1
|
Wang H, Zhang W, Sun Y, Xu X, Chen X, Zhao K, Yang Z, Liu H. Nanotherapeutic strategies exploiting biological traits of cancer stem cells. Bioact Mater 2025; 50:61-94. [PMID: 40242505 PMCID: PMC12002948 DOI: 10.1016/j.bioactmat.2025.03.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 03/08/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer stem cells (CSCs) represent a distinct subpopulation of cancer cells that orchestrate cancer initiation, progression, metastasis, and therapeutic resistance. Despite advances in conventional therapies, the persistence of CSCs remains a major obstacle to achieving cancer eradication. Nanomedicine-based approaches have emerged for precise CSC targeting and elimination, offering unique advantages in overcoming the limitations of traditional treatments. This review systematically analyzes recent developments in nanomedicine for CSC-targeted therapy, emphasizing innovative nanomaterial designs addressing CSC-specific challenges. We first provide a detailed examination of CSC biology, focusing on their surface markers, signaling networks, microenvironmental interactions, and metabolic signatures. On this basis, we critically evaluate cutting-edge nanomaterial engineering designed to exploit these CSC traits, including stimuli-responsive nanodrugs, nanocarriers for drug delivery, and multifunctional nanoplatforms capable of generating localized hyperthermia or reactive oxygen species. These sophisticated nanotherapeutic approaches enhance selectivity and efficacy in CSC elimination, potentially circumventing drug resistance and cancer recurrence. Finally, we present an in-depth analysis of current challenges in translating nanomedicine-based CSC-targeted therapies from bench to bedside, offering critical insights into future research directions and clinical implementation. This review aims to provide a comprehensive framework for understanding the intersection of nanomedicine and CSC biology, contributing to more effective cancer treatment modalities.
Collapse
Affiliation(s)
- Hongyu Wang
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Wenjing Zhang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Yun Sun
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xican Xu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Xiaoyang Chen
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Kexu Zhao
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Zhao Yang
- State Key Laboratory of Green Biomanufacturing, Innovation Center of Molecular Diagnostics, College of Life Science and Technology, Beijing University of Chemical Technology, 100029, Beijing, China
| | - Huiyu Liu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Advanced Innovation Center for Soft Matter Science and Engineering, Beijing Key Laboratory of Bioprocess, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, 100029, Beijing, China
| |
Collapse
|
2
|
Khidr WA, Alfarttoosi KH, Taher WM, Alwan M, Ali Al-Nuaimi AM, Jawad MJ. A review of the role of tumor-derived exosomes in cancers treatment and progression. Int Immunopharmacol 2025; 157:114782. [PMID: 40334624 DOI: 10.1016/j.intimp.2025.114782] [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: 03/11/2025] [Revised: 04/23/2025] [Accepted: 04/29/2025] [Indexed: 05/09/2025]
Abstract
Tumor cells (TCs) produce exosomes (EXOs), nanovesicles formed in endosomes. Tumor-derived exosomes (TDEs) are tiny, bubble-shaped structures formed by TCs that include microRNAs (miRNA), proteins, enzymes, and copies of DNA and RNA. Many different kinds of cancer rely on TDEs. For instance, TDEs play a large role in the tumor microenvironment (TME) and promote tumor spread via many pathways. Furthermore, TDEs impact the efficacy of cancer treatments. Additionally, because of their low immunogenicity, high biocompatibility, and low toxicity, TDEs have been extensively used as drug delivery vehicles for cancer immunotherapy. Consequently, future cancer treatments may benefit from focusing on both the therapeutic function and the tumorigenic pathways of TDEs. Consequently, in this work, we have examined the roles of TDEs in cancer development, such as tumor angiogenesis, immune system evasion, and tumor metastasis. Then, we reviewed TDEs used to transport anticancer medicines, including chemotherapeutic medications, therapeutic compounds (including miRNA), and anticancer nanoparticles. We have concluded by outlining the challenges of clinical translation, including carcinogenicity and medication resistance, and by offering some suggestions for addressing these issues.
Collapse
Affiliation(s)
- Wajida Ataallah Khidr
- Department of Medical Laboratory Technics, College of Health and Medical Technology, Alnoor University, Mosul, Iraq
| | | | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
3
|
Dietz L, Simon J, Speth KR, Landfester K, Mailänder V. Plasma protein corona on silica nanoparticles enhances exocytosis. Biomater Sci 2025. [PMID: 40433828 DOI: 10.1039/d4bm01189a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/29/2025]
Abstract
While the influence of the protein corona on nanoparticle uptake in mammalian cells is well understood, little is known about the influence of the protein corona on nanoparticle exocytosis. However, the exocytosis of nanoparticles also contributes to the therapeutic efficacy as it influences the net delivery of nanoparticles to a cell. In this study we demonstrate that the exocytosis of silica nanoparticles from HCT 116 cells is enhanced by the pre-adsorption of a human plasma protein corona. This pre-adsorption effect also depends on the diameter of the nanoparticles. The exocytosis of small silica nanoparticles (10 nm) is less pronounced, while the exocytosis of larger silica nanoparticles (100 nm) is significantly increased in the presence of a protein corona. A proteomic analysis of the plasma protein corona of the different-sized silica nanoparticles (10 nm, 30 nm, 50 nm, and 100 nm) reveals different protein compositions. Apolipoproteins and coagulation proteins are enriched in a size-dependent manner with high amounts of apolipoproteins adsorbed to small silica nanoparticles. The findings underscore the significance of the nanoparticle protein corona for exocytosis and demonstrate the need to engineer nanocarriers that are not exocytosed rapidly to enhance the efficacy in drug delivery.
Collapse
Affiliation(s)
- Laura Dietz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Julia Simon
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Kai R Speth
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| | - Katharina Landfester
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | - Volker Mailänder
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
- Department of Dermatology, University Medical Center Mainz, Langenbeckstraße 1, 55131 Mainz, Germany
| |
Collapse
|
4
|
Zhang Y, Li W, Chen S, Zhang Y, Zhu Y, Lan F, Du H, Fan R, Zhu J, Pan W, Situ B, Zheng L, Luo S, Yan X. Layered-Responsive Multivalent Tetrahedral DNA Framework-Decorated CRISPR-Cas12a Nanocapsule Enables Precise and Enhanced Tumor Chemotherapy. ACS NANO 2025; 19:19274-19286. [PMID: 40366179 DOI: 10.1021/acsnano.5c01747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
The lack of selective tumor targeting and the high toxicity of conventional chemotherapy treatments remain major challenges in cancer therapy. Here, we develop a self-controlled DNA nanostructure-CRISPR-12a system, a triple-locked cascade tumor therapy nanocapsule (Tatna), for efficient and targeted tumor treatment. Tatna integrates structural DNA tetrahedrons (DTs) with high drug-loading capacity, Cas12a/crRNA ribonucleoprotein (Cas12a RNP), and doxorubicin (DOX) to enable multisite response for precise drug delivery and augmented tumor treatment. By incorporation of a nucleolin-targeting aptamer, Tatna achieves selective targeting and efficient tumor cell internalization. Encapsulation in pH-responsive poly l-lactic-co-glycolic acid (PLGA) nanocapsule ensures stable circulation and controlled release of both DOX and Cas12a until tumor-specific activation in the acidic microenvironment. The Cas12a RNP, triggered by APE1 mRNA overexpression in tumor cells, induces trans-cleavage of DTs, releasing DOX and Cas12a to transport into the nucleus and induce enhanced cell apoptosis. This self-regulating and multifunctional approach enhances the efficacy of chemotherapy while reducing off-target effects. Tatna's programmable, tumor-specific delivery system represents a powerful strategy for advancing precision medicine and personalized cancer treatment.
Collapse
Affiliation(s)
- Ye Zhang
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Wenbin Li
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Siting Chen
- School of Basic Medical Sciences, Medical Research Center of Nanfang Hospital, Department of Laboratory Medicine, Southern Medical University, Guangzhou 510515, P. R. China
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, P. R. China
| | - Yu Zhang
- School of Basic Medical Sciences, Medical Research Center of Nanfang Hospital, Department of Laboratory Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Yitong Zhu
- School of Basic Medical Sciences, Medical Research Center of Nanfang Hospital, Department of Laboratory Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| | - Fei Lan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Huijun Du
- The Affiliated Qingyuan Hospital (Qingyuan People's Hospital), Guangzhou Medical University, Qingyuan, Guangdong 511518, P. R. China
| | - Rui Fan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Junfang Zhu
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, P. R. China
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, P. R. China
| | - Weilun Pan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Bo Situ
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Lei Zheng
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
| | - Shihua Luo
- Center for Clinical Laboratory Diagnosis and Research, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, P. R. China
- Key Laboratory of Research on Clinical Molecular Diagnosis for High Incidence Diseases in Western Guangxi of Guangxi Higher Education Institutions, Affiliated Hospital of Youjiang Medical University for Nationalities, Baise 533000, Guangxi, P. R. China
| | - Xiaohui Yan
- Department of Laboratory Medicine, Medical Research Center of Nanfang Hospital, Guangdong Provincial Key Laboratory of Precision Medical Diagnostics, Guangdong Engineering and Technology Research Center for Rapid Diagnostic Biosensors, Guangdong Provincial Key Laboratory of Single-cell and Extracellular Vesicles, Nanfang Hospital, Southern Medical University, Guangzhou 510515, P. R. China
- School of Basic Medical Sciences, Medical Research Center of Nanfang Hospital, Department of Laboratory Medicine, Southern Medical University, Guangzhou 510515, P. R. China
| |
Collapse
|
5
|
Zaiki Y, Yap PG, Gan CY, Rani MFA, Traini D, Wong TW. "Actual" peptide properties required for nanoparticle development in precision cancer therapeutic delivery. J Control Release 2025:113866. [PMID: 40412661 DOI: 10.1016/j.jconrel.2025.113866] [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: 02/19/2025] [Revised: 04/27/2025] [Accepted: 05/18/2025] [Indexed: 05/27/2025]
Abstract
Functionalizing nanoparticles with peptides (3-30 amino acids) reduces premature clearance and increases colloidal stability and targeting capacity of cancer therapeutics. Glutamate/lysine-rich zwitterionic and hydrophilic/neutral peptides minimize reticuloendothelial digestion of nanomedicine through reducing particle hydrophobicity and depressing plasma anti-PEG immunoglobulin that disrupts the PEG-based particle stealth. Anionic peptides negate protein corona formation and subsequent particle aggregation in vivo enabling efficient nanoparticles biodistribution and drug targeting by facilitating their endothelial/extracellular matrix pore diffusion. Cationic and hydrophobic peptides display a strong affinity for anionic cancer cell membrane and mediate membrane porosification or receptor binding leading to particle uptake and endocytosis. The peptide ionic and hydrophobicity/hydrophilicity attributes collectively facilitate endosomal escape, and nuclear and mitochondria targeting of nanoparticles. Peptides are required to present with different physicochemical attributes from administration site, through blood and extracellular matrix, to cancer site of action. Charge/hydrophilicity-hydrophobicity switching and projection of receptor-specific domain of peptides are attainable through pH-pKa interplay and labile bond hydrolysis of "unwanted" domain to give rise to new functional domains in response to pH, thermal and enzymatic stimuli. Co-introducing all functional attributes on a single peptide is challenging. Use of peptide blends risks leaching during nanoparticles production. Peptides-nanoparticles conjugation risks peptide conformational alterations and loss of acidic/basic termini affecting their roles in nanoparticle stabilization, targeting, membrane permeabilization and subcellular delivery.
Collapse
Affiliation(s)
- Yazid Zaiki
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia
| | - Pei Gee Yap
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia, University Innovation Incubator Building, SAINS@USM campus, Lebuh Bukit Jambul, Bayan Lepas, 11900, Penang, Malaysia
| | - Chee Yuen Gan
- Analytical Biochemistry Research Centre (ABrC), Universiti Sains Malaysia, University Innovation Incubator Building, SAINS@USM campus, Lebuh Bukit Jambul, Bayan Lepas, 11900, Penang, Malaysia
| | | | - Daniela Traini
- Woolcock Institute of Medical Research, 431 Glebe Point Road, Glebe, Sydney 2037, Australia; Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Campus Macquarie Park, Sydney 2019, Australia
| | - Tin Wui Wong
- Non-Destructive Biomedical and Pharmaceutical Research Centre, Smart Manufacturing Research Institute, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Particle Design Research Group, Faculty of Pharmacy, Universiti Teknologi MARA Selangor, Puncak Alam 42300, Selangor, Malaysia; Department of Industrial Pharmacy, Faculty of Pharmacy, Silpakorn University, Nakhon Pathom 73000, Thailand.
| |
Collapse
|
6
|
Wang X, Liu Y, Jiang Y, Li Q. Tumor-derived exosomes as promising tools for cancer diagnosis and therapy. Front Pharmacol 2025; 16:1596217. [PMID: 40444049 PMCID: PMC12119533 DOI: 10.3389/fphar.2025.1596217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Accepted: 05/06/2025] [Indexed: 06/02/2025] Open
Abstract
Mounting evidences indicated that cancer cell-derived exosomes (TDEs) contribute to cancer progression and metastasis by reshaping the tumor microenvironment (TME) and interfering immunity response. TDEs contain unique biomolecular cargo, consisting of protein, nucleic acid, and lipids. In recent years, TDEs have been used as potential disease therapeutics and diagnosis biomarkers and prime candidates as delivery tools for cancer treatment. In the present review, we firstly summarized TDEs biogenesis and characteristic. Also, the role of TDEs in cancer cell metastasis and invasiveness, drug resistance, and immunosuppression was mentioned via cell-cell communication. Additionally, we concluded the current strategies for TDE-based cancer therapy, including TDEs inhibition and clearance, usage as therapeutic drug delivery vector and cancer vaccines. Furthermore, combination therapy with engineered TDEs were summarized, such as radiotherapy, photodynamic therapy, photothermal therapy, and sonodynamic therapy. Consequently, the above opens up novel and interesting opportunities for cancer diagnosis and prognosis based on TDEs, which is prospective to accelerate the clinical translation of TDEs for cancer therapy.
Collapse
Affiliation(s)
- Xirui Wang
- Department of Biomedical Engineering, School of Medical Imaging Xuzhou Medical University, Xuzhou, China
| | - Yanfang Liu
- Department of Central Laboratory, Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Yaowen Jiang
- Department of Biomedical Engineering, School of Medical Imaging Xuzhou Medical University, Xuzhou, China
| | - Qinghua Li
- Institute of Medical Imaging and Artificial Intelligence, Jiangsu University, Zhenjiang, China
| |
Collapse
|
7
|
Wu KY, Dave A, Nirwal GK, Giunta M, Nguyen VDH, Tran SD. Exosome Innovations in Ophthalmology and Sjögren's Syndrome. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025. [PMID: 40360847 DOI: 10.1007/5584_2025_865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Exosomes, a subset of extracellular vesicles, have emerged as potential therapeutic agents in ophthalmology due to their ability to modulate immune responses, facilitate cellular communication, and promote tissue repair. This chapter explores the potential applications of exosome-based therapies in corneal and anterior segment disorders, retinal diseases, glaucoma, and Sjögren's syndrome. In corneal disorders, mesenchymal stem cell (MSC)-derived secretomes have shown promise in accelerating wound healing, reducing fibrosis, and modulating inflammation, with hydrogel encapsulation strategies potentially enhancing their efficacy. In retinal diseases, exosomes may provide neuroprotective effects in age-related macular degeneration, diabetic retinopathy, and retinitis pigmentosa by modulating oxidative stress and inflammation. In glaucoma, secretome-based therapies could support retinal ganglion cell survival and optic nerve regeneration, though their impact on intraocular pressure via the trabecular meshwork remains uncertain. Additionally, exosomal biomarkers in aqueous humor are being investigated as potential diagnostic tools. In Sjögren's syndrome, exosomal biomarkers may facilitate earlier detection, while stem cell-derived exosomes hold promise in modulating immune responses and restoring glandular function. Despite encouraging preclinical and early clinical findings, standardization, scalability, and long-term safety must be addressed before clinical translation. Future research will focus on optimizing exosome-based therapies and exploring their feasibility for ophthalmic applications.
Collapse
Affiliation(s)
- Kevin Y Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC, Canada.
| | - Archan Dave
- Department of Medicine, University of British Columbia, Vancouver, BC, Canada
| | - Gurleen K Nirwal
- Department of Zoology, University of British Columbia, Vancouver, BC, Canada
| | - Michel Giunta
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC, Canada
| | | | - Simon D Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC, Canada
| |
Collapse
|
8
|
Ling LA, Boukhalfa A, Kung AH, Yang VK, Chen HH. Advances in Targeted Autophagy Modulation Strategies to Treat Cancer and Associated Treatment-Induced Cardiotoxicity. Pharmaceuticals (Basel) 2025; 18:671. [PMID: 40430490 PMCID: PMC12114528 DOI: 10.3390/ph18050671] [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: 03/31/2025] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Autophagy, an evolutionarily conserved process, plays an important role in cellular homeostasis and human diseases. Cardiovascular dysfunction, which presents during cancer treatment or in cancer-free individuals years after treatment, is a growing clinical challenge. Millions of cancer survivors and patients face an unpredictable risk of developing cardiotoxicity. Cardiotoxicity due to cancer treatment, as well as cancer progression, has been linked to autophagy dysregulation. Modulating autophagy has been further proposed as a therapeutic treatment for both cancer and cardiovascular disorders. The safe and effective use of autophagy modulation as a cardioprotective strategy during cancer treatment especially requires careful consideration and experimentation to minimize the impact on cancer treatment. We focus here on recent advances in targeted autophagy modulation strategies that utilize interdisciplinary approaches in biomedical sciences and are potentially translatable to treat cardiotoxicity and improve cancer treatment outcomes. This review highlights non-small molecule autophagy modulators to enhance targeted therapy, nanomedicine for autophagy modulation and monitoring, and in vitro models and future experiments needed to bring novel autophagy discoveries from basic research to clinical translation.
Collapse
Affiliation(s)
- Lauren A. Ling
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA; (L.A.L.); (A.B.)
- School of Medicine, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA
| | - Asma Boukhalfa
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA; (L.A.L.); (A.B.)
| | - Andrew H. Kung
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA; (L.A.L.); (A.B.)
| | - Vicky K. Yang
- Cummings School of Veterinary Medicine, Tufts University, 200 Westboro Rd., North Grafton, MA 01536, USA;
| | - Howard H. Chen
- Molecular Cardiology Research Institute, Tufts Medical Center, 800 Washington Street, #80, Boston, MA 02111, USA; (L.A.L.); (A.B.)
- School of Medicine, Tufts University, 145 Harrison Avenue, Boston, MA 02111, USA
| |
Collapse
|
9
|
Song G, Zeng C, Li J, Liu J, Zhao J, Liu B, Fan J, Xie H. Exosome-based nanomedicines for digestive system tumors therapy. Nanomedicine (Lond) 2025; 20:1167-1180. [PMID: 40248953 PMCID: PMC12068745 DOI: 10.1080/17435889.2025.2493037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/10/2025] [Indexed: 04/19/2025] Open
Abstract
Digestive system tumors constitute a major subset of malignancies, consistently ranking among the leading causes of mortality globally. Despite limitations inherent in current therapeutic modalities, recent advancements in targeted therapy and drug delivery systems have led to significant improvements in the efficacy of pharmacotherapy for digestive system tumors. In this context, exosomes - naturally occurring nanoscale vesicles - have emerged as promising drug delivery candidates due to their intrinsic molecular transport capabilities, superior biocompatibility, and targeted recognition of tumor cells. The integration of exosomes into cancer therapeutics represents a novel and potentially transformative approach for treating digestive system tumors, which may drive further progress in this field. This review comprehensively examines the sources, loading mechanisms, and therapeutic efficacy of exosomes in the context of digestive system tumor treatment. Furthermore, it discusses the opportunities and challenges associated with exosomes, offering insights into their future role within the therapeutic armamentarium against digestive tumors.
Collapse
Affiliation(s)
- Ge Song
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Chenlu Zeng
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Junru Li
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Jiajia Liu
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Juanxia Zhao
- Department of Pathology, The Affiliated Nanhua Hospital, Hengyang Medical College, University of South China, Hengyang, Hunan, China
| | - Bin Liu
- College of Biology, Hunan University, Changsha, Hunan, China
| | - Jialong Fan
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Hailong Xie
- Key Laboratory of Cancer Cellular and Molecular Pathology in Hunan Province, Cancer Research Institute of Hengyang Medical College, University of South China, Hengyang, Hunan, China
| |
Collapse
|
10
|
Lizama-Muñoz A, Plaza-Diaz J. Bispecific Antibodies, Nanobodies and Extracellular Vesicles: Present and Future to Cancer Target Therapy. Biomolecules 2025; 15:639. [PMID: 40427532 PMCID: PMC12109199 DOI: 10.3390/biom15050639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/28/2025] [Accepted: 04/28/2025] [Indexed: 05/29/2025] Open
Abstract
Cancer remains one of the leading causes of mortality worldwide, with a growing need for precise and effective treatments. Traditional therapies such as chemotherapy and radiotherapy have limitations, including off-target effects and drug resistance. In recent years, targeted therapies have emerged as promising alternatives, aiming to improve treatment specificity and reduce systemic toxicity. Among the most innovative approaches, bispecific antibodies, nanobodies, and extracellular vesicles offer distinct and complementary mechanisms for cancer therapy. Bispecific antibodies enhance immune responses and enable dual-targeting of cancer cells, nanobodies provide superior tumor penetration due to their small size, and extracellular vesicles present a novel platform for drug and RNA delivery. This work aims to review and analyze these three approaches, assessing their current applications, advantages, challenges, and future perspectives.
Collapse
Affiliation(s)
- Asier Lizama-Muñoz
- Department of Biochemistry, Molecular Biology and Immunology III, Faculty of Medicine, University of Granada, 18016 Granada, Spain;
- Clinical Analysis and Immunology Department, University Hospital Virgen de las Nieves, 18014 Granada, Spain
| | - Julio Plaza-Diaz
- ANUT-DSM (Alimentaciò, Nutrició Desenvolupament i Salut Mental), Departament de Bioquímica i Biotecnologia, Universitat Rovira i Virgili, 43201 Reus, Spain
- School of Health Sciences, Universidad Internacional de La Rioja, Avenida de la Paz 137, 26006 Logroño, Spain
| |
Collapse
|
11
|
Fu G, Zhao Y, Mao C, Liu Y. Enhancing nano-immunotherapy of cancer through cGAS-STING pathway modulation. Biomater Sci 2025; 13:2235-2260. [PMID: 40111213 DOI: 10.1039/d4bm01532k] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Activation of the cyclic guanosine monophosphate-adenosine monophosphate synthase (cGAS)-stimulator of interferon genes (STING) pathway plays a critical role in cancer immunotherapy due to the secretion of multiple pro-inflammatory cytokines and chemokines. Numerous cGAS-STING agonists have been developed for preclinical and clinical trials in tumor immunity. However, several obstacles, such as agonist molecules requiring multiple doses, rapid degradation and poor targeting, weaken STING activation at the tumor site. The advancement of nanotechnology provides an optimized platform for the clinical application of STING agonists. In this review, we summarize events of cGAS-STING pathway activation, the dilemma of delivering STING agonists, and recent advances in the nano-delivery of cGAS-STING agonist formulations for enhancing tumor immunity. Furthermore, we address the future challenges associated with STING-based therapies and offer insights to guide subsequent clinical applications.
Collapse
Affiliation(s)
- Gaohong Fu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
| | - Yanan Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
| | - Chengqiong Mao
- Key Laboratory of Biological Targeting Diagnosis, Therapy and Rehabilitation of Guangdong Higher Education Institutes, The Fifth Affiliated Hospital, School of Biomedical Engineering, Guangzhou Medical University, Guangzhou 510180, P. R. China
| | - Yang Liu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, P. R. China.
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, P. R. China
- Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, P.R. China
| |
Collapse
|
12
|
Yang YP, Nicol CJB, Chiang MC. A Review of the Neuroprotective Properties of Exosomes Derived from Stem Cells and Exosome-Coated Nanoparticles for Treating Neurodegenerative Diseases and Stroke. Int J Mol Sci 2025; 26:3915. [PMID: 40332773 PMCID: PMC12028030 DOI: 10.3390/ijms26083915] [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: 02/18/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 05/08/2025] Open
Abstract
Neurological diseases, including neurodegenerative disorders and stroke, represent significant medical challenges due to their complexity and the limitations of current treatment approaches. This review explores the potential of stem cell (SC)-derived exosomes (Exos) as a transformative therapeutic strategy for these diseases. Exos, especially those derived from SCs, exhibit natural targeting ability, biocompatibility, and the capacity to cross the blood-brain barrier (BBB), making them ideal vehicles for drug delivery. This review provides an in-depth discussion of the properties and advantages of SC-Exos. It highlights their potential synergistic benefits in therapeutic approaches to treat neurological diseases. This article discusses the mechanisms of action of SC-Exos, highlighting their ability to target specific cells, modulate disease pathways, and provide controlled release of therapeutic agents. Applications in specific neurological disorders have been investigated, demonstrating the potential to improve outcomes in conditions such as Alzheimer's Disease (AD), Parkinson's Disease (PD), and stroke. Moreover, Exos-coated nanoparticles (NPs) combine the natural properties of Exos with the multifunctionality of NPs. This integration takes advantage of exosome membrane biocompatibility and targeting capabilities while preserving NPs' beneficial features, such as drug loading and controlled release. As a result, Exos-coated NPs may enhance the precision, efficacy, and safety of therapeutic interventions. In conclusion, SC-Exos represent a promising and innovative approach to treating neurological diseases.
Collapse
Affiliation(s)
- Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Christopher J. B. Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, and Cancer Biology and Genetics Division, Sinclair Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
13
|
Hsu CW, Fang YC, Li JF, Cheng CA. Decoding Complex Biological Milieus: SHINER's Approach to Profiling and Functioning of Extracellular Vesicle Subpopulations. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025:e2503638. [PMID: 40255212 DOI: 10.1002/smll.202503638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2025] [Indexed: 04/22/2025]
Abstract
Extracellular vesicles (EVs) are celebrated for their pivotal roles in cellular communication and their potential in disease diagnosis and therapeutic applications. However, their inherent heterogeneity acts as a double-edged sword, complicating the isolation of specific EV subpopulations. Conventional EV isolation methods often fall short, relying on biophysical properties, while affinity-based techniques may compromise EV integrity and utility with harsh recovery conditions. To address these limitations, the SHINER (subpopulation homogeneous isolation and nondestructive EV release) workflow is introduced, which redefines how EVs are isolated and recoverd, featuring the innovative SWITCHER (switchable extracellular vesicle releaser) tool. The SHINER workflow facilitates the precise purification and gentle recovery of target EV subpopulations from complex biological mixtures, preserving their structural integrity and biological functionality. Importantly, SHINER demonstrates exceptional adaptability to multiple markers and clinical applications. It not only enhances the ability to trace EV origins for accurate disease diagnosis but also advances fundamental EV research and provides standardized EV materials for therapeutic innovations. By improving the understanding of EVs and enabling the development of personalized diagnostics and treatments, SHINER propels EV-based science into new frontiers of advanced medicine, offering transformative potential for healthcare.
Collapse
Affiliation(s)
- Chen-Wei Hsu
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Yao-Ching Fang
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Jhih-Fong Li
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| | - Chi-An Cheng
- School of Pharmacy, College of Medicine, National Taiwan University, Taipei, 10050, Taiwan
| |
Collapse
|
14
|
Li X, Deng Z, Zhang W, Zhou W, Liu X, Quan H, Li J, Li P, Li Y, Hu C, Li F, Niu L, Tian Z, Meng L, Zheng H. Oscillating microbubble array-based metamaterials (OMAMs) for rapid isolation of high-purity exosomes. SCIENCE ADVANCES 2025; 11:eadu8915. [PMID: 40238867 PMCID: PMC12002133 DOI: 10.1126/sciadv.adu8915] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 03/12/2025] [Indexed: 04/18/2025]
Abstract
Exosomes secreted by cells hold substantial potential for disease diagnosis and treatment. However, the rapid isolation of high-purity exosomes and their subpopulations from biofluids (e.g., undiluted whole blood) remains challenging. This study presents oscillating microbubble array-based metamaterials (OMAMs) for enabling the rapid isolation of high-purity exosomes and their subpopulations from biofluids without labeling or preprocessing. Particularly, leveraging acoustically excited microbubble oscillation, OMAMs can generate numerous acoustofluidic traps for filtering in-fluid micro/nanoparticles, thus allowing for removing bioparticles larger than exosomes to obtain high-purity (93%) exosomes from undiluted whole blood in ~3 minutes. Moreover, exosome subpopulations in different size ranges can be isolated by tuning the microbubble oscillation amplitude. Additionally, as each oscillating microbubble functions as an ultradeep subwavelength (~λ/186) acoustic amplifier and a nonlinear source, OMAMs can generate high-resolution complex acoustic energy patterns and tune the patterns by activating different-sized microbubbles at their distinct resonance frequencies.
Collapse
Affiliation(s)
- Xinjia Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110016, China
| | - Zhiting Deng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Wenjun Zhang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
- School of Intelligent Manufacturing and Materials Engineering, Gannan University of Science and Technology, 156 Kejia Avenue, Ganzhou 341000, China
| | - Wei Zhou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Xiufang Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Hao Quan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110016, China
| | - Jiali Li
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Pengqi Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Yingyin Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110016, China
| | - Cai Hu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Fei Li
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Lili Niu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Zhenhua Tian
- Department of Mechanical Engineering, Virginia Polytechnic Institute and State University, Blacksburg, VA 24061, USA
| | - Long Meng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning 110016, China
- Guangdong Provincial Key Laboratory of Multimodality Non-Invasive Brain-Computer Interfaces, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Shenzhen 518055, China
| |
Collapse
|
15
|
Makkar S, Rana N, Priyadarshi N, Bajaj G, Kumar S, Singhal NK. Unravelling the therapeutic properties of aptamer-modified exosome nanocomposite. Adv Colloid Interface Sci 2025; 342:103517. [PMID: 40245577 DOI: 10.1016/j.cis.2025.103517] [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: 12/30/2024] [Revised: 04/08/2025] [Accepted: 04/09/2025] [Indexed: 04/19/2025]
Abstract
Exosomes are naturally occurring nanocarriers derived from various cells. In recent years, they have attained significant attention for their potential in precise drug delivery and therapeutic applications. Exosomes exhibit several advantages, remarkably improved stability, bioavailability, and delivery efficiency, which are further augmented by integration with nanomaterials. Functionalizing the aptamer and nanomaterial on the exosomal surface significantly improves the binding affinity and specificity. Here in this review, we examine the synergistic therapeutic effect of exosome-nanomaterial-aptamer conjugate with particular attention to their uses in cancer therapy, bone fracture regeneration, wound healing, etc. Recent advances in the field demonstrated that the amalgamation of different nanomaterials, aptamers, and exosomes has proven to be a transformative approach in the field of therapeutics. Here in the nanocomposite, the aptamer is exclusively used as a recognition molecule to provide specificity to the target cells. Exosomes serve as biocompatible nanocarriers, and different nanomaterials (AuNPs, AuNRs, SiNPs, Graphene, etc.) complement the therapeutic efficiency by PTT/PDT/ROS generation/SO generation, etc. Briefly, the above-mentioned nanocomposite serves as the perfect therapeutic agent by utilizing the exosome's biocompatibility, aptamer's high affinity and nanomaterial's multifunctionality. Furthermore, the challenges and limitations of this nanocomposite have been discussed, along with its prospects in clinical practices.
Collapse
Affiliation(s)
- Simran Makkar
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India; Department of Biotechnology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Niket Rana
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Nitesh Priyadarshi
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India
| | - Geetika Bajaj
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India; Department of Biotechnology, Panjab University, Sector 25, Chandigarh 160014, India
| | - Sandeep Kumar
- Department of Physics, Punjab Engineering College (Deemed to be University), Chandigarh 160012, India
| | - Nitin Kumar Singhal
- National Agri-Food and Biomanufacturing Institute (NABI), Sector-81, S.A.S. Nagar, Mohali 140306, Punjab, India.
| |
Collapse
|
16
|
Liu YY, Liu J, Guo Y, Zhang Q, Cao A, Wang H. Interactions between polystyrene nanoparticles and human intestinal epithelial Caco-2 cells. NANOIMPACT 2025; 38:100559. [PMID: 40220994 DOI: 10.1016/j.impact.2025.100559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/18/2025] [Accepted: 04/08/2025] [Indexed: 04/14/2025]
Abstract
Nanoplastics enter the human body mainly by ingestion through the gastrointestinal tract and thus the uptake and release of nanoplastics in intestinal cells have been studied. However, the fate of nanoplastics in intestinal cells remains poorly understood, particularly how they are exocytosed. Herein, we investigated the uptake, distribution, and exocytosis of nanoplastics in Caco-2 cells using 70 nm red fluorescent polystyrene (R70PS) as a nanoplastic model. The results show that R70PS readily enters Caco-2 cells and the content per cell peaks at around 24 h, but the total intracellular content in all cells increases continuously over 72 h. In addition, the uptake mechanisms change over incubation time, i.e. R70PS entered Caco-2 cells via both the energy-independent pathway and the energy-dependent caveolae-mediated endocytosis and macropinocytosis at 4 h incubation, but almost all R70PS entered cells in an energy-dependent manner via caveolae-mediated endocytosis, macropinocytosis, and clathrin-mediated endocytosis at 12 h incubation. Most of the intracellular R70PS accumulated in lysosomes, but R70PS also entered the mitochondria and its level increased over time. Approximately 45 % of the intracellular R70PS could be cleared from the cells within 12 h, mainly via the lysosomal pathway. Exocytosis was also associated with autophagy and was facilitated by the increase in the number of mitochondria and lysosomes, but inhibited by serum in the medium. Our findings deepen the understanding of the interaction between nanoplastics and intestinal cells, which is helpful for the risk assessment of nanoplastics.
Collapse
Affiliation(s)
- Yuan-Yuan Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Jie Liu
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Yuan Guo
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Qiangqiang Zhang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Aoneng Cao
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China
| | - Haifang Wang
- Institute of Nanochemistry and Nanobiology, Shanghai University, Shanghai 200444, China.
| |
Collapse
|
17
|
Tang K, Tang Z, Niu M, Kuang Z, Xue W, Wang X, Liu X, Yu Y, Jeong S, Ma Y, Wu A, Kim BYS, Jiang W, Yang Z, Li C. Allosteric targeted drug delivery for enhanced blood-brain barrier penetration via mimicking transmembrane domain interactions. Nat Commun 2025; 16:3410. [PMID: 40210849 PMCID: PMC11986143 DOI: 10.1038/s41467-025-58746-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 04/01/2025] [Indexed: 04/12/2025] Open
Abstract
Current strategies for active targeting in the brain are entirely based on the effective interaction of the ligand with the orthosteric sites of specific receptors on the blood-brain barrier (BBB), which is highly susceptible to various pathophysiological factors and limits the efficacy of drug delivery. Here, we propose an allosteric targeted drug delivery strategy that targets classical BBB transmembrane receptors by designing peptide ligands that specifically bind to their transmembrane domains. This strategy prevents competitive interference from endogenous ligands and antibodies by using the insulin receptor and integrin αv as model targets, respectively, and can effectively overcome pseudotargets or target loss caused by shedding or mutating the extracellular domain of target receptors. Moreover, these ligands can be spontaneously embedded in the phospholipid layer of lipid carriers using a plug-and-play approach without chemical modification, with excellent tunability and immunocompatibility. Overall, this allosteric targeted drug delivery strategy can be applied to multiple receptor targets and drug carriers and offers promising therapeutic benefits in brain diseases.
Collapse
Affiliation(s)
- Kaicheng Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China
- School of Pharmacy, Sichuan Industrial Institute of Antibiotics, Chengdu University, Chengdu, China
| | - Zhongjie Tang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Miaomiao Niu
- School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Nanjing, China
| | - Zuyin Kuang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Weiwei Xue
- School of Pharmaceutical Sciences, Chongqing University, Chongqing, China
| | - Xinyu Wang
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Xinlong Liu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Yang Yu
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China
| | - Seongdong Jeong
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yifan Ma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Annette Wu
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Zhaogang Yang
- School of Life Sciences, Jilin University, Changchun, China.
| | - Chong Li
- Medical Research Institute, College of Pharmaceutical Sciences, Southwest University, Chongqing, China.
- Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou, China.
| |
Collapse
|
18
|
Feng S, Xu Q, Liu B, He Y, Song L, Zhao Q, Wang S. Modulating Intracellular Autophagy and Macropinocytosis for Increased Neighboring Drug Delivery. ACS NANO 2025; 19:13175-13190. [PMID: 40162609 DOI: 10.1021/acsnano.4c18465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Neighboring effects provided a valuable direction for in-depth penetration of nanoparticles into tumors. However, the uncontrollable drug transcytosis and limited drug uptake hindered by viscous cancer-associated fibroblasts (CAFs) greatly limit their in-depth penetration. Here, we proposed and demonstrated that intracellular autophagosomes could carry the remaining drugs to neighboring cells, and the enhanced macropinocytosis played a major role in neighboring delivery. To enhance the autophagosome-based neighboring delivery, Ca2+-doped polydopamine was prepared to load GLS1 inhibitor CB-839 and modified glutamine (839/CG) for triggering macropinocytosis-based active cells uptake. After Ca2+-release caused lysosome damage, 839/CG escaped from lysosomes and hindered the autophagosome maturation. Then, Ca2+-induced endoplasmic reticulum oscillations and glutamine starvation both increased and blocked autophagy flow, causing 839/CG-contained autophagosome accumulation. Meanwhile, the tumor increased its macropinocytosis in response to mTOR downregulation-induced glutamine hunger, causing "the more you eat, the hungrier you get". After tumor death, the 839/CG-contained autophagosomes were released and actively ingested by neighboring hungry tumor cells through macropinocytosis. Combined with the photothermal effect triggered CAF decrease, neighboring cells repeated the above process for in-depth tumor delivery. Also, immunogenic death enhanced the antigen presentation of DCs and infiltration of T cells, thereby inhibiting tumor growth and lung metastasis.
Collapse
Affiliation(s)
- Shuaipeng Feng
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Qingqing Xu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Bin Liu
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Ye He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Luming Song
- Department of Microbial and Biochemical Pharmacy, School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Qinfu Zhao
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| | - Siling Wang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning 110016, PR China
| |
Collapse
|
19
|
Pareja Tello R, Lamparelli EP, Ciardulli MC, Hirvonen J, Barreto G, Mafulli N, Della Porta G, Santos HA. Hybrid lipid nanoparticles derived from human mesenchymal stem cell extracellular vesicles by microfluidic sonication for collagen I mRNA delivery to human tendon progenitor stem cells. Biomater Sci 2025; 13:2066-2081. [PMID: 40033856 DOI: 10.1039/d4bm01405g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Tendon degeneration remains an intricate pathological process characterized by the coexistence of multiple dysregulated homeostasis processes, including the increase in collagen III production in comparison with collagen I. Mesenchymal stem cell-derived extracellular vesicles (MSC-EVs) remain a promising therapeutic tool thanks to their pro-regenerative properties and applicability as drug delivery systems, despite their drug loading limitations. Herein, we developed MSC-EV-derived hybrid lipid nanoparticles (MSC-Hyb NPs) using a microfluidic-sonication technique as an alternative platform for the delivery of collagen type I (COL 1A1) mRNA into pathological TSPCs. The MSC-Hyb NPs produced had LNP-like physicochemical characteristics and were 178.6 nm in size with a PDI value of 0.245. Moreover, MSC-Hyb NPs encapsulated mRNA and included EV-derived surface proteins such as CD63, CD81 and CD144. MSC-Hyb NPs remained highly biocompatible with TSPCs and proved to be functional mRNA delivery agents with certain limitations in comparison with lipid nanoparticles (LNPs). In vitro efficacy studies on TSPCs showed a 2-fold increase in procollagen type I carboxy-terminal peptide production comparable with the effect caused by LNPs. Therefore, our work provides an alternative production method for MSC-EV-derived hybrid NPs and supports their potential use as drug delivery systems for tendon regeneration.
Collapse
Affiliation(s)
- Rubén Pareja Tello
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, University of Helsinki, Helsinki FI-00014, Finland.
- Department of Medicine, Surgery and Dentistry, University of Salerno, via S. Allende, 84081 Baronissi, SA, Italy.
| | - Erwin Pavel Lamparelli
- Department of Medicine, Surgery and Dentistry, University of Salerno, via S. Allende, 84081 Baronissi, SA, Italy.
| | - Maria Camilla Ciardulli
- Department of Medicine, Surgery and Dentistry, University of Salerno, via S. Allende, 84081 Baronissi, SA, Italy.
| | - Jouni Hirvonen
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, University of Helsinki, Helsinki FI-00014, Finland.
| | - Goncalo Barreto
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, 00014 Helsinki, Finland
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, 02150 Espoo, Finland
- Orton Orthopedic Hospital, Tenholantie 10, 00280 Helsinki, Finland
| | - Nicola Mafulli
- Department of Trauma and Orthopaedics, Faculty of Medicine and Psychology, Sant' Andrea Hospital, Sapienza University, 00189 Rome, Italy
| | - Giovanna Della Porta
- Department of Medicine, Surgery and Dentistry, University of Salerno, via S. Allende, 84081 Baronissi, SA, Italy.
- Interdepartment Centre BIONAM, University of Salerno, Via Giovanni Paolo II, 84084 Fisciano, SA, Italy
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, University of Helsinki, Helsinki FI-00014, Finland.
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen (UMCG), University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands.
| |
Collapse
|
20
|
Liu H, Yong T, Zhang X, Wei Z, Bie N, Xu S, Zhang X, Li S, Zhang J, Zhou P, Yang X, Gan L. Spatial Regulation of Cancer-Associated Fibroblasts and Tumor Cells via pH-Responsive Bispecific Antibody Delivery for Enhanced Chemo-Immunotherapy Synergy. ACS NANO 2025; 19:11756-11773. [PMID: 40114589 DOI: 10.1021/acsnano.4c13277] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
The effectiveness of chemotherapy is often compromised by physiological barriers and an immunosuppressive tumor microenvironment. Cancer-associated fibroblasts (CAFs) significantly contribute to the reconfiguration of the tumor extracellular matrix (ECM) and the suppression of immune responses, making them crucial targets for therapeutic intervention. Here, a tumor acidic microenvironment-responsive delivery system that utilizes tumor cell-derived microparticles (MPs) as carriers for the chemotherapeutic agent doxorubicin (DOX) and the bispecific antibody YM101 targeting both TGF-β and PD-L1 is developed (DOX@MPs-YM101) to spatially regulate both CAFs and tumor cells for enhanced chemotherapeutic efficacy. DOX@MPs-YM101 efficiently targets tumor tissues and releases DOX@MPs and YM101 in response to the acidic tumor microenvironment. YM101 reprograms CAFs and reduces the tumor ECM, facilitating tumor accumulation and deep penetration of DOX@MPs-YM101. DOX@MPs are highly internalized into tumor cells, triggering immunogenic cell death (ICD) and activating CD8+ T cell-mediated antitumor immunity. The reprogramming of CAFs by YM101 further promotes the accumulation of CD8+ T cells and reduces the number of immunosuppressive cells within the tumors. Additionally, YM101 effectively neutralizes PD-L1 on tumor cells induced by DOX@MPs, restoring CD8+ T cell activity and generating long-term antitumor immune memory to prevent tumor recurrence. Our findings highlight the potential of DOX@MPs-YM101 to improve chemotherapy in cancer treatment.
Collapse
Affiliation(s)
- Haojie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaoqiong Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaojuan Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Jing Zhang
- Wuhan YZY Biopharma Co., Ltd., Wuhan 430074, China
| | - Pengfei Zhou
- Wuhan YZY Biopharma Co., Ltd., Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
21
|
Li M, Liu Y, Liu F, Chen Q, Xu L, Cheng Z, Tan Y, Liu Z. Extracellular Vesicle-Based Antitumor Nanomedicines. Adv Healthc Mater 2025; 14:e2403903. [PMID: 39935134 DOI: 10.1002/adhm.202403903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/30/2024] [Indexed: 02/13/2025]
Abstract
Extracellular vesicles (EVs) have emerged as promising bioactive carriers for delivering therapeutic agents, including nucleic acids, proteins, and small-molecule drugs, owing to their excellent physicochemical stability and biocompatibility. However, comprehensive reviews on the various types of EV-based nanomedicines for cancer therapy remain scarce. This review explores the potential of EVs as antitumor nanomedicines. Methods for EV extraction, drug loading, and engineering modifications are systematically examined, and the strengths and limitations of these technical approaches are critically assessed. Additionally, key strategies for developing EV-based antitumor therapies are highlighted. Finally, the opportunities and challenges associated with advancing EVs toward clinical translation are discussed. With the integration of multiple disciplines, robust EV-based therapeutic platforms are expected to be manufactured to provide more personalized and effective solutions for oncology patients.
Collapse
Affiliation(s)
- Mingfeng Li
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yanfei Liu
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Fei Liu
- Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Qiwen Chen
- Department of Pharmaceutical Engineering, College of Chemistry and Chemical Engineering, Central South University, Changsha, Hunan, 410083, P. R. China
| | - Lishang Xu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhongyu Cheng
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Yifu Tan
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
| | - Zhenbao Liu
- Department of Pharmaceutics, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, 410013, P. R. China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, 410008, P. R. China
| |
Collapse
|
22
|
Firouzjaei AA, Mohammadi-Yeganeh S. Advancements in Targeted Therapies for Colorectal Cancer: Innovative Drug Formulation and Delivery Strategies. Arch Pharm (Weinheim) 2025; 358:e202400969. [PMID: 40259467 DOI: 10.1002/ardp.202400969] [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: 12/23/2024] [Revised: 03/08/2025] [Accepted: 03/12/2025] [Indexed: 04/23/2025]
Abstract
Colorectal cancer (CRC) is a leading cause of cancer-related mortality globally, with increasing incidence presenting significant treatment challenges. Traditional nontargeted therapies often result in high toxicity and limited efficacy, underscoring the need for improved treatment modalities. This review highlights recent advancements in drug delivery systems to enhance therapeutic outcomes for CRC. We examine innovative strategies, including computer-assisted pharmaceutical formulation, sustained-release matrices, and prodrugs, as well as targeted delivery mechanisms such as exosomes, liposomes, hydrogels, antibody-drug conjugates, and stimuli-responsive systems. These methodologies offer improved drug biodistribution, enhanced targeting of cancer cells, and reduced off-target effects, promising better clinical outcomes. Additionally, we discuss the development of novel formulations designed to optimize the delivery of therapeutic agents in advanced CRC. Ongoing clinical trials investigating these innovative systems signify a shift toward more effective patient treatment options. While challenges remain in the clinical application of these targeted therapies, continued research offers promising avenues for improving patient outcomes in CRC. This study aims to inform future strategies for managing this aggressive disease, ultimately enhancing survival rates and quality of life for affected individuals.
Collapse
Affiliation(s)
- Ali Ahmadizad Firouzjaei
- Bioinformatics Research Center, Basic Sciences Research Institute, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Samira Mohammadi-Yeganeh
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
23
|
Rahimkhoei V, Akbari A, Jassim AY, Hussein UAR, Salavati-Niasari M. Recent advances in targeting cancer stem cells by using nanomaterials. Int J Pharm 2025; 673:125381. [PMID: 39988213 DOI: 10.1016/j.ijpharm.2025.125381] [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: 12/25/2024] [Revised: 02/12/2025] [Accepted: 02/19/2025] [Indexed: 02/25/2025]
Abstract
Cancer stem cells (CSCs) are a special group of cells that start, regenerate, and maintain the growth of tumors. Cancer stem cells (CSCs) contribute to the dissemination of tumors, their recurrence following treatment, and the mechanisms by which cancers develop resistance to therapies. CSCs reside in a unique microenvironment influenced by a variety of factors from their immediate surroundings. These factors include low oxygen levels, too much new blood vessel growth, a shift in how cells use energy from breathing oxygen to breaking down glucose, and an increase in certain markers and signals related to stem cells that help remove drugs from the body. Antibodies and special molecules that focus on the unique features keeping the environment stable are used to deliver cancer treatments to CSCs. As a result, nanoparticles are extremely effective in delivering drugs that combat cancer directly to cancer stem cells. Right now, stem cell nanotechnology is a new and interesting area of study. Some experiments on how stem cells interact with tiny structures or materials have shown good results. The importance of tiny structures and materials in creating treatments using stem cells for diseases and injuries has been clearly understood. The way nanomaterials are built and their characteristics influence how stem cells grow and change. This area of study is a new and exciting field where material science meets medicine. This review talks about the biology of CSCs and new ways to create nanoparticles (NPs) that can deliver cancer drugs specifically to these CSCs. This review talks about the creation of different types of tiny particles, including synthetic and natural polymer particles, lipid particles, inorganic particles, protein particles that can assemble themselves, combined antibody-drug particles, and small bubbles called nanovesicles, all aimed at targeting cancer stem cells. This paper talks about recent progress and opinions on using nanotechnology in stem cell research and therapy. It also covers how nanoparticles can help track, control, and improve the retention of stem cells.
Collapse
Affiliation(s)
- Vahid Rahimkhoei
- Institute of Nano Science and Nano Technology, University of Kashan, Kashan 87317-51167, Islamic Republic of Iran
| | - Ali Akbari
- Solid Tumor Research Center, Cellular and Molecular Medicine Research Institute, Urmia University of Medical Sciences, Urmia, Iran.
| | - Amar Yasser Jassim
- Department of Marine Vertebrate, Marine Science Center, University of Basrah, Iraq
| | | | - Masoud Salavati-Niasari
- Institute of Nano Science and Nano Technology, University of Kashan, Kashan 87317-51167, Islamic Republic of Iran.
| |
Collapse
|
24
|
Cheng M, Chai Y, Rong G, Xin C, Gu L, Zhou X, Hong J. Nanotechnology-based strategies for vaccine development: accelerating innovation and delivery. BIOMATERIALS TRANSLATIONAL 2025; 6:55-72. [PMID: 40313573 PMCID: PMC12041807 DOI: 10.12336/biomatertransl.2025.01.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 07/08/2024] [Accepted: 12/03/2024] [Indexed: 05/03/2025]
Abstract
The key role and impact of nanotechnology in vaccine development became particularly prominent following the outbreak of the coronavirus disease 2019 (COVID-19) pandemic in 2019. Especially in the process of designing and optimising COVID-19 vaccines, the application of nanomaterials significantly accelerated vaccine development and efficient delivery. In this review, we categorised and evaluated conventional vaccines, including attenuated live vaccines, inactivated vaccines, and subunit vaccines, highlighting their advantages and limitations. We summarised the development history, mechanisms, and latest technologies of vaccine adjuvants, emphasising their critical role in immune responses. Furthermore, we focused on the application of nanotechnology in the vaccine field, detailing the characteristics of nanoparticle vaccines, including virus-like particles, lipid-based carriers, inorganic nanoparticles, and polymer-based carriers. We emphasised their potential advantages in enhancing vaccine stability and immunogenicity, as well as their ability to deliver vaccines and present antigens through various routes. Despite facing challenges such as low drug loading efficiency, issues with long-term storage, high costs, and difficulties in large-scale production, nano-vaccines hold promise for the future. This review underscores the pivotal role and prospects of nanotechnology in vaccine development, offering new pathways and strategies to address current and future disease challenges.
Collapse
Affiliation(s)
- Mingrui Cheng
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Gene Editing and Cell Therapy Key Lab for Rare Disease; Shanghai Engineering Research Center of Synthetic Immunology, Shanghai, China
| | - Yawei Chai
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Gene Editing and Cell Therapy Key Lab for Rare Disease; Shanghai Engineering Research Center of Synthetic Immunology, Shanghai, China
| | - Guangyu Rong
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Gene Editing and Cell Therapy Key Lab for Rare Disease; Shanghai Engineering Research Center of Synthetic Immunology, Shanghai, China
| | - Changchang Xin
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Gene Editing and Cell Therapy Key Lab for Rare Disease; Shanghai Engineering Research Center of Synthetic Immunology, Shanghai, China
| | - Lei Gu
- Epigenetics Laboratory, Max Planck Institute for Heart and Lung Research & Cardiopulmonary Institute (CPI), Bad Nauheim, Germany
| | - Xujiao Zhou
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Gene Editing and Cell Therapy Key Lab for Rare Disease; Shanghai Engineering Research Center of Synthetic Immunology, Shanghai, China
| | - Jiaxu Hong
- Department of Ophthalmology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
- NHC Key Laboratory of Myopia and Related Eye Diseases, Shanghai, China
- Shanghai Gene Editing and Cell Therapy Key Lab for Rare Disease; Shanghai Engineering Research Center of Synthetic Immunology, Shanghai, China
| |
Collapse
|
25
|
Zhou Q, Gao J, Wu G, Wang C, Yang Y, Huang T, Wang Y, Yue T, Gao Z, Xie H, Xiong F, Xiang K, Yong T, Zhang W, Zhang T, Kong W, Chen C, Zhang S, Yu Q, Fan X, Liu S, Liu Y, Wang CY. Adipose progenitor cell-derived extracellular vesicles suppress macrophage M1 program to alleviate midlife obesity. Nat Commun 2025; 16:2743. [PMID: 40113754 PMCID: PMC11926339 DOI: 10.1038/s41467-025-57444-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 02/19/2025] [Indexed: 03/22/2025] Open
Abstract
Among different age groups, middle-aged individuals are particularly susceptible to obesity, with a 22% higher risk of all-cause mortality. However, the underlying mechanisms remain unclear. In this study, we identify adipose progenitor cells (APCs) in the white adipose tissue (WAT) of middle-aged subjects as potential causes of midlife obesity. Specifically, the extracellular vesicles (EVs) derived from APCs display an impaired ability to mitigate the inflammaging of adipose tissue macrophages (ATMs) in middle-aged individuals. Mechanistically, these EVs, lacking miR-145-5p, fail to suppress the expression of L-selectin in ATMs, thereby facilitating their M1 program via the NF-κB signaling pathway. In contrast, EVs from young APCs effectively inhibit M1 macrophage polarization. Accordingly, targeted liposomes are designed to deliver miR-145-5p mimics to ATMs, which effectively prevent the obesity in middle-aged mice. Collectively, our findings highlight the role of APC-derived EVs in midlife obesity and propose miR-145-5pas a promising therapeutic target for clinical applications.
Collapse
Affiliation(s)
- Qing Zhou
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia Gao
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Guorao Wu
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chenwei Wang
- Lester and Sue Smith Breast Center, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Yan Yang
- Department of Endocrinology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Teng Huang
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yi Wang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tiantian Yue
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhichao Gao
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xie
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Xiong
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ke Xiang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, China
| | - Wanguang Zhang
- Department of Hepatic Surgery, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Tongtong Zhang
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Chengdu, China
| | - Wen Kong
- Department of Endocrinology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Cai Chen
- Department of Endocrinology, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shu Zhang
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qilin Yu
- Department of Respiratory and Critical Care Medicine, the Center for Biomedical Research, NHC Key Laboratory of Respiratory Diseases, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xuemei Fan
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China
| | - Shiwei Liu
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China.
| | - Yanjun Liu
- Center of Gastrointestinal and Minimally Invasive Surgery, Department of General Surgery, The Third People's Hospital of Chengdu, Chengdu, China.
| | - Cong-Yi Wang
- Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, the Key Laboratory of Endocrine and Metabolic Diseases of Shanxi Province, Taiyuan, China.
- The Center for Biomedical Research, Tongji Hospital Research Building, Tongji Hospital, Tongji Medical College, Huazhong University of Sciences and Technology, Wuhan, China.
| |
Collapse
|
26
|
Chen H, Law J, Wang Y, Chen Z, Du X, Fang K, Wang Z, Duan F, Sun Y, Yu J. Active microgel particle swarms for intrabronchial targeted delivery. SCIENCE ADVANCES 2025; 11:eadr3356. [PMID: 40073130 PMCID: PMC11900871 DOI: 10.1126/sciadv.adr3356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Accepted: 02/05/2025] [Indexed: 03/14/2025]
Abstract
Intrabronchial delivery of therapeutic agents is critical to the treatment of respiratory diseases. Targeted delivery is demanded because of the off-target accumulation of drugs in normal lung tissues caused by inhalation and the limited motion dexterity of clinical bronchoscopes in tortuous bronchial trees. Herein, we developed microrobotic swarms consisting of magnetic hydrogel microparticles to achieve intrabronchial targeted delivery. Under programmed magnetic fields, the microgel particle swarms performed controllable locomotion and adaptative structure reconfiguration in tortuous and air-filled environments. The swarms were further integrated with imaging contrast agents for precise tracking under x-ray fluoroscopy and computed tomography imaging. Magnetic navigation of the swarms in an ex vivo lung phantom and in vivo delivery into deep branches of the bronchial trees were achieved. The on-demand reconfiguration of swarms for avoiding the microgel particles from entering nontarget bronchi and the precise delivery into tilted bronchi through climbing motion were validated.
Collapse
Affiliation(s)
- Hui Chen
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
| | - Junhui Law
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
- Institute of Robotics and Intelligent Systems, Dalian University of Technology, Dalian, China
| | - Yibin Wang
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
| | - Ziheng Chen
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
- School of Mechatronics Engineering and Automation, Shanghai University, Shanghai, China
| | - Xingzhou Du
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
| | - Kaiwen Fang
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
| | - Zhe Wang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Feng Duan
- Department of Interventional Radiology, Chinese PLA General Hospital First Medical Center, Beijing, China
| | - Yu Sun
- Department of Mechanical and Industrial Engineering, University of Toronto, Toronto, Canada
- Institute of Biomedical Engineering, University of Toronto, Toronto, Canada
- Robotics Institute, University of Toronto, Toronto, Canada
| | - Jiangfan Yu
- School of Science and Engineering, Chinese University of Hong Kong, Shenzhen, China
- Shenzhen Institute of Artificial Intelligence and Robotics for Society, Shenzhen, China
| |
Collapse
|
27
|
Jia X, Wang E, Wang J. Rational Design of Nanozymes for Engineered Cascade Catalytic Cancer Therapy. Chem Rev 2025; 125:2908-2952. [PMID: 39869790 DOI: 10.1021/acs.chemrev.4c00882] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2025]
Abstract
Nanozymes have shown significant potential in cancer catalytic therapy by strategically catalyzing tumor-associated substances and metabolites into toxic reactive oxygen species (ROS) in situ, thereby inducing oxidative stress and promoting cancer cell death. However, within the complex tumor microenvironment (TME), the rational design of nanozymes and factors like activity, reaction substrates, and the TME itself significantly influence the efficiency of ROS generation. To address these limitations, recent research has focused on exploring the factors that affect activity and developing nanozyme-based cascade catalytic systems, which can trigger two or more cascade catalytic processes within tumors, thereby producing more therapeutic substances and achieving efficient and stable cancer therapy with minimal side effects. This area has shown remarkable progress. This Perspective provides a comprehensive overview of nanozymes, covering their classification and fundamentals. The regulation of nanozyme activity and efficient strategies of rational design are discussed in detail. Furthermore, representative paradigms for the successful construction of cascade catalytic systems for cancer treatment are summarized with a focus on revealing the underlying catalytic mechanisms. Finally, we address the current challenges and future prospects for the development of nanozyme-based cascade catalytic systems in biomedical applications.
Collapse
Affiliation(s)
- Xiuna Jia
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Erkang Wang
- State Key Laboratory of Electroanalytical Chemistry, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, Jilin 130022, P. R. China
| | - Jin Wang
- Center for Theoretical Interdisciplinary Sciences Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang 325001, P. R. China
- Department of Chemistry and Physics, State University of New York at Stony Brook, Stony Brook, New York 11794-3400, United States
| |
Collapse
|
28
|
Ning S, Shangguan P, Zhu X, Ou X, Wang K, Suo M, Shen H, Lu X, Wei X, Zhang T, Chen X, Tang BZ. Pyridinium Rotor Strategy toward a Robust Photothermal Agent for STING Activation and Multimodal Image-Guided Immunotherapy for Triple-Negative Breast Cancer. J Am Chem Soc 2025; 147:7433-7444. [PMID: 39977833 PMCID: PMC11887044 DOI: 10.1021/jacs.4c15534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 02/08/2025] [Accepted: 02/10/2025] [Indexed: 02/22/2025]
Abstract
The immunosuppressive tumor microenvironment in triple-negative breast cancer could hinder the response to thorough immunotherapy and diminish the antitumor efficacy. Although the STING pathway emerges as a promising target to remedy defects, uncertain drug delivery might lead to off-target inflammatory reactions. Here, we manifest a novel phototheranostic agent with an aggregation-induced emission property that guided the pharmacological activation of a STING agonist for photothermal immunotherapy to create an immunologically "hot" tumor. A pyridinium rotor strategy is proposed to develop a positively charged TBTP-Bz, which is stably coincorporated with a STING agonist MSA-2 into thermal-responsive exosome-liposome hybrid nanoparticles for tumor-targeting delivery. TBTP-Bz exhibits aggregation-enhanced NIR-II emission and a photoacoustic signal, accomplishing real-time tumor tracking. Its photothermal stimulation induces immunogenic cancer cell death and promotes the precise release of MSA-2, thus boosting STING activation and STING-mediated type I interferon production. Significantly, single-dose photoimmunotherapy effectively suppresses abscopal tumor growth and provokes an immune memory effect to inhibit postsurgical recurrent and rechallenged tumors. This demonstrates promising clinical potential for poorly immunogenic breast cancer.
Collapse
Affiliation(s)
- Shipeng Ning
- Department
of Breast Surgery, The Second Affiliated
Hospital of Guangxi Medical University, Nanning 530000, China
| | - Ping Shangguan
- Guangzhou
Institute of Cancer Research, the Affiliated Cancer Hospital, School
of Biomedical Engineering, Guangzhou Medical
University, Guangdong 511436, China
| | - Xinyan Zhu
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Xinwen Ou
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Kaiyuan Wang
- Department
of Pharmaceutics, Wuya College of Innovation, Shenyang Pharmaceutical University, Shenyang, Liaoning 110016, P. R. China
- Departments
of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering,
and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty
of Engineering, National University of Singapore, Singapore 119074, Singapore
| | - Meng Suo
- Guangzhou
Institute of Cancer Research, the Affiliated Cancer Hospital, School
of Biomedical Engineering, Guangzhou Medical
University, Guangdong 511436, China
| | - Hanchen Shen
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
| | - Xiuxin Lu
- Department
of Breast Surgery, The Second Affiliated
Hospital of Guangxi Medical University, Nanning 530000, China
| | - Xianqing Wei
- Department
of Breast Surgery, The Second Affiliated
Hospital of Guangxi Medical University, Nanning 530000, China
| | - Tianfu Zhang
- Guangzhou
Institute of Cancer Research, the Affiliated Cancer Hospital, School
of Biomedical Engineering, Guangzhou Medical
University, Guangdong 511436, China
| | - Xiaoyuan Chen
- Departments
of Diagnostic Radiology, Surgery, Chemical and Biomolecular Engineering,
and Biomedical Engineering, Yong Loo Lin School of Medicine and Faculty
of Engineering, National University of Singapore, Singapore 119074, Singapore
- Clinical
Imaging Research Centre, Centre for Translational Medicine, Yong Loo
Lin School of Medicine, National University
of Singapore, Singapore 117599, Singapore
- Nanomedicine
Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore
- Institute
of Molecular and Cell Biology, Agency for Science, Technology, and
Research (A*STAR), 61
Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Ben Zhong Tang
- Department
of Chemistry, Hong Kong Branch of Chinese National Engineering Research
Center for Tissue Restoration and Reconstruction, Division of Life
Science, and State Key Laboratory of Molecular Neuroscience, The Hong Kong University of Science and Technology, Kowloon, Hong Kong, China
- School
of Science and Engineering, Shenzhen Institute of Aggregate Science
and Technology, The Chinese University of
Hong Kong, Shenzhen (CUHK-Shenzhen), Guangdong 518172, China
| |
Collapse
|
29
|
Solovicová V, Ďatková A, Bertók T, Kasák P, Vikartovská A, Lorencová L, Tkac J. Advances in magnetic affinity-based isolation/detection of exosomes for robust diagnostics. Mikrochim Acta 2025; 192:206. [PMID: 40042696 PMCID: PMC11882713 DOI: 10.1007/s00604-025-07048-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/13/2025] [Indexed: 03/09/2025]
Abstract
The review article provides a short introduction to exosomes with the focus to use exosomes as disease markers itself (i.e. their concentration or presence of some specific receptors) or a source of disease biomarkers such as proteins and metabolites. In detail, we are discussing various methods of exosome isolation and the main focus of the review paper is on affinity capture of exosomes, since some of them can be applied to the isolation of specific sub-populations of exosomes produced by some specific organs. The article provides a comprehensive overview of magnetic (bio)affinity capture applied to the detection of exosomes or exosomal cargo using different (bio)affinity capture ligands such as antibodies, DNA aptamers, peptides, glycan-based recognition, transferrin-based approaches, affinity based on recognition of phospholipids of exosomes and other approaches including electrostatic interactions. The review in detail provides key analytical and clinical parameters of such approaches in a form of an extensive table summarising outcomes published in the last two years (2023-2024). Finally, the review paper also provides conclusions sections discussing pros and cons of magnetic (bio)affinity capture for exosome isolation and/or determination of exosomal content.
Collapse
Affiliation(s)
- Veronika Solovicová
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 5807/9, 845 38, Bratislava, Slovak Republic
| | - Anna Ďatková
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 5807/9, 845 38, Bratislava, Slovak Republic
| | - Tomáš Bertók
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 5807/9, 845 38, Bratislava, Slovak Republic
| | - Peter Kasák
- Center for Advanced Materials, Qatar University, P.O. Box 2713, Doha, Qatar
| | - Alica Vikartovská
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 5807/9, 845 38, Bratislava, Slovak Republic
| | - Lenka Lorencová
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 5807/9, 845 38, Bratislava, Slovak Republic
| | - Jan Tkac
- Institute of Chemistry, Slovak Academy of Sciences, Dúbravská cesta 5807/9, 845 38, Bratislava, Slovak Republic.
| |
Collapse
|
30
|
Pang L, Tian C, Wang Q, Zhao Z, Pan B, Luo Z, Wu S, Li X, Fan J. An Integrating Microfluidic System for Concentration Gradient Generation of Exosomes and Exosome-Assisted Single-Cell-Derived Tumor-Sphere Formation. ACS Sens 2025; 10:678-688. [PMID: 39866075 DOI: 10.1021/acssensors.4c01542] [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: 01/28/2025]
Abstract
To enhance exploration on tumor stem-like cells (TSCs) without altering their cellular biological characteristics, researchers advocate for application of single-cell-derived tumor-spheres (STSs). TSCs are regulated by their surrounding microenvironment, making it crucial to simulate a tumor microenvironment to facilitate STS formation. Recently, exosomes that originated from the tumor microenvironment have emerged as a promising approach for mimicking the tumor microenvironment. In the tumor microenvironment, various associated cells (such as fibroblasts, endothelial cells, and immune cells) play crucial roles. Utilizing exosomes derived from these cells enabled us to simulate the tumor microenvironment and promote STS formation. Herein, we have developed an integrated microfluidic platform to generate serial concentration gradients and evaluate the effects of multiple exosomes on STS formation. To demonstrate the feasibility of our approach, we generated serial concentration gradients of exosomes derived from two different cell types (HUVEC and NIH/3T3 cells) and assessed their effects on STS formation. Subsequently, we investigated the drug resistance of STSs treated with free doxorubicin and doxorubicin-loaded poly(lactic-co-glycolic acid) (PLGA) nanoparticles. Our findings revealed that the serial concentration gradients of mixed exosomes could be successfully generated, leading to an enhanced formation rate and size of STSs. Compared to exosomes derived from one cell type, the mixed exosomes exhibited superior promotion of STS formation. Additionally, nanomedicines demonstrated a reduction in the drug resistance of TSCs compared to free drug treatment, particularly in smaller and/or more deformable TSCs. This platform provides an innovative approach for STS formation enhancement and tumor microenvironment simulation.
Collapse
Affiliation(s)
- Long Pang
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| | - Chang Tian
- Public Health School, Anhui University of Science & Technology, Huainan, Anhui 232001, China
| | - Qirui Wang
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Zhaohua Zhao
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Bofeng Pan
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Zichun Luo
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Shuqiang Wu
- Oncology Department of Xi'an Fengcheng Hospital, Xi'an 710021, China
| | - Xueping Li
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
| | - Jianglin Fan
- School of Basic Medical Science, Xi'an Key Laboratory of Pathogenic Microorganism and Tumor Immunity, Xi'an Medical University, Xi'an 710021, China
- School of Biotechnology and Health Sciences, Wuyi University, Jiangmen 529020, China
| |
Collapse
|
31
|
Xia W, Tan Y, Liu Y, Xie N, Zhu H. Prospect of extracellular vesicles in tumor immunotherapy. Front Immunol 2025; 16:1525052. [PMID: 40078996 PMCID: PMC11897508 DOI: 10.3389/fimmu.2025.1525052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 01/28/2025] [Indexed: 03/14/2025] Open
Abstract
Extracellular vesicles (EVs), as cell-derived small vesicles, facilitate intercellular communication within the tumor microenvironment (TME) by transporting biomolecules. EVs from different sources have varied contents, demonstrating differentiated functions that can either promote or inhibit cancer progression. Thus, regulating the formation, secretion, and intake of EVs becomes a new strategy for cancer intervention. Advancements in EV isolation techniques have spurred interest in EV-based therapies, particularly for tumor immunotherapy. This review explores the multifaceted functions of EVs from various sources in tumor immunotherapy, highlighting their potential in cancer vaccines and adoptive cell therapy. Furthermore, we explore the potential of EVs as nanoparticle delivery systems in tumor immunotherapy. Finally, we discuss the current state of EVs in clinical settings and future directions, aiming to provide crucial information to advance the development and clinical application of EVs for cancer treatment.
Collapse
Affiliation(s)
- Wenbo Xia
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yunhan Tan
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Yongen Liu
- State Key Laboratory of Oral Diseases and National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, China
| | - Na Xie
- West China School of Basic Medical Sciences and Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu, China
| | - Huili Zhu
- Department of Reproductive Medicine, Key Laboratory of Birth Defects and Related Diseases of Women and Children of Ministry of Education, West China Second University Hospital of Sichuan University, Chengdu, China
| |
Collapse
|
32
|
Liu J, Srivastava S, Li T, Moujane F, Lee JY, Chen Y, Liu H, Deng SX, Xie YH. On the Feasibility of SERS-Based Monitoring of Drug Loading Efficiency in Exosomes for Targeted Delivery. BIOSENSORS 2025; 15:141. [PMID: 40136938 PMCID: PMC11939968 DOI: 10.3390/bios15030141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/18/2025] [Accepted: 02/19/2025] [Indexed: 03/27/2025]
Abstract
Cancer, a significant cause of mortality, necessitates improved drug delivery strategies. Exosomes, as natural drug carriers, offer a more efficient, targeted, and less toxic drug delivery system compared to direct dispersal methods via ingestion or injection. To be successfully implemented as drug carriers, efficient loading of drugs into exosomes is crucial, and a deeper understanding of the loading mechanism remains to be solved. This study introduces surface-enhanced Raman scattering (SERS) to monitor drug loading efficacy at the single vesicle level. By enhancing the Raman signal, SERS overcomes limitations in Raman spectroscopy. A gold nanopyramids array-based SERS substrate assesses exosome heterogeneity in drug-loading capabilities with the help of single-layer graphene for precise quantification. This research advances targeted drug delivery by presenting a more efficient method of evaluating drug-loading efficiency into individual exosomes through SERS-based monitoring. Furthermore, the study explores leveraging osmotic pressure variations, enhancing the efficiency of drug loading into exosomes.
Collapse
Affiliation(s)
- Jun Liu
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Siddharth Srivastava
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Tieyi Li
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Faycal Moujane
- Cornea Division, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - John Y. Lee
- Cornea Division, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Yiqing Chen
- Department of Bioengineering, University of California Riverside, Riverside, CA 92521, USA
| | - Huinan Liu
- Department of Bioengineering, University of California Riverside, Riverside, CA 92521, USA
| | - Sophie X. Deng
- Cornea Division, Stein Eye Institute, University of California Los Angeles, Los Angeles, CA 90095, USA
| | - Ya-Hong Xie
- Department of Materials Science and Engineering, University of California Los Angeles, Los Angeles, CA 90095, USA
| |
Collapse
|
33
|
Wang J, Jin N, Xie Z, Cheng Q, Jiang B, Shuai Y, Xu Z, Wan Q, Chen Y, Mao C, Yang M. Gold nanorods coated with self-assembled silk fibroin for improving their biocompatibility and facilitating targeted photothermal-photodynamic cancer therapy. NANOSCALE 2025; 17:4624-4635. [PMID: 39811885 DOI: 10.1039/d4nr03641g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/16/2025]
Abstract
Gold nanorods (AuNRs) have shown great potential as photothermal agents for cancer therapy. However, the biosafety of AuNRs ordinarily synthesized using a cationic ligand assistance procedure has always been a subject of controversy, which limits their application in tumor therapy. In this study, we propose a novel strategy to enhance the biocompatibility of AuNRs by constructing a biological coating derived from silk fibroin (SF) on their surface. The SF coating could be easily and precisely manipulated using a layer-by-layer (LBL) assembly method. The resulting SF-coated gold nanorods (AuNRs@SF) exhibited reduced cytotoxicity and improved hemocompatibility compared to untreated AuNRs. Moreover, the treated nanorods were easily modified with a tumor-targeting peptide (AuNRs@MTSF) and efficiently loaded indocyanine green (ICG). Both in vitro and in vivo analyses demonstrated that AuNRs@MTSF could more effectively reach tumor tissue and enter MCF-7 cells. Furthermore, after loading ICG, AuNRs@MTSF exhibited superior antitumor efficacy compared to other groups by combining photodynamic therapy (PDT) with photothermal therapy (PTT) under near-infrared (NIR) irradiation without inducing any side effects. This work suggests that SF coating of gold nanorods is a potential approach for improving their biocompatibility, and that function-modified AuNRs@SF are effective nanoplatforms for targeted and multimodal tumor therapy.
Collapse
Affiliation(s)
- Jie Wang
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Na Jin
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Zheyu Xie
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Qichao Cheng
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Bo Jiang
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Yajun Shuai
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Zongpu Xu
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Quan Wan
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Yuyin Chen
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| | - Mingying Yang
- Zhejiang Provincial Key Laboratory of Utilization and Innovation of Silkworm and Bee Resources, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Yuhangtang Road 866, Hangzhou, 310058 Zhejiang, P. R. China
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong SAR, P. R. China
| |
Collapse
|
34
|
Luo X, McAndrews KM, Kalluri R. Natural and Bioengineered Extracellular Vesicles in Diagnosis, Monitoring and Treatment of Cancer. ACS NANO 2025; 19:5871-5896. [PMID: 39869032 PMCID: PMC12002402 DOI: 10.1021/acsnano.4c11630] [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] [Indexed: 01/28/2025]
Abstract
Extracellular vesicles (EVs) are cell derived nanovesicles which are implicated in both physiological and pathological intercellular communication, including the initiation, progression, and metastasis of cancer. The exchange of biomolecules between stromal cells and cancer cells via EVs can provide a window to monitor cancer development in real time for better diagnostic and interventional strategies. In addition, the process of secretion and internalization of EVs by stromal and cancer cells in the tumor microenvironment (TME) can be exploited for delivering therapeutics. EVs have the potential to provide a targeted, biocompatible, and efficient delivery platform for the treatment of cancer and other diseases. Natural as well as engineered EVs as nanomedicine have immense potential for disease intervention. Here, we provide an overview of current knowledge of EVs' function in cancer progression, diagnostic and therapeutic applications for EVs in the cancer setting, as well as current EV engineering strategies.
Collapse
Affiliation(s)
- Xin Luo
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
| | - Kathleen M. McAndrews
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
| | - Raghu Kalluri
- Department of Cancer Biology, University of Texas MD Anderson Cancer Center, Houston, Texas 77054, United States
- Department of Bioengineering, Rice University, Houston, Texas 77005, United States
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas 77030, United States
| |
Collapse
|
35
|
Sych T, Görgens A, Steiner L, Gucluler G, Huge Y, Alamdari F, Johansson M, Aljabery F, Sherif A, Gabrielsson S, El Andaloussi S, Sezgin E. Imaging Single Particle Profiler to Study Nanoscale Bioparticles Using Conventional Confocal Microscopy. NANO LETTERS 2025; 25:2173-2180. [PMID: 39878336 PMCID: PMC11827106 DOI: 10.1021/acs.nanolett.4c05117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 01/31/2025]
Abstract
Single particle profiling (SPP) is a unique methodology to study nanoscale bioparticles such as liposomes, lipid nanoparticles, extracellular vesicles, and lipoproteins in a single particle and high throughput manner. The initial version requires the single photon counting modules for data acquisition, which limits its adoptability. Here, we present imaging-based SPP (iSPP) that can be performed by imaging a spot over time in the common imaging mode with confocal detectors. We also provide user-friendly software with a graphical user interface to analyze such data and give quantitative insights on the content and properties of nanoscale bioparticles. We use iSPP to decipher lipid-protein interactions, membrane modifications by drugs, and the heterogeneity of extracellular vesicles isolated from cell lines and human urine. This easily applicable modality of the single particle profiler will facilitate nanoscale bioparticle research in laboratories with access to any confocal microscope.
Collapse
Affiliation(s)
- Taras Sych
- Science
for Life Laboratory, Department of Women’s and Children’s
Health, Karolinska Institutet, Tomtebodavägen 23, 17165 Solna, Sweden
| | - André Görgens
- Division
of Biomolecular and Cellular Medicine, Department of Laboratory Medicine,
Karolinska ATMP Center, Karolinska Institutet, 14152 Huddinge, Sweden
- Department
of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, 14152 Huddinge, Sweden
| | - Loïc Steiner
- Division
of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, 17164 405 Stockholm, Sweden
- Department
of Clinical Immunology and Transfusion Medicine, Center for Molecular
Medicine, Karolinska University Hospital, 17164 Stockholm, Sweden
| | - Gozde Gucluler
- Division
of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, 17164 405 Stockholm, Sweden
- Department
of Clinical Immunology and Transfusion Medicine, Center for Molecular
Medicine, Karolinska University Hospital, 17164 Stockholm, Sweden
| | - Ylva Huge
- Department
of Urology in Östergötland and Department of Biomedical
and Clinical Sciences, Linköping
University, 58225 Linköping, Sweden
| | - Farhood Alamdari
- Department
of Urology, Vastmanland Hospital, 72189 Västerås, Sweden
| | - Markus Johansson
- Departement
of Surgery and Urology, County Hospital
of Sundsvall-Härnösand, 85643 Sundsvall, Sweden
| | - Firas Aljabery
- Department
of Urology in Östergötland, and Department of Biomedical
and Clinical Sciences, Linköping
University, 58225 Linköping, Sweden
| | - Amir Sherif
- Department
of Diagnostics and Intervention, Umeå
University, 90187 Umeå, Sweden
| | - Susanne Gabrielsson
- Division
of Immunology and Respiratory Medicine, Department of Medicine Solna, Karolinska Institutet, 17164 405 Stockholm, Sweden
- Department
of Clinical Immunology and Transfusion Medicine, Center for Molecular
Medicine, Karolinska University Hospital, 17164 Stockholm, Sweden
| | - Samir El Andaloussi
- Division
of Biomolecular and Cellular Medicine, Department of Laboratory Medicine,
Karolinska ATMP Center, Karolinska Institutet, 14152 Huddinge, Sweden
- Department
of Cellular Therapy and Allogeneic Stem Cell Transplantation (CAST), Karolinska University Hospital, 14152 Huddinge, Sweden
| | - Erdinc Sezgin
- Science
for Life Laboratory, Department of Women’s and Children’s
Health, Karolinska Institutet, Tomtebodavägen 23, 17165 Solna, Sweden
| |
Collapse
|
36
|
Jia L, Wang W, Zhao H, Ding X, Zheng M, Cai D, Wang Y, Wang Z, Liu H. Innovative Nano Delivery Systems for Astaxanthin: Enhancing Stability, Bioavailability, and Targeted Therapeutic Applications. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:3286-3304. [PMID: 39886831 DOI: 10.1021/acs.jafc.4c09415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/01/2025]
Abstract
Astaxanthin (AST), as a natural antioxidant, has broad application prospects in medicine and health products. However, its highly unsaturated structure and significant lipophilic characteristics limit its dispersibility and bioavailability, thereby restricting its application in food, medicines, and nutraceuticals. To overcome these limitations, researchers have proposed the use of nano delivery systems. This review summarizes various nanocarriers, including liposomes, nanostructured lipid carriers, nanoparticles, and others, and analyzes their advantages in enhancing the solubility, stability, and bioavailability of AST. Furthermore, the study focuses on targeted delivery systems achieved through biomolecular modifications, which enable precise delivery of AST to specific cells or tissues, enhancing therapeutic effects. Additionally, smart-responsive delivery systems, such as pH-responsive and light-sensitive systems, are also discussed, showing their immense potential in precise release and targeted therapy. These findings provide new perspectives for the precise nutrition and clinical applications of AST. Future research should further optimize the design of nanocarriers to enable broader applications.
Collapse
Affiliation(s)
- Lei Jia
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
- National Engineering Research Center for Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Wei Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
- National Engineering Research Center for Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Hongyu Zhao
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Xiaoyu Ding
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
- National Engineering Research Center for Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Mingzhu Zheng
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
- National Engineering Research Center for Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Dan Cai
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
- National Engineering Research Center for Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Yuhua Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Zhitong Wang
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
| | - Huimin Liu
- College of Food Science and Engineering, Jilin Agricultural University, Changchun, Jilin 130118, China
- National Engineering Research Center for Wheat and Corn Deep Processing, Jilin Agricultural University, Changchun, Jilin 130118, China
| |
Collapse
|
37
|
Zhang W, Jiang Y, Liu L, Shen H, Huang X, Zheng W, Chu Z, Wang W, Guo Y, Qian H. Implantable Microneedles Loaded with Nanoparticles Surface Engineered Escherichia coli for Efficient Eradication of Triple-Negative Breast Cancer Stem Cells. NANO LETTERS 2025; 25:2041-2051. [PMID: 39836986 DOI: 10.1021/acs.nanolett.4c06052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Eliminating cancer stem cells (CSCs) is essential for the effective treatment of triple-negative breast cancer (TNBC). This study synthesized Au@cerium-zinc composite core@shell nanoparticles (Au@Zn/CeO) that were subsequently conjugated with Escherichia coli (E. coli) to create the engineered bacterium AZCE, which was then combined with microneedle carriers and freeze-dried to obtain AZCE-MN. Upon implantation into TNBC tumors, the inherent properties of E. coli facilitate AZCE to penetrate the extracellular matrix and break through the basement membrane, enabling effective delivery of AZC to CSCs-enriched regions deep within the tumor. The released Zn2+ induces mitochondrial dysfunction and amplifies reactive oxygen species (ROS) production. The redox cycling between Ce3+/Ce4+ effectively depleted glutathione, which further increased ROS generation. Under near-infrared laser irradiation, Au nanorods initiated photothermal therapy, effectively ablating CSCs while amplifying catalytic reactions and ionic effects. This microneedle-mediated engineered bacteria delivery improved nanodrug penetration in tumor tissues, providing new insights for TNBC clinical treatment.
Collapse
Affiliation(s)
- Weinan Zhang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Yechun Jiang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Litao Liu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Hui Shen
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Xianyu Huang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Wang Zheng
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Zhaoyou Chu
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Wanni Wang
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
| | - Yanchuan Guo
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
- Key Laboratory of Photochemical Conversion and Optoelectronic Materials, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing 100190, P. R. China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, P. R. China
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei 230601, P. R. China
| |
Collapse
|
38
|
Wang TY, Hu HG, Zhao L, Zhuo SH, Su JY, Feng GH, Li YM. EXO TLR1/2-STING: A Dual-Mechanism Stimulator of Interferon Genes Activator for Cancer Immunotherapy. ACS NANO 2025; 19:5017-5028. [PMID: 39846950 DOI: 10.1021/acsnano.4c18056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
As natural agonists of the stimulator of interferon genes (STING) protein, cyclic dinucleotides (CDNs) can activate the STING pathway, leading to the expression of type I interferons and various cytokines. Efficient activation of the STING pathway in antigen-presenting cells (APCs) and tumor cells is crucial for antitumor immune response. Tumor-derived exosomes can be effectively internalized by APCs and tumor cells and have excellent potential to deliver CDNs to the cytoplasm of APCs and tumor cells. Here, we leverage tumor exosomes as a delivery platform, designing an EXOTLR1/2-STING loaded with CDNs. To achieve efficient loading of CDNs onto exosomes, we chemically conjugated CDNs with Pam3CSK4, a compound featuring multiple fatty acid chains, resulting in Pam3CSK4-CDGSF. Utilizing the high lipophilicity of Pam3CSK4, Pam3CSK4-CDGSF could be efficiently loaded onto the exosomes through simple incubation. Moreover, as an agonist for Toll-like receptor 1/2, Pam3CSK4 also exhibits robust immunological synergistic effects in conjunction with CDNs. EXOTLR1/2-STING effectively induced the activation of APCs and triggered tumor cell death, producing a favorable antitumor therapeutic effect. It also demonstrated significant synergistic effects with immune checkpoint therapies.
Collapse
Affiliation(s)
- Tian-Yang Wang
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Hong-Guo Hu
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Lang Zhao
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Shao-Hua Zhuo
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Jing-Yun Su
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Geng-Hui Feng
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
| | - Yan-Mei Li
- Key Lab of Bioorganic Phosphorus Chemistry & Chemical Biology, Department of Chemistry, Tsinghua University, Beijing 100084, China
- Beijing Institute for Brain Disorders, Beijing 100069, China
- Center for Synthetic and Systems Biology, Tsinghua University, Beijing 100084, China
| |
Collapse
|
39
|
Bie N, Li S, Liang Q, Zheng W, Xu S, Liu H, Zhang X, Wei Z, Yong T, Yang X, Gan L. Tumor-Repopulating Cell-Derived Microparticle-Based Therapeutics Amplify the Antitumor Effect through Synergistic Inhibition of Chemoresistance and Immune Evasion. Mol Pharm 2025; 22:733-746. [PMID: 39772575 DOI: 10.1021/acs.molpharmaceut.4c00709] [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: 01/11/2025]
Abstract
Traditional chemotherapy often encounters failure attributed to drug resistance mediated by tumor-repopulating cells (TRCs) and chemotherapy-triggered immune suppression. The effective inhibition of TRCs and the mitigation of drug-induced immune suppression are pivotal for the successful chemotherapy. Here, TRC-derived microparticles (3D-MPs), characterized by excellent tumor-targeting and high TRC uptake properties, are utilized to deliver metformin and the chemotherapeutic drug doxorubicin ((DOX+Met)@3D-MPs). (DOX+Met)@3D-MPs efficiently enhance tumor accumulation and are highly internalized in tumor cells and TRCs. Additionally, (DOX+Met)@3D-MPs significantly decrease the chemotherapy-triggered upregulation in P-glycoprotein expression to enhance intracellular doxorubicin retention, resulting in increased chemotherapy sensitivity and immunogenic cell death in tumor cells and TRCs for improved antitumor immunity. Importantly, (DOX+Met)@3D-MPs also remarkably reduce chemotherapy-induced PD-L1 expression, efficiently alleviating immune suppression facilitated by the PD-L1/PD-1 axis to further enhance immunological response against malignancy. These results underscore the (DOX+Met)@3D-MPs' potential as a viable platform for augmenting the efficacy of antitumor therapies.
Collapse
Affiliation(s)
- Nana Bie
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyu Li
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Qingle Liang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Wenxia Zheng
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Shiyi Xu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Haojie Liu
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiaojuan Zhang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Zhaohan Wei
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Tuying Yong
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Xiangliang Yang
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Lu Gan
- National Engineering Research Center for Nanomedicine, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Key Laboratory of Molecular Biophysics of the Ministry of Education, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
40
|
Sancho-Albero M, Decio A, Akpinar R, De Luigi A, Giavazzi R, Terracciano LM, De Cola L. Melanoma extracellular vesicles membrane coated nanoparticles as targeted delivery carriers for tumor and lungs. Mater Today Bio 2025; 30:101433. [PMID: 39866783 PMCID: PMC11764275 DOI: 10.1016/j.mtbio.2024.101433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/17/2024] [Accepted: 12/27/2024] [Indexed: 01/28/2025] Open
Abstract
Targeting is the most challenging problem to solve for drug delivery systems. Despite the use of targeting units such as antibodies, peptides and proteins to increase their penetration in tumors the amount of therapeutics that reach the target is very small, even with the use of nanoparticles (NPs). Nature has solved the selectivity problem using a combination of proteins and lipids that are exposed on the cell membranes and are able to recognize specific tissues as demonstrated by cancer metastasis. Extracellular vesicles (EVs) have a similar ability in target only certain organs or to return to their original cells, showing home behavior. Here we report a strategy inspired by nature, using a combination of NPs and the targeting cell membranes of EVs. We implement the EV membranes, extracted by the EVs produced by melanoma B16-BL6 cells, as a coating of organosilica porous particles with the aim of targeting tumors and lung metastasis, while avoiding systemic effects and accumulation of the NPs in undesired organs. The tissue-specific fingerprint provided by the EVs-derived membranes from melanoma cells provides preferential uptake into the tumor and selective targeting of lungs. The ability of the EVs hybrid systems to behave as the natural EVs was demonstrated in vitro and in vivo in two different tumor models. As a proof of concept, the loading and release of doxorubicin, was investigated and its accumulation demonstrated in the expected tissues.
Collapse
Affiliation(s)
- María Sancho-Albero
- Department of Biochemistry and Molecular Pharmacology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
| | - Alessandra Decio
- Department of Oncology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
| | - Reha Akpinar
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
| | - Ada De Luigi
- Department of Biochemistry and Molecular Pharmacology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
| | - Raffaella Giavazzi
- Department of Oncology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
| | - Luigi M. Terracciano
- IRCCS Humanitas Research Hospital, via Manzoni 56, Rozzano, 20089, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, Pieve Emanuele, 20072, Milan, Italy
| | - Luisa De Cola
- Department of Biochemistry and Molecular Pharmacology. Istituto di Ricerche Farmacologiche Mario Negri, IRCCS, Via Mario Negri, 2, Milan, Italy
- Department of Pharmaceutical Science, DISFARM. Università degli Studi di Milano, Milan, 20133, Italy
| |
Collapse
|
41
|
Song Y, Kong H, Oh S, Kim SB. Plant-derived extracellular vesicles as nanocarriers for combination therapy enhancing paclitaxel-based regimens in breast cancer. BMB Rep 2025; 58:53-63. [PMID: 39978779 PMCID: PMC11875746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/26/2024] [Accepted: 01/15/2025] [Indexed: 02/22/2025] Open
Abstract
Breast cancer remains a leading cause of morbidity and mortality worldwide. Triple-negative breast cancer (TNBC) presents unique challenges owing to its aggressiveness and limited treatment options. Paclitaxel-based chemotherapy is widely used in breast cancer treatment. However, its efficacy is often limited by toxicity, multidrug resistance, and lack of targeted delivery. In response to these challenges, recent studies have focused on the use of extracellular vesicles (EVs), particularly plant-derived EVs, as innovative drug delivery systems capable of enhancing therapeutic outcomes and reducing adverse effects. Plant-derived EVs offer significant advantages owing to their biocompatibility, low immunogenicity, and scalability. They provide a natural platform for delivering chemotherapeutics such as paclitaxel and doxorubicin directly to tumor cells. This review explores the therapeutic potential of plant-derived EVs in breast cancer treatment, focusing on TNBC by examining their ability to improve drug stability, bioavailability, and selective targeting of cancer cells. Key studies on EVs derived from plants such as grapefruit, ginger, and tea leaves have demonstrated their capacity to deliver chemotherapeutic agents effectively while mitigating common side effects associated with conventional delivery methods. Although the use of plantderived EVs is still in early stages of research, findings suggest that that these nanocarriers can serve as transformative tools in oncology, providing a versatile and efficient platform for precise cancer treatment. This review highlights current landscape of research on plant-derived EVs, their application in breast cancer therapy, and future directions required to translate these findings into clinical practice. [BMB Reports 2025; 58(2): 53-63].
Collapse
Affiliation(s)
- Youngcheon Song
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| | - Hyunseok Kong
- Department of Animal Science, Sahmyook University, Seoul 01795, Korea
| | - Soohwan Oh
- College of Pharmacy, Korea University, Sejong 30019, Korea
| | - Sang Bum Kim
- College of Pharmacy, Sahmyook University, Seoul 01795, Korea
| |
Collapse
|
42
|
Chen Z, Yun X, Tian J, Li F, Zhang Z, Meng J, Li N, Bian H, Duan S, Zhang L. Engineering Macrophage-Derived Exosome to Deliver Pirfenidone: A Novel Approach to Combat Silicotic Pulmonary Fibrosis. Adv Healthc Mater 2025; 14:e2403227. [PMID: 39382242 DOI: 10.1002/adhm.202403227] [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: 08/27/2024] [Revised: 09/19/2024] [Indexed: 10/10/2024]
Abstract
Silicosis is a severe lung disease characterized by diffuse pulmonary fibrosis, for which there is currently no effective treatment. Pirfenidone (PFD) shows great antifibrotic potential but is clinically hindered by low bioavailability and gastrointestinal side effects. To address these limitations, this study develops a PFD delivery system (PFD-Exo) using J774A.1 macrophage-derived exosomes. Firstly, PFD is loaded via sonication, then PFD-Exo is characterized using Raman spectral imaging and UV absorption spectroscopy. Finally, in vitro and in vivo silicosis models are established to evaluate its antifibrotic effects. Results show that PFD-Exo outperforms free PFD in inhibiting TGF-β1-induced transdifferentiation of primary lung fibroblasts in vitro. In a mouse model of silicosis, PFD-Exo is found to be accumulated in the lungs following intratracheal administration and significantly ameliorates pulmonary inflammation and fibrosis while minimizing gastrointestinal side effects. Mechanistic studies reveal that PFD-Exo modulates the TGF-β signaling pathway by downregulating SMAD3 and upregulating SMAD7 and NOGGIN. In conclusion, this study provides the first evidence of macrophage-derived exosomes as an effective PFD delivery system for silicosis treatment and offers a promising strategy for other refractory pulmonary diseases.
Collapse
Affiliation(s)
- Zhen Chen
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250001, China
- Jinan (Preparatory) Key Laboratory of Women's Diseases and Fertility Preservation, Jinan, 250001, China
| | - Xiang Yun
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China
| | - Jiaqi Tian
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250001, China
- Jinan (Preparatory) Key Laboratory of Women's Diseases and Fertility Preservation, Jinan, 250001, China
| | - Fei Li
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China
| | - Zitong Zhang
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250001, China
- Jinan (Preparatory) Key Laboratory of Women's Diseases and Fertility Preservation, Jinan, 250001, China
- School of Public Health, Qingdao University, Qingdao, 266071, China
| | - Jiahua Meng
- School of Public Health, North China University of Science and Technology, Tangshan, 063000, China
| | - Ning Li
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250001, China
- Jinan (Preparatory) Key Laboratory of Women's Diseases and Fertility Preservation, Jinan, 250001, China
| | - Hongying Bian
- National Institute of Occupational Health and Poison Control, Chinese Center for Disease Control and Prevention, Beijing, 102206, China
| | - Shuyin Duan
- School of Public Health, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, 250001, China
| | - Lin Zhang
- Clinical Medical Research Center for Women and Children Diseases, Key Laboratory of Birth Regulation and Control Technology of National Health Commission of China, Shandong Provincial Maternal and Child Health Care Hospital Affiliated to Qingdao University, Jinan, 250001, China
- Jinan (Preparatory) Key Laboratory of Women's Diseases and Fertility Preservation, Jinan, 250001, China
| |
Collapse
|
43
|
Wen X, Hao Z, Yin H, Min J, Wang X, Sun S, Ruan G. Engineered Extracellular Vesicles as a New Class of Nanomedicine. CHEM & BIO ENGINEERING 2025; 2:3-22. [PMID: 39975802 PMCID: PMC11835263 DOI: 10.1021/cbe.4c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Revised: 10/19/2024] [Accepted: 10/20/2024] [Indexed: 02/21/2025]
Abstract
Extracellular vesicles (EVs) are secreted from biological cells and contain many molecules with diagnostic values or therapeutic functions. There has been great interest in academic and industrial communities to utilize EVs as tools for diagnosis or therapeutics. In addition, EVs can also serve as delivery vehicles for therapeutic molecules. An indicator of the enormous interest in EVs is the large number of review articles published on EVs, with the focus ranging from their biology to their applications. An emerging trend in EV research is to produce and utilize "engineered EVs", which are essentially the enhanced version of EVs. EV engineering can be conducted by cell culture condition control, genetic engineering, or chemical engineering. Given their nanometer-scale sizes and therapeutic potentials, engineered EVs are an emerging class of nanomedicines. So far, an overwhelming majority of the research on engineered EVs is preclinical studies; there are only a very small number of reported clinical trials. This Review focuses on engineered EVs, with a more specific focus being their applications in therapeutics. The various approaches to producing engineered EVs and their applications in various diseases are reviewed. Furthermore, in vivo imaging of EVs, the mechanistic understandings, and the clinical translation aspects are discussed. The discussion is primarily on preclinical studies while briefly mentioning the clinical trials. With continued interdisciplinary research efforts from biologists, pharmacists, physicians, bioengineers, and chemical engineers, engineered EVs could become a powerful solution for many major diseases such as neurological, immunological, and cardiovascular diseases.
Collapse
Affiliation(s)
- Xiaowei Wen
- Institute
of Analytical Chemistry and Instrument for Life Science, The Key Laboratory
of Biomedical Information Engineering of Ministry of Education, School
of Life Science and Technology, Xi’an
Jiaotong University, Xi’an, China 710049
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Zerun Hao
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Haofan Yin
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Jie Min
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Xueying Wang
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Sihan Sun
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| | - Gang Ruan
- Wisdom
Lake Academy of Pharmacy, Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Jiangsu
Province Higher Education Key Laboratory of Cell Therapy Nanoformulation
(Construction), Xi’an Jiaotong-Liverpool
University, Suzhou, China 215123
- Xi’an
Jiaotong-Liverpool University & University of Liverpool Joint
Center of Pharmacology and Therapeutics, Suzhou, China 215123
| |
Collapse
|
44
|
Paranandi KS, Amar-Lewis E, Mirkin CA, Artzi N. Nomadic Nanomedicines: Medicines Enabled by the Paracrine Transfer Effect. ACS NANO 2025; 19:21-30. [PMID: 39746105 DOI: 10.1021/acsnano.4c15052] [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] [Indexed: 01/04/2025]
Abstract
In nanomedicine, the cellular export of nanomaterials has been less explored than uptake. Traditionally viewed in a negative light, recent findings highlight the potential of nanomedicine export to enhance therapeutic effects. This Perspective examines key pathways for export and how nanomaterial design affects removal rates. We present the idea of the "paracrine transfer effect" (PTE), where nanomaterials are first internalized by a "waypoint" cell and then exported to a "destination" cell, influencing both in potentially exploitable ways. Essential characteristics for nanomedicines to leverage the PTE are discussed, along with two case studies: STING-stimulating polymeric nanoparticles and TLR9-stimulating liposomal spherical nucleic acids. We propose future research directions to better understand and utilize the PTE in developing more effective nanomedicines.
Collapse
Affiliation(s)
- Krishna S Paranandi
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Medical Scientist Training Program, Northwestern University Feinberg School of Medicine, Chicago, Illinois 60611, United States
| | - Eliz Amar-Lewis
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| | - Chad A Mirkin
- International Institute for Nanotechnology, Northwestern University, Evanston, Illinois 60208, United States
- Department of Chemistry, Northwestern University, Evanston, Illinois 60208, United States
| | - Natalie Artzi
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts 02139, United States
- Department of Medicine, Division of Engineering in Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, Massachusetts 02215, United States
| |
Collapse
|
45
|
Shao X, Zhao X, Wang B, Fan J, Wang J, An H. Tumor microenvironment targeted nano-drug delivery systems for multidrug resistant tumor therapy. Theranostics 2025; 15:1689-1714. [PMID: 39897552 PMCID: PMC11780529 DOI: 10.7150/thno.103636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/10/2024] [Indexed: 02/04/2025] Open
Abstract
In recent years, nano-drug delivery systems (Nano-DDS) that target the tumor microenvironment (TME) to overcome multidrug resistance (MDR) have become a research hotspot in the field of cancer therapy. By precisely targeting the TME and regulating its unique pathological features, such as hypoxia, weakly acidic pH, and abnormally expressed proteins, etc., these Nano-DDS enable effective delivery of therapeutic agents and reversal of MDR. This scientific research community is increasing its investment in the development of diversified systems and exploring their anti-drug resistance potential. Therefore, it is particularly important to conduct a comprehensive review of the research progress of TME-targeted Nano-DDS in recent years. After a brief introduction of TME and tumor MDR, the design principle and structure of liposomes, polymer micelles and inorganic nanocarriers are focused on, and their characteristics as TME-targeted nanocarriers are described. It also demonstrates how these systems break through the cancer MDR treatment through various targeting mechanisms, discusses their synthetic innovation, research results and resistance overcoming mechanisms. The review was concluded with deliberations on the key challenges and future outlooks of targeting TME Nano-DDS in cancer therapy.
Collapse
Affiliation(s)
| | | | | | | | - Jinping Wang
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, PR China
| | - Hailong An
- Key Laboratory of Molecular Biophysics of Hebei Province, Institute of Biophysics, School of Health Sciences and Biomedical Engineering, Hebei University of Technology, 300401, Tianjin, PR China
| |
Collapse
|
46
|
Ma Y, Dong S, Grippin AJ, Teng L, Lee AS, Kim BYS, Jiang W. Engineering therapeutical extracellular vesicles for clinical translation. Trends Biotechnol 2025; 43:61-82. [PMID: 39227240 PMCID: PMC11717644 DOI: 10.1016/j.tibtech.2024.08.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 09/05/2024]
Abstract
Cell-based therapies are revolutionizing medicine by replacing or modifying dysfunctional cells with healthy cells or engineered derivatives, offering disease reversal and cure. One promising approach is using cell-derived extracellular vesicles (EVs), which offer therapeutic benefits similar to cell transplants without the biosafety risks. Although EV applications face challenges like limited production, inadequate therapeutic loading, and poor targeting efficiency, recent advances in bioengineering have enhanced their effectiveness. Herein, we summarize technological breakthroughs in EV bioengineering over the past 5 years, highlighting their improved therapeutic functionalities and potential clinical prospects. We also discuss biomanufacturing processes, regulation, and safety considerations for bioengineered EV therapies, emphasizing the significance of establishing robust frameworks to ensure translation capability, safety, and therapeutic effectiveness for successful clinical adoption.
Collapse
Affiliation(s)
- Yifan Ma
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiyan Dong
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Adam J Grippin
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lesheng Teng
- School of Life Sciences, Jilin University, Changchun, China
| | - Andrew S Lee
- Peking University Shenzhen Graduate School, Shenzhen, China; Institute of Cancer Research, Shenzhen Bay Laboratory, Shenzhen, China
| | - Betty Y S Kim
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Wen Jiang
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
47
|
Zhang Y, Sun X, Guan Y, Sun Y. Exosome-Derived Cargos in Immune Microenvironment in Esophageal Carcinoma: A Mini-Review. Recent Pat Anticancer Drug Discov 2025; 20:137-144. [PMID: 38173209 DOI: 10.2174/0115748928280161231123060159] [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: 09/29/2023] [Revised: 11/09/2023] [Accepted: 11/16/2023] [Indexed: 01/05/2024]
Abstract
Esophageal carcinoma, a lethal malignancy with limited treatment options and poor prognosis, necessitates understanding its underlying mechanisms and identifying novel therapeutic targets. Recent studies have highlighted the critical role of the immune microenvironment in esophageal carcinoma, particularly the interplay between tumor cells and immune cells mediated by exosomes and their cargos. Exosomes, small extracellular vesicles secreted by various cells, including tumor cells, facilitate intercellular communication by transferring bioactive molecules such as proteins, nucleic acids, and lipids to recipient cells. In the context of esophageal carcinoma, tumor-derived exosomes have been shown to play a significant role in shaping the immune microenvironment. In esophageal carcinoma, exosomal cargos have been found to modulate immune cell function and impact tumor progression. These cargos can carry immune inhibitory molecules, such as programmed death-ligand 1 (PD-L1), to suppress T-cell activity and promote immune evasion by tumor cells. Furthermore, exosomal cargos can activate antigen- presenting cells, enhancing their ability to present tumor-specific antigens to T cells and thereby promoting anti-tumor immune responses. Additionally, the cargos of exosomes have been implicated in the induction of immune regulatory T cells (Tregs) and myeloid-derived suppressor cells (MDSCs) within the esophageal carcinoma microenvironment. These immunosuppressive effectors inhibit the activity of T cells, contributing to tumor immune evasion and resistance to immune therapies. In summary, exosomes and their cargo play a crucial role in the immune microenvironment of esophageal carcinoma. Understanding the mechanisms by which exosomal cargos regulate immune cell function and tumor progression may reveal novel therapeutic targets for this devastating disease.
Collapse
Affiliation(s)
- Yakun Zhang
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao Municipal Hospital, Shandong University, Qingdao, Shandong, 266012, China
| | - Xiaoyan Sun
- Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, 266012, China
| | - Yan Guan
- Shandong First Medical University & Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 250117, Jinan, PR China
| | - Ying Sun
- Department of Oncology, Qingdao Hospital, University of Health and Rehabilitation Sciences (Qingdao Municipal Hospital), Qingdao, Shandong, 266012, China
| |
Collapse
|
48
|
Zhu H, Huang D, Nie M, Zhao Y, Sun L. Dexamethasone loaded DNA scavenger nanogel for systemic lupus erythematosus treatment. Bioact Mater 2025; 43:330-339. [PMID: 40115883 PMCID: PMC11923376 DOI: 10.1016/j.bioactmat.2024.08.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 08/09/2024] [Accepted: 08/27/2024] [Indexed: 03/23/2025] Open
Abstract
Lupus nephritis (LN) poses a severe risk for individuals with systemic lupus erythematosus (SLE), prompting extensive research into targeted delivery systems capable of modulating immune responses and clearing cell-free DNA (cfDNA). Here, we propose a novel renal homing nanogel that acts as a cfDNA scavenger and a dexamethasone (DXM) delivery carrier for LN treatment. Based on the generation 3 polylysine dendrimers, the created cationic nanogels (G3DSP) exhibit minimal toxicity and outstanding DXM loading efficiency. Our studies confirm that these nanogels can competitively bind with anionic cfDNA in vitro, leading to the suppression of toll-like receptor 9 (TLR9) activation. When administered systemically to MRL/lpr mice, the nanogels preferentially localize to and are retained in the inflamed kidneys, releasing their payload in response to reactive oxygen species (ROS), therefore effectively ameliorating SLE symptoms. Consequently, G3DSP nanogels emerge as a promising effective combined therapy for LN, minimizing cfDNA accumulation in vital organs and delivering immunomodulatory benefits through DXM.
Collapse
Affiliation(s)
- Haofang Zhu
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Danqing Huang
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Min Nie
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| | - Lingyun Sun
- Department of Rheumatology and Immunology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
- Department of Rheumatology and Immunology, Institute of Translational Medicine, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
49
|
Namdari M, McDonnell FS. Extracellular vesicles as emerging players in glaucoma: Mechanisms, biomarkers, and therapeutic targets. Vision Res 2025; 226:108522. [PMID: 39581065 PMCID: PMC11640964 DOI: 10.1016/j.visres.2024.108522] [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: 07/01/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/26/2024]
Abstract
In recent years, extracellular vesicles (EVs) have attracted significant scientific interest due to their widespread distribution, their potential as disease biomarkers, and their promising applications in therapy. Encapsulated by lipid bilayers these nanovesicles include small extracellular vesicles (sEV) (30-150 nm), microvesicles (100-1000 nm), and apoptotic bodies (100-5000 nm) and are essential for cellular communication, immune responses, biomolecular transport, and physiological regulation. As they reflect the condition and functionality of their originating cells, EVs play critical roles in numerous physiological processes and diseases. Therefore, EVs offer valuable opportunities for uncovering disease mechanisms, enhancing drug delivery systems, and identifying novel biomarkers. In the context of glaucoma, a leading cause of irreversible blindness, the specific roles of EVs are still largely unexplored. This review examines the emerging role of EVs in the pathogenesis of glaucoma, with a focus on their potential as diagnostic biomarkers and therapeutic agents. Through a thorough analysis of current literature, we summarize key advancements in EV research and identify areas where further investigation is needed to fully understand their function in glaucoma.
Collapse
Affiliation(s)
- Maral Namdari
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA
| | - Fiona S McDonnell
- John A. Moran Eye Center, University of Utah, Salt Lake City, UT, USA; Biomedical Engineering, University of Utah, Salt Lake City, UT, USA; Pharmacology and Toxicology, University of Utah Salt Lake City, UT, USA.
| |
Collapse
|
50
|
Liang QL, Liu H, Wang T, Lau CH, Wang J, Mo ZY, Zhou ZM, Zhou ZY, Zhu H, Chen G, Tong S. UV radiation enhanced encapsulation of superparamagnetic iron oxide nanoparticles (MNPs) in microparticles derived from tumor repopulating cells. Biochem Biophys Res Commun 2024; 741:151050. [PMID: 39586131 DOI: 10.1016/j.bbrc.2024.151050] [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: 09/18/2024] [Revised: 11/06/2024] [Accepted: 11/21/2024] [Indexed: 11/27/2024]
Abstract
Extracellular vesicles (EVs) such as microparticles secreted by the cells can be manipulated and used for delivering therapeutic drugs to target and eradicate cancer cells. However, high encapsulation efficiency and mass production of the microparticles remain difficult to achieve. Efficient and targeted delivery to cancer cells is another hurdle to be addressed. To overcome these issues, we integrated superparamagnetic iron oxide nanoparticles (MNPs) with microparticles. First of all, exposure of highly aggressive tumor-repopulating cells (TRC) to UV radiation dramatically improved microparticle production. These TRC cells were selected from diverse cancer cell lines that are 3D culturing in soft fibrin gel. These microparticles derived from 3D-cultured TRCs have lower membrane stiffness than 2D-cultured cells. Ferrozine assay showed that endocytosis and encapsulation of MNPs during microparticle production were higher in 3D-cultured TRC cells than in 2D cultured cells. Packaging of MNPs into microparticles also enhanced cellular uptake of MNPs without inducing cytotoxicity to treated cells. Compared to the naked MNPs, ex vivo fluorescence imaging shows that mice tail-vein injected with microparticle-encapsulated MNPs displayed continuous increments of intratumoral accumulation of MNPs. Furthermore, MRI images revealed a higher T2 contrast and an uneven distribution of the T2 contrast in the tumor of mice tail-vein injected with microparticle-encapsulated MNPs than naked MNPs. This study provides a new platform for cancer imaging by integrating MNPs and microparticles derived from tumor-repopulating cells.
Collapse
Affiliation(s)
- Qing-Le Liang
- Department of Clinical Laboratory Medicine, Chongqing University Jiangjin Hospital, Chongqing, China
| | - He Liu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Tao Wang
- Department of Biology, College of Science, Shantou University, Shantou, Guangdong, China
| | - Cia-Hin Lau
- Department of Biology, College of Science, Shantou University, Shantou, Guangdong, China
| | - Jianchao Wang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Zheng-Ying Mo
- Department of Oncology, Tai-He Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Zhang-Ming Zhou
- Department of Neurosurgery, Dujiangyan Medical Center, Chengdu, China
| | - Zhe-Yu Zhou
- Department of Radiology, Tai-He Hospital, Hubei University of Medicine, Shiyan, Hubei, China
| | - Haibao Zhu
- Department of Biology, College of Science, Shantou University, Shantou, Guangdong, China; Guangdong Provincial Key Laboratory of Marine Biotechnology, Shantou University, Shantou, Guangdong, China; Shantou Key Laboratory of Marine Microbial Resources and Interactions with Environment, Shantou University, Shantou, Guangdong, China.
| | - Gang Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China.
| | - Sheng Tong
- Department of Biomedical Engineering, University of Kentucky, USA.
| |
Collapse
|