1
|
Liu Y, Ling Y, Tai W. Mechanical Extrusion of the Plasma Membrane to Generate Ectosome-Mimetic Nanovesicles for Lung Targeting. Mol Pharm 2025; 22:304-315. [PMID: 39591514 DOI: 10.1021/acs.molpharmaceut.4c00927] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2024]
Abstract
Extracellular vehicles (EVs) are naturally occurring nanocarriers that participate in the transportation of biologics between cells. Despite their potential in drug delivery, their optimal use in therapy remains a challenge, which comes from the difficulty in preparation scale-up and cargo loading efficiency. As a membrane-enclosed nanoscale system, EVs are reluctant to be transfected with cargos and purified by conventional methods. In the present study, we proposed an EV-mimetic nanovesicle system to overcome the challenges. Using the easy-culture mammalian cells as raw materials, we isolated the plasma membrane sheets and vesiculated them into membrane-enclosed nanovesicles as an EV mimic by the mechanical extrusion through porous membranes. In order to controllably load the cargos in the lumen of vesicles, the endogenous actin filament was chosen as an anchor to capture the cargos (fused with an anti-actin nanobody) in the inner leaflet of plasma membrane sheets and vesiculated inside after extrusion. By loading the bioluminescent tracer nano-luciferase (Nluc) and tracking biodistribution in mice, we unclosed the lung-tropic nature of these nanovesicles. Furthermore, we demonstrated that nanovesicles can be genetically engineered with chimeric antigen receptors to achieve the active targeting of lung cancer cells. In conclusion, our study indicated that plasma membrane extrusion might be an applicable approach to generate EV mimics for drug delivery, especially to the lung tissue.
Collapse
Affiliation(s)
- Yahui Liu
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Yuelei Ling
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| | - Wanyi Tai
- Department of Pharmaceutical Engineering, School of Pharmaceutical Sciences, Wuhan University, Wuhan, Hubei 430071, China
| |
Collapse
|
2
|
Wang J, Zhang X, Xing J, Gao L, Lu H. Nanomedicines in diagnosis and treatment of prostate cancers: an updated review. Front Bioeng Biotechnol 2024; 12:1444201. [PMID: 39318666 PMCID: PMC11420853 DOI: 10.3389/fbioe.2024.1444201] [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: 06/07/2024] [Accepted: 08/05/2024] [Indexed: 09/26/2024] Open
Abstract
Prostate cancer (PC) is the third most common male cancer in the world, which occurs due to various mutations leading to the loss of chromatin structure. There are multiple treatments for this type of cancer, of which chemotherapy is one of the most important. Sometimes, a combination of different treatments, such as chemotherapy, radiotherapy, and surgery, are used to prevent tumor recurrence. Among other treatments, androgen deprivation therapy (ADT) can be mentioned, which has had promising results. One of the drawbacks of chemotherapy and ADT treatments is that they are not targeted to the tumor tissue. For this reason, their use can cause extensive side effects. Treatments based on nanomaterials, known as nanomedicine, have attracted much attention today. Nanoparticles (NPs) are one of the main branches of nanomedicine, and they can be made of different materials such as polymer, metal, and carbon, each of which has distinct characteristics. In addition to NPs, nanovesicles (NVs) also have therapeutic applications in PC. In treating PC, synthetic NVs (liposomes, micelles, and nanobubbles) or produced from cells (exosomes) can be used. In addition to the role that NPs and NVs have in treating PC, due to being targeted, they can be used to diagnose PC and check the treatment process. Knowing the characteristics of nanomedicine-based treatments can help design new treatments and improve researchers' understanding of tumor biology and its rapid diagnosis. In this study, we will discuss conventional and nanomedicine-based treatments. The results of these studies show that the use of NPs and NVs in combination with conventional treatments has higher efficacy in tumor treatment than the individual use of each of them.
Collapse
Affiliation(s)
- Jiajia Wang
- Department of Oncology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Xuan Zhang
- Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Jiazhen Xing
- Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Lijian Gao
- Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| | - Hua Lu
- Department of Urology, Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, China
| |
Collapse
|
3
|
Smack C, Johnson B, Nyalwidhe JO, Semmes OJ, Yang L. Small extracellular vesicles: Roles and clinical application in prostate cancer. Adv Cancer Res 2024; 161:119-190. [PMID: 39032949 DOI: 10.1016/bs.acr.2024.05.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/23/2024]
Abstract
Prostate cancer is a significant health problem in the United States. It is remarkably heterogenous, ranging from slow growing disease amenable to active surveillance to highly aggressive forms requiring active treatments. Therefore, being able to precisely determine the nature of disease and appropriately match patients to available and/or novel therapeutics is crucial to improve patients' overall outcome and quality of life. Recently small extracellular vesicles (sEVs), a subset of nanoscale membranous vesicles secreted by various cells, have emerged as important analytes for liquid biopsy and promising vehicles for drug delivery. sEVs contain various biomolecules such as genetic material, proteins, and lipids that recapitulate the characteristics and state of their donor cells. The application of existing and newly developed technologies has resulted in an increased depth of knowledge about biophysical structures, biogenesis, and functions of sEVs. In prostate cancer patients, tumor-derived sEVs can be isolated from biofluids, commonly urine and blood. They mediate intercellular signaling within the tumor microenvironment and distal organ-specific sites, supporting cancer initiation, progression, and metastasis. A mounting body of evidence suggests that sEV components can be potent biomarkers for prostate cancer diagnosis, prognosis, and prediction of disease progression and treatment response. Due to enhanced circulation stability and bio-barrier permeability, sEVs can be also used as effective drug delivery carriers to improve the efficacy and specificity of anti-tumor therapies. This review discusses recent studies on sEVs in prostate cancer and is focused on their role as biomarkers and drug delivery vehicles in the clinical management of prostate cancer.
Collapse
Affiliation(s)
- Caleb Smack
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Benjamin Johnson
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Julius O Nyalwidhe
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - O John Semmes
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States
| | - Lifang Yang
- Leroy T. Canoles Jr. Cancer Research Center, Eastern Virginia Medical School, Norfolk, VA, United States; Department of Microbiology and Molecular Cell Biology, Eastern Virginia Medical School, Norfolk, VA, United States.
| |
Collapse
|
4
|
Si C, Gao J, Ma X. Engineered exosomes in emerging cell-free therapy. Front Oncol 2024; 14:1382398. [PMID: 38595822 PMCID: PMC11003191 DOI: 10.3389/fonc.2024.1382398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 03/14/2024] [Indexed: 04/11/2024] Open
Abstract
The discovery and use of exosomes ushered in a new era of cell-free therapy. Exosomes are a subgroup of extracellular vesicles that show great potential in disease treatment. Engineered exosomes. with their improved functions have attracted intense interests of their application in translational medicine research. However, the technology of engineering exosomes still faces many challenges which have been the great limitation for their clinical application. This review summarizes the current status of research on engineered exosomes and the difficulties encountered in recent years, with a view to providing new approaches and ideas for future exosome modification and new drug development.
Collapse
Affiliation(s)
| | - Jianen Gao
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xu Ma
- National Research Institute for Family Planning, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
5
|
Hu R, Lan J, Zhang D, Shen W. Nanotherapeutics for prostate cancer treatment: A comprehensive review. Biomaterials 2024; 305:122469. [PMID: 38244344 DOI: 10.1016/j.biomaterials.2024.122469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 01/22/2024]
Abstract
Prostate cancer (PCa) is the most prevalent solid organ malignancy and seriously affects male health. The adverse effects of prostate cancer therapeutics can cause secondary damage to patients. Nanotherapeutics, which have special targeting abilities and controlled therapeutic release profiles, may serve as alternative agents for PCa treatment. At present, many nanotherapeutics have been developed to treat PCa and have shown better treatment effects in animals than traditional therapeutics. Although PCa nanotherapeutics are highly attractive, few successful cases have been reported in clinical practice. To help researchers design valuable nanotherapeutics for PCa treatment and avoid useless efforts, herein, we first reviewed the strategies and challenges involved in prostate cancer treatment. Subsequently, we presented a comprehensive review of nanotherapeutics for PCa treatment, including their targeting methods, controlled release strategies, therapeutic approaches and mechanisms. Finally, we proposed the future prospects of nanotherapeutics for PCa treatment.
Collapse
Affiliation(s)
- Ruimin Hu
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University (Army Medical University), Chongqing, 400038, China
| | - Jin Lan
- Department of Ultrasound, Second Affiliated Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Dinglin Zhang
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Department of Chemistry, College of Basic Medicine, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| | - Wenhao Shen
- Department of Urology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
| |
Collapse
|
6
|
Chen H, Pang B, Zhou C, Han M, Gong J, Li Y, Jiang J. Prostate cancer-derived small extracellular vesicle proteins: the hope in diagnosis, prognosis, and therapeutics. J Nanobiotechnology 2023; 21:480. [PMID: 38093355 PMCID: PMC10720096 DOI: 10.1186/s12951-023-02219-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Accepted: 11/18/2023] [Indexed: 12/17/2023] Open
Abstract
Current diagnostic tools for prostate cancer (PCa) diagnosis and risk stratification are insufficient. The hidden onset and poor efficacy of traditional therapies against metastatic PCa make this disease a heavy burden in global men's health. Prostate cancer-derived extracellular vesicles (PCDEVs) have garnered attention in recent years due to their important role in communications in tumor microenvironment. Recent advancements have demonstrated PCDEVs proteins play an important role in PCa invasion, progression, metastasis, therapeutic resistance, and immune escape. In this review, we briefly discuss the applications of sEV proteins in PCa diagnosis and prognosis in liquid biopsy, focus on the roles of the PCa-derived small EVs (sEVs) proteins in tumor microenvironment associated with cancer progression, and explore the therapeutic potential of sEV proteins applied for future metastatic PCa therapy.
Collapse
Affiliation(s)
- Haotian Chen
- Health Science Center, Ningbo University, Ningbo, 315211, Zhejiang, People's Republic of China
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Bairen Pang
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Cheng Zhou
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Meng Han
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Jie Gong
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China
| | - Yong Li
- Cancer Care Centre, St George Hospital, Kogarah, NSW, 2217, Australia.
- School of Clinical Medicine, St. George and Sutherland Clinical Campuses, UNSW Sydney, Kensington, NSW, 2052, Australia.
| | - Junhui Jiang
- Ningbo Clinical Research Center for Urological Disease, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China.
- Translational Research Laboratory for Urology, Department of Urology, The First Affiliated Hospital of Ningbo University, Ningbo, 315010, Zhejiang, People's Republic of China.
- Department of Urology, Ningbo First Hospital, The First Affiliated Hospital of Ningbo University, Haishu District, Ningbo, 315600, Zhejiang, People's Republic of China.
| |
Collapse
|
7
|
Bhatta R, Han J, Liu Y, Bo Y, Lee D, Zhou J, Wang Y, Nelson ER, Chen Q, Zhang XS, Hassaneen W, Wang H. Metabolic tagging of extracellular vesicles and development of enhanced extracellular vesicle based cancer vaccines. Nat Commun 2023; 14:8047. [PMID: 38052869 PMCID: PMC10697976 DOI: 10.1038/s41467-023-43914-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 11/23/2023] [Indexed: 12/07/2023] Open
Abstract
As key mediators of cellular communication, extracellular vesicles (EVs) have been actively explored for diagnostic and therapeutic applications. However, effective methods to functionalize EVs and modulate the interaction between EVs and recipient cells are still lacking. Here we report a facile and universal metabolic tagging technology that can install unique chemical tags (e.g., azido groups) onto EVs. The surface chemical tags enable conjugation of molecules via efficient click chemistry, for the tracking and targeted modulation of EVs. In the context of tumor EV vaccines, we show that the conjugation of toll-like receptor 9 agonists onto EVs enables timely activation of dendritic cells and generation of superior antitumor CD8+ T cell response. These lead to 80% tumor-free survival against E.G7 lymphoma and 33% tumor-free survival against B16F10 melanoma. Our study yields a universal technology to generate chemically tagged EVs from parent cells, modulate EV-cell interactions, and develop potent EV vaccines.
Collapse
Affiliation(s)
- Rimsha Bhatta
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Joonsu Han
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yusheng Liu
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yang Bo
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - David Lee
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jiadiao Zhou
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Yueji Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
| | - Erik Russell Nelson
- Cancer Center at Illinois (CCIL), Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Molecular and Integrative Physiology, University of Illinois at Urbana-, Champaign, IL, USA
| | - Qian Chen
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Xiaojia Shelly Zhang
- Department of Mechanical Science and Engineering, University of Illinois Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Civil and Environmental Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- National Center for Supercomputing Applications, Urbana, IL, 61801, USA
| | - Wael Hassaneen
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Carle Foundation Hospital, Urbana, IL, 61801, USA
| | - Hua Wang
- Department of Materials Science and Engineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Cancer Center at Illinois (CCIL), Urbana, IL, 61801, USA.
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Materials Research Laboratory, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Carle College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA.
| |
Collapse
|
8
|
Patel G, Agnihotri TG, Gitte M, Shinde T, Gomte SS, Goswami R, Jain A. Exosomes: a potential diagnostic and treatment modality in the quest for counteracting cancer. Cell Oncol (Dordr) 2023; 46:1159-1179. [PMID: 37040056 PMCID: PMC10088756 DOI: 10.1007/s13402-023-00810-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/31/2023] [Indexed: 04/12/2023] Open
Abstract
BACKGROUND Exosomes are nanosized bio vesicles formed when multivesicular bodies and the plasma membrane merge and discharge into bodily fluids. They are well recognized for facilitating intercellular communication by transporting numerous biomolecules, including DNA, RNAs, proteins, and lipids, and have been implicated in varied diseases including cancer. Exosomes may be altered to transport a variety of therapeutic payloads, including as short interfering RNAs, antisense oligonucleotides, chemotherapeutic drugs, and immunological modulators, and can be directed to a specific target. Exosomes also possess the potential to act as a diagnostic biomarker in cancer, in addition to their therapeutic potential. CONCLUSION In this review, the physiological roles played by exosomes were summarized along with their biogenesis process. Different isolation techniques of exosomes including centrifugation-based, size-based, and polymer precipitation-based techniques have also been described in detail with a special focus on cancer therapeutic applications. The review also shed light on techniques of incubation of drugs with exosomes and their characterization methods covering the most advanced techniques. Myriad applications of exosomes in cancer as diagnostic biomarkers, drug delivery carriers, and chemoresistance-related issues have been discussed at length. Furthermore, a brief overview of exosome-based anti-cancer vaccines and a few prominent challenges concerning exosomal delivery have been concluded at the end.
Collapse
Affiliation(s)
- Gayatri Patel
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Tejas Girish Agnihotri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Manoj Gitte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Tanuja Shinde
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Shyam Sudhakar Gomte
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India
| | - Roshan Goswami
- Biological E Limited, Plot No-1, Phase 2, Kolthur Village, Medchal District, Shameerpet Mdl, Hyderabad, Telangana, 500078, India
| | - Aakanchha Jain
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Ahmedabad, Palaj, Gandhinagar, Gujarat, 382355, India.
| |
Collapse
|
9
|
Ribeiro J, Lopes I, Gomes AC. A New Perspective for the Treatment of Alzheimer's Disease: Exosome-like Liposomes to Deliver Natural Compounds and RNA Therapies. Molecules 2023; 28:6015. [PMID: 37630268 PMCID: PMC10458935 DOI: 10.3390/molecules28166015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
With the increment of the aging population in recent years, neurodegenerative diseases exert a major global disease burden, essentially as a result of the lack of treatments that stop the disease progression. Alzheimer's Disease (AD) is an example of a neurodegenerative disease that affects millions of people globally, with no effective treatment. Natural compounds have emerged as a viable therapy to fill a huge gap in AD management, and in recent years, mostly fueled by the COVID-19 pandemic, RNA-based therapeutics have become a hot topic in the treatment of several diseases. Treatments of AD face significant limitations due to the complex and interconnected pathways that lead to their hallmarks and also due to the necessity to cross the blood-brain barrier. Nanotechnology has contributed to surpassing this bottleneck in the treatment of AD by promoting safe and enhanced drug delivery to the brain. In particular, exosome-like nanoparticles, a hybrid delivery system combining exosomes and liposomes' advantageous features, are demonstrating great potential in the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Joana Ribeiro
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (J.R.); (I.L.)
- Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| | - Ivo Lopes
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (J.R.); (I.L.)
| | - Andreia Castro Gomes
- Centre of Molecular and Environmental Biology (CBMA)/Aquatic Research Network (ARNET) Associate Laboratory, Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal; (J.R.); (I.L.)
- Institute of Science and Innovation for Sustainability (IB-S), Universidade do Minho, Campus de Gualtar, 4710-057 Braga, Portugal
| |
Collapse
|
10
|
Kumar SK, Sasidhar MV. Recent Trends in the Use of Small Extracellular Vesicles as Optimal Drug Delivery Vehicles in Oncology. Mol Pharm 2023; 20:3829-3842. [PMID: 37410017 DOI: 10.1021/acs.molpharmaceut.3c00363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/07/2023]
Abstract
Small extracellular vesicles (sEVs) are produced by most cells and play an important role in cell-to-cell communication and maintaining cellular homeostasis. Their ability to transfer biological cargo to target cells makes them a promising tool for cancer drug delivery. Advances in sEV engineering, EV mimetics, and ligand-directed targeting have improved the efficacy of anticancer drug delivery and functionality. EV-based RNA interference and hybrid miRNA transfer have also been extensively used in various preclinical cancer models. Despite these developments, gaps still exist in our understanding of using sEVs to treat solid tumor malignancies effectively. This article provides an overview of the last five years of sEV research and its current status for the efficient and targeted elimination of cancer cells, which could advance cancer research and bring sEV formulations into clinical use.
Collapse
Affiliation(s)
- Sarwareddy Kartik Kumar
- Apollo Hospitals Educational and Research Foundation (AHERF), Apollo Hospitals, Jubilee Hills, Hyderabad 500033, India
| | - Manda Venkata Sasidhar
- Apollo Hospitals Educational and Research Foundation (AHERF), Apollo Hospitals, Jubilee Hills, Hyderabad 500033, India
- Urvogelbio Private Limited, AHERF, Jubilee Hills, Hyderabad 500033, India
| |
Collapse
|
11
|
Zhang Y, Dou Y, Liu Y, Di M, Bian H, Sun X, Yang Q. Advances in Therapeutic Applications of Extracellular Vesicles. Int J Nanomedicine 2023; 18:3285-3307. [PMID: 37346366 PMCID: PMC10281276 DOI: 10.2147/ijn.s409588] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/08/2023] [Indexed: 06/23/2023] Open
Abstract
Extracellular vesicles (EVs) are nanoscale bilayer phospholipid membrane vesicles released by cells. Contained large molecules such as nucleic acid, protein, and lipid, EVs are an integral part of cell communication. The contents of EVs vary based on the cell source and play an important role in both pathological and physiological conditions. EVs can be used as drugs or targets in disease treatment, and changes in the contents of EVs can indicate the progression of diseases. In recent years, with the continuous exploration of the structure, characteristics, and functions of EVs, the potential of engineered EVs for drug delivery and therapy being constantly explored. This review provides a brief overview of the structure, characteristics and functions of EVs, summarizes the advanced application of EVs and outlook on the prospect of it. It is our hope that this review will increase understanding of the current development of medical applications of EVs and help us overcome future challenges.
Collapse
Affiliation(s)
- Yiming Zhang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yiming Dou
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Yang Liu
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Mingyuan Di
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Hanming Bian
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Xun Sun
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| | - Qiang Yang
- Department of Spine Surgery, Tianjin Hospital, Tianjin University, Tianjin, People’s Republic of China
- Clinical School of Orthopedics, Tianjin Medical University, Tianjin, People’s Republic of China
| |
Collapse
|
12
|
Johnson V, Vasu S, Kumar US, Kumar M. Surface-Engineered Extracellular Vesicles in Cancer Immunotherapy. Cancers (Basel) 2023; 15:2838. [PMID: 37345176 PMCID: PMC10216164 DOI: 10.3390/cancers15102838] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Revised: 05/14/2023] [Accepted: 05/15/2023] [Indexed: 06/23/2023] Open
Abstract
Extracellular vesicles (EVs) are lipid bilayer-enclosed bodies secreted by all cell types. EVs carry bioactive materials, such as proteins, lipids, metabolites, and nucleic acids, to communicate and elicit functional alterations and phenotypic changes in the counterpart stromal cells. In cancer, cells secrete EVs to shape a tumor-promoting niche. Tumor-secreted EVs mediate communications with immune cells that determine the fate of anti-tumor therapeutic effectiveness. Surface engineering of EVs has emerged as a promising tool for the modulation of tumor microenvironments for cancer immunotherapy. Modification of EVs' surface with various molecules, such as antibodies, peptides, and proteins, can enhance their targeting specificity, immunogenicity, biodistribution, and pharmacokinetics. The diverse approaches sought for engineering EV surfaces can be categorized as physical, chemical, and genetic engineering strategies. The choice of method depends on the specific application and desired outcome. Each has its advantages and disadvantages. This review lends a bird's-eye view of the recent progress in these approaches with respect to their rational implications in the immunomodulation of tumor microenvironments (TME) from pro-tumorigenic to anti-tumorigenic ones. The strategies for modulating TME using targeted EVs, their advantages, current limitations, and future directions are discussed.
Collapse
Affiliation(s)
- Vinith Johnson
- Department of Chemical Engineering, Indian Institute of Technology, Tirupati 517619, India
| | - Sunil Vasu
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| | - Uday S. Kumar
- Department of Chemical Engineering, Indian Institute of Technology, Tirupati 517619, India
| | - Manoj Kumar
- Department of Radiology, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
13
|
Ma GL, Lin WF. Immune checkpoint inhibition mediated with liposomal nanomedicine for cancer therapy. Mil Med Res 2023; 10:20. [PMID: 37106400 PMCID: PMC10142459 DOI: 10.1186/s40779-023-00455-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 04/08/2023] [Indexed: 04/29/2023] Open
Abstract
Immune checkpoint blockade (ICB) therapy for cancer has achieved great success both in clinical results and on the market. At the same time, success drives more attention from scientists to improve it. However, only a small portion of patients are responsive to this therapy, and it comes with a unique spectrum of side effects termed immune-related adverse events (irAEs). The use of nanotechnology could improve ICBs' delivery to the tumor, assist them in penetrating deeper into tumor tissues and alleviate their irAEs. Liposomal nanomedicine has been investigated and used for decades, and is well-recognized as the most successful nano-drug delivery system. The successful combination of ICB with liposomal nanomedicine could help improve the efficacy of ICB therapy. In this review, we highlighted recent studies using liposomal nanomedicine (including new emerging exosomes and their inspired nano-vesicles) in associating ICB therapy.
Collapse
Affiliation(s)
- Guang-Long Ma
- Faculty of Medicine, Centre for Cancer Immunology, University of Southampton, Southampton, SO16 6YD UK
| | - Wei-Feng Lin
- Department of Molecular Chemistry and Materials Science, Weizmann Institute of Science, 76100 Rehovot, Israel
- Key Laboratory of Bio-Inspired Smart Interfacial Science and Technology of Ministry of Education, School of Chemistry, Beihang University, Beijing, 100191 China
| |
Collapse
|
14
|
Mohammadi AH, Ghazvinian Z, Bagheri F, Harada M, Baghaei K. Modification of Extracellular Vesicle Surfaces: An Approach for Targeted Drug Delivery. BioDrugs 2023; 37:353-374. [PMID: 37093521 DOI: 10.1007/s40259-023-00595-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/25/2023]
Abstract
Extracellular vesicles (EVs) are a promising drug delivery vehicle candidate because of their natural origin and intrinsic function of transporting various molecules between different cells. Several advantages of the EV delivery platform include enhanced permeability and retention effect, efficient interaction with recipient cells, the ability to traverse biological barriers, high biocompatibility, high biodegradability, and low immunogenicity. Furthermore, EV membranes share approximately similar structures and contents to the cell membrane, which allows surface modification of EVs, an approach to enable specific targeting. Enhanced drug accumulation in intended sites and reduced adverse effects of chemotherapeutic drugs are the most prominent effects of targeted drug delivery. In order to improve the targeting ability of EVs, chemical modification and genetic engineering are the most adopted methods to date. Diverse chemical methods are employed to decorate EV surfaces with various ligands such as aptamers, carbohydrates, peptides, vitamins, and antibodies. In this review, we introduce the biogenesis, content, and cellular pathway of natural EVs and further discuss the genetic modification of EVs, and its challenges. Furthermore, we provide a comprehensive deliberation on the various chemical modification methods for improved drug delivery, which are directly related to increasing the therapeutic index.
Collapse
Affiliation(s)
- Amir Hossein Mohammadi
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran
| | - Zeinab Ghazvinian
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Bagheri
- Department of Biotechnology, Faculty of Chemical Engineering, Tarbiat Modares University, Tehran, Iran.
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
- Department of Biomedical Engineering, Michigan State University, East Lansing, MI, USA.
| | - Kaveh Baghaei
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
15
|
Sun Y, Sun F, Xu W, Qian H. Engineered Extracellular Vesicles as a Targeted Delivery Platform for Precision Therapy. Tissue Eng Regen Med 2023; 20:157-175. [PMID: 36637750 PMCID: PMC10070595 DOI: 10.1007/s13770-022-00503-y] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2022] [Revised: 10/13/2022] [Accepted: 10/23/2022] [Indexed: 01/14/2023] Open
Abstract
Extracellular vesicles (EVs)-based cell-free strategy has shown therapeutic potential in tissue regeneration. Due to their important roles in intercellular communications and their natural ability to shield cargos from degradation, EVs are also emerged as novel delivery vehicles for various bioactive molecules and drugs. Accumulating studies have revealed that EVs can be modified to enhance their efficacy and specificity for the treatment of many diseases. Engineered EVs are poised as the next generation of targeted delivery platform in the field of precision therapy. In this review, the unique properties of EVs are overviewed in terms of their biogenesis, contents, surface features and biological functions, and the recent advances in the strategies of engineered EVs construction are summarized. Additionally, we also discuss the potential applications of engineered EVs in targeted therapy of cancer and damaged tissues, and evaluate the opportunities and challenges for translating them into clinical practice.
Collapse
Affiliation(s)
- Yuntong Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Fengtian Sun
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Wenrong Xu
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China
| | - Hui Qian
- Jiangsu Key Laboratory of Medical Science and Laboratory Medicine, School of Medicine, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, China.
| |
Collapse
|
16
|
Zhang M, Hu S, Liu L, Dang P, Liu Y, Sun Z, Qiao B, Wang C. Engineered exosomes from different sources for cancer-targeted therapy. Signal Transduct Target Ther 2023; 8:124. [PMID: 36922504 PMCID: PMC10017761 DOI: 10.1038/s41392-023-01382-y] [Citation(s) in RCA: 125] [Impact Index Per Article: 62.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 01/31/2023] [Accepted: 02/22/2023] [Indexed: 03/17/2023] Open
Abstract
Exosome is a subgroup of extracellular vesicles, which has been serving as an efficient therapeutic tool for various diseases. Engineered exosomes are the sort of exosomes modified with surface decoration and internal therapeutic molecules. After appropriate modification, engineered exosomes are able to deliver antitumor drugs to tumor sites efficiently and precisely with fewer treatment-related adverse effects. However, there still exist many challenges for the clinical translation of engineered exosomes. For instance, what sources and modification strategies could endow exosomes with the most efficient antitumor activity is still poorly understood. Additionally, how to choose appropriately engineered exosomes in different antitumor therapies is another unresolved problem. In this review, we summarized the characteristics of engineered exosomes, especially the spatial and temporal properties. Additionally, we concluded the recent advances in engineered exosomes in the cancer fields, including the sources, isolation technologies, modification strategies, and labeling and imaging methods of engineered exosomes. Furthermore, the applications of engineered exosomes in different antitumor therapies were summarized, such as photodynamic therapy, gene therapy, and immunotherapy. Consequently, the above provides the cancer researchers in this community with the latest ideas on engineered exosome modification and new direction of new drug development, which is prospective to accelerate the clinical translation of engineered exosomes for cancer-targeted therapy.
Collapse
Affiliation(s)
- Menghui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Shengyun Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Lin Liu
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.,Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Pengyuan Dang
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China
| | - Yang Liu
- Department of Radiotherapy, Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, Henan, 450001, China
| | - Zhenqiang Sun
- Department of Colorectal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China. .,Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Bingbing Qiao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| | - Chengzeng Wang
- Henan Institute of Interconnected Intelligent Health Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China. .,Department of Ultrasound, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450001, China.
| |
Collapse
|
17
|
Sen S, Xavier J, Kumar N, Ahmad MZ, Ranjan OP. Exosomes as natural nanocarrier-based drug delivery system: recent insights and future perspectives. 3 Biotech 2023; 13:101. [PMID: 36860361 PMCID: PMC9970142 DOI: 10.1007/s13205-023-03521-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2022] [Accepted: 02/13/2023] [Indexed: 03/03/2023] Open
Abstract
Exosomes are nanosized (size ~ 30-150 nm) natural vesicular structures released from cells by physiological processes or pathological circumstances. Exosomes are growing in popularity as a result of their many benefits over conventional nanovehicles, including their ability to escape homing in the liver or metabolic destruction and their lack of undesired accumulation before reaching their intended targets. Various therapeutic molecules, including nucleic acids, have been incorporated into exosomes by different techniques, many of which have shown satisfactory performance in various diseases. Surface-modified exosomes are a potentially effective strategy, and it increases the circulation time and produces the specific drug target vehicle. In this comprehensive review, we describe composition exosomes biogenesis and the role of exosomes in intercellular signaling and cell-cell communications, immune responses, cellular homeostasis, autophagy, and infectious diseases. In addition, we discuss the role of exosomes as diagnostic markers, and their therapeutic and clinical implications. Furthermore, we addressed the challenges and outstanding developments in exosome research and discuss future perspectives. In addition to the current status of exosomes as a therapeutic carrier, the lacuna in the clinical development lifecycles along with the possible strategies to fill the lacuna have been addressed.
Collapse
Affiliation(s)
- Srijita Sen
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101 India
| | - Joyal Xavier
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar 844102 India
| | - Nitesh Kumar
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Hajipur, Bihar 844102 India
| | - Mohammad Zaki Ahmad
- Department of Pharmaceutics, College of Pharmacy, Najran University, Najran, 11001 Kingdom of Saudi Arabia
| | - Om Prakash Ranjan
- Department of Pharmaceutical Technology (Formulations), National Institute of Pharmaceutical Education and Research (NIPER), Guwahati, Assam 781101 India
| |
Collapse
|
18
|
Cheng W, Sun Y, Zhao G, Khan A, Zhang J, Zhang Z, Yi Y, Kong D, Li J. A novel peptide-templated AgNPs nanoprobe for theranostics of prostate cancer. Biosens Bioelectron 2023; 223:114978. [PMID: 36586149 DOI: 10.1016/j.bios.2022.114978] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/17/2022] [Accepted: 11/29/2022] [Indexed: 12/23/2022]
Abstract
Prostate specific membrane antigen (PSMA)-positive exosomes have the potential to serve as highly sensitive biomarkers for prostate cancer detection. Herein, a sensitive electrochemical biosensor for the ultrasensitive detection of PSMA-positive exosomes has been constructed based on a peptide-templated AgNPs nanoprobe. In this work, PSMA-specific binding peptides immobilized on a gold electrode were responsible for prostate cancer-derived exosomes capturing. Well-designed peptide (CCY- LWYIKC) serves a dual role: as a signal probe and as a recognizer in the exosomes-identification process. Specifically, LWYIKC bind to cholesterol at the exosome membranes, and CCY function as peptide templates to host a large number of silver nanoparticles, leading to a strong electrochemical signal. Thus, the concentration of exosomes can be quantified via electrochemical signal. This innovative method displayed a wide detection range of 102 to 108 particles/μL and a detection limit as low as 37 particles/μL. Notably, the method has shown outstanding performance when validated using clinical samples, suggesting its potential for clinical applications.
Collapse
Affiliation(s)
- Wenting Cheng
- Department of Clinical Laboratory, Gaochun People's Hospital, Nanjing, 211300, China
| | - Ying Sun
- Department of Clinical Laboratory, Gaochun People's Hospital, Nanjing, 211300, China
| | - Guiping Zhao
- Department of Clinical Laboratory, Gaochun People's Hospital, Nanjing, 211300, China
| | - Adeel Khan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Jianchun Zhang
- Department of Clinical Laboratory, Gaochun People's Hospital, Nanjing, 211300, China
| | - Zhaoli Zhang
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China
| | - Yongxiang Yi
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| | - Dehua Kong
- Department of Clinical Laboratory, Gaochun People's Hospital, Nanjing, 211300, China.
| | - Jinlong Li
- The Second Hospital of Nanjing, Nanjing University of Chinese Medicine, Nanjing, 210003, China.
| |
Collapse
|
19
|
Sun JX, Xu JZ, An Y, Ma SY, Liu CQ, Zhang SH, Luan Y, Wang SG, Xia QD. Future in precise surgery: Fluorescence-guided surgery using EVs derived fluorescence contrast agent. J Control Release 2023; 353:832-841. [PMID: 36496053 DOI: 10.1016/j.jconrel.2022.12.013] [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: 10/28/2022] [Revised: 12/04/2022] [Accepted: 12/05/2022] [Indexed: 12/23/2022]
Abstract
Surgery is the only cure for many solid tumors, but positive resection margins, damage to vital nerves, vessels and organs during surgery, and the range and extent of lymph node dissection are significant concerns which hinder the development of surgery. The emergence of fluorescence-guided surgery (FGS) means a farewell to the era when surgeons relied only on visual and tactile feedback, and it gives surgeons another eye to distinguish tumors from normal tissues for precise resection and helps to find a balance between complete tumor lesions removal and maximal organ function conservation. However, the existing synthetic fluorescence contrast agent has flaws in safety, specificity and biocompatibility to various extents. Extracellular vesicles (EVs) are a group of heterogeneous types of cell-derived membranous structures present in all biological fluids. EVs, especially engineered targeting EVs, play an increasingly important role in drug delivery because of their good biocompatibility, validated safety and targeting ability. Nevertheless, few studies have employed EVs loaded with fluorophores to construct fluorescence contrast agents and used them in FGS. Here, we systematically reviewed the current state of knowledge regarding FGS, fundamental characteristics of EVs, and the development of engineered targeting EVs, and put forward a novel strategy and procedures to produce EVs-based fluorescence contrast agent used in fluorescence-guided surgery.
Collapse
Affiliation(s)
- Jian-Xuan Sun
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Jin-Zhou Xu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Ye An
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Si-Yang Ma
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Chen-Qian Liu
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Si-Han Zhang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China
| | - Yang Luan
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China.
| | - Shao-Gang Wang
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China.
| | - Qi-Dong Xia
- Department and Institute of Urology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, 430030 Wuhan, China.
| |
Collapse
|
20
|
Zhang M, Lu Y, Wang L, Mao Y, Hu X, Chen Z. Current Status of Research on Small Extracellular Vesicles for the Diagnosis and Treatment of Urological Tumors. Cancers (Basel) 2022; 15:cancers15010100. [PMID: 36612097 PMCID: PMC9817817 DOI: 10.3390/cancers15010100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 12/17/2022] [Accepted: 12/20/2022] [Indexed: 12/28/2022] Open
Abstract
Extracellular vesicles (EVs) are important mediators of communication between tumor cells and normal cells. These vesicles are rich in a variety of contents such as RNA, DNA, and proteins, and can be involved in angiogenesis, epithelial-mesenchymal transition, the formation of pre-metastatic ecological niches, and the regulation of the tumor microenvironment. Small extracellular vesicles (sEVs) are a type of EVs. Currently, the main treatments for urological tumors are surgery, radiotherapy, and targeted therapy. However, urological tumors are difficult to diagnose and treat due to their high metastatic rate, tendency to develop drug resistance, and the low sensitivity of liquid biopsies. Numerous studies have shown that sEVs offer novel therapeutic options for tumor treatment, such as tumor vaccines and tumor drug carriers. sEVs have attracted a great deal of attention owing to their contribution to in intercellular communication, and as novel biomarkers, and role in the treatment of urological tumors. This article reviews the research and applications of sEVs in the diagnosis and treatment of urological tumors.
Collapse
Affiliation(s)
- Mengting Zhang
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Yukang Lu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Lanfeng Wang
- Department of Nephrology, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Yiping Mao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Xinyi Hu
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - Zhiping Chen
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou 341000, China
- Department of Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
- Correspondence: ; Tel.: +86-150-8373-7280
| |
Collapse
|
21
|
Yang F, Wang M, Guan X. Exosomes and mimics as novel delivery platform for cancer therapy. Front Pharmacol 2022; 13:1001417. [PMID: 36313380 PMCID: PMC9602403 DOI: 10.3389/fphar.2022.1001417] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Accepted: 09/30/2022] [Indexed: 11/15/2022] Open
Abstract
Exosomes are nano-sized biological extracellular vesicles transmitting information between cells and constituting a new intercellular communication mode. Exosomes have many advantages as an ideal drug delivery nanocarrier, including good biocompatibility, permeability, low toxicity, and low immunogenicity. Recently, exosomes have been used to deliver chemotherapeutic agents, natural drugs, nucleic acid drugs, and other antitumor drugs to treat many types of tumors. Due to the limited production of exosomes, synthetic exosome-mimics have been developed as an ideal platform for drug delivery. This review summarizes recent advances in the application of exosomes and exosome-mimics delivering therapeutic drugs in treating cancers.
Collapse
Affiliation(s)
- Fuxu Yang
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
- Key Laboratory of Pharmaceutics and Bioengineering, School of Medical Technology, Beihua University, Jilin, China
| | - Mingyue Wang
- Key Laboratory of Pharmaceutics and Bioengineering, School of Medical Technology, Beihua University, Jilin, China
- Center of Reproductive Medicine and Center of Prenatal Diagnosis, First Hospital, Jilin University, Changchun, China
| | - Xingang Guan
- Department of Basic Medicine, School of Medicine, Taizhou University, Taizhou, China
- *Correspondence: Xingang Guan,
| |
Collapse
|
22
|
Cui X, Fu Q, Wang X, Xia P, Cui X, Bai X, Lu Z. Molecular mechanisms and clinical applications of exosomes in prostate cancer. Biomark Res 2022; 10:56. [PMID: 35906674 PMCID: PMC9338661 DOI: 10.1186/s40364-022-00398-w] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 07/11/2022] [Indexed: 11/30/2022] Open
Abstract
Prostate cancer (PC) is a common tumor in men, and the incidence rate is high worldwide. Exosomes are nanosized vesicles released by all types of cells into multiple biological fluid types. These vesicles contribute to intercellular communication by delivering both nucleic acids and proteins to recipient cells. In recent years, many studies have explored the mechanisms by which exosomes mediate the epithelial-mesenchymal transition, angiogenesis, tumor microenvironment establishment, and drug resistance acquisition in PC, and the mechanisms that have been identified and the molecules involved have provided new perspectives for the possible discovery of novel diagnostic markers in PC. Furthermore, the excellent biophysical properties of exosomes, such as their high stability, high biocompatibility and ability to cross biological barriers, have made exosomes promising candidates for use in novel targeted drug delivery system development. In this review, we summarize the roles of exosomes in the growth and signal transmission in PC and show the promising future of exosome contributions to PC diagnostics and treatment.
Collapse
Affiliation(s)
- Xiaolin Cui
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.,Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Qiang Fu
- Department of Urology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Xueying Wang
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.,Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Pengcheng Xia
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.,Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Xianglun Cui
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.,Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Xiaohui Bai
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.,Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China
| | - Zhiming Lu
- Department of Clinical Laboratory Medicine, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China. .,Department of Clinical Laboratory, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|
23
|
Panigrahi AR, Srinivas L, Panda J. Exosomes: Insights and therapeutic applications in cancer. Transl Oncol 2022; 21:101439. [PMID: 35551002 PMCID: PMC9108525 DOI: 10.1016/j.tranon.2022.101439] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 04/21/2022] [Indexed: 12/19/2022] Open
Abstract
Cancer refers to the division of abnormal cells at an uncontrollable rate that possesses the ability to infiltrate and destroy normal tissues. It frequently spreads to normal tissues throughout the body, a condition known as metastasis, which is a significant concern. It is the second leading cause of mortality globally and treatment therapy can assist in improving survival rates. Exosomes are the extracellular vesicles secreted by several cells that act as messengers between cells. When engineered, exosomes act as promising drug delivery vehicles that help achieve targeted action at the tumour site and reduce the limitations of conventional treatments such as castration, chemotherapy, radiation, etc. The present review provides an overview of exosomes, the biogenesis, sources, isolation methods and characterization. The current status and applications of chemotherapeutic agents loaded, engineered exosomes in cancer treatment were convoluted.
Collapse
Affiliation(s)
- Anita Raj Panigrahi
- GITAM Institute of Pharmacy, GITAM Deemed to be University, Rushikonda, Visakhapatnam, 530045, India
| | - Lankalapalli Srinivas
- GITAM Institute of Pharmacy, GITAM Deemed to be University, Rushikonda, Visakhapatnam, 530045, India.
| | - Jagadeesh Panda
- Raghu College of Pharmacy, Dakamarri, Visakhapatnam - 531162, India
| |
Collapse
|
24
|
Pereira S, Ma G, Na L, Hudoklin S, Kreft ME, Kostevsek N, Al-Jamal WT. Encapsulation of doxorubicin prodrug in heat-triggered liposomes overcomes off-target activation for advanced prostate cancer therapy. Acta Biomater 2022; 140:530-546. [PMID: 34954416 DOI: 10.1016/j.actbio.2021.12.019] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Revised: 12/14/2021] [Accepted: 12/17/2021] [Indexed: 12/29/2022]
Abstract
L-377,202 prodrug consists of doxorubicin (Dox) conjugated to a prostate-specific antigen (PSA) peptide substrate that can be cleaved by enzymatically active PSA at the tumor site. Despite the initial promise in phase I trial, further testing of L-377,202 (herein called Dox-PSA) was ceased due to some degree of non-specific activation and toxicity concerns. To improve safety of Dox-PSA, we encapsulated it into low temperature-sensitive liposomes (LTSL) to bypass systemic activation, while maintaining its biological activity upon controlled release in response to mild hyperthermia (HT). A time-dependent accumulation of activated prodrug in the nuclei of PSA-expressing cells exposed to mild HT was observed, showing that Dox-PSA was efficiently released from the LTSL, cleaved by PSA and entering the cell nucleus as free Dox. Furthermore, we have shown that Dox-PSA loading in LTSL can block its biological activity at 37°C, while the combination with mild HT resulted in augmented cytotoxicity in both 2D and 3D PC models compared to the free Dox-PSA. More importantly, Dox-PSA encapsulation in LTSL prolonged its blood circulation and reduced Dox accumulation in the heart of C4-2B tumor-bearing mice over the free Dox-PSA, thus significantly improving Dox-PSA therapeutic window. Finally, Dox-PSA-loaded LTSL combined with HT significantly delayed tumor growth at a similar rate as mice treated with free Dox-PSA in both solid and metastatic PC tumor models. This indicates this strategy could block the systemic cleavage of Dox-PSA without reducing its efficacy in vivo, which could represent a safer option to treat patients with locally advanced PC. STATEMENT OF SIGNIFICANCE: This study investigates a new tactic to tackle non-specific cleavage of doxorubicin PSA-activatable prodrug (L-377,202) to treat advanced prostate cancer. In the present study, we report a nanoparticle-based approach to overcome the non-specific activation of L-377,202 in the systemic circulation. This includes encapsulating Dox-PSA in low temperature-sensitive liposomes to prevent its premature hydrolysis and non-specific cleavage. This class of liposomes offers payload protection against degradation in plasma, improved pharmacokinetics and tumor targeting, and an efficient and controlled drug release triggered by mild hyperthermia (HT) (∼42°C). We believe that this strategy holds great promise in bypassing any systemic toxicity concerns that could arise from the premature activation of the prodrug whilst simultaneously being able to control the spatiotemporal context of Dox-PSA cleavage and metabolism.
Collapse
|
25
|
Filipović L, Kojadinović M, Popović M. Exosomes and exosome-mimetics as targeted drug carriers: Where we stand and what the future holds? J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2021.103057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
26
|
Dually targeted bioinspired nanovesicle delays advanced prostate cancer tumour growth in vivo. Acta Biomater 2021; 134:559-575. [PMID: 34274531 DOI: 10.1016/j.actbio.2021.07.021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2021] [Revised: 07/07/2021] [Accepted: 07/08/2021] [Indexed: 01/03/2023]
Abstract
Prostate cancer (PC) is second-leading cancer in men, with limited treatment options available for men with advanced and metastatic PC. Prostate-specific antigen (PSA) and prostate-specific membrane antigen (PSMA) have been exploited as therapeutic targets in PC due to their upregulation in the advanced stages of the disease. To date, several PSA- and PSMA-activatable prodrugs have been developed to reduce the systemic toxicity of existing chemotherapeutics. Bioinspired nanovesicles have been exploited in drug delivery, offering prolonged drug blood circulation and higher tumour accumulation. For the first time, this study describes the engineering of dually targeted PSA/PSMA nanovesicles for advanced PC. PSMA-targeted bioinspired hybrids were prepared by hydrating a lipid film with anti-PSMA-U937 cell membranes and DOX-PSA prodrug, followed by extrusion. The bioinspired hybrids were characterised using dynamic light scattering, transmission electron microscopy, Dot blot, flow cytometry and Western blot. Cellular binding and toxicity studies in PC cancer cell lines were carried out using flow cytometry, confocal microscopy, and resazurin assay. Finally, tumour targeting and therapeutic efficacy studies were performed in solid and metastatic C4-2B-tumor-bearing mice. Interestingly, our PSMA-targeted hybrids demonstrated high cell uptake in PSMA-expressing cells with significant accumulation in solid and metastatic C4-2B tumour tissues following intravenous administration. More promisingly, our dually targeted PSA/PSMA hybrid significantly slowed down the C4-2B tumour growth in vivo, compared to free DOX-PSA and non-targeted PSA-hybrid. Our PSA/PSMA bioinspired hybrid could offer a highly selective treatment for advanced PC with lower side effects. STATEMENT OF SIGNIFICANCE: This study investigates a new approach to treat prostate cancer using dually targeted bioinspired nanovesicle . Our bioinspired vesicles are made mainly of a human blood cell membrane with a ligand recognising a specific marker (PSMA) on the surface of the prostate cancer cells. The present work describes the successful loading of a doxorubicin prodrug linked to a PSA- activatable peptide into these targeted bioinspired nanovesicle , where the active PSA enzyme presents in these cells converts the drug to its active form. Our dually targeted PSA/PSMA hybrid vesicles has successfully improved site-specific prodrug delivery to tackle advanced prostate cancer, offering a novel and effective prostate cancer treatment.
Collapse
|
27
|
Fu Y, Wang L, Liu W, Yang L, Li L, Wang L, Sun X, Zhang ZR, Lin Q, Zhang L. OX40L blockade cellular nanovesicles for autoimmune diseases therapy. J Control Release 2021; 337:557-570. [PMID: 34371056 DOI: 10.1016/j.jconrel.2021.08.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 08/03/2021] [Accepted: 08/04/2021] [Indexed: 12/13/2022]
Abstract
Current clinical agents for autoimmunity disorders treatment often cause substantial adverse effects and safety concerns, owing to non-specific immune modulation. Due to the prominent contribution of effector T cells in pathogenesis of rheumatoid arthritis (RA) and inflammatory bowel disease (IBD), and preferential location of co-stimulatory receptor-ligand pair OX40-OX40L at the inflamed sites, selectively targeting autoaggressive T cells by blockade OX40-OX40L, might represent an alternative strategy. Herein, we developed a new strategy to antagonize OX40-OX40L interaction by engineering a cell membrane derived nanovesicles (NVs) expressing OX40 receptors (OX40 NVs), and explored their potential for autoimmune disorders therapy. OX40 NVs showed specific binding capability to inflamed HUVECs in vitro, it also possessed distinct arthritic-targeting capacity in RA inflamed joints, and preferential accumulation in IBD inflamed colon. OX40 NVs efficiently suppressed the progression of both RA and IBD diseases through reducing CD4+OX40+ T cells population, and proinflammatory cytokines (i.e., TNF-α and IL-1β), while reinforcing Tregs immune-suppressive effect, with superior therapeutic efficacy than anti-OX40L. Additionally, dexamethasone (DEX) loading can further enhance the potential of OX40 NVs for RA treatment. Owing to their preferential localization to inflamed sites, and potent immune-suppression ability, targeting OX40-OX40L blockade by OX40 NVs for autoimmune therapy is highly promising.
Collapse
Affiliation(s)
- Yu Fu
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china; College of Pharmaceutical Sciences, Southwest University, 400715, China
| | - Leilei Wang
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china
| | - Wei Liu
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china
| | - Lan Yang
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china
| | - Lin Li
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china
| | - Luyao Wang
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china
| | - Xun Sun
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china
| | - Zhi-Rong Zhang
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china
| | - Qing Lin
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china.
| | - Ling Zhang
- College of Polymer Science and Engineering, Key Laboratory of Drug Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, 640041, china.
| |
Collapse
|