1
|
Notarbartolo M, Alfieri ML, Avolio R, Ball V, Errico ME, Massaro M, Puglisi R, Sànchez-Espejo R, Viseras C, Riela S. Design of innovative and low-cost dopamine-biotin conjugate sensor for the efficient detection of protein and cancer cells. J Colloid Interface Sci 2025; 678:766-775. [PMID: 39307064 DOI: 10.1016/j.jcis.2024.09.145] [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: 06/13/2024] [Revised: 08/30/2024] [Accepted: 09/15/2024] [Indexed: 10/27/2024]
Abstract
The rapid, precise identification and quantification of specific biomarkers, toxins, or pathogens is currently a key strategy for achieving more efficient diagnoses. Herein a dopamine-biotin monomer was synthetized and oxidized in the presence of hexamethylenediamine, to obtain adhesive coatings based on polydopamine-biotin (PDA-BT) on different materials to be used in targeted molecular therapy. Insight into the structure of the PDA-BT coating was obtained by solid-state 13C NMR spectroscopy acquired, for the first time, directly onto the coating, deposited on alumina spheres. The receptor binding capacity of the PDA-BT coating toward 4-hydroxyazobenzene-2-carboxylic acid/Avidin complex was verified by means of UV-vis spectroscopy. Different deposition cycles of avidin onto the PDA-BT coating by layer-by-layer assembly showed that the film retains its receptor binding capacity for at least eight consecutive cycles. Finally, the feasibility of PDA-BT coating to recognize cell lines with different grade of overexpression of biotin receptors (BR) was investigated by tumor cell capture experiments by using MCF-7 (BR+) and HL-60 (BR-) cell lines. The results show that the developed system can selectively capture MCF-7 cells indicating that it could represent a first approach for the development of future more sophisticated biosensors easily accessible, low cost and recyclable with the dual and rapid detection of both proteins and cells.
Collapse
Affiliation(s)
- Monica Notarbartolo
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, Parco d'Orleans II, Ed. 16-17, 90128 Palermo, Italy
| | - Maria Laura Alfieri
- Department of Chemical Sciences, University of Naples "Federico II", I-80126 Naples, Italy.
| | - Roberto Avolio
- Institute of Chemistry and Technology of Polymers, National Council of Research (CNR), via Campi Flegrei 34, Pozzuoli I-80078, Italy
| | - Vincent Ball
- Université de Strasbourg, Faculté de Chirurgie Dentaire, 8 rue Sainte Elizabeth, 67000 Strasbourg. France; Institut National de la Santé et de la Recherche Médicale, Unité mixte de rechere 1121. 1 rue Eugène Boeckel, 67084 Strasbourg Cedex, France
| | - Maria Emanuela Errico
- Institute of Chemistry and Technology of Polymers, National Council of Research (CNR), via Campi Flegrei 34, Pozzuoli I-80078, Italy
| | - Marina Massaro
- Dipartimento di Scienze e Tecnologie Biologiche, Chimiche e Farmaceutiche (STEBICEF), Università di Palermo, Viale delle Scienze, Parco d'Orleans II, Ed. 16-17, 90128 Palermo, Italy.
| | - Roberta Puglisi
- Dipartimento di Scienze Chimiche (DSC), Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| | - Rita Sànchez-Espejo
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Campus Universitario de Cartuja, 18071 Granada, Spain
| | - César Viseras
- Department of Pharmacy and Pharmaceutical Technology, Faculty of Pharmacy, University of Granada, Campus Universitario de Cartuja, 18071 Granada, Spain; Andalusian Institute of Earth Sciences, CSIC-UGR, 18100 Armilla, Granada, Spain
| | - Serena Riela
- Dipartimento di Scienze Chimiche (DSC), Università di Catania, Viale Andrea Doria 6, 95125 Catania, Italy
| |
Collapse
|
2
|
Hadi Barhaghtalab R, Tanimowo Aiyelabegan H, Maleki H, Mirzavi F, Gholizadeh Navashenaq J, Abdi F, Ghaffari F, Vakili-Ghartavol R. Recent advances with erythrocytes as therapeutics carriers. Int J Pharm 2024; 665:124658. [PMID: 39236775 DOI: 10.1016/j.ijpharm.2024.124658] [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: 06/06/2024] [Revised: 08/24/2024] [Accepted: 08/31/2024] [Indexed: 09/07/2024]
Abstract
Erythrocytes have gained popularity as a natural option for in vivo drug delivery due to their advantages, which include lengthy circulation times, biocompatibility, and biodegradability. Consequently, the drug's pharmacokinetics and pharmacodynamics in red blood cells can be considerably up the dosage. Here, we provide an overview of the erythrocyte membrane's structure and discuss the characteristics of erythrocytes that influence their suitability as carrier systems. We also cover current developments in the erythrocyte-based nanocarrier, which could be used for both active and passive targeting of disease tissues, particularly those of the reticuloendothelial system (RES) and cancer tissues. We also go over the most recent discoveries about the in vivo and in vitro uses of erythrocytes for medicinal and diagnostic purposes. Moreover, the clinical relevance of erythrocytes is discussed in order to improve comprehension and enable the potential use of erythrocyte carriers in the management of various disorders.
Collapse
Affiliation(s)
| | | | - Hassan Maleki
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Farshad Mirzavi
- Cardiovascular Diseases Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Fereshteh Abdi
- Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran; Noncommunicable Diseases Research Center, Bam University of Medical Sciences, Bam, Iran
| | - Faezeh Ghaffari
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Roghayyeh Vakili-Ghartavol
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran; Department of Medical Nanotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
3
|
Wang Y, Zhou Q, Luo W, Yang X, Zhang J, Lou Y, Mao J, Chen J, Wu F, Hou J, Tang G, Bai H, Yu R. A collagenase-decorated Cu-based nanotheranostics: remodeling extracellular matrix for optimizing cuproptosis and MRI in pancreatic ductal adenocarcinoma. J Nanobiotechnology 2024; 22:689. [PMID: 39523309 PMCID: PMC11552245 DOI: 10.1186/s12951-024-02968-6] [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: 08/17/2024] [Accepted: 11/03/2024] [Indexed: 11/16/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC), characterized by a dense extracellular matrix (ECM), presents significant therapeutic challenges due to its poor prognosis and high resistance to chemotherapy. Current chemodrugs and diagnostic agents largely fail to cross the barrier posed by the ECM, which severely limits the PDAC theranostics. This study introduces a novel theranostic strategy using thioether-hybridized hollow mesoporous organosilica nanoparticles (dsMNs) for the co-delivery of copper (Cu) and disulfiram (DSF), aiming to induce cuproptosis in PDAC cells. Our approach leverages the ECM-degrading enzyme collagenase, integrated with dsMNs, to enhance drug penetration by reducing matrix stiffness. Furthermore, the innovative use of a pancreatic cancer cell membrane coating on the nanoparticles enhances tumor targeting and stability (dsMCu-D@M-Co). The multifunctional platform not only facilitates deep drug penetration and triggers cuproptosis effectively but also utilizes the inherent properties of Cu to serve as a T1-weighted magnetic resonance imaging (MRI) contrast agent. In vitro and in vivo assessments demonstrate significant tumor size reduction in PDAC-bearing mice, highlighting the dual functionality of our platform in improving therapeutic efficacy and diagnostic precision. This integrated strategy represents a significant advancement in the management of PDAC, offering a promising new direction for overcoming one of the most lethal cancers.
Collapse
Affiliation(s)
- Yining Wang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Qiaomei Zhou
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Wangping Luo
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Xiaoyan Yang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Jinguo Zhang
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Yijie Lou
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, People's Republic of China
| | - Jin Mao
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Jiayi Chen
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, People's Republic of China
| | - Fan Wu
- Department of Neurosurgery, First Affiliated Hospital, School of Medicine, Zhejiang University, 79 Qingchun Road, Hangzhou, Zhejiang, 310009, People's Republic of China
| | - Jue Hou
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, People's Republic of China
| | - Guping Tang
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, People's Republic of China
| | - Hongzhen Bai
- Department of Chemistry, Zhejiang University, Hangzhou, 310028, People's Republic of China.
| | - Risheng Yu
- Department of Radiology, Second Affiliated Hospital, School of Medicine, Zhejiang University, 88 Jiefang Road, Hangzhou, Zhejiang, 310009, People's Republic of China.
| |
Collapse
|
4
|
Tikhonov A, Kachanov A, Yudaeva A, Danilik O, Ponomareva N, Karandashov I, Kostyusheva A, Zamyatnin AA, Parodi A, Chulanov V, Brezgin S, Kostyushev D. Biomimetic Nanoparticles for Basic Drug Delivery. Pharmaceutics 2024; 16:1306. [PMID: 39458635 PMCID: PMC11510494 DOI: 10.3390/pharmaceutics16101306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 10/28/2024] Open
Abstract
Biomimetic nanoparticles (BMNPs) are innovative nanovehicles that replicate the properties of naturally occurring extracellular vesicles, facilitating highly efficient drug delivery across biological barriers to target organs and tissues while ensuring maximal biocompatibility and minimal-to-no toxicity. BMNPs can be utilized for the delivery of therapeutic payloads and for imparting novel properties to other nanotechnologies based on organic and inorganic materials. The application of specifically modified biological membranes for coating organic and inorganic nanoparticles has the potential to enhance their therapeutic efficacy and biocompatibility, presenting a promising pathway for the advancement of drug delivery technologies. This manuscript is grounded in the fundamentals of biomimetic technologies, offering a comprehensive overview and analytical perspective on the preparation and functionalization of BMNPs, which include cell membrane-coated nanoparticles (CMCNPs), artificial cell-derived vesicles (ACDVs), and fully synthetic vesicles (fSVs). This review examines both "top-down" and "bottom-up" approaches for nanoparticle preparation, with a particular focus on techniques such as cell membrane coating, cargo loading, and microfluidic fabrication. Additionally, it addresses the technological challenges and potential solutions associated with the large-scale production and clinical application of BMNPs and related technologies.
Collapse
Affiliation(s)
- Andrey Tikhonov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Artyom Kachanov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Alexandra Yudaeva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Oleg Danilik
- Department of Pharmaceutical and Toxicological Chemistry, First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
| | - Natalia Ponomareva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
- Department of Pharmaceutical and Toxicological Chemistry, First Moscow State Medical University (Sechenov University), 119146 Moscow, Russia;
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Ivan Karandashov
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Anastasiya Kostyusheva
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Andrey A. Zamyatnin
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119992 Moscow, Russia
| | - Alessandro Parodi
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
| | - Vladimir Chulanov
- Department of Infectious Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia;
| | - Sergey Brezgin
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
| | - Dmitry Kostyushev
- Laboratory of Genetic Technologies, Martsinovsky Institute of Medical Parasitology, Tropical and Vector-Borne Diseases, First Moscow State Medical University (Sechenov University), 119991 Moscow, Russia; (A.T.); (A.K.); (A.Y.); (N.P.); (I.K.); (A.K.); (S.B.)
- Division of Biotechnology, Sirius University of Science and Technology, 354340 Sochi, Russia;
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, 119234 Moscow, Russia;
| |
Collapse
|
5
|
Meng Z, Ouyang H, Hu Y, Chen B, Dong X, Wang T, Wu M, Yu N, Lou X, Wang S, Xia F, Dai J. Surface-engineered erythrocyte membrane-camouflage fluorescent bioprobe for precision ovarian cancer surgery. Eur J Nucl Med Mol Imaging 2024; 51:3532-3544. [PMID: 38867107 DOI: 10.1007/s00259-024-06793-9] [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: 01/21/2024] [Accepted: 06/05/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE Fluorescence imaging-guided surgery has been used in oncology. However, for tiny tumors, the current imaging probes are still difficult to achieve high-contrast imaging, leading to incomplete resection. In this study, we achieved precise surgical resection of tiny metastatic cancers by constructing an engineering erythrocyte membrane-camouflaged bioprobe (AR-M@HMSN@P). METHODS AR-M@HMSN@P combined the properties of aggregation-induced emission luminogens (AIEgens) named PF3-PPh3 (P), with functional erythrocyte membrane modified by a modular peptide (AR). Interestingly, AR was composed of an asymmetric tripodal pentapeptide scaffold (GGKGG) with three appended modulars: KPSSPPEE (A6) peptide, RRRR (R4) peptide and cholesterol. To verify the specificity of the probe in vitro, SKOV3 cells with overexpression of CD44 were used as the positive group, and HLF cells with low expression of CD44 were devoted as the control group. The AR-M@HMSN@P fluorescence imaging was utilized to provide surgical guidance for the removal of micro-metastatic lesions. RESULTS In vivo, the clearance of AR-M@HMSN@P by the immune system was reduced due to the natural properties inherited from erythrocytes. Meanwhile, the A6 peptide on AR-M@HMSN@P was able to specifically target CD44 on ovarian cancer cells, and the electrostatic attraction between the R4 peptide and the cell membrane enhanced the firmness of this targeting. Benefiting from these multiple effects, AR-M@HMSN@P achieved ultra-precise tumor imaging with a signal-to-noise ratio (SNR) of 15.2, making it possible to surgical resection of tumors < 1 mm by imaging guidance. CONCLUSION We have successfully designed an engineered fluorescent imaging bioprobe (AR-M@HMSN@P), which can target CD44-overexpressing ovarian cancers for precise imaging and guide the resection of minor tumors. Notably, this work holds significant promise for developing biomimetic probes for clinical imaging-guided precision cancer surgery by exploiting their externally specified functional modifications.
Collapse
Affiliation(s)
- Zijuan Meng
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Hanzhi Ouyang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Yuxin Hu
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Biao Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430034, China
| | - Xiyuan Dong
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430034, China
| | - Tingting Wang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Meng Wu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430034, China
| | - Nan Yu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430034, China
| | - Xiaoding Lou
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Shixuan Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430034, China
| | - Fan Xia
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
| | - Jun Dai
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430034, China.
| |
Collapse
|
6
|
Rezaei B, Harun A, Wu X, Iyer PR, Mostufa S, Ciannella S, Karampelas IH, Chalmers J, Srivastava I, Gómez-Pastora J, Wu K. Effect of Polymer and Cell Membrane Coatings on Theranostic Applications of Nanoparticles: A Review. Adv Healthc Mater 2024; 13:e2401213. [PMID: 38856313 DOI: 10.1002/adhm.202401213] [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: 04/01/2024] [Revised: 05/28/2024] [Indexed: 06/11/2024]
Abstract
The recent decade has witnessed a remarkable surge in the field of nanoparticles, from their synthesis, characterization, and functionalization to diverse applications. At the nanoscale, these particles exhibit distinct physicochemical properties compared to their bulk counterparts, enabling a multitude of applications spanning energy, catalysis, environmental remediation, biomedicine, and beyond. This review focuses on specific nanoparticle categories, including magnetic, gold, silver, and quantum dots (QDs), as well as hybrid variants, specifically tailored for biomedical applications. A comprehensive review and comparison of prevalent chemical, physical, and biological synthesis methods are presented. To enhance biocompatibility and colloidal stability, and facilitate surface modification and cargo/agent loading, nanoparticle surfaces are coated with different synthetic polymers and very recently, cell membrane coatings. The utilization of polymer- or cell membrane-coated nanoparticles opens a wide variety of biomedical applications such as magnetic resonance imaging (MRI), hyperthermia, photothermia, sample enrichment, bioassays, drug delivery, etc. With this review, the goal is to provide a comprehensive toolbox of insights into polymer or cell membrane-coated nanoparticles and their biomedical applications, while also addressing the challenges involved in translating such nanoparticles from laboratory benchtops to in vitro and in vivo applications. Furthermore, perspectives on future trends and developments in this rapidly evolving domain are provided.
Collapse
Affiliation(s)
- Bahareh Rezaei
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, 79409, United States
| | - Asma Harun
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, 79409, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, Texas, 79106, United States
| | - Xian Wu
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, United States
| | - Poornima Ramesh Iyer
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, United States
| | - Shahriar Mostufa
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, 79409, United States
| | - Stefano Ciannella
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, 79409, United States
| | | | - Jeffrey Chalmers
- Department of Chemical and Biomolecular Engineering, The Ohio State University, Columbus, OH, 43210, United States
| | - Indrajit Srivastava
- Department of Mechanical Engineering, Texas Tech University, Lubbock, TX, 79409, United States
- Texas Center for Comparative Cancer Research (TC3R), Amarillo, Texas, 79106, United States
| | - Jenifer Gómez-Pastora
- Department of Chemical Engineering, Texas Tech University, Lubbock, TX, 79409, United States
| | - Kai Wu
- Department of Electrical and Computer Engineering, Texas Tech University, Lubbock, TX, 79409, United States
| |
Collapse
|
7
|
Bezze A, Mattioda C, Ciardelli G, Mattu C. Harnessing cells to improve transport of nanomedicines. Eur J Pharm Biopharm 2024; 203:114446. [PMID: 39122052 DOI: 10.1016/j.ejpb.2024.114446] [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: 06/07/2024] [Revised: 07/18/2024] [Accepted: 08/05/2024] [Indexed: 08/12/2024]
Abstract
Efficient tumour treatment is hampered by the poor selectivity of anticancer drugs, resulting in scarce tumour accumulation and undesired off-target effects. Nano-sized drug-delivery systems in the form of nanoparticles (NPs) have been proposed to improve drug distribution to solid tumours, by virtue of their ability of passive and active tumour targeting. Despite these advantages, literature studies indicated that less than 1% of the administered NPs can successfully reach the tumour mass, highlighting the necessity for more efficient drug transporters in cancer treatment. Living cells, such as blood cells, circulating immune cells, platelets, and stem cells, are often found as an infiltrating component in most solid tumours, because of their ability to naturally circumvent immune recognition, bypass biological barriers, and reach inaccessible tissues through innate tropism and active motility. Therefore, the tumour-homing ability of these cells can be harnessed to design living cell carriers able to improve the transport of drugs and NPs to tumours. Albeit promising, this approach is still in its beginnings and suffers from difficult scalability, high cost, and poor reproducibility. In this review, we present an overview of the most common cell transporters of drugs and NPs, and we discuss how different cell types interact with biological barriers to deliver cargoes of various natures to tumours. Finally, we analyse the different techniques used to load drugs or NPs in living cells and discuss their advantages and disadvantages.
Collapse
Affiliation(s)
- Andrea Bezze
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Carlotta Mattioda
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Gianluca Ciardelli
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Clara Mattu
- Politecnico di Torino - DIMEAS, C.so Duca degli Abruzzi 24, 10129 Torino, Italy.
| |
Collapse
|
8
|
Duan M, Cao R, Yang Y, Chen X, Liu L, Ren B, Wang L, Goh BC. Blood-Brain Barrier Conquest in Glioblastoma Nanomedicine: Strategies, Clinical Advances, and Emerging Challenges. Cancers (Basel) 2024; 16:3300. [PMID: 39409919 PMCID: PMC11475686 DOI: 10.3390/cancers16193300] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is a prevalent type of malignancy within the central nervous system (CNS) that is associated with a poor prognosis. The standard treatment for GBM includes the surgical resection of the tumor, followed by radiotherapy and chemotherapy; yet, despite these interventions, overall treatment outcomes remain suboptimal. The blood-brain barrier (BBB), which plays a crucial role in maintaining the stability of brain tissue under normal physiological conditions of the CNS, also poses a significant obstacle to the effective delivery of therapeutic agents to GBMs. Recent preclinical studies have demonstrated that nanomedicine delivery systems (NDDSs) offer promising results, demonstrating both effective GBM targeting and safety, thereby presenting a potential solution for targeted drug delivery. In this review, we first explore the various strategies employed in preclinical studies to overcome the BBB for drug delivery. Subsequently, the results of the clinical translation of NDDSs are summarized, highlighting the progress made. Finally, we discuss potential strategies for advancing the development of NDDSs and accelerating their translational research through well-designed clinical trials in GBM therapy.
Collapse
Affiliation(s)
- Mengyun Duan
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Ruina Cao
- Department of Anesthesiology, Maternal and Child Health Hospital of Hubei Province, Wuhan 430070, China;
| | - Yuan Yang
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430060, China;
| | - Xiaoguang Chen
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lian Liu
- Department of Pharmacology, Health Science Center, Yangtze University, Jingzhou 434023, China;
| | - Boxu Ren
- Department of Medical Imaging, Health Science Center, Yangtze University, Jingzhou 434023, China; (M.D.); (X.C.)
| | - Lingzhi Wang
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| | - Boon-Cher Goh
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore;
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117600, Singapore
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
- Department of Haematology-Oncology, National University Cancer Institute, Singapore 119228, Singapore
| |
Collapse
|
9
|
Hao X, Gai W, Zhang Y, Zhao D, Zhou W, Feng Y. Peptide functionalized biomimetic gene complexes enhance specificity for vascular endothelial regeneration. Colloids Surf B Biointerfaces 2024; 241:114020. [PMID: 38878659 DOI: 10.1016/j.colsurfb.2024.114020] [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: 03/22/2024] [Revised: 05/28/2024] [Accepted: 06/06/2024] [Indexed: 07/29/2024]
Abstract
Gene delivery presents great potential in endothelium regeneration and prevention of vascular diseases, but its outcome is inevitably limited by high shear stress and instable microenvironment. Highly efficient nanosystems may alleviate the problem with strong dual-specificity for diseased site and targeted cells. Hence, biomimetic coatings incorporating EC-targeting peptides were constructed by platelets and endothelial cells (ECs) for surface modification. A series of biomimetic gene complexes were fabricated by the biomimetic coatings to deliver pcDNA3.1-VEGF165 plasmid (pVEGF) for rapid recovery of endothelium. The gene complexes possessed good biocompatibility with macrophages, stability with serum and showed no evident cytotoxicity for ECs even at very high concentrations. Furthermore, the peptide modified gene complexes achieved selective internalization in ECs and significant accumulation in endothelium-injured site, especially the REDV-modified and EC-derived gene complexes. They substantially enhanced VEGF expression at mRNA and protein levels, thereby enabling a wound to heal completely within 24 h according to wound healing assay. In an artery endothelium-injured mouse model, the REDV-modified and EC-derived gene complexes presented efficient re-endothelialization with the help of enhanced specificity. The biomimetic gene complexes offer an efficient dual-targeting strategy for rapid recovery of endothelium, and hold potential in vascular tissue regeneration.
Collapse
Affiliation(s)
- Xuefang Hao
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, China.
| | - Weiwei Gai
- College of Animal Science and Technology, Inner Mongolia Minzu University, Tongliao 028000, China
| | - Yanping Zhang
- Dalian Institute of Chemical Physics, Chinese Academy of Sciences, Dalian 116023, China
| | - Dandan Zhao
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, China
| | - Weitong Zhou
- State Key Laboratory of New Pharmaceutical Preparations and Excipients, Key Laboratory of Medicinal Chemistry and Molecular Diagnosis of Ministry of Education, Hebei University, Baoding 071002, China
| | - Yakai Feng
- School of Chemical Engineering and Technology, Tianjin University, Yaguan Road 135, Tianjin 300350, China; Frontiers Science Center for Synthetic Biology, Tianjin University, Weijin Road 92, Tianjin 300072, China; Key Laboratory of Systems Bioengineering (Ministry of Education), Tianjin University, Tianjin 300072, China.
| |
Collapse
|
10
|
Yu T, Wang J, Zhou Y, Ma C, Bai R, Huang C, Wang S, Liu K, Han B. Harnessing Engineered Extracellular Vesicles from Mesenchymal Stem Cells as Therapeutic Scaffolds for Bone‐Related Diseases. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202402861] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Indexed: 10/05/2024]
Abstract
AbstractMesenchymal stem cells (MSCs) play a crucial role in maintaining bone homeostasis and are extensively explored for cell therapy in various bone‐related diseases. In addition to direct cell therapy, the secretion of extracellular vesicles (EVs) by MSCs has emerged as a promising alternative approach. MSC‐derived EVs (MSC‐EVs) offer equivalent therapeutic efficacy to MSCs while mitigating potential risks. These EVs possess unique properties that enable them to traverse biological barriers and deliver bioactive cargos to target cells. Furthermore, by employing modification and engineering strategies, the therapeutic effects and tissue targeting specificity of MSC‐EVs can be further enhanced to meet specific therapeutic needs. In this review, the mechanisms and advantages of MSC‐EV therapy in diseased bone tissues are highlighted. Through simple isolation and modification techniques, MSC‐EV‐based biomaterials have demonstrated great promise for bone regeneration. Finally, future perspectives on MSC‐EV therapy are presented, envisioning the development of next‐generation regenerative materials and bioactive agents for clinical translation in the field of bone regeneration.
Collapse
Affiliation(s)
- Tingting Yu
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Jingwei Wang
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Yusai Zhou
- School of Materials Science and Engineering Beihang University Beijing 100191 P. R. China
| | - Chao Ma
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 P. R. China
| | - Rushui Bai
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Cancan Huang
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| | - Shidong Wang
- Musculoskeletal Tumor Center Peking University People's Hospital No.11 Xizhimen South St. Beijing 100044 P. R. China
| | - Kai Liu
- Engineering Research Center of Advanced Rare Earth Materials (Ministry of Education) Department of Chemistry Tsinghua University Beijing 100084 P. R. China
| | - Bing Han
- Department of Orthodontics Cranial‐Facial Growth and Development Center Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
- National Center for Stomatology National Clinical Research Center for Oral Diseases National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing Key Laboratory for Digital Stomatology NMPA Key Laboratory for Dental Materials NHC Key Laboratory of Digital Stomatology Peking University School and Hospital of Stomatology 22 Zhongguancun South Avenue, Haidian District Beijing 100081 P. R. China
| |
Collapse
|
11
|
Misra R, Sanjana Sharath N. Red blood cells based nanotheranostics: A smart biomimetic approach for fighting against cancer. Int J Pharm 2024; 661:124401. [PMID: 38986966 DOI: 10.1016/j.ijpharm.2024.124401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/12/2024]
Abstract
The technique of engineering drug delivery vehicles continues to develop, which bring enhancements in working more efficiently and minimizing side effects to make it more effective and safer. The intense capability of therapeutic agents to remain undamaged in a harsh extracellular environment is helpful to the success of drug development efforts. With this in mind, alterations of biopharmaceuticals with enhanced stability and decreased immunogenicity have been an increasingly active focus of such efforts. Red blood cells (RBCs), also known as erythrocytes have undergone extensive scrutiny as potential vehicles for drug delivery due to their remarkable attributes over the years of research. These include intrinsic biocompatibility, minimal immunogenicity, flexibility, and prolonged systemic circulation. Throughout the course of investigation, a diverse array of drug delivery platforms based on RBCs has emerged. These encompass genetically engineered RBCs, non-genetically modified RBCs, and RBC membrane-coated nanoparticles, each devised to cater to a range of biomedical objectives. Given their prevalence in the circulatory system, RBCs have gained significant attention for their potential to serve as biomimetic coatings for artificial nanocarriers. By virtue of their surface emulation capabilities and customizable core materials, nanocarriers mimicking these RBCs, hold considerable promise across a spectrum of applications, spanning drug delivery, imaging, phototherapy, immunomodulation, sensing, and detection. These multifaceted functionalities underscore the considerable therapeutic and diagnostic potential across various diseases. Our proposed review provides the synthesis of recent strides in the theranostic utilization of erythrocytes in the context of cancer. It also delves into the principal challenges and prospects intrinsic to this realm of research. The focal point of this review pertains to accentuating the significance of erythrocyte-based theranostic systems in combating cancer. Furthermore, it precisely records the latest and the most specific methodologies for tailoring the attributes of these biomimetic nanoscale formulations, attenuating various discoveries for the treatment and management of cancer.
Collapse
Affiliation(s)
- Ranjita Misra
- Department of Biotechnology, Centre for Research in Pure and Applied Sciences, School of Sciences, Jain (Deemed-to-be University), JC Road, Bengaluru 560027, Karnataka, India.
| | - Naomi Sanjana Sharath
- Department of Biotechnology, Centre for Research in Pure and Applied Sciences, School of Sciences, Jain (Deemed-to-be University), JC Road, Bengaluru 560027, Karnataka, India
| |
Collapse
|
12
|
Le QV, Shim G. Biorobotic Drug Delivery for Biomedical Applications. Molecules 2024; 29:3663. [PMID: 39125066 PMCID: PMC11314275 DOI: 10.3390/molecules29153663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/24/2024] [Accepted: 06/27/2024] [Indexed: 08/12/2024] Open
Abstract
Despite extensive efforts, current drug-delivery systems face biological barriers and difficulties in bench-to-clinical use. Biomedical robotic systems have emerged as a new strategy for drug delivery because of their innovative diminutive engines. These motors enable the biorobots to move independently rather than relying on body fluids. The main components of biorobots are engines controlled by external stimuli, chemical reactions, and biological responses. Many biorobot designs are inspired by blood cells or microorganisms that possess innate swimming abilities and can incorporate living materials into their structures. This review explores the mechanisms of biorobot locomotion, achievements in robotic drug delivery, obstacles, and the perspectives of translational research.
Collapse
Affiliation(s)
- Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City 700000, Vietnam;
| | - Gayong Shim
- School of Systems Biomedical Science, Soongsil University, Seoul 06978, Republic of Korea
- Integrative Institute of Basic Sciences, Soongsil University, Seoul 06978, Republic of Korea
| |
Collapse
|
13
|
Ye Z, Liu J, Liu Y, Zhao Y, Li Z, Xu B, Chen D, Wang B, Wang Q, Shen Y. Hybrid nanopotentiators with dual cascade amplification for glioma combined interventional therapy. J Control Release 2024; 372:95-112. [PMID: 38851536 DOI: 10.1016/j.jconrel.2024.06.016] [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: 09/24/2023] [Revised: 06/02/2024] [Accepted: 06/05/2024] [Indexed: 06/10/2024]
Abstract
Glioma is an aggressive malignant brain tumor with a very poor prognosis for survival. The poor tumor targeting efficiency and tumor microenvironment penetration barrier also as troubles inhibited the effective glioma chemotherapy. Here, we design a core-shell structure cascade amplified hybrid catalytic nanopotentiators CFpAD with DM1 encapsulated to overcome the glioma therapeutic obstacles. NIR laser-based BBB penetrating enhances the tumor accumulation of CFpAD. When CFpAD, as the cascade amplified drug, is treated on the cancer cells, the bomb-like CFpAD releases gold nanoparticles as glucose oxidase (GOx) and ferric oxide nanoparticles (FNPs) as peroxides (POx) after blasting, producing ROS via a cascade amplification for tumor cell apoptosis. Gold nanoparticles can rest CAFs and reduce ECM secretion, achieving deep penetration of CFpAD. Moreover, CFpAD also cuts off the nutritional supply of the tumor, reduces the pH value, and releases free radicals to destroy the cancer. The glioma cell viability was significantly decreased through DNA damage and ROS aggregation due to the DM1-based chemotherapy synergistically combined with interventional photothermal therapy (IPTT) and radiotherapy (RT). This domino cascade amplified loop, combined with starvation therapy with IPTT and RT, has good tumor penetration and outstanding antitumor efficacy, and is a promising glioma treatment system.
Collapse
Affiliation(s)
- Zixuan Ye
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Ji Liu
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yanyan Liu
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Yan Zhao
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Zhen Li
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China
| | - Bohui Xu
- School of Pharmacy, Nantong University, No.19 Qixiu Road, Nantong 226001,China
| | - Daquan Chen
- School of Pharmacy, Yantai University, 30 Qingquan Road, Yantai 264005, China
| | - Buhai Wang
- Cancer Institute of Northern Jiangsu People's Hospital Affiliated to Yangzhou University, Yangzhou 225000, China.
| | - Qiyue Wang
- School of Pharmaceutical Science, Nanjing Tech University, Nanjing 211816, China.
| | - Yan Shen
- Department of Pharmaceutics, State Key Laboratory of Nature Medicines, China Pharmaceutical University, 24 Tong Jia Xiang, Nanjing 210009, China.
| |
Collapse
|
14
|
Ma Y, Yi J, Ruan J, Ma J, Yang Q, Zhang K, Zhang M, Zeng G, Jin L, Huang X, Li J, Yang H, Wu W, Sun D. Engineered Cell Membrane-Coated Nanoparticles: New Strategies in Glioma Targeted Therapy and Immune Modulation. Adv Healthc Mater 2024; 13:e2400514. [PMID: 38652681 DOI: 10.1002/adhm.202400514] [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: 02/08/2024] [Revised: 04/09/2024] [Indexed: 04/25/2024]
Abstract
Gliomas, the most prevalent primary brain tumors, pose considerable challenges due to their heterogeneity, intricate tumor microenvironment (TME), and blood-brain barrier (BBB), which restrict the effectiveness of traditional treatments like surgery and chemotherapy. This review provides an overview of engineered cell membrane technologies in glioma therapy, with a specific emphasis on targeted drug delivery and modulation of the immune microenvironment. This study investigates the progress in engineered cell membranes, encompassing physical, chemical, and genetic alterations, to improve drug delivery across the BBB and effectively target gliomas. The examination focuses on the interaction of engineered cell membrane-coated nanoparticles (ECM-NPs) with the TME in gliomas, emphasizing their potential to modulate glioma cell behavior and TME to enhance therapeutic efficacy. The review further explores the involvement of ECM-NPs in immunomodulation techniques, highlighting their impact on immune reactions. While facing obstacles related to membrane stability and manufacturing scalability, the review outlines forthcoming research directions focused on enhancing membrane performance. This review underscores the promise of ECM-NPs in surpassing conventional therapeutic constraints, proposing novel approaches for efficacious glioma treatment.
Collapse
Affiliation(s)
- Yilei Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Jia Yi
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jing Ruan
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325000, China
| | - Kun Zhang
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Maolan Zhang
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Guoming Zeng
- Chongqing Engineering Laboratory of Nano/Micro Biological Medicine Detection Technology, Chongqing University of Science and Technology, Chongqing, 401331, China
| | - Libo Jin
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
| | - Xiaobei Huang
- Chongqing Institute of Green and Intelligent Technology, Chinese Academy of Sciences, Chongqing, 400714, China
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu, 610065, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Haifeng Yang
- JinFeng Laboratory, Chongqing, 401329, China
- Department of Neuro-Oncology, Chongqing University Cancer Hospital, Chongqing, 400044, China
| | - Wei Wu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
- JinFeng Laboratory, Chongqing, 401329, China
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou, 325035, China
- Key Lab of Biohealth Materials and Chemistry of Wenzhou, Wenzhou University, Wenzhou, 325035, China
- JinFeng Laboratory, Chongqing, 401329, China
| |
Collapse
|
15
|
Guo Q, Qian ZM. Macrophage based drug delivery: Key challenges and strategies. Bioact Mater 2024; 38:55-72. [PMID: 38699242 PMCID: PMC11061709 DOI: 10.1016/j.bioactmat.2024.04.004] [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: 12/11/2023] [Revised: 03/14/2024] [Accepted: 04/05/2024] [Indexed: 05/05/2024] Open
Abstract
As a natural immune cell and antigen presenting cell, macrophages have been studied and engineered to treat human diseases. Macrophages are well-suited for use as drug carriers because of their biological characteristics, such as excellent biocompatibility, long circulation, intrinsic inflammatory homing and phagocytosis. Meanwhile, macrophages' uniquely high plasticity and easy re-education polarization facilitates their use as part of efficacious therapeutics for the treatment of inflammatory diseases or tumors. Although recent studies have demonstrated promising advances in macrophage-based drug delivery, several challenges currently hinder further improvement of therapeutic effect and clinical application. This article focuses on the main challenges of utilizing macrophage-based drug delivery, from the selection of macrophage sources, drug loading, and maintenance of macrophage phenotypes, to drug migration and release at target sites. In addition, corresponding strategies and insights related to these challenges are described. Finally, we also provide perspective on shortcomings on the road to clinical translation and production.
Collapse
Affiliation(s)
- Qian Guo
- Laboratory of Drug Delivery, School of Medicine, Shanghai University, 99 Shangda Road, Shanghai, 200444, China
| | - Zhong-Ming Qian
- Institute of Translational and Precision Medicine, Nantong University, 19 Qi Xiu Road, Nantong, Jiangsu, 226019, China
- National Clinical Research Center for Aging and Medicine of Huashan Hospital, Fudan University, Shanghai, 201203, China
| |
Collapse
|
16
|
Kang W, Xu Z, Lu H, Liu S, Li J, Ding C, Lu Y. Advances in biomimetic nanomaterial delivery systems: harnessing nature's inspiration for targeted drug delivery. J Mater Chem B 2024; 12:7001-7019. [PMID: 38919030 DOI: 10.1039/d4tb00565a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/27/2024]
Abstract
The properties of nanomaterials make them promising and advantageous for use in drug delivery systems, but challenges arise from the immune system's recognition of exogenous nanoparticles, leading to their clearance and reduced targeting efficiency. Drawing inspiration from nature, this paper explores biomimetic strategies to transform recognizable nanomaterials into a "camouflaged state." The focal point of this paper is the exploration of bionic nanoparticles, with a focus on cell membrane-coated nanoparticles. These biomimetic structures, particularly those mimicking red blood cells (RBCs), white blood cells (WBCs), platelets, and cancer cells, demonstrate enhanced drug delivery efficiency and prolonged circulation. This article underscores the versatility of these biomimetic structures across diverse diseases and explores the use of hybrid cell membrane-coated nanoparticles as a contemporary trend. This review also investigated exosomes and protein bionic nanoparticles, emphasizing their potential for specific targeting, immune evasion, and improved therapeutic outcomes. We expect that this continued development based on biomimetic nanomaterials will contribute to the efficiency and safety of disease treatment.
Collapse
Affiliation(s)
- Weiqi Kang
- Guangyuan Central Hospital, Guangyuan 628000, P. R. China.
| | - Zhe Xu
- Guangyuan Central Hospital, Guangyuan 628000, P. R. China.
| | - Haiying Lu
- Guangyuan Central Hospital, Guangyuan 628000, P. R. China.
| | - Siwei Liu
- Guangyuan Central Hospital, Guangyuan 628000, P. R. China.
| | - Jianshu Li
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Chunmei Ding
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials Engineering, Sichuan University, Chengdu 610065, P. R. China.
| | - Yongping Lu
- Guangyuan Central Hospital, Guangyuan 628000, P. R. China.
| |
Collapse
|
17
|
Branco F, Cunha J, Mendes M, Vitorino C, Sousa JJ. Peptide-Hitchhiking for the Development of Nanosystems in Glioblastoma. ACS NANO 2024; 18:16359-16394. [PMID: 38861272 PMCID: PMC11223498 DOI: 10.1021/acsnano.4c01790] [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: 02/05/2024] [Revised: 05/15/2024] [Accepted: 05/23/2024] [Indexed: 06/12/2024]
Abstract
Glioblastoma (GBM) remains the epitome of aggressiveness and lethality in the spectrum of brain tumors, primarily due to the blood-brain barrier (BBB) that hinders effective treatment delivery, tumor heterogeneity, and the presence of treatment-resistant stem cells that contribute to tumor recurrence. Nanoparticles (NPs) have been used to overcome these obstacles by attaching targeting ligands to enhance therapeutic efficacy. Among these ligands, peptides stand out due to their ease of synthesis and high selectivity. This article aims to review single and multiligand strategies critically. In addition, it highlights other strategies that integrate the effects of external stimuli, biomimetic approaches, and chemical approaches as nanocatalytic medicine, revealing their significant potential in treating GBM with peptide-functionalized NPs. Alternative routes of parenteral administration, specifically nose-to-brain delivery and local treatment within the resected tumor cavity, are also discussed. Finally, an overview of the significant obstacles and potential strategies to overcome them are discussed to provide a perspective on this promising field of GBM therapy.
Collapse
Affiliation(s)
- Francisco Branco
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Joana Cunha
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
| | - Maria Mendes
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - Carla Vitorino
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| | - João J. Sousa
- Faculty
of Pharmacy, University of Coimbra, Pólo das Ciências
da Saúde, Azinhaga de Santa Comba, 3000-548 Coimbra, Portugal
- Coimbra
Chemistry Centre, Institute of Molecular Sciences − IMS, Faculty
of Sciences and Technology, University of
Coimbra, 3004-535 Coimbra, Portugal
| |
Collapse
|
18
|
Jia G, Wang J, Wang H, Hu X, Long F, Yuan C, Liang C, Wang F. New insights into red blood cells in tumor precision diagnosis and treatment. NANOSCALE 2024; 16:11863-11878. [PMID: 38841898 DOI: 10.1039/d4nr01454e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2024]
Abstract
Red blood cells (RBCs), which function as material transporters in organisms, are rich in materials that are exchanged with metabolically active tumor cells. Recent studies have demonstrated that tumor cells can regulate biological changes in RBCs, including influencing differentiation, maturation, and morphology. RBCs play an important role in tumor development and immune regulation. Notably, the novel scientific finding that RBCs absorb fragments of tumor-carrying DNA overturns the conventional wisdom that RBCs do not contain nucleic acids. RBC membranes are excellent biomimetic materials with significant advantages in terms of their biocompatibility, non-immunogenicity, non-specific adsorption resistance, and biodegradability. Therefore, RBCs provide a new research perspective for the development of tumor liquid biopsies, molecular imaging, drug delivery, and other tumor precision diagnosis and treatment technologies.
Collapse
Affiliation(s)
- Gaihua Jia
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Jun Wang
- Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, China.
| | - Hu Wang
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Xin Hu
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Fei Long
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Chunhui Yuan
- Department of Laboratory Medicine, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430016, China.
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Chen Liang
- Department of Radiation and Medical Oncology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| | - Fubing Wang
- Department of Laboratory Medicine, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
- Center for Single-Cell Omics and Tumor Liquid Biopsy, Zhongnan Hospital of Wuhan University, Wuhan 430071, China.
| |
Collapse
|
19
|
Susa F, Arpicco S, Pirri CF, Limongi T. An Overview on the Physiopathology of the Blood-Brain Barrier and the Lipid-Based Nanocarriers for Central Nervous System Delivery. Pharmaceutics 2024; 16:849. [PMID: 39065547 PMCID: PMC11279990 DOI: 10.3390/pharmaceutics16070849] [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: 05/11/2024] [Revised: 06/12/2024] [Accepted: 06/20/2024] [Indexed: 07/28/2024] Open
Abstract
The state of well-being and health of our body is regulated by the fine osmotic and biochemical balance established between the cells of the different tissues, organs, and systems. Specific districts of the human body are defined, kept in the correct state of functioning, and, therefore, protected from exogenous or endogenous insults of both mechanical, physical, and biological nature by the presence of different barrier systems. In addition to the placental barrier, which even acts as a linker between two different organisms, the mother and the fetus, all human body barriers, including the blood-brain barrier (BBB), blood-retinal barrier, blood-nerve barrier, blood-lymph barrier, and blood-cerebrospinal fluid barrier, operate to maintain the physiological homeostasis within tissues and organs. From a pharmaceutical point of view, the most challenging is undoubtedly the BBB, since its presence notably complicates the treatment of brain disorders. BBB action can impair the delivery of chemical drugs and biopharmaceuticals into the brain, reducing their therapeutic efficacy and/or increasing their unwanted bioaccumulation in the surrounding healthy tissues. Recent nanotechnological innovation provides advanced biomaterials and ad hoc customized engineering and functionalization methods able to assist in brain-targeted drug delivery. In this context, lipid nanocarriers, including both synthetic (liposomes, solid lipid nanoparticles, nanoemulsions, nanostructured lipid carriers, niosomes, proniosomes, and cubosomes) and cell-derived ones (extracellular vesicles and cell membrane-derived nanocarriers), are considered one of the most successful brain delivery systems due to their reasonable biocompatibility and ability to cross the BBB. This review aims to provide a complete and up-to-date point of view on the efficacy of the most varied lipid carriers, whether FDA-approved, involved in clinical trials, or used in in vitro or in vivo studies, for the treatment of inflammatory, cancerous, or infectious brain diseases.
Collapse
Affiliation(s)
- Francesca Susa
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| | - Candido Fabrizio Pirri
- Department of Applied Science and Technology, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy; (F.S.); (C.F.P.)
| | - Tania Limongi
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, 10125 Turin, Italy;
| |
Collapse
|
20
|
Hu W, Yang L, Liao H, Sun D, Ouyang XK, Wang N, Yang G. Disulfiram-loaded CuO 2 nanocarriers for enhanced synergistic chemodynamic chemotherapy. J Colloid Interface Sci 2024; 674:9-18. [PMID: 38908062 DOI: 10.1016/j.jcis.2024.06.147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2024] [Revised: 06/07/2024] [Accepted: 06/20/2024] [Indexed: 06/24/2024]
Abstract
Disulfiram (DSF) metabolites exhibit antitumor properties when bound to Cu2+. This combination also promotes the generation of reactive oxygen species (ROS), ultimately leading to tumor cell death. In this study, CuO2 served as a carrier for DSF, forming a dual-drug delivery system with Cu2+ and DSF encapsulated in polydopamine (PDA). In the final delivery system, CuO2 (DSF-CuO2@PDA) was hydrolyzed at the tumor site, releasing both Cu2+ and H2O2. Cu2+ reacts with DSF metabolites to form Bis(diethyldithiocarbamate)-Cu (CuET), which triggers a Fenton-like reaction that generates ROS. Chemotherapy and chemodynamic therapy exhibited significant tumor-suppressive capabilities, with an inhibition rate of 61 %. In addition, the DSF-CuO2@PDA complex demonstrated superlative tumor-targeting ability and biocompatibility.
Collapse
Affiliation(s)
- Wei Hu
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, PR China
| | - Lianlian Yang
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, PR China
| | - Hongtao Liao
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Wenzhou Medical University, Zhoushan 316000, PR China
| | - Deguan Sun
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Wenzhou Medical University, Zhoushan 316000, PR China
| | - Xiao-Kun Ouyang
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, PR China.
| | - Nan Wang
- School of Food and Pharmacy, Zhejiang Ocean University, Zhoushan 316022, PR China.
| | - Guocai Yang
- Department of Cardiothoracic Surgery, Zhoushan Hospital, Wenzhou Medical University, Zhoushan 316000, PR China.
| |
Collapse
|
21
|
Chi S, Wang C, Liu Z. Biomimetic Nanocomposites for Glioma Imaging and Therapy. Chemistry 2024; 30:e202304338. [PMID: 38538540 DOI: 10.1002/chem.202304338] [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: 12/27/2023] [Indexed: 04/24/2024]
Abstract
Glioma, the most common primary brain tumor, is highly invasive and grows rapidly. As such, the survival of glioma patients is relatively short, highlighting the vital importance of timely diagnosis and treatment of glioma. However, the blood brain barrier (BBB) and the non-targeting delivery systems of contrast agents and drugs greatly hinder the effective glioma imaging and therapy. Fortunately, in recent years, investigators have constructed various biomimetic delivery platforms utilizing the exceptional advantages of biomimetic nanocomposites, such as immune evasion, homologous targeting ability, and BBB penetrating ability, to achieve efficient and precise delivery of substances to glioma sites for improved diagnosis and treatment. In this concept, we present the application of these biomimetic nanocomposites in fluorescence imaging (FI), magnetic resonance imaging (MRI), and multi-modal imaging, as well as in chemotherapy, phototherapy, and combined therapy for glioma. Lastly, we provide our perspective on this research field.
Collapse
Affiliation(s)
- Siyu Chi
- College of Health Science and Engineering, Hubei University, Wuhan, 430062, P. R. China
| | - Caixia Wang
- College of Health Science and Engineering, Hubei University, Wuhan, 430062, P. R. China
| | - Zhihong Liu
- College of Health Science and Engineering, Hubei University, Wuhan, 430062, P. R. China
| |
Collapse
|
22
|
Liu C, Xu X, Chen Y, Yin M, Mäkilä E, Zhou W, Su W, Zhang H. Metabolism-Regulating Nanozyme System for Advanced Nanocatalytic Cancer Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2307794. [PMID: 38168483 DOI: 10.1002/smll.202307794] [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: 09/07/2023] [Revised: 12/17/2023] [Indexed: 01/05/2024]
Abstract
Nanocatalytic therapy, an emerging approach in cancer treatment, utilizes nanomaterials to initiate enzyme-mimetic catalytic reactions within tumors, inducing tumor-suppressive effects. However, the targeted and selective catalysis within tumor cells is challenging yet critical for minimizing the adverse effects. The distinctive reliance of tumor cells on glycolysis generates abundant lactate, influencing the tumor's pH, which can be manipulated to selectively activate nanozymatic catalysis. Herein, small interfering ribonucleic acid (siRNA) targeting lactate transporter-mediated efflux is encapsulated within the iron-based metal-organic framework (FeMOF) and specifically delivered to tumor cells through cell membrane coating. This approach traps lactate within the cell, swiftly acidifying the tumor cytoplasm and creating an environment for boosting the catalysis of the FeMOF nanozyme. The nanozyme generates hydroxyl radical (·OH) in the reversed acidic environment, using endogenous hydrogen peroxide (H2O2) produced by mitochondria as a substrate. The induced cytoplasmic acidification disrupts calcium homeostasis, leading to mitochondrial calcium overload, resulting in mitochondrial dysfunction and subsequent tumor cell death. Additionally, the tumor microenvironment is also remodeled, inhibiting migration and invasion, thus preventing metastasis. This groundbreaking strategy combines metabolic regulation with nanozyme catalysis in a toxic drug-free approach for tumor treatment, holding promise for future clinical applications.
Collapse
Affiliation(s)
- Chang Liu
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
| | - Xiaoyu Xu
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
- ENT Institute and Department of Otorhinolaryngology, Eye & ENT Hospital, State Key Laboratory of Medical Neurobiology, Institutes of Biomedical Sciences, Fudan University, Shanghai, 200031, China
| | - Yongyang Chen
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Miao Yin
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Ermei Mäkilä
- Industrial Physics Laboratory, Department of Physics and Astronomy, University of Turku, Turku, 20014, Finland
| | - Wenhui Zhou
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| | - Wenmei Su
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
| | - Hongbo Zhang
- Department of Pulmonary Oncology, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-communicable Diseases, Affiliated Hospital of Guangdong Medical University, Zhanjiang, 524001, China
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, Turku, 20520, Finland
- Turku Bioscience Centre, University of Turku and Åbo Akademi University, Turku, 20520, Finland
| |
Collapse
|
23
|
Huang D, Wang X, Wang W, Li J, Zhang X, Xia B. Cell-membrane engineering strategies for clinic-guided design of nanomedicine. Biomater Sci 2024; 12:2865-2884. [PMID: 38686665 DOI: 10.1039/d3bm02114a] [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: 05/02/2024]
Abstract
Cells are the fundamental units of life. The cell membrane primarily composed of two layers of phospholipids (a bilayer) structurally defines the boundary of a cell, which can protect its interior from external disturbances and also selectively exchange substances and conduct signals from the extracellular environment. The complexity and particularity of transmembrane proteins provide the foundation for versatile cellular functions. Nanomedicine as an emerging therapeutic strategy holds tremendous potential in the healthcare field. However, it is susceptible to recognition and clearance by the immune system. To overcome this bottleneck, the technology of cell membrane coating has been extensively used in nanomedicines for their enhanced therapeutic efficacy, attributed to the favorable fluidity and biocompatibility of cell membranes with various membrane-anchored proteins. Meanwhile, some engineering strategies of cell membranes through various chemical, physical and biological ways have been progressively developed to enable their versatile therapeutic functions against complex diseases. In this review, we summarized the potential clinical applications of four typical cell membranes, elucidated their underlying therapeutic mechanisms, and outlined their current engineering approaches. In addition, we further discussed the limitation of this technology of cell membrane coating in clinical applications, and possible solutions to address these challenges.
Collapse
Affiliation(s)
- Di Huang
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | - Xiaoyu Wang
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | - Wentao Wang
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | - Jiachen Li
- Department of Biomedical Engineering, W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen/University of Groningen, Ant. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Xiaomei Zhang
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
| | - Bing Xia
- College of Science, State Key Laboratory of Tree Genetics and Breeding, Nanjing Forestry University, Nanjing 210037, P. R. China.
- Department of Geriatric Oncology, Affiliated Nanjing Drum Tower Hospital of Nanjing University Medical School, Nanjing 210008, P. R. China
| |
Collapse
|
24
|
Blaya-Cánovas JL, Griñán-Lisón C, Blancas I, Marchal JA, Ramírez-Tortosa C, López-Tejada A, Benabdellah K, Cortijo-Gutiérrez M, Cano-Cortés MV, Graván P, Navarro-Marchal SA, Gómez-Morales J, Delgado-Almenta V, Calahorra J, Agudo-Lera M, Sagarzazu A, Rodríguez-González CJ, Gallart-Aragón T, Eich C, Sánchez-Martín RM, Granados-Principal S. Autologous patient-derived exhausted nano T-cells exploit tumor immune evasion to engage an effective cancer therapy. Mol Cancer 2024; 23:83. [PMID: 38730475 PMCID: PMC11084007 DOI: 10.1186/s12943-024-01997-x] [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: 01/18/2024] [Accepted: 04/05/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Active targeting by surface-modified nanoplatforms enables a more precise and elevated accumulation of nanoparticles within the tumor, thereby enhancing drug delivery and efficacy for a successful cancer treatment. However, surface functionalization involves complex procedures that increase costs and timelines, presenting challenges for clinical implementation. Biomimetic nanoparticles (BNPs) have emerged as unique drug delivery platforms that overcome the limitations of actively targeted nanoparticles. Nevertheless, BNPs coated with unmodified cells show reduced functionalities such as specific tumor targeting, decreasing the therapeutic efficacy. Those challenges can be overcome by engineering non-patient-derived cells for BNP coating, but these are complex and cost-effective approaches that hinder their wider clinical application. Here we present an immune-driven strategy to improve nanotherapeutic delivery to tumors. Our unique perspective harnesses T-cell exhaustion and tumor immune evasion to develop a groundbreaking new class of BNPs crafted from exhausted T-cells (NExT) of triple-negative breast cancer (TNBC) patients by specific culture methods without sophisticated engineering. METHODS NExT were generated by coating PLGA (poly(lactic-co-glycolic acid)) nanoparticles with TNBC-derived T-cells exhausted in vitro by acute activation. Physicochemical characterization of NExT was made by dynamic light scattering, electrophoretic light scattering and transmission electron microscopy, and preservation and orientation of immune checkpoint receptors by flow cytometry. The efficacy of chemotherapy-loaded NExT was assessed in TNBC cell lines in vitro. In vivo toxicity was made in CD1 mice. Biodistribution and therapeutic activity of NExT were determined in cell-line- and autologous patient-derived xenografts in immunodeficient mice. RESULTS We report a cost-effective approach with a good performance that provides NExT naturally endowed with immune checkpoint receptors (PD1, LAG3, TIM3), augmenting specific tumor targeting by engaging cognate ligands, enhancing the therapeutic efficacy of chemotherapy, and disrupting the PD1/PDL1 axis in an immunotherapy-like way. Autologous patient-derived NExT revealed exceptional intratumor accumulation, heightened chemotherapeutic index and efficiency, and targeted the tumor stroma in a PDL1+ patient-derived xenograft model of triple-negative breast cancer. CONCLUSIONS These advantages underline the potential of autologous patient-derived NExT to revolutionize tailored adoptive cancer nanotherapy and chemoimmunotherapy, which endorses their widespread clinical application of autologous patient-derived NExT.
Collapse
Grants
- PRDJA19001BLAY Fundación Científica Asociación Española Contra el Cáncer
- POSTDOC_21_638 Consejería de Economía, Innovación, Ciencia y Empleo, Junta de Andalucía
- RTI2018.101309B-C22 Ministerio de Ciencia, Innovación y Universidades
- FPU19/04450 Ministerio de Ciencia, Innovación y Universidades
- DOC_01686 Consejería de Transformación Económica, Industria, Conocimiento y Universidades
- PI19/01533 Instituto de Salud Carlos III
- P29/22/02 Consejería de Economía, Conocimiento, Empresas y Universidad, Junta de Andalucía, Spain
Collapse
Affiliation(s)
- José L Blaya-Cánovas
- UGC de Oncología Médica, Hospital Universitario de Jaén, Jaén, 23007, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Carmen Griñán-Lisón
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18100, Spain
| | - Isabel Blancas
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- UGC de Oncología, Hospital Universitario San Cecilio, Granada, 18016, Spain
- Department of Medicine, University of Granada, Granada, 18016, Spain
| | - Juan A Marchal
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18100, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM), University of Granada, Granada, 18100, Spain
- Department of Human Anatomy and Embryology, Faculty of Medicine, University of Granada, Granada, 18016, Spain
| | - César Ramírez-Tortosa
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- UGC de Anatomía Patológica, Hospital San Cecilio, Granada, 18016, Spain
| | - Araceli López-Tejada
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain
| | - Karim Benabdellah
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Marina Cortijo-Gutiérrez
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - M Victoria Cano-Cortés
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry Applied to Biomedicine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain
| | - Pablo Graván
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18100, Spain
- Department of Applied Physics, Faculty of Science, University of Granada, Granada, 18071, Spain
| | - Saúl A Navarro-Marchal
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Excellence Research Unit "Modeling Nature" (MNat), University of Granada, Granada, 18100, Spain
- Biopathology and Regenerative Medicine Institute (IBIMER), Centre for Biomedical Research, (CIBM), University of Granada, Granada, 18100, Spain
- Department of Applied Physics, Faculty of Science, University of Granada, Granada, 18071, Spain
| | - Jaime Gómez-Morales
- Laboratorio de Estudios Cristalográficos IACT-CSIC-UGR, Armilla, 18100, Spain
| | - Violeta Delgado-Almenta
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Jesús Calahorra
- UGC de Oncología Médica, Hospital Universitario de Jaén, Jaén, 23007, Spain
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - María Agudo-Lera
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | - Amaia Sagarzazu
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
| | | | - Tania Gallart-Aragón
- Department of Medicine, University of Granada, Granada, 18016, Spain
- UGC de Cirugía General y del Aparato Digestivo, Hospital Universitario San Cecilio, Granada, 18016, Spain
| | - Christina Eich
- Translational Nanobiomaterials and Imaging, Department of Radiology, Leiden University Medical Center, Leiden, 2333, The Netherlands
| | - Rosario M Sánchez-Martín
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain
- Department of Medicinal & Organic Chemistry and Excellence Research Unit of "Chemistry Applied to Biomedicine and the Environment", Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain
| | - Sergio Granados-Principal
- Instituto de Investigación Biosanitaria ibs.GRANADA, University Hospitals of Granada- University of Granada, Granada, 18100, Spain.
- Centre for Genomics and Oncological Research, GENYO, Pfizer/University of Granada/Andalusian Regional Government, Granada, 18016, Spain.
- Department of Biochemistry and Molecular Biology 2, Faculty of Pharmacy, University of Granada, Campus de Cartuja s/n, Granada, 18071, Spain.
| |
Collapse
|
25
|
Ding Y, Xu Q, Chai Z, Wu S, Xu W, Wang J, Zhou J, Luo Z, Liu Y, Xie C, Lu L, Lu W. All-stage targeted red blood cell membrane-coated docetaxel nanocrystals for glioma treatment. J Control Release 2024; 369:325-334. [PMID: 38565395 DOI: 10.1016/j.jconrel.2024.03.055] [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: 10/23/2023] [Revised: 03/08/2024] [Accepted: 03/29/2024] [Indexed: 04/04/2024]
Abstract
Challenges for glioma treatment with nanomedicines include physio-anatomical barriers (the blood-brain barrier and blood-brain tumor barrier), low drug loading capacity, and limited circulation time. Here, a red blood cell membrane-coated docetaxel drug nanocrystal (pV-RBCm-NC(DTX)), modified with pHA-VAP (pV) for all-stage targeting of glioma, was designed. The NC(DTX) core exhibited a high drug loading capacity but low in vivo stability, and the RBCm coating significantly enhanced the stability and prolonged in vivo circulation. Moreover, the Y-shaped targeting ligand pV was modified by a mild avidin-biotin interaction, which endowed RBCm-NC(DTX) with superior barrier-crossing ability and therapeutic efficacy. The integration of nanocrystal technology, cell membrane coating, and the avidin-biotin insertion method into this active targeting biomimetic formulation represents a promising drug delivery strategy for glioma.
Collapse
Affiliation(s)
- Yuan Ding
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Qianzhu Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Zhilan Chai
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Sunyi Wu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Weixia Xu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jun Wang
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Jianfen Zhou
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Zimiao Luo
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Yu Liu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China
| | - Cao Xie
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd., Shanghai, 201314, China
| | - Linwei Lu
- Department of Integrative Medicine, Huashan Hospital, Fudan University, Shanghai 200040, China; Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China.
| | - Weiyue Lu
- Department of Pharmaceutics, School of Pharmacy, Fudan University & Key Laboratory of Smart Drug Delivery (Fudan University), Ministry of Education, Shanghai 201203, China; Department of Research and Development, Shanghai Tayzen PharmLab Co., Ltd., Shanghai, 201314, China; Institutes of Integrative Medicine, Fudan University, Shanghai 200040, China; Shanghai Engineering Technology Research Center for Pharmaceutical Intelligent Equipment, Shanghai Frontiers Science Center for Druggability of Cardiovascular non-coding RNA, Institute for Frontier Medical Technology, Shanghai University of Engineering Science, Shanghai 201620, China.
| |
Collapse
|
26
|
Wang Y, Wang J, Ye R, Jin Q, Yin F, Liu N, Wang Y, Zhang Q, Gao T, Zhao Y. Cancer Cell-Mimicking Prussian Blue Nanoplatform for Synergistic Mild Photothermal/Chemotherapy via Heat Shock Protein Inhibition. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38624164 DOI: 10.1021/acsami.4c00873] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2024]
Abstract
Combined mild-temperature photothermal/chemotherapy has emerged as a highly promising modality for tumor therapy. However, its therapeutic efficacy is drastically compromised by the heat-induced overexpression of heat shock proteins (HSPs) by the cells, which resist heat stress and apoptosis. The purpose of this study was to downregulate HSPs and enhance the mild-temperature photothermal/chemotherapy effect. In detail, the colon cancer cell membrane (CT26M)-camouflaged HSP90 inhibitor ganetespib and the chemotherapeutic agent doxorubicin (DOX)-coloaded hollow mesoporous Prussian blue (HMPB) nanoplatform (named PGDM) were designed for synergistic mild photothermal/chemotherapy via HSP inhibition. In addition to being a photothermal agent with a high efficiency of photothermal conversion (24.13%), HMPB offers a hollow hole that can be filled with drugs. Concurrently, the cancer cell membrane camouflaging enhances tumor accumulation through a homologous targeting mechanism and gives the nanoplatform the potential to evade the immune system. When exposed to NIR radiation, HMPB's photothermal action (44 °C) not only causes tumor cells to undergo apoptosis but also causes ganetespib to be released on demand. This inhibits the formation of HSP90, which enhances the mild photothermal/chemotherapy effect. The results confirmed that the combined treatment regimen of mild photothermal therapy (PTT) and chemotherapy showed a better therapeutic efficacy than the individual treatment methods. Therefore, this multimodal nanoparticle can advance the development of drugs for the treatment of malignancies, such as colon cancer, and has prospects for clinical application.
Collapse
Affiliation(s)
- Yun Wang
- Department of Gastroenterology, Jiamusi Central Hospital, Jiamus 154003, P. R. China
- Department of Internal Medicine, School of Clinical Medicine, Jiamusi University, Jiamusi 154007, P. R. China
| | - Jinling Wang
- Department of Emergency and Critical Care Center, The Second Affiliated Hospital of Guangdong Medical University, No.12 Minyou Road, Xiashan, Zhanjiang, Guangdong 524003, P. R. China
| | - Roumei Ye
- Department of Pharmacy, Medical College of Guangxi University, Nanning 530004, P. R. China
| | - Quanyi Jin
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, P. R. China
| | - Fengyue Yin
- Department of Pharmacy, Medical College of Guangxi University, Nanning 530004, P. R. China
| | - Nian Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361005, P. R. China
| | - Yubo Wang
- Department of Biomedical Engineering, Medical College of Guangxi University, Nanning 530004, P. R. China
| | - Quan Zhang
- Department of Gastroenterology, Jiamusi Central Hospital, Jiamus 154003, P. R. China
- Department of Internal Medicine, School of Clinical Medicine, Jiamusi University, Jiamusi 154007, P. R. China
| | - Ting Gao
- Department of Pharmaceutical Preparation, General Hospital of Ningxia Medical University, Yinchuan 750004, P. R. China
| | - Yilin Zhao
- Department of Oncology and Vascular Interventional Radiology, Zhongshan Hospital of Xiamen University, Xiamen 361004, P. R. China
| |
Collapse
|
27
|
He X, Wu W, Hu Y, Wu M, Li H, Ding L, Huang S, Fan Y. Visualizing the global trends of peptides in wound healing through an in-depth bibliometric analysis. Int Wound J 2024; 21:e14575. [PMID: 38116897 PMCID: PMC10961903 DOI: 10.1111/iwj.14575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 12/04/2023] [Indexed: 12/21/2023] Open
Abstract
Wound healing is a complicated and multistage biological process for the repair of damaged/injured tissues, which requires intelligent designs to provide comprehensive and convenient treatment. Peptide-based wound dressings have received extensive attention for further development and application due to their excellent biocompatibility and multifunctionality. However, the current lack of intuitive analysis of the development trend and research hotspots of peptides applied in wound healing, as well as detailed elaboration of possible research hotspots, restricted obtaining a comprehensive understanding and development in this field. The present study analysed publications from the Web of Science (WOS) Core Collection database and visualized the hotspots and current trends of peptide research in wound healing. Data between January 1st, 2003, and December 31st, 2022, were collected and subjected to a bibliometric analysis. The countries, institutions, co-authorship, co-citation reference, and co-occurrence of keywords in this subject were examined using VOSviewer and CiteSpace. We provided an intuitive, timely, and logical overview of the development prospects and challenges of peptide application in wound healing and some solutions to the major obstacles, which will help researchers gain insights into the investigation of this promising field.
Collapse
Affiliation(s)
- Xinyan He
- Department of Pharmaceutics, Chongqing University Jiangjin Hospital, Chongqing University, Chongqing, China
| | - Wen Wu
- Chongqing key Laboratory of High Active Traditional Chinese Drug Delivery System, Chongqing Engineering Research Center of Pharmaceutical Sciences, Chongqing Medical and Pharmaceutical College, Chongqing, China
| | - Yuchen Hu
- School of Biological Engineering and Food, Hubei University of Technology, Wuhan, China
| | - Meiling Wu
- Université de Lorraine, CITHEFOR, Nancy, France
| | - Hong Li
- School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Ling Ding
- Department of Pharmaceutics, Chongqing University Jiangjin Hospital, Chongqing University, Chongqing, China
| | - Shiqin Huang
- Department of Pharmaceutics, Chongqing University Jiangjin Hospital, Chongqing University, Chongqing, China
| | - Ying Fan
- Department of Pharmaceutics, Chongqing University Jiangjin Hospital, Chongqing University, Chongqing, China
| |
Collapse
|
28
|
Wang S, Yang L, He W, Zheng M, Zou Y. Cell Membrane Camouflaged Biomimetic Nanoparticles as a Versatile Platform for Brain Diseases Treatment. SMALL METHODS 2024:e2400096. [PMID: 38461538 DOI: 10.1002/smtd.202400096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Revised: 02/27/2024] [Indexed: 03/12/2024]
Abstract
Although there are various advancements in biomedical in the past few decades, there are still challenges in the treatment of brain diseases. The main difficulties are the inability to deliver a therapeutic dose of the drug to the brain through the blood-brain barrier (BBB) and the serious side effects of the drug. Thus, it is essential to select biocompatible drug carriers and novel therapeutic tools to better enhance the effect of brain disease treatment. In recent years, biomimetic nanoparticles (BNPs) based on natural cell membranes, which have excellent biocompatibility and low immunogenicity, are widely used in the treatment of brain diseases to enable the drug to successfully cross the BBB and target brain lesions. BNPs can prolong the circulation time in vivo, are more conducive to drug aggregation in brain lesions. Cell membranes (CMs) from cancer cells (CCs), red blood cells (RBCs), white blood cells (WBCs), and so on are used as biomimetic coatings for nanoparticles (NPs) to achieve the ability to target, evade clearance, or stimulate the immune system. This review summarizes the application of different cell sources as BNPs coatings in the treatment of brain diseases and discusses the possibilities and challenges of clinical translation.
Collapse
Affiliation(s)
- Shiyu Wang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Longfei Yang
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Wenya He
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Meng Zheng
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
| | - Yan Zou
- Henan-Macquarie Uni Joint Centre for Biomedical Innovation, Academy for Advanced Interdisciplinary Studies, Henan Key Laboratory of Brain Targeted Bio-nanomedicine, School of Life Sciences, Henan University, Kaifeng, Henan, 475004, China
- Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, Sydney, NSW, 2109, Australia
| |
Collapse
|
29
|
Huang Y, Ji W, Zhang J, Huang Z, Ding A, Bai H, Peng B, Huang K, Du W, Zhao T, Li L. The involvement of the mitochondrial membrane in drug delivery. Acta Biomater 2024; 176:28-50. [PMID: 38280553 DOI: 10.1016/j.actbio.2024.01.027] [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: 10/10/2023] [Revised: 12/23/2023] [Accepted: 01/18/2024] [Indexed: 01/29/2024]
Abstract
Treatment effectiveness and biosafety are critical for disease therapy. Bio-membrane modification facilitates the homologous targeting of drugs in vivo by exploiting unique antibodies or antigens, thereby enhancing therapeutic efficacy while ensuring biosafety. To further enhance the precision of disease treatment, future research should shift focus from targeted cellular delivery to targeted subcellular delivery. As the cellular powerhouses, mitochondria play an indispensable role in cell growth and regulation and are closely involved in many diseases (e.g., cancer, cardiovascular, and neurodegenerative diseases). The double-layer membrane wrapped on the surface of mitochondria not only maintains the stability of their internal environment but also plays a crucial role in fundamental biological processes, such as energy generation, metabolite transport, and information communication. A growing body of evidence suggests that various diseases are tightly related to mitochondrial imbalance. Moreover, mitochondria-targeted strategies hold great potential to decrease therapeutic threshold dosage, minimize side effects, and promote the development of precision medicine. Herein, we introduce the structure and function of mitochondrial membranes, summarize and discuss the important role of mitochondrial membrane-targeting materials in disease diagnosis/treatment, and expound the advantages of mitochondrial membrane-assisted drug delivery for disease diagnosis, treatment, and biosafety. This review helps readers understand mitochondria-targeted therapies and promotes the application of mitochondrial membranes in drug delivery. STATEMENT OF SIGNIFICANCE: Bio-membrane modification facilitates the homologous targeting of drugs in vivo by exploiting unique antibodies or antigens, thereby enhancing therapeutic efficacy while ensuring biosafety. Compared to cell-targeted treatment, targeting of mitochondria for drug delivery offers higher efficiency and improved biosafety and will promote the development of precision medicine. As a natural material, the mitochondrial membrane exhibits excellent biocompatibility and can serve as a carrier for mitochondria-targeted delivery. This review provides an overview of the structure and function of mitochondrial membranes and explores the potential benefits of utilizing mitochondrial membrane-assisted drug delivery for disease treatment and biosafety. The aim of this review is to enhance readers' comprehension of mitochondrial targeted therapy and to advance the utilization of mitochondrial membrane in drug delivery.
Collapse
Affiliation(s)
- Yinghui Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Wenhui Ji
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Jiaxin Zhang
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Ze Huang
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China; Future Display Institute in Xiamen, Xiamen 361005, China
| | - Aixiang Ding
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China
| | - Hua Bai
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE) and Xi'an Institute of Biomedical Materials & Engineering, Northwestern Polytechnical University, Xi'an 710072, China
| | - Kai Huang
- Future Display Institute in Xiamen, Xiamen 361005, China
| | - Wei Du
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Tingting Zhao
- School of Basic Medical Sciences, Anhui Medical University, Hefei 230032, China.
| | - Lin Li
- The Institute of Flexible Electronics (IFE, Future Technologies), Xiamen University, Xiamen 361005, China; Future Display Institute in Xiamen, Xiamen 361005, China.
| |
Collapse
|
30
|
Guan X, Xing S, Liu Y. Engineered Cell Membrane-Camouflaged Nanomaterials for Biomedical Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:413. [PMID: 38470744 DOI: 10.3390/nano14050413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2023] [Revised: 02/05/2024] [Accepted: 02/07/2024] [Indexed: 03/14/2024]
Abstract
Recent strides in nanomaterials science have paved the way for the creation of reliable, effective, highly accurate, and user-friendly biomedical systems. Pioneering the integration of natural cell membranes into sophisticated nanocarrier architectures, cell membrane camouflage has emerged as a transformative approach for regulated drug delivery, offering the benefits of minimal immunogenicity coupled with active targeting capabilities. Nevertheless, the utility of nanomaterials with such camouflage is curtailed by challenges like suboptimal targeting precision and lackluster therapeutic efficacy. Tailored cell membrane engineering stands at the forefront of biomedicine, equipping nanoplatforms with the capacity to conduct more complex operations. This review commences with an examination of prevailing methodologies in cell membrane engineering, spotlighting strategies such as direct chemical modification, lipid insertion, membrane hybridization, metabolic glycan labeling, and genetic engineering. Following this, an evaluation of the unique attributes of various nanomaterials is presented, delivering an in-depth scrutiny of the substantial advancements and applications driven by cutting-edge engineered cell membrane camouflage. The discourse culminates by recapitulating the salient influence of engineered cell membrane camouflage within nanomaterial applications and prognosticates its seminal role in transformative healthcare technologies. It is envisaged that the insights offered herein will catalyze novel avenues for the innovation and refinement of engineered cell membrane camouflaged nanotechnologies.
Collapse
Affiliation(s)
- Xiyuan Guan
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Kay Lab of Bioorganic Phosphorus Chemistry and Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Simin Xing
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Kay Lab of Bioorganic Phosphorus Chemistry and Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| | - Yang Liu
- Department of Chemistry, Beijing Key Laboratory for Analytical Methods and Instrumentation, Kay Lab of Bioorganic Phosphorus Chemistry and Chemical Biology of Ministry of Education, Tsinghua University, Beijing 100084, China
| |
Collapse
|
31
|
Zhao G, Zhang Y, Xu CF, Wang J. In vivo production of CAR-T cells using virus-mimetic fusogenic nanovesicles. Sci Bull (Beijing) 2024; 69:354-366. [PMID: 38072706 DOI: 10.1016/j.scib.2023.11.055] [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: 05/11/2023] [Revised: 08/20/2023] [Accepted: 11/27/2023] [Indexed: 01/24/2024]
Abstract
Engineered T cells expressing chimeric antigen receptor (CAR) exhibit high response rates in B-cell malignancy treatments and possess therapeutic potentials against various diseases. However, the complicated ex vivo production process of CAR-T cells limits their application. Herein, we use virus-mimetic fusogenic nanovesicles (FuNVs) to produce CAR-T cells in vivo via membrane fusion-mediated CAR protein delivery. Briefly, the FuNVs are modified using T-cell fusogen, adapted from measles virus or reovirus fusogens via displaying anti-CD3 single-chain variable fragment. The FuNVs can efficiently fuse with the T-cell membrane in vivo, thereby delivering the loaded anti-CD19 (αCD19) CAR protein onto T-cells to produce αCD19 CAR-T cells. These αCD19 CAR-T cells alone or in combination with anti-OX40 antibodies can treat B-cell lymphoma without inducing cytokine release syndrome. Thus, our strategy provides a novel method for engineering T cells into CAR-T cells in vivo and can further be employed to deliver other therapeutic membrane proteins.
Collapse
Affiliation(s)
- Gui Zhao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Yue Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China
| | - Cong-Fei Xu
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Biomedical Engineering, South China University of Technology, Guangzhou 510006, China.
| | - Jun Wang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China; National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou 510006, China; Key Laboratory of Biomedical Materials and Engineering of the Ministry of Education, South China University of Technology, Guangzhou 510006, China.
| |
Collapse
|
32
|
Geng F, Fan X, Liu Y, Lu W, Wei G. Recent advances in nanocrystal-based technologies applied for ocular drug delivery. Expert Opin Drug Deliv 2024; 21:211-227. [PMID: 38271023 DOI: 10.1080/17425247.2024.2311119] [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: 10/21/2023] [Accepted: 01/24/2024] [Indexed: 01/27/2024]
Abstract
INTRODUCTION The intricate physiological barriers of the eye and the limited volume of eye drops impede efficient delivery of poorly water-soluble drugs. In the last decade, nanocrystals have emerged as versatile drug delivery systems in various administration routes from bench to bedside. The unique superiorities of nanocrystals, mainly embodied in high drug-loading capacity, good mucosal adhesion and penetration, and greatly improved drug solubility, reveal a promising prospect for ocular delivery of poorly water-soluble drugs. AREAS COVERED This article focuses on the ophthalmic nanocrystal technologies and products that are in the literature, clinical trials, and even on the market. The recent research progress in the preparation, ocular application, and absorption of nanocrystals are highlighted, and the pros and cons of nanocrystals in overcoming the physiological barriers of the eye are also summarized. EXPERT OPINION Nanocrystals have demonstrated success as glucocorticoid eye drops in the treatment of anterior segment diseases. However, the thermodynamic stability of nanocrystals remains the major challenge in product development. New technologies for efficiently optimizing stabilizers and sterilization processes are still expected. Strategies to confer more diverse functions via surface modification are also worth exploration to improve the potential of nanocrystals in delivering poorly water-soluble drugs to posterior segment of the eye.
Collapse
Affiliation(s)
- Feiyang Geng
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Xingyan Fan
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Yu Liu
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
| | - Weiyue Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
- The Institutes of Integrative Medicine of Fudan University, Shanghai, China
| | - Gang Wei
- Key Laboratory of Smart Drug Delivery, Ministry of Education; Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai, China
- The Institutes of Integrative Medicine of Fudan University, Shanghai, China
- Shanghai Engineering Research Center of ImmunoTherapeutics, Shanghai, China
| |
Collapse
|
33
|
Gu J, Yan C, Yin S, Wu H, Liu C, Xue A, Lei X, Zhang N, Geng F. Erythrocyte membrane-coated nanocarriers modified by TGN for Alzheimer's disease. J Control Release 2024; 366:448-459. [PMID: 38128884 DOI: 10.1016/j.jconrel.2023.12.030] [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: 08/10/2023] [Revised: 12/12/2023] [Accepted: 12/18/2023] [Indexed: 12/23/2023]
Abstract
Alzheimer's disease (AD) is an aging-related neurodegenerative disease, and the main pathological feature was β-amyloid protein (Aβ) deposition. Recently, bioactive materials-based drug delivery system has been widely investigated for the treatment of AD. In this study, we developed a red blood cells (RBC) membrane-coated polycaprolactone (PCL) nanoparticles (NPs) loading with a therapeutic agent for AD, curcumin (Cur). A functional peptide TGNYKALHPHN (TGN) was conjugated to the surface of membrane for blood-brain barrier (BBB) transport (TGN-RBC-NPs-Cur). TGN peptide can be recognized by receptors on the BBB and has great potential for brain transport. To confirm the targeted delivery of Cur to the brain, a cell co-culturing immortalized human cerebral microvascular endothelial cells and human brain astrocytes glioblastoma (hCMEC/D3 and U-118MG) in vitro model was established. As a result, the BBB transporting ratio of TGN-RBC-NPs-FITC was 29.64% at 12 h which was approximately eight-fold than RBC-NPs-FITC. The improvement of drug accumulation in the AD lesion was confirmed by the NPs modified with the BBB-penetrating peptide in the fluorescence imaging and quantitative analysis with UPLC-MS/MS in vivo. The neuroprotective effects were evaluated with new object recognition behavioral test, in vitro AD cell model, dendritic spine stain, GFAP and IBA1 immunofluorescence stain. The spatial learning and memory abilities of the AD model mice treated with TGN-RBC-NPs-Cur were obviously enhanced compared with the AD control mice and were also better than Cur at the same dosage. These results were consistent with the values of protection index of rat adrenal pheochromocytoma cells (PC12 cells) treated by Aβ25-35. TGN-RBC-NPs-Cur increased the dendritic segments densities and restrained activation of microglia and astrocytes of AD mice, as well as reversed cognitive function of AD mice. All of the results demonstrated TGN-RBC-NPs-Cur a promising therapeutic strategy for delaying the progression of AD by designing biomimetic nanosystems to deliver drugs into the brain.
Collapse
Affiliation(s)
- Jinlian Gu
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Chang Yan
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Shun Yin
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Hao Wu
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Chi Liu
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China
| | - Ao Xue
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150004, China
| | - Xia Lei
- Wuxi Traditional Chinese Medicine Hospital, Wuxi, Jiangsu 214071, China.
| | - Ning Zhang
- College of Pharmacy, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang 150004, China; Wuxi Traditional Chinese Medicine Hospital, Wuxi, Jiangsu 214071, China.
| | - Fang Geng
- School of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, Heilongjiang 150025, China.
| |
Collapse
|
34
|
Yang Y, Jiang Q, Zhang F. Nanocrystals for Deep-Tissue In Vivo Luminescence Imaging in the Near-Infrared Region. Chem Rev 2024; 124:554-628. [PMID: 37991799 DOI: 10.1021/acs.chemrev.3c00506] [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: 11/23/2023]
Abstract
In vivo imaging technologies have emerged as a powerful tool for both fundamental research and clinical practice. In particular, luminescence imaging in the tissue-transparent near-infrared (NIR, 700-1700 nm) region offers tremendous potential for visualizing biological architectures and pathophysiological events in living subjects with deep tissue penetration and high imaging contrast owing to the reduced light-tissue interactions of absorption, scattering, and autofluorescence. The distinctive quantum effects of nanocrystals have been harnessed to achieve exceptional photophysical properties, establishing them as a promising category of luminescent probes. In this comprehensive review, the interactions between light and biological tissues, as well as the advantages of NIR light for in vivo luminescence imaging, are initially elaborated. Subsequently, we focus on achieving deep tissue penetration and improved imaging contrast by optimizing the performance of nanocrystal fluorophores. The ingenious design strategies of NIR nanocrystal probes are discussed, along with their respective biomedical applications in versatile in vivo luminescence imaging modalities. Finally, thought-provoking reflections on the challenges and prospects for future clinical translation of nanocrystal-based in vivo luminescence imaging in the NIR region are wisely provided.
Collapse
Affiliation(s)
- Yang Yang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Qunying Jiang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
| | - Fan Zhang
- College of Energy Materials and Chemistry, State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Inner Mongolia University, Hohhot 010021, China
- Department of Chemistry, State Key Laboratory of Molecular Engineering of Polymers, Shanghai Key Laboratory of Molecular Catalysis and Innovative Materials, Fudan University, Shanghai 200433, China
| |
Collapse
|
35
|
Han X, Gong C, Yang Q, Zheng K, Wang Z, Zhang W. Biomimetic Nano-Drug Delivery System: An Emerging Platform for Promoting Tumor Treatment. Int J Nanomedicine 2024; 19:571-608. [PMID: 38260239 PMCID: PMC10802790 DOI: 10.2147/ijn.s442877] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 12/12/2023] [Indexed: 01/24/2024] Open
Abstract
With the development of nanotechnology, nanoparticles (NPs) have shown broad prospects as drug delivery vehicles. However, they exhibit certain limitations, including low biocompatibility, poor physiological stability, rapid clearance from the body, and nonspecific targeting, which have hampered their clinical application. Therefore, the development of novel drug delivery systems with improved biocompatibility and high target specificity remains a major challenge. In recent years, biofilm mediated biomimetic nano-drug delivery system (BNDDS) has become a research hotspot focus in the field of life sciences. This new biomimetic platform uses bio-nanotechnology to encapsulate synthetic NPswithin biomimetic membrane, organically integrating the low immunogenicity, low toxicity, high tumor targeting, good biocompatibility of the biofilm with the adjustability and versatility of the nanocarrier, and shows promising applications in the field of precision tumor therapy. In this review, we systematically summarize the new progress in BNDDS used for optimizing drug delivery, providing a theoretical reference for optimizing drug delivery and designing safe and efficient treatment strategies to improve tumor treatment outcomes.
Collapse
Affiliation(s)
- Xiujuan Han
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Chunai Gong
- Department of Pharmacy, Shanghai Ninth People’s Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, 201999, People’s Republic of China
| | - Qingru Yang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Kaile Zheng
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
| | - Zhuo Wang
- Department of Pharmacy, First Affiliated Hospital of Naval Medical University (Shanghai Changhai Hospital), Shanghai, 200433, People’s Republic of China
- School of Life Sciences and Biopharmaceuticals, Shenyang Pharmaceutical University, Shenyang, 110016, People’s Republic of China
| | - Wei Zhang
- Department of Pharmacy, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, People’s Republic of China
| |
Collapse
|
36
|
He Y, Cheng C, Liu Y, Chen FM, Chen Y, Yang C, Zhao Z, Dawulieti J, Shen Z, Zhang Y, Du JZ, Guan S, Shao D. Intravenous Senescent Erythrocyte Vaccination Modulates Adaptive Immunity and Splenic Complement Production. ACS NANO 2024; 18:470-482. [PMID: 38146673 DOI: 10.1021/acsnano.3c07943] [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: 12/27/2023]
Abstract
Targeted delivery of vaccines to the spleen remains a challenge. Inspired by the erythrophagocytotic process in the spleen, we herein report that intravenous administration of senescent erythrocyte-based vaccines profoundly alters their tropism toward splenic antigen-presenting cells (APCs) for imprinting adaptive immune responses. Compared with subcutaneous inoculation, intravenous vaccination significantly upregulated splenic complement expression in vivo and demonstrated synergistic antibody killing in vitro. Consequently, intravenous senescent erythrocyte vaccination produces potent SARS-CoV-2 antibody-neutralizing effects, with potential protective immune responses. Moreover, the proposed senescent erythrocyte can deliver antigens from resected tumors and adjuvants to splenic APCs, thereby inducing a personalized immune reaction against tumor recurrence after surgery. Hence, our findings suggest that senescent erythrocyte-based vaccines can specifically target splenic APCs and evoke adaptive immunity and complement production, broadening the tools for modulating immunity, helping to understand adaptive response mechanisms to senescent erythrocytes better, and developing improved vaccines against cancer and infectious diseases.
Collapse
Affiliation(s)
- Yan He
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 511442, China
| | - Chuanxu Cheng
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 511442, China
| | - Yuheng Liu
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
- Department of Pharmacology, College of Pharmacy, Chongqing Medical University, Chongqing, 400016, China
| | - Fang-Man Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 511442, China
| | - Yinglu Chen
- School of Medicine, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
| | - Chao Yang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 511442, China
- Department of Orthopedics, Academy of Orthopedics-Guangdong Province, Orthopedic Hospital of Guangdong Province, Guangdong Provincial Key Laboratory of Bone and Joint Degenerative Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou, 510665, China
| | - Zhibin Zhao
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Jianati Dawulieti
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 511442, China
| | - Zikun Shen
- School of Medicine, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
| | - Yunjiao Zhang
- School of Medicine, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
| | - Jin-Zhi Du
- School of Medicine, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
| | - Shan Guan
- National Engineering Research Center of Immunological Products, Third Military Medical University, Chongqing 400038, China
| | - Dan Shao
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou, Guangdong 511442, China
- School of Medicine, South China University of Technology, Guangzhou International Campus, Guangzhou, Guangdong 510006, China
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China
| |
Collapse
|
37
|
Mohammad-Rafiei F, Khojini JY, Ghazvinian F, Alimardan S, Norioun H, Tahershamsi Z, Tajbakhsh A, Gheibihayat SM. Cell membrane biomimetic nanoparticles in drug delivery. Biotechnol Appl Biochem 2023; 70:1843-1859. [PMID: 37387120 DOI: 10.1002/bab.2487] [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: 09/02/2022] [Accepted: 06/05/2023] [Indexed: 07/01/2023]
Abstract
Despite the efficiency of nanoparticle (NP) therapy, in vivo investigations have shown that it does not perform as well as in vitro. In this case, NP confronts many defensive hurdles once they enter the body. The delivery of NP to sick tissue is inhibited by these immune-mediated clearance mechanisms. Hence, using a cell membrane to hide NP for active distribution offers up a new path for focused treatment. These NPs are better able to reach the disease's target location, leading to enhanced therapeutic efficacy. In this emerging class of drug delivery vehicles, the inherent relation between the NPs and the biological components obtained from the human body was utilized, which mimic the properties and activities of native cells. This new technology has shown the viability of using biomimicry to evade immune system-provided biological barriers, with an emphasis on restricting clearance from the body before reaching its intended target. Furthermore, by providing signaling cues and transplanted biological components that favorably change the intrinsic immune response at the disease site, the NPs would be capable interacting with immune cells regarding the biomimetic method. Thus, we aimed to provide a current landscape and future trends of biomimetic NPs in drug delivery.
Collapse
Affiliation(s)
- Fatemeh Mohammad-Rafiei
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Javad Yaghmoorian Khojini
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| | - Fatemeh Ghazvinian
- Department of Life science and biotechnology, Faculty of Natural Sciences, University of Shahid Beheshti, Tehran, Iran
| | - Sajad Alimardan
- Department of Medical Biotechnology, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamid Norioun
- Medical Genetics Department, Institute of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran, Iran
| | - Zahra Tahershamsi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Amir Tajbakhsh
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Molecular Medicine, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Seyed Mohammad Gheibihayat
- Department of Medical Biotechnology, School of Medicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
- Network of Immunity in Infection, Malignancy and Autoimmunity (NIIMA), Universal Scientific Education and Research Network (USERN), Munich, Germany
| |
Collapse
|
38
|
Liu Z, Xia Q, Ma D, Wang Z, Li L, Han M, Yin X, Ji X, Wang S, Xin T. Biomimetic nanoparticles in ischemic stroke therapy. DISCOVER NANO 2023; 18:40. [PMID: 36969494 PMCID: PMC10027986 DOI: 10.1186/s11671-023-03824-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Accepted: 03/07/2023] [Indexed: 05/28/2023]
Abstract
Abstract Ischemic stroke is one of the most severe neurological disorders with limited therapeutic strategies. The utilization of nanoparticle drug delivery systems is a burgeoning field and has been widely investigated. Among these, biomimetic drug delivery systems composed of biogenic membrane components and synthetic nanoparticles have been extensively highlighted in recent years. Biomimetic membrane camouflage presents an effective strategy to prolong circulation, reduce immunogenicity and enhance targeting. For one thing, biomimetic nanoparticles reserve the physical and chemical properties of intrinsic nanoparticle. For another, the biological functions of original source cells are completely inherited. Compared to conventional surface modification methods, this approach is more convenient and biocompatible. In this review, membrane-based nanoparticles derived from different donor cells were exemplified. The prospect of future biomimetic nanoparticles in ischemic stroke therapy was discussed. Graphic abstract
Collapse
Affiliation(s)
- Zihao Liu
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021 China
| | - Qian Xia
- Department of Endocrinology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012 China
| | - Dengzhen Ma
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021 China
| | - Zhihai Wang
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021 China
| | - Longji Li
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021 China
| | - Min Han
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 China
| | - Xianyong Yin
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 China
| | - Xiaoshuai Ji
- Department of Neurosurgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021 China
| | - Shan Wang
- Shandong Key Laboratory of Reproductive Medicine, Department of Obstetrics and Gynecology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021 Shandong China
| | - Tao Xin
- Department of Neurosurgery, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, 250021 China
- Department of Neurosurgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, 250014 China
- Medical Science and Technology Innovation Center, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 250117 China
| |
Collapse
|
39
|
Tiwari P, Yadav K, Shukla RP, Gautam S, Marwaha D, Sharma M, Mishra PR. Surface modification strategies in translocating nano-vesicles across different barriers and the role of bio-vesicles in improving anticancer therapy. J Control Release 2023; 363:290-348. [PMID: 37714434 DOI: 10.1016/j.jconrel.2023.09.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 09/03/2023] [Accepted: 09/06/2023] [Indexed: 09/17/2023]
Abstract
Nanovesicles and bio-vesicles (BVs) have emerged as promising tools to achieve targeted cancer therapy due to their ability to overcome many of the key challenges currently being faced with conventional chemotherapy. These challenges include the diverse and often complex pathophysiology involving the progression of cancer, as well as the various biological barriers that circumvent therapeutic molecules reaching their target site in optimum concentration. The scientific evidence suggests that surface-functionalized nanovesicles and BVs camouflaged nano-carriers (NCs) both can bypass the established biological barriers and facilitate fourth-generation targeting for the improved regimen of treatment. In this review, we intend to emphasize the role of surface-functionalized nanovesicles and BVs camouflaged NCs through various approaches that lead to an improved internalization to achieve improved and targeted oncotherapy. We have explored various strategies that have been employed to surface-functionalize and biologically modify these vesicles, including the use of biomolecule functionalized target ligands such as peptides, antibodies, and aptamers, as well as the targeting of specific receptors on cancer cells. Further, the utility of BVs, which are made from the membranes of cells such as mesenchymal stem cells (MSCs), white blood cells (WBCs), red blood cells (RBCs), platelets (PLTs) as well as cancer cells also been investigated. Lastly, we have discussed the translational challenges and limitations that these NCs can encounter and still need to be overcome in order to fully realize the potential of nanovesicles and BVs for targeted cancer therapy. The fundamental challenges that currently prevent successful cancer therapy and the necessity of novel delivery systems are in the offing.
Collapse
Affiliation(s)
- Pratiksha Tiwari
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Krishna Yadav
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Ravi Prakash Shukla
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Shalini Gautam
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Disha Marwaha
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Madhu Sharma
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India
| | - Prabhat Ranjan Mishra
- Division of Pharmaceutics and Pharmacokinetics, CSIR-Central Drug Research Institute, Lucknow, India; Academy of Scientific and Innovation Research (AcSIR), Ghaziabad 201002, U.P., India.
| |
Collapse
|
40
|
Song X, Qian H, Yu Y. Nanoparticles Mediated the Diagnosis and Therapy of Glioblastoma: Bypass or Cross the Blood-Brain Barrier. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2023; 19:e2302613. [PMID: 37415556 DOI: 10.1002/smll.202302613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2023] [Revised: 06/19/2023] [Indexed: 07/08/2023]
Abstract
Glioblastoma is one of the most aggressive central nervous system malignancies with high morbidity and mortality. Current clinical approaches, including surgical resection, radiotherapy, and chemotherapy, are limited by the difficulty of targeting brain lesions accurately, leading to disease recurrence and fatal outcomes. The lack of effective treatments has prompted researchers to continuously explore novel therapeutic strategies. In recent years, nanomedicine has made remarkable progress and expanded its application in brain drug delivery, providing a new treatment for brain tumors. Against this background, this article reviews the application and progress of nanomedicine delivery systems in brain tumors. In this paper, the mechanism of nanomaterials crossing the blood-brain barrier is summarized. Furthermore, the specific application of nanotechnology in glioblastoma is discussed in depth.
Collapse
Affiliation(s)
- Xiaowei Song
- Department of Radiology, Anhui Provincial Institute of Translational Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, P. R. China
- Research Center of Clinical Medical Imaging, Hefei, 230022, China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei, 230011, P. R. China
- Anhui Engineering Research Center for Medical Micro-Nano Devices, Hefei, 230011, China
| | - Yongqiang Yu
- Department of Radiology, Anhui Provincial Institute of Translational Medicine, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, No. 218, Jixi Road, Shushan District, Hefei, 230022, P. R. China
- Research Center of Clinical Medical Imaging, Hefei, 230022, China
| |
Collapse
|
41
|
Nguyen-Thi PT, Nguyen TT, Phan HL, Ho TT, Vo TV, Vo GV. Cell membrane-based nanomaterials for therapeutics of neurodegenerative diseases. Neurochem Int 2023; 170:105612. [PMID: 37714337 DOI: 10.1016/j.neuint.2023.105612] [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: 10/14/2022] [Revised: 04/20/2023] [Accepted: 09/10/2023] [Indexed: 09/17/2023]
Abstract
Central nervous system (CNS) diseases such as Alzheimer's disease (AD), Parkinson's disease (PD), glioblastoma (GBM), and peripheral nerve injury have been documented as incurable diseases, which lead to serious impacts on human health especially prevalent in the aging population worldwide. Most of the treatment strategies fail due to low efficacy, toxicity, and poor brain penetration. Recently, advancements in nanotechnology have helped alleviate the challenges associated with the application of cell membrane-based nanomaterials against CNS diseases. In the following review, the existing types of cell membrane-based nanomaterials systems which have improved therapeutic efficacy for CNS diseases would be described. A summary of recent progress in the incorporation of nanomaterials in cell membrane-based production, separation, and analysis will be provided. Addition to, challenges relate to large-scale manufacturing of cell membrane-based nanomaterials and future clinical trial of such platforms will be discussed.
Collapse
Affiliation(s)
| | - Thuy Trang Nguyen
- Faculty of Chemical Engineering, Industrial University of Ho Chi Minh City, Ho Chi Minh City, 71420, Viet Nam.
| | - Hoang Long Phan
- Faculty of Pharmacy, Van Lang University, Ho Chi Minh City, 700000, Viet Nam
| | - Thanh-Tam Ho
- Institute for Global Health Innovations, Duy Tan University, Da Nang, 550000, Viet Nam; Faculty of Pharmacy, Duy Tan University, Da Nang, 550000, Viet Nam.
| | - Toi Van Vo
- Tissue Engineering and Regenerative Medicine Department, School of Biomedical Engineering, International University, Ho Chi Minh City, Viet Nam; Vietnam National University - Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Viet Nam
| | - Giau Van Vo
- Department of Biomedical Engineering, School of Medicine, Vietnam National University -Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Viet Nam; Research Center for Genetics and Reproductive Health (CGRH), School of Medicine, Vietnam National University, Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 70000, Viet Nam; Vietnam National University - Ho Chi Minh City (VNU-HCM), Ho Chi Minh City, 700000, Viet Nam
| |
Collapse
|
42
|
Li X, Pu X, Wang X, Wang J, Liao X, Huang Z, Yin G. A dual-targeting peptide for glioblastoma screened by phage display peptide library biopanning combined with affinity-adaptability analysis. Int J Pharm 2023; 644:123306. [PMID: 37572856 DOI: 10.1016/j.ijpharm.2023.123306] [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: 03/30/2023] [Revised: 06/26/2023] [Accepted: 08/10/2023] [Indexed: 08/14/2023]
Abstract
The obstruction of blood-brain barrier (BBB) and the poor specific targeting are still the major obstacles and challenges of targeted nano-pharmaceutical therapy for glioblastoma (GBM) up to now. It is critical to find appropriate targeting ligands that can effectively mediate the nano-pharmaceuticals to penetrate brain capillary endothelial cells (BCECs) and then specifically bind to glioblastoma cells (GCs). Herein, a dual-targeting ligand for GBM was screened by the combination of phage display peptide library biopanning and affinity-adaptability analysis. Based on the acquisition of sub-library of peptide which exhibited the specific affinity to both BCECs and GCs, a comparison parameter of relative affinity was deliberately introduced to evaluate the relative affinity of candidate peptides to U251-MG cells and bEnd.3 cells. The optimized WTW peptide (sequenced as WTWEYTK) was provided with a high relative affinity (RU/B = 2.44), implying that its high affinity to U251-MG cells and moderate affinity to bEnd.3 cells might synergistically promote its receptor-mediated internalization and transport, the dissociation from bEnd.3, and the binding to U251-MG. The results of BBB model trials in vitro showed that the BBB penetration efficiency and GBM accumulation of WTW peptide were significantly higher than those of WSL peptide, GNH peptide, and REF peptide. Results of orthotopic GBM xenograft model assays in vivo also indicated that WTW peptide had successfully penetrated the BBB and improved accumulation in GBM. The screened WTW peptide might be the potential dual-targeting ligand to motivate the advancement of GBM targeted therapy.
Collapse
Affiliation(s)
- Xiaoxu Li
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, PR China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, PR China
| | - Xingming Wang
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, PR China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, PR China
| | - Xiaoming Liao
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, PR China
| | - Zhongbin Huang
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, PR China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, PR China.
| |
Collapse
|
43
|
Jiang X, Wu L, Zhang M, Zhang T, Chen C, Wu Y, Yin C, Gao J. Biomembrane nanostructures: Multifunctional platform to enhance tumor chemoimmunotherapy via effective drug delivery. J Control Release 2023; 361:510-533. [PMID: 37567505 DOI: 10.1016/j.jconrel.2023.08.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 07/02/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023]
Abstract
Chemotherapeutic drugs have been found to activate the immune response against tumors by inducing immunogenic cell death, in addition to their direct cytotoxic effects toward tumors, therefore broadening the application of chemotherapy in tumor immunotherapy. The combination of other therapeutic strategies, such as phototherapy or radiotherapy, could further strengthen the therapeutic effects of immunotherapy. Nanostructures can facilitate multimodal tumor therapy by integrating various active agents and combining multiple types of therapeutics in a single nanostructure. Biomembrane nanostructures (e.g., exosomes and cell membrane-derived nanostructures), characterized by superior biocompatibility, intrinsic targeting ability, intelligent responsiveness and immune-modulating properties, could realize superior chemoimmunotherapy and represent next-generation nanostructures for tumor immunotherapy. This review summarizes recent advances in biomembrane nanostructures in tumor chemoimmunotherapy and highlights different types of engineering approaches and therapeutic mechanisms. A series of engineering strategies for combining different biomembrane nanostructures, including liposomes, exosomes, cell membranes and bacterial membranes, are summarized. The combination strategy can greatly enhance the targeting, intelligence and functionality of biomembrane nanostructures for chemoimmunotherapy, thereby serving as a stronger tumor therapeutic method. The challenges associated with the clinical translation of biomembrane nanostructures for chemoimmunotherapy and their future perspectives are also discussed.
Collapse
Affiliation(s)
- Xianghe Jiang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China
| | - Lili Wu
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Mengya Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Cuimin Chen
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang 157011, China.
| | - Chuan Yin
- Department of Gastroenterology, Shanghai Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China.
| |
Collapse
|
44
|
Hao Y, Li H, Ge X, Liu Y, Yin J, Li X, Liu Y, Chen H, Huang L, Zou J, Zhang S, Wu H, Zhang Z. Site-specific nanoswitch circumventing immune resistance via activating TLR and inhibiting PD-L1/PD-1 axis. J Control Release 2023; 361:64-76. [PMID: 37532143 DOI: 10.1016/j.jconrel.2023.07.048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2023] [Revised: 07/14/2023] [Accepted: 07/29/2023] [Indexed: 08/04/2023]
Abstract
Immunotherapy has fundamentally altered cancer treatment; however, its effectiveness is clinically hampered by insufficient intratumoral T lymphocyte infiltration and failed T lymphocyte priming. Additionally, inducing cancer-specific immune responses while sparing normal cells remains challenging. Herein, we developed a redox-activatable polymeric nanoswitch (c-N@IM/JQ) that remained 'off' status in circulation but rapidly switched 'on' after entering the tumor. Toll-like receptor (TLR) 7/8 agonist (imidazoquinoline, IMQ) and bromodomain and extraterminal inhibitor (JQ1) are locked in c-N@IM/JQ with a redox-cleavable linker (switch off). Upon systemic administration, c-N@IM/JQ with c-RGD peptide modification preferentially accumulated at tumor sites and responded to the high glutathione levels to release native IMQ for fully mobilizing T lymphocyte army, and JQ1 for removing the programmed death ligand (PD-L)-1 protection on tumor cells (switch on). These strengthened T lymphocyte armies are easily accessible to these de-protected tumor cells, revitalizing the immune response against tumors.
Collapse
Affiliation(s)
- Yanyun Hao
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Hui Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Xiaoyan Ge
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Yang Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jialin Yin
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Xia Li
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Yutong Liu
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Hongfei Chen
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Lingling Huang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Jing Zou
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Shiying Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Hao Wu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China
| | - Zhiyue Zhang
- NMPA Key Laboratory for Technology Research and Evaluation of Drug Products, Key Laboratory of Chemical Biology (Ministry of Education), Department of Pharmaceutics, School of Pharmaceutical Sciences, Cheeloo College of Medicine, Shandong University, Jinan 250012, PR China.
| |
Collapse
|
45
|
Li W, Cheng J, He F, Zhang P, Zhang N, Wang J, Song Q, Hou Y, Gan Z. Cell membrane-based nanomaterials for theranostics of central nervous system diseases. J Nanobiotechnology 2023; 21:276. [PMID: 37596631 PMCID: PMC10439658 DOI: 10.1186/s12951-023-02004-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 07/13/2023] [Indexed: 08/20/2023] Open
Abstract
Central nervous system (CNS) diseases have been widely acknowledged as one of the major healthy concerns globally, which lead to serious impacts on human health. There will be about 135 million CNS diseases cases worldwide by mid-century, and CNS diseases will become the second leading cause of death after the cardiovascular disease by 2040. Most CNS diseases lack of effective diagnostic and therapeutic strategies with one of the reasons that the biological barrier extremely hampers the delivery of theranostic agents. In recent years, nanotechnology-based drug delivery is a quite promising way for CNS diseases due to excellent properties. Among them, cell membrane-based nanomaterials with natural bio-surface, high biocompatibility and biosafety, are of great significance in both the diagnosis and treatment of different CNS diseases. In this review, the state of art of the fabrication of cell membranes-based nanomaterials is introduced. The characteristics of different CNS diseases, and the application of cell membranes-based nanomaterials in the theranostics are summarized. In addition, the future prospects and limitations of cell membrane nanotechnology are anticipated. Through summarizing the state of art of the fabrication, giving examples of CNS diseases, and highlighting the applications in theranostics, the current review provides designing methods and ideas for subsequent cell membrane nanomaterials.
Collapse
Affiliation(s)
- Wenyue Li
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Junwei Cheng
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fangfei He
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Peisen Zhang
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Ni Zhang
- Department of Psychiatry, West China Hospital of Sichuan University, Chengdu, 610041, China.
| | - Jian Wang
- Department of Head and Neck Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, 100021, China.
| | - Qiliang Song
- Shandong Peninsula Engineering Research Center of Comprehensive Brine Utilization, Weifang University of Science and Technology, Shouguang, 262700, China.
| | - Yi Hou
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Zhihua Gan
- College of Materials Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
46
|
Hu Y, Zhao M, Wang H, Guo Y, Cheng X, Zhao T, Wang H, Zhang Y, Ma Y, Tao W. Exosome-sheathed ROS-responsive nanogel to improve targeted therapy in perimenopausal depression. J Nanobiotechnology 2023; 21:261. [PMID: 37553718 PMCID: PMC10408189 DOI: 10.1186/s12951-023-02005-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2023] [Accepted: 07/13/2023] [Indexed: 08/10/2023] Open
Abstract
The development of natural membranes as coatings for nanoparticles to traverse the blood-brain barrier (BBB) presents an effective approach for treating central nervous system (CNS) disorders. In this study, we have designed a nanogel loaded with PACAP and estrogen (E2), sheathed with exosomes and responsive to reactive oxygen species (ROS), denoted as HA NGs@exosomes. The objective of this novel design is to serve as a potent drug carrier for the targeted treatment of perimenopausal depression. The efficient cellular uptake and BBB penetration of HA NGs@exosomes has been demonstrated in vitro and in vivo. Following intranasal intervention with HA NGs@exosomes, ovariectomized mice under chronic unpredictable mild stress (CUMS) have shown improved behavioral performance, indicating that HA NGs@exosomes produced a rapid-onset antidepressant effect. Moreover, HA NGs@exosomes exhibit notable antioxidant and anti-inflammatory properties and may regulate the expression of pivotal proteins in the PACAP/PAC1 pathway to promote synaptic plasticity. Our results serve as a proof-of-concept for the utility of exosome-sheathed ROS-responsive nanogel as a promising drug carrier for the treatment of perimenopausal depression.
Collapse
Affiliation(s)
- Yue Hu
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 8 Zhongnan West Road, Wuxi, 214071, China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Min Zhao
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 8 Zhongnan West Road, Wuxi, 214071, China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Hui Wang
- School of pharmacology, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Yang Guo
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 8 Zhongnan West Road, Wuxi, 214071, China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Xiaolan Cheng
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 8 Zhongnan West Road, Wuxi, 214071, China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Tong Zhao
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 8 Zhongnan West Road, Wuxi, 214071, China
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China
| | - Hanqing Wang
- School of pharmacology, Ningxia Medical University, 1160 Shengli Street, Xingqing District, Yinchuan, China
| | - Yafeng Zhang
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 8 Zhongnan West Road, Wuxi, 214071, China.
| | - Yong Ma
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 8 Zhongnan West Road, Wuxi, 214071, China.
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| | - Weiwei Tao
- Jiangsu CM Clinical Innovation Center of Degenerative Bone & Joint Disease, Wuxi TCM Hospital Affiliated to Nanjing University of Chinese Medicine, 8 Zhongnan West Road, Wuxi, 214071, China.
- School of Chinese Medicine, School of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, Nanjing University of Chinese Medicine, 138 Xianlin Road, Nanjing, 210023, China.
| |
Collapse
|
47
|
Yenurkar D, Nayak M, Mukherjee S. Recent advances of nanocrystals in cancer theranostics. NANOSCALE ADVANCES 2023; 5:4018-4040. [PMID: 37560418 PMCID: PMC10408581 DOI: 10.1039/d3na00397c] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 07/10/2023] [Indexed: 08/11/2023]
Abstract
Emerging cancer cases across the globe and treating them with conventional therapies with multiple limitations have been challenging for decades. Novel drug delivery systems and alternative theranostics are required for efficient detection and treatment. Nanocrystals (NCs) have been established as a significant cancer diagnosis and therapeutic tool due to their ability to deliver poorly water-soluble drugs with sustained release, low toxicity, and flexibility in the route of administration, long-term sustainable drug release, and noncomplicated excretion. This review summarizes several therapies of NCs, including anticancer, immunotherapy, radiotherapy, biotheranostics, targeted therapy, photothermal, and photodynamic. Further, different imaging and diagnostics using NCs are mentioned, including imaging, diagnosis through magnetic resonance imaging (MRI), computed tomography (CT), biosensing, and luminescence. In addition, the limitations and potential solutions of NCs in the field of cancer theranostics are discussed. Preclinical and clinical data depicting the importance of NCs in the spotlight of cancer, its current status, future aspects, and challenges are covered in detail.
Collapse
Affiliation(s)
- Devyani Yenurkar
- School of Biomedical Engineering, Indian Institute of Technology, BHU Varanasi-221005 UP India
| | - Malay Nayak
- School of Biomedical Engineering, Indian Institute of Technology, BHU Varanasi-221005 UP India
| | - Sudip Mukherjee
- School of Biomedical Engineering, Indian Institute of Technology, BHU Varanasi-221005 UP India
| |
Collapse
|
48
|
Wu H, Zhang T, Li N, Gao J. Cell membrane-based biomimetic vehicles for effective central nervous system target delivery: Insights and challenges. J Control Release 2023; 360:169-184. [PMID: 37343724 DOI: 10.1016/j.jconrel.2023.06.023] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Revised: 06/14/2023] [Accepted: 06/17/2023] [Indexed: 06/23/2023]
Abstract
Central nervous system (CNS) disorders, including brain tumor, ischemic stroke, Alzheimer's disease, and Parkinson's disease, threaten human health. And the existence of the blood-brain barrier (BBB) hinders the delivery of drugs and the design of drug targeting delivery vehicles. Over the past decades, great interest has been given to cell membrane-based biomimetic vehicles since the rise of targeting drug delivery systems and biomimetic nanotechnology. Cell membranes are regarded as natural multifunction biomaterials, and provide potential for targeting delivery design and modification. Cell membrane-based biomimetic vehicles appear timely with the participation of cell membranes and nanoparticles, and raises new lights for BBB recognition and transport, and effective therapy with its biological multifunction and high biocompatibility. This review summarizes existing challenges in CNS target delivery and recent advances of different kinds of cell membrane-based biomimetic vehicles for effective CNS target delivery, and deliberates the BBB targeting mechanism. It also discusses the challenges and possibility of clinical translation, and presents new insights for development.
Collapse
Affiliation(s)
- Honghui Wu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, PR China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, PR China
| | - Tianyuan Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, PR China
| | - Ni Li
- Department of Cardiothoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315041, Zhejiang, PR China
| | - Jianqing Gao
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, Zhejiang, PR China; Jinhua Institute of Zhejiang University, Jinhua 321299, Zhejiang, PR China; Department of Cardiothoracic Surgery, Ningbo Medical Centre Lihuili Hospital, Ningbo University, Ningbo 315041, Zhejiang, PR China.
| |
Collapse
|
49
|
Wu Y, Qian Y, Peng W, Qi X. Functionalized nanoparticles crossing the brain-blood barrier to target glioma cells. PeerJ 2023; 11:e15571. [PMID: 37426416 PMCID: PMC10327649 DOI: 10.7717/peerj.15571] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/25/2023] [Indexed: 07/11/2023] Open
Abstract
Glioma is the most common tumor of the central nervous system (CNS), with a 5-year survival rate of <35%. Drug therapy, such as chemotherapeutic and immunotherapeutic agents, remains one of the main treatment modalities for glioma, including temozolomide, doxorubicin, bortezomib, cabazitaxel, dihydroartemisinin, immune checkpoint inhibitors, as well as other approaches such as siRNA, ferroptosis induction, etc. However, the filter function of the blood-brain barrier (BBB) reduces the amount of drugs needed to effectively target CNS tumors, making it one of the main reasons for poor drug efficacies in glioma. Thus, finding a suitable drug delivery platform that can cross the BBB, increase drug aggregation and retainment in tumoral areas and avoid accumulation in non-targeted areas remains an unsolved challenge in glioma drug therapy. An ideal drug delivery system for glioma therapy should have the following features: (1) prolonged drug life in circulation and effective penetration through the BBB; (2) adequate accumulation within the tumor (3) controlled-drug release modulation; (4) good clearance from the body without significant toxicity and immunogenicity, etc. In this regard, due to their unique structural features, nanocarriers can effectively span the BBB and target glioma cells through surface functionalization, providing a new and effective strategy for drug delivery. In this article, we discuss the characteristics and pathways of different nanocarriers for crossing the BBB and targeting glioma by listing different materials for drug delivery platforms, including lipid materials, polymers, nanocrystals, inorganic nanomaterials, etc.
Collapse
Affiliation(s)
- Yongyan Wu
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
| | - Yufeng Qian
- Department of Neurosurgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, People’s Republic of China
| | - Wei Peng
- Medical Research Center, Shaoxing People’s Hospital, Shaoxing, Zhejiang Province, People’s Republic of China
| | - Xuchen Qi
- Department of Neurosurgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, People’s Republic of China
- Department of Neurosurgery, Shaoxing People’s Hospital, Shaoxing, Zhejiang, People’s Republic of China
| |
Collapse
|
50
|
Yang Y, Cheng N, Luo Q, Shao N, Ma X, Chen J, Luo L, Xiao Z. How Nanotherapeutic Platforms Play a Key Role in Glioma? A Comprehensive Review of Literature. Int J Nanomedicine 2023; 18:3663-3694. [PMID: 37427368 PMCID: PMC10327925 DOI: 10.2147/ijn.s414736] [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: 03/29/2023] [Accepted: 06/15/2023] [Indexed: 07/11/2023] Open
Abstract
Glioblastoma (GBM), a highly aggressive form of brain cancer, is considered one of the deadliest cancers, and even with the most advanced medical treatments, most affected patients have a poor prognosis. However, recent advances in nanotechnology offer promising avenues for the development of versatile therapeutic and diagnostic nanoplatforms that can deliver drugs to brain tumor sites through the blood-brain barrier (BBB). Despite these breakthroughs, the use of nanoplatforms in GBM therapy has been a subject of great controversy due to concerns over the biosafety of these nanoplatforms. In recent years, biomimetic nanoplatforms have gained unprecedented attention in the biomedical field. With advantages such as extended circulation times, and improved immune evasion and active targeting compared to conventional nanosystems, bionanoparticles have shown great potential for use in biomedical applications. In this prospective article, we endeavor to comprehensively review the application of bionanomaterials in the treatment of glioma, focusing on the rational design of multifunctional nanoplatforms to facilitate BBB infiltration, promote efficient accumulation in the tumor, enable precise tumor imaging, and achieve remarkable tumor suppression. Furthermore, we discuss the challenges and future trends in this field. Through careful design and optimization of nanoplatforms, researchers are paving the way toward safer and more effective therapies for GBM patients. The development of biomimetic nanoplatform applications for glioma therapy is a promising avenue for precision medicine, which could ultimately improve patient outcomes and quality of life.
Collapse
Affiliation(s)
- Yongqing Yang
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Nianlan Cheng
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Qiao Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Ni Shao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Xiaocong Ma
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Jifeng Chen
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Liangping Luo
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People’s Republic of China
| | - Zeyu Xiao
- The Guangzhou Key Laboratory of Molecular and Functional Imaging for Clinical Translation, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, People’s Republic of China
| |
Collapse
|