang H, Wang C. Recent advances in therapeutic engineered extracellular vesicles. NANOSCALE 2024; 16:7825-7840. [PMID: 38533676 DOI: 10.1039/d3nr05470e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/28/2024]
Abstract
Extracellular vesicles (EVs) are natural particles secreted by living cells, which hold significant potential for various therapeutic applications. Native EVs have specific components and structures, allowing them to cross biological barriers, and circulate in vivo for a long time. Native EVs have also been bioengineered to enhance their therapeutic efficacy and targeting affinity. Recently, the therapeutic potential of surface-engineered EVs has been explored in the treatment of tumors, autoimmune diseases, infections and other diseases by ongoing research and clinical trials. In this review, we will introduce the modified methods of engineered EVs, summarize the application of engineered EVs in preclinical and clinical trials, and discuss the opportunities and challenges for the clinical translation of surface-engineered EVs.
Collapse
Affiliation(s)
- Chenlu Yao
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| | - Hong Zhang
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| | - Chao Wang
- Laboratory for Biomaterial and ImmunoEngineering, Institute of Functional Nano & Soft Materials (FUNSOM), Soochow University, Suzhou, Jiangsu 215123, China.
| |
Collapse
|
12
|
Yue Y, Dai W, Wei Y, Cao S, Liao S, Li A, Liu P, Lin J, Zeng H. Unlocking the potential of exosomes: a breakthrough in the theranosis of degenerative orthopaedic diseases. Front Bioeng Biotechnol 2024; 12:1377142. [PMID: 38699435 PMCID: PMC11064847 DOI: 10.3389/fbioe.2024.1377142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Accepted: 04/01/2024] [Indexed: 05/05/2024] Open
Abstract
Degenerative orthopaedic diseases pose a notable worldwide public health issue attributable to the global aging population. Conventional medical approaches, encompassing physical therapy, pharmaceutical interventions, and surgical methods, face obstacles in halting or reversing the degenerative process. In recent times, exosome-based therapy has gained widespread acceptance and popularity as an effective treatment for degenerative orthopaedic diseases. This therapeutic approach holds the potential for "cell-free" tissue regeneration. Exosomes, membranous vesicles resulting from the fusion of intracellular multivesicles with the cell membrane, are released into the extracellular matrix. Addressing challenges such as the rapid elimination of natural exosomes in vivo and the limitation of drug concentration can be effectively achieved through various strategies, including engineering modification, gene overexpression modification, and biomaterial binding. This review provides a concise overview of the source, classification, and preparation methods of exosomes, followed by an in-depth analysis of their functions and potential applications. Furthermore, the review explores various strategies for utilizing exosomes in the treatment of degenerative orthopaedic diseases, encompassing engineering modification, gene overexpression, and biomaterial binding. The primary objective is to provide a fresh viewpoint on the utilization of exosomes in addressing bone degenerative conditions and to support the practical application of exosomes in the theranosis of degenerative orthopaedic diseases.
Collapse
Affiliation(s)
- Yaohang Yue
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Wei Dai
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Yihao Wei
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Siyang Cao
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shuai Liao
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Aikang Li
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Peng Liu
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Jianjing Lin
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Sports Medicine and Rehabilitation, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Hui Zeng
- School of Clinical Medicine, Shandong Second Medical University, Weifang, Shandong, China
- Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Department of Bone and Joint Surgery, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- National and Local Joint Engineering Research Centre of Orthopaedic Biomaterials, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Key Laboratory of Orthopaedic Diseases and Biomaterials Research, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
- Shenzhen Second People’s Hospital, Shenzhen, Guangdong, China
| |
Collapse
|
13
|
Boudna M, Campos AD, Vychytilova-Faltejskova P, Machackova T, Slaby O, Souckova K. Strategies for labelling of exogenous and endogenous extracellular vesicles and their application for in vitro and in vivo functional studies. Cell Commun Signal 2024; 22:171. [PMID: 38461237 PMCID: PMC10924393 DOI: 10.1186/s12964-024-01548-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Accepted: 02/29/2024] [Indexed: 03/11/2024] Open
Abstract
This review presents a comprehensive overview of labelling strategies for endogenous and exogenous extracellular vesicles, that can be utilised both in vitro and in vivo. It covers a broad spectrum of approaches, including fluorescent and bioluminescent labelling, and provides an analysis of their applications, strengths, and limitations. Furthermore, this article presents techniques that use radioactive tracers and contrast agents with the ability to track EVs both spatially and temporally. Emphasis is also placed on endogenous labelling mechanisms, represented by Cre-lox and CRISPR-Cas systems, which are powerful and flexible tools for real-time EV monitoring or tracking their fate in target cells. By summarizing the latest developments across these diverse labelling techniques, this review provides researchers with a reference to select the most appropriate labelling method for their EV based research.
Collapse
Affiliation(s)
- Marie Boudna
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Andres Delgado Campos
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | | | - Tana Machackova
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic
| | - Ondrej Slaby
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic.
- Department of Biology, Faculty of Medicine, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic.
| | - Kamila Souckova
- Central European Institute of Technology, Masaryk University, Kamenice 753/5, 625 00, Brno, Czech Republic.
| |
Collapse
|
14
|
Meng Y, Sun J, Zhang G. Harnessing the power of goat milk-derived extracellular vesicles for medical breakthroughs: A review. Int J Biol Macromol 2024; 262:130044. [PMID: 38340922 DOI: 10.1016/j.ijbiomac.2024.130044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 02/03/2024] [Accepted: 02/06/2024] [Indexed: 02/12/2024]
Abstract
Research into goat milk-derived extracellular vesicles (GMVs) has grown in popularity in recent years owing to their potential uses in several sectors, including medicine. GMVs are tiny, lipid-bound structures that cells secrete and use to transport bioactive substances like proteins, lipids, and nucleic acids. They may be extracted from different body fluids, including blood, urine, and milk, and have been found to play crucial roles in cell-to-cell communication. GMVs are a promising field of study with applications in preventing and treating various disorders. Their immune-modulating properties, for instance, have been investigated, and they have shown promise in treating autoimmune illnesses and cancer. They may be loaded with therapeutic compounds and directed to particular cells or tissues, but they have also been studied for their potential use as drug-delivery vehicles. Goat milk extracellular vesicles are an intriguing study topic with many possible benefits. Although more study is required to thoroughly understand their functioning and prospective applications, they provide a promising path for creating novel medical treatments and technology.
Collapse
Affiliation(s)
- Yiming Meng
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan road, Dadong district, Shenyang 110042, China.
| | - Jing Sun
- Department of Biobank, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan road, Dadong district, Shenyang 110042, China
| | - Guirong Zhang
- Department of Central Laboratory, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No. 44, Xiaoheyan road, Dadong district, Shenyang 110042, China
| |
Collapse
|
15
|
Asfiya R, Xu L, Paramanantham A, Kabytaev K, Chernatynskaya A, McCully G, Yang H, Srivastava A. Physio-chemical Modifications to Re-engineer Small Extracellular Vesicles for Targeted Anticancer Therapeutics Delivery and Imaging. ACS Biomater Sci Eng 2024; 10:697-722. [PMID: 38241003 PMCID: PMC10956554 DOI: 10.1021/acsbiomaterials.3c01404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2024]
Abstract
Cancer theranostics developed through nanoengineering applications are essential for targeted oncologic interventions in the new era of personalized and precision medicine. Recently, small extracellular vesicles (sEVs) have emerged as an attractive nanoengineering platform for tumor-directed anticancer therapeutic delivery and imaging of malignant tumors. These natural nanoparticles have multiple advantages over synthetic nanoparticle-based delivery systems, such as intrinsic targeting ability, less immunogenicity, and a prolonged circulation time. Since the inception of sEVs as a viable replacement for liposomes (synthetic nanoparticles) as a drug delivery vehicle, many studies have attempted to further the therapeutic efficacy of sEVs. This article discusses engineering strategies for sEVs using physical and chemical methods to enhance their anticancer therapeutic delivery performance. We review physio-chemical techniques of effective therapeutic loading into sEV, sEV surface engineering for targeted entry of therapeutics, and its cancer environment sensitive release inside the cells/organ. Next, we also discuss the novel hybrid sEV systems developed by a combination of sEVs with lipid and metal nanoparticles to garner each component's benefits while overcoming their drawbacks. The article extensively analyzes multiple sEV labeling techniques developed and investigated for live tracking or imaging sEVs. Finally, we discuss the theranostic potential of engineered sEVs in future cancer care regimens.
Collapse
Affiliation(s)
- Rahmat Asfiya
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Lei Xu
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Anjugam Paramanantham
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Kuanysh Kabytaev
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Anna Chernatynskaya
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Grace McCully
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| | - Hu Yang
- Linda and Bipin Doshi Department of Chemical and Biochemical Engineering, Missouri University of Science and Technology, Rolla, Missouri 65409, United States
| | - Akhil Srivastava
- Department of Pathology and Anatomical Sciences, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
- Ellis Fischel Cancer Centre, University of Missouri School of Medicine, Columbia, Missouri 65212, United States
| |
Collapse
|
16
|
Zhou X, Huang Q, Jiang Y, Tang H, Zhang L, Li D, Xu Y. Emerging technologies for engineering of extracellular vesicles. Front Bioeng Biotechnol 2023; 11:1298746. [PMID: 38026881 PMCID: PMC10666158 DOI: 10.3389/fbioe.2023.1298746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 10/30/2023] [Indexed: 12/01/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bilayer membrane-enclosed vesicles that are secreted by all cell types. Natural EVs contain biological information such as proteins, nucleic acids, and lipids from their parent cells. Therefore, EVs have been extensively studied as diagnostic biomarkers and therapeutic tools under normal and pathological conditions. However, some drawbacks, including low yield, poor therapeutic effects, lack of imaging, and targeting capacity of natural EVs, still need to be improved. Emerging engineering technologies have rendered EVs new properties or functionalities that broadened their applications in the biomedical field. Herein, in this review, we gave a brief overview of advanced strategies for EV engineering. We focused on pre-treatment of parent cells to regulate their released EVs. Meanwhile, we summarized and discussed the direct modification of EVs to achieve drug loading, imaging, and targeting functionalities for downstream applications.
Collapse
Affiliation(s)
- Xin Zhou
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Qing Huang
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yang Jiang
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Huijing Tang
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Luhan Zhang
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Danyang Li
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Yunsheng Xu
- Research Center, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
- Department of Dermatovenereology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
17
|
Lucchetti D, Colella F, Artemi G, Haque S, Sgambato A, Pellicano R, Fagoonee S. Smart nano-sized extracellular vesicles for cancer therapy: Potential theranostic applications in gastrointestinal tumors. Crit Rev Oncol Hematol 2023; 191:104121. [PMID: 37690633 DOI: 10.1016/j.critrevonc.2023.104121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 01/27/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023] Open
Abstract
Extracellular vesicles (EVs) have gained tremendous interest in the search for next-generation therapeutics for the treatment of a range of pathologies, including cancer, especially due to their small size, biomolecular cargo, ability to mediate intercellular communication, high physicochemical stability, low immunogenicity and biocompatibility. The theranostic potential of EVs have been enhanced by adopting several strategies such as genetic or metabolic engineering, parental cell modification or direct functionalization to incorporate therapeutic compounds into these nanoplatforms. The smart nano-sized EVs indeed offer huge opportunities in the field of cancer, and current research is set at overcoming the existing pitfalls. Smart EVs are already being applied in the clinics despite the challenges faced. We provide, herein, an update on the technologies employed for EV functionalization in order to achieve optimal tumor cell targeting and EV tracking in vivo with bio-imaging modalities, as well as the preclinical and clinical studies making use of these modified EVs, in the context of gastrointestinal tumors.
Collapse
Affiliation(s)
- Donatella Lucchetti
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Filomena Colella
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy
| | - Giulia Artemi
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy
| | - Shafiul Haque
- Research and Scientific Studies Unit, College of Nursing and Allied Health Sciences, Jazan University, Jazan 45142, Saudi Arabia; Gilbert and Rose-Marie Chagoury School of Medicine, Lebanese American University, Beirut 1102 2801, Lebanon; Centre of Medical and Bio-Allied Health Sciences Research, Ajman University, Ajman 13306, United Arab Emirates
| | - Alessandro Sgambato
- Fondazione Policlinico Universitario 'Agostino Gemelli' IRCCS, Rome, Italy; Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, Rome, Italy.
| | - Rinaldo Pellicano
- Gastroenterology Unit, Città della salute e della Scienza Hospital, Turin, Italy
| | - Sharmila Fagoonee
- Institute of Biostructure and Bioimaging (CNR), Molecular Biotechnology Center, Turin, Italy
| |
Collapse
|
18
|
Chen Q, Yuan L, Chou WC, Cheng YH, He C, Monteiro-Riviere NA, Riviere JE, Lin Z. Meta-Analysis of Nanoparticle Distribution in Tumors and Major Organs in Tumor-Bearing Mice. ACS NANO 2023; 17:19810-19831. [PMID: 37812732 PMCID: PMC10604101 DOI: 10.1021/acsnano.3c04037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 08/24/2023] [Indexed: 10/11/2023]
Abstract
Low tumor delivery efficiency is a critical barrier in cancer nanomedicine. This study reports an updated version of "Nano-Tumor Database", which increases the number of time-dependent concentration data sets for different nanoparticles (NPs) in tumors from the previous version of 376 data sets with 1732 data points from 200 studies to the current version of 534 data sets with 2345 data points from 297 studies published from 2005 to 2021. Additionally, the current database includes 1972 data sets for five major organs (i.e., liver, spleen, lung, heart, and kidney) with a total of 8461 concentration data points. Tumor delivery and organ distribution are calculated using three pharmacokinetic parameters, including delivery efficiency, maximum concentration, and distribution coefficient. The median tumor delivery efficiency is 0.67% injected dose (ID), which is low but is consistent with previous studies. Employing the best regression model for tumor delivery efficiency, we generate hypothetical scenarios with different combinations of NP factors that may lead to a higher delivery efficiency of >3%ID, which requires further experimentation to confirm. In healthy organs, the highest NP accumulation is in the liver (10.69%ID/g), followed by the spleen 6.93%ID/g and the kidney 3.22%ID/g. Our perspective on how to facilitate NP design and clinical translation is presented. This study reports a substantially expanded "Nano-Tumor Database" and several statistical models that may help nanomedicine design in the future.
Collapse
Affiliation(s)
- Qiran Chen
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Long Yuan
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Wei-Chun Chou
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| | - Yi-Hsien Cheng
- Department
of Anatomy and Physiology, Kansas State
University, Manhattan, Kansas 66506, United States
- Institute
of Computational Comparative Medicine, Kansas
State University, Manhattan, Kansas 66506, United States
| | - Chunla He
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Department
of Biostatistics College of Public Health and Health Professions, University of Florida, Gainesville, Florida 32608, United States
| | - Nancy A. Monteiro-Riviere
- Nanotechnology
Innovation Center of Kansas State, Kansas
State University, Manhattan, Kansas 66506, United States
- Center
for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Jim E. Riviere
- Center
for Chemical Toxicology Research and Pharmacokinetics, North Carolina State University, Raleigh, North Carolina 27606, United States
- 1
Data Consortium, Kansas State University, Olathe, Kansas 66061, United States
| | - Zhoumeng Lin
- Department
of Environmental and Global Health, College of Public Health and Health
Professions, University of Florida, Gainesville, Florida 32608, United States
- Center
for Environmental and Human Toxicology, University of Florida, Gainesville, Florida 32610, United States
| |
Collapse
|
19
|
Cheng Z, Shang J, Wang H, Yu L, Yuan Z, Zhang Y, Du Y, Tian J. Molecular imaging-guided extracellular vesicle-based drug delivery for precise cancer management: Current status and future perspectives. J Control Release 2023; 362:97-120. [PMID: 37625599 DOI: 10.1016/j.jconrel.2023.08.040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 08/16/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023]
Abstract
Extracellular vesicles (EVs), the mediators of intercellular communication, have attracted the attention of researchers for the important roles they play in cancer treatment. Compared with other inorganic nano-materials, EVs possess the advantages of higher biocompatibility, better physiochemical stability, easier surface modification, and excellent biosafety. They can be used as an advanced drug delivery system with an improved therapeutic index for various therapeutic agents. Engineered EV-based imaging and therapeutic agents (engineered EVs) have emerged as useful tools in targeted cancer diagnosis and therapy. Non-invasive tracing of engineered EVs contributes to a better evaluation of their functions in cancer progression, in vivo dynamic biodistribution, therapeutic response, and drug-loading efficiency. Recent advances in real-time molecular imaging (MI), and innovative EV labeling strategies have led to the development of novel tools that can evaluate the pharmacokinetics of engineered EVs in cancer management, which may accelerate further clinical translation of novel EV-based drug delivery platforms. Herein, we review the latest advances in EVs, their characteristics, and current examples of EV-based targeted drug delivery for cancer. Then, we discuss the prominent applications of MI for tracing both natural and engineered EVs. Finally, we discuss the current challenges and considerations of EVs in targeted cancer treatment and the limitations of different MI modalities. In the coming decades, EV-based therapeutic applications for cancer with improved drug loading and targeting abilities will be developed, and better anti-cancer effects of drug delivery nanoplatform will be achieved.
Collapse
Affiliation(s)
- Zhongquan Cheng
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China; CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China
| | - Jihuan Shang
- School of Clinical Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Huarong Wang
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China
| | - Leyi Yu
- Beijing Haidian Hospital, Beijing 100080, China
| | - Zhu Yuan
- Department of General Surgery, Capital Medical University, Beijing Friendship Hospital, Beijing 100050, China.
| | - Yinlong Zhang
- School of Nanoscience and Engineering, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Yang Du
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; University of Chinese Academy of Sciences, Beijing 100080, China.
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Beijing Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing 100190, China; Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Medicine, China; Science and Engineering, Beihang University, Beijing 100191, China.
| |
Collapse
|
20
|
Zou Z, Li H, Xu G, Hu Y, Zhang W, Tian K. Current Knowledge and Future Perspectives of Exosomes as Nanocarriers in Diagnosis and Treatment of Diseases. Int J Nanomedicine 2023; 18:4751-4778. [PMID: 37635911 PMCID: PMC10454833 DOI: 10.2147/ijn.s417422] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 07/29/2023] [Indexed: 08/29/2023] Open
Abstract
Exosomes, as natural nanocarriers, characterized with low immunogenicity, non-cytotoxicity and targeted delivery capability, which have advantages over synthetic nanocarriers. Recently, exosomes have shown great potential as diagnostic markers for diseases and are also considered as a promising cell-free therapy. Engineered exosomes have significantly enhanced the efficacy and precision of delivering therapeutic agents, and are currently being extensively employed in targeted therapeutic investigations for various ailments, including oncology, inflammatory disorders, and degenerative conditions. Particularly, engineered exosomes enable therapeutic agent loading, targeted modification, evasion of MPS phagocytosis, intelligent control, and bioimaging, and have been developed as multifunctional nano-delivery platforms in recent years. The utilization of bioactive scaffolds that are loaded with exosome delivery has been shown to substantially augment retention, extend exosome release, and enhance efficacy. This approach has advanced from conventional hydrogels to nanocomposite hydrogels, nanofiber hydrogels, and 3D printing, resulting in superior physical and biological properties that effectively address the limitations of natural scaffolds. Additionally, plant-derived exosomes, which can participate in gut flora remodeling via oral administration, are considered as an ideal delivery platform for the treatment of intestinal diseases. Consequently, there is great interest in exosomes and exosomes as nanocarriers for therapeutic and diagnostic applications. This comprehensive review provides an overview of the biogenesis, composition, and isolation methods of exosomes. Additionally, it examines the pathological and diagnostic mechanisms of exosomes in various diseases, including tumors, degenerative disorders, and inflammatory conditions. Furthermore, this review highlights the significance of gut microbial-derived exosomes. Strategies and specific applications of engineered exosomes and bioactive scaffold-loaded exosome delivery are further summarized, especially some new techniques such as large-scale loading technique, macromolecular loading technique, development of multifunctional nano-delivery platforms and nano-scaffold-loaded exosome delivery. The potential benefits of using plant-derived exosomes for the treatment of gut-related diseases are also discussed. Additionally, the challenges, opportunities, and prospects of exosome-based nanocarriers for disease diagnosis and treatment are summarized from both preclinical and clinical viewpoints.
Collapse
Affiliation(s)
- Zaijun Zou
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Han Li
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Gang Xu
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| | - Yunxiang Hu
- School of Graduates, Dalian Medical University, Dalian, Liaoning, 116000, People’s Republic of China
| | - Weiguo Zhang
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| | - Kang Tian
- Department of Sports Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, 116011, People’s Republic of China
- Key Laboratory of Molecular Mechanism for Repair and Remodeling of Orthopaedic Disease, Dalian, Liaoning Province, 116011, People’s Republic of China
| |
Collapse
|
21
|
Bao C, Xiang H, Chen Q, Zhao Y, Gao Q, Huang F, Mao L. A Review of Labeling Approaches Used in Small Extracellular Vesicles Tracing and Imaging. Int J Nanomedicine 2023; 18:4567-4588. [PMID: 37588627 PMCID: PMC10426735 DOI: 10.2147/ijn.s416131] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 07/26/2023] [Indexed: 08/18/2023] Open
Abstract
Small extracellular vesicles (sEVs), a subset of extracellular vesicles (EVs) originating from the endosomal compartment, are a kind of lipid bilayer vesicles released by almost all types of cells, serving as natural carriers of nucleic acids, proteins, and lipids for intercellular communication and transfer of bioactive molecules. The current findings suggest their vital role in physiological and pathological processes. Various sEVs labeling techniques have been developed for the more advanced study of the function, mode of action, bio-distribution, and related information of sEVs. In this review, we summarize the existing and emerging sEVs labeling techniques, including fluorescent labeling, radioisotope labeling, nanoparticle labeling, chemical contrast agents labeling, and label-free technique. These approaches will pave the way for an in-depth study of sEVs. We present a systematic and comprehensive review of the principles, advantages, disadvantages, and applications of these techniques, to help promote applications of these labeling approaches in future research on sEVs.
Collapse
Affiliation(s)
- Chenxuan Bao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Huayuan Xiang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Qiaoqiao Chen
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Yuxue Zhao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| | - Qianqian Gao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Feng Huang
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
| | - Lingxiang Mao
- Department of Laboratory Medicine, Affiliated Kunshan Hospital of Jiangsu University, Kunshan, Jiangsu, People’s Republic of China
- Department of Laboratory Medicine, the Affiliated People’s Hospital, Jiangsu University, Zhenjiang, Jiangsu, People’s Republic of China
| |
Collapse
|
22
|
Banerjee A, Lino M, Jesus C, Ribeiro Q, Abrunhosa A, Ferreira L. Imaging platforms to dissect the in vivo communication, biodistribution and controlled release of extracellular vesicles. J Control Release 2023; 360:549-563. [PMID: 37406818 DOI: 10.1016/j.jconrel.2023.06.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/07/2023]
Abstract
Extracellular vesicles (EVs) work as communication vehicles, allowing the exchange of bioactive molecules (microRNAs, mRNAs, proteins, etc) between neighbouring and distant cells in the organism. EVs are thus important players in several physiological and pathological processes. Thus, it is critical to understand their role in cellular/organ communication to fully evaluate their biological, diagnosis and therapeutic potential. In addition, recent studies have explored the controlled release of EVs for regenerative medicine applications and thus the evaluation of their release profile is important to correlate with biological activity. Here, we give a brief introduction about EV imaging platforms in terms of their sensitivity, penetration depth, cost, and operational simplicity, followed by a discussion of different EV labelling processes with their advantages and limitations. Next, we cover the relevance of these imaging platforms to dissect the tropism and biological role of endogenous EVs. We also cover the relevance of imaging platforms to monitor the accumulation of exogenous EVs and their potential cellular targets. Finally, we highlight the importance of imaging platforms to investigate the release profile of EVs from different controlled systems.
Collapse
Affiliation(s)
- Arnab Banerjee
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Miguel Lino
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Carlos Jesus
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Quélia Ribeiro
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal
| | - Antero Abrunhosa
- ICNAS/CIBIT - Institute for Nuclear Sciences Applied to Health/Coimbra Institute for Biomedical Imaging and Translational research, University of Coimbra, 3000-548 Coimbra, Portugal
| | - Lino Ferreira
- CNC-Center for Neurosciences and Cell Biology, University of Coimbra, 3004-517 Coimbra, Portugal; Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal.
| |
Collapse
|
23
|
Petroni D, Fabbri C, Babboni S, Menichetti L, Basta G, Del Turco S. Extracellular Vesicles and Intercellular Communication: Challenges for In Vivo Molecular Imaging and Tracking. Pharmaceutics 2023; 15:1639. [PMID: 37376087 PMCID: PMC10301899 DOI: 10.3390/pharmaceutics15061639] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/23/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Extracellular vesicles (EVs) are a heterogeneous class of cell-derived membrane vesicles released by various cell types that serve as mediators of intercellular signaling. When released into circulation, EVs may convey their cargo and serve as intermediaries for intracellular communication, reaching nearby cells and possibly also distant organs. In cardiovascular biology, EVs released by activated or apoptotic endothelial cells (EC-EVs) disseminate biological information at short and long distances, contributing to the development and progression of cardiovascular disease and related disorders. The significance of EC-EVs as mediators of cell-cell communication has advanced, but a thorough knowledge of the role that intercommunication plays in healthy and vascular disease is still lacking. Most data on EVs derive from in vitro studies, but there are still little reliable data available on biodistribution and specific homing EVs in vivo tissues. Molecular imaging techniques for EVs are crucial to monitoring in vivo biodistribution and the homing of EVs and their communication networks both in basal and pathological circumstances. This narrative review provides an overview of EC-EVs, trying to highlight their role as messengers of cell-cell interaction in vascular homeostasis and disease, and describes emerging applications of various imaging modalities for EVs visualization in vivo.
Collapse
Affiliation(s)
- Debora Petroni
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Costanza Fabbri
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
- Institute of Life Sciences, Scuola Superiore Sant’Anna, 56127 Pisa, Italy
| | - Serena Babboni
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Luca Menichetti
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Giuseppina Basta
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| | - Serena Del Turco
- Institute of Clinical Physiology, CNR San Cataldo Research Area, Via Moruzzi 1, 56124 Pisa, Italy
| |
Collapse
|
24
|
Ahmadi M, Emzhik M, Mosayebnia M. Nanoparticles labeled with gamma-emitting radioisotopes: an attractive approach for in vivo tracking using SPECT imaging. Drug Deliv Transl Res 2023; 13:1546-1583. [PMID: 36811810 DOI: 10.1007/s13346-023-01291-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2023] [Indexed: 02/24/2023]
Abstract
Providing accurate molecular imaging of the body and biological process is critical for diagnosing disease and personalizing treatment with the minimum side effects. Recently, diagnostic radiopharmaceuticals have gained more attention in precise molecular imaging due to their high sensitivity and appropriate tissue penetration depth. The fate of these radiopharmaceuticals throughout the body can be traced using nuclear imaging systems, including single-photon emission computed tomography (SPECT) and positron emission tomography (PET) modalities. In this regard, nanoparticles are attractive platforms for delivering radionuclides into targets because they can directly interfere with the cell membranes and subcellular organelles. Moreover, applying radiolabeled nanomaterials can decrease their toxicity concerns because radiopharmaceuticals are usually administrated at low doses. Therefore, incorporating gamma-emitting radionuclides into nanomaterials can provide imaging probes with valuable additional properties compared to the other carriers. Herein, we aim to review (1) the gamma-emitting radionuclides used for labeling different nanomaterials, (2) the approaches and conditions adopted for their radiolabeling, and (3) their application. This study can help researchers to compare different radiolabeling methods in terms of stability and efficiency and choose the best way for each nanosystem.
Collapse
Affiliation(s)
- Mahnaz Ahmadi
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Marjan Emzhik
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mona Mosayebnia
- Department of Pharmaceutical Chemistry and Radiopharmacy, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Niayesh Junction, Vali-E-Asr Ave, Tehran, 14155-6153, Iran.
| |
Collapse
|
25
|
Avgoulas DI, Tasioulis KS, Papi RM, Pantazaki AA. Therapeutic and Diagnostic Potential of Exosomes as Drug Delivery Systems in Brain Cancer. Pharmaceutics 2023; 15:pharmaceutics15051439. [PMID: 37242681 DOI: 10.3390/pharmaceutics15051439] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 04/26/2023] [Accepted: 05/03/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer is designated as one of the principal causes of mortality universally. Among different types of cancer, brain cancer remains the most challenging one due to its aggressiveness, the ineffective permeation ability of drugs through the blood-brain barrier (BBB), and drug resistance. To overcome the aforementioned issues in fighting brain cancer, there is an imperative need for designing novel therapeutic approaches. Exosomes have been proposed as prospective "Trojan horse" nanocarriers of anticancer theranostics owing to their biocompatibility, increased stability, permeability, negligible immunogenicity, prolonged circulation time, and high loading capacity. This review provides a comprehensive discussion on the biological properties, physicochemical characteristics, isolation methods, biogenesis and internalization of exosomes, while it emphasizes their therapeutic and diagnostic potential as drug vehicle systems in brain cancer, highlighting recent advances in the research field. A comparison of the biological activity and therapeutic effectiveness of several exosome-encapsulated cargo including drugs and biomacromolecules underlines their great supremacy over the non-exosomal encapsulated cargo in the delivery, accumulation, and biological potency. Various studies on cell lines and animals give prominence to exosome-based nanoparticles (NPs) as a promising and alternative approach in the management of brain cancer.
Collapse
Affiliation(s)
- Dimitrios I Avgoulas
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Konstantinos S Tasioulis
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Rigini M Papi
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| | - Anastasia A Pantazaki
- Laboratory of Biochemistry, Department of Chemistry, Aristotle University of Thessaloniki, 54124 Thessaloniki, Greece
| |
Collapse
|
26
|
Ashique S, Anand K. Radiolabelled Extracellular Vesicles as Imaging Modalities for Precise Targeted Drug Delivery. Pharmaceutics 2023; 15:pharmaceutics15051426. [PMID: 37242668 DOI: 10.3390/pharmaceutics15051426] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/20/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023] Open
Abstract
Extracellular vesicles (ECVs) have been abandoned as bio-inspired drug delivery systems (DDS) in the biomedical field. ECVs have a natural ability to cross over extracellular and intracellular barriers, making them superior to manufactured nanoparticles. Additionally, they have the ability to move beneficial biomolecules among far-flung bodily cells. These advantages and the accomplishment of favorable in vivo results convincingly show the value of ECVs in medication delivery. The usage of ECVs is constantly being improved, as it might be difficult to develop a consistent biochemical strategy that is in line with their useful clinical therapeutic uses. Extracellular vesicles (ECVs) have the potential to enhance the therapy of diseases. Imaging technologies, particularly radiolabelled imaging, have been exploited for non-invasive tracking to better understand their in vivo activity.
Collapse
Affiliation(s)
- Sumel Ashique
- Department of Pharmaceutics, Bharat Institute of Technology (BIT), School of Pharmacy, Meerut 250103, India
| | - Krishnan Anand
- Department of Chemical Pathology, School of Pathology, Faculty of Health Sciences, University of the Free State, Bloemfontein 9300, South Africa
| |
Collapse
|
27
|
Aafreen S, Feng J, Wang W, Liu G. Theranostic extracellular vesicles: a concise review of current imaging technologies and labeling strategies. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2023; 4:107-132. [PMID: 37829171 PMCID: PMC10568980 DOI: 10.20517/evcna.2023.01] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Extracellular vesicles (EVs), or exosomes, are naturally occurring nano- and micro-sized membrane vesicles playing an essential role in cell-to-cell communication. There is a recent increasing interest in harnessing the therapeutic potential of these natural nanoparticles to develop cell-free regenerative medicine and manufacture highly biocompatible and targeted drug and gene delivery vectors, amongst other applications. In the context of developing novel and effective EV-based therapy, imaging tools are of paramount importance as they can be used to not only elucidate the underlying mechanisms but also provide the basis for optimization and clinical translation. In this review, recent efforts and knowledge advances on EV-based therapies have been briefly introduced, followed by an outline of currently available labeling strategies by which EVs can be conjugated with various imaging agents and/or therapeutic drugs and genes. A comprehensive review of prevailing EV imaging technologies is then presented along with examples and applications, with emphasis on imaging probes and agents, corresponding labeling methods, and the pros and cons of each imaging modality. Finally, the potential of theranostic EVs as a powerful new weapon in the arsenal of regenerative medicine and nanomedicine is summarized and envisioned.
Collapse
Affiliation(s)
- Safiya Aafreen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD 21218, USA
| | - Jonathan Feng
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
| | - Wenshen Wang
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guanshu Liu
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, USA
- Russell H. Morgan Department of Radiology and Radiological Sciences, The Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| |
Collapse
|
28
|
Chen M, Lin S, Zhou C, Cui D, Haick H, Tang N. From Conventional to Microfluidic: Progress in Extracellular Vesicle Separation and Individual Characterization. Adv Healthc Mater 2023; 12:e2202437. [PMID: 36541411 DOI: 10.1002/adhm.202202437] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 12/07/2022] [Indexed: 12/24/2022]
Abstract
Extracellular vesicles (EVs) are nanoscale membrane vesicles, which contain a wide variety of cargo such as proteins, miRNAs, and lipids. A growing body of evidence suggests that EVs are promising biomarkers for disease diagnosis and therapeutic strategies. Although the excellent clinical value, their use in personalized healthcare practice is not yet feasible due to their highly heterogeneous nature. Taking the difficulty of isolation and the small size of EVs into account, the characterization of EVs at a single-particle level is both imperative and challenging. In a bid to address this critical point, more research has been directed into a microfluidic platform because of its inherent advantages in sensitivity, specificity, and throughput. This review discusses the biogenesis and heterogeneity of EVs and takes a broad view of state-of-the-art advances in microfluidics-based EV research, including not only EV separation, but also the single EV characterization of biophysical detection and biochemical analysis. To highlight the advantages of microfluidic techniques, conventional technologies are included for comparison. The current status of artificial intelligence (AI) for single EV characterization is then presented. Furthermore, the challenges and prospects of microfluidics and its combination with AI applications in single EV characterization are also discussed. In the foreseeable future, recent breakthroughs in microfluidic platforms are expected to pave the way for single EV analysis and improve applications for precision medicine.
Collapse
Affiliation(s)
- Mingrui Chen
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Shujing Lin
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Cheng Zhou
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Daxiang Cui
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| | - Hossam Haick
- Department of Chemical Engineering and Russell Berrie Nanotechnology Institute, Technion-Israel Institute of Technology, Haifa, 3200003, Israel
| | - Ning Tang
- School of Electronic Information and Electrical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, P. R. China
| |
Collapse
|
29
|
Crum RJ, Capella-Monsonís H, Chang J, Dewey MJ, Kolich BD, Hall KT, El-Mossier SO, Nascari DG, Hussey GS, Badylak SF. Biocompatibility and biodistribution of matrix-bound nanovesicles in vitro and in vivo. Acta Biomater 2023; 155:113-122. [PMID: 36423817 DOI: 10.1016/j.actbio.2022.11.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
Abstract
Matrix-bound nanovesicles (MBV) are a distinct subtype of extracellular vesicles that are firmly embedded within biomaterials composed of extracellular matrix (ECM). MBV both store and transport a diverse, tissue specific portfolio of signaling molecules including proteins, miRNAs, and bioactive lipids. MBV function as a key mediator in ECM-mediated control of the local tissue microenvironment. One of the most important mechanisms by which MBV in ECM bioscaffolds support constructive tissue remodeling following injury is immunomodulation and, specifically, the promotion of an anti-inflammatory, pro-remodeling immune cell activation state. Recent in vivo studies have shown that isolated MBV have therapeutic efficacy in rodent models of both retinal damage and rheumatoid arthritis through the targeted immunomodulation of pro-inflammatory macrophages towards an anti-inflammatory activation state. While these results show the therapeutic potential of MBV administered independent of the rest of the ECM, the in vitro and in vivo safety and biodistribution profile of MBV remain uncharacterized. The purpose of the present study was to thoroughly characterize the pre-clinical safety profile of MBV through a combination of in vitro cytotoxicity and MBV uptake studies and in vivo toxicity, immunotoxicity, and imaging studies. The results showed that MBV isolated from porcine urinary bladder are well-tolerated and are not cytotoxic in cell culture, are non-toxic to the whole organism, and are not immunosuppressive compared to the potent immunosuppressive drug cyclophosphamide. Furthermore, this safety profile was sustained across a wide range of MBV doses. STATEMENT OF SIGNIFICANCE: Matrix-bound nanovesicles (MBV) are a distinct subtype of bioactive extracellular vesicles that are embedded within biomaterials composed of extracellular matrix (ECM). Recent studies have shown therapeutic efficacy of MBV in models of both retinal damage and rheumatoid arthritis through the targeted immunomodulation of pro-inflammatory macrophages towards an anti-inflammatory activation state. While these results show the therapeutic potential of MBV, the in vitro and in vivo biocompatibility and biodistribution profile of MBV remain uncharacterized. The results of the present study showed that MBV are a well-tolerated ECM-derived therapy that are not cytotoxic in cell culture, are non-toxic to the whole organism, and are not immunosuppressive. Collectively, these data highlight the translational feasibility of MBV therapeutics across a wide variety of clinical applications.
Collapse
Affiliation(s)
- Raphael J Crum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Héctor Capella-Monsonís
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Jordan Chang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Marley J Dewey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Brian D Kolich
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Kelsey T Hall
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Salma O El-Mossier
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - David G Nascari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - George S Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
30
|
Zhang L, Cheng Y, Liu YG, Chen X, Liu H. Anticancer Effect of Chlorambucil Enhanced by Chiral Phthalidyl Promoiety. Chem Biodivers 2023; 20:e202201025. [PMID: 36427041 DOI: 10.1002/cbdv.202201025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2022] [Accepted: 11/23/2022] [Indexed: 11/27/2022]
Abstract
Phthalidyl promoiety has been used in several drugs, but they were all marketed in racemic form. The pharmaceutical effects of each enantiomer have not been clearly demonstrated. In this project, an anticancer chemotherapy drug, chlorambucil, was modified as enantiopure phthalidyl prodrugs. The enantiomers, together with phthalidyl unit and their racemic mixture, were then subject to the in vivo bioactivity tests against B16F10 melanoma cells. It was found that proper chirality within the promoiety had noticeably better in vivo pharmacological effects than the parent drug, the enantiomer and racemic mixture. This merit perhaps could be extended from the phthalidyl prodrugs to other chirality containing prodrugs.
Collapse
Affiliation(s)
- Long Zhang
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, P. R. China
| | - Yisa Cheng
- First Affiliated Hospital of Zhengzhou University, and Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Ying-Guo Liu
- First Affiliated Hospital of Zhengzhou University, and Henan Institute of Advanced Technology, Zhengzhou University, Zhengzhou, 450001, P. R. China
| | - Xingkuan Chen
- Guangdong Provincial Key Laboratory of Functional Supramolecular Coordination Materials and Applications, Department of Chemistry, Jinan University, Guangzhou, Guangdong, 510632, P. R. China
| | - Hongmei Liu
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, P. R. China
| |
Collapse
|
31
|
Preeclampsia and syncytiotrophoblast membrane extracellular vesicles (STB-EVs). Clin Sci (Lond) 2022; 136:1793-1807. [PMID: 36511102 DOI: 10.1042/cs20220149] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2022] [Revised: 10/03/2022] [Accepted: 10/21/2022] [Indexed: 12/15/2022]
Abstract
Preeclampsia (PE) is a hypertensive complication of pregnancy that affects 2-8% of women worldwide and is one of the leading causes of maternal deaths and premature birth. PE can occur early in pregnancy (<34 weeks gestation) or late in pregnancy (>34 weeks gestation). Whilst the placenta is clearly implicated in early onset PE (EOPE), late onset PE (LOPE) is less clear with some believing the disease is entirely maternal whilst others believe that there is an interplay between maternal systems and the placenta. In both types of PE, the syncytiotrophoblast (STB), the layer of the placenta in direct contact with maternal blood, is stressed. In EOPE, the STB is oxidatively stressed in early pregnancy (leading to PE later in gestation- the two-stage model) whilst in LOPE the STB is stressed because of villous overcrowding and senescence later in pregnancy. It is this stress that perturbs maternal systems leading to the clinical manifestations of PE. Whilst some of the molecular species driving this stress have been identified, none completely explain the multisystem nature of PE. Syncytiotrophoblast membrane vesicles (STB-EVs) are a potential contributor to this multisystem disorder. STB-EVs are released into the maternal circulation in increasing amounts with advancing gestational age, and this release is further exacerbated with stress. There are good in vitro evidence that STB-EVs are taken up by macrophages and liver cells with additional evidence supporting endothelial cell uptake. STB-EV targeting remains in the early stages of discovery. In this review, we highlight the role of STB-EVs in PE. In relation to current research, we discuss different protocols for ex vivo isolation of STB-EVs, as well as specific issues involving tissue preparation, isolation (some of which may be unique to STB-EVs), and methods for their analysis. We suggest potential solutions for these challenges.
Collapse
|
32
|
Almeida SFF, Fonseca A, Sereno J, Ferreira HRS, Lapo-Pais M, Martins-Marques T, Rodrigues T, Oliveira RC, Miranda C, Almeida LP, Girão H, Falcão A, Abrunhosa AJ, Gomes CM. Osteosarcoma-Derived Exosomes as Potential PET Imaging Nanocarriers for Lung Metastasis. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2203999. [PMID: 36316233 DOI: 10.1002/smll.202203999] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 10/08/2022] [Indexed: 06/16/2023]
Abstract
Lung metastases represent the most adverse clinical factor and rank as the leading cause of osteosarcoma-related death. Nearly 80% of patients present lung micrometastasis at diagnosis not detected with current clinical tools. Herein, an exosome (EX)-based imaging tool is developed for lung micrometastasis by positron emission tomography (PET) using osteosarcoma-derived EXs as natural nanocarriers of the positron-emitter copper-64 (64 Cu). Exosomes are isolated from metastatic osteosarcoma cells and functionalized with the macrocyclic chelator NODAGA for complexation with 64 Cu. Surface functionalization has no effect on the physicochemical properties of EXs, or affinity for donor cells and endows them with favorable pharmacokinetics for in vivo studies. Whole-body PET/magnetic resonance imaging (MRI) images in xenografted models show a specific accumulation of 64 Cu-NODAGA-EXs in metastatic lesions as small as 2-3 mm or in a primary tumor, demonstrating the exquisite tropism of EXs for homotypic donor cells. The targetability for lung metastasis is also observed by optical imaging using indocyanine green (ICG)-labeled EXs and D-luciferin-loaded EXs. These findings show that tumor-derived EXs hold great potential as targeted imaging agents for the noninvasive detection of small lung metastasis by PET. This represents a step forward in the biomedical application of EXs in imaging diagnosis with increased translational potential.
Collapse
Affiliation(s)
- Sara F F Almeida
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548, Coimbra, Portugal
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548, Coimbra, Portugal
| | - Alexandra Fonseca
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548, Coimbra, Portugal
| | - José Sereno
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548, Coimbra, Portugal
- Chemistry Department, University of Coimbra, 3004-535, Coimbra, Portugal
| | - Hugo R S Ferreira
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075, Coimbra, Portugal
| | - Mariana Lapo-Pais
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548, Coimbra, Portugal
| | - Tânia Martins-Marques
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075, Coimbra, Portugal
| | - Teresa Rodrigues
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075, Coimbra, Portugal
| | - Rui C Oliveira
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075, Coimbra, Portugal
- Pathology Department, Centro Hospitalar e Universitário de Coimbra, 3004-561, Coimbra, Portugal
| | - Catarina Miranda
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504, Coimbra, Portugal
| | - Luís P Almeida
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504, Coimbra, Portugal
| | - Henrique Girão
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075, Coimbra, Portugal
| | - Amílcar Falcão
- Faculty of Pharmacy, University of Coimbra, 3000-548, Coimbra, Portugal
| | - Antero J Abrunhosa
- Institute for Nuclear Sciences Applied to Health (ICNAS) and Coimbra Institute for Biomedical Imaging and Translational Research (CIBIT), University of Coimbra, 3000-548, Coimbra, Portugal
| | - Célia M Gomes
- Faculty of Medicine, Coimbra Institute for Clinical and Biomedical Research (iCBR), University of Coimbra, 3000-548, Coimbra, Portugal
- Center for Innovative Biomedicine and Biotechnology Consortium (CIBB), University of Coimbra, 3000-548, Coimbra, Portugal
- Clinical Academic Center of Coimbra (CACC), 3000-075, Coimbra, Portugal
| |
Collapse
|
33
|
Liu Q, Huang J, Xia J, Liang Y, Li G. Tracking tools of extracellular vesicles for biomedical research. Front Bioeng Biotechnol 2022; 10:943712. [PMID: 36466335 PMCID: PMC9716315 DOI: 10.3389/fbioe.2022.943712] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2022] [Accepted: 11/03/2022] [Indexed: 08/02/2023] Open
Abstract
Imaging of extracellular vesicles (EVs) will facilitate a better understanding of their biological functions and their potential as therapeutics and drug delivery vehicles. In order to clarify EV-mediated cellular communication in vitro and to track the bio-distribution of EV in vivo, various strategies have been developed to label and image EVs. In this review, we summarized recent advances in the tracking of EVs, demonstrating the methods for labeling and imaging of EVs, in which the labeling methods include direct and indirect labeling and the imaging modalities include fluorescent imaging, bioluminescent imaging, nuclear imaging, and nanoparticle-assisted imaging. These techniques help us better understand the mechanism of uptake, the bio-distribution, and the function of EVs. More importantly, we can evaluate the pharmacokinetic properties of EVs, which will help promote their further clinical application.
Collapse
Affiliation(s)
- Qisong Liu
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopaedic Surgery, Shenzhen People’s Hospital (The Second Clinical Medical College of Jinan University), Shenzhen, China
| | - Jianghong Huang
- Department of Orthopedics, Shenzhen Second People’s Hospital (First Affiliated Hospital of Shenzhen University, Health Science Center), Shenzhen, China
- Tsinghua University Shenzhen International Graduate School, Shenzhen, China
| | - Jiang Xia
- Department of Chemistry, The Chinese University of Hong Kong, Hong Kong, China
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, China
- Affiliated Hospital of Jining Medical University, Jining Medical University, Jining, China
| | - Guangheng Li
- Shenzhen Key Laboratory of Musculoskeletal Tissue Reconstruction and Function Restoration, Department of Orthopaedic Surgery, Shenzhen People’s Hospital (The Second Clinical Medical College of Jinan University), Shenzhen, China
| |
Collapse
|
34
|
Santos-Coquillat A, Herreros-Pérez D, Samaniego R, González MI, Cussó L, Desco M, Salinas B. Dual-labeled nanoparticles based on small extracellular vesicles for tumor detection. Biol Direct 2022; 17:31. [DOI: 10.1186/s13062-022-00345-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
Abstract
Background
Small extracellular vesicles (sEVs) are emerging natural nanoplatforms in cancer diagnosis and therapy, through the incorporation of signal components or drugs in their structure. However, for their translation into the clinical field, there is still a lack of tools that enable a deeper understanding of their in vivo pharmacokinetics or their interactions with the cells of the tumor microenvironment. In this study, we have designed a dual-sEV probe based on radioactive and fluorescent labeling of goat milk sEVs.
Results
The imaging nanoprobe was tested in vitro and in vivo in a model of glioblastoma. In vitro assessment of the uptake of the dual probe in different cell populations (RAW 264.7, U87, and HeLa) by optical and nuclear techniques (gamma counter, confocal imaging, and flow cytometry) revealed the highest uptake in inflammatory cells (RAW 264.7), followed by glioblastoma U87 cells. In vivo evaluation of the pharmacokinetic properties of nanoparticles confirmed a blood circulation time of ~ 8 h and primarily hepatobiliary elimination. The diagnostic capability of the dual nanoprobe was confirmed in vivo in a glioblastoma xenograft model, which showed intense in vivo uptake of the SEV-based probe in tumor tissue. Histological assessment by confocal imaging enabled quantification of tumor populations and confirmed uptake in tumor cells and tumor-associated macrophages, followed by cancer-associated fibroblasts and endothelial cells.
Conclusions
We have developed a chemical approach for dual radioactive and fluorescent labeling of sEVs. This methodology enables in vivo and in vitro study of these vesicles after exogenous administration. The dual nanoprobe would be a promising technology for cancer diagnosis and a powerful tool for studying the biological behavior of these nanosystems for use in drug delivery.
Graphical Abstract
Collapse
|
35
|
Srivastava A, Rathore S, Munshi A, Ramesh R. Organically derived exosomes as carriers of anticancer drugs and imaging agents for cancer treatment. Semin Cancer Biol 2022; 86:80-100. [PMID: 35192929 PMCID: PMC9388703 DOI: 10.1016/j.semcancer.2022.02.020] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 02/15/2022] [Accepted: 02/17/2022] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs), is the umbrella term used for different types of vesicles produced by the cells, among which exosomes form the largest group. Exosomes perform intercellular communication by carrying several biologics from donor or parental cells and delivering them to recipient cells. Their unique cargo-carrying capacity has recently been explored for use as delivery vehicles of anticancer drugs and imaging agents. Being naturally produced, exosomes have many advantages over synthetic lipid-based nanoparticles currently being used clinically to treat cancer and other diseases. The finding of the role of exosomes in human diseases has led to numerous preclinical and clinical studies exploring their use as an amenable drug delivery vehicle and a theranostic in cancer diagnosis and treatment. However, there are certain limitations associated with exosomes, with the most important being the selection of the biological source for producing highly biocompatible exosomes on a large scale. This review article explores the various sources from which therapeutically viable exosomes can be isolated for use as drug carriers for cancer treatment. The methods of exosome isolation and the process of loading them with cancer therapeutics and imaging agents are also discussed in the follow-up sections. Finally, the article concludes with future directions for exosome-based applications in cancer diagnosis and treatment.
Collapse
Affiliation(s)
- Akhil Srivastava
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Shipra Rathore
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Anupama Munshi
- Radiation Oncology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA
| | - Rajagopal Ramesh
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, USA; Graduate Program in Biomedical Sciences, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
36
|
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: 6.0] [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
|
37
|
Aimaletdinov AM, Gomzikova MO. Tracking of Extracellular Vesicles' Biodistribution: New Methods and Approaches. Int J Mol Sci 2022; 23:11312. [PMID: 36232613 PMCID: PMC9569979 DOI: 10.3390/ijms231911312] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 09/15/2022] [Accepted: 09/21/2022] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs) are nanosized lipid bilayer vesicles that are released by almost all cell types. They range in diameter from 30 nm to several micrometres and have the ability to carry biologically active molecules such as proteins, lipids, RNA, and DNA. EVs are natural vectors and play an important role in many physiological and pathological processes. The amount and composition of EVs in human biological fluids serve as biomarkers and are used for diagnosing diseases and monitoring the effectiveness of treatment. EVs are promising for use as therapeutic agents and as natural vectors for drug delivery. However, the successful use of EVs in clinical practice requires an understanding of their biodistribution in an organism. Numerous studies conducted so far on the biodistribution of EVs show that, after intravenous administration, EVs are mostly localized in organs rich in blood vessels and organs associated with the reticuloendothelial system, such as the liver, lungs, spleen, and kidneys. In order to improve resolution, new dyes and labels are being developed and detection methods are being optimized. In this work, we review all available modern methods and approaches used to assess the biodistribution of EVs, as well as discuss their advantages and limitations.
Collapse
Affiliation(s)
| | - Marina O. Gomzikova
- Laboratory of Intercellular Communication, Institute of Fundamental Medicine and Biology, Kazan Federal University, Kazan 420008, Russia
| |
Collapse
|
38
|
Jiang A, Nie W, Xie H. In Vivo Imaging for the Visualization of Extracellular Vesicle-Based Tumor Therapy. ChemistryOpen 2022; 11:e202200124. [PMID: 36101512 PMCID: PMC9471060 DOI: 10.1002/open.202200124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/01/2022] [Indexed: 12/04/2022] Open
Abstract
Extracellular vesicles (EVs) exhibiting versatile biological functions provide promising prospects as natural therapeutic agents and drug delivery vehicles. For future clinical translation, revealing the fate of EVs in vivo, especially their accumulation at lesion sites, is very important. The continuous development of in vivo imaging technology has made it possible to track the real-time distribution of EVs. This article reviews the applications of mammal-, plant-, and bacteria-derived EVs in tumor therapy, the labeling methods of EVs for in vivo imaging, the advantages and disadvantages of different imaging techniques, and possible improvements for future work.
Collapse
Affiliation(s)
- Anqi Jiang
- School of Life ScienceBeijing Institute of TechnologyBeijing100081P. R. China
| | - Weidong Nie
- School of Life ScienceBeijing Institute of TechnologyBeijing100081P. R. China
| | - Hai‐Yan Xie
- School of Life ScienceBeijing Institute of TechnologyBeijing100081P. R. China
| |
Collapse
|
39
|
Toghiani R, Abolmaali SS, Najafi H, Tamaddon AM. Bioengineering exosomes for treatment of organ ischemia-reperfusion injury. Life Sci 2022; 302:120654. [PMID: 35597547 DOI: 10.1016/j.lfs.2022.120654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2022] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 11/30/2022]
Abstract
Ischemia-reperfusion (I/R) injury is a leading cause of death worldwide. It arises from blood reflowing after tissue hypoxia induced by ischemia that causes severe damages due to the accumulation of reactive oxygen species and the activation of inflammatory responses. Exosomes are the smallest members of the extracellular vesicles' family, which originate from nearly all eukaryotic cells. Exosomes have a great potential in the treatment of I/R injury either in native or modified forms. Native exosomes are secreted by different cell types, such as stem cells, and contain components such as specific miRNA molecules with tissue protective properties. On the other hand, exosome bioengineering has recently received increased attention in context of current advances in the purification, manipulation, biological characterization, and pharmacological applications. There are various pre-isolation and post-isolation manipulation approaches that can be utilized to increase the circulation half-life of exosomes or the availability of their bioactive cargos in the target site. In this review, the various therapeutic actions of native exosomes in different I/R injury will be discussed first. Exosome bioengineering approaches will then be explained, including pre- and post-isolation manipulation methods, applicability for delivery of bioactive agents to injured tissue, clinical translation issues, and future perspectives.
Collapse
Affiliation(s)
- Reyhaneh Toghiani
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Haniyeh Najafi
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
40
|
Arifin DR, Witwer KW, Bulte JWM. Non-Invasive imaging of extracellular vesicles: Quo vaditis in vivo? J Extracell Vesicles 2022; 11:e12241. [PMID: 35844061 PMCID: PMC9289215 DOI: 10.1002/jev2.12241] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 06/12/2022] [Accepted: 06/16/2022] [Indexed: 01/11/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid-bilayer delimited vesicles released by nearly all cell types that serve as mediators of intercellular signalling. Recent evidence has shown that EVs play a key role in many normal as well as pathological cellular processes. EVs can be exploited as disease biomarkers and also as targeted, cell-free therapeutic delivery and signalling vehicles for use in regenerative medicine and other clinical settings. Despite this potential, much remains unknown about the in vivo biodistribution and pharmacokinetic profiles of EVs after administration into living subjects. The ability to non-invasively image exogeneous EVs, especially in larger animals, will allow a better understanding of their in vivo homing and retention patterns, blood and tissue half-life, and excretion pathways, all of which are needed to advance clinical diagnostic and/or therapeutic applications of EVs. We present the current state-of-the-art methods for labeling EVs with various diagnostic contrast agents and tracers and the respective imaging modalities that can be used for their in vivo visualization: magnetic resonance imaging (MRI), X-ray computed tomography (CT) imaging, magnetic particle imaging (MPI), single-photon emission computed tomography (SPECT), positron emission tomography (PET), and optical imaging (fluorescence and bioluminescence imaging). We review here the strengths and weaknesses of each of these EV imaging approaches, with special emphasis on clinical translation.
Collapse
Affiliation(s)
- Dian R. Arifin
- Russell H. Morgan Department of Radiology and Radiological ScienceDivision of MR Researchthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Kenneth W. Witwer
- Department of Molecular and Comparative Pathobiologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Neurologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jeff W. M. Bulte
- Russell H. Morgan Department of Radiology and Radiological ScienceDivision of MR Researchthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Cellular Imaging Section and Vascular Biology ProgramInstitute for Cell Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Oncologythe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Chemical & Biomolecular Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
- Department of Biomedical Engineeringthe Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
41
|
Skotland T, Iversen TG, Llorente A, Sandvig K. Biodistribution, pharmacokinetics and excretion studies of intravenously injected nanoparticles and extracellular vesicles: Possibilities and challenges. Adv Drug Deliv Rev 2022; 186:114326. [PMID: 35588953 DOI: 10.1016/j.addr.2022.114326] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 04/25/2022] [Accepted: 05/05/2022] [Indexed: 12/16/2022]
Abstract
There is a large interest in developing nanoparticles and extracellular vesicles for delivery of therapeutics or imaging agents. Regulatory approval of such products requires knowledge about their biodistribution, metabolism and excretion. We here discuss possibilities and challenges of methods used for such studies, which most often are performed after labelling with radioactive isotopes or fluorescent molecules. It is important to evaluate if the labelled and unlabeled products can be expected to behave similarly in the body. Furthermore, one needs to critically consider whether the labels are still associated with the product at the time of analyses. We discuss advantages and disadvantages of different imaging modalities such as PET, SPECT, MRI, CT, ultrasound and optical imaging for whole-body biodistribution, and describe how to estimate the amount of labelled product in harvested organs and tissue. Microscopy of cells and tissues and various mass spectrometry methods are also discussed in this review.
Collapse
|
42
|
Choi H, Kim MY, Kim DH, Yun H, Oh BK, Kim SB, Song IH, Park HS, Kim SE, Park C, Choi C. Quantitative Biodistribution and Pharmacokinetics Study of GMP-Grade Exosomes Labeled with 89Zr Radioisotope in Mice and Rats. Pharmaceutics 2022; 14:pharmaceutics14061118. [PMID: 35745690 PMCID: PMC9229812 DOI: 10.3390/pharmaceutics14061118] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 05/13/2022] [Accepted: 05/18/2022] [Indexed: 12/13/2022] Open
Abstract
For the successful clinical advancement of exosome therapeutics, the biodistribution and pharmacokinetic profile of exogenous exosomes in various animal models must be determined. Compared with fluorescence or bioluminescence imaging, radionuclide imaging confers multiple advantages for the in vivo tracking of biomolecular therapeutics because of its excellent sensitivity for deep tissue imaging and potential for quantitative measurement. Herein, we assessed the quantitative biodistribution and pharmacokinetics of good manufacturing practice-grade therapeutic exosomes labeled with zirconium-89 (89Zr) after systemic intravenous administration in mice and rats. Quantitative biodistribution analysis by positron emission tomography/computed tomography and gamma counting in mice and rats revealed that the total 89Zr signals in the organs were lower in rats than in mice, suggesting a higher excretion rate of exosomes in rats. A prolonged 89Zr signal for up to 7 days in most organs indicated that substantial amounts of exosomes were taken up by the parenchymal cells in those organs, highlighting the therapeutic potential of exosomes for the intracellular delivery of therapeutics. Exosomes were mainly distributed in the liver and to a lesser extent in the spleen, while a moderately distributed in the kidney, lung, stomach, intestine, urinary bladder, brain, and heart. Exosomes were rapidly cleared from the blood circulation, with a rate greater than that of free 89Zr, indicating that exosomes might be rapidly taken up by cells and tissues.
Collapse
Affiliation(s)
- Hojun Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (M.-Y.K.); (D.-H.K.); (H.Y.); (B.-K.O.)
| | - Myung-Yoon Kim
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (M.-Y.K.); (D.-H.K.); (H.Y.); (B.-K.O.)
| | - Dae-Hwan Kim
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (M.-Y.K.); (D.-H.K.); (H.Y.); (B.-K.O.)
| | - Hanoul Yun
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (M.-Y.K.); (D.-H.K.); (H.Y.); (B.-K.O.)
| | - Byung-Koo Oh
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (M.-Y.K.); (D.-H.K.); (H.Y.); (B.-K.O.)
| | - Su-Bin Kim
- Department of Applied Bioengineering, Graduate School of Convergence Science and Technology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea;
- Department of Nuclear Medicine, Seoul National University Bundang Hospital 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea; (I.-H.S.); (H.-S.P.); (S.-E.K.)
| | - In-Ho Song
- Department of Nuclear Medicine, Seoul National University Bundang Hospital 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea; (I.-H.S.); (H.-S.P.); (S.-E.K.)
| | - Hyun-Soo Park
- Department of Nuclear Medicine, Seoul National University Bundang Hospital 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea; (I.-H.S.); (H.-S.P.); (S.-E.K.)
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
| | - Sang-Eun Kim
- Department of Nuclear Medicine, Seoul National University Bundang Hospital 82, Gumi-ro 173 Beon-gil, Bundang-gu, Seongnam 13620, Korea; (I.-H.S.); (H.-S.P.); (S.-E.K.)
- Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, Korea
- Advanced Institutes of Convergence Technology 145, Gwanggyo-ro, Yeongtong-gu, Suwon 16229, Korea
| | - Cheolhyoung Park
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (M.-Y.K.); (D.-H.K.); (H.Y.); (B.-K.O.)
- Correspondence: (C.P.); (C.C.); Tel.: +82-42-863-4450 (C.C.)
| | - Chulhee Choi
- ILIAS Biologics Inc., Daejeon 34014, Korea; (H.C.); (M.-Y.K.); (D.-H.K.); (H.Y.); (B.-K.O.)
- Department of Bio and Brain Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon 34141, Korea
- Correspondence: (C.P.); (C.C.); Tel.: +82-42-863-4450 (C.C.)
| |
Collapse
|
43
|
Lai CH, Lee CL, Vu CA, Vu VT, Tsai YH, Chen WY, Cheng CM. Paper-Based Devices for Capturing Exosomes and Exosomal Nucleic Acids From Biological Samples. Front Bioeng Biotechnol 2022; 10:836082. [PMID: 35497368 PMCID: PMC9039228 DOI: 10.3389/fbioe.2022.836082] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Accepted: 03/08/2022] [Indexed: 12/13/2022] Open
Abstract
Exosomes, nanovesicles derived from cells, contain a variety of biomolecules that can be considered biomarkers for disease diagnosis, including microRNAs (miRNAs). Given knowledge and demand, inexpensive, robust, and easy-to-use tools that are compatible with downstream nucleic acid detection should be developed to replace traditional methodologies for point-of-care testing (POCT) applications. This study deploys a paper-based extraction kit for exosome and exosomal miRNA analytical system with some quantifying methods to serve as an easy sample preparation for a possible POCT process. Exosomes concentrated from HCT116 cell cultures were arrested on paper-based immunoaffinity devices, which were produced by immobilizing anti-CD63 antibodies on Whatman filter paper, before being subjected to paper-based silica devices for nucleic acids to be trapped by silica nanoparticles adsorbed onto Whatman filter paper. Concentrations of captured exosomes were quantified by enzyme-linked immunosorbent assay (ELISA), demonstrating that paper-based immunoaffinity devices succeeded in capturing and determining exosome levels from cells cultured in both neutral and acidic microenvironments, whereas microRNA 21 (miR-21), a biomarker for various types of cancers and among the nucleic acids absorbed onto the silica devices, was determined by reverse transcription quantitative polymerase chain reaction (RT-qPCR) to prove that paper-based silica devices were capable of trapping exosomal nucleic acids. The developed paper-based kit and the devised procedure was successfully exploited to isolate exosomes and exosomal nucleic acids from different biological samples (platelet-poor plasma and lesion fluid) as clinical applications.
Collapse
Affiliation(s)
- Chi-Hung Lai
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Chih-Ling Lee
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Cao-An Vu
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Van-Truc Vu
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
| | - Yao-Hung Tsai
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Wen-Yih Chen
- Department of Chemical and Materials Engineering, National Central University, Taoyuan, Taiwan
- *Correspondence: Chao-Min Cheng, ; Wen-Yih Chen,
| | - Chao-Min Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- *Correspondence: Chao-Min Cheng, ; Wen-Yih Chen,
| |
Collapse
|
44
|
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: 1.5] [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
|
45
|
Khan A, Man F, Faruqu FN, Kim J, Al-Salemee F, Carrascal-Miniño A, Volpe A, Liam-Or R, Simpson P, Fruhwirth GO, Al-Jamal KT, T. M. de Rosales R. PET Imaging of Small Extracellular Vesicles via [ 89Zr]Zr(oxinate) 4 Direct Radiolabeling. Bioconjug Chem 2022; 33:473-485. [PMID: 35224973 PMCID: PMC8931726 DOI: 10.1021/acs.bioconjchem.1c00597] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 12/30/2021] [Indexed: 12/25/2022]
Abstract
Exosomes or small extracellular vesicles (sEVs) are increasingly gaining attention for their potential as drug delivery systems and biomarkers of disease. Therefore, it is important to understand their in vivo biodistribution using imaging techniques that allow tracking over time and at the whole-body level. Positron emission tomography (PET) allows short- and long-term whole-body tracking of radiolabeled compounds in both animals and humans and with excellent quantification properties compared to other nuclear imaging techniques. In this report, we explored the use of [89Zr]Zr(oxinate)4 (a cell and liposome radiotracer) for direct and intraluminal radiolabeling of several types of sEVs, achieving high radiolabeling yields. The radiosynthesis and radiolabeling protocols were optimized for sEV labeling, avoiding sEV damage, as demonstrated using several characterizations (cryoEM, nanoparticle tracking analysis, dot blot, and flow cytometry) and in vitro techniques. Using pancreatic cancer sEVs (PANC1) in a healthy mouse model, we showed that it is possible to track 89Zr-labeled sEVs in vivo using PET imaging for at least up to 24 h. We also report differential biodistribution of intact sEVs compared to intentionally heat-damaged sEVs, with significantly reduced spleen uptake for the latter. Therefore, we conclude that 89Zr-labeled sEVs using this method can reliably be used for in vivo PET tracking and thus allow efficient exploration of their potential as drug delivery systems.
Collapse
Affiliation(s)
- Azalea
A. Khan
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Francis Man
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
- Institute
of Pharmaceutical Sciences, School of Cancer & Pharmaceutical
Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, U.K.
| | - Farid N. Faruqu
- Institute
of Pharmaceutical Sciences, School of Cancer & Pharmaceutical
Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, U.K.
| | - Jana Kim
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Fahad Al-Salemee
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Amaia Carrascal-Miniño
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Alessia Volpe
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Revadee Liam-Or
- Institute
of Pharmaceutical Sciences, School of Cancer & Pharmaceutical
Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, U.K.
| | - Paul Simpson
- Electron
Microscopy Centre, Department of Life Sciences, Faculty of Natural
Sciences, Imperial College London, Flowers Building, London SW7 2AZ, U.K.
| | - Gilbert O. Fruhwirth
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| | - Khuloud T. Al-Jamal
- Institute
of Pharmaceutical Sciences, School of Cancer & Pharmaceutical
Sciences, King’s College London, Franklin Wilkins Building, London SE1 9NH, U.K.
| | - Rafael T. M. de Rosales
- Department
of Imaging Chemistry and Biology, School of Biomedical Engineering
and Imaging Sciences, King’s College London, St. Thomas’ Hospital, London SE1 7EH, U.K.
| |
Collapse
|
46
|
Quadri Z, Elsherbini A, Bieberich E. Extracellular vesicles in pharmacology: Novel approaches in diagnostics and therapy. Pharmacol Res 2022; 175:105980. [PMID: 34863822 PMCID: PMC8760625 DOI: 10.1016/j.phrs.2021.105980] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 11/03/2021] [Accepted: 11/09/2021] [Indexed: 01/03/2023]
Abstract
Exosomes are nano-sized lipid vesicles that are produced by all eukaryotic cells, and they typically range in size from 30 to 150 nm. Exosomes were discovered almost 40 years ago; however, the last two decades have attracted considerable attention due to exosomes' inherent abilities to shuttle nucleic acids, lipids and proteins between cells, along with their natural affinity to exosome target cells. From a pharmaceutical perspective, exosomes are regarded as naturally produced nanoparticle drug delivery vehicles. The application of exosomes as a means of drug delivery offers critical advantages compared to other nanoparticulate drug delivery systems, such as liposomes and polymeric nanoparticles. These advantages are due to the exosomes' intrinsic features, such as low immunogenicity, biocompatibility, stability, and their ability to overcome biological barriers. Herein, we outline the structure and origin of exosomes, as well as their biological functions. We also touch upon recent advances in exosome labeling, imaging and drug loading. Finally, we discuss exosomes in targeted drug delivery and clinical trial development.
Collapse
Affiliation(s)
- Zainuddin Quadri
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, United States; Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Ahmed Elsherbini
- Veterans Affairs Medical Center, Lexington, KY 40502, United States
| | - Erhard Bieberich
- Department of Physiology, University of Kentucky College of Medicine, Lexington, KY 40536, United States; Veterans Affairs Medical Center, Lexington, KY 40502, United States.
| |
Collapse
|
47
|
Thanh Nguyen TD, Marasini R, Aryal S. Re-engineered imaging agent using biomimetic approaches. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2022; 14:e1762. [PMID: 34698438 PMCID: PMC8758533 DOI: 10.1002/wnan.1762] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/25/2021] [Indexed: 01/03/2023]
Abstract
Recent progress in biomedical technology, the clinical bioimaging, has a greater impact on the diagnosis, treatment, and prevention of disease, especially by early intervention and precise therapy. Varieties of organic and inorganic materials either in the form of small molecules or nano-sized materials have been engineered as a contrast agent (CA) to enhance image resolution among different tissues for the detection of abnormalities such as cancer and vascular occlusion. Among different innovative imaging agents, contrast agents coupled with biologically derived endogenous platform shows the promising application in the biomedical field, including drug delivery and bioimaging. Strategy using biocomponents such as cells or products of cells as a delivery system predominantly reduces the toxic behavior of its cargo, as these systems reduce non-specific distribution by navigating its cargo toward the targeted location. The hypothesis is that depending on the original biological role of the naïve cell, the contrast agents carried by such a system can provide corresponding natural designated behavior. Therefore, by combining properties of conventional synthetic molecules and nanomaterials with endogenous cell body, new solutions in the field of bioimaging to overcome biological barriers have been offered as innovative bioengineering. In this review, we will discuss the engineering of cell and cell-derived components as a delivery system for various contrast agents to achieve clinically relevant contrast for diagnosis and study underlining mechanism of disease progression. This article is categorized under: Nanotechnology Approaches to Biology > Cells at the Nanoscale Diagnostic Tools > In Vivo Nanodiagnostics and Imaging Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
- Tuyen Duong Thanh Nguyen
- Department of Experimental Radiation Oncology, University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | - Ramesh Marasini
- Department of Chemistry, Nanotechnology Innovation Center of Kansas State, Kansas State Univeristy, Manhattan, KS
| | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, Texas 75799, USA
| |
Collapse
|
48
|
Radionuclide-Based Imaging of Breast Cancer: State of the Art. Cancers (Basel) 2021; 13:cancers13215459. [PMID: 34771622 PMCID: PMC8582396 DOI: 10.3390/cancers13215459] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 10/14/2021] [Accepted: 10/14/2021] [Indexed: 12/21/2022] Open
Abstract
Simple Summary Breast cancer is one of the most commonly diagnosed malignant tumors, possessing high incidence and mortality rates that threaten women’s health. Thus, early and effective breast cancer diagnosis is crucial for enhancing the survival rate. Radionuclide molecular imaging displays its advantages for detecting breast cancer from a functional perspective. Noninvasive visualization of biological processes with radionuclide-labeled small metabolic compounds helps elucidate the metabolic state of breast cancer, while radionuclide-labeled ligands/antibodies for receptor-targeted radionuclide molecular imaging is sensitive and specific for visualization of the overexpressed molecular markers in breast cancer. This review focuses on the most recent developments of novel radiotracers as promising tools for early breast cancer diagnosis. Abstract Breast cancer is a malignant tumor that can affect women worldwide and endanger their health and wellbeing. Early detection of breast cancer can significantly improve the prognosis and survival rate of patients, but with traditional anatomical imagine methods, it is difficult to detect lesions before morphological changes occur. Radionuclide-based molecular imaging based on positron emission tomography (PET) and single-photon emission computed tomography (SPECT) displays its advantages for detecting breast cancer from a functional perspective. Radionuclide labeling of small metabolic compounds can be used for imaging biological processes, while radionuclide labeling of ligands/antibodies can be used for imaging receptors. Noninvasive visualization of biological processes helps elucidate the metabolic state of breast cancer, while receptor-targeted radionuclide molecular imaging is sensitive and specific for visualization of the overexpressed molecular markers in breast cancer, contributing to early diagnosis and better management of cancer patients. The rapid development of radionuclide probes aids the diagnosis of breast cancer in various aspects. These probes target metabolism, amino acid transporters, cell proliferation, hypoxia, estrogen receptor (ER), progesterone receptor (PR), human epidermal growth factor receptor 2 (HER2), gastrin-releasing peptide receptor (GRPR) and so on. This article provides an overview of the development of radionuclide molecular imaging techniques present in preclinical or clinical studies, which are used as tools for early breast cancer diagnosis.
Collapse
|
49
|
Dually targeted bioinspired nanovesicle delays advanced prostate cancer tumour growth in vivo. Acta Biomater 2021; 134:559-575. [PMID: 34274531 DOI: 10.1016/j.actbio.2021.07.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/03/2023]
Abstract
Prostate cancer (PC) is second-leading cancer in men, with limited treatment options available for men with advanced and metastatic PC. Prostate-specific antigen (PSA) and prostate-specific membrane antigen (PSMA) have been exploited as therapeutic targets in PC due to their upregulation in the advanced stages of the disease. To date, several PSA- and PSMA-activatable prodrugs have been developed to reduce the systemic toxicity of existing chemotherapeutics. Bioinspired nanovesicles have been exploited in drug delivery, offering prolonged drug blood circulation and higher tumour accumulation. For the first time, this study describes the engineering of dually targeted PSA/PSMA nanovesicles for advanced PC. PSMA-targeted bioinspired hybrids were prepared by hydrating a lipid film with anti-PSMA-U937 cell membranes and DOX-PSA prodrug, followed by extrusion. The bioinspired hybrids were characterised using dynamic light scattering, transmission electron microscopy, Dot blot, flow cytometry and Western blot. Cellular binding and toxicity studies in PC cancer cell lines were carried out using flow cytometry, confocal microscopy, and resazurin assay. Finally, tumour targeting and therapeutic efficacy studies were performed in solid and metastatic C4-2B-tumor-bearing mice. Interestingly, our PSMA-targeted hybrids demonstrated high cell uptake in PSMA-expressing cells with significant accumulation in solid and metastatic C4-2B tumour tissues following intravenous administration. More promisingly, our dually targeted PSA/PSMA hybrid significantly slowed down the C4-2B tumour growth in vivo, compared to free DOX-PSA and non-targeted PSA-hybrid. Our PSA/PSMA bioinspired hybrid could offer a highly selective treatment for advanced PC with lower side effects. STATEMENT OF SIGNIFICANCE: This study investigates a new approach to treat prostate cancer using dually targeted bioinspired nanovesicle . Our bioinspired vesicles are made mainly of a human blood cell membrane with a ligand recognising a specific marker (PSMA) on the surface of the prostate cancer cells. The present work describes the successful loading of a doxorubicin prodrug linked to a PSA- activatable peptide into these targeted bioinspired nanovesicle , where the active PSA enzyme presents in these cells converts the drug to its active form. Our dually targeted PSA/PSMA hybrid vesicles has successfully improved site-specific prodrug delivery to tackle advanced prostate cancer, offering a novel and effective prostate cancer treatment.
Collapse
|
50
|
Lino MM, Simões S, Tomatis F, Albino I, Barrera A, Vivien D, Sobrino T, Ferreira L. Engineered extracellular vesicles as brain therapeutics. J Control Release 2021; 338:472-485. [PMID: 34428481 DOI: 10.1016/j.jconrel.2021.08.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 08/18/2021] [Accepted: 08/19/2021] [Indexed: 12/26/2022]
Abstract
Extracellular vesicles (EVs) are communication channels between different cell types in the brain, between the brain and the periphery and vice-versa, playing a fundamental role in physiology and pathology. The evidence that EVs might be able to cross the blood-brain barrier (BBB) make them very promising candidates as nanocarriers to treat brain pathologies. EVs contain a cocktail of bioactive factors, yet their content and surface can be further engineered to enhance their biological activity, stability and targeting ability. Native and engineered EVs have been reported for the treatment of different brain pathologies, although issues related to their modest accumulation and limited local therapeutic effect in the brain still need to be addressed. In this review, we cover the therapeutic applications of native and bioengineered EVs for brain diseases. We also review recent data about the interaction between EVs and the BBB and discuss the challenges and opportunities in clinical translation of EVs as brain therapeutics.
Collapse
Affiliation(s)
- Miguel M Lino
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Coimbra, Portugal; Faculty of Medicine, University Coimbra, 3000-548 Coimbra, Portugal
| | - Susana Simões
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Coimbra, Portugal
| | - Francesca Tomatis
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Coimbra, Portugal
| | - Inês Albino
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Coimbra, Portugal
| | - Angela Barrera
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Coimbra, Portugal
| | - Denis Vivien
- Normandie Univ, UNICAEN, INSERM U1237, Etablissement Français du Sang (EFS), Physiopathology and Imaging of Neurological Disorders (PhIND), Cyceron, Institut Blood and Brain @ Caen-Normandie (BB@C), 14000 Caen, France; Department of clinical research, Caen-Normandie University Hospital, CHU, Avenue de la côte de Nacre, Caen, France
| | - Tomas Sobrino
- Clinical Neurosciences Research Laboratory, Health Research Institute of Santiago de Compostela (IDIS), Santiago de Compostela, Spain
| | - Lino Ferreira
- CNC-Center for Neuroscience and Cell Biology, CIBB-Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, UC, Biotech Parque Tecnológico de Cantanhede, 3060-197 Coimbra, Portugal; Faculty of Medicine, University Coimbra, 3000-548 Coimbra, Portugal.
| |
Collapse
|