1
|
Zhang B, Zhang C, Chen C, Hong R, Shen Y, Yao C, Sun J, Zhang Y. Hybrid membranes-mediated biomimetic-nanoparticle carrying miR-665 for effective tumor treatment by remodeling tumor microenvironment. Int J Pharm 2025; 675:125479. [PMID: 40090634 DOI: 10.1016/j.ijpharm.2025.125479] [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: 11/07/2024] [Revised: 03/03/2025] [Accepted: 03/13/2025] [Indexed: 03/18/2025]
Abstract
BACKGROUND Osteosarcomas (OS) are malignant bone tumors prevalent in adolescents, characterized by aggressiveness and early metastasis. Current treatments including surgery and chemotherapy face challenges due to drug limitations and the complex tumor microenvironment (TME). METHODS Tumor membranes (TM) derived from OS cells and macrophage membranes (MM) derived from macrophages were mixed to create hybrid membranes (HM), which were subsequently used to encapsulate microRNA-665(miR-665)-loaded Poly lactic-co-glycolic acid (PLGA) nanoparticles, forming HM@PLGA/miR-665 complexes. In vitro characterization included physical properties, colocalization studies, and assessment of macrophage polarization. In vivo experiments involved a nude mouse model to evaluate tumor targeting, biosafety, and therapeutic efficacy. RESULTS The HM@PLGA/miR-665 complexes exhibited good physical characteristics and stability. In vitro, the complexes significantly altered the M1/M2 macrophage ratio, promoting M1 polarization and inhibiting M2 polarization. Supernatants from HM@PLGA/miR-665-treated macrophages inhibited proliferation, migration, and induced apoptosis in MG-63 osteosarcoma cells. In vivo, the complexes effectively targeted tumor tissues, showed good biosafety, and significantly inhibited OS progression, promoting tumor cell apoptosis and altering the M1/M2 macrophage ratio. CONCLUSION The HM@PLGA/miR-665 delivery system successfully targeted OS by modulating macrophages in the TME, exhibiting potential as a novel therapeutic strategy for OS.
Collapse
Affiliation(s)
- Bo Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, PR China
| | - Chi Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, PR China
| | - Cao Chen
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, PR China
| | - Ru Hong
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, PR China
| | - Yongping Shen
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, PR China
| | - Chen Yao
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, PR China
| | - Jie Sun
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, PR China.
| | - Yafeng Zhang
- Department of Orthopaedics, Affiliated Hospital of Nantong University, Nantong City, Jiangsu Province, PR China.
| |
Collapse
|
2
|
Xie M, Zhao J, Feng X, Gao X, Cheng W, Kong L, Liang F. Cell membrane-inspired chitosan nanoparticles for prolonged circulation and tumor-targeted drug delivery. Int J Biol Macromol 2025; 304:140934. [PMID: 39947555 DOI: 10.1016/j.ijbiomac.2025.140934] [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: 11/08/2024] [Revised: 01/25/2025] [Accepted: 02/10/2025] [Indexed: 02/16/2025]
Abstract
Conventional chemotherapy is challenged by its inherent limitations. As an emerging drug delivery system, nanoparticles are easily eliminated by the immune system due to their exogenous nature. Moreover, the lack of long-circulation and tumor-targeting greatly limits their application. In our research, to address this problem, the biomimetic nanocarriers combine cell membranes, targeting ligand and nanoparticles into one system. Firstly, an active ester monomer and three terpolymers PMAN containing different proportions of phosphorylcholine, sulfonic acid and active ester group were successfully synthesized. Subsequently, aminated-folate was synthesized and grafted onto the terpolymers to prepare the folate-targeted polymers FA-PMAN. Finally, cell membrane biomimetic and folate-targeted chitosan nanoparticles (FA-PMAN/CS) with average particle size of 100 to 200 nm were prepared through electrostatic interactions. The in vivo blood circulation results showed that the long-circulating properties of PMAN/CS nanoparticles were significantly enhanced, compared with TPP/CS nanoparticle as a control. The half-life extended from 2.71 h to 12.95 h, with five-fold increase. Additionally, the uptake efficiency of FA-PMAN/CS nanoparticles by rat breast cancer cells (MADB-106) was 5-6 times greater than that of nanoparticles without folate. The biological distribution in mice demonstrated that the signal intensity in tumors treated with FA-PMAN/CS nanoparticle was substantially higher than in other organs, indicating that folate on the surface of nanoparticles increased the uptake into cancer cells. This work aims to design more efficient nanocarriers through biomimetic simulation of cell membranes, reduce recognition and clearance by the immune system, prolong blood circulation time and achieve tumor-targeted drug delivery. It is expected to significantly improve efficacy and provide an innovative strategy for clinical treatment.
Collapse
Affiliation(s)
- Mingzhu Xie
- School of Environmental and Chemical Engineering, Xi'an Polytechnic University, Xi'an 710048, China
| | - Jing Zhao
- School of Environmental and Chemical Engineering, Xi'an Polytechnic University, Xi'an 710048, China.
| | - Xiaokai Feng
- School of Environmental and Chemical Engineering, Xi'an Polytechnic University, Xi'an 710048, China
| | - Xu Gao
- School of Environmental and Chemical Engineering, Xi'an Polytechnic University, Xi'an 710048, China
| | - Wenwen Cheng
- School of Environmental and Chemical Engineering, Xi'an Polytechnic University, Xi'an 710048, China
| | - Lingheng Kong
- Department of Pharmacy, Xi'an Medical University, Xi'an 710021, China
| | - Fei Liang
- Department of Pharmacy, Xi'an Medical University, Xi'an 710021, China
| |
Collapse
|
3
|
Ahmed T, Alam KT. Biomimetic Nanoparticle Based Targeted mRNA Vaccine Delivery as a Novel Therapy for Glioblastoma Multiforme. AAPS PharmSciTech 2025; 26:68. [PMID: 39984771 DOI: 10.1208/s12249-025-03065-z] [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: 09/24/2024] [Accepted: 02/06/2025] [Indexed: 02/23/2025] Open
Abstract
The prognosis for patients with glioblastoma multiforme (GBM), an aggressive and deadly brain tumor, is poor due to the limited therapeutic options available. Biomimetic nanoparticles have emerged as a promising vehicle for targeted mRNA vaccine delivery, thanks to recent advances in nanotechnology. This presents a novel treatment method for GBM. This review explores the potential of using biomimetic nanoparticles to improve the specificity and effectiveness of mRNA vaccine against GBM. These nanoparticles can evade immune detection, cross the blood-brain barrier, & deliver mRNA directly to glioma cells by mimicking natural biological structures. This allows glioma cells to produce tumor-specific antigens that trigger strong immune responses against the tumor. This review discusses biomimetic nanoparticle design strategies, which are critical for optimizing transport and ensuring targeted action. These tactics include surface functionalization and encapsulation techniques. It also highlights the ongoing preclinical research and clinical trials that demonstrate the therapeutic advantages and challenges of this strategy. Biomimetic nanoparticles for mRNA vaccine delivery represent a new frontier in GBM treatment, which could impact the management of this deadly disease and improve patient outcomes by integrating cutting-edge nanotechnology with immunotherapy.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, School of Health and Life Sciences, North South University, Plot 15, Block B, Bashundhara R/A, Dhaka, 1229, Bangladesh.
| | - Kazi Tasnuva Alam
- Department of Pharmaceutical Sciences, School of Health and Life Sciences, North South University, Plot 15, Block B, Bashundhara R/A, Dhaka, 1229, Bangladesh
| |
Collapse
|
4
|
Wahengbam GS, Nirmal S, Nandwana J, Kar S, Kumari V, Mishra R, Singh A. Polymeric Nanoparticles Revolutionizing Brain Cancer Therapy: A Comprehensive Review of Strategies and Advances. Crit Rev Ther Drug Carrier Syst 2025; 42:73-106. [PMID: 39819464 DOI: 10.1615/critrevtherdrugcarriersyst.2024051822] [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: 01/19/2025]
Abstract
Brain cancer continues to be one of the most formidable malignancies to manage, mainly attributable to the presence of the blood-brain barrier (BBB) limiting the permeability of drugs and the diverse characteristics of brain tumors complicating treatment. The management of brain tumors has been hampered by many different factors, including the impermeability of the BBB, which restricts the delivery of chemotherapeutic agents to the tumor site, as well as intertumoral heterogeneity and the influence of brain tumor stem cells. In addition, small molecular weight drugs cannot specifically accumulate in malignant cells and have a limited circulation half-life. Nanoparticles (NPs) can be engineered to traverse the BBB and transport therapeutic medications directly into the brain, enhancing their efficacy compared with the conventional delivery of unbound drugs. Surface modifications of NPs can boost their efficiency by increasing their selectivity towards tumor receptors. This review covers treatment methods for malignant gliomas, associated risk factors, and improvements in brain drug administration, emphasizing the future potential of polymeric NPs and their mechanism for crossing the BBB. To surmount these obstacles, the newly formulated drug-delivery approach utilizing NPs, particularly those coated with cell membranes, has demonstrated potential in treating brain cancer. These NPs provide targeted tumor specificity, biocompatibility, extended circulation, enhanced BBB penetration, and immune evasion. This review focuses on coating strategies for PLGA NPs, particularly dual-targeting methods, to enhance BBB permeability and tumor-targeted delivery of drugs in brain cancer.
Collapse
Affiliation(s)
| | - Sakshi Nirmal
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Jai Nandwana
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Swatileena Kar
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Vandana Kumari
- Cancer Research Laboratory, Department of Biosciences, Manipal University Jaipur, India
| | - Rajeev Mishra
- Department of Life Sciences and Biotechnology, Chhatrapati Shahu Ji Maharaj University, Kanpur, India
| | | |
Collapse
|
5
|
Zeng Q, Zhang S, Leng N, Xing Y. Advancing tumor vaccines: Overcoming TME challenges, delivery strategies, and biomaterial-based vaccine for enhanced immunotherapy. Crit Rev Oncol Hematol 2025; 205:104576. [PMID: 39581246 DOI: 10.1016/j.critrevonc.2024.104576] [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: 10/08/2024] [Revised: 11/03/2024] [Accepted: 11/16/2024] [Indexed: 11/26/2024] Open
Abstract
Tumor vaccines, as an immunotherapeutic approach, harness the body's immune cells to provoke antitumor responses, which have shown promising efficacy in clinical settings. However, the immunosuppressive tumor microenvironment (TME) and the ineffective vaccine delivery systems hinder the progression of many vaccines beyond phase II trials. This article begins with a comprehensive review of the complex interactions between tumor vaccines and TME, summarizing the current state of vaccine clinical research. Subsequently, we review recent advancements in targeted vaccine delivery systems and explore biomaterial-based tumor vaccines as a strategy to improve the efficacy of both delivery systems and treatment. Finally, we have presented our perspectives on tumor vaccine development, aiming to advance the field towards the creation of more effective tumor vaccines.
Collapse
Affiliation(s)
- Qingsong Zeng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Shibo Zhang
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Ning Leng
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China
| | - Yingying Xing
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, 210009, PR China.
| |
Collapse
|
6
|
Marinho A, Reis S, Nunes C. On the design of cell membrane-coated nanoparticles to treat inflammatory conditions. NANOSCALE HORIZONS 2024; 10:38-55. [PMID: 39499543 DOI: 10.1039/d4nh00457d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2024]
Abstract
Biomimetic-based drug delivery systems (DDS) attempt to recreate the complex interactions that occur naturally between cells. Cell membrane-coated nanoparticles (CMCNPs) have been one of the main strategies in this area to prevent opsonization and clearance. Moreover, coating nanoparticles with cell membranes allows them to acquire functions and properties inherent to the mother cells. In particular, cells from bloodstream show to have specific advantages depending on the cell type to be used for that application, specifically in cases of chronic inflammation. Thus, this review focuses on the biomimetic strategies that use membranes from blood cells to target and treat inflammatory conditions.
Collapse
Affiliation(s)
- Andreia Marinho
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
- LAQV, REQUIMTE, Faculdade de Ciências, Universidade do Porto, R. Campo Alegre s/n, 4169-007 Porto, Portugal
| | - Salette Reis
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
| | - Cláudia Nunes
- LAQV, REQUIMTE, Faculdade de Farmácia, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal.
- LAQV, REQUIMTE, Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, R. Jorge de Viterbo Ferreira 228, 4500-313 Porto, Portugal
| |
Collapse
|
7
|
Wang H, Bo W, Feng X, Zhang J, Li G, Chen Y. Strategies and Recent Advances on Improving Efficient Antitumor of Lenvatinib Based on Nanoparticle Delivery System. Int J Nanomedicine 2024; 19:5581-5603. [PMID: 38882543 PMCID: PMC11177867 DOI: 10.2147/ijn.s460844] [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: 01/23/2024] [Accepted: 05/28/2024] [Indexed: 06/18/2024] Open
Abstract
Lenvatinib (LVN) is a potentially effective multiple-targeted receptor tyrosine kinase inhibitor approved for treating hepatocellular carcinoma, metastatic renal cell carcinoma and thyroid cancer. Nonetheless, poor pharmacokinetic properties including poor water solubility and rapid metabolic, complex tumor microenvironment, and drug resistance have impeded its satisfactory therapeutic efficacy. This article comprehensively reviews the uses of nanotechnology in LVN to improve antitumor effects. With the characteristic of high modifiability and loading capacity of the nano-drug delivery system, an active targeting approach, controllable drug release, and biomimetic strategies have been devised to deliver LVN to target tumors in sequence, compensating for the lack of passive targeting. The existing applications and advances of LVN in improving therapeutic efficacy include improving longer-term efficiency, achieving higher efficiency, combination therapy, tracking and diagnosing application and reducing toxicity. Therefore, using multiple strategies combined with photothermal, photodynamic, and immunoregulatory therapies potentially overcomes multi-drug resistance, regulates unfavorable tumor microenvironment, and yields higher synergistic antitumor effects. In brief, the nano-LVN delivery system has brought light to the war against cancer while at the same time improving the antitumor effect. More intelligent and multifunctional nanoparticles should be investigated and further converted into clinical applications in the future.
Collapse
Affiliation(s)
- Haiqing Wang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Wentao Bo
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Xielin Feng
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Jinliang Zhang
- Department of Hepatopancreatobiliary Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Ge Li
- Department of Emergency, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| | - Yan Chen
- Department of Pharmacy, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, People’s Republic of China
| |
Collapse
|
8
|
Chheda D, Shete S, Tanisha T, Devrao Bahadure S, Sampathi S, Junnuthula V, Dyawanapelly S. Multifaceted therapeutic applications of biomimetic nanovaccines. Drug Discov Today 2024; 29:103991. [PMID: 38663578 DOI: 10.1016/j.drudis.2024.103991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
The development of vaccines has had a crucial role in preventing and controlling infectious diseases on a global scale. Innovative formulations of biomimetic vaccines inspired by natural defense mechanisms combine long-term antigen stability, immunogenicity, and targeted delivery with sustained release. Types of biomimetic nanoparticle (NP) include bacterial outer membrane vesicles (OMVs), cell membrane-decorated NPs, liposomes, and exosomes. These approaches have shown potential for cancer immunotherapy, and in antibacterial and antiviral applications. Despite current challenges, nanovaccines have immense potential to transform disease prevention and treatment, promising therapeutic approaches for the future. In this review, we highlight recent advances in biomimetic vaccine design, mechanisms of action, and clinical applications, emphasizing their role in personalized medicine, targeted drug delivery, and immunomodulation.
Collapse
Affiliation(s)
- Dev Chheda
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Sukhen Shete
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Tanisha Tanisha
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Hajipur, India
| | - Sumedh Devrao Bahadure
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, Guwahati, India
| | - Sunitha Sampathi
- Department of Pharmacy, School of Pharmacy, Vishwakarma University, Pune, Maharashtra, India.
| | | | - Sathish Dyawanapelly
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India.
| |
Collapse
|
9
|
Ozsoy F, Mohammed M, Jan N, Lulek E, Ertas YN. T Cell and Natural Killer Cell Membrane-Camouflaged Nanoparticles for Cancer and Viral Therapies. ACS APPLIED BIO MATERIALS 2024; 7:2637-2659. [PMID: 38687958 PMCID: PMC11110059 DOI: 10.1021/acsabm.4c00074] [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: 01/16/2024] [Revised: 04/17/2024] [Accepted: 04/19/2024] [Indexed: 05/02/2024]
Abstract
Extensive research has been conducted on the application of nanoparticles in the treatment of cancer and infectious diseases. Due to their exceptional characteristics and flexible structure, they are classified as highly efficient drug delivery systems, ensuring both safety and targeted delivery. Nevertheless, nanoparticles still encounter obstacles, such as biological instability, absence of selectivity, recognition as unfamiliar elements, and quick elimination, which restrict their remedial capacity. To surmount these drawbacks, biomimetic nanotechnology has been developed that utilizes T cell and natural killer (NK) cell membrane-encased nanoparticles as sophisticated methods of administering drugs. These nanoparticles can extend the duration of drug circulation and avoid immune system clearance. During the membrane extraction and coating procedure, the surface proteins of immunological cells are transferred to the biomimetic nanoparticles. Such proteins present on the surface of cells confer several benefits to nanoparticles, including prolonged circulation, enhanced targeting, controlled release, specific cellular contact, and reduced in vivo toxicity. This review focuses on biomimetic nanosystems that are derived from the membranes of T cells and NK cells and their comprehensive extraction procedure, manufacture, and applications in cancer treatment and viral infections. Furthermore, potential applications, prospects, and existing challenges in their medical implementation are highlighted.
Collapse
Affiliation(s)
- Fatma Ozsoy
- ERNAM−Nanotechnology
Research and Application Center, Erciyes
University, Kayseri 38039, Turkey
- Department
of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
| | - Mahir Mohammed
- ERNAM−Nanotechnology
Research and Application Center, Erciyes
University, Kayseri 38039, Turkey
| | - Nasrullah Jan
- Department
of Pharmacy, The University of Chenab, Gujrat, Punjab 50700, Pakistan
| | - Elif Lulek
- ERNAM−Nanotechnology
Research and Application Center, Erciyes
University, Kayseri 38039, Turkey
- Department
of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
| | - Yavuz Nuri Ertas
- ERNAM−Nanotechnology
Research and Application Center, Erciyes
University, Kayseri 38039, Turkey
- Department
of Biomedical Engineering, Erciyes University, Kayseri 38039, Turkey
- UNAM−National
Nanotechnology Research Center, Bilkent
University, Ankara 06800, Turkey
| |
Collapse
|
10
|
Peng J, Zhou J, Liu X, Zhang X, Zhou X, Gong Z, Chen Y, Shen X, Chen Y. A biomimetic nanocarrier facilitates glucose consumption and reactive oxide species accumulation in enzyme therapy for colorectal cancer. J Control Release 2024; 367:76-92. [PMID: 38262488 DOI: 10.1016/j.jconrel.2024.01.041] [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: 10/13/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 01/25/2024]
Abstract
Glucose oxidase (GOx)-based enzyme therapeutics are potential alternatives for colorectal cancer (CRC) treatment via glucose consumption and accumulation of hydrogen peroxide (H2O2). Given that H2O2 can be eliminated by cytoprotective autophagy, autophagy inhibitors that can interrupt autolysosome-induced H2O2 elimination are promising combination drugs of GOx. Here, we developed a multifunctional biomimetic nanocarrier for effective co-delivery of an autophagy inhibitor-chloroquine phosphate (CQP) and GOx to exert their synergistic effect by irreversibly upregulating intracellular reactive oxygen species (ROS) levels. Poly (D, l-lactide-co-glycolide) (PLGA) nanoparticles (NPs) were used to encapsulate both GOx and CQP using a W/O/W multi-emulsion method. Calcium phosphate (CaP) was used to "fix" CQP to GOx in the internal water phase, where it served as a pH-sensitive unit to facilitate intracellular drug release. Folic acid-modified red blood cell membranes (FR) were used to camouflage the GOx/CQP/CaP encapsulated PLGA NPs (referred to as PLGA/GCC@FR). In an AOM/DSS-induced CRC mouse model, PLGA/GCC@FR exhibited improved antitumor effects, in which the number of tumor nodes were only a quarter of that in the free drug combination group. The enhanced therapeutic effects of PLGA/GCC@FR were attributed to the prolonged tumor retention which was verified by both dynamic in vivo imaging and drug biodistribution. This multifunctional biomimetic nanocarrier facilitated combined enzyme therapeutics by depleting glucose and augmenting intracellular ROS levels in tumor cells, which exerted a synergistic inhibitory effect on tumor growth. Therefore, this study proposed a novel strategy for the enhancement of combined enzyme therapeutics, which provided a promising method for effective CRC treatment.
Collapse
Affiliation(s)
- Jianqing Peng
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Jia Zhou
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xing Liu
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xiaobo Zhang
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Xiang Zhou
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Zipeng Gong
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; Guizhou Provincial Key Laboratory of Pharmaceutics, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China
| | - Yi Chen
- State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Xiangchun Shen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| | - Yan Chen
- The High Efficacy Application of Natural Medicinal Resources Engineering Center of Guizhou Province, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; State Key Laboratory of Functions and Applications of Medicinal Plants, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China; Key Laboratory of Novel Anti-Cancer Drug Targets Discovery and Application, School of Pharmaceutical Sciences, Guizhou Medical University, Guizhou 561113, China.
| |
Collapse
|
11
|
Liu Z, Xie H, Wang T. Erythrocyte-Cancer Hybrid Membrane-Camouflaged Prussian Blue Nanoparticles with Enhanced Photothermal Therapy in Tumors. ACS OMEGA 2023; 8:23056-23066. [PMID: 37396272 PMCID: PMC10308386 DOI: 10.1021/acsomega.3c02370] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Accepted: 05/26/2023] [Indexed: 07/04/2023]
Abstract
Prussian blue (PB) nanoparticles have been widely used in photothermal therapy research due to the efficient photothermal conversion ability. In this study, PB was modified with a bionic coating using a hybrid membrane of red blood cell membranes and tumor cell membranes to prepare bionic photothermal nanoparticles (PB/RHM), which can further improve the blood circulation ability and tumor targeting of the nanoparticles to achieve efficient photothermal therapy for tumor treatment. In vitro formulation characterization showed that PB/RHM was a monodisperse spherical core-shell structured nanoparticle with a diameter of 207.2 nm and effectively retained the cell membrane proteins. The in vivo biological evaluation results showed that PB/RHM could effectively accumulate into the tumor tissue, inducing a rapid temperature increase in the tumor site to 50.9 °C within 10 min, inhibiting tumor growth efficiently with a tumor inhibition rate of 93.56% and with good therapeutic safety. In summary, this paper provided a hybrid film-modified Prussian blue nanoparticle with efficient photothermal anti-tumor capacity and safety.
Collapse
Affiliation(s)
- Zhining Liu
- Ultrasound
Department, First Affiliated Hospital of
Jinzhou Medical University, Jinzhou 121001, China
| | - Huichao Xie
- College
of Pharmacy, Shenzhen Technology University, Shenzhen 518118, China
| | - Tianyi Wang
- Ultrasound
Department, First Affiliated Hospital of
Jinzhou Medical University, Jinzhou 121001, China
| |
Collapse
|
12
|
Avancini G, Menilli L, Visentin A, Milani C, Mastrotto F, Moret F. Mesenchymal Stem Cell Membrane-Coated TPCS 2a-Loaded Nanoparticles for Breast Cancer Photodynamic Therapy. Pharmaceutics 2023; 15:1654. [PMID: 37376102 PMCID: PMC10302938 DOI: 10.3390/pharmaceutics15061654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 05/26/2023] [Accepted: 05/31/2023] [Indexed: 06/29/2023] Open
Abstract
Despite substantial improvements in breast cancer (BC) treatment there is still an urgent need to find alternative treatment options to improve the outcomes for patients with advanced-stage disease. Photodynamic therapy (PDT) is gaining a lot of attention as a BC therapeutic option because of its selectivity and low off-target effects. However, the hydrophobicity of photosensitizers (PSs) impairs their solubility and limits the circulation in the bloodstream, thus representing a major challenge. The use of polymeric nanoparticles (NPs) to encapsulate the PS may represent a valuable strategy to overcome these issues. Herein, we developed a novel biomimetic PDT nanoplatform (NPs) based on a polymeric core of poly(lactic-co-glycolic)acid (PLGA) loaded with the PS meso-tetraphenylchlorin disulfonate (TPCS2a). TPCS2a@NPs of 98.89 ± 18.56 nm with an encapsulation efficiency percentage (EE%) of 81.9 ± 7.92% were obtained and coated with mesenchymal stem cells-derived plasma membranes (mMSCs) (mMSC-TPCS2a@NPs, size of 139.31 ± 12.94 nm). The mMSC coating armed NPs with biomimetic features to impart long circulation times and tumor-homing capabilities. In vitro, biomimetic mMSC-TPCS2a@NPs showed a decrease in macrophage uptake of 54% to 70%, depending on the conditions applied, as compared to uncoated TPCS2a@NPs. Both NP formulations efficiently accumulated in MCF7 and MDA-MB-231 BC cells, while the uptake was significantly lower in normal breast epithelial MCF10A cells with respect to tumor cells. Moreover, encapsulation of TPCS2a in mMSC-TPCS2a@NPs effectively prevents its aggregation, ensuring efficient singlet oxygen (1O2) production after red light irradiation, which resulted in a considerable in vitro anticancer effect in both BC cell monolayers (IC50 < 0.15 µM) and three-dimensional spheroids.
Collapse
Affiliation(s)
- Greta Avancini
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Luca Menilli
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Adele Visentin
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Celeste Milani
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| | - Francesca Mastrotto
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy;
| | - Francesca Moret
- Department of Biology, University of Padova, Via Ugo Bassi 58/B, 35131 Padova, Italy; (G.A.); (L.M.); (C.M.)
| |
Collapse
|
13
|
Li H, Qiao W, Shen Y, Xu H, Fan Y, Liu Y, Lan Y, Gong Y, Chen F, Feng S. Biomimetic Boron Nitride Nanoparticles for Targeted Drug Delivery and Enhanced Antitumor Activity. Pharmaceutics 2023; 15:pharmaceutics15041269. [PMID: 37111754 PMCID: PMC10145272 DOI: 10.3390/pharmaceutics15041269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 04/05/2023] [Accepted: 04/17/2023] [Indexed: 04/29/2023] Open
Abstract
Boron nitride nanomaterials are being increasingly recognized as vehicles for cancer drug delivery that increase drug loading and control drug release because of their excellent physicochemical properties and biocompatibility. However, these nanoparticles are often cleared rapidly by the immune system and have poor tumor targeting effects. As a result, biomimetic nanotechnology has emerged to address these challenges in recent times. Cell-derived biomimetic carriers have the characteristics of good biocompatibility, long circulation time, and strong targeting ability. Here, we report a biomimetic nanoplatform (CM@BN/DOX) prepared by encapsulating boron nitride nanoparticles (BN) and doxorubicin (DOX) together using cancer cell membrane (CCM) for targeted drug delivery and tumor therapy. The CM@BN/DOX nanoparticles (NPs) were able to target cancer cells of the same type on its own initiative through homologous targeting of cancer cell membranes. This led to a remarkable increase in cellular uptake. In vitro simulation of an acidic tumor microenvironment could effectively promote drug release from CM@BN/DOX. Furthermore, the CM@BN/DOX complex exhibited an excellent inhibitory effect against homotypic cancer cells. These findings suggest that CM@BN/DOX are promising in targeted drug delivery and potentially personalized therapy against their homologous tumor.
Collapse
Affiliation(s)
- Hui Li
- School of Environmental and Chemical Engineering, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Wei Qiao
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Yizhe Shen
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Huashan Xu
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Yuan Fan
- School of Environmental and Chemical Engineering, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Yuxiang Liu
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Yadi Lan
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Yan Gong
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Fuxue Chen
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| | - Shini Feng
- School of Life Sciences, Shanghai University, 333 Nanchen Road, Shanghai 200444, China
| |
Collapse
|
14
|
Wu Y, Zhu R, Zhou M, Liu J, Dong K, Zhao S, Cao J, Wang W, Sun C, Wu S, Wang F, Shi Y, Sun Y. Homologous cancer cell membrane-camouflaged nanoparticles target drug delivery and enhance the chemotherapy efficacy of hepatocellular carcinoma. Cancer Lett 2023; 558:216106. [PMID: 36841418 DOI: 10.1016/j.canlet.2023.216106] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 02/27/2023]
Abstract
Hepatocellular carcinoma (HCC) is a common digestive tract malignancy that seriously threatens human life and health. Early HCC may be treated by intervention, surgery, and internal radiotherapy, while the choice for late HCC is primarily chemotherapy to prolong patient survival. Lenvatinib (LT) is a Food and Drug Administration (FDA)-approved frontline drug for the treatment of advanced liver cancer and has achieved excellent clinical efficacy. However, its poor solubility and severe side effects cannot be ignored. In this study, a bionic nanodrug delivery platform was successfully constructed. The platform consists of a core of Lenvatinib wrapped with a pH-sensitive polymer, namely, poly(β-amino ester)-polyethylene glycol-amine (PAE-PEG-NH2), and a shell formed by a cancer cell membrane (CCM). The prepared nanodrugs have high drug loading capacity, long-term stability, good biocompatibility, and a long retention time. In addition, the targeting effect of tumor cell membranes and the pH-responsive characteristics of the polymer materials enable them to precisely target tumor cells and achieve responsive release in the tumor microenvironment, which makes them suitable for effective drug delivery. In vivo experiments revealed that the nanodrug showed superior tumor accumulation and therapeutic effects in subcutaneous tumor mice model and could effectively eliminate tumors within 21 days. As a result, it opens up a new way to reduce side effects and improve the specific therapeutic effect of first-line clinical medications to treat tumors.
Collapse
Affiliation(s)
- Yahui Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China
| | - Rongtao Zhu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China
| | - Mengyang Zhou
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Jingjing Liu
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Kai Dong
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China
| | - Senfeng Zhao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China
| | - Jiahui Cao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China
| | - Weijie Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China
| | - Chenguang Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China
| | - Shitao Wu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China
| | - Fan Wang
- Experimental Animal Platform in Academy of Medical Sciences of Zhengzhou University, Zhengzhou, 450052, China
| | - Yupeng Shi
- Department of Magnetic Resonance Imaging, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Yuling Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China; Institute of Hepatobiliary and Pancreatic Diseases, Zhengzhou University, Zhengzhou, 450052, China; Zhengzhou Basic and Clinical Key Laboratory of Hepatopancreatobiliary Diseases, Zhengzhou, 450052, China.
| |
Collapse
|
15
|
Shao M, Lopes D, Lopes J, Yousefiasl S, Macário-Soares A, Peixoto D, Ferreira-Faria I, Veiga F, Conde J, Huang Y, Chen X, Paiva-Santos AC, Makvandi P. Exosome membrane-coated nanosystems: Exploring biomedical applications in cancer diagnosis and therapy. MATTER 2023; 6:761-799. [DOI: 10.1016/j.matt.2023.01.012] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
16
|
Wang S, Wang Y, Jin K, Zhang B, Peng S, Nayak AK, Pang Z. Recent advances in erythrocyte membrane-camouflaged nanoparticles for the delivery of anti-cancer therapeutics. Expert Opin Drug Deliv 2022; 19:965-984. [PMID: 35917435 DOI: 10.1080/17425247.2022.2108786] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022]
Abstract
INTRODUCTION Red blood cell (or erythrocyte) membrane-camouflaged nanoparticles (RBC-NPs) not only have a superior circulation life and do not induce accelerated blood clearance, but also possess special functions, which offers great potential in cancer therapy. AREAS COVERED This review focuses on the recent advances of RBC-NPs for delivering various agents to treat cancers in light of their vital role in improving drug delivery. Meanwhile, the construction and in vivo behavior of RBC-NPs are discussed to provide an in-depth understanding of the basis of RBC-NPs for improved cancer drug delivery. EXPERT OPINION Although RBC-NPs are quite prospective in delivering anti-cancer therapeutics, they are still in their infancy stage and many challenges need to be overcome for successful translation into the clinic. The preparation and modification of RBC membranes, the optimization of coating methods, the scale-up production and the quality control of RBC-NPs, and the drug loading and release should be carefully considered in the clinical translation of RBC-NPs for cancer therapy.
Collapse
Affiliation(s)
- Siyu Wang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| | - Yiwei Wang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Kai Jin
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| | - Bo Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science & Technology, Wuhan 430022, China
| | - Shaojun Peng
- Zhuhai Institute of Translational Medicine, Zhuhai Precision Medical Center, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Zhuhai, Guangdong 519000, China
| | - Amit Kumar Nayak
- Department of Pharmaceutics, Seemanta Institute of Pharmaceutical Sciences, Mayurbhanj-757086, Odisha, India
| | - Zhiqing Pang
- School of Pharmacy, Fudan University, Key Laboratory of Smart Drug Delivery, Ministry of Education, 826 Zhangheng Road, Shanghai, 201203, China
| |
Collapse
|
17
|
Gareev KG, Grouzdev DS, Koziaeva VV, Sitkov NO, Gao H, Zimina TM, Shevtsov M. Biomimetic Nanomaterials: Diversity, Technology, and Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2485. [PMID: 35889709 PMCID: PMC9316400 DOI: 10.3390/nano12142485] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 07/15/2022] [Accepted: 07/18/2022] [Indexed: 02/04/2023]
Abstract
Biomimetic nanomaterials (BNMs) are functional materials containing nanoscale components and having structural and technological similarities to natural (biogenic) prototypes. Despite the fact that biomimetic approaches in materials technology have been used since the second half of the 20th century, BNMs are still at the forefront of materials science. This review considered a general classification of such nanomaterials according to the characteristic features of natural analogues that are reproduced in the preparation of BNMs, including biomimetic structure, biomimetic synthesis, and the inclusion of biogenic components. BNMs containing magnetic, metal, or metal oxide organic and ceramic structural elements (including their various combinations) were considered separately. The BNMs under consideration were analyzed according to the declared areas of application, which included tooth and bone reconstruction, magnetic and infrared hyperthermia, chemo- and immunotherapy, the development of new drugs for targeted therapy, antibacterial and anti-inflammatory therapy, and bioimaging. In conclusion, the authors' point of view is given about the prospects for the development of this scientific area associated with the use of native, genetically modified, or completely artificial phospholipid membranes, which allow combining the physicochemical and biological properties of biogenic prototypes with high biocompatibility, economic availability, and scalability of fully synthetic nanomaterials.
Collapse
Affiliation(s)
- Kamil G. Gareev
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia; (N.O.S.); (T.M.Z.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Denis S. Grouzdev
- SciBear OU, Tartu mnt 67/1-13b, Kesklinna Linnaosa, 10115 Tallinn, Estonia;
| | - Veronika V. Koziaeva
- Research Center of Biotechnology of the Russian Academy of Sciences, Institute of Bioengineering, 119071 Moscow, Russia;
| | - Nikita O. Sitkov
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia; (N.O.S.); (T.M.Z.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Huile Gao
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, West China School of Pharmacy, Sichuan University, Chengdu 610041, China;
| | - Tatiana M. Zimina
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia; (N.O.S.); (T.M.Z.)
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
| | - Maxim Shevtsov
- Laboratory of Biomedical Nanotechnologies, Institute of Cytology of the Russian Academy of Sciences, 194064 Saint Petersburg, Russia
- Center of Translational Cancer Research (TranslaTUM), Klinikum Rechts der Isar, Technical University Munich, 81675 Munich, Germany
- Personalized Medicine Centre, Almazov National Medical Research Centre, 197341 Saint Petersburg, Russia
- National Center for Neurosurgery, Nur-Sultan 010000, Kazakhstan
| |
Collapse
|
18
|
Cell-based drug delivery systems and their in vivo fate. Adv Drug Deliv Rev 2022; 187:114394. [PMID: 35718252 DOI: 10.1016/j.addr.2022.114394] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/17/2022] [Accepted: 06/07/2022] [Indexed: 11/22/2022]
Abstract
Cell-based drug delivery systems (DDSs) have received attention recently because of their unique biological properties and self-powered functions, such as excellent biocompatibility, low immunogenicity, long circulation time, tissue-homingcharacteristics, and ability to cross biological barriers. A variety of cells, including erythrocytes, stem cells, and lymphocytes, have been explored as functional vectors for the loading and delivery of various therapeutic payloads (e.g., small-molecule and nucleic acid drugs) for subsequent disease treatment. These cell-based DDSs have their own unique in vivo fates, which are attributed to various factors, including their biological properties and functions, the loaded drugs and loading process, physiological and pathological circumstances, and the body's response to these carrier cells, which result in differences in drug delivery efficiency and therapeutic effect. In this review, we summarize the main cell-based DDSs and their biological properties and functions, applications in drug delivery and disease treatment, and in vivo fate and influencing factors. We envision that the unique biological properties, combined with continuing research, will enable development of cell-based DDSs as friendly drug vectors for the safe, effective, and even personalized treatment of diseases.
Collapse
|