1
|
Sindeeva OA, Kozyreva ZV, Abdurashitov AS, Sukhorukov GB. Engineering colloidal systems for cell manipulation, delivery, and tracking. Adv Colloid Interface Sci 2025; 340:103462. [PMID: 40037017 DOI: 10.1016/j.cis.2025.103462] [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: 10/14/2024] [Revised: 02/22/2025] [Accepted: 02/23/2025] [Indexed: 03/06/2025]
Abstract
Men-made colloidal systems are widely presented across various aspects of biomedical science. There is a strong demand for engineering colloids to tailor their functions and properties to meet the requirements of biological and medical tasks. These requirements are not only related to size, shape, capacity to carry bioactive compounds as drug delivery systems, and the ability to navigate via chemical and physical targeting. Today, the more challenging aspects of colloid design are how the colloidal particles interact with biological cells, undergo internalization by cells, how they reside in the cell interior, and whether we can explore cells with colloids, intervene with biochemical processes, and alter cell functionality. Cell tracking, exploitation of cells as natural transporters of internalized colloidal carriers loaded with drugs, and exploring physical methods as external triggers of cell functions are ongoing topics in the research agenda. In this review, we summarize recent advances in these areas, focusing on how colloidal particles interact and are taken up by mesenchymal stem cells, dendritic cells, neurons, macrophages, neutrophils and lymphocytes, red blood cells, and platelets. The engineering of colloidal vesicles with cell membrane fragments and exosomes facilitates their application. The perspectives of different approaches in colloid design, their limitations, and obstacles on the biological side are discussed.
Collapse
Affiliation(s)
- Olga A Sindeeva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia.
| | - Zhanna V Kozyreva
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia
| | - Arkady S Abdurashitov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia; Life Improvement by Future Technologies (LIFT) Center, Bolshoy Boulevard 30, Moscow 121205, Russia
| | - Gleb B Sukhorukov
- Skolkovo Institute of Science and Technology, Bolshoy Boulevard 30, Moscow 121205, Russia.
| |
Collapse
|
2
|
Zhang Z, Ma T, Liu Q, Nan J, Liu G, Yang Y, Hu Y, Xie J. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells Encapsulated in M2 Macrophage Cell Membrane Targeted to Inhibit Joint Periprosthetic Inflammation. ACS APPLIED MATERIALS & INTERFACES 2025; 17:22279-22292. [PMID: 40168527 DOI: 10.1021/acsami.4c22304] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2025]
Abstract
Periprosthetic osteolysis (PPOL) is a serious complication following total joint replacement surgery, and exploring treatments for this complication is of significant societal importance. Exosomes derived from bone marrow mesenchymal stem cells (BMSC-Exos, Exos) have diverse cellular functions, such as inhibiting osteoclast formation, suppressing inflammation progression, and promoting M2 macrophage polarization. However, standalone Exosomes are easily recognized and phagocytosed by the immune system, have a short half-life, and lack specificity. This study is based on the homing effect possessed by M2 macrophages under the regulation of various factors. By combining this with cell membrane encapsulation technology and embedding BMSC-Exos within the membrane of M2 macrophages (M2M-Exos), the aim is to inhibit inflammation and treat PPOL. It was found that M2M-Exos can target the PPOL area, enhancing the therapeutic effects of the BMSC-Exos and reducing wear particle-induced cranial osteolysis. Additionally, M2M-Exos provide immune camouflage through the cell membrane, allowing the BMSC-Exos to evade clearance by the mononuclear macrophage system in the body. Therefore, the study demonstrates the targeting ability of M2M-Exos and their unique role in preventing PPOL. These biomimetic nanoparticles establish a targeted nanodrug delivery system for PPOL treatment.
Collapse
Affiliation(s)
- Zheyu Zhang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Tianliang Ma
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Qimeng Liu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jiangyu Nan
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Guanzhi Liu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yute Yang
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Yihe Hu
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| | - Jie Xie
- Department of Orthopedics, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, China
| |
Collapse
|
3
|
Wu Z, Yang S, Wei Z, Wang Y, Yang X, Zhu J, Hong H, Li P, Yu XF, Peng C, Zhi C. Constructing Lipid-Like Biomimetic Structure via Electrolyte Designation for Stable Zinc-Ion Batteries. ACS NANO 2025; 19:14085-14096. [PMID: 40189849 PMCID: PMC12005047 DOI: 10.1021/acsnano.4c18796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Revised: 03/24/2025] [Accepted: 03/24/2025] [Indexed: 04/16/2025]
Abstract
Zinc-ion batteries (ZIBs) have attracted widespread attention in recent years. However, due to the aqueous electrolyte's high activity, the zinc anode is affected by severe side reactions such as corrosion and hydrogen evolution, resulting in poor reversibility. Inspired by the structure of a lipid bilayer in biology, in this paper, we introduce lithium nonafluorobutylsulfonate to inhibit the water activity via vigorous binding between S═O and H2O and form a bilayer lipid-like protective structure on the surface of the zinc anode, thereby improving the reversibility of the zinc anode and extending the lifespan of the ZIBs. The zinc anode in the biomimetic electrolyte demonstrated outstanding reversibility with a 880 h cycle life and 99.91% average Comlombic efficiency in the Zn||Cu asymmetric battery, as well as a 2460 h cycle life and a cumulative capacity of 6 Ah cm-2 in the Zn||Zn symmetric battery (5 mA cm-2 and 5 mAh cm-2). In addition, full cells with Zn0.25V2O5·nH2O and MnO2 show excellent capacity retention of 91.67% after 1200 cycles and 100% after 1000 cycles, respectively. After cycles, the ampere-hour-level pouch cell showed a capacity retention rate of 93%. This method provides a biomimetic strategy for constructing biomimetic electrolytes to improve the reversibility of zinc anodes.
Collapse
Affiliation(s)
- Zhuoxi Wu
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
| | - Shuo Yang
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
| | - Zhiquan Wei
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
| | - Yiqiao Wang
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
| | - Xinru Yang
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
| | - Jiaxiong Zhu
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
| | - Hu Hong
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
| | - Pei Li
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
| | - Xue-Feng Yu
- Materials
Interfaces Center, Shenzhen Institute of
Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Chao Peng
- Materials
Interfaces Center, Shenzhen Institute of
Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, Guangdong, China
| | - Chunyi Zhi
- Department
of Materials Science and Engineering, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
- Hong
Kong Institute for Advanced Study, City
University of Hong Kong, Kowloon 999077, Hong Kong
SAR, China
- Hong
Kong Institute for Clean Energy, City University
of Hong Kong, Kowloon 999077, Hong Kong SAR, China
- Centre
for Advanced Nuclear Safety and Sustainable Development, City University of Hong Kong, Kowloon 999077, Hong Kong SAR, China
| |
Collapse
|
4
|
Xiang X, Shao Y, Xiang L, Jiao Q, Zhang W, Qin Y, Chen Y. Suppression of Liver Fibrogenesis with Photothermal Sorafenib Nanovesicles via Selectively Inhibiting Glycolysis and Amplification of Active HSCs. Mol Pharm 2025; 22:1939-1957. [PMID: 40053386 DOI: 10.1021/acs.molpharmaceut.4c01135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/08/2025]
Abstract
As the major driving factor of hepatic fibrosis, the activated hepatic stellate cells (aHSCs) rely on active glycolysis to support their aberrant proliferation and secretion of the extracellular matrix. Sorafenib (Sor) can combat liver fibrosis by suppressing HIF-1α and glycolysis, but its poor solubility, rapid metabolism, and low bioavailability restrict such a clinical application. Here, Sor was loaded onto polydopamine nanoparticles and then encapsulated by a retinoid-decorated red blood cell membrane, yielding HSC-targeted Sor nanovesicles (PDA/Sor@RMV-VA) with a high Sor-loading capacity and photothermally controlled drug release for antifibrotic treatment. These Sor RMVs not only exhibited a good particle size, dispersity and biocompatibility, prolonged circulation time, enhanced aHSC targetability, and hepatic accumulation both in vitro and in vivo, but also displayed a mild photothermal activity proper for promoting sorafenib release and accumulation in CCl4-induced fibrotic mouse livers without incurring phototoxicity. Compared with nontargeting Sor formulations, PDA/Sor@RMV-VA more effectively downregulated HIF-1α and glycolytic enzyme in both cultured aHSCs and fibrotic mice and reversed myofibroblast phenotype and amplification of aHSCs and thus more significantly improved liver damage, inflammation, and fibrosis, all of which could be even further advanced with NIR irradiation. These results fully demonstrate the antifibrotic power and therapeutic potential of PDA/Sor@RMV-VA as an antifibrotic nanomedicine, which would support a new clinical treatment for hepatic fibrosis.
Collapse
Affiliation(s)
- Xianjing Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yaru Shao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Li Xiang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| | - Qiangqiang Jiao
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Wenhui Zhang
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yuting Qin
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
| | - Yuping Chen
- School of Pharmaceutical Sciences, University of South China, Hengyang 410001, China
- Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
- MOE Key Laboratory of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan 410001, China
| |
Collapse
|
5
|
Chen J, Xu X, Wang K, Xue M, Wang Q, Zhong W, Wan Y, Liu X, Zheng J, Gao G, Wang G. Hypoxia-Activated Liposomes Enable Synergistic Photodynamic Therapy for Oral Cancer. Adv Healthc Mater 2025; 14:e2404395. [PMID: 39950552 DOI: 10.1002/adhm.202404395] [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: 11/07/2024] [Revised: 02/02/2025] [Indexed: 04/08/2025]
Abstract
Oral cancer is a significant public health problem, which is one of the most common malignancy with high mortality in the world. Apart from surgery, photodynamic therapy (PDT) is perceived as another capable method for the treatment of oral cancer. However, limited oxygen content of tumor microenvironment decreased the efficiency of current PDT. Hence, a complementary tumor-killing system is developed to overcome PDT-induced oxygen depletion by introducing hypoxia-activated chemotherapy drugs into tumor-targeting liposomes. First, liposomes derived from red blood cell (RBC) membrane are decorated with tumor-targeting formic acid (FA) and designed to carrying photosensitizer TPP and the hypoxia-activated drug TPZ (TPZ/TPP@RBC-FA). Laser irradiation on tumor initiated the discharge of chemotherapeutic drugs (TPZ) from TPZ/TPP@RBC-FA liposomes by increasing the reactive oxygen species, causing the aggravated intratumoral hypoxia and the further generation of the toxic TPZ radicals. The combination of hypoxia-activated chemotherapy and photodynamic therapy will not only induced efficient cell apoptosis but also caused immunogenic cell death and immunocyte activation. The strategy of loading drugs into liposomes also demonstrated acceptable biosafety. It is believed that the strategy of combining of photodynamic therapy and hypoxia-activated chemotherapy with liposomes can be implemented for the oral cancer treatment in the near future.
Collapse
Affiliation(s)
- Jiaxuan Chen
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Xiaodi Xu
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Kexuan Wang
- School of Medical Imaging, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Mingxia Xue
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Qing Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Weixiang Zhong
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Yan Wan
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Xiaoyang Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu, 210096, China
| | - Junnian Zheng
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Ge Gao
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, 99 Huaihai Road, Xuzhou, Jiangsu, 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, 209 Tongshan Road, Xuzhou, Jiangsu, 221004, China
| |
Collapse
|
6
|
Li W, Zhang H, Chen L, Huang C, Jiang Z, Zhou H, Zhu X, Liu X, Zheng Z, Yu Q, He Y, Gao Y, Ma J, Yang L. Cell membrane-derived nanovesicles as extracellular vesicle-mimetics in wound healing. Mater Today Bio 2025; 31:101595. [PMID: 40104636 PMCID: PMC11914519 DOI: 10.1016/j.mtbio.2025.101595] [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: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Cell membrane-derived nanovesicles (NVs) have emerged as promising alternatives to extracellular vesicles (EVs) for wound healing applications, addressing the limitations of traditional EVs, which include insufficient targeting capability, low production yield, and limited drug-loading capacity. Through mechanical cell extrusion methods, NVs exhibit superior characteristics, demonstrating enhanced yield, stability, and purity compared to natural EVs. These NVs can be derived from various membrane sources, including single cell types (stem cells, blood cells, immune cells, and bacterial membranes), hybrid cell membranes and cell membranes mixed with liposomes, with each offering unique therapeutic properties. The integration of genetic engineering and surface modifications has further enhanced NV functionality, enabling precise targeting and improved drug delivery capabilities. Recent advances in NV-based therapies have demonstrated their potential across multiple biomedical applications. Although challenges persist in terms of standardization, storage stability, and clinical translation, the combination of natural cell-derived functions with artificial modification potential positions NVs as a promising platform for next-generation therapeutic delivery systems, thereby offering new possibilities in wound healing applications. Finally, we explore the challenges and future prospects of translating NV-based therapeutics into clinical practice, providing insights into the future development of this innovative approach in wound healing and tissue repair.
Collapse
Affiliation(s)
- Wenwen Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huihui Zhang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoyang Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hai Zhou
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinxi Zhu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoyang Liu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zesen Zheng
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiuyi Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yufang He
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
7
|
Xi ZY, Fan CY, Jiang YY, Xi XR, Nie GY, Zhu S, Zhang JJ, Xu L. Nanocatalytic system releases overloaded zinc ions and ROS to induce Znproptosis and interrupt cell cycle through inhibiting Akt/mTOR pathway. Theranostics 2025; 15:4734-4762. [PMID: 40225560 PMCID: PMC11984402 DOI: 10.7150/thno.107025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/09/2025] [Indexed: 04/15/2025] Open
Abstract
Background: Traditional programmed cell death, including ferroptosis, cuproptosis, and apoptosis, has demonstrated excellent anti-tumor effects and declared their complete mechanisms, however, the zinc ion-mediated tumor inhibiting mechanisms remain insufficiently explored. In this study, a self-generated oxygen nanocatalytic system (ZnO@COF@EM, ZCE) was developed to stimulate cascade amplified effect (CAE) of reactive oxygen species (ROS) generation leading to Znproptosis. The underlying Znproptosis mechanism to disrupt mitochondrial (Mito) metabolism was also investigated. Methods: Specifically, the principle of Znproptosis caused by accumulated zinc and ROS, which served as key factors, was declared through western blot analysis and genetic testing. The mechanism of generated ROS (·OH and 1O2) under NIR irradiation by ZCE was detected by UV scanning curves, confocal laser scanning microscopy (CLSM) images, and density functional analysis. The injury condition of Fe-S protein of mitochondria metabolism, which triggered Znproptosis with FDX2/LIAS pathway by zinc and ROS, was examined by PCR test and MTT assay. Notably, a Mito-targeting strategy for ZCE was proposed by using molecular docking technology, wherein Zn2+ was recognized by zinc finger proteins (ZFPs) with the Mito. Results: ZCE, along with CAE, produced abundant ROS (2.42-time more than control group). At the quantum chemical level, the CAE mechanism was associated with a narrower highest occupied molecular orbital-lowest unoccupied molecular orbital (HOMO-LUMO) gap and increased electronic energy motion within ZCE, which prolonged the excited triplet state (ETS). At the gene level, Znproptosis was achieved by regulating FDX2 and ZIP7 proteins to damage Fe-S protein. The cell cycle was interrupted by Chk2/Cdc25C/Cdc2 and Chk2/p21/Cyclin B1 pathways, leading to the arrest of G1/S and G2/M phases of the cell cycle and inhibition of the Akt/mTOR signaling pathway. Moreover, Znproptosis induced by overloading zinc ions and ROS resulted in a significant antitumor effect (up to 83.81%). Conclusion: Hence, the research reveals a detailed Znproptosis mechanism in nanocatalytic system. Through regulating FDX2/LIAS pathway, Znproptosis could improve the death rate of mitochondria by decreasing the production of Fe-S protein, contributing to advancements in the field of antitumor therapy.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Lu Xu
- School of Pharmacy, Shenyang Pharmaceutical University, Shenyang, 110016, China
| |
Collapse
|
8
|
Karabaliev M, Paarvanova B, Savova G, Tacheva B, Georgieva R. In Situ Monitoring of Morphology Changes and Oxygenation State of Human Erythrocytes During Surfactant-Induced Hemolysis. Cells 2025; 14:469. [PMID: 40214423 PMCID: PMC11987960 DOI: 10.3390/cells14070469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/19/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Erythrocytes, the most abundant blood cells, are a prevalent cell model for the analysis of the membrane-damaging effects of different molecules, including drugs. In response to stimuli, erythrocytes can change their morphology, e.g., shape or volume, which in turns influences their main function to transport oxygen. Membrane active molecules can induce hemolysis, i.e., release of hemoglobin into the blood plasma. Free hemoglobin in the blood circulation is toxic causing serious health problems including vasoconstriction, high blood pressure and kidney damage. Therefore, early recognition of the risk of massive hemolysis is highly important. Here, we investigated surfactant induced hemolysis applying UV-vis spectrophotometry. Saponin, sodium dodecyl sulfate and Triton X-100, detergents known to provoke hemolysis at different concentrations and by different mechanisms, were applied to initiate the process. Whole absorption spectra of erythrocyte suspensions in the range 300-750 nm were recorded every 15 s for following the process in real-time. The hemolysis process, with respect to morphological changes in the erythrocytes and their influence on the oxygenation state of hemoglobin, was characterized by the absorbance at 700 nm, the height relative to the background and the wavelength of the Soret peak. The results suggest that these UV-vis spectrophotometry parameters provide reliable information in real-time; not only about the process of hemolysis itself, but also about pre-hemolytic changes in the erythrocytes, even at sub-hemolytic surfactant concentrations.
Collapse
Affiliation(s)
- Miroslav Karabaliev
- Department of Physics and Biophysics, Faculty of Medicine, Trakia University, 11 Armeiska, 6000 Stara Zagora, Bulgaria; (B.P.); (G.S.); (B.T.)
| | - Boyana Paarvanova
- Department of Physics and Biophysics, Faculty of Medicine, Trakia University, 11 Armeiska, 6000 Stara Zagora, Bulgaria; (B.P.); (G.S.); (B.T.)
| | - Gergana Savova
- Department of Physics and Biophysics, Faculty of Medicine, Trakia University, 11 Armeiska, 6000 Stara Zagora, Bulgaria; (B.P.); (G.S.); (B.T.)
| | - Bilyana Tacheva
- Department of Physics and Biophysics, Faculty of Medicine, Trakia University, 11 Armeiska, 6000 Stara Zagora, Bulgaria; (B.P.); (G.S.); (B.T.)
| | - Radostina Georgieva
- Department of Physics and Biophysics, Faculty of Medicine, Trakia University, 11 Armeiska, 6000 Stara Zagora, Bulgaria; (B.P.); (G.S.); (B.T.)
- Institute of Transfusion Medicine, Charité-Universitätsmedizin Berlin, Charitéplatz 1, 10117 Berlin, Germany
| |
Collapse
|
9
|
Miranda M, Brandsma E, Robben L, Van Dender H, van Alphen FPJ, Fijnvandraat K, van den Biggelaar M, Lacroix-Desmazes S, van Bruggen R, Voorberg J. Exploring red blood cells as an antigen delivery system to modulate the immune response towards FVIII in hemophilia A. J Thromb Haemost 2025; 23:836-848. [PMID: 39617188 DOI: 10.1016/j.jtha.2024.11.012] [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: 08/15/2024] [Revised: 10/23/2024] [Accepted: 11/14/2024] [Indexed: 03/10/2025]
Abstract
BACKGROUND The main complication in hemophilia A treatment is the development of inhibitory antibodies against factor (F)VIII. Immune tolerance induction, the gold standard for eradicating anti-FVIII antibodies, is efficient in only 60% to 80% of cases. This underscores the need for more efficient induction of tolerance in patients with hemophilia A with FVIII inhibitors. OBJECTIVES In this study, we explored whether red blood cells (RBCs) can be utilized as antigen delivery system to modulate the immune response against FVIII. METHODS Two promiscuously HLA-DR-presented peptides derived from the A2 and C1 domains of FVIII were fused to the TAT cell-penetrating peptide and incubated with RBCs. RESULTS Biotinylated TAT-A2 and TAT-C1 peptides were found to interact with RBCs as shown by flow cytometry and imaging flow cytometry. Moreover, macrophages efficiently phagocytosed TAT-FVIII peptide-treated RBCs. Using mass spectrometry-based immunopeptidomics we established that TAT-FVIII peptides were presented on major histocompatibility complex class II of macrophages that phagocytosed TAT peptide-pulsed RBCs. Specifically, the TAT-A2 peptide exhibited efficient processing and presentation on HLA-DR molecules. Importantly, incubation of TAT-C1 peptide-treated RBCs-loaded macrophages with a FVIII-specific T-cell hybridoma led to a significant increase in IL-2 production, suggesting functional presentation of TAT-C1-derived peptides by macrophages. CONCLUSION Our findings indicate that RBCs can serve as effective vehicle for the delivery of FVIII-derived peptides to antigen-presenting cells. The successful display of T-cell epitopes on antigen-presenting cell using ex vivo-loaded RBC may be potentially utilized to modulate pathogenic immune responses such as observed in a subset of patients with hemophilia A.
Collapse
Affiliation(s)
- Mariarosaria Miranda
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Eelke Brandsma
- Department of Hematopoiesis, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Lotte Robben
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Helena Van Dender
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Floris P J van Alphen
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Karin Fijnvandraat
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands; Department of Pediatric Hematology, Amsterdam University Medical Center Location, University of Amsterdam, Amsterdam, The Netherlands
| | - Maartje van den Biggelaar
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Sebastien Lacroix-Desmazes
- Institut National de la Santé et de la Recherche Médicale, Centre de Recherche des Cordeliers, CNRS, Sorbonne Université, Université Paris Cité, Paris, France
| | - Robin van Bruggen
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands
| | - Jan Voorberg
- Department of Molecular Hematology, Sanquin Research and Landsteiner Laboratory, Amsterdam, The Netherlands.
| |
Collapse
|
10
|
Zhu L, Ahn BC. Natural Killer Cell-Derived Exosome Mimetics as Natural Nanocarriers for In Vitro Delivery of Chemotherapeutics to Thyroid Cancer Cells. Exp Oncol 2025; 46:358-367. [PMID: 39985349 DOI: 10.15407/exp-oncology.2024.04.358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Indexed: 02/24/2025]
Abstract
BACKGROUND Exosomes have become a potential field of nanotechnology for the treatment and identification of many disorders. However, the generation of exosomes is a difficult, time-consuming, and low-yielding procedure. At the same time, exosome mimetics (EM) resemble exosomes in their characteristics but have higher production yields. The aim of this study was to produce natural killer (NK) cell-derived EM (NKEM) loaded with sorafenib and test their killing ability against thyroid cancer cell lines. MATERIALS AND METHODS Sorafenib was loaded into NKEM by mixing sorafenib with NK cells during NKEM production (NKEM-S). Then, these two types of nanoparticles were characterized with nanoparticle tracking analysis (NTA) to measure their sizes. In addition, the cellular uptake and in vitro killing effect of NKEM-S on thyroid cancer cell lines were investigated using confocal laser microscopy and bioluminescence imaging (BLI) techniques. RESULTS The uptake of NKEM and NKEM-S by the thyroid cancer cells was observed. Moreover, BLI confirmed the killing and anti-proliferation effect of NKEM-S on two thyroid cancer cell lines. Especially important, the NKEM-S demonstrated a desirable killing effect even for anaplastic thyroid cancer (ATC) cells. CONCLUSION Sorafenib-loaded NKEM showed the ability to kill thyroid cancer cells in vitro, even against ATC. This provides a new opportunity for drug delivery systems and thyroid cancer treatment.
Collapse
Affiliation(s)
- L Zhu
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, South Korea
| | - B-C Ahn
- Department of Nuclear Medicine, School of Medicine, Kyungpook National University, Daegu, South Korea
- Cardiovascular Research Institute, Kyungpook National University, Daegu, South Korea
- Department of Nuclear Medicine, Kyungpook National University Hospital, Daegu, South Korea
- BK21 FOUR KNU Convergence Educational Program of Biomedical Sciences for Creative Future Talents, Department of Biomedical Sciences, School of Medicine, Kyungpook National University, Daegu, South Korea
| |
Collapse
|
11
|
Sun L, Ma B, Yang F, Zou H, Guo Y, Wang X, Han M. Anti-hepatoma effect of homologous delivery of doxorubicin by HepG2 cells. Int J Pharm 2025; 670:125113. [PMID: 39710309 DOI: 10.1016/j.ijpharm.2024.125113] [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: 08/08/2024] [Revised: 12/13/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Compared to conventional polymer-based and biomaterial carriers, cells as vehicles for delivering bioactive molecules in the treatment of tumor diseases offer characteristics such as non-toxicity, biocompatibility, low immunogenicity, and prolonged in vivo circulation. However, the focus of current cell drug delivery systems predominantly lies on live cells, such as red blood cells, white blood cells and others. Here, a drug delivery strategy targeting liver cancer utilizing cryo-shocked liver cancer cells (HepG2) as carriers was presented, and non-proliferative HepG2 cells particles loaded with DOX (HepG2-DOX) was effectively prepared, which has good homologous targeting. Subsequent in vitro and in vivo experiments demonstrated the non-proliferative and non-pathogenic nature of this drug delivery system. The outcomes of in vitro experiments revealed that the inhibitory effect of HepG2-DOX on HepG2 was approximately five times higher than that of free DOX, with the IC50 value of HepG2-DOX being 0.0739 µg/mL and free DOX being 0.3606 µg/mL. Furthermore, in comparison to the positive DOX group, the HepG2-DOX group has a very significant advantage in tumor inhibition rate (91.34 % vs. 64.20 %). Cell uptake experiments indicated significant HepG2-DOX uptake by HepG2 cells compared to 4T1, LO2, and Raw cell groups, highlighting the excellent cell specificity of HepG2-DOX. Fluorescence imaging conducted in mice following the administration of HepG2-DOX demonstrated prompt drug localization within the tumor region, highlighting exceptional in vivo targeting precision. To sum up, this study introduced a novel strategy utilizing cryo-shocked liver cancer cells as a drug delivery system, effectively treating liver tumor by enhancing tumor targeting specificity.
Collapse
Affiliation(s)
- Lina Sun
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China; School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing 100029, China
| | - Baonan Ma
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Fangzhou Yang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Hang Zou
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Yifei Guo
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China
| | - Xiangtao Wang
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China.
| | - Meihua Han
- Institute of Medicinal Plant Development, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing 100193, China.
| |
Collapse
|
12
|
Skalickova M, Hadrava Vanova K, Uher O, Leischner Fialova J, Petrlakova K, Masarik M, Kejík Z, Martasek P, Pacak K, Jakubek M. Injecting hope: the potential of intratumoral immunotherapy for locally advanced and metastatic cancer. Front Immunol 2025; 15:1479483. [PMID: 39850897 PMCID: PMC11754201 DOI: 10.3389/fimmu.2024.1479483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/17/2024] [Indexed: 01/25/2025] Open
Abstract
Despite enormous progress, advanced cancers are still one of the most serious medical problems in current society. Although various agents and therapeutic strategies with anticancer activity are known and used, they often fail to achieve satisfactory long-term patient outcomes and survival. Recently, immunotherapy has shown success in patients by harnessing important interactions between the immune system and cancer. However, many of these therapies lead to frequent side effects when administered systemically, prompting treatment modifications or discontinuation or, in severe cases, fatalities. New therapeutic approaches like intratumoral immunotherapy, characterized by reduced side effects, cost, and systemic toxicity, offer promising prospects for future applications in clinical oncology. In the context of locally advanced or metastatic cancer, combining diverse immunotherapeutic and other treatment strategies targeting multiple cancer hallmarks appears crucial. Such combination therapies hold promise for improving patient outcomes and survival and for promoting a sustained systemic response. This review aims to provide a current overview of immunotherapeutic approaches, specifically focusing on the intratumoral administration of drugs in patients with locally advanced and metastatic cancers. It also explores the integration of intratumoral administration with other modalities to maximize therapeutic response. Additionally, the review summarizes recent advances in intratumoral immunotherapy and discusses novel therapeutic approaches, outlining future directions in the field.
Collapse
Affiliation(s)
- Marketa Skalickova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Katerina Hadrava Vanova
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Ondrej Uher
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Jindriska Leischner Fialova
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Katerina Petrlakova
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Michal Masarik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czechia
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Pavel Martasek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, United States
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec, Czechia
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague, Czechia
| |
Collapse
|
13
|
Cao L, Du M, Cai M, Feng Y, Miao J, Sun J, Song J, Du B. Neutrophil membrane-coated nanoparticles for targeted delivery of toll-like receptor 4 siRNA ameliorate LPS-induced acute lung injury. Int J Pharm 2025; 668:124960. [PMID: 39551221 DOI: 10.1016/j.ijpharm.2024.124960] [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: 10/29/2024] [Accepted: 11/13/2024] [Indexed: 11/19/2024]
Abstract
Pulmonary delivery of small interfering RNAs (siRNAs) is an effective treatment for acute lung injury (ALI), which can modulate the expression of pro-inflammatory cytokines and alleviate the symptoms of ALI. However, the rapid degradation of siRNA in vivo and its limited ability to target and validate cells are important challenges it faces in clinical practice. In this work, we developed neutrophil membrane-coated Poly (lactic-co-glycolic acid) nanoparticles loaded with TLR4 siRNA (si-TLR4) (Neutrophil-NP-TLR4), which can target both inflammatory and macrophage cells to alleviate the pulmonary inflammation in lipopolysaccharide (LPS)-induced ALI mice. These Neutrophil-NP-TLR4 effectively reduce the TNF-α and IL-1β expressions both in vitro and in vivo. Meanwhile, they also reduced the expression of TLR4, and its downstream genes including TNF receptor-associated factor 6 (TRAF6), X-linked inhibitor of apoptosis protein (XIAP), and Nuclear Factor kappa-B (NF-κB), but elevated the levels of Aquaporin 1 (AQP1) and Aquaporin 5 (AQP5). Moreover, the Neutrophil-NP-TLR4 precisely targets the inflammatory site to attenuate the lung injury without causing toxicity to normal tissue. This system provides a promising approach to effective delivery of siRNA to precisely treat the ALI.
Collapse
Affiliation(s)
- Liang Cao
- Department of ICU, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Min Du
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Mengmeng Cai
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Yan Feng
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Juanjuan Miao
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Jiafeng Sun
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Jie Song
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China
| | - Boxiang Du
- Department of Anesthesiology, Affiliated Hospital 2 of Nantong University, Nantong, Jiangsu 226014, China.
| |
Collapse
|
14
|
Aalhate M, Mahajan S, Dhuri A, Singh PK. Biohybrid nano-platforms manifesting effective cancer therapy: Fabrication, characterization, challenges and clinical perspective. Adv Colloid Interface Sci 2025; 335:103331. [PMID: 39522420 DOI: 10.1016/j.cis.2024.103331] [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: 03/13/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Nanotechnology-based delivery systems have brought a paradigm shift in the management of cancer. However, the main obstacles to nanocarrier-based delivery are their limited circulation duration, excessive immune clearance, inefficiency in interacting effectively in a biological context and overcoming biological barriers. This demands effective engineering of nanocarriers to achieve maximum efficacy. Nanocarriers can be maneuvered with biological components to acquire biological identity for further regulating their biodistribution and cell-to-cell cross-talk. Thus, the integration of synthetic and biological components to deliver therapeutic cargo is called a biohybrid delivery system. These delivery systems possess the advantage of synthetic nanocarriers, such as high drug loading, engineerable surface, reproducibility, adequate communication and immune evasion ability of biological constituents. The biohybrid delivery vectors offer an excellent opportunity to harness the synergistic properties of the best entities of the two worlds for improved therapeutic outputs. The major spotlights of this review are different biological components, synthetic counterparts of biohybrid nanocarriers, recent advances in hybridization techniques, and the design of biohybrid delivery systems for cancer therapy. Moreover, this review provides an overview of biohybrid systems with therapeutic and diagnostic applications. In a nutshell, this article summarizes the advantages and limitations of various biohybrid nano-platforms, their clinical potential and future directions for successful translation in cancer management.
Collapse
Affiliation(s)
- Mayur Aalhate
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Srushti Mahajan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Anish Dhuri
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India
| | - Pankaj Kumar Singh
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research, (NIPER), Hyderabad 500037, India.
| |
Collapse
|
15
|
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 2025; 9:e2400096. [PMID: 38461538 DOI: 10.1002/smtd.202400096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [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
|
16
|
Wu S, Zhu L, Ni S, Zhao X, Yan M, Zhong Y, Kusuma K, Liang Y, Bao K, Qu K, Qin X, Zhang K, Deng W, Sun D, Wu W. Hemoglobin integrated red blood cell membrane-coated metal-organic framework nano-platform for improving the self-adaptive blood glucose management. NANO RESEARCH 2025; 18:94907078. [DOI: 10.26599/nr.2025.94907078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2025]
|
17
|
Meng X, Wang X, Zhang Z, Song L, Chen J. Recent Advancements of Nanomedicine in Breast Cancer Surgery. Int J Nanomedicine 2024; 19:14143-14169. [PMID: 39759962 PMCID: PMC11699852 DOI: 10.2147/ijn.s494364] [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: 09/03/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
Breast cancer surgery plays a pivotal role in the multidisciplinary approaches. Surgical techniques and objectives are gradually shifting from tumor complete resection towards prolonging survival, improving cosmetic outcomes, and restoring the social and psychological well-being of patients. However, surgical treatment still faces challenges such as inadequate sensitivity in sentinel lymph node localization, the need to improve intraoperative tumor boundary localization imaging, postoperative scar healing, and the risk of recurrence, necessitating other adjunct measures for improvement. To address these challenges, specificity-optimized nanomedicines have been introduced into the surgical therapeutic landscape of breast cancer. In particular, this review involves starting with an overview of breast structure and the composition of the tumor microenvironment and then introducing the guiding principle and foundation for the design of nanomedicine. Moreover, we will take the order process of breast cancer surgery diagnosis and treatment as the starting point, and adaptively propose the roles and advantages of nanomedicine in addressing the corresponding issues. Furthermore, we also involved the prospects of utilizing advanced technological approaches. Overall, this review seeks to uncover the sophisticated design and strategies of nanomedicine from a clinical standpoint, address the challenges faced in surgical treatment, and provide insights into this subject matter.
Collapse
Affiliation(s)
- Xiangyue Meng
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Xin Wang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Zhihao Zhang
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| | - Linlin Song
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, 610041, People’s Republic of China
- Department of Ultrasound, Laboratory of Ultrasound Medicine, West China Hospital, Sichuan University, Chengdu, Sichuan, 610041, People’s Republic of China
| | - Jie Chen
- Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
- Breast Center, West China Hospital, Sichuan University, Chengdu, 610041, People’s Republic of China
| |
Collapse
|
18
|
He Y, Zhang S, She Y, Liu Z, Zhu Y, Cheng Q, Ji X. Innovative utilization of cell membrane-coated nanoparticles in precision cancer therapy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230164. [PMID: 39713200 PMCID: PMC11655310 DOI: 10.1002/exp.20230164] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 03/03/2024] [Indexed: 12/24/2024]
Abstract
Cell membrane-coated nanoparticles (CMNPs) have recently emerged as a promising platform for cancer therapy. By encapsulating therapeutic agents within a cell membrane-derived coating, these nanoparticles combine the advantages of synthetic nanoparticles and natural cell membranes. This review provides a comprehensive overview of the recent advancements in utilizing CMNPs as effective drug delivery vehicles for cancer therapy. The synthesis and fabrication methods of CMNPs are comprehensively discussed. Various techniques, such as extrusion, sonication, and self-assembly, are employed to coat synthetic nanoparticles with cell membranes derived from different cell types. The cell membrane coating enables biocompatibility, reducing the risk of an immune response and enhancing the stability of the nanoparticles in the bloodstream. Moreover, functionalization strategies for CMNPs, primarily chemical modification, genetic engineering, and external stimuli, are highlighted. The presence of specific cell surface markers on the coated membrane allows targeted drug delivery to cancer cells and maximizes therapeutic efficacy. Preclinical studies utilizing CMNPs for cancer therapy demonstrated the successful delivery of various therapeutic agents, such as chemotherapeutic drugs, nucleic acids, and immunotherapeutic agents, using CMNPs. Furthermore, the article explores the future directions and challenges of this technology while offering insights into its clinical potential.
Collapse
Affiliation(s)
- Yiling He
- Department of PharmacyJinhua Municipal Central HospitalJinhuaZhejiangChina
| | - Shuquan Zhang
- Department of OrthopedicsIntegrated Chinese and Western Medicine HospitalTianjin UniversityTianjinChina
- Department of OrthopedicsTianjin Nankai HospitalTianjinChina
- Department of OrthopedicsTianjin Hospital of Integrated Chinese and Western MedicineTianjinChina
| | - Yaoguang She
- Department of General SurgeryFirst Medical CenterChinese People's Liberation Army (PLA) General HospitalBeijingChina
| | - Zhaoshan Liu
- Laboratory of Immune Cell BiologyCenter for Cancer ResearchNational Cancer InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Yalan Zhu
- Department of PharmacyJinhua Municipal Central HospitalJinhuaZhejiangChina
| | - Qinzhen Cheng
- Department of PharmacyJinhua Municipal Central HospitalJinhuaZhejiangChina
| | - Xiaoyuan Ji
- Academy of Medical Engineering and Translational MedicineMedical CollegeTianjin UniversityTianjinChina
| |
Collapse
|
19
|
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
|
20
|
Tong J, Wang Z, Zhang J, Gao R, Liu X, Liao Y, Guo X, Wei Y. Advanced Applications of Nanomaterials in Atherosclerosis Diagnosis and Treatment: Challenges and Future Prospects. ACS APPLIED MATERIALS & INTERFACES 2024; 16:58072-58099. [PMID: 39432384 DOI: 10.1021/acsami.4c13607] [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: 10/23/2024]
Abstract
Atherosclerosis-induced coronary artery disease is a major cause of cardiovascular mortality. Clinically, conservative treatment strategies for atherosclerosis still focus on lifestyle interventions and the use of lipid-lowering and anticoagulant medications. Despite achieving some therapeutic effects, these approaches are limited by low bioavailability, long intervention periods, and significant side effects. With the advancement of nanotechnology, nanomaterials have demonstrated extraordinary potential in the biomedical field. Their excellent biocompatibility, surface modifiability, and high targeting capability not only enable efficient diagnosis of plaque progression but also allow precise drug delivery within atherosclerotic plaques, significantly enhancing drug bioavailability and reducing systemic side effects. Here, we systematically review the current research progress of nanomaterials in the field of atherosclerosis to summarize not only the types of nanomaterials but also their applications in both the diagnosis and treatment of atherosclerosis. Notably, in the context of plaque therapy, we provide a comprehensive overview of current nanomaterial applications based on their targeted therapeutic systems for different cell types within plaques. Additionally, we address the persistent challenge of clinical translation of nanomaterials by summarizing current issues and providing directions for innovation and improvement in nanomaterial design. Overall, we believe that this review systematically summarizes the applications and challenges of biomedical nanomaterials in atherosclerosis diagnosis and therapy, thereby offering insights and references for the development of therapeutic materials for atherosclerosis.
Collapse
Affiliation(s)
- Junran Tong
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zhiwen Wang
- Department of Nephrology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Jiahui Zhang
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Ran Gao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiangfei Liu
- Department of Cardiology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100005, China
| | - Yuhan Liao
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xiaopeng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Yumiao Wei
- Department of Cardiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Engineering Research Center for Immunological Diagnosis and Therapy of Cardiovascular Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
21
|
Song Q, Li J, Li T, Li H. Nanomaterials that Aid in the Diagnosis and Treatment of Alzheimer's Disease, Resolving Blood-Brain Barrier Crossing Ability. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403473. [PMID: 39101248 PMCID: PMC11481234 DOI: 10.1002/advs.202403473] [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: 05/06/2024] [Revised: 07/04/2024] [Indexed: 08/06/2024]
Abstract
As a form of dementia, Alzheimer's disease (AD) suffers from no efficacious cure, yet AD treatment is still imperative, as it ameliorates the symptoms or prevents it from deteriorating or maintains the current status to the longest extent. The human brain is the most sensitive and complex organ in the body, which is protected by the blood-brain barrier (BBB). This yet induces the difficulty in curing AD as the drugs or nanomaterials that are much inhibited from reaching the lesion site. Thus, BBB crossing capability of drug delivery system remains a significant challenge in the development of neurological therapeutics. Fortunately, nano-enabled delivery systems possess promising potential to achieve multifunctional diagnostics/therapeutics against various targets of AD owing to their intriguing advantages of nanocarriers, including easy multifunctionalization on surfaces, high surface-to-volume ratio with large payloads, and potential ability to cross the BBB, making them capable of conquering the limitations of conventional drug candidates. This review, which focuses on the BBB crossing ability of the multifunctional nanomaterials in AD diagnosis and treatment, will provide an insightful vision that is conducive to the development of AD-related nanomaterials.
Collapse
Affiliation(s)
- Qingting Song
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Junyou Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Ting Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| | - Hung‐Wing Li
- Department of ChemistryThe Chinese University of Hong KongHong KongChina
| |
Collapse
|
22
|
Lan J, Chen L, Li Z, Liu L, Zeng R, He Y, Shen Y, Zhang T, Ding Y. Multifunctional Biomimetic Liposomes with Improved Tumor-Targeting for TNBC Treatment by Combination of Chemotherapy, Antiangiogenesis and Immunotherapy. Adv Healthc Mater 2024; 13:e2400046. [PMID: 38767575 DOI: 10.1002/adhm.202400046] [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: 01/05/2024] [Revised: 04/11/2024] [Indexed: 05/22/2024]
Abstract
Triple negative breast cancer (TNBC) featuring high relapses and metastasis shows limited clinical therapeutic efficiency with chemotherapy for the extremely complex tumor microenvironment, especially angiogenesis and immunosuppression. Combination of antiangiogenesis and immunotherapy holds promise for effective inhibition of tumor proliferation and invasion, while it remains challenging for specific targeting drug delivery to tumors and metastatic lesions. Here, a multifunctional biomimetic liposome loading Gambogic acid (G/R-MLP) is developed using Ginsenoside Rg3 (Rg3) to substitute cholesterol and cancer cell membrane coating, which is designed to increase long-circulating action by a low immunogenicity and specifically deliver gambogic acid (GA) to tumor site and metastatic lesions by homologous targeting and glucose transporter targeting. After G/R-MLP accumulates in the primary tumors and metastatic nodules, it synergistically enhances the antitumor efficacy of GA, effectively suppressing the tumor growth and lung metastasis by killing tumor cells, inhibiting tumor cell migration and invasion, achieving antiangiogenesis and improving the antitumor immunity. All in all, the strategy combining chemotherapy, antiangiogenesis, and immunotherapy improves therapeutic efficiency and prolonged survival, providing a new perspective for the clinical treatment of TNBC.
Collapse
Affiliation(s)
- Jinshuai Lan
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Lixia Chen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Zhe Li
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Li Liu
- Institute of Interdisciplinary Integrative Medicine Research, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Ruifeng Zeng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yitian He
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yi Shen
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Tong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yue Ding
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- The MOE Innovation Centre for Basic Medicine Research on Qi-Blood TCM Theories, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
- State Key Laboratory of Integration and Innovation of Classic Formula and Modern Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| |
Collapse
|
23
|
Zhou R, Xue S, Cheng Y, Chen Y, Wang Y, Xing J, Liu H, Xu Y, Lin Y, Pei Z, Wei X, Ding J, Li S, Wang K, Yao F, Zhao Y, Ding C, Hu W. Macrophage membrane-camouflaged biomimetic nanoparticles for rheumatoid arthritis treatment via modulating macrophage polarization. J Nanobiotechnology 2024; 22:578. [PMID: 39300463 DOI: 10.1186/s12951-024-02822-9] [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: 05/26/2024] [Accepted: 08/30/2024] [Indexed: 09/22/2024] Open
Abstract
Rheumatoid arthritis (RA) is a debilitating autoimmune disease characterized by chronic joint inflammation and cartilage damage. Current therapeutic strategies often result in side effects, necessitating the development of targeted and safer treatment options. This study introduces a novel nanotherapeutic system, 2-APB@DGP-MM, which utilizes macrophage membrane (MM)-encapsulated nanoparticles (NPs) for the targeted delivery of 2-Aminoethyl diphenylborinate (2-APB) to inflamed joints more effectively. The NPs are designed with a matrix metalloproteinase (MMP)-cleavable peptide, allowing for MMP-responsive drug release within RA microenvironment. Comprehensive in vitro and in vivo assays confirmed the successful synthesis and loading of 2-APB into the DSPE-GPLGVRGC-PEG (DGP) NPs, as well as their ability to repolarize macrophages from a pro-inflammatory M1 to an anti-inflammatory M2 phenotype. The NPs demonstrated high biocompatibility, low cytotoxicity, and enhanced cellular uptake. In a collagen-induced arthritis (CIA) mouse model, intra-articular injection of 2-APB@DGP-MM significantly reduced synovial inflammation and cartilage destruction. Histological analysis corroborated these findings, demonstrating marked improvements in joint structure and delayed disease progression. Above all, the 2-APB@DGP-MM nanotherapeutic system offers a promising and safe approach for RA treatment by modulating macrophage polarization and delivering effective agents to inflamed joints.
Collapse
Affiliation(s)
- Renpeng Zhou
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China
| | - Song Xue
- Department of Sports Medicine, Peking University Shenzhen Hospital, Shenzhen Peking University-The Hong Kong University of Science and Technology Medical Center, Shenzhen, 518036, China
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510200, China
| | - Yuanzhi Cheng
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Yong Chen
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yan Wang
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Jing Xing
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Hao Liu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Yucai Xu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Yi Lin
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Zejun Pei
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Xin Wei
- Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, People's Republic of China
| | - Jie Ding
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China
| | - Shufang Li
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Ke Wang
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Feng Yao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Yingjie Zhao
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China.
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| | - Changhai Ding
- Clinical Research Centre, Zhujiang Hospital, Southern Medical University, Guangzhou, 510200, China.
- Menzies Institute for Medical Research, University of Tasmania, Hobart, TAS, Australia.
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China.
- The Key Laboratory of Major Autoimmune Diseases, School of Pharmacy, Anhui Institute of Innovative Drugs, Anhui Medical University, Hefei, 230032, China.
- The Key Laboratory of Anti-Inflammatory and Immune Medicine, Anhui Medical University, Ministry of Education, Hefei, 230032, China.
| |
Collapse
|
24
|
Coronado S, Herrera J, Pino MG, Martín S, Ballesteros-Rueda L, Cea P. Advancements in Engineering Planar Model Cell Membranes: Current Techniques, Applications, and Future Perspectives. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1489. [PMID: 39330645 PMCID: PMC11434481 DOI: 10.3390/nano14181489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/28/2024] [Accepted: 09/05/2024] [Indexed: 09/28/2024]
Abstract
Cell membranes are crucial elements in living organisms, serving as protective barriers and providing structural support for cells. They regulate numerous exchange and communication processes between cells and their environment, including interactions with other cells, tissues, ions, xenobiotics, and drugs. However, the complexity and heterogeneity of cell membranes-comprising two asymmetric layers with varying compositions across different cell types and states (e.g., healthy vs. diseased)-along with the challenges of manipulating real cell membranes represent significant obstacles for in vivo studies. To address these challenges, researchers have developed various methodologies to create model cell membranes or membrane fragments, including mono- or bilayers organized in planar systems. These models facilitate fundamental studies on membrane component interactions as well as the interactions of membrane components with external agents, such as drugs, nanoparticles (NPs), or biomarkers. The applications of model cell membranes have extended beyond basic research, encompassing areas such as biosensing and nanoparticle camouflage to evade immune detection. In this review, we highlight advancements in the engineering of planar model cell membranes, focusing on the nanoarchitectonic tools used for their fabrication. We also discuss approaches for incorporating challenging materials, such as proteins and enzymes, into these models. Finally, we present our view on future perspectives in the field of planar model cell membranes.
Collapse
Affiliation(s)
- Sara Coronado
- Departamento de Química Física, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Centro de Investigaciones en Catálisis (CICAT), Escuela de Ingeniería Química, Universidad Industrial de Santander, Parque Tecnológico de Guatiguará, Km 2 vía El Refugio, Piedecuesta, Santander 681911, Colombia
| | - Johan Herrera
- Departamento de Química Física, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Centro de Investigaciones en Catálisis (CICAT), Escuela de Ingeniería Química, Universidad Industrial de Santander, Parque Tecnológico de Guatiguará, Km 2 vía El Refugio, Piedecuesta, Santander 681911, Colombia
| | - María Graciela Pino
- Departamento de Química Física, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Santiago Martín
- Departamento de Química Física, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Luz Ballesteros-Rueda
- Departamento de Química Física, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
- Centro de Investigaciones en Catálisis (CICAT), Escuela de Ingeniería Química, Universidad Industrial de Santander, Parque Tecnológico de Guatiguará, Km 2 vía El Refugio, Piedecuesta, Santander 681911, Colombia
| | - Pilar Cea
- Departamento de Química Física, Facultad de Ciencias, Universidad de Zaragoza, Pedro Cerbuna 12, 50009 Zaragoza, Spain
| |
Collapse
|
25
|
Sulthana S, Shrestha D, Aryal S. Maximizing liposome tumor delivery by hybridizing with tumor-derived extracellular vesicles. NANOSCALE 2024; 16:16652-16663. [PMID: 39171636 PMCID: PMC11401241 DOI: 10.1039/d4nr02191f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/23/2024]
Abstract
Extracellular vesicles (EVs) have gained widespread interest due to their potential in the diagnosis and treatment of inflammation, autoimmune diseases, and cancers. EVs are lipidic vesicles comprising vesicles of endosomal origin called exosomes, microvesicles from membrane shedding, and apoptotic bodies from programmed cell death membrane blebbing that carry complex sets of cargo from their cells of origin, including proteins, lipids, mRNA, and DNA. EVs are rich in integrin proteins that facilitate intrinsic cellular communication to deliver their cargo contents and can also be used as biomarkers to study respective cellular conditions. Within this background, we hypothesized that when these EVs are hybridized with synthetic liposomes, it would help navigate the hybrid construct in the complex biological environment to find its target. Toward this endeavor, we have hybridized a synthetic liposome with EVs (herein called LEVs) derived from mouse breast cancer (4T1 tumors) cells and incorporated a rhodamine-B/near-infrared fluorescent dye to investigate their potential for cellular targeting and tumor delivery. Using membrane extrusion, we have successfully hybridized both entities resulting in the formation of LEVs and characterized their colloidal properties and stability over a period. While EVs are broadly dispersed nano- and micron-sized vesicles, LEVs are engineered as monodispersed with an average hydrodynamic size of 140 ± 5. Using immunoblotting and ELISA, we monitored and quantified the EV-specific protein CD63 and other characteristic proteins such as CD9 and CD81, which were taken as a handle to ensure the reproducibility of EVs and thus LEVs. These LEVs were further challenged with mice bearing orthotopic 4T1 breast tumors and the LEV uptake was found to be maximum in tumors and organs like the liver, spleen, and lungs when compared to control PEGylated liposomes in live animal imaging. Likewise, the constructs were capable of finding lung metastasis as observed in ex vivo imaging. We anticipate that this study can open avenues for drug delivery solutions that are superior in target recognition.
Collapse
Affiliation(s)
- Shoukath Sulthana
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX 75799, USA.
| | - Dinesh Shrestha
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX 75799, USA.
| | - Santosh Aryal
- Department of Pharmaceutical Sciences and Health Outcomes, The Ben and Maytee Fisch College of Pharmacy, University of Texas at Tyler, Tyler, TX 75799, USA.
| |
Collapse
|
26
|
Kumar J, Karim A, Sweety UH, Sarma H, Nurunnabi M, Narayan M. Bioinspired Approaches for Central Nervous System Targeted Gene Delivery. ACS APPLIED BIO MATERIALS 2024; 7:4975-4997. [PMID: 38100377 DOI: 10.1021/acsabm.3c00842] [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] [Indexed: 12/17/2023]
Abstract
Disorders of the central nervous system (CNS) which include a wide range of neurodegenerative and neurological conditions have become a serious global issue. The presence of CNS barriers poses a significant challenge to the progress of designing effective therapeutic delivery systems, limiting the effectiveness of drugs, genes, and other therapeutic agents. Natural nanocarriers present in biological systems have inspired researchers to design unique delivery systems through biomimicry. As natural resource derived delivery systems are more biocompatible, current research has been focused on the development of delivery systems inspired by bacteria, viruses, fungi, and mammalian cells. Despite their structural potential and extensive physiological function, making them an excellent choice for biomaterial engineering, the delivery of nucleic acids remains challenging due to their instability in biological systems. Similarly, the efficient delivery of genetic material within the tissues of interest remains a hurdle due to a lack of selectivity and targeting ability. Considering that gene therapies are the holy grail for intervention in diseases, including neurodegenerative disorders such as Alzheimer's disease, Parkinson's Disease, and Huntington's disease, this review centers around recent advances in bioinspired approaches to gene delivery for the prevention of CNS disorders.
Collapse
Affiliation(s)
- Jyotish Kumar
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Afroz Karim
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Ummy Habiba Sweety
- Environmental Science and Engineering, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| | - Hemen Sarma
- Bioremediation Technology Research Group, Department of Botany, Bodoland University, Rangalikhata, Deborgaon, 783370, Kokrajhar (BTR), Assam, India
| | - Md Nurunnabi
- The Department of Pharmaceutical Sciences, School of Pharmacy, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, The University of Texas at El Paso (UTEP), El Paso, Texas 79968, United States
| |
Collapse
|
27
|
Chen Y, Li X, Shang H, Sun Y, Wang C, Wang X, Tian H, Yang H, Zhang L, Deng L, Yang K, Wu B, Cheng W. Mechanism exploration of synergistic photo-immunotherapy strategy based on a novel exosome-like nanosystem for remodeling the immune microenvironment of HCC. NANO CONVERGENCE 2024; 11:31. [PMID: 39141072 PMCID: PMC11324638 DOI: 10.1186/s40580-024-00441-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 07/30/2024] [Indexed: 08/15/2024]
Abstract
The immunosuppressive tumor microenvironment (TME) has become a major challenge in cancer immunotherapy, with abundant tumor-associated macrophages (TAMs) playing a key role in promoting tumor immune escape by displaying an immunosuppressive (M2) phenotype. Recently, it was reported that M1 macrophage-derived nanovesicles (M1NVs) can reprogram TAMs to an anti-tumor M1 phenotype, thereby significantly alleviating the immunosuppressive TME and enhancing the anti-tumor efficacy of immunotherapy. Herein, we developed M1NVs loaded with mesoporous dopamine (MPDA) and indocyanine green (ICG), which facilitated the recruitment of M2 TAMs through synergistic photothermal and photodynamic therapy. Thereafter, M1NVs can induce M1 repolarization of TAMs, resulting in increased infiltration of cytotoxic T lymphocytes within the tumor to promote tumor regression. This study investigated the effect of phototherapy on the immune environment of liver cancer using single-cell RNA sequencing (scRNA-seq) by comparing HCC tissues before and after MPDA/ICG@M1NVs + NIR treatment. The results showed significant shifts in cell composition and gene expression, with decreases in epithelial cells, B cells, and macrophages and increases in neutrophils and myeloid cells. Additionally, gene analysis indicated a reduction in pro-inflammatory signals and immunosuppressive functions, along with enhanced B-cell function and anti-tumor immunity, downregulation of the Gtsf1 gene in the epithelial cells of the MPDA/ICG @M1NVs + NIR group, and decreased expression of the lars2 gene in immune subpopulations. Eno3 expression is reduced in M1 macrophages, whereas Clec4a3 expression is downregulated in M2 macrophages. Notably, the B cell population decreased, whereas Pou2f2 expression increased. These genes regulate cell growth, death, metabolism, and tumor environment, indicating their key role in HCC progression. This study highlights the potential for understanding cellular and molecular dynamics to improve immunotherapy.
Collapse
Affiliation(s)
- Yichi Chen
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Xudong Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China
| | - Haitao Shang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Yucao Sun
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Chunyue Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Xiaodong Wang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Huimin Tian
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Huajing Yang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Lei Zhang
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Liwen Deng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China
| | - Kuikun Yang
- School of Life Science and Technology, Harbin Institute of Technology, No. 92, West Dazhi Street, Nangang District, Harbin, Heilongjiang, 150080, P. R. China.
| | - Bolin Wu
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China.
| | - Wen Cheng
- Department of Ultrasound, Harbin Medical University Cancer Hospital, No.150, Haping Road, Nangang District, Harbin, 150081, China.
| |
Collapse
|
28
|
Zheng B, Shi Y, Xiao L, Li B, Chen Z, Zhao J, Li S, Hou H, Li J, Cai X, Wang H, Wu P, Zheng X. Simultaneously Modulating HIF-1α and HIF-2α and Optimizing Macrophage Polarization through the Biomimetic Gene Vector toward the Treatment of Osteoarthritis. Biomater Res 2024; 28:0059. [PMID: 39076894 PMCID: PMC11283864 DOI: 10.34133/bmr.0059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Accepted: 06/20/2024] [Indexed: 07/31/2024] Open
Abstract
In osteoarthritis (OA), articular cartilage is continuously submerged in a hypoxic environment throughout life, and hypoxia-inducible factors (HIFs) play a crucial role in OA progression. Among the various HIF phenotypes, HIF-1α positively contributes to maintaining the stability of the articular cartilage matrix. In contrast, HIF-2α has a detrimental effect, leading to chondrocyte apoptosis and exacerbating inflammation. Notably, there is currently no simultaneous regulation of HIF-1α and HIF-2α for OA treatment. Thus, the biomimetic gene vector (MENP) was developed for co-delivery of siHIF-2α and Mg2+ to the inflamed regions in OA joints, comprising an inner core consisting of siHIF-2α and Mg2+ and an outer M2 macrophage membrane. In vitro and in vivo studies demonstrate that MENP effectively targets inflamed areas, efficiently silences HIF-2α, and facilitates HIF-1α-mediated cartilage restoration through Mg2+. Furthermore, it indirectly promotes the polarization of macrophages toward an anti-inflammatory M2 phenotype through its action on inflamed synoviocytes. Overall, MENP is an efficient biomimetic vehicle for alleviating inflammation and promoting cartilage repair, representing an appealing approach for OA treatment.
Collapse
Affiliation(s)
- Boyuan Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| | - Yiwan Shi
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| | - Lei Xiao
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| | - Bowei Li
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| | - Zihang Chen
- Department of Psychology, Li Ka Shing Faculty of Medicine, State Key Laboratory of Brain and Cognitive Sciences, The University of Hong Kong, Hong Kong SAR, China
| | - Jing Zhao
- Joint Laboratory of Chinese Herbal Glycoengineering and Testing Technology, University of Macau and National Glycoengineering Research Center, Macao, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China
| | - Shaoping Li
- Joint Laboratory of Chinese Herbal Glycoengineering and Testing Technology, University of Macau and National Glycoengineering Research Center, Macao, China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Macao, China
| | - Huige Hou
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| | - Jieruo Li
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| | - Xianlong Cai
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| | - Huajun Wang
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| | - Peng Wu
- Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, 200072 Shanghai, China
| | - Xiaofei Zheng
- Department of Sports Medicine, The First Affiliated Hospital, Guangdong Provincial Key Laboratory of Speed Capability, The Guangzhou Key Laboratory of Precision Orthopedics and Regenerative Medicine, State Key Laboratory of Frigid Zone Cardiovascular Diseases, Jinan University, 510630 Guangzhou, China
| |
Collapse
|
29
|
Ye J, Yu Y, Li Y, Yao B, Gu M, Li Y, Yin S. Nanoparticles Encapsulated in Red Blood Cell Membranes for Near-Infrared Second Window Imaging-Guided Photothermal-Enhanced Immunotherapy on Tumors. ACS APPLIED MATERIALS & INTERFACES 2024; 16:34607-34619. [PMID: 38924764 DOI: 10.1021/acsami.4c05334] [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: 06/28/2024]
Abstract
Photothermal therapy (PTT), which uses the high thermal conversion ability of photothermal agents to ablate tumor cells at high temperatures, has gained significant attention because it has the advantages of high selectivity and specificity, precise targeting of tumor sites, and low invasiveness and trauma. However, PTT guided by the NIR-I has limitations in tissue penetration depth, resulting in limited imaging monitoring and therapeutic effects on deep-seated tumor tissues. Moreover, nanoparticles are easily cleared by the immune system and difficult to passively target tumor sites during the process of treatment. To address these issues, we prepared nanoparticles using NIR-II dyes IR1048 and DSPE-PEG-OH and further encapsulated them in red blood cell membranes derived from mice. These biomimetic nanoparticles, called RDIR1048, showed reduced clearance by the immune system and had long circulation characteristics. They effectively accumulated at tumor sites, and strong fluorescence could still be observed at the tumor site 96 h after administration. Furthermore, through mouse thermal imaging experiments, we found that RDIR1048 exhibited good PTT ability. When used in combination with an immune checkpoint inhibitor, anti-PD-L1 antibodies, it enhanced the immunogenic cell death of tumor cells caused by PTT and improved the therapeutic effect of immunotherapy, which demonstrated good therapeutic efficacy in the treatment of tumor-bearing mice. This study provides a feasible basis for the future development of NIR-II nanoparticles with long circulation properties.
Collapse
Affiliation(s)
- Jingtao Ye
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Yanlu Yu
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Yaojun Li
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Bo Yao
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Meier Gu
- Laboratory Animal Center, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Yang Li
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| | - Shouchun Yin
- Key Laboratory of Organosilicon Chemistry and Materials Technology of Ministry of Education, College of Materials, Chemistry and Chemical Engineering, Hangzhou Normal University, Hangzhou 311121, P. R. China
| |
Collapse
|
30
|
Baig MS, Ahmad A, Pathan RR, Mishra RK. Precision Nanomedicine with Bio-Inspired Nanosystems: Recent Trends and Challenges in Mesenchymal Stem Cells Membrane-Coated Bioengineered Nanocarriers in Targeted Nanotherapeutics. J Xenobiot 2024; 14:827-872. [PMID: 39051343 PMCID: PMC11270309 DOI: 10.3390/jox14030047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/09/2024] [Accepted: 06/15/2024] [Indexed: 07/27/2024] Open
Abstract
In the recent past, the formulation and development of nanocarriers has been elaborated into the broader fields and opened various avenues in their preclinical and clinical applications. In particular, the cellular membrane-based nanoformulations have been formulated to surpass and surmount the limitations and restrictions associated with naïve or free forms of therapeutic compounds and circumvent various physicochemical and immunological barriers including but not limited to systemic barriers, microenvironmental roadblocks, and other cellular or subcellular hinderances-which are quite heterogeneous throughout the diseases and patient cohorts. These limitations in drug delivery have been overcome through mesenchymal cells membrane-based precision therapeutics, where these interventions have led to the significant enhancements in therapeutic efficacies. However, the formulation and development of nanocarriers still focuses on optimization of drug delivery paradigms with a one-size-fits-all resolutions. As mesenchymal stem cell membrane-based nanocarriers have been engineered in highly diversified fashions, these are being optimized for delivering the drug payloads in more and better personalized modes, entering the arena of precision as well as personalized nanomedicine. In this Review, we have included some of the advanced nanocarriers which have been designed and been utilized in both the non-personalized as well as precision applicability which can be employed for the improvements in precision nanotherapeutics. In the present report, authors have focused on various other aspects of the advancements in stem cells membrane-based nanoparticle conceptions which can surmount several roadblocks and barriers in drug delivery and nanomedicine. It has been suggested that well-informed designing of these nanocarriers will lead to appreciable improvements in the therapeutic efficacy in therapeutic payload delivery applications. These approaches will also enable the tailored and customized designs of MSC-based nanocarriers for personalized therapeutic applications, and finally amending the patient outcomes.
Collapse
Affiliation(s)
- Mirza Salman Baig
- Anjuman-I-Islam Kalsekar Technical Campus School of Pharmacy, Sector-16, Near Thana Naka, Khandagao, New Panvel, Navi Mumbai 410206, Maharashtra, India;
| | - Anas Ahmad
- Julia McFarlane Diabetes Research Centre (JMDRC), Department of Microbiology, Immunology and Infectious Diseases, Snyder Institute for Chronic Diseases, Hotchkiss Brain Institute, Cumming School of Medicine, Foothills Medical Centre, University of Calgary, Calgary, AB T2N 4N1, Canada
| | | | - Rakesh Kumar Mishra
- School of Health Sciences and Technology, University of Petroleum and Energy Studies (UPES), Bidholi, Dehradun 248007, Uttarakhand, India;
| |
Collapse
|
31
|
Shahid S, Khan A, Shahid W, Rehan M, Asif R, Nisar H, Kanwal Q, Choi JR. Nanoenzymes: A Radiant Hope for the Early Diagnosis and Effective Treatment of Breast and Ovarian Cancers. Int J Nanomedicine 2024; 19:5813-5835. [PMID: 38895143 PMCID: PMC11184228 DOI: 10.2147/ijn.s460712] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Accepted: 04/16/2024] [Indexed: 06/21/2024] Open
Abstract
Breast and ovarian cancers, despite having chemotherapy and surgical treatment, still have the lowest survival rate. Experimental stages using nanoenzymes/nanozymes for ovarian cancer diagnosis and treatment are being carried out, and correspondingly the current treatment approaches to treat breast cancer have a lot of adverse side effects, which is the reason why researchers and scientists are looking for new strategies with less side effects. Nanoenzymes have intrinsic enzyme-like activities and can reduce the shortcomings of naturally occurring enzymes due to the ease of storage, high stability, less expensive, and enhanced efficiency. In this review, we have discussed various ways in which nanoenzymes are being used to diagnose and treat breast and ovarian cancer. For breast cancer, nanoenzymes and their multi-enzymatic properties can control the level of reactive oxygen species (ROS) in cells or tissues, for example, oxidase (OXD) and peroxidase (POD) activity can be used to generate ROS, while catalase (CAT) or superoxide dismutase (SOD) activity can scavenge ROS. In the case of ovarian cancer, most commonly nanoceria is being investigated, and also when folic acid is combined with nanoceria there are additional advantages like inhibition of beta galactosidase. Nanocarriers are also used to deliver small interfering RNA that are effective in cancer treatment. Studies have shown that iron oxide nanoparticles are actively being used for drug delivery, similarly ferritin carriers are used for the delivery of nanozymes. Hypoxia is a major factor in ovarian cancer, therefore MnO2-based nanozymes are being used as a therapy. For cancer diagnosis and screening, nanozymes are being used in sonodynamic cancer therapy for cancer diagnosis and screening, whereas biomedical imaging and folic acid gold particles are also being used for image guided treatments. Nanozyme biosensors have been developed to detect ovarian cancer. This review article summarizes a detailed insight into breast and ovarian cancers in light of nanozymes-based diagnostic and therapeutic approaches.
Collapse
Affiliation(s)
- Samiah Shahid
- Research Centre for Health Sciences (RCHS), The University of Lahore, Lahore, Pakistan
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Ayesha Khan
- Research Centre for Health Sciences (RCHS), The University of Lahore, Lahore, Pakistan
| | - Wajeehah Shahid
- Department of Physics, The University of Lahore, Lahore, Pakistan
| | - Mehvesh Rehan
- Institute of Molecular Biology and Biotechnology (IMBB), The University of Lahore, Lahore, Pakistan
| | - Roha Asif
- Research Centre for Health Sciences (RCHS), The University of Lahore, Lahore, Pakistan
| | - Haseeb Nisar
- School of Life Sciences, University of Management and Technology, Lahore, Pakistan
| | - Qudsia Kanwal
- Department of Chemistry, The University of Lahore, Lahore, Pakistan
| | - Jeong Ryeol Choi
- School of Electronic Engineering, Kyonggi University, Suwon, Kyeonggi-do, 16227, Republic of Korea
| |
Collapse
|
32
|
López Angulo D, Lourenço RV, Bridi A, Chaves MA, da Silveira JC, Sobral PJDA. Enhancing Bovine Embryo Development In Vitro Using Oil-in-Water Nanoemulsions as Specific Carriers for Essential Lipids. BIOTECH 2024; 13:19. [PMID: 38921051 PMCID: PMC11201380 DOI: 10.3390/biotech13020019] [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: 04/19/2024] [Revised: 05/20/2024] [Accepted: 05/30/2024] [Indexed: 06/27/2024] Open
Abstract
Worldwide meat consumption and production have nearly quintupled in the last 60 years. In this context, research and the application of new technologies related to animal reproduction have evolved in an accelerated way. The objective of the present study was to apply nanoemulsions (NEs) as carriers of lipids to feed bovine embryos in culture media and verify their impact on the development of embryos produced in vitro. The NEs were characterized by particle size, polydispersity, size distribution, physical stability, morphology using atomic force microscopy (AFM), surface tension, density, pH, and rheological behavior. The NEs were prepared by the emulsification/evaporation technique. A central composite rotatable design (CCRD) was used to optimize the NE fabrication parameters. The three optimized formulations used in the embryo application showed an emulsion stability index (ESI) between 0.046 and 0.086, which reflects high stability. The mean droplet diameter analyzed by laser diffraction was approximately 70-80 nm, suggesting a possible transit across the embryonic zona pellucida with pores of an average 90 nm in diameter. AFM images clearly confirm the morphology of spherical droplets with a mean droplet diameter of less than 100 nm. The optimized formulations added during the higher embryonic genome activation phase in bovine embryos enhanced early embryonic development.
Collapse
Affiliation(s)
- Daniel López Angulo
- Department of Food Engineering, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil
| | - Rodrigo Vinicius Lourenço
- Department of Food Engineering, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil
| | - Alessandra Bridi
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil; (A.B.)
| | - Matheus Andrade Chaves
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil; (A.B.)
| | - Juliano Coelho da Silveira
- Department of Veterinary Medicine, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil; (A.B.)
| | - Paulo José do Amaral Sobral
- Department of Food Engineering, Faculty of Animal Science and Food Engineering, University of São Paulo, Pirassununga 13635-900, SP, Brazil
- Food Research Center (FoRC), University of São Paulo, São Paulo 05508-000, SP, Brazil
| |
Collapse
|
33
|
Wu T, Wang L, Gao C, Jian C, Liu Y, Fu Z, Shi C. Treg-Derived Extracellular Vesicles: Roles in Diseases and Theranostics. Mol Pharm 2024; 21:2659-2672. [PMID: 38695194 DOI: 10.1021/acs.molpharmaceut.4c00233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
Regulatory T cells (Tregs), a subset of CD4+ T cells, are indispensable in maintaining immune self-tolerance and have been utilized in various diseases. Treg-derived extracellular vesicles (Treg-EVs) have been discovered to play an important role in the mechanism of Treg functions. As cell-derived membranous particles, EVs carry multiple bioactive substances that possess tremendous potential for theranostics. Treg-EVs are involved in numerous physiological and pathological processes, carrying proteins and miRNAs inherited from the parental cells. To comprehensively understand the function of Treg-EVs, here we reviewed the classification of Treg-EVs, the active molecules in Treg-EVs, their various applications in diseases, and the existing challenges for Treg-EVs based theranostics. This Review aims to clarify the feasibility and potential of Treg-EVs in diseases and theranostics, facilitating further research and application of Treg-EVs.
Collapse
Affiliation(s)
- Tingting Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Lulu Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Chen Gao
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Chen Jian
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Yajing Liu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Zhiwen Fu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| | - Chen Shi
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, Wuhan, 430022, China
| |
Collapse
|
34
|
Choi Y, Seok SH, Yoon HY, Ryu JH, Kwon IC. Advancing cancer immunotherapy through siRNA-based gene silencing for immune checkpoint blockade. Adv Drug Deliv Rev 2024; 209:115306. [PMID: 38626859 DOI: 10.1016/j.addr.2024.115306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Revised: 04/01/2024] [Accepted: 04/09/2024] [Indexed: 05/23/2024]
Abstract
Cancer immunotherapy represents a revolutionary strategy, leveraging the patient's immune system to inhibit tumor growth and alleviate the immunosuppressive effects of the tumor microenvironment (TME). The recent emergence of immune checkpoint blockade (ICB) therapies, particularly following the first approval of cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors like ipilimumab, has led to significant growth in cancer immunotherapy. The extensive explorations on diverse immune checkpoint antibodies have broadened the therapeutic scope for various malignancies. However, the clinical response to these antibody-based ICB therapies remains limited, with less than 15% responsiveness and notable adverse effects in some patients. This review introduces the emerging strategies to overcome current limitations of antibody-based ICB therapies, mainly focusing on the development of small interfering ribonucleic acid (siRNA)-based ICB therapies and innovative delivery systems. We firstly highlight the diverse target immune checkpoint genes for siRNA-based ICB therapies, incorporating silencing of multiple genes to boost anti-tumor immune responses. Subsequently, we discuss improvements in siRNA delivery systems, enhanced by various nanocarriers, aimed at overcoming siRNA's clinical challenges such as vulnerability to enzymatic degradation, inadequate pharmacokinetics, and possible unintended target interactions. Additionally, the review presents various combination therapies that integrate chemotherapy, phototherapy, stimulatory checkpoints, ICB antibodies, and cancer vaccines. The important point is that when used in combination with siRNA-based ICB therapy, the synergistic effect of traditional therapies is strengthened, improving host immune surveillance and therapeutic outcomes. Conclusively, we discuss the insights into innovative and effective cancer immunotherapeutic strategies based on RNA interference (RNAi) technology utilizing siRNA and nanocarriers as a novel approach in ICB cancer immunotherapy.
Collapse
Affiliation(s)
- Youngjin Choi
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Su Hyun Seok
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Hong Yeol Yoon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science &Technology, KIST School, University of Science and Technology, Seoul 02792, Republic of Korea
| | - Ju Hee Ryu
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea.
| | - Ick Chan Kwon
- Medicinal Materials Research Center, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| |
Collapse
|
35
|
Li B, Yuan D, Chen H, Wang X, Liang Y, Wong CTT, Xia J. Site-selective antibody-lipid conjugates for surface functionalization of red blood cells and targeted drug delivery. J Control Release 2024; 370:302-309. [PMID: 38663752 DOI: 10.1016/j.jconrel.2024.04.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Revised: 03/23/2024] [Accepted: 04/23/2024] [Indexed: 05/04/2024]
Abstract
Displaying antibodies on carrier surfaces facilitates precise targeting and delivery of drugs to diseased cells. Here, we report the synthesis of antibody-lipid conjugates (ALCs) through site-selective acetylation of Lys 248 in human Immunoglobulin G (IgG) and the development of antibody-functionalized red blood cells (immunoRBC) for targeted drug delivery. ImmunoRBC with the HER2-selective antibody trastuzumab displayed on the surface (called Tras-RBC) was constructed following a three-step procedure. First, a peptide-guided, proximity-induced reaction transferred an azidoacetyl group to the ε-amino group of Lys 248 in the Fc domain. Second, the azide-modified IgG was subsequently conjugated with dibenzocyclooctyne (DBCO)-functionalized lipids via strain-promoted azide-alkyne cycloaddition (SPAAC) to result in ALCs. Third, the lipid portion of ALCs was then inserted into the cell membranes, and IgGs were displayed on red blood cells (RBCs) to construct immunoRBCs. We then loaded Tras-RBC with a photosensitizer (PS), Zinc phthalocyanine (ZnPc), to selectively target HER2-overexpressing cells, release ZnPc into cancer cells following photolysis, and induce photodynamic cytotoxicity in the cancer cells. This work showcases assembling immunoRBCs following site-selective lipid conjugation on therapeutic antibodies and the targeted introduction of PS into cancer cells. This method could apply to the surface functionalization of other membrane-bound vesicles or lipid nanoparticles for antibody-directed drug delivery.
Collapse
Affiliation(s)
- Biquan Li
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Dingdong Yuan
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Hongfei Chen
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Xun Wang
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Yujie Liang
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Clarence T T Wong
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Kowloon, Hong Kong SAR, China.
| | - Jiang Xia
- Department of Chemistry and Center for Cell & Developmental Biology, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
36
|
Siddiquee T, Bhaskaran NA, Nathani K, Sawarkar SP. Empowering lung cancer treatment: Harnessing the potential of natural phytoconstituent-loaded nanoparticles. Phytother Res 2024. [PMID: 38806412 DOI: 10.1002/ptr.8241] [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: 10/16/2023] [Revised: 04/29/2024] [Accepted: 04/29/2024] [Indexed: 05/30/2024]
Abstract
Lung cancer, the second leading cause of cancer-related deaths, accounts for a substantial portion, representing 18.4% of all cancer fatalities. Despite advances in treatment modalities such as chemotherapy, surgery, and immunotherapy, significant challenges persist, including chemoresistance, non-specific targeting, and adverse effects. Consequently, there is an urgent need for innovative therapeutic approaches to overcome these limitations. Natural compounds, particularly phytoconstituents, have emerged as promising candidates due to their potent anticancer properties and relatively low incidence of adverse effects compared to conventional treatments. However, inherent challenges such as poor solubility, rapid metabolism, and enzymatic degradation hinder their clinical utility. To address these obstacles, researchers have increasingly turned to nanotechnology-based drug delivery systems (DDS). Nanocarriers offer several advantages, including enhanced drug stability, prolonged circulation time, and targeted delivery to tumor sites, thereby minimizing off-target effects. By encapsulating phytoconstituents within nanocarriers, researchers aim to optimize their bioavailability and therapeutic efficacy while reducing systemic toxicity. Moreover, the integration of nanotechnology with phytoconstituents allows for a nuanced understanding of the intricate molecular pathways involved in lung cancer pathogenesis. This integrated approach holds promise for modulating key cellular processes implicated in tumor growth and progression. Additionally, by leveraging the synergistic effects of phytoconstituents and nanocarriers, researchers seek to develop tailored therapeutic strategies that maximize efficacy while minimizing adverse effects. In conclusion, the integration of phytoconstituents with nanocarriers represents a promising avenue for advancing lung cancer treatment. This synergistic approach has the potential to revolutionize current therapeutic paradigms by offering targeted, efficient, and minimally toxic interventions. Continued research in this field holds the promise of improving patient outcomes and addressing unmet clinical needs in lung cancer management.
Collapse
Affiliation(s)
- Taufique Siddiquee
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Navya Ajitkumar Bhaskaran
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Khushali Nathani
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| | - Sujata P Sawarkar
- Department of Pharmaceutics, SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai, Mumbai, India
| |
Collapse
|
37
|
Sarkar R, Biswas S, Ghosh R, Samanta P, Pakhira S, Mondal M, Dutta Gupta Y, Bhandary S, Saha P, Bhowmik A, Hajra S. Exosome-sheathed porous silica nanoparticle-mediated co-delivery of 3,3'-diindolylmethane and doxorubicin attenuates cancer stem cell-driven EMT in triple negative breast cancer. J Nanobiotechnology 2024; 22:285. [PMID: 38796426 PMCID: PMC11127288 DOI: 10.1186/s12951-024-02518-0] [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: 01/29/2024] [Accepted: 05/01/2024] [Indexed: 05/28/2024] Open
Abstract
BACKGROUND Therapeutic management of locally advanced and metastatic triple negative breast cancer (TNBC) is often limited due to resistance to conventional chemotherapy. Metastasis is responsible for more than 90% of breast cancer-associated mortality; therefore, the clinical need to prevent or target metastasis is immense. The epithelial to mesenchymal transition (EMT) of cancer stem cells (CSCs) is a crucial determinant in metastasis. Doxorubicin (DOX) is the frequently used chemotherapeutic drug against TNBC that may increase the risk of metastasis in patients. After cancer treatment, CSCs with the EMT characteristic persist, which contributes to advanced malignancy and cancer recurrence. The latest developments in nanotechnology for medicinal applications have raised the possibility of using nanomedicines to target these CSCs. Hence, we present a novel approach of combinatorial treatment of DOX with dietary indole 3,3'-diindolylmethane (DIM) which is an intriguing field of research that may target CSC mediated EMT induction in TNBC. For efficient delivery of both the compounds to the tumor niche, advance method of drug delivery based on exosomes sheathed with mesoporous silica nanoparticles may provide an attractive strategy. RESULTS DOX, according to our findings, was able to induce EMT in CSCs, making the breast cancer cells more aggressive and metastatic. In CSCs produced from spheres of MDAMB-231 and 4T1, overexpression of N-cadherin, Snail, Slug, and Vimentin as well as downregulation of E-cadherin by DOX treatment not only demonstrated EMT induction but also underscored the pressing need for a novel chemotherapeutic combination to counteract this detrimental effect of DOX. To reach this goal, DIM was combined with DOX and delivered to the CSCs concomitantly by loading them in mesoporous silica nanoparticles encapsulated in exosomes (e-DDMSNP). These exosomes improved the specificity, stability and better homing ability of DIM and DOX in the in vitro and in vivo CSC niche. Furthermore, after treating the CSC-enriched TNBC cell population with e-DDMSNP, a notable decrease in DOX mediated EMT induction was observed. CONCLUSION Our research seeks to propose a new notion for treating TNBC by introducing this unique exosomal nano-preparation against CSC induced EMT.
Collapse
Affiliation(s)
- Rupali Sarkar
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India
| | - Souradeep Biswas
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India
| | - Rituparna Ghosh
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India
| | - Priya Samanta
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India
| | - Shampa Pakhira
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India
| | - Mrinmoyee Mondal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India
| | - Yashaswi Dutta Gupta
- Department of Biological Sciences, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, West Bengal, India
| | - Suman Bhandary
- Department of Biological Sciences, School of Life Science and Biotechnology, Adamas University, Kolkata, 700126, West Bengal, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India.
| | - Subhadip Hajra
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, S.P. Mukherjee Road, Kolkata, West Bengal, 700 026, India.
| |
Collapse
|
38
|
Chen Q, Yang Z, Liu H, Man J, Oladejo AO, Ibrahim S, Wang S, Hao B. Novel Drug Delivery Systems: An Important Direction for Drug Innovation Research and Development. Pharmaceutics 2024; 16:674. [PMID: 38794336 PMCID: PMC11124876 DOI: 10.3390/pharmaceutics16050674] [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: 04/07/2024] [Revised: 05/12/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
The escalating demand for enhanced therapeutic efficacy and reduced adverse effects in the pharmaceutical domain has catalyzed a new frontier of innovation and research in the field of pharmacy: novel drug delivery systems. These systems are designed to address the limitations of conventional drug administration, such as abbreviated half-life, inadequate targeting, low solubility, and bioavailability. As the disciplines of pharmacy, materials science, and biomedicine continue to advance and converge, the development of efficient and safe drug delivery systems, including biopharmaceutical formulations, has garnered significant attention both domestically and internationally. This article presents an overview of the latest advancements in drug delivery systems, categorized into four primary areas: carrier-based and coupling-based targeted drug delivery systems, intelligent drug delivery systems, and drug delivery devices, based on their main objectives and methodologies. Additionally, it critically analyzes the technological bottlenecks, current research challenges, and future trends in the application of novel drug delivery systems.
Collapse
Affiliation(s)
- Qian Chen
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Q.C.); (Z.Y.); (H.L.); (J.M.); (A.O.O.); (S.I.)
| | - Zhen Yang
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Q.C.); (Z.Y.); (H.L.); (J.M.); (A.O.O.); (S.I.)
| | - Haoyu Liu
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Q.C.); (Z.Y.); (H.L.); (J.M.); (A.O.O.); (S.I.)
| | - Jingyuan Man
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Q.C.); (Z.Y.); (H.L.); (J.M.); (A.O.O.); (S.I.)
| | - Ayodele Olaolu Oladejo
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Q.C.); (Z.Y.); (H.L.); (J.M.); (A.O.O.); (S.I.)
- Department of Animal Health Technology, Oyo State College of Agriculture and Technology, Igboora 201003, Nigeria
| | - Sally Ibrahim
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Q.C.); (Z.Y.); (H.L.); (J.M.); (A.O.O.); (S.I.)
- Department of Animal Reproduction and AI, Veterinary Research Institute, National Research Centre, Dokki 12622, Egypt
| | - Shengyi Wang
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Q.C.); (Z.Y.); (H.L.); (J.M.); (A.O.O.); (S.I.)
| | - Baocheng Hao
- Key Laboratory of New Animal Drug Project, Gansu Province, Key Laboratory of Veterinary Pharmaceutical Development, Ministry of Agriculture and Rural Affairs, Lanzhou Institute of Husbandry and Pharmaceutical Sciences of Chinese Academy of Agriculture Sciences, Lanzhou 730050, China; (Q.C.); (Z.Y.); (H.L.); (J.M.); (A.O.O.); (S.I.)
| |
Collapse
|
39
|
Bai L, Gao J, Zhang P, Lin S, Zhang C. Immunotherapy of M2 macrophage derived from exosome-based nanoparticles for spinal cord injury. Int Immunopharmacol 2024; 132:111983. [PMID: 38593504 DOI: 10.1016/j.intimp.2024.111983] [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: 08/12/2023] [Revised: 03/27/2024] [Accepted: 03/28/2024] [Indexed: 04/11/2024]
Abstract
Developing biomimetic nanoparticles without off-target side-effects remains a major challenge in spinal cord injury (SCI) immunotherapy. In this paper, we have conducted a drug carrier which is biocompatible macrophages-exocytosed exosome-biomimetic manganese (Mn)-iron prussian blue analogues (MPBs) for SCI immunotherapy. Exosome-sheathed MPBs (E-MPBs) exhibit promoted microglia accumulation, alleviation from H2O2-induced microenvironment and inhibition of apoptosis and inflammation in vitro. In addition, E-MPBs possessed significant tissue repair and neuroprotection in vivo. These properties endowed E-MPBs with great improvement in vivo in function recovery, resulting in anti-neuroinflammation activity and excellent biocompatibility in mice SCI model. As a promising treatment for efficient SCI immunotherapy, these results demonstrate the use of exosome-sheathed biomimetic nanoparticles exocytosed by anti-inflammation cells is feasible.
Collapse
Affiliation(s)
- Lu Bai
- Department of Gastroenterology, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Jinpeng Gao
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Peng Zhang
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China
| | - Sen Lin
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China.
| | - Chuanjie Zhang
- Department of Orthopedics, Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, PR China.
| |
Collapse
|
40
|
Pan H, Yang S, Gao L, Zhou J, Cheng W, Chen G, Shuhang W, Li N, Veranič P, Musiol R, Cai Q, Shubhra QT. At the crossroad of nanotechnology and cancer cell membrane coating: Expanding horizons with engineered nanoplatforms for advanced cancer therapy harnessing homologous tumor targeting. Coord Chem Rev 2024; 506:215712. [DOI: 10.1016/j.ccr.2024.215712] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
41
|
Chen K, Zhou A, Zhou X, He J, Xu Y, Ning X. Cellular Trojan Horse initiates bimetallic Fe-Cu MOF-mediated synergistic cuproptosis and ferroptosis against malignancies. SCIENCE ADVANCES 2024; 10:eadk3201. [PMID: 38598629 PMCID: PMC11006215 DOI: 10.1126/sciadv.adk3201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/16/2023] [Accepted: 03/05/2024] [Indexed: 04/12/2024]
Abstract
Disruptions in metal balance can trigger a synergistic interplay of cuproptosis and ferroptosis, offering promising solutions to enduring challenges in oncology. Here, we have engineered a Cellular Trojan Horse, named MetaCell, which uses live neutrophils to stably internalize thermosensitive liposomal bimetallic Fe-Cu MOFs (Lip@Fe-Cu-MOFs). MetaCell can instigate cuproptosis and ferroptosis, thereby enhancing treatment efficacy. Mirroring the characteristics of neutrophils, MetaCell can evade the immune system and not only infiltrate tumors but also respond to inflammation by releasing therapeutic components, thereby surmounting traditional treatment barriers. Notably, Lip@Fe-Cu-MOFs demonstrate notable photothermal effects, inciting a targeted release of Fe-Cu-MOFs within cancer cells and amplifying the synergistic action of cuproptosis and ferroptosis. MetaCell has demonstrated promising treatment outcomes in tumor-bearing mice, effectively eliminating solid tumors and forestalling recurrence, leading to extended survival. This research provides great insights into the complex interplay between copper and iron homeostasis in malignancies, potentially paving the way for innovative approaches in cancer treatment.
Collapse
Affiliation(s)
- Kerong Chen
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, P. R. China
| | - Anwei Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, School of Physics, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, P. R. China
| | - Xinyuan Zhou
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, P. R. China
| | - Jielei He
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, P. R. China
| | - Yurui Xu
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, P. R. China
| | - Xinghai Ning
- National Laboratory of Solid State Microstructures, Collaborative Innovation Center of Advanced Microstructures, Chemistry and Biomedicine Innovation Center, College of Engineering and Applied Sciences, Jiangsu Key Laboratory of Artificial Functional Materials, Nanjing University, Nanjing 210093, P. R. China
| |
Collapse
|
42
|
Siddique AR, Bhagwat GS. Erythrocytes Nanoparticle Delivery: A Boon for Targeting Tumor. Adv Pharm Bull 2024; 14:132-146. [PMID: 38585450 PMCID: PMC10997934 DOI: 10.34172/apb.2023.080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 01/14/2023] [Accepted: 04/24/2023] [Indexed: 04/09/2024] Open
Abstract
Although nanoparticles (NPs) have many advantages as drug delivery systems, their poor stability in circulation, premature drug release, and nonspecific uptake in non-target organs have prompted biomimetic approaches to camouflage nano vehicles using natural cell membranes. Among them, which are extensively studied in erythrocytes, are the most abundant circulating blood cells. They are specially used for biomimetic coating on artificial NPs due to their excellent properties of good biocompatibility, biodegradability, non-immunogenicity, and long-term blood circulation. Erythrocyte-mimicking nanoparticles (EM-NPs) are prepared by combining nanoparticle cores with naturally derived erythrocyte (red blood cell or RBC) membranes. Compared with conventional nanosystems, EM-NPs hold the preferable characteristics of prolonged blood circulation time and immune evasion. In this review, the biomimetic platform of erythrocyte membrane-coated NPs is described in various aspects, with particular focus placed on the coating mechanism, preparation methods, characterization method, and recent advances in the biomedical applications of EM-NPs concerning cancer and targeted delivery.
Collapse
Affiliation(s)
| | - Geeta Sameer Bhagwat
- Pharmaceutics Department, DY Patil University School of Pharmacy, Sector-7, Nerul, Navi Mumbai 400706, India
| |
Collapse
|
43
|
Arcos Rosero WA, Bueno Barbezan A, Daruich de Souza C, Chuery Martins Rostelato ME. Review of Advances in Coating and Functionalization of Gold Nanoparticles: From Theory to Biomedical Application. Pharmaceutics 2024; 16:255. [PMID: 38399309 PMCID: PMC10892584 DOI: 10.3390/pharmaceutics16020255] [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: 10/27/2023] [Revised: 11/27/2023] [Accepted: 01/02/2024] [Indexed: 02/25/2024] Open
Abstract
Nanoparticles, especially gold nanoparticles (Au NPs) have gained increasing interest in biomedical applications. Used for disease prevention, diagnosis and therapies, its significant advantages in therapeutic efficacy and safety have been the main target of interest. Its application in immune system prevention, stability in physiological environments and cell membranes, low toxicity and optimal bioperformances are critical to the success of engineered nanomaterials. Its unique optical properties are great attractors. Recently, several physical and chemical methods for coating these NPs have been widely used. Biomolecules such as DNA, RNA, peptides, antibodies, proteins, carbohydrates and biopolymers, among others, have been widely used in coatings of Au NPs for various biomedical applications, thus increasing their biocompatibility while maintaining their biological functions. This review mainly presents a general and representative view of the different types of coatings and Au NP functionalization using various biomolecules, strategies and functionalization mechanisms.
Collapse
|
44
|
Ding J, Ding X, Liao W, Lu Z. Red blood cell-derived materials for cancer therapy: Construction, distribution, and applications. Mater Today Bio 2024; 24:100913. [PMID: 38188647 PMCID: PMC10767221 DOI: 10.1016/j.mtbio.2023.100913] [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: 09/30/2023] [Revised: 12/02/2023] [Accepted: 12/11/2023] [Indexed: 01/09/2024] Open
Abstract
Cancer has become an increasingly important public health issue owing to its high morbidity and mortality rates. Although traditional treatment methods are relatively effective, they have limitations such as highly toxic side effects, easy drug resistance, and high individual variability. Meanwhile, emerging therapies remain limited, and their actual anti-tumor effects need to be improved. Nanotechnology has received considerable attention for its development and application. In particular, artificial nanocarriers have emerged as a crucial approach for tumor therapy. However, certain deficiencies persist, including immunogenicity, permeability, targeting, and biocompatibility. The application of erythrocyte-derived materials will help overcome the above problems and enhance therapeutic effects. Erythrocyte-derived materials can be acquired via the application of physical and chemical techniques from natural erythrocyte membranes, or through the integration of these membranes with synthetic inner core materials using cell membrane biomimetic technology. Their natural properties such as biocompatibility and long circulation time make them an ideal choice for drug delivery or nanoparticle biocoating. Thus, red blood cell-derived materials are widely used in the field of biomedicine. However, further studies are required to evaluate their efficacy, in vivo metabolism, preparation, design, and clinical translation. Based on the latest research reports, this review summarizes the biology, synthesis, characteristics, and distribution of red blood cell-derived materials. Furthermore, we provide a reference for further research and clinical transformation by comprehensively discussing the applications and technical challenges faced by red blood cell-derived materials in the treatment of malignant tumors.
Collapse
Affiliation(s)
- Jianghua Ding
- Department of Hematology & Oncology, Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, 332005, China
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, 332005, China
| | - Xinjing Ding
- Oncology of Department, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 332000, China
| | - Weifang Liao
- Jiujiang Clinical Precision Medicine Research Center, Jiujiang, Jiangxi, 332005, China
- Department of Medical Laboratory, Clinical Medical College/Affiliated Hospital of Jiujiang University, Jiujiang, Jiangxi, 332005, China
| | - Zhihui Lu
- Oncology of Department, First Affiliated Hospital of Nanchang University, Nanchang, Jiangxi, 332000, China
| |
Collapse
|
45
|
He X, Chang Z, Chen F, Zhang W, Sun M, Shi T, Liu J, Chen P, Zhang K, Guan S, Zhao Z, Li M, Dong WF, Shao D, Yang C. Engineering a biomimetic system for hepatocyte-specific RNAi treatment of non-alcoholic fatty liver disease. Acta Biomater 2024; 174:281-296. [PMID: 37951519 DOI: 10.1016/j.actbio.2023.10.038] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 10/10/2023] [Accepted: 10/29/2023] [Indexed: 11/14/2023]
Abstract
RNA interference (RNAi) presents great potential against intractable liver diseases. However, the establishment of specific, efficient, and safe delivery systems targeting hepatocytes remains a great challenge. Herein, we described a promising hepatocytes-targeting system through integrating triantennary N-acetylgalactosamine (GalNAc)-engineered cell membrane with biodegradable mesoporous silica nanoparticles, which efficiently and safely delivered siRNA to hepatocytes and silenced the target PCSK9 gene expression for the treatment of non-alcoholic fatty liver disease. Having optimized the GalNAc-engineering strategy, insertion orders, and cell membrane source, we obtained the best-performing GalNAc-formulations allowing strong hepatocyte-specific internalization with reduced Kupffer cell capture, resulting in robust gene silencing and less hepatotoxicity when compared with cationic lipid-based GalNAc-formulations. Consequently, a durable reduction of lipid accumulation and damage was achieved by systemic administering siRNAs targeting PCSK9 in high-fat diet-fed mice, accompanied by displaying desirable safety profiles. Taken together, this GalNAc-engineering biomimetics represented versatile, efficient, and safe carriers for the development of hepatocyte-specific gene therapeutics, and prevention of metabolic diseases. STATEMENT OF SIGNIFICANCE: Compared to MSN@LP-GN3 (MC3-LNP), MSN@CM-GN3 exhibited strong hepatocyte targeting and Kupffer cell escaping, as well as good biocompatibility for safe and efficient siRNA delivery. Furthermore, siPCSK9 delivered by MSN@CM-GN3 reduced both serum and liver LDL-C, TG, TC levels and lipid droplets in HFD-induced mice, resulting in better performance than MSN/siPCSK9@LP-GN3 in terms of lipid-lowering effect and safety profiles. These findings indicated promising advantages of our biomimetic GN3-based systems for hepatocyte-specific gene delivery in chronic liver diseases. Our work addressed the challenges associated with the lower targeting efficiency of cell membrane-mimetic drug delivery systems and the immunogenicity of traditional GalNAc delivery systems. In conclusion, this study provided an effective and versatile approach for efficient and safe gene editing using ligand-integrated biomimetic nanoplatforms.
Collapse
Affiliation(s)
- Xuan He
- 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, Guangdong 510006, China
| | - Zhimin Chang
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou 215163, China
| | - Fangman Chen
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou 215163, China.
| | - Wensheng Zhang
- Department of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Madi Sun
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Tongfei Shi
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Jie Liu
- Department of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China
| | - Peiyu Chen
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Kunbao Zhang
- School of Biomedical Sciences and Engineering, South China University of Technology, Guangzhou International Campus, Guangzhou 511442, China
| | - Shan Guan
- National Engineering Research Center of Immunological Products, The Third Military Medical University, Chongqing 400038, China
| | - Zhibin Zhao
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
| | - Mingqiang Li
- Laboratory of Biomaterials and Translational Medicine, Center for Nanomedicine, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou 510630, China
| | - Wen-Fei Dong
- CAS Key Laboratory of Bio Medical Diagnostics, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Suzhou 215163, China
| | - Dan Shao
- National Engineering Research Center for Tissue Restoration and Reconstruction, South China University of Technology, Guangzhou, Guangdong 510006, China; School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China.
| | - Chao Yang
- Department of Orthopedics, Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, The Third Affiliated Hospital of Southern Medical University, Guangzhou 510630, China.
| |
Collapse
|
46
|
Zhang Y, Wang Y, Zhu A, Yu N, Xia J, Li J. Dual-Targeting Biomimetic Semiconducting Polymer Nanocomposites for Amplified Theranostics of Bone Metastasis. Angew Chem Int Ed Engl 2024; 63:e202310252. [PMID: 38010197 DOI: 10.1002/anie.202310252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/03/2023] [Accepted: 11/24/2023] [Indexed: 11/29/2023]
Abstract
Bone metastasis is a type of metastatic tumors that involves the spreads of malignant tumor cells into skeleton, and its diagnosis and treatment remain a big challenge due to the unique tumor microenvironment. We herein develop osteoclast and tumor cell dual-targeting biomimetic semiconducting polymer nanocomposites (SPFeNOC ) for amplified theranostics of bone metastasis. SPFeNOC contain semiconducting polymer and iron oxide (Fe3 O4 ) nanoparticles inside core and surface camouflaged hybrid membrane of cancer cells and osteoclasts. The hybrid membrane camouflage enables their targeting to both metastatic tumor cells and osteoclasts in bone metastasis through homologous targeting mechanism, thus achieving an enhanced nanoparticle accumulation in tumors. The semiconducting polymer mediates near-infrared (NIR) fluorescence imaging and sonodynamic therapy (SDT), and Fe3 O4 nanoparticles are used for magnetic resonance (MR) imaging and chemodynamic therapy (CDT). Because both cancer cells and osteoclasts are killed synchronously via the combinational action of SDT and CDT, the vicious cycle in bone metastasis is broken to realize high antitumor efficacy. Therefore, 4T1 breast cancer-based bone metastasis can be effectively detected and cured by using SPFeNOC as dual-targeting theranostic nanoagents. This study provides an unusual biomimetic nanoplatform that simultaneously targets osteoclasts and cancer cells for amplified theranostics of bone metastasis.
Collapse
Affiliation(s)
- Yijing Zhang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yue Wang
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai, 201600, China
| | - Anni Zhu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Ningyue Yu
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jindong Xia
- Department of Radiology, Shanghai Songjiang District Central Hospital, Shanghai, 201600, China
| | - Jingchao Li
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| |
Collapse
|
47
|
Liu XY, Li RF, Jia J, Yu ZL. Antibacterial micro/nanomotors: current research progress, challenges, and opportunities. Theranostics 2024; 14:1029-1048. [PMID: 38250044 PMCID: PMC10797294 DOI: 10.7150/thno.92449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 12/23/2023] [Indexed: 01/23/2024] Open
Abstract
Bacterial infections remain a formidable threat to human health, a situation exacerbated by the escalating problem of antibiotic resistance. While alternative antibacterial strategies such as oxidants, heat treatments, and metal nanoparticles (NPs) have shown potential, they come with significant drawbacks, ranging from non-specificity to potential environmental concerns. In the face of these challenges, the rapid evolution of micro/nanomotors (MNMs) stands out as a revolutionary development in the antimicrobial arena. MNMs harness various forms of energy and convert it into a substantial driving force, offering bright prospects for combating microbial threats. MNMs' mobility allows for swift and targeted interaction with bacteria, which not only improves the carrying potential of therapeutic agents but also narrows the required activation range for non-drug antimicrobial interventions like photothermal and photodynamic therapies, substantially improving their bacterial clearance rates. In this review, we summarized the diverse propulsion mechanisms of MNMs employed in antimicrobial applications and articulated their multiple functions, which include direct bactericidal action, capture and removal of microorganisms, detoxification processes, and the innovative detection of bacteria and associated toxins. Despite MNMs' potential to revolutionize antibacterial research, the translation from laboratory to clinical use remains challenging. Based on the current research status, we summarized the potential challenges and possible solutions and also prospected several key directions for future studies of MNMs for antimicrobial purposes. Collectively, by highlighting the important knowns and unknowns of antimicrobial MNMs, our present review would help to light the way forward for the field of antimicrobial MNMs and prevent unnecessary blindness and detours.
Collapse
Affiliation(s)
- Xin-Yang Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
| | - Rui-Fang Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, 430079 Wuhan, China
| | - Jun Jia
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, 430079 Wuhan, China
| | - Zi-Li Yu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University
- Department of Oral and Maxillofacial Surgery, School and Hospital of Stomatology, Wuhan University, 430079 Wuhan, China
| |
Collapse
|
48
|
Xia D, Li J, Feng L, Gao Z, Liu J, Wang X, Hu Y. Advances in Targeting Drug Biological Carriers for Enhancing Tumor Therapy Efficacy. Macromol Biosci 2023; 23:e2300178. [PMID: 37466216 DOI: 10.1002/mabi.202300178] [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: 04/24/2023] [Revised: 06/27/2023] [Accepted: 07/17/2023] [Indexed: 07/20/2023]
Abstract
Chemotherapy drugs continue to be the main component of oncology treatment research and have been proven to be the main treatment modality in tumor therapy. However, the poor delivery efficiency of cancer therapeutic drugs and their potential off-target toxicity significantly limit their effectiveness and extensive application. The recent integration of biological carriers and functional agents is expected to camouflage synthetic biomimetic nanoparticles for targeted delivery. The promising candidates, including but not limited to red blood cells and their membranes, platelets, tumor cell membrane, bacteria, immune cell membrane, and hybrid membrane are typical representatives of biological carriers because of their excellent biocompatibility and biodegradability. Biological carriers are widely used to deliver chemotherapy drugs to improve the effectiveness of drug delivery and therapeutic efficacy in vivo, and tremendous progress is made in this field. This review summarizes recent developments in biological vectors as targeted drug delivery systems based on microenvironmental stimuli-responsive release, thus highlighting the potential applications of target drug biological carriers. The review also discusses the possibility of clinical translation, as well as the exploitation trend of these target drug biological carriers.
Collapse
Affiliation(s)
- Donglin Xia
- School of Public Health, Nantong University, Nantong, Jiangsu, 226019, P.R. China
| | - Jia Li
- School of Public Health, Nantong University, Nantong, Jiangsu, 226019, P.R. China
| | - Lingzi Feng
- School of Public Health, Nantong University, Nantong, Jiangsu, 226019, P.R. China
| | - Ziqing Gao
- School of Public Health, Nantong University, Nantong, Jiangsu, 226019, P.R. China
| | - Jun Liu
- Department of Laboratory Medicine, Wuxi No. 5 People's Hospital Affiliated Jiangnan University, Wuxi, Jiangsu, 214005, P.R. China
| | - Xiangqian Wang
- Department of Radiotherapy, Nantong Tumor Hospital, Tumor Hospital Affiliated to Nantong University, Nantong, Jiangsu, 226361, P.R. China
| | - Yong Hu
- College of Engineering and Applied Sciences, Nanjing University, Nanjing, Jiangsu, 210023, P.R. China
| |
Collapse
|
49
|
Li C, Gao D, Gao Y, Zhang R, Qu X, Li S, Xing C. NIR-II Regulation of Mitochondrial Potassium Channel with Dual-Targeted Conjugated Oligomer Nanoparticles for Efficient Cancer Theranostics In Vivo. Adv Healthc Mater 2023; 12:e2301954. [PMID: 37722719 DOI: 10.1002/adhm.202301954] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 09/13/2023] [Indexed: 09/20/2023]
Abstract
Cell fate can be efficiently modulated by switching ion channels. However, the precise regulation of ion channels in cells, especially in specific organelles, remains challenging. Herein, biomimetic second near-infrared (NIR-II) responsive conjugated oligomer nanoparticles with dual-targeted properties are designed and prepared to modulate the ion channels of mitochondria to selectively kill malignant cells in vivo. Upon 1060 nm laser irradiation, the mitochondria-located nanoparticles photothermally release a specific ion inhibitor of the potassium channel via a temperature-sensitive liposome, thus altering the redox balance and pathways of mitochondria. NIR-II responsive nanoparticles can effectively regulate the potassium channels of mitochondria and fully suppress tumor growth. This work provides a new modality based on the NIR-II nanoplatform to regulate ion channels in specific organelles and proposes an effective therapeutic mechanism for malignant tumors.
Collapse
Affiliation(s)
- Chaoqun Li
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
| | - Dong Gao
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
| | - Yijian Gao
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, P. R. China
| | - Ran Zhang
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
| | - Xiongwei Qu
- School of Chemical Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
| | - Shengliang Li
- College of Pharmaceutical Sciences, Soochow University, Suzhou, 215000, P. R. China
| | - Chengfen Xing
- School of Materials Science and Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
- School of Health Sciences and Biomedical Engineering, Hebei University of Technology, Tianjin, 300130, P. R. China
| |
Collapse
|
50
|
Wu YW, Lee DY, Lu YL, Delila L, Nebie O, Barro L, Changou CA, Lu LS, Goubran H, Burnouf T. Platelet extracellular vesicles are efficient delivery vehicles of doxorubicin, an anti-cancer drug: preparation and in vitro characterization. Platelets 2023; 34:2237134. [PMID: 37580876 DOI: 10.1080/09537104.2023.2237134] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Revised: 07/10/2023] [Accepted: 07/10/2023] [Indexed: 08/16/2023]
Abstract
Platelet extracellular vesicles (PEVs) are an emerging delivery vehi for anticancer drugs due to their ability to target and remain in the tumor microenvironment. However, there is still a lack of understanding regarding yields, safety, drug loading efficiencies, and efficacy of PEVs. In this study, various methods were compared to generate PEVs from clinical-grade platelets, and their properties were examined as vehicles for doxorubicin (DOX). Sonication and extrusion produced the most PEVs, with means of 496 and 493 PEVs per platelet (PLT), respectively, compared to 145 and 33 by freeze/thaw and incubation, respectively. The PEVs were loaded with DOX through incubation and purified by chromatography. The size and concentration of the PEVs and PEV-DOX were analyzed using dynamic light scattering and nanoparticle tracking analysis. The results showed that the population sizes and concentrations of PEVs and PEV-DOX were in the ranges of 120-150 nm and 1.2-6.2 × 1011 particles/mL for all preparations. The loading of DOX determined using fluorospectrometry was found to be 2.1 × 106, 1.7 × 106, and 0.9 × 106 molecules/EV using freeze/thaw, extrusion, and sonication, respectively. The internalization of PEVs was determined to occur through clathrin-mediated endocytosis. PEV-DOX were more efficiently taken up by MDA-MB-231 breast cancer cells compared to MCF7/ADR breast cancer cells and NIH/3T3 cells. DOX-PEVs showed higher anticancer activity against MDA-MB-231 cells than against MCF7/ADR or NIH/3T3 cells and better than acommercial liposomal DOX formulation. In conclusion, this study demonstrates that PEVs generated by PLTs using extrusion, freeze/thaw, or sonication can efficiently load DOX and kill breast cancer cells, providing a promising strategy for further evaluation in preclinical animal models. The study findings suggest that sonication and extrusion are the most efficient methods to generate PEVs and that PEVs loaded with DOX exhibit significant anticancer activity against MDA-MB-231 breast cancer cells.
Collapse
Affiliation(s)
- Yu-Wen Wu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Deng-Yao Lee
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Yeh-Lin Lu
- Core Facility Center, Office of Research and Development, Taipei Medical University, Taipei, Taiwan
| | - Liling Delila
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Ouada Nebie
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Lassina Barro
- International PhD Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
| | - Chun Austin Changou
- Core Facility Center, Office of Research and Development, Taipei Medical University, Taipei, Taiwan
- Graduate Institute of Translational Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
- The Ph.D. Program for Cancer Biology and Drug Discovery, Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
| | - Long-Sheng Lu
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- The Ph.D. Program for Cancer Biology and Drug Discovery, Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
- Department of Radiation Oncology, Taipei Medical University Hospital, Taipei, Taiwan
- Translational Laboratory, Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Hadi Goubran
- Saskatoon Cancer Centre and College of Medicine, University of Saskatchewan, Saskatoon, Saskatchewan, Canada
| | - Thierry Burnouf
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei, Taiwan
- The Ph.D. Program for Cancer Biology and Drug Discovery, Center for Translational Medicine, Taipei Medical University, Taipei, Taiwan
- International PhD Program in Cell Therapy and Regeneration Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|