1
|
Afridi S, Sharma P, Choudhary F, Rizwan A, Nizam A, Parvez A, Farooqi H. Extracellular Vesicles: A New Approach to Study the Brain's Neural System and Its Diseases. Cell Biochem Biophys 2024; 82:521-534. [PMID: 38727784 DOI: 10.1007/s12013-024-01271-3] [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: 04/08/2024] [Indexed: 08/25/2024]
Abstract
In normal and pathophysiological conditions our cells secrete vesicular bodies known as extracellular particles. Extracellular vesicles are lipid-bound extracellular particles. A majority of these extracellular vesicles are linked to cell-to-cell communication. Brain consists of tightly packed neural cells. Neural cell releases extracellular vesicles in cerebrospinal fluid. Extracellular vesicle mediated crosstalk maintains neural homeostasis in the central nervous system via transferring cargos between neural cells. In neurodegenerative diseases, small extracellular vesicle transfer misfolded proteins to healthy cells in the neural microenvironment. They can also cross blood-brain barrier (BBB) and stimulate peripheral immune response inside central nervous system. In today's world different approaches employ extracellular vesicle in various therapeutics. This review gives a brief knowledge about the biological relevance of extracellular vesicles in the central nervous system and relevant advances in the translational application of EV in brain disorders.
Collapse
Affiliation(s)
- Shahid Afridi
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Pradakshina Sharma
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Furqan Choudhary
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Amber Rizwan
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Anam Nizam
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Adil Parvez
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India
| | - Humaira Farooqi
- Department of Biotechnology, School of Chemical and Life Sciences, Jamia Hamdard, Hamdard Nagar, New Delhi, 110062, India.
| |
Collapse
|
2
|
Che Shaffi S, Hairuddin ON, Mansor SF, Syafiq TMF, Yahaya BH. Unlocking the Potential of Extracellular Vesicles as the Next Generation Therapy: Challenges and Opportunities. Tissue Eng Regen Med 2024; 21:513-527. [PMID: 38598059 PMCID: PMC11087396 DOI: 10.1007/s13770-024-00634-4] [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: 11/14/2022] [Revised: 08/25/2023] [Accepted: 08/25/2023] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Mesenchymal stem cells (MSCs) have undergone extensive investigation for their potential therapeutic applications, primarily attributed to their paracrine activity. Recently, researchers have been exploring the therapeutic potential of extracellular vesicles (EVs) released by MSCs. METHODS MEDLINE/PubMed and Google scholar databases were used for the selection of literature. The keywords used were mesenchymal stem cells, extracellular vesicles, clinical application of EVs and challenges EVs production. RESULTS These EVs have demonstrated robust capabilities in transporting intracellular cargo, playing a critical role in facilitating cell-to-cell communication by carrying functional molecules, including proteins, RNA species, DNAs, and lipids. Utilizing EVs as an alternative to stem cells offers several benefits, such as improved safety, reduced immunogenicity, and the ability to traverse biological barriers. Consequently, EVs have emerged as an increasingly attractive option for clinical use. CONCLUSION From this perspective, this review delves into the advantages and challenges associated with employing MSC-EVs in clinical settings, with a specific focus on their potential in treating conditions like lung diseases, cancer, and autoimmune disorders.
Collapse
Affiliation(s)
- Syahidatulamali Che Shaffi
- Lung Stem Cell and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Kepala Batas, Penang, Malaysia
| | - Omar Nafiis Hairuddin
- Lung Stem Cell and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Kepala Batas, Penang, Malaysia
| | - Siti Farizan Mansor
- Lung Stem Cell and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Kepala Batas, Penang, Malaysia
- Faculty of Health Sciences, Universiti Teknologi MARA, Cawangan Pulau Pinang, Kampus Bertam, 13200, Kepala Batas, Penang, Malaysia
| | - Tengku Muhamad Faris Syafiq
- IIUM Molecular and Cellular Biology Research, Department of Basic Medical Sciences, Kulliyyah of Nursing, International Islamic University Malaysia, 25100, Kuantan, Pahang, Malaysia
| | - Badrul Hisham Yahaya
- Lung Stem Cell and Gene Therapy Group, Department of Biomedical Sciences, Advanced Medical and Dental Institute (IPPT), SAINS@BERTAM, Universiti Sains Malaysia, 13200, Kepala Batas, Penang, Malaysia.
| |
Collapse
|
3
|
Louka E, Koumandou VL. The Emerging Role of Human Gut Bacteria Extracellular Vesicles in Mental Disorders and Developing New Pharmaceuticals. Curr Issues Mol Biol 2024; 46:4751-4767. [PMID: 38785554 PMCID: PMC11120620 DOI: 10.3390/cimb46050286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 05/08/2024] [Accepted: 05/13/2024] [Indexed: 05/25/2024] Open
Abstract
In recent years, further evidence has emerged regarding the involvement of extracellular vesicles in various human physiopathological conditions such as Alzheimer's disease, Parkinson's disease, irritable bowel syndrome, and mental disorders. The biogenesis and cargo of such vesicles may reveal their impact on human health nd disease and set the underpinnings for the development of novel chemical compounds and pharmaceuticals. In this review, we examine the link between bacteria-derived exosomes in the gastrointestinal tract and mental disorders, such as depression and anxiety disorders. Crucially, we focus on whether changes in the gut environment affect the human mental state or the other way around. Furthermore, the possibility of handling bacteria-derived exosomes as vectors of chemicals to treat such conditions is examined.
Collapse
Affiliation(s)
- Effrosyni Louka
- Genetics Laboratory, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| | - Vassiliki Lila Koumandou
- Genetics Laboratory, Department of Biotechnology, Agricultural University of Athens, Iera Odos 75, 11855 Athens, Greece
| |
Collapse
|
4
|
Spadin FS, Gergely LP, Kämpfer T, Frenz M, Vermathen M. Fluorescence lifetime imaging and phasor analysis of intracellular porphyrinic photosensitizers applied with different polymeric formulations. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY. B, BIOLOGY 2024; 254:112904. [PMID: 38579534 DOI: 10.1016/j.jphotobiol.2024.112904] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 03/06/2024] [Accepted: 04/01/2024] [Indexed: 04/07/2024]
Abstract
The fluorescence lifetime of a porphyrinic photosensitizer (PS) is an important parameter to assess the aggregation state of the PS even in complex biological environments. Aggregation-induced quenching of the PS can significantly reduce the yield of singlet oxygen generation and thus its efficiency as a medical drug in photodynamic therapy (PDT) of diseased tissues. Hydrophobicity and the tendency to form aggregates pose challenges on the development of efficient PSs and often require carrier systems. A systematic study was performed to probe the impact of PS structure and encapsulation into polymeric carriers on the fluorescence lifetime in solution and in the intracellular environment. Five different porphyrinic PSs including chlorin e6 (Ce6) derivatives and tetrakis(m-hydroxyphenyl)-porphyrin and -chlorin were studied in free form and combined with polyvinylpyrrolidone (PVP) or micelles composed of triblock-copolymers or Cremophor. Following incubation of HeLa cells with these systems, fluorescence lifetime imaging combined with phasor analysis and image segmentation was applied to study the lifetime distribution in the intracellular surrounding. The data suggest that for free PSs, the structure-dependent cell uptake pathways determine their state and emission lifetimes. PS localization in the plasma membrane yielded mostly monomers with long fluorescence lifetimes whereas the endocytic pathway with subsequent lysosomal deposition adds a short-lived component for hydrophilic anionic PSs. Prolonged incubation times led to increasing contributions from short-lived components that derive from aggregates mainly localized in the cytoplasm. Encapsulation of PSs into polymeric carriers led to monomerization and mostly fluorescence emission decays with long fluorescence lifetimes in solution. However, the efficiency depended on the binding strength that was most pronounced for PVP. In the cellular environment, PVP was able to maintain monomeric long-lived species over prolonged incubation times. This was most pronounced for Ce6 derivatives with a logP value around 4.5. Micellar encapsulation led to faster release of the PSs resulting in multiple components with long and short fluorescence lifetimes. The hydrophilic hardly aggregating PS exhibited a mostly stable invariant lifetime distribution over time with both carriers. The presented data are expected to contribute to optimized PDT treatment protocols and improved PS-carrier design for preventing intracellular fluorescence quenching. In conclusion, amphiphilic and concurrent hydrophobic PSs with high membrane affinity as well as strong binding to the carrier have best prospects to maintain their photophysical properties in vivo and serve thus as efficient photodynamic diagnosis and PDT drugs.
Collapse
Affiliation(s)
- Florentin S Spadin
- Institute of Applied Physics, University of Bern, Sidlerstrasse 5, 3012 Bern, Switzerland
| | - Lea P Gergely
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, Freiestrasse 3, 3012 Bern, Switzerland
| | - Tobias Kämpfer
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, Freiestrasse 3, 3012 Bern, Switzerland
| | - Martin Frenz
- Institute of Applied Physics, University of Bern, Sidlerstrasse 5, 3012 Bern, Switzerland.
| | - Martina Vermathen
- Department of Chemistry, Biochemistry and Pharmaceutical Sciences, Freiestrasse 3, 3012 Bern, Switzerland.
| |
Collapse
|
5
|
Khaled YS, Khot MI, Aiyappa-Maudsley R, Maisey T, Pramanik A, Tiernan J, Lintern N, Al-Enezi E, Shamsuddin SH, Tomlinson D, Coletta L, Millner PA, Hughes TA, Jayne DG. Photoactive imaging and therapy for colorectal cancer using a CEA-Affimer conjugated Foslip nanoparticle. NANOSCALE 2024; 16:7185-7199. [PMID: 38506227 PMCID: PMC10993305 DOI: 10.1039/d3nr04118b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 10/08/2023] [Indexed: 03/21/2024]
Abstract
Theranostic nanoparticles hold promise for simultaneous imaging and therapy in colorectal cancer. Carcinoembryonic antigen can be used as a target for these nanoparticles because it is overexpressed in most colorectal cancers. Affimer reagents are synthetic proteins capable of binding specific targets, with additional advantages over antibodies for targeting. We fabricated silica nanoparticles using a water-in-oil microemulsion technique, loaded them with the photosensitiser Foslip, and functionalised the surface with anti-CEA Affimers to facilitate fluorescence imaging and photodynamic therapy of colorectal cancer. CEA-specific fluorescence imaging and phototoxicity were quantified in colorectal cancer cell lines and a LS174T murine xenograft colorectal cancer model. Anti-CEA targeted nanoparticles exhibited CEA-specific fluorescence in the LoVo, LS174T and HCT116 cell lines when compared to control particles (p < 0.0001). No toxicity was observed in LS174T cancer mouse xenografts or other organs. Following photo-irradiation, the anti-CEA targeted particles caused significant cell death in LoVo (60%), LS174T (90%) and HCT116 (70%) compared to controls (p < 0.0001). Photodynamic therapy (PDT) at 24 h in vivo showed a 4-fold reduction in tumour volume compared to control mouse xenografts (p < 0.0001). This study demonstrates the efficacy of targeted fluorescence imaging and PDT using Foslip nanoparticles conjugated to anti-CEA Affimer nanoparticles in in vitro and in vivo colorectal cancer models.
Collapse
Affiliation(s)
- Yazan S Khaled
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom.
| | - M Ibrahim Khot
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom.
| | | | - Thomas Maisey
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom.
| | - Arindam Pramanik
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom.
| | - Jim Tiernan
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom.
| | - Nicole Lintern
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Eiman Al-Enezi
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Shazana H Shamsuddin
- Department of Pathology, School of Medical Sciences, University Sains Malaysia, Malaysia
| | - Darren Tomlinson
- School of Molecular and Cellular Biology, University of Leeds, Leeds, UK
| | - Louise Coletta
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom.
| | - Paul A Millner
- School of Biomedical Sciences, University of Leeds, Leeds, UK
| | - Thomas A Hughes
- School of Medicine, University of Leeds, Leeds, UK
- School of Science, Technology and Health, York St John University, York, UK
| | - David G Jayne
- Leeds Institute of Medical Research, St James's University Hospital, Leeds, United Kingdom.
| |
Collapse
|
6
|
Cheng HT, Ngoc Ta YN, Hsia T, Chen Y. A quantitative review of nanotechnology-based therapeutics for kidney diseases. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1953. [PMID: 38500369 DOI: 10.1002/wnan.1953] [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: 01/12/2023] [Revised: 02/20/2024] [Accepted: 02/21/2024] [Indexed: 03/20/2024]
Abstract
Kidney-specific nanocarriers offer a targeted approach to enhance therapeutic efficacy and reduce off-target effects in renal treatments. The nanocarriers can achieve organ or cell specificity via passive targeting and active targeting mechanisms. Passive targeting capitalizes on the unique physiological traits of the kidney, with factors like particle size, charge, shape, and material properties enhancing organ specificity. Active targeting, on the other hand, achieves renal specificity through ligand-receptor interactions, modifying nanocarriers with molecules, peptides, or antibodies for receptor-mediated delivery. Nanotechnology-enabled therapy targets diseased kidney tissue by modulating podocytes and immune cells to reduce inflammation and enhance tissue repair, or by inhibiting myofibroblast differentiation to mitigate renal fibrosis. This review summarizes the current reports of the drug delivery systems that have been tested in vivo, identifies the nanocarriers that may preferentially accumulate in the kidney, and quantitatively compares the efficacy of various cargo-carrier combinations to outline optimal strategies and future research directions. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Hui-Teng Cheng
- Department of Internal Medicine, National Taiwan University Hospital Hsin-Chu Branch, Zhu Bei City, Taiwan
| | - Yen-Nhi Ngoc Ta
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- International Intercollegiate Ph.D. Program, National Tsing Hua University, Hsinchu, Taiwan
| | - Tiffaney Hsia
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, Taiwan
- Department of Chemistry, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
7
|
Jing Z, Zhang G, Cai Y, Liang J, Lv L, Dang X. Engineered extracellular vesicle-delivered TGF-β inhibitor for attenuating osteoarthritis by targeting subchondral bone. J Tissue Eng 2024; 15:20417314241257781. [PMID: 39071897 PMCID: PMC11273819 DOI: 10.1177/20417314241257781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 05/13/2024] [Indexed: 07/30/2024] Open
Abstract
Osteoarthritis (OA) is a disease that affects the entire joint. To treat OA, it may be beneficial to inhibit the activity of TGF-β in the subchondral bone. However, delivering drugs to the subchondral bone using conventional methods is challenging. In this study, we developed an extracellular vesicle delivery system. The utilization of macrophage-derived extracellular vesicles as a drug-carrying platform enables drugs to evade immune clearance and cross biological barriers. By incorporating targeting peptides on the surface of extracellular vesicles, the drug platform becomes targeted. The combination of these two factors results in the successful delivery of the drug to the subchondral bone. The study evaluated the stability, cytotoxicity, and bone targeting capability of the engineered extracellular vesicle platform (BT-EV-G). It also assessed the effects of BT-EV-G on the differentiation, proliferation, and migration of bone mesenchymal stem cells (BMSCs). Additionally, the researchers administered BT-EV-G to anterior cruciate ligament transection (ACLT)-induced OA mice. The results showed that BT-EV-G had low toxicity and high bone targeting ability both in vitro and in vivo. BT-EV-G can restore coupled bone remodeling in subchondral bone by inhibiting pSmad2/3-dependent TGF-β signaling. This work provides new insights into the treatment of OA.
Collapse
Affiliation(s)
- Zhaopu Jing
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Guangyang Zhang
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yuanqing Cai
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Jialin Liang
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Leifeng Lv
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Xiaoqian Dang
- The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
8
|
Parhi R, Kaishap PP, Jena GK. Recent advances in nanomaterial-based drug delivery systems for melanoma therapy. ADMET AND DMPK 2023; 12:107-150. [PMID: 38560710 PMCID: PMC10974823 DOI: 10.5599/admet.2088] [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: 09/15/2023] [Revised: 10/22/2023] [Indexed: 04/04/2024] Open
Abstract
Background and Purpose Safe and effective drug delivery is crucial for the treatment of cancer, which is quite impossible to achieve through traditional methods. Among all types of cancer, skin melanoma is known for its aggressive metastasizing ability and an unprecedented higher degree of lethalness, limiting the overall therapeutic efficacy. Here, we focus on the different types of nanomaterials (NMs) and their drug delivery applications against melanoma. Experimental Approach All relevant publications, including research papers, reviews, chapters and patents, were assessed using search engines such as Scopus and PubMed, up to the end of August of 2023. The keywords used in the search were: nanomaterials, melanoma, drug delivery routes for melanoma, and nanomaterial-based drug delivery systems (DDS). Most of the publications out of 234 cited in this review are from the last five years. Key Results The recent advancement and mechanism of action of various NMs against melanoma, including inorganic metallic and carbon-based NMs, organic polymeric and lipid-based NMs, and cell-derived vesicles are discussed. We also focus on the application of different NMs in the delivery of therapeutic agents for melanoma therapy. In addition, the skin and melanoma, genetic mutation and pathways for melanoma, conventional treatment options, and delivery routes for therapeutic agents are also discussed briefly. Conclusion There are few NM-based DDS developed in the lab set up recently. The findings of this review will pave the path for the development of NM-based DDS on an industrial scale and help in the better management of skin melanoma.
Collapse
Affiliation(s)
- Rabinarayan Parhi
- Department of Pharmaceutical Sciences, Susruta School of Medical and Paramedical Sciences, Assam University (A Central University), Silchar-788011, Assam, India
| | - Partha Pratim Kaishap
- Department of Pharmaceutical Sciences, Susruta School of Medical and Paramedical Sciences, Assam University (A Central University), Silchar-788011, Assam, India
| | - Goutam Kumar Jena
- Roland Institute of Pharmaceutical Sciences, Berhampur-7600010, Odisha, India
| |
Collapse
|
9
|
Millard M, Bernhard Y, Canilho N, Grandemange S, Parant S, Mourer M, Lassalle HP, Pasc A. Enhanced stability and photothermal efficiency of Indocyanine Green J-aggregates by nanoformulation with Calix[4]arene for photothermal therapy of cancers. Colloids Surf B Biointerfaces 2023; 230:113516. [PMID: 37660516 DOI: 10.1016/j.colsurfb.2023.113516] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 07/17/2023] [Accepted: 08/13/2023] [Indexed: 09/05/2023]
Abstract
Photothermal therapy (PTT) is a method of growing attention, owing to its controllable process, high efficiency and minimal side effect. Indocyanine Green (ICG) is as Food and Drug Administration (FDA) approved agent that stands on the frontline of further developments of PTT toward clinics. However, the applicability of ICG-mediated PTT is limited by the rapid in vivo clearance and photo-degradation of ICG. To improve those parameters, nanosized ICG-loaded nanoparticles (ICG-J/CX) were fabricated in this study by co-assembly of anionic ICG J-aggregates (ICG-J) with cationic tetraguanidinium calix[4]arene (CX). This very simple approach produces ICG-J/CX with a well-defined nanometer range size and a close to neutral charge. The nanoparticles demonstrate high photothermal conversion efficiency (PCE) and dramatically improved photostability, as compared with ICG. The in vitro cellular uptake and cytotoxicity studies further demonstrated that the ICG-J/CX nanoparticles enhance uptake and photothermal efficiency in comparison with ICG or non-formulated ICG-J, overall demonstrating that ICG-J/CX mediated photothermal therapy have significant potential for attaining cancer treatment.
Collapse
Affiliation(s)
- Marie Millard
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France; Université de Lorraine, CNRS UMR 7039, CRAN, F-54000 Nancy, France
| | - Yann Bernhard
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France
| | - Nadia Canilho
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France
| | | | - Stéphane Parant
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France
| | - Maxime Mourer
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France
| | - Henri-Pierre Lassalle
- Université de Lorraine, CNRS UMR 7039, CRAN, F-54000 Nancy, France; Institut de Cancérologie de Lorraine, Unité de Recherche Translationnelle F-54000 Nancy, France.
| | - Andreea Pasc
- Université de Lorraine, CNRS, L2CM UMR 7053, F-54506 Vandoeuvre-lès-Nancy, France.
| |
Collapse
|
10
|
Wang J, Liu Y, Liu F, Gan S, Roy S, Hasan I, Zhang B, Guo B. Emerging extracellular vesicle-based carriers for glioblastoma diagnosis and therapy. NANOSCALE 2023. [PMID: 37337814 DOI: 10.1039/d3nr01667f] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/21/2023]
Abstract
Glioblastoma (GBM) treatment is still a big clinical challenge because of its highly malignant, invasive, and lethal characteristics. After treatment with the conventional therapeutic paradigm of surgery combined with radio- and chemotherapy, patients bearing GBMs generally exhibit a poor prognosis, with high mortality and a high disability rate. The main reason is the existence of the formidable blood-brain barrier (BBB), aggressive growth, and the infiltration nature of GBMs. Especially, the BBB suppresses the delivery of imaging and therapeutic agents to lesion sites, and thus this leads to difficulties in achieving a timely diagnosis and treatment. Recent studies have demonstrated that extracellular vesicles (EVs) exhibit favorable merits including good biocompatibility, a strong drug loading capacity, long circulation time, good BBB crossing efficiency, specific targeting to lesion sites, and high efficiency in the delivery of a variety of cargos for GBM therapy. Importantly, EVs inherit physiological and pathological molecules from the source cells, which are ideal biomarkers for molecularly tracking the malignant progression of GBMs. Herein, we start by introducing the pathophysiology and physiology of GBMs, followed by presenting the biological functions of EVs in GBMs with a special focus on their role as biomarkers for GBM diagnosis and as messengers in the modulation of the GBM microenvironment. Furthermore, we provide an update on the recent progress of using EVs in biology, functionality, and isolation applications. More importantly, we systematically summarize the most recent advances of EV-based carriers for GBM therapy by delivering different drugs including gene/RNA-based drugs, chemotherapy drugs, imaging agents, and combinatory drugs. Lastly, we point out the challenges and prospects of future research on EVs for diagnosing and treating GBMs. We hope this review will stimulate interest from researchers with different backgrounds and expedite the progress of GBM treatment paradigms.
Collapse
Affiliation(s)
- Jingjing Wang
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Yue Liu
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Fengbo Liu
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shaoyan Gan
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Shubham Roy
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Ikram Hasan
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| | - Baozhu Zhang
- Department of Oncology, People's Hospital of Shenzhen Baoan District, The Second Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong 518055, China.
| | - Bing Guo
- Shenzhen Key Laboratory of Advanced Functional Carbon Materials Research and Comprehensive Application, Shenzhen Key Laboratory of Flexible Printed Electronics Technology, and School of Science, Harbin Institute of Technology, Shenzhen 518055, China.
- Shenzhen Key Laboratory of Flexible Printed Electronics Technology, Harbin Institute of Technology, Shenzhen 518055, China
| |
Collapse
|
11
|
Ulpiano C, da Silva CL, Monteiro GA. Bioengineered Mesenchymal-Stromal-Cell-Derived Extracellular Vesicles as an Improved Drug Delivery System: Methods and Applications. Biomedicines 2023; 11:biomedicines11041231. [PMID: 37189850 DOI: 10.3390/biomedicines11041231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/30/2023] [Accepted: 04/12/2023] [Indexed: 05/17/2023] Open
Abstract
Extracellular vesicles (EVs) are cell-derived nano-sized lipid membranous structures that modulate cell-cell communication by transporting a variety of biologically active cellular components. The potential of EVs in delivering functional cargos to targeted cells, their capacity to cross biological barriers, as well as their high modification flexibility, make them promising drug delivery vehicles for cell-free therapies. Mesenchymal stromal cells (MSCs) are known for their great paracrine trophic activity, which is largely sustained by the secretion of EVs. MSC-derived EVs (MSC-EVs) retain important features of the parental cells and can be bioengineered to improve their therapeutic payload and target specificity, demonstrating increased therapeutic potential in numerous pre-clinical animal models, including in the treatment of cancer and several degenerative diseases. Here, we review the fundamentals of EV biology and the bioengineering strategies currently available to maximize the therapeutic value of EVs, focusing on their cargo and surface manipulation. Then, a comprehensive overview of the methods and applications of bioengineered MSC-EVs is presented, while discussing the technical hurdles yet to be addressed before their clinical translation as therapeutic agents.
Collapse
Affiliation(s)
- Cristiana Ulpiano
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Cláudia L da Silva
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Gabriel A Monteiro
- Department of Bioengineering and iBB-Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy at Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| |
Collapse
|
12
|
Pourmadadi M, Mahdi Eshaghi M, Ostovar S, Mohammadi Z, K. Sharma R, Paiva-Santos AC, Rahmani E, Rahdar A, Pandey S. Innovative nanomaterials for cancer diagnosis, imaging, and therapy: Drug deliveryapplications. J Drug Deliv Sci Technol 2023. [DOI: 10.1016/j.jddst.2023.104357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2023]
|
13
|
Karmacharya M, Kumar S, Cho YK. Tuning the Extracellular Vesicles Membrane through Fusion for Biomedical Applications. J Funct Biomater 2023; 14:jfb14020117. [PMID: 36826916 PMCID: PMC9960107 DOI: 10.3390/jfb14020117] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 02/22/2023] Open
Abstract
Membrane fusion is one of the key phenomena in the living cell for maintaining the basic function of life. Extracellular vesicles (EVs) have the ability to transfer information between cells through plasma membrane fusion, making them a promising tool in diagnostics and therapeutics. This study explores the potential applications of natural membrane vesicles, EVs, and their fusion with liposomes, EVs, and cells and introduces methodologies for enhancing the fusion process. EVs have a high loading capacity, bio-compatibility, and stability, making them ideal for producing effective drugs and diagnostics. The unique properties of fused EVs and the crucial design and development procedures that are necessary to realize their potential as drug carriers and diagnostic tools are also examined. The promise of EVs in various stages of disease management highlights their potential role in future healthcare.
Collapse
Affiliation(s)
- Mamata Karmacharya
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
- Department of Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
| | - Sumit Kumar
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Correspondence: (S.K.); (Y.-K.C.)
| | - Yoon-Kyoung Cho
- Center for Soft and Living Matter, Institute for Basic Science (IBS), Ulsan 44919, Republic of Korea
- Department of Biomedical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan 44919, Republic of Korea
- Correspondence: (S.K.); (Y.-K.C.)
| |
Collapse
|
14
|
Expanding therapeutic strategies for intracellular bacterial infections through conjugates of apoptotic body-antimicrobial peptides. Drug Discov Today 2023; 28:103444. [PMID: 36400344 DOI: 10.1016/j.drudis.2022.103444] [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: 04/30/2022] [Revised: 11/03/2022] [Accepted: 11/10/2022] [Indexed: 11/17/2022]
Abstract
Macrophage intracellular infections are difficult to treat because conventional antibiotics tend to have poor penetration of mammalian cells. As a consequence, the immune response is affected and bacteria remain protected inside macrophages. The use of antimicrobial peptides (AMPs) is one of the alternatives developed as new treatments because of their broad spectrum of action. To improve drug delivery into the intracellular space, extracellular vesicles (EVs) have emerged as an innovative strategy for drug delivery. In particular, apoptotic bodies (ApoBDs) are EVs that exhibit attraction to macrophages, which makes them a promising means of improving AMP delivery to treat macrophage intracellular infections. Here, we review important aspects that should be taken into account when developing ApoBD-AMP conjugates.
Collapse
|
15
|
Huis in ‘t Veld RV, Heuts J, Ma S, Cruz LJ, Ossendorp FA, Jager MJ. Current Challenges and Opportunities of Photodynamic Therapy against Cancer. Pharmaceutics 2023; 15:pharmaceutics15020330. [PMID: 36839652 PMCID: PMC9965442 DOI: 10.3390/pharmaceutics15020330] [Citation(s) in RCA: 42] [Impact Index Per Article: 42.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/06/2023] [Accepted: 01/12/2023] [Indexed: 01/20/2023] Open
Abstract
BACKGROUND Photodynamic therapy (PDT) is an established, minimally invasive treatment for specific types of cancer. During PDT, reactive oxygen species (ROS) are generated that ultimately induce cell death and disruption of the tumor area. Moreover, PDT can result in damage to the tumor vasculature and induce the release and/or exposure of damage-associated molecular patterns (DAMPs) that may initiate an antitumor immune response. However, there are currently several challenges of PDT that limit its widespread application for certain indications in the clinic. METHODS A literature study was conducted to comprehensively discuss these challenges and to identify opportunities for improvement. RESULTS The most notable challenges of PDT and opportunities to improve them have been identified and discussed. CONCLUSIONS The recent efforts to improve the current challenges of PDT are promising, most notably those that focus on enhancing immune responses initiated by the treatment. The application of these improvements has the potential to enhance the antitumor efficacy of PDT, thereby broadening its potential application in the clinic.
Collapse
Affiliation(s)
- Ruben V. Huis in ‘t Veld
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
- Department of Radiology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
- Correspondence:
| | - Jeroen Heuts
- Department of Immunology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Sen Ma
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Luis J. Cruz
- Department of Radiology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Ferry A. Ossendorp
- Department of Immunology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| | - Martine J. Jager
- Department of Ophthalmology, Leiden University Medical Centre (LUMC), 2333 ZA Leiden, Zuid-Holland, The Netherlands
| |
Collapse
|
16
|
Ni W, Ramalingam M, Li Y, Park JH, Dashnyam K, Lee JH, Bloise N, Fassina L, Visai L, De Angelis MGC, Pedraz JL, Kim HW, Hu J. Immunomodulatory and Anti-inflammatory effect of Neural Stem/Progenitor Cells in the Central Nervous System. Stem Cell Rev Rep 2023; 19:866-885. [PMID: 36650367 DOI: 10.1007/s12015-022-10501-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2022] [Indexed: 01/19/2023]
Abstract
Neuroinflammation is a critical event that responds to disturbed homeostasis and governs various neurological diseases in the central nervous system (CNS). The excessive inflammatory microenvironment in the CNS can adversely affect endogenous neural stem cells, thereby impeding neural self-repair. Therapies with neural stem/progenitor cells (NSPCs) have shown significant inhibitory effects on inflammation, which is mainly achieved through intercellular contact and paracrine signalings. The intercellular contact between NSPCs and immune cells, the activated CNS- resident microglia, and astrocyte plays a critical role in the therapeutic NSPCs homing and immunomodulatory effects. Moreover, the paracrine effect mainly regulates infiltrating innate and adaptive immune cells, activated microglia, and astrocyte through the secretion of bioactive molecules and extracellular vesicles. However, the molecular mechanism involved in the immunomodulatory effect of NSPCs is not well discussed. This article provides a systematic analysis of the immunomodulatory mechanism of NSPCs, discusses efficient ways to enhance its immunomodulatory ability, and gives suggestions on clinical therapy.
Collapse
Affiliation(s)
- Wei Ni
- Department of Clinical Laboratory, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, 213000, Jiangsu, China.,Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China
| | - Murugan Ramalingam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea. .,School of Basic Medical Sciences, Chengdu University, Chengdu, 610106, People's Republic of China.
| | - Yumeng Li
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jeong-Hui Park
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Khandmaa Dashnyam
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea.,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea.,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Nora Bloise
- Department of Molecular Medicine, Centre for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, 27100, Pavia, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100, Pavia, Italy
| | - Lorenzo Fassina
- Department of Electrical, Computer and Biomedical Engineering, University of Pavia, 27100, Pavia, Italy
| | - Livia Visai
- Department of Molecular Medicine, Centre for Health Technologies (CHT), INSTM UdR of Pavia, University of Pavia, 27100, Pavia, Italy.,Medicina Clinica-Specialistica, UOR5 Laboratorio di Nanotecnologie, ICS Maugeri, IRCCS, 27100, Pavia, Italy
| | | | - Jose Luis Pedraz
- NanoBioCel Research Group, Laboratory of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of the Basque Country (UPV/EHU), 01006, Vitoria-Gasteiz, Spain.,Networking Research Centre of Bioengineering, Biomaterials and Nanomedicine, Institute of Health Carlos III, 28029, Madrid, Spain
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering, Dankook University, Cheonan, 31116, Republic of Korea. .,Department of Nanobiomedical Science, BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea. .,Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Jiabo Hu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
17
|
Delova A, Losantos R, Pecourneau J, Bernhard Y, Mourer M, Pasc A, Monari A. Perturbation of Lipid Bilayers by Biomimetic Photoswitches Based on Cyclocurcumin. J Chem Inf Model 2023; 63:299-307. [PMID: 36479861 DOI: 10.1021/acs.jcim.2c01152] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The use of photoswitches which may be activated by suitable electromagnetic radiation is an attractive alternative to conventional photodynamic therapy. Here, we report all-atom molecular dynamics simulation of a biomimetic photoswitch derived from cyclocurcumin and experiencing E/Z photoisomerization. In particular, we show that the two isomers interact persistently with a lipid bilayer modeling a cellular membrane. Furthermore, the interaction with the membrane is strongly dependent on the concentration, and a transition between ordered and disordered arrangements of the photoswitches is observed. We also confirm that the structural parameters of the bilayer are differently affected by the two isomers and hence can be modulated through photoswitching, offering interesting perspectives for future applications.
Collapse
Affiliation(s)
| | - Raúl Losantos
- Université Paris Cité and CNRS, ITODYS, F-75006 Paris, France.,Departamento de Química, Centro de Investigación en Síntesis Química, Universidad de La Rioja, 26006 Logroño, Spain
| | | | - Yann Bernhard
- Université de Lorraine CNRS, L2CM UMR 7053, F-54000 Nancy, France
| | - Maxime Mourer
- Université de Lorraine CNRS, L2CM UMR 7053, F-54000 Nancy, France
| | - Andreea Pasc
- Université de Lorraine CNRS, L2CM UMR 7053, F-54000 Nancy, France
| | - Antonio Monari
- Université Paris Cité and CNRS, ITODYS, F-75006 Paris, France
| |
Collapse
|
18
|
Focus on organoids: cooperation and interconnection with extracellular vesicles - Is this the future of in vitro modeling? Semin Cancer Biol 2022; 86:367-381. [PMID: 34896267 DOI: 10.1016/j.semcancer.2021.12.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/29/2021] [Accepted: 12/07/2021] [Indexed: 01/27/2023]
Abstract
Organoids are simplified in vitro model systems of organs that are used for modeling tissue development and disease, drug screening, cell therapy, and personalized medicine. Despite considerable success in the design of organoids, challenges remain in achieving real-life applications. Organoids serve as unique and organized groups of micro physiological systems that are capable of self-renewal and self-organization. Moreover, they exhibit similar organ functionality(ies) as that of tissue(s) of origin. Organoids can be designed from adult stem cells, induced pluripotent stem cells, or embryonic stem cells. They consist of most of the important cell types of the desired tissue/organ along with the topology and cell-cell interactions that are highly similar to those of an in vivo tissue/organ. Organoids have gained interest in human biomedical research, as they demonstrate high promise for use in basic, translational, and applied research. As in vitro models, organoids offer significant opportunities for reducing the reliance and use of experimental animals. In this review, we will provide an overview of organoids, as well as those intercellular communications mediated by extracellular vesicles (EVs), and discuss the importance of organoids in modeling a tumor immune microenvironment (TIME). Organoids can also be exploited to develop a better understanding of intercellular communications mediated by EVs. Also, organoids are useful in mimicking TIME, thereby offering a better-controlled environment for studying various associated biological processes and immune cell types involved in tumor immunity, such as T-cells, macrophages, dendritic cells, and myeloid-derived suppressor cells, among others.
Collapse
|
19
|
Liu A, Yang G, Liu Y, Liu T. Research progress in membrane fusion-based hybrid exosomes for drug delivery systems. Front Bioeng Biotechnol 2022; 10:939441. [PMID: 36051588 PMCID: PMC9424752 DOI: 10.3389/fbioe.2022.939441] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 07/27/2022] [Indexed: 12/12/2022] Open
Abstract
Liposomes are the earliest and most widely used nanoparticles for targeted drug delivery. Exosomes are nanosized membrane-bound particles and important mediators of intercellular communication. Combining liposomes and exosomes using various membrane fusion methods gives rise to a novel potential drug delivery system called membrane fusion-based hybrid exosomes (MFHE). These novel MFHEs not only exhibit potential advantageous features, such as high drug loading rate and targeted cellular uptake via surface modification, but are also endowed with high biocompatibility and low immunogenicity. Here, we provide an overview of MFHEs’ various preparation methods, characterization strategies, and their applications for disease treatment and scientific research.
Collapse
Affiliation(s)
- Anqi Liu
- Department of Orthodontics, Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Oral Pathology, Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, China
| | - Gang Yang
- Department of Orthodontics, Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
| | - Yuehua Liu
- Department of Orthodontics, Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- *Correspondence: Yuehua Liu, ; Tingjiao Liu,
| | - Tingjiao Liu
- Shanghai Key Laboratory of Craniomaxillofacial Development and Diseases, Fudan University, Shanghai, China
- Department of Oral Pathology, Shanghai Stomatological Hospital and School of Stomatology, Fudan University, Shanghai, China
- *Correspondence: Yuehua Liu, ; Tingjiao Liu,
| |
Collapse
|
20
|
Tang TT, Wang B, Lv LL, Dong Z, Liu BC. Extracellular vesicles for renal therapeutics: State of the art and future perspective. J Control Release 2022; 349:32-50. [PMID: 35779658 DOI: 10.1016/j.jconrel.2022.06.049] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/24/2022] [Accepted: 06/25/2022] [Indexed: 12/21/2022]
Abstract
With the ever-increasing burden of kidney disease, the need for developing new therapeutics to manage this disease has never been greater. Extracellular vesicles (EVs) are natural membranous nanoparticles present in virtually all organisms. Given their excellent delivery capacity in the body, EVs have emerged as a frontier technology for drug delivery and have the potential to usher in a new era of nanomedicine for kidney disease. This review is focused on why EVs are such compelling drug carriers and how to release their fullest potentiality in renal therapeutics. We discuss the unique features of EVs compared to artificial nanoparticles and outline the engineering technologies and steps in developing EV-based therapeutics, with an emphasis on the emerging approaches to target renal cells and prolong kidney retention. We also explore the applications of EVs as natural therapeutics or as drug carriers in the treatment of renal disorders and present our views on the critical challenges in manufacturing EVs as next-generation renal therapeutics.
Collapse
Affiliation(s)
- Tao-Tao Tang
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China; Department of Pathology and Pathophysiology, Southeast University School of Medicine, Nanjing, China
| | - Bin Wang
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China
| | - Lin-Li Lv
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China.
| | - Zheng Dong
- Department of Cellular Biology and Anatomy, Medical College of Georgia at Augusta University and Charlie Norwood VA Medical Center, Augusta, GA, USA
| | - Bi-Cheng Liu
- Institute of Nephrology, Zhong Da Hospital, Nanjing, China.
| |
Collapse
|
21
|
Wang J, Wang X, Zhang X, Shao T, Luo Y, Wang W, Han Y. Extracellular Vesicles and Hepatocellular Carcinoma: Opportunities and Challenges. Front Oncol 2022; 12:884369. [PMID: 35692794 PMCID: PMC9175035 DOI: 10.3389/fonc.2022.884369] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Accepted: 04/25/2022] [Indexed: 12/05/2022] Open
Abstract
The incidence of hepatocellular carcinoma (HCC) is increasing worldwide. Extracellular vesicles (EVs) contain sufficient bioactive substances and are carriers of intercellular information exchange, as well as delivery vehicles for nucleic acids, proteins and drugs. Although EVs show great potential for the treatment of HCC and their role in HCC progression has been extensively studied, there are still many challenges such as time-consuming extraction, difficult storage, easy contamination, and low drug loading rate. We focus on the biogenesis, morphological characteristics, isolation and extraction of EVs and their significance in the progression of HCC, tumor invasion, immune escape and cancer therapy for a review. EVs may be effective biomarkers for molecular diagnosis of HCC and new targets for tumor-targeted therapy.
Collapse
Affiliation(s)
- Juan Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiaoya Wang
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Xintong Zhang
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Tingting Shao
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yanmei Luo
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Wei Wang
- Clinical Medicine, Southwest Medical University, Luzhou, China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China.,Department of Oncology, The Affiliated Hospital of Southwest Medical University, Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Academician (Expert) Workstation of Sichuan Province, Luzhou, China.,Nuclear Medicine and Molecular Imaging Key Laboratory of Sichuan Province, Luzhou, China.,School of Basic Medical Sciences, Shandong University, Jinan, China
| |
Collapse
|
22
|
Fisher WS, Tchounwou C, Wei S, Roberts L, Ewert KK, Safinya CR. Exosomes are secreted at similar densities by M21 and PC3 human cancer cells and show paclitaxel solubility. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183841. [PMID: 34953781 PMCID: PMC8896395 DOI: 10.1016/j.bbamem.2021.183841] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 12/08/2021] [Accepted: 12/11/2021] [Indexed: 06/14/2023]
Abstract
Exosomes are cell-secreted vesicles less than ≈150 nm in size that contain gene-encoding and gene-silencing RNA and cytosolic proteins with roles in intercellular communication. Interest in the use of exosomes as targeted drug delivery vehicles has grown since it was shown that they can bind specific cells and deliver intact genetic material to the cytosol of target cells. We isolated extracellular vesicles (EVs), consisting of a mixture of exosomes and microvesicles, from prostate (PC3) and melanoma (M21) cancer cell lines using serial ultracentrifugation. Interrogation via western blot analysis confirmed enrichment of CD63, a widely recognized EV surface protein, in the EV pellet from both cell lines. Nanoparticle tracking analysis (NTA) of EV pellets revealed that the two cell lines produced distinct vesicle size profiles in the ≈30 nm to ≈400 nm range. NTA further showed that the fraction of exosomes to all EVs was constant, suggesting cellular mechanisms that control the fraction of secreted vesicles that are exosomes. Transmission electron microscopy (TEM) images of the unmodified PC3 EVs showed vesicles with cup-like (i.e., nanocapsule) and previously unreported prolate morphologies. The observed non-spherical morphologies for dehydrated exosomal vesicles (size ≈30-100 nm) are most likely related to the dense packing of proteins in exosome membranes. Solubility phase diagram data showed that EVs enhanced the solubility of paclitaxel (PTX) in aqueous solution compared to a water-only control. Combined with their inherent targeting and cytosol delivery properties, these findings highlight the potential advantages of using exosomes as chemotherapeutic drug carriers in vivo.
Collapse
Affiliation(s)
- William S Fisher
- Materials Department, Molecular, Cellular, and Developmental Biology Department, Physics Department, and Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA 93106, USA
| | - Christine Tchounwou
- Materials Department, Molecular, Cellular, and Developmental Biology Department, Physics Department, and Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA 93106, USA
| | - Sophia Wei
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| | - Logan Roberts
- Molecular, Cellular, and Developmental Biology Department, University of California, Santa Barbara, CA 93106, USA
| | - Kai K Ewert
- Materials Department, Molecular, Cellular, and Developmental Biology Department, Physics Department, and Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA 93106, USA
| | - Cyrus R Safinya
- Materials Department, Molecular, Cellular, and Developmental Biology Department, Physics Department, and Biomolecular Science and Engineering Program, University of California, Santa Barbara, CA 93106, USA.
| |
Collapse
|
23
|
van der Koog L, Gandek TB, Nagelkerke A. Liposomes and Extracellular Vesicles as Drug Delivery Systems: A Comparison of Composition, Pharmacokinetics, and Functionalization. Adv Healthc Mater 2022; 11:e2100639. [PMID: 34165909 PMCID: PMC11468589 DOI: 10.1002/adhm.202100639] [Citation(s) in RCA: 132] [Impact Index Per Article: 66.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Revised: 05/27/2021] [Indexed: 12/11/2022]
Abstract
Over the past decades, lipid-based nanoparticle drug delivery systems (DDS) have caught the attention of researchers worldwide, encouraging the field to rapidly develop improved ways for effective drug delivery. One of the most prominent examples is liposomes, which are spherical shaped artificial vesicles composed of lipid bilayers and able to encapsulate both hydrophilic and hydrophobic materials. At the same time, biological nanoparticles naturally secreted by cells, called extracellular vesicles (EVs), have emerged as promising more complex biocompatible DDS. In this review paper, the differences and similarities in the composition of both vesicles are evaluated, and critical mediators that affect their pharmacokinetics are elucidate. Different strategies that have been assessed to tweak the pharmacokinetics of both liposomes and EVs are explored, detailing the effects on circulation time, targeting capacity, and cytoplasmic delivery of therapeutic cargo. Finally, whether a hybrid system, consisting of a combination of only the critical constituents of both vesicles, could offer the best of both worlds is discussed. Through these topics, novel leads for further research are provided and, more importantly, gain insight in what the liposome field and the EV field can learn from each other.
Collapse
Affiliation(s)
- Luke van der Koog
- Molecular PharmacologyGroningen Research Institute of PharmacyGRIAC Research Institute, University Medical Center GroningenUniversity of GroningenP.O. Box 196, XB10Groningen9700 ADThe Netherlands
| | - Timea B. Gandek
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| | - Anika Nagelkerke
- Pharmaceutical AnalysisGroningen Research Institute of PharmacyUniversity of GroningenP.O. Box 196, XB20Groningen9700 ADThe Netherlands
| |
Collapse
|
24
|
Wang BZ, Luo L, Vunjak-Novakovic G. RNA and Protein Delivery by Cell-Secreted and Bioengineered Extracellular Vesicles. Adv Healthc Mater 2022; 11:e2101557. [PMID: 34706168 PMCID: PMC8891029 DOI: 10.1002/adhm.202101557] [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] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Revised: 09/28/2021] [Indexed: 12/22/2022]
Abstract
Extracellular vesicles (EVs) are carriers of biological signals through export and delivery of RNAs and proteins. Of increasing interest is the use of EVs as a platform for delivery of biomolecules. Preclinical studies have effectively used EVs to treat a number of diseases. Uniquely, endogenous machinery within cells can be manipulated in order to produce desirable loading of cargo within secreted EVs. In order to inform the development of such approaches, an understanding of the cellular mechanisms by which cargo is sorted to EVs is required. Here, the current knowledge of cargo sorting within EVs is reviewed. Here is given an overview of recent bioengineering approaches that leverage these advances. Methods of externally manipulating EV cargo are also discussed. Finally, a perspective on the current challenges of EVs as a drug delivery platform is offered. It is proposed that standardized bioengineering methods for therapeutic EV preparation will be required to create a well-defined clinical product.
Collapse
Affiliation(s)
- Bryan Z. Wang
- Department of Biomedical Engineering, 622 West 168th Street VC12-234, 10032, U.S.A
- Department of Medicine, 622 West 168th Street VC12-234, 10032, U.S.A
| | - Lori Luo
- Department of Medicine, 622 West 168th Street VC12-234, 10032, U.S.A
| | - Gordana Vunjak-Novakovic
- Department of Biomedical Engineering, 622 West 168th Street VC12-234, 10032, U.S.A
- Department of Medicine, 622 West 168th Street VC12-234, 10032, U.S.A
| |
Collapse
|
25
|
Kamerkar S, Leng C, Burenkova O, Jang SC, McCoy C, Zhang K, Dooley K, Kasera S, Zi T, Sisó S, Dahlberg W, Sia CL, Patel S, Schmidt K, Economides K, Soos T, Burzyn D, Sathyanarayanan S. Exosome-mediated genetic reprogramming of tumor-associated macrophages by exoASO-STAT6 leads to potent monotherapy antitumor activity. SCIENCE ADVANCES 2022; 8:eabj7002. [PMID: 35179953 PMCID: PMC8856615 DOI: 10.1126/sciadv.abj7002] [Citation(s) in RCA: 120] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 12/23/2021] [Indexed: 05/04/2023]
Abstract
Effectiveness of checkpoint immunotherapy in cancer can be undermined by immunosuppressive tumor-associated macrophages (TAMs) with an M2 phenotype. Reprogramming TAMs toward a proinflammatory M1 phenotype is a novel approach to induce antitumor immunity. The M2 phenotype is controlled by key transcription factors such as signal transducer and activator of transcription 6 (STAT6), which have been "undruggable" selectively in TAMs. We describe an engineered exosome therapeutic candidate delivering an antisense oligonucleotide (ASO) targeting STAT6 (exoASO-STAT6), which selectively silences STAT6 expression in TAMs. In syngeneic models of colorectal cancer and hepatocellular carcinoma, exoASO-STAT6 monotherapy results in >90% tumor growth inhibition and 50 to 80% complete remissions. Administration of exoASO-STAT6 leads to induction of nitric oxide synthase 2 (NOS2), an M1 macrophage marker, resulting in remodeling of the tumor microenvironment and generation of a CD8 T cell-mediated adaptive immune response. Collectively, exoASO-STAT6 represents the first platform targeting transcription factors in TAMs in a highly selective manner.
Collapse
Affiliation(s)
| | - Charan Leng
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | | | - Su Chul Jang
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | | | - Kelvin Zhang
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | - Kevin Dooley
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | | | - Tong Zi
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | - Sílvia Sisó
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | | | | | - Shil Patel
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | - Karl Schmidt
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | | | - Timothy Soos
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | - Dalia Burzyn
- Codiak BioSciences Inc., Cambridge, MA 02140, USA
| | | |
Collapse
|
26
|
Thomas SC, Kim JW, Pauletti GM, Hassett DJ, Kotagiri N. Exosomes: Biological Pharmaceutical Nanovectors for Theranostics. Front Bioeng Biotechnol 2022; 9:808614. [PMID: 35096795 PMCID: PMC8790084 DOI: 10.3389/fbioe.2021.808614] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Accepted: 12/24/2021] [Indexed: 12/12/2022] Open
Abstract
Exosomes are natural cell-derived nanovesicles of endocytic origin that enable cellular crosstalk by transferring encapsulated molecular cargos across biological barriers, thereby holding significantly complex implications in the etiology and progression of diverse disease states. Consequently, the development of exosomes-based nano-theranostic strategies has received immense consideration for advancing therapeutic interventions and disease prognosis. Their favorable biopharmaceutical properties make exosomes a unique nanoparticulate carrier for pharmaceutical drug delivery. This review provides an update on the contemporary strategies utilizing exosomes for theranostic applications in nanomedicine. In addition, we provide a synopsis of exosomal features and insights into strategic modifications that control in vivo biodistribution. We further discuss their opportunities, merits and pitfalls for cell/tissue targeted drug delivery in personalized nanotherapy.
Collapse
Affiliation(s)
- Shindu C Thomas
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| | - Jin-Woo Kim
- Department of Biological and Agricultural Engineering, Institute for Nanoscience and Engineering, University of Arkansas, Fayetteville, AR, United States
| | - Giovanni M Pauletti
- St. Louis College of Pharmacy, University of Health Sciences and Pharmacy in St. Louis, St. Louis, MO, United States
| | - Daniel J Hassett
- Department of Molecular Genetics, Biochemistry and Microbiology, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Nalinikanth Kotagiri
- Division of Pharmaceutical Sciences, James L. Winkle College of Pharmacy, University of Cincinnati, Cincinnati, OH, United States
| |
Collapse
|
27
|
Limongi T, Susa F, Marini M, Allione M, Torre B, Pisano R, di Fabrizio E. Lipid-Based Nanovesicular Drug Delivery Systems. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:3391. [PMID: 34947740 PMCID: PMC8707227 DOI: 10.3390/nano11123391] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Revised: 12/07/2021] [Accepted: 12/13/2021] [Indexed: 12/14/2022]
Abstract
In designing a new drug, considering the preferred route of administration, various requirements must be fulfilled. Active molecules pharmacokinetics should be reliable with a valuable drug profile as well as well-tolerated. Over the past 20 years, nanotechnologies have provided alternative and complementary solutions to those of an exclusively pharmaceutical chemical nature since scientists and clinicians invested in the optimization of materials and methods capable of regulating effective drug delivery at the nanometer scale. Among the many drug delivery carriers, lipid nano vesicular ones successfully support clinical candidates approaching such problems as insolubility, biodegradation, and difficulty in overcoming the skin and biological barriers such as the blood-brain one. In this review, the authors discussed the structure, the biochemical composition, and the drug delivery applications of lipid nanovesicular carriers, namely, niosomes, proniosomes, ethosomes, transferosomes, pharmacosomes, ufasomes, phytosomes, catanionic vesicles, and extracellular vesicles.
Collapse
|
28
|
Engineering and loading therapeutic extracellular vesicles for clinical translation: A data reporting frame for comparability. Adv Drug Deliv Rev 2021; 178:113972. [PMID: 34509573 DOI: 10.1016/j.addr.2021.113972] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/06/2021] [Accepted: 09/07/2021] [Indexed: 02/08/2023]
Abstract
Extracellular vesicles (EVs) have emerged as new drug delivery systems as well as a regenerative cell-free effectors going beyond academic research to reach industrial research and development (R&D). Many proof-of-concept studies are now published describing the delivery of drugs, nanoparticles or biologics among which nucleic acids, proteins, viruses, etc. Their main interests rely on their intrinsic biocompatibility, targeting capabilities and biological activities. The possibility of loading EVs with exogenous therapeutic drug/nanoparticles or imaging tracers opens up the perspectives to extend EV therapeutic properties and enable EV tracking. Clinical translation is still hampered by the difficulty to produce and load EVs with large scale, efficient and cGMP methods. In this review, we critically discuss important notions related to EV engineering and the methods available with a particular focus on technologies fitted for clinical translation. Besides, we provide a tentative data reporting frame in order to support comparability and standardization in the field.
Collapse
|
29
|
Functional intersections between extracellular vesicles and oncolytic therapies. Trends Pharmacol Sci 2021; 42:883-896. [PMID: 34598797 DOI: 10.1016/j.tips.2021.09.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 08/30/2021] [Accepted: 09/01/2021] [Indexed: 01/15/2023]
Abstract
Minimally invasive focal therapies for nonviral oncolysis are a cornerstone of cancer therapeutics. Our ability to optimally deploy oncolytic therapies and identify synergistic combination approaches requires a deeper understanding of elicited biological responses. Extracellular vesicles (EV), which orchestrate a variety of pathophysiological processes and have a critical role in the evolution of primary and disseminated tumors, are now known to be potently modulated by oncolytic focal therapies, such as radiotherapy, photodynamic therapy (PDT), and therapeutic ultrasound (TUS). In this review, we summarize the diverse impacts of the aforementioned therapeutic modalities on EV biology, and highlight the most recent advances in EV-based drug delivery systems leveraging these modalities.
Collapse
|
30
|
Zinc-Phthalocyanine-Loaded Extracellular Vesicles Increase Efficacy and Selectivity of Photodynamic Therapy in Co-Culture and Preclinical Models of Colon Cancer. Pharmaceutics 2021; 13:pharmaceutics13101547. [PMID: 34683840 PMCID: PMC8537141 DOI: 10.3390/pharmaceutics13101547] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/07/2021] [Accepted: 09/13/2021] [Indexed: 12/19/2022] Open
Abstract
Photodynamic therapy (PDT) is a promising and clinically approved method for the treatment of cancer. However, the efficacy of PDT is often limited by the poor selectivity and distribution of the photosensitizers (PS) toward the malignant tumors, resulting in prolonged periods of skin photosensitivity. In this work, we present a simple and straightforward strategy to increase the tumor distribution, selectivity, and efficacy of lipophilic PS zinc phthalocyanine (ZnPc) in colon cancer by their stabilization in purified, naturally secreted extracellular vesicles (EVs). The PS ZnPc was incorporated in EVs (EV-ZnPc) by a direct incubation strategy that did not affect size distribution or surface charge. By using co-culture models simulating a tumor microenvironment, we determined the preferential uptake of EV-ZnPc toward colon cancer cells when compared with macrophages and dendritic cells. We observed that PDT promoted total tumor cell death in normal and immune cells, but showed selectivity against cancer cells in co-culture models. In vivo assays showed that after a single intravenous or intratumoral injection, EV-ZnPc were able to target the tumor cells and strongly reduce tumor growth over 15 days. These data expose opportunities to enhance the potential and efficacy of PDT using simple non-synthetic strategies that might facilitate translation into clinical practice.
Collapse
|
31
|
Jurgielewicz B, Stice S, Yao Y. Therapeutic Potential of Nucleic Acids when Combined with Extracellular Vesicles. Aging Dis 2021; 12:1476-1493. [PMID: 34527423 PMCID: PMC8407886 DOI: 10.14336/ad.2021.0708] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Accepted: 07/08/2021] [Indexed: 12/12/2022] Open
Abstract
Extracellular vesicles (EVs), endogenous nanocarriers of proteins, lipids, and genetic material, have been harnessed as intrinsic delivery vectors for nucleic acid therapies. EVs are nanosized lipid bilayer bound vesicles released from most cell types responsible for delivery of functional biologic material to mediate intercellular communication and to modulate recipient cell phenotypes. Due to their innate biological role and composition, EVs possess several advantages as delivery vectors for nucleic acid based therapies including low immunogenicity and toxicity, high bioavailability, and ability to be engineered to enhance targeting to specific recipient cells in vivo. In this review, the current understanding of the biological role of EVs as well as the advancements in loading EVs to deliver nucleic acid therapies are summarized. We discuss the current methods and associated challenges in loading EVs and the prospects of utilizing the inherent characteristics of EVs as a delivery vector of nucleic acid therapies for genetic disorders.
Collapse
Affiliation(s)
- Brian Jurgielewicz
- 1Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA.,2Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| | - Steven Stice
- 1Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA.,2Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA.,3ArunA Bio, Athens, GA 30602, USA
| | - Yao Yao
- 1Regenerative Bioscience Center, University of Georgia, Athens, GA 30602, USA.,2Department of Animal and Dairy Science, College of Agricultural and Environmental Sciences, University of Georgia, Athens, GA 30602, USA
| |
Collapse
|
32
|
Nucleic acid delivery with extracellular vesicles. Adv Drug Deliv Rev 2021; 173:89-111. [PMID: 33746014 DOI: 10.1016/j.addr.2021.03.005] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 02/16/2021] [Accepted: 03/08/2021] [Indexed: 12/14/2022]
Abstract
Extracellular vesicles (EVs) are membrane-enclosed particles, heterogeneous in size, shape, contents, biogenesis and structure. They are released by eukaryotic and prokaryotic cells and exert (patho-)physiological roles as mediators for transmitting molecular information from the producer (donor) to a recipient cell. This review focuses on the potential of EVs for delivering nucleic acids, as particularly problematic cargoes with regard to stability/protection and uptake efficacy. It highlights important properties of EVs for nucleic acid delivery and discusses their physiological and pathophysiological roles with regard to various cellular RNA species. It then describes the application of EVs for delivering a broad selection of nucleic acids/oligonucleotides, in particular giving a comprehensive overview of preclinical in vivo studies and the various strategies explored. In this context, different techniques for EV loading are discussed, as well as other important technical aspects related to EV preparation, characterization and in particular, the various approaches of artificial EV modification.
Collapse
|
33
|
Pinto A, Marangon I, Méreaux J, Nicolás-Boluda A, Lavieu G, Wilhelm C, Sarda-Mantel L, Silva AKA, Pocard M, Gazeau F. Immune Reprogramming Precision Photodynamic Therapy of Peritoneal Metastasis by Scalable Stem-Cell-Derived Extracellular Vesicles. ACS NANO 2021; 15:3251-3263. [PMID: 33481565 DOI: 10.1021/acsnano.0c09938] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
The dissemination of tumor metastasis in the peritoneal cavity, also called peritoneal metastasis (PM) or carcinomatosis, represents a late stage of gastrointestinal and gynecological cancer with very poor prognosis, even when cytoreductive surgery is effective, due to residual microscopic disease. Photodynamic therapy (PDT) in the management of peritoneal metastasis has been clinically limited by the low tumor selectivity of photosensitizers (PS) and important adverse effects. Here, we propose extracellular nanovesicles (EVs) derived from mesenchymal stem/stromal cells (MSCs) as the fourth generation of immune active PS vectors that are able to target peritoneal metastasis with superior selectivity, potentiate PDT cytotoxicity at the tumor site without affecting healthy tissues, modulate the tumor microenvironment of immunocompetent colorectal and ovarian carcinomatosis models, and promote an antitumor immune response. A pioneering strategy was developed for high yield, large-scale production of MSC-EVs encapsulating the drug meta(tetrahydroxyphenyl)chlorin (mTHPC) (EVs-mTHPC) that is compatible with requirements of clinical translation and also preserves the topology and integrity of naturally produced EVs. Intraperitoneal injection of EVs-mTHPC showed an impressive enhancement of tumoral selectivity in comparison to the free drug and to the liposomal formulation Foslip (mean ratio of PS in tumors/organs of 40 for EVs-mTHPC versus 1.5 for the free PS and 5.5 for Foslip). PDT mediated by EVs-mTHPC permitted an important tumoral necrosis (55% of necrotic tumoral nodules versus 18% for Foslip (p < 0.0001)) and promoted antitumor immune cell infiltration, mainly proinflammatory M1-like CD80+ and CD8+ T cell effector. Intratumor proliferation was significantly decreased after PDT with EVs-mTHPC. Overall EVs vectorization of mTHPC afforded important tumoral selectivity while overcoming the PDT toxicity of the free drug and prolonged mice survival in the colorectal carcinomatosis model. MSC-EVs produced by our scalable manufacturing method appears like the clinically relevant fourth-generation PDT vehicle to overcome current limitations of PDT in the treatment of peritoneal metastasis and promote a hot tumor immune environment in PM.
Collapse
Affiliation(s)
- Amandine Pinto
- Inserm UMR 1275 CAP Paris-Tech, Université de Paris, F-75010 Paris, France
- Service de Chirurgie Digestive et Cancérologique, Hôpital Lariboisière AP-HP, 2 rue Ambroise Paré, F-75010 Paris, France
| | - Iris Marangon
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - Julie Méreaux
- Inserm UMR 1275 CAP Paris-Tech, Université de Paris, F-75010 Paris, France
- Service de Chirurgie Digestive et Cancérologique, Hôpital Lariboisière AP-HP, 2 rue Ambroise Paré, F-75010 Paris, France
| | - Alba Nicolás-Boluda
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - Grégory Lavieu
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - Claire Wilhelm
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - Laure Sarda-Mantel
- Service de Médecine Nucléaire, Université de Paris, Hôpital Lariboisière AP-HP, 2 rue Ambroise Paré, F-75010 Paris, France
| | - Amanda K A Silva
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| | - Marc Pocard
- Inserm UMR 1275 CAP Paris-Tech, Université de Paris, F-75010 Paris, France
- Service de Chirurgie Digestive et Cancérologique, Hôpital Lariboisière AP-HP, 2 rue Ambroise Paré, F-75010 Paris, France
| | - Florence Gazeau
- Laboratoire MSC Matière et Systèmes Complexes, CNRS UMR 7057, Université de Paris, 10 Rue Alice Domon et Léonie Duquet, 75205 Paris Cedex 13, France
| |
Collapse
|
34
|
Han Y, Jones TW, Dutta S, Zhu Y, Wang X, Narayanan SP, Fagan SC, Zhang D. Overview and Update on Methods for Cargo Loading into Extracellular Vesicles. Processes (Basel) 2021; 9. [PMID: 33954091 PMCID: PMC8096148 DOI: 10.3390/pr9020356] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The enormous library of pharmaceutical compounds presents endless research avenues. However, several factors limit the therapeutic potential of these drugs, such as drug resistance, stability, off-target toxicity, and inadequate delivery to the site of action. Extracellular vesicles (EVs) are lipid bilayer-delimited particles and are naturally released from cells. Growing evidence shows that EVs have great potential to serve as effective drug carriers. Since EVs can not only transfer biological information, but also effectively deliver hydrophobic drugs into cells, the application of EVs as a novel drug delivery system has attracted considerable scientific interest. Recently, EVs loaded with siRNA, miRNA, mRNA, CRISPR/Cas9, proteins, or therapeutic drugs show improved delivery efficiency and drug effect. In this review, we summarize the methods used for the cargo loading into EVs, including siRNA, miRNA, mRNA, CRISPR/Cas9, proteins, and therapeutic drugs. Furthermore, we also include the recent advance in engineered EVs for drug delivery. Finally, both advantages and challenges of EVs as a new drug delivery system are discussed. Here, we encourage researchers to further develop convenient and reliable loading methods for the potential clinical applications of EVs as drug carriers in the future.
Collapse
Affiliation(s)
- Yohan Han
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Timothy W. Jones
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Saugata Dutta
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Yin Zhu
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Xiaoyun Wang
- Center for Vaccines and Immunology, University of Georgia, Athens, GA 30602, USA
| | - S. Priya Narayanan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
- James and Jean Culver Vision Discovery Institute, Augusta University, Augusta, GA 30912, USA
| | - Susan C. Fagan
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
| | - Duo Zhang
- Clinical and Experimental Therapeutics, College of Pharmacy, University of Georgia and Charlie Norwood VA Medical Center, Augusta, GA 30912, USA
- Vascular Biology Center, Augusta University, Augusta, GA 30912, USA
- Correspondence: ; Tel.: +1-706-721-6491; Fax: +1-706-721-3994
| |
Collapse
|
35
|
Khan AA, T. M. de Rosales R. Radiolabelling of Extracellular Vesicles for PET and SPECT imaging. Nanotheranostics 2021; 5:256-274. [PMID: 33654653 PMCID: PMC7914338 DOI: 10.7150/ntno.51676] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Accepted: 01/09/2021] [Indexed: 12/13/2022] Open
Abstract
Extracellular vesicles (EVs) such as exosomes and microvesicles have gained recent attention as potential biomarkers of disease as well as nanomedicinal tools, but their behaviour in vivo remains mostly unexplored. In order to gain knowledge of their in vivo biodistribution it is important to develop imaging tools that allow us to track EVs over time and at the whole-body level. Radionuclide-based imaging (PET and SPECT) have properties that allow us to do so efficiently, mostly due to their high sensitivity, imaging signal tissue penetration, and accurate quantification. Furthermore, they can be easily translated from animals to humans. In this review, we summarise and discuss the different studies that have used PET or SPECT to study the behaviour of EVs in vivo. With a focus on the different radiolabelling methods used, we also discuss the advantages and disadvantages of each one, and the challenges of imaging EVs due to their variable stability and heterogeneity.
Collapse
Affiliation(s)
| | - Rafael T. M. de Rosales
- Dept. of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, King's College London, St. Thomas' Hospital, London, United Kingdom
| |
Collapse
|
36
|
Bordanaba-Florit G, Madarieta I, Olalde B, Falcón-Pérez JM, Royo F. 3D Cell Cultures as Prospective Models to Study Extracellular Vesicles in Cancer. Cancers (Basel) 2021; 13:307. [PMID: 33467651 PMCID: PMC7830667 DOI: 10.3390/cancers13020307] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2020] [Revised: 01/10/2021] [Accepted: 01/12/2021] [Indexed: 12/12/2022] Open
Abstract
The improvement of culturing techniques to model the environment and physiological conditions surrounding tumors has also been applied to the study of extracellular vesicles (EVs) in cancer research. EVs role is not only limited to cell-to-cell communication in tumor physiology, they are also a promising source of biomarkers, and a tool to deliver drugs and induce antitumoral activity. In the present review, we have addressed the improvements achieved by using 3D culture models to evaluate the role of EVs in tumor progression and the potential applications of EVs in diagnostics and therapeutics. The most employed assays are gel-based spheroids, often utilized to examine the cell invasion rate and angiogenesis markers upon EVs treatment. To study EVs as drug carriers, a more complex multicellular cultures and organoids from cancer stem cell populations have been developed. Such strategies provide a closer response to in vivo physiology observed responses. They are also the best models to understand the complex interactions between different populations of cells and the extracellular matrix, in which tumor-derived EVs modify epithelial or mesenchymal cells to become protumor agents. Finally, the growth of cells in 3D bioreactor-like systems is appointed as the best approach to industrial EVs production, a necessary step toward clinical translation of EVs-based therapy.
Collapse
Affiliation(s)
- Guillermo Bordanaba-Florit
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Exosomes Laboratory, Basque Research and Technology Alliance (BRTA), E48160 Derio, Spain; (G.B.-F.); (J.M.F.-P.)
| | - Iratxe Madarieta
- TECNALIA Basque Research and Technology Alliance (BRTA), E20009 Donostia San Sebastian, Spain; (I.M.); (B.O.)
| | - Beatriz Olalde
- TECNALIA Basque Research and Technology Alliance (BRTA), E20009 Donostia San Sebastian, Spain; (I.M.); (B.O.)
| | - Juan M. Falcón-Pérez
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Exosomes Laboratory, Basque Research and Technology Alliance (BRTA), E48160 Derio, Spain; (G.B.-F.); (J.M.F.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), E28029 Madrid, Spain
- Ikerbasque, Basque Foundation for Science, E48009 Bilbao, Spain
| | - Félix Royo
- Center for Cooperative Research in Biosciences (CIC bioGUNE), Exosomes Laboratory, Basque Research and Technology Alliance (BRTA), E48160 Derio, Spain; (G.B.-F.); (J.M.F.-P.)
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (Ciberehd), E28029 Madrid, Spain
| |
Collapse
|
37
|
Yakavets I, Francois A, Lamy L, Piffoux M, Gazeau F, Wilhelm C, Zorin V, Silva AKA, Bezdetnaya L. Effect of stroma on the behavior of temoporfin-loaded lipid nanovesicles inside the stroma-rich head and neck carcinoma spheroids. J Nanobiotechnology 2021; 19:3. [PMID: 33407564 PMCID: PMC7789590 DOI: 10.1186/s12951-020-00743-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Accepted: 11/30/2020] [Indexed: 01/12/2023] Open
Abstract
Background Despite the highly expected clinical application of nanoparticles (NPs), the translation of NPs from lab to the clinic has been relatively slow. Co-culture 3D spheroids account for the 3D arrangement of tumor cells and stromal components, e.g., cancer-associated fibroblasts (CAFs) and extracellular matrix, recapitulating microenvironment of head and neck squamous cell carcinoma (HNSCC). In the present study, we investigated how the stroma-rich tumor microenvironment affects the uptake, penetration, and photodynamic efficiency of three lipid-based nanoformulations of approved in EU photosensitizer temoporfin (mTHPC): Foslip® (mTHPC in conventional liposomes), drug-in-cyclodextrin-in-liposomes (mTHPC-DCL) and extracellular vesicles (mTHPC-EVs). Results Collagen expression in co-culture stroma-rich 3D HNSCC spheroids correlates with the amount of CAFs (MeWo cells) in individual spheroid. The assessment of mTHPC loading demonstrated that Foslip®, mTHPC-DCL and mTHPC-EVs encapsulated 0.05 × 10− 15 g, 0.07 × 10− 15 g, and 1.3 × 10− 15 g of mTHPC per nanovesicle, respectively. The mid-penetration depth of mTHPC NPs in spheroids was 47.8 µm (Foslip®), 87.8 µm (mTHPC-DCL), and 49.7 µm (mTHPC-EVs), irrespective of the percentage of stromal components. The cellular uptake of Foslip® and mTHPC-DCL was significantly higher in stroma-rich co-culture spheroids and was increasing upon the addition of serum in the culture medium. Importantly, we observed no significant difference between PDT effect in monoculture and co-culture spheroids treated with lipid-based NPs. Overall, in all types of spheroids mTHPC-EVs demonstrated outstanding total cellular uptake and PDT efficiency comparable to other NPs. Conclusions The stromal microenvironment strongly affects the uptake of NPs, while the penetration and PDT efficacy are less sensitive to the presence of stromal components. mTHPC-EVs outperform other lipid nanovesicles due to the extremely high loading capacity. The results of the present study enlarge our understanding of how stroma components affect the delivery of NPs into the tumors. ![]()
Collapse
Affiliation(s)
- Ilya Yakavets
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique, UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506, Vandoeuvre-lès-Nancy, France. .,Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandoeuvre-lès-Nancy, France. .,Laboratory of Biophysics and Biotechnology, Belarusian State University, 4 Nezavisimosti Avenue, 220030, Minsk, Belarus.
| | - Aurelie Francois
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique, UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506, Vandoeuvre-lès-Nancy, France.,Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandoeuvre-lès-Nancy, France
| | - Laureline Lamy
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique, UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506, Vandoeuvre-lès-Nancy, France.,Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandoeuvre-lès-Nancy, France
| | - Max Piffoux
- Laboratoire Matière et systèmes complexes, CNRS UMR 7057, Université de Paris, 75205, Paris Cedex 13, France
| | - Florence Gazeau
- Laboratoire Matière et systèmes complexes, CNRS UMR 7057, Université de Paris, 75205, Paris Cedex 13, France
| | - Claire Wilhelm
- Laboratoire Matière et systèmes complexes, CNRS UMR 7057, Université de Paris, 75205, Paris Cedex 13, France
| | - Vladimir Zorin
- Laboratory of Biophysics and Biotechnology, Belarusian State University, 4 Nezavisimosti Avenue, 220030, Minsk, Belarus
| | - Amanda K A Silva
- Laboratoire Matière et systèmes complexes, CNRS UMR 7057, Université de Paris, 75205, Paris Cedex 13, France
| | - Lina Bezdetnaya
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique, UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506, Vandoeuvre-lès-Nancy, France. .,Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519, Vandoeuvre-lès-Nancy, France.
| |
Collapse
|
38
|
Wu R, Yow C, Law E, Chu E, Huang Z. Effect of Foslip® mediated photodynamic therapy on 5-fluorouracil resistant human colorectal cancer cells. Photodiagnosis Photodyn Ther 2020; 31:101945. [PMID: 32768589 DOI: 10.1016/j.pdpdt.2020.101945] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Revised: 07/27/2020] [Accepted: 07/31/2020] [Indexed: 02/07/2023]
Abstract
Colorectal cancer (CRC) is the commonest cancer in Hong Kong and is often treated with 5-fluorouracil (5-FU). However the clinical application of 5-FU was limited by drug resistance in CRC. Photodynamic therapy (PDT) is a novel treatment combating CRC via the combination of photosensitizer, molecular oxygen and light activation. In this study, 5-FU resistant HT29 (HT29FU) was established and its susceptibility to Foslip® PDT tested. Effect of 5-FU to HT29 cells was measured via qPCR. Efficacy of Foslip® PDT on HT29 and HT29FU cells were measured via photosensitizer uptake, cellular localization, cytotoxicity, cell cycle distribution and signal proteins expression. 5-FU significantly induced ABCB1 mRNA expression in HT29 cells; whereas with a 24 fold increase in HT29FU cells. Both cells responded similarly to Foslip® PDT, with the inhibitory concentration IC20, IC50 and IC70 achieved at 1 ng/mL, 2 ng/mL and 5 ng/mL with 2 J/cm2 light activation respectively. Foslip® PDT triggered apoptosis and reduced JNK protein expression at IC70 on both cells. Effect of Foslip® PDT on HT29 cells was independent to 5-FU resistance properties. Therefore, Foslip® PDT could be a potential treatment for 5-FU resistant cancer patients. Further investigations on the Foslip® PDT mediated molecular changes in HT29FU cells deserve to be explored.
Collapse
Affiliation(s)
- Rwk Wu
- School of Medical and Health Sciences, Tung Wah College, HKSAR, Hong Kong, China.
| | - Cmn Yow
- Medical Laboratory Science, Department of Health Technology & Informatics, Hong Kong Polytechnic University, HKSAR, Hong Kong, China
| | - Eric Law
- Medical Laboratory Science, Department of Health Technology & Informatics, Hong Kong Polytechnic University, HKSAR, Hong Kong, China
| | - Esm Chu
- School of Medical and Health Sciences, Tung Wah College, HKSAR, Hong Kong, China
| | - Zheng Huang
- MOE Key Laboratory of Photonics Science and Technology for Medicine, Fujian Normal University, Fuzhou, China.
| |
Collapse
|
39
|
Wu RWK, Chu ESM, Yuen JWM, Huang Z. Comparative study of FosPeg® photodynamic effect on nasopharyngeal carcinoma cells in 2D and 3D models. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY B-BIOLOGY 2020; 210:111987. [PMID: 32801063 DOI: 10.1016/j.jphotobiol.2020.111987] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Revised: 07/21/2020] [Accepted: 08/04/2020] [Indexed: 12/24/2022]
Abstract
Photodynamic Therapy (PDT) offers an alternative option for the treatment of nasopharyngeal carcinoma (NPC). The utilization of 3-dimensional (3D) culture model might provide better understanding of PDT effects on NPC cells. The aim of this in vitro study was to compare PDT effect on NPC cells using 2D and 3D models. Two 3D culture models were established using liquid overlay method with agarose base (MCL) and hanging drop method (MCS). PDT was carried out using the combination of FosPeg® and 652 nm laser in 3D and conventional 2D models. The effects of 3D culture size and morphology on the uptake and distribution of sensitizer and gene expression were examined. Photocytotoxity, mode of cell death, and protein expression were compared for 2D and 3D models. Regular and irregular NPC spheroids were obtained from MCL and MCS methods, respectively. A significantly down-regulation of LMP1 mRNA were observed in MCL spheroid. The sensitizer uptake in 3D spheroids was half of 2D culture. More sensitizers were required to obtain IC50 in 3D spheroids. Apoptosis, necrosis and autophagosomes were detected in PDT treated 2D and 3D cells. Different protein expression patterns were observed in 2D and 3D models. FosPeg® PDT is effective in killing NPC cells. MCL-derived 3D spheroid model is more suitable for the evaluation of PDT killing mechanisms.
Collapse
Affiliation(s)
- Ricky Wing Kei Wu
- School of Medical and Health Sciences, Tung Wah College, Hong Kong, China.
| | | | - John Wai Man Yuen
- School of Nursing, Hong Kong Polytechnic University, Hong Kong, China
| | - Zheng Huang
- Biomedical Photonics Center, MOE Key Laboratory of Photonics Science and Technology for Medicine, School of OptoElectronic and Information Engineering, Fujian Normal University, Fuzhou, Hong Kong, China
| |
Collapse
|
40
|
mTHPC-Loaded Extracellular Vesicles Significantly Improve mTHPC Diffusion and Photodynamic Activity in Preclinical Models. Pharmaceutics 2020; 12:pharmaceutics12070676. [PMID: 32709026 PMCID: PMC7407764 DOI: 10.3390/pharmaceutics12070676] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 07/10/2020] [Accepted: 07/15/2020] [Indexed: 12/23/2022] Open
Abstract
Extracellular vesicles (EVs), derived from the cell, display a phospholipid bilayer membrane that protects the cargo molecules from degradation and contributes to increasing their stability in the bloodstream and tumor targeting. EVs are interesting in regard to the delivery of photosensitizers (PSs) used in the photodynamic therapy (PDT), as they allow us to overcome the limitations observed with liposomes. In fact, liposomal formulation of meta-tetra(hydroxyphenyl)chlorin (mTHPC) (Foslip®), one of the most potent clinically approved PSs, is rapidly destroyed in circulation, thus decreasing in vivo PDT efficacy. mTHPC-EV uptake was evaluated in vitro in a 3D human colon HT-29 microtumor and in vivo study was performed in HT-29 xenografted mice. The obtained data were compared with Foslip®. After intravenous injection of the mTHPC formulations, biodistribution, pharmacokinetics and PDT-induced tumor regrowth were evaluated. In a 3D model of cells, mTHPC-EV uptake featured a deeper penetration after 24h incubation compared to liposomal mTHPC. In vivo results showed a considerable improvement of 33% tumor cure with PDT treatment applied 24h after injection, while 0% was observed after Foslip®/PDT. Moreover, 47 days were required to obtain ten times the initial tumor volume after mTHPC-EVs/PDT compared to 30 days for liposomal mTHPC. In conclusion, compared to Foslip®, mTHPC-EVs improved mTHPC biodistribution and PDT efficacy in vivo. We deduced that a major determinant factor for the improved in vivo PDT efficacy is the deep mTHPC intratumor penetration.
Collapse
|
41
|
Aslanoglu B, Yakavets I, Zorin V, Lassalle HP, Ingrosso F, Monari A, Catak S. Optical properties of photodynamic therapy drugs in different environments: the paradigmatic case of temoporfin. Phys Chem Chem Phys 2020; 22:16956-16964. [PMID: 32672774 DOI: 10.1039/d0cp02055a] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Computational tools have been used to study the photophysical and photochemical features of photosensitizers in photodynamic therapy (PDT) - a minimally invasive, less aggressive alternative for cancer treatment. PDT is mainly based on the activation of molecular oxygen through the action of a photoexcited sensitizer (photosensitizer). Temoporfin, widely known as mTHPC, is a second-generation photosensitizer, which produces the cytotoxic singlet oxygen when irradiated with visible light and hence destroys tumor cells. However, the bioavailability of the mostly hydrophobic photosensitizer, and hence its incorporation into cells, is fundamental to achieve the desired effect on malignant tissues via PDT. In this study, we focus on the optical properties of the temoporfin chromophore in different environments -in vacuo, in solution, encapsulated in drug delivery agents, namely cyclodextrin, and interacting with a lipid bilayer.
Collapse
Affiliation(s)
- Busenur Aslanoglu
- Bogazici University, Department of Chemistry, Bebek 34342, Istanbul, Turkey.
| | | | | | | | | | | | | |
Collapse
|
42
|
Cyclodextrin nanosponge as a temoporfin nanocarrier: Balancing between accumulation and penetration in 3D tumor spheroids. Eur J Pharm Biopharm 2020; 154:33-42. [PMID: 32634570 DOI: 10.1016/j.ejpb.2020.06.022] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/15/2020] [Accepted: 06/30/2020] [Indexed: 01/19/2023]
Abstract
As the intertissue delivery of hydrophobic temoporfin (mTHPC) remains inefficient, we propose the use of cyclodextrin-based nanosponges as a smart, advanced system for improved mTHPC delivery. Recently, we demonstrated that cyclodextrins (CDs) allow mTHPC to penetrate into tumor spheroids via a nanoshuttle mechanism. However, the CD complexes were very sensitive to the dilution, thus limiting their translation invivo. Hypercrosslinked CD monomers in a three-dimensional network (namely, CD nanosponges), however, may form both inclusion and non-inclusion complexes with drug molecules, providing controlled release and prolonged exposure to the drug. In the present work, we demonstrate that epichlorohydrin-crosslinked CD nanosponges based on β-CD (βCDp) and carboxymethyl-β-CD (CMβCDp) monomers efficiently encapsulated mTHPC. We calculated the apparent binding constants between mTHPC and CD polymers (K=(6.3-8.8) × 106M-1 and K=(1.2-1.7) × 106M-1 for βCDp and CMβCDp, respectively) using fluorescence titration curve fitting. The encapsulation of mTHPC in a CD polymer matrix had slower photosensitizer (PS) release compared to monomer CD units, providing deep penetration of mTHPC in 3D tumor spheroids in a concentration-dependent manner. However, the improvement of mTHPC penetration in 3D human pharynx squamous cell carcinoma (FaDu) spheroids using CD polymers was strongly accompanied by the inhibition of PS cellular uptake, demonstrating the delicate balance between the accumulation and the penetration of PS in FaDu spheroids. In summary, mTHPC-loaded CD nanosponges are a strong candidate for further invivo study in preclinical models, which could be considered as an advanced smart system for mTHPC delivery.
Collapse
|
43
|
Elsharkasy OM, Nordin JZ, Hagey DW, de Jong OG, Schiffelers RM, Andaloussi SEL, Vader P. Extracellular vesicles as drug delivery systems: Why and how? Adv Drug Deliv Rev 2020; 159:332-343. [PMID: 32305351 DOI: 10.1016/j.addr.2020.04.004] [Citation(s) in RCA: 631] [Impact Index Per Article: 157.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/09/2020] [Accepted: 04/14/2020] [Indexed: 12/16/2022]
Abstract
Over the past decades, a multitude of synthetic drug delivery systems has been developed and introduced to the market. However, applications of such systems are limited due to inefficiency, cytotoxicity and/or immunogenicity. At the same time, the field of natural drug carrier systems has grown rapidly. One of the most prominent examples of such natural carriers are extracellular vesicles (EVs). EVs are cell-derived membranous particles which play important roles in intercellular communication. EVs possess a number of characteristics that qualify them as promising vehicles for drug delivery. In order to take advantage of these attributes, an in-depth understanding of why EVs are such unique carrier systems and how we can exploit their qualities is pivotal. Here, we review unique EV features that are relevant for drug delivery and highlight emerging strategies to make use of those features for drug loading and targeted delivery.
Collapse
|
44
|
Sokolova EA, Senatskaya AO, Lermontova SA, Akinchits EK, Klapshina LG, Brilkina AA, Balalaeva IV. Model of Ovarian Adenocarcinoma Spheroids for Assessing Photodynamic Cytotoxicity. Sovrem Tekhnologii Med 2020; 12:34-40. [PMID: 34513035 PMCID: PMC8353712 DOI: 10.17691/stm2020.12.1.04] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Indexed: 11/14/2022] Open
Abstract
The aim of the study was to compare the relevance of ovarian adenocarcinoma spheroids with that of a monolayer culture for assessing photodynamic effect of the tetrakis(4-benzyloxyphenyl)tetracyanoporphyrazine photosensitizer. Materials and Methods The work was performed on SKOV-3 human ovary adenocarcinoma cells grown in vitro in a monolayer culture and in the form of tumor spheroids obtained using culture plates with ultra-low attachment. We determined the photoinduced toxicity of porphyrazine on a monolayer culture using the MTT assay; the effect on the spheroids was tested by assessing the dynamics of their growth. Cellular uptake of porphyrazine was analyzed by confocal microscopy. Results Porphyrazine has a pronounced photodynamic effect on SKOV-3 cells. When exposed to light at a dose of 20 J/cm2, the IC50 value 24 h after exposure was 2.3 μM for SKOV-3 monolayer culture. For the spheroids, the effect manifested after a latency period: significant growth retardation of the treated spheroids appeared no sooner than 5 and 9 days after exposure. Notably, no decrease in the initial size of the treated spheroids was observed under any of the photodynamic regimes. The penetration depth of porphyrazine into spheroids was 50-100 μm during 24 h incubation. Conclusion The limited penetration of the photosensitizer into the body of spheroids and its predominant accumulation in the surface layers can be one of the key factors behind the significant differences in the photodynamic response between the surface and deep layers of a spheroid. For cells located close to the spheroid surface, the photodynamic effect is comparable to that for a monolayer culture, while in deeper layers, the cells remain viable and support/maintain the growth of the spheroid even under intense photo-exposure. The fact that the in vitro distribution is similar to the inhomogeneous accumulation of photosensitizers in tumors in vivo allows us to consider spheroids more relevant than a monolayer culture for studying photodynamic anti-tumor effects.
Collapse
Affiliation(s)
- E A Sokolova
- Junior Researcher, Laboratory of Optical Theranostics, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia
| | - A O Senatskaya
- Graduate Student, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia
| | - S A Lermontova
- Researcher, Laboratory for Search and Applied Research, G.A. Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, 49 Tropinina St., Nizhny Novgorod, 603137, Russia
| | - E K Akinchits
- Researcher, Department of Biophysics, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia
| | - L G Klapshina
- Senior Researcher, Sector of Chromophore Compounds for Medicine, G.A. Razuvaev Institute of Organometallic Chemistry, Russian Academy of Sciences, 49 Tropinina St., Nizhny Novgorod, 603137, Russia
| | - A A Brilkina
- Associate Professor, Senior Researcher, Laboratory of Optical Theranostics, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia
| | - I V Balalaeva
- Associate Professor, Senior Researcher, Laboratory of Optical Theranostics, Institute of Biology and Biomedicine, National Research Lobachevsky State University of Nizhni Novgorod, 23 Prospekt Gagarina, Nizhny Novgorod, 603950, Russia
| |
Collapse
|
45
|
Patient-Derived Head and Neck Cancer Organoids Recapitulate EGFR Expression Levels of Respective Tissues and Are Responsive to EGFR-Targeted Photodynamic Therapy. J Clin Med 2019; 8:jcm8111880. [PMID: 31694307 PMCID: PMC6912517 DOI: 10.3390/jcm8111880] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 10/31/2019] [Accepted: 11/03/2019] [Indexed: 01/10/2023] Open
Abstract
Patients diagnosed with head and neck squamous cell carcinoma (HNSCC) are currently treated with surgery and/or radio- and chemotherapy. Despite these therapeutic interventions, 40% of patients relapse, urging the need for more effective therapies. In photodynamic therapy (PDT), a light-activated photosensitizer produces reactive oxygen species that ultimately lead to cell death. Targeted PDT, using a photosensitizer conjugated to tumor-targeting molecules, has been explored as a more selective cancer therapy. Organoids are self-organizing three-dimensional structures that can be grown from both normal and tumor patient-material and have recently shown translational potential. Here, we explore the potential of a recently described HNSCC–organoid model to evaluate Epidermal Growth Factor Receptor (EGFR)-targeted PDT, through either antibody- or nanobody-photosensitizer conjugates. We find that EGFR expression levels differ between organoids derived from different donors, and recapitulate EGFR expression levels of patient material. EGFR expression levels were found to correlate with the response to EGFR-targeted PDT. Importantly, organoids grown from surrounding normal tissues showed lower EGFR expression levels than their tumor counterparts, and were not affected by the treatment. In general, nanobody-targeted PDT was more effective than antibody-targeted PDT. Taken together, patient-derived HNSCC organoids are a useful 3D model for testing in vitro targeted PDT.
Collapse
|
46
|
Stroma-Rich Co-Culture Multicellular Tumor Spheroids as a Tool for Photoactive Drugs Screening. J Clin Med 2019; 8:jcm8101686. [PMID: 31618880 PMCID: PMC6832590 DOI: 10.3390/jcm8101686] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/28/2022] Open
Abstract
Conventional 3D multicellular tumor spheroids of head and neck squamous cell carcinoma (HNSCC) consisting exclusively of cancer cells have some limitations. They are compact cell aggregates that do not interact with their extracellular milieu, thus suffering from both insufficient extracellular matrix (ECM) deposition and absence of different types of stromal cells. In order to better mimic in vivo HNSCC tumor microenvironment, we have constructed a 3D stroma-rich in vitro model of HNSCC, using cancer-associated MeWo skin fibroblasts and FaDu pharynx squamous cell carcinoma. The expression of stromal components in heterospheroids was confirmed by immunochemical staining. The generated co-culture FaDu/MeWo spheroids were applied to study penetration, distribution and antitumor efficacy of photoactive drugs such as Temoporfin and Chlorin e6 used in the photodynamic therapy flow cytometry and fluorescence microscopy techniques. We also investigated the distribution of photodiagnostic agent Indocyanine Green. We demonstrated that the presence of stroma influences the behavior of photoactive drugs in different ways: (i) No effect on Indocyanine Green distribution; (ii) lower accumulation of Chlorin e6; (iii) better penetration and PDT efficiency of Temoporfin. Overall, the developed stroma-rich spheroids enlarge the arsenal of in vitro pre-clinical models for high-throughput screening of anti-cancer drugs.
Collapse
|
47
|
Mol EA, Lei Z, Roefs MT, Bakker MH, Goumans M, Doevendans PA, Dankers PYW, Vader P, Sluijter JPG. Injectable Supramolecular Ureidopyrimidinone Hydrogels Provide Sustained Release of Extracellular Vesicle Therapeutics. Adv Healthc Mater 2019; 8:e1900847. [PMID: 31559704 DOI: 10.1002/adhm.201900847] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2019] [Revised: 08/23/2019] [Indexed: 12/15/2022]
Abstract
Extracellular vesicles (EVs) are small vesicles secreted by cells and have gained increasing interest as both drug delivery vehicles or as cell-free therapeutics for regenerative medicine. To achieve optimal therapeutic effects, strategies are being developed to prolong EV exposure to target organs. One promising approach to achieve this is through EV-loaded injectable hydrogels. In this study, the use of a hydrogel based on ureido-pyrimidinone (UPy) units coupled to poly(ethylene glycol) chains (UPy-hydrogel) is examined as potential delivery platform for EVs. The UPy-hydrogel undergoes a solution-to-gel transition upon switching from a high to neutral pH, allowing immediate gelation upon administration into physiological systems. Here, sustained EV release from the UPy-hydrogel measured over a period of 4 d is shown. Importantly, EVs retain their functional capacity after release. Upon local administration of fluorescently labeled EVs incorporated in a UPy-hydrogel in vivo, EVs are still detected in the UPy-hydrogel after 3 d, whereas in the absence of a hydrogel, EVs are internalized by fat and skin tissue near the injection site. Together, these data demonstrate that UPy-hydrogels provide sustained EV release over time and enhance local EV retention in vivo, which could contribute to improved therapeutic efficacy upon local delivery and translation toward new applications.
Collapse
Affiliation(s)
- Emma A. Mol
- Department of CardiologyLaboratory of Experimental CardiologyUniversity Medical Center Utrecht Utrecht 3584 The Netherlands
- Laboratory of Cardiovascular Cell BiologyDepartment of Cell and Chemical BiologyLeiden University Medical Center Leiden 2333ZA The Netherlands
| | - Zhiyong Lei
- Department of CardiologyLaboratory of Experimental CardiologyUniversity Medical Center Utrecht Utrecht 3584 The Netherlands
| | - Marieke T. Roefs
- Department of CardiologyLaboratory of Experimental CardiologyUniversity Medical Center Utrecht Utrecht 3584 The Netherlands
| | - Maarten H. Bakker
- Institute for Complex Molecular SystemsDepartment of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of Technology 5600 MB Eindhoven The Netherlands
| | - Marie‐José Goumans
- Laboratory of Cardiovascular Cell BiologyDepartment of Cell and Chemical BiologyLeiden University Medical Center Leiden 2333ZA The Netherlands
| | - Pieter A. Doevendans
- Department of CardiologyLaboratory of Experimental CardiologyUniversity Medical Center Utrecht Utrecht 3584 The Netherlands
- UMC Utrecht Regenerative Medicine CenterUniversity Medical Center Utrecht 3584CT The Netherlands
- CMH NL‐HI Utrecht 3584CX The Netherlands
| | - Patricia Y. W. Dankers
- Institute for Complex Molecular SystemsDepartment of Biomedical EngineeringLaboratory of Chemical BiologyEindhoven University of Technology 5600 MB Eindhoven The Netherlands
| | - Pieter Vader
- Department of CardiologyLaboratory of Experimental CardiologyUniversity Medical Center Utrecht Utrecht 3584 The Netherlands
- Laboratory of Clinical Chemistry and HaematologyUniversity Medical Center Utrecht Utrecht 3584 The Netherlands
| | - Joost P. G. Sluijter
- Department of CardiologyLaboratory of Experimental CardiologyUniversity Medical Center Utrecht Utrecht 3584 The Netherlands
- UMC Utrecht Regenerative Medicine CenterUniversity Medical Center Utrecht 3584CT The Netherlands
- University Utrecht Utrecht 3508TC The Netherlands
| |
Collapse
|
48
|
Matryoshka-Type Liposomes Offer the Improved Delivery of Temoporfin to Tumor Spheroids. Cancers (Basel) 2019; 11:cancers11091366. [PMID: 31540319 PMCID: PMC6770699 DOI: 10.3390/cancers11091366] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 09/10/2019] [Accepted: 09/12/2019] [Indexed: 12/12/2022] Open
Abstract
The balance between the amount of drug delivered to tumor tissue and the homogeneity of its distribution is a challenge in the efficient delivery of photosensitizers (PSs) in photodynamic therapy (PDT) of cancer. To date, many efforts have been made using various nanomaterials to efficiently deliver temoporfin (mTHPC), one of the most potent photosensitizers. The present study aimed to develop double-loaded matryoshka-type hybrid nanoparticles encapsulating mTHPC/cyclodextrin inclusion complexes in mTHPC-loaded liposomes. This system was expected to improve the transport of mTHPC to target tissues and to strengthen its accumulation in the tumor tissue. Double-loaded hybrid nanoparticles (DL-DCL) were prepared, characterized, and tested in 2D and 3D in vitro models and in xenografted mice in vivo. Our studies indicated that DL-DCL provided deep penetration of mTHPC into the multicellular tumor spheroids via cyclodextrin nanoshuttles once the liposomes had been destabilized by serum proteins. Unexpectedly, we observed similar PDT efficiency in xenografted HT29 tumors for liposomal mTHPC formulation (Foslip®) and DL-DCL.
Collapse
|
49
|
Yakavets I, Millard M, Zorin V, Lassalle HP, Bezdetnaya L. Current state of the nanoscale delivery systems for temoporfin-based photodynamic therapy: Advanced delivery strategies. J Control Release 2019; 304:268-287. [PMID: 31136810 DOI: 10.1016/j.jconrel.2019.05.035] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 05/21/2019] [Accepted: 05/23/2019] [Indexed: 12/22/2022]
Abstract
Enthusiasm for photodynamic therapy (PDT) as a promising technique to eradicate various cancers has increased exponentially in recent decades. The majority of clinically approved photosensitizers are hydrophobic in nature, thus, the effective delivery of photosensitizers at the targeted site is the main hurdle associated with PDT. Temoporfin (mTHPC, medicinal product name: Foscan®), is one of the most potent clinically approved photosensitizers, is not an exception. Successful temoporfin-PDT requires nanoscale delivery systems for selective delivery of photosensitizer. Over the last 25 years, the number of papers on nanoplatforms developed for mTHPC delivery such as conjugates, host-guest inclusion complexes, lipid-and polymer-based nanoparticles and carbon nanotubes is burgeoning. However, none of them appeared to be "ultimate". The present review offers the description of different challenges and achievements in nanoparticle-based mTHPC delivery focusing on the synergetic combination of various nano-platforms to improve temoporfin delivery at all stages of biodistribution. Furthermore, the association of different nanoparticles in one nanoplatform might be considered as an advanced strategy allowing the combination of several treatment modalities.
Collapse
Affiliation(s)
- Ilya Yakavets
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France; Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France; Laboratory of Biophysics and Biotechnology, Belarusian State University, 4 Nezavisimosti Avenue, 220030 Minsk, Belarus.
| | - Marie Millard
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France; Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France.
| | - Vladimir Zorin
- Laboratory of Biophysics and Biotechnology, Belarusian State University, 4 Nezavisimosti Avenue, 220030 Minsk, Belarus; International Sakharov Environmental Institute, Belarusian State University, Dauhabrodskaja 23, 220030 Minsk, Belarus.
| | - Henri-Pierre Lassalle
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France; Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France.
| | - Lina Bezdetnaya
- Centre de Recherche en Automatique de Nancy, Centre National de la Recherche Scientifique UMR 7039, Université de Lorraine, Campus Sciences, Boulevard des Aiguillette, 54506 Vandoeuvre-lès-Nancy, France; Research Department, Institut de Cancérologie de Lorraine, 6 avenue de Bourgogne, 54519 Vandoeuvre-lès-Nancy, France.
| |
Collapse
|