634 results on '"Pang, Zhiqing"'
Search Results
202. Encapsulation of cationic peptides into polymersomes throughin situgelatinization
- Author
-
Gao, Huile, primary, Pang, Zhiqing, additional, Lu, Wei, additional, Pan, Shuaiqi, additional, and Jiang, Xinguo, additional
- Published
- 2011
- Full Text
- View/download PDF
203. A novel combination of TRAIL and doxorubicin enhances antitumor effect based on passive tumor-targeting of liposomes
- Author
-
Guo, Liangran, primary, Fan, Li, additional, Ren, Jinfeng, additional, Pang, Zhiqing, additional, Ren, Yulong, additional, Li, Jingwei, additional, Wen, Ziyi, additional, and Jiang, Xinguo, additional
- Published
- 2011
- Full Text
- View/download PDF
204. Enhanced Intracellular Delivery and Chemotherapy for Glioma Rats by Transferrin-Conjugated Biodegradable Polymersomes Loaded with Doxorubicin
- Author
-
Pang, Zhiqing, primary, Gao, Huile, additional, Yu, Yuan, additional, Guo, Liangran, additional, Chen, Jun, additional, Pan, Shuaiqi, additional, Ren, Jinfeng, additional, Wen, Ziyi, additional, and Jiang, Xinguo, additional
- Published
- 2011
- Full Text
- View/download PDF
205. Facile synthesis of superparamagnetic Fe3O4@Au nanoparticles for photothermal destruction of cancer cells
- Author
-
Ren, Jinfeng, primary, Shen, Shun, additional, Pang, Zhiqing, additional, Lu, Xiaohui, additional, Deng, Chunhui, additional, and Jiang, Xinguo, additional
- Published
- 2011
- Full Text
- View/download PDF
206. Lactoferrin-Conjugated Biodegradable Polymersome Holding Doxorubicin and Tetrandrine for Chemotherapy of Glioma Rats
- Author
-
Pang, Zhiqing, primary, Feng, Liang, additional, Hua, Rongrong, additional, Chen, Jun, additional, Gao, Huile, additional, Pan, Shuaiqi, additional, Jiang, Xinguo, additional, and Zhang, Peng, additional
- Published
- 2010
- Full Text
- View/download PDF
207. Full encapsulation of oncolytic virus using hybrid erythroctye-liposome membranes for augmented anti-refractory tumor effectiveness.
- Author
-
Huang, Hanwei, Sun, Mengchi, Liu, Mingyang, Pan, Siwei, Liu, Pengfei, Cheng, Zhenguo, Li, Jia, Xu, Huimian, Liu, Funan, and Pang, Zhiqing
- Subjects
LIPOSOMES ,CYTOKINE release syndrome ,CELL surface antigens ,CELL membranes ,ARTIFICIAL cells ,ARTIFICIAL membranes ,NANOMEDICINE - Abstract
Intravenous delivery of oncolytic virus (OVs) is promising in cancer treatment. Unfortunately, fast clearance of OVs and the severe cytokine release syndrome impede its wide application. It has been shown that nanoparticles coated with cell membranes display less toxicity and slower clearance. However, different from conventional nanoparticles, the characteristic spike-like structure and abundant antigens on the surface make it difficult for intravenously delivered OVs to take advantage of cell membrane coating to shield their surface antigens. To overcome this challenge, we, for the first time, used erythrocyte-lipid hybrid membrane vesicle (erythroliposome) to fully encapsulate OVs for their intravenous delivery. We found that adding artificial membranes to cell membranes reduced the fluidity of the membranes, leading to an extraordinary shielding effect on OV antigens. Consequently, circulation of OVs was significantly prolonged and their oncolytic efficacy to metastatic and refractory tumors was markedly enhanced. [Display omitted] • We firstly found that only cell membranes or liposomes showed no effect on shielding the antigen of oncolytic virus (OVs). • We firstly used erythroliposomes formed by fusing liposomes with RBC membranes to fully coat OVs. • We firstly treat metastatic tumors successfully by intravenous delivery of OVs. • We firstly solved the problem that neutralizing antibodies produced during multiple OV injections reduced the anti-tumor efficacy of OVs. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
208. Advances of nanoparticles as drug delivery systems for disease diagnosis and treatment
- Author
-
Liu, Rui, Luo, Cong, Pang, Zhiqing, Zhang, Jinming, Ruan, Shaobo, Wu, Meiying, Wang, Lei, Sun, Tao, Li, Nan, Han, Liang, Shi, Jinjin, Huang, Yuanyu, Guo, Weisheng, Peng, Shaojun, Zhou, Wenhu, and Gao, Huile
- Abstract
Decades have passed since the first nanoparticles-base medicine was approved for human cancer treatment, and the research and development of nanoparticles for drug delivery are always undergoing. Nowadays, the significant advances complicate nanoparticles’ branches, including liposomes, solid lipid nanoparticles, inorganic nanoparticles, micelles, nanovaccines and nano-antibodies, etc.These nanoparticles show numerous capabilities in treatment and diagnosis of stubborn diseases like cancer and neurodegenerative diseases, emerging as novel drug carriers or therapeutic agents in future. In this review, the complicated branches of nanoparticles are classified and summarized, with their property and functions concluded. Besides, there are also some delivery strategies that make nanoparticles smarter and more efficient in drug delivery, and frontiers in these strategies are also summarized in this review. Except these excellent works in newly-produced drug delivery nanoparticles, some points of view and future expectations are made in the end.
- Published
- 2023
- Full Text
- View/download PDF
209. Virus-Protein Corona Replacement Strategy to Improve the Antitumor Efficacy of Intravenously Injected Oncolytic Adenovirus
- Author
-
Huang, Hanwei, Liu, Mingyang, Sun, Mengchi, Duan, Shijie, Pan, Siwei, Liu, Pengfei, Cheng, Zhenguo, Ergonul, Onder, Can, Füsun, Wang, Zhenning, Pang, Zhiqing, and Liu, Funan
- Abstract
Intravenous administration of oncolytic adenoviruses (OVs) is a hopeful tumor therapeutic modality. However, the sharp clearance of OVs by the immune system dampens its effectiveness. Many studies have attempted to extend the circulation of intravenously administered OVs, almost all by preventing OVs from binding to neutralizing antibodies and complements in the blood, but the results have not been satisfactory. In contrast to previous conclusions, we found that the key to improving the circulation of OVs is to prevent the formation of the virus-protein corona rather than simply preventing the binding of neutralizing antibodies or complements to OVs. After identifying the key protein components of the virus-protein corona, we proposed a virus-protein corona replacement strategy, where an artificial virus-protein corona was formed on OVs to completely prevent the interaction of OVs with key virus-protein corona components in the plasma. It was found that this strategy dramatically prolonged the circulation time of OVs by over 30 fold and increased the distribution of OVs in tumors by over 10-fold, resulting in superior antitumor efficacy in primary and metastatic tumor models. Our finding provides a perspective on intravenous delivery of OVs, shifting the focus of future studies from preventing OV binding with neutralization antibodies and complements to preventing OVs from interacting with key virus-protein corona components in the plasma.
- Published
- 2023
- Full Text
- View/download PDF
210. Quantification of Dopamine in Brain Microdialysates with High-Performance Liquid Chromatography-Tandem Mass Spectrometry
- Author
-
Qi, Dawei, Zhang, Qian, Zhou, Wanhong, Zhao, Jingjing, Zhang, Bo, Sha, Yunfei, and Pang, Zhiqing
- Abstract
The dopamine level in the brain and the mesolimbick dopaminergic system are responsible for nicotine addiction. In the present study, extracellular dopamine in nucleus accumbens was collected by a brain microdialysis technique. Also a sensitive high-performance liquid chromatography–tandem mass spectrometry (HPLC-MS/MS) method with a gradient elution procedure was developed for a precise determination of dopamine in brain dialysate. The retention time of dopamine was about 11.32 min. The linear range was 20–1000 pg/mL. The limit of detection (LOD) and the limit of qualification (LOQ) were 5 and 20 pg/mL, respectively. The recovery ranged from 98.2 to 109.0%, and both the intraRSD and inter-RSD were below <9%, respectively. The probe recovery for dopamine in this brain microdialysis experiment was about 25%. Finally, the dopamine concentrations in the rat brain microdialysates were determined, and the pharmacokinetics of extracellular dopamine in the brain nucleus accumbens after an intravenous injection of nicotine was successfully evaluated.
- Published
- 2016
- Full Text
- View/download PDF
211. Study and evaluation of mechanisms of dual targeting drug delivery system with tumor microenvironment assays compared with normal assays.
- Author
-
Gao, Huile, Yang, Zhi, Zhang, Shuang, Pang, Zhiqing, Liu, Qingfeng, and Jiang, Xinguo
- Subjects
DRUG delivery systems ,TARGETED drug delivery ,BIOLOGICAL assay ,GLIOMAS ,NANOMEDICINE ,LABORATORY mice - Abstract
Abstract: A dual targeting delivery system was developed to completely conquer the two barriers that glioma treatment faces: the blood–brain barrier (BBB) and the brain–glioma barrier. Recently, a system comprising AS1411 aptamer (for glioma targeting) and TGN peptide (for BBB targeting) modified nanoparticles (AsTNPs) was developed, which can effectively target brain glioma and improve the survival of glioma-bearing mice. However, the in vitro models currently used are far too different from the in vivo tumor microenvironment that the glioma targeting delivery system actually faces. In this study, the pharmacology mechanisms of AsTNPs were explored in several models that imitated the tumor microenvironment. AsTNPs can be selectively taken up by endothelial and glioma cells, effectively penetrating the BBB and brain–glioma barriers to reach glioma cells and display their anti-glioma effect. The cell monolayers, tumor spheroids and coculture systems were more suitable in vitro models for the pharmacology evaluation of targeted drug delivery systems. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
212. Encapsulation of cationic peptides into polymersomes through in situ gelatinization.
- Author
-
Gao, Huile, Pang, Zhiqing, Lu, Wei, Pan, Shuaiqi, and Jiang, Xinguo
- Subjects
- *
MICROENCAPSULATION , *GELATION , *POLYETHYLENE glycol , *GEL permeation chromatography , *TETRAHYDROFURAN - Abstract
Encapsulation of peptides and proteins remains an obstacle in drug nanoformulations. Here, we established an alternative method to encapsulate peptides and proteins into polymersomes (POs). NC-1900, a type of cationic peptide that can induce the gelatinization of deacetylated gellan gum (DGG), was selected as a model peptide. DGG was first trapped in POs to serve as a reservoir to capture NC-1900. Analysis of the optimized formulation revealed that the drug-loading capability of NC-1900-loaded POs was 1.20%%, and the encapsulation efficiency was 30%%. The release of NC-1900 from the gel was the rate-limiting step and could be expressed by Fick's law of diffusion. These results indicated that the preparation of POs encapsulated with gelatin could be employed as an effective loading method for ionic peptides and proteins. [ABSTRACT FROM AUTHOR]
- Published
- 2011
- Full Text
- View/download PDF
213. List of contributors
- Author
-
Barui, Ayan K., Bollu, Vishnu S., Buitrago, Diana M., Cheng, Weiren, de La Torre, Lucimara G., Eş, Ismail, García, Yenny M., Ibla, José F., Jain, Keerti, Jana, Sougata, Jana, Subrata, Jiang, Ting, Jin, Kai, Kotcherlakota, Rajesh, Lafaurie, Gloria I., Liu, Xianpping, Liu, Ye, Maheshwari, Rahul, Maiti, Sabyasachi, Mody, Nishi, Morantes, Sandra J., F. Naves, Thays, Nethi, Susheel K., Oliveira, Aline F., Pang, Zhiqing, Parraga, Jenny E., Patra, Chitta R., Pessoa, Amanda C.S.N., Prabaharan, M., Sharma, Rajeev, Sipoli, Caroline C., Sivashankari, P.R., Tekade, Muktika, Tekade, Rakesh K., Vit, Franciele F., Vitor, Micaela T., and Vyas, Suresh P.
- Published
- 2017
- Full Text
- View/download PDF
214. Intravenous Delivery of Living Listeria monocytogenesElicits Gasdmermin-Dependent Tumor Pyroptosis and Motivates Anti-Tumor Immune Response
- Author
-
Liu, Yao, Lu, Yiping, Ning, Bo, Su, Xiaomin, Yang, Binru, Dong, Haiqing, Yin, Bo, Pang, Zhiqing, and Shen, Shun
- Abstract
The facultative intracellular bacterium Listeria monocytogenes(Lmo) has great potential for development as a cancer vaccine platform given its properties. However, the clinical application of Lmohas been severely restricted due to its rapid clearance, compromised immune response in tumors, and inevitable side effects such as severe systemic inflammation after intravenous administration. Herein, an immunotherapy system was developed on the basis of natural red blood cell (RBC) membranes encapsulated Lmowith selective deletion of virulence factors (Lmo@RBC). The biomimetic Lmo@RBC not only generated a low systemic inflammatory response but also enhanced the accumulation in tumors due to the long blood circulation and tumor hypoxic microenvironment favoring anaerobic Lmocolonization. After genome screening of tumors treated with intravenous PBS, Lmo, or Lmo@RBC, it was first found that Lmo@RBC induced extensive pore-forming protein gasdermin C (GSDMC)-dependent pyroptosis, which reversed immunosuppressive tumor microenvironment and promoted a systemic strong and durable anti-tumor immune response, resulting in an excellent therapeutic effect on solid tumors and tumor metastasis. Overall, Lmo@RBC, as an intravenous living bacterial therapy for the selective initiation of tumor pyrolysis, provided a proof-of-concept of live bacteria vaccine potentiating tumor immune therapy.
- Published
- 2022
- Full Text
- View/download PDF
215. Nanoplateletsomes for rapid hemostasis performance
- Author
-
Wang, Honglan, Zhu, Yuefei, Zhang, Longlong, Liu, Huiwen, Liu, Chunying, Zhang, Bo, Song, Yanan, Hu, Yu, and Pang, Zhiqing
- Abstract
Uncontrollable hemorrhage remains staple trouble in surgical procedures and a leading cause after major trauma. The bleeding issue may trigger various pathologic scenarios that can lead to tissue morbidities and mortalities, and currently available on-site hemostatic agents are confined to a narrow therapeutic index and may carry the risk of immunogenicity. Inspired by the crucial role of platelets in the process of thrombus, a platelet-mimetic plateletsome with wound targeting and blood coagulation properties is developed for hemorrhage control. Plateletsome is formulated by integrating platelet membranes with functionalized synthetic liposomes and exhibits superior wound targeting and effective hemostasis properties. It presents less blood loss and shorter hemostasis time than the platelet membrane vehicles or the conventional liposomes in the mouse tail transection model. The strong homing of the biomimetic plateletsome to the thrombus was also confirmed, demonstrating the potential of this engineered cell membrane vesicle as a biomimetic hemostat for bleeding treatment.
- Published
- 2022
- Full Text
- View/download PDF
216. Biomimetic Targeted Theranostic Nanoparticles for Breast Cancer Treatment.
- Author
-
Marshall, Suphalak Khamruang, Angsantikul, Pavimol, Pang, Zhiqing, Nasongkla, Norased, Hussen, Rusnah Syahila Duali, and Thamphiwatana, Soracha D.
- Subjects
- *
CELL adhesion molecules , *BREAST cancer , *CANCER treatment , *ERYTHROCYTES , *THERAPEUTICS , *BIOMIMETIC materials , *DRUG delivery systems , *CANCER cells - Abstract
The development of biomimetic drug delivery systems for biomedical applications has attracted significant research attention. As the use of cell membrane as a surface coating has shown to be a promising platform for several disease treatments. Cell-membrane-coated nanoparticles exhibit enhanced immunocompatibility and prolonged circulation time. Herein, human red blood cell (RBC) membrane-cloaked nanoparticles with enhanced targeting functionality were designed as a targeted nanotheranostic against cancer. Naturally, derived human RBC membrane modified with targeting ligands coated onto polymeric nanoparticle cores containing both chemotherapy and imaging agent. Using epithelial cell adhesion molecule (EpCAM)-positive MCF-7 breast cancer cells as a disease model, the nature-inspired targeted theranostic human red blood cell membrane-coated polymeric nanoparticles (TT-RBC-NPs) platform was capable of not only specifically binding to targeted cancer cells, effectively delivering doxorubicin (DOX), but also visualizing the targeted cancer cells. The TT-RBC-NPs achieved an extended-release profile, with the majority of the drug release occurring within 5 days. The TT-RBC-NPs enabled enhanced cytotoxic efficacy against EpCAM positive MCF-7 breast cancer over the non-targeted NPs. Additionally, fluorescence images of the targeted cancer cells incubated with the TT-RBC-NPs visually indicated the increased cellular uptake of TT-RBC-NPs inside the breast cancer cells. Taken together, this TT-RBC-NP platform sets the foundation for the next-generation stealth theranostic platforms for systemic cargo delivery for treatment and diagnostic of cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
217. High Intensity Focused Ultrasound‐Driven Nanomotor for Effective Ferroptosis‐Immunotherapy of TNBC.
- Author
-
Yu, Xiangrong, Li, Xuejing, Chen, Qingwang, Wang, Siyu, Xu, Ruizhe, He, Ying, Qin, Xifeng, Zhang, Jun, Yang, Wuli, Shi, Leming, Lu, Ligong, Zheng, Yuanting, Pang, Zhiqing, and Peng, Shaojun
- Subjects
- *
HIGH-intensity focused ultrasound , *TRIPLE-negative breast cancer , *NANOMOTORS - Abstract
The heterogeneity of triple‐negative breast cancers (TNBC) remains challenging for various treatments. Ferroptosis, a recently identified form of cell death resulting from the unrestrained peroxidation of phospholipids, represents a potential vulnerability in TNBC. In this study, a high intensity focused ultrasound (HIFU)‐driven nanomotor is developed for effective therapy of TNBC through induction of ferroptosis. Through bioinformatics analysis of typical ferroptosis‐associated genes in the FUSCCTNBC dataset, gambogic acid is identified as a promising ferroptosis drug and loaded it into the nanomotor. It is found that the rapid motion of nanomotors propelled by HIFU significantly enhanced tumor accumulation and penetration. More importantly, HIFU not only actuated nanomotors to trigger effective ferroptosis of TNBC cells, but also drove nanomotors to activate ferroptosis‐mediated antitumor immunity in primary and metastatic TNBC models, resulting in effective tumor regression and prevention of metastases. Overall, HIFU‐driven nanomotors show great potential for ferroptosis‐immunotherapy of TNBC. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
218. Recent insights for the emerging COVID-19: drug discovery, therapeutic options and vaccine development
- Author
-
Zhu, Yuefei, primary, Li, Jia, additional, and pang, zhiqing, additional
- Full Text
- View/download PDF
219. Erythrocyte‐Leveraged Oncolytic Virotherapy (ELeOVt): Oncolytic Virus Assembly on Erythrocyte Surface to Combat Pulmonary Metastasis and Alleviate Side Effects.
- Author
-
Liu, Mingyang, Zhang, Ruizhe, Huang, Hanwei, Liu, Pengfei, Zhao, Xu, Wu, Hu, He, Ying, Xu, Ruizhe, Qin, Xifeng, Cheng, Zhenguo, Liu, Hongyu, Ergonul, Onder, Can, Füsun, Ouyang, Defang, Wang, Zhenning, Pang, Zhiqing, and Liu, Funan
- Subjects
- *
ONCOLYTIC virotherapy , *CYTOKINE release syndrome , *ERYTHROCYTES , *INTRAVENOUS injections , *METASTASIS , *CANCER treatment - Abstract
Despite being a new promising tool for cancer therapy, intravenous delivery of oncolytic viruses (OVs) is greatly limited by poor tumor targeting, rapid clearance in the blood, severe organ toxicity, and cytokine release syndrome. Herein, a simple and efficient strategy of erythrocyte‐leveraged oncolytic virotherapy (ELeOVt) is reported, which for the first time assembled OVs on the surface of erythrocytes with up to near 100% efficiency and allowed targeted delivery of OVs to the lung after intravenous injection to achieve excellent treatment of pulmonary metastases while greatly improving the biocompatibility of OVs as a drug. Polyethyleneimine (PEI) as a bridge to assemble OVs on erythrocytes also played an important role in promoting the transfection of OVs. It is found that ELeOVt approach significantly prolonged the circulation time of OVs and increased the OVs distribution in the lung by more than tenfold, thereby significantly improving the treatment of lung metastases while reducing organ and systemic toxicity. Taken together, these findings suggest that the ELeOVt provides a biocompatible, efficient, and widely available approach to empower OVs to combat lung metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
220. Targeted delivery of platelet membrane modified extracellular vesicles into atherosclerotic plaque to regress atherosclerosis.
- Author
-
Li, Qiyu, Huang, Zheyong, Pang, Zhiqing, Wang, Qiaozi, Gao, Jinfeng, Chen, Jing, Wang, Zhengmin, Tan, Haipeng, Li, Su, Xu, Fei, Chen, Jinxiang, Liu, Muyin, Weng, Xueyi, Yang, Hongbo, Song, Yanan, Qian, Juying, and Ge, Junbo
- Subjects
- *
ATHEROSCLEROTIC plaque , *EXTRACELLULAR vesicles , *ENDOCYTOSIS , *MACROPHAGES , *MESENCHYMAL stem cells , *BLOOD platelets , *ATHEROSCLEROSIS - Abstract
• Platelet membrane modified extracellular vesicles (P-EVs) inherit the targeting ability towards injured endothelial cells which makes P-EVs accumulate in the plaques. • P-EVs are able to be endocytosed by inflammatory macrophages in plaques and transfer miRNAs into cytosol for further treatment. Atherosclerosis is a major underlying cause of cardiovascular disease. Although certain success has been achieved in treatment for atherosclerosis, the risk of cardiovascular diseases still remains high. Anti-inflammation therapy was proved to halt atherosclerosis progression, but their clinical application was hindered by the systemic immunosuppressive effect. Since macrophages play a key role in the development of atherosclerosis, targeted delivery of anti-inflammatory therapeutics to lesional macrophages might be an effective therapeutic strategy to resolve localized inflammation while reducing the adverse effects. Mesenchymal stem cells derived extracellular vesicles (MSC-EVs) are believed to possess anti-atherosclerosis effects by reducing inflammation. In this study, we fabricated platelet mimetic MSC-EVs (P-EVs) by decorating EVs with platelets membranes. P-EVs inherited the natural homing ability of platelets to plaques and therapeutic effects of MSC-EVs. When injected into atherosclerotic mice, P-EVs were recruited to injured endothelium, then penetrated into plaques followed with endocytosis by inflammatory macrophages through the interaction between platelet membranes proteins and macrophages surface proteins. Subsequently, P-EVs delivered miRNAs into the cytoplasm of macrophages through lysosomal escape, which attenuated the progression of atherosclerosis eventually by switching the macrophage into anti-inflammatory phenotype, scavenging the overproduced ROS and reducing lipid deposition in lesional macrophages. In general, our study demonstrated the pro-resolving potential of P-EVs for targeted anti-inflammation therapy of atherosclerosis. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
221. Targeted delivery and ROS-responsive release of Resolvin D1 by platelet chimeric liposome ameliorates myocardial ischemia–reperfusion injury.
- Author
-
Weng, Xueyi, Tan, Haipeng, Huang, Zheyong, Chen, Jing, Zhang, Ning, Wang, Qiaozi, Li, Qiyu, Gao, Jinfeng, Sun, Dili, Yakufu, Wusiman, Wang, Zhengmin, Li, Weiyan, Zhu, Guangrui, Pang, Zhiqing, Song, Yanan, Qian, Juying, and Ge, Junbo
- Subjects
- *
LIPOSOMES , *REPERFUSION injury , *MYOCARDIAL reperfusion , *BLOOD platelets , *REACTIVE oxygen species , *INTRAVENOUS injections , *DRUG delivery systems - Abstract
Resolvin D1 (RvD1) has been shown to provide effective protection against ischemia–reperfusion injury in multiple vital organs such as the heart, brain, kidney. However, the clinical translational potential of systemic administration of RvD1 in the treatment of ischemia–reperfusion injury is greatly limited due to biological instability and lack of targeting ability. Combining the natural inflammatory response and reactive oxygen species (ROS) overproduction after reperfusion injury, we developed a platelet-bionic, ROS-responsive RvD1 delivery platform. The resulting formulation enables targeted delivery of RvD1 to the injury site by hijacking circulating chemotactic monocytes, while achieving locally controlled release. In a mouse model of myocardial ischemia repefusuin (MI/R) injury, intravenous injection of our formula resulted in the enrichment of RvD1 in the injured area, which in turn promotes clearance of dead cells, production of specialized proresolving mediators (SPMs), and angiogenesis during injury repair, effectively improving cardiac function. This delivery system integrates drug bio-protection, targeted delivery and controlled release, which endow it with great clinical translational value. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
222. Aptamer-Modified Erythrocyte Membrane-Coated pH-Sensitive Nanoparticles for c-Met-Targeted Therapy of Glioblastoma Multiforme.
- Author
-
Liu, Xianping, Chen, Yixin, Geng, Daoying, Li, Haichun, Jiang, Ting, Luo, Zimiao, Wang, Jianhong, Pang, Zhiqing, and Zhang, Jun
- Published
- 2022
- Full Text
- View/download PDF
223. Low-temperature photothermal irradiation triggers alkyl radicals burst for potentiating cancer immunotherapy.
- Author
-
Ning, Bo, Liu, Yao, Ouyang, Boshu, Su, Xiaomin, Guo, Huishu, Pang, Zhiqing, and Shen, Shun
- Subjects
- *
ALKYL radicals , *IMMUNE checkpoint inhibitors , *IMMUNE checkpoint proteins , *REACTIVE oxygen species , *IMMUNOTHERAPY , *PROGRAMMED cell death 1 receptors , *NEURAL stimulation , *CYTOTOXIC T cells - Abstract
[Display omitted] Although low-temperature photothermal therapy (PTT) can sensitize tumors to immune checkpoint inhibition, its efficacy is still restricted in the deep and internal tumors without enough oxygen and lymphocytic infiltration. Non-oxygen-dependent alkyl radicals have been demonstrated to synergistically enhance PTT through up-regulating lipid peroxidation and reactive oxygen species (ROS). Herein, an innovative strategy based on alkyl radicals to augment immunogenetic cell death (ICD) caused by mild PTT was proposed to improve poor efficacy of immunotherapy, which composed of a photothermal material of Chinse ink, an azo-initiator of 2,2-azobis[2-(2- imidazoline -2- acyl) propane ]dihydrochloride (AIPH) and a PD-L1 inhibitor of HY19991 (HY). Upon near-infrared-II laser irradiation, low-temperature (<45℃) stimulation induced a high expression of immune checkpoint receptor (PD-L1) in tumors and triggered a large amount alkyl radicals generated by AIPH. Significantly, the alkyl radicals augmented the ICD and increased the recruitment of tumor-infiltrating lymphocytes against tumors after transformation of the immunologically cold tumor microenvironment into hot by mild PTT. The released HY further enhanced the immunotherapy effect by blocking the binding of activated T lymphocytes and PD-L1. In vivo studies exhibited that the all-in-one hydrogel with synergistic mechanisms had an extraordinary ability to reverse the immunosuppressive microenvironment, stimulate innate and adaptive immune responses to eliminate tumors and prevent metastasis. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
224. Targeted neutrophil-mimetic liposomes promote cardiac repair by adsorbing proinflammatory cytokines and regulating the immune microenvironment.
- Author
-
Chen, Jing, Song, Yanan, Wang, Qiaozi, Li, Qiyu, Tan, Haipeng, Gao, Jinfeng, Zhang, Ning, Weng, Xueyi, Sun, Dili, Yakufu, Wusiman, Wang, Zhengmin, Qian, Juying, Pang, Zhiqing, Huang, Zheyong, and Ge, Junbo
- Subjects
- *
LIPOSOMES , *CELL surface antigens , *BIOMOLECULES , *CYTOKINE receptors , *MYOCARDIAL infarction , *CYTOKINES , *ANTI-inflammatory agents - Abstract
Acute myocardial infarction (MI) induces a sterile inflammatory response that may result in poor cardiac remodeling and dysfunction. Despite the progress in anti-cytokine biologics, anti-inflammation therapy of MI remains unsatisfactory, due largely to the lack of targeting and the complexity of cytokine interactions. Based on the nature of inflammatory chemotaxis and the cytokine-binding properties of neutrophils, we fabricated biomimetic nanoparticles for targeted and broad-spectrum anti-inflammation therapy of MI. By fusing neutrophil membranes with conventional liposomes, we fabricated biomimetic liposomes (Neu-LPs) that inherited the surface antigens of the source cells, making them ideal decoys of neutrophil-targeted biological molecules. Based on their abundant chemokine and cytokine membrane receptors, Neu-LPs targeted infarcted hearts, neutralized proinflammatory cytokines, and thus suppressed intense inflammation and regulated the immune microenvironment. Consequently, Neu-LPs showed significant therapeutic efficacy by providing cardiac protection and promoting angiogenesis in a mouse model of myocardial ischemia–reperfusion. Therefore, Neu-LPs have high clinical translation potential and could be developed as an anti-inflammatory agent to remove broad-spectrum inflammatory cytokines during MI and other neutrophil-involved diseases. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
225. Dual targeting micelles loaded with paclitaxel and lapatinib for combinational therapy of brain metastases from breast cancer.
- Author
-
Lu, Heng, Chen, Tianran, Wang, Yiran, He, Yuwei, Pang, Zhiqing, and Wang, Yajie
- Subjects
- *
METASTATIC breast cancer , *BLOOD-brain barrier , *PACLITAXEL , *BRAIN metastasis , *LAPATINIB , *DRUG delivery systems , *MICELLES - Abstract
Due to the presence of the blood–brain barrier (BBB), the delivery of general drugs into the brain tissue remains to be a tricky problem. For patients with brain metastases from breast cancer, drug delivery systems must overcome this physical barrier. Targeted nano vehicles arise as a promising alternative to deliver drugs to brain tissues successively. Herein, a dual targeting micelle drug delivery system loaded with paclitaxel (PTX) and lapatinib (LPTN) was developed for combinational therapy of brain metastases. In our study, it was shown the micelles modified with Angiopep-2 had high loading efficiency of paclitaxel and lapatinib (Ang-MIC-PTX/LP). In addition, Ang-MIC-PTX/LP could transport across the in vitro BBB model and accumulate in breast cancer cells. After intravenous injection, Ang-MIC significantly accumulated in the brain metastasis. Ang-MIC-PTX/LP could also extend the life span of brain metastasis mouse models. Overall, this study provided a promising method for treatment of brain metastases from breast cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
226. Ruxolitinib-loaded cytokine nanosponge alleviated the cytokine storm and dampened macrophage overactivation for the treatment of hemophagocytic lymphohistiocytosis.
- Author
-
Wang, Honglan, Wang, Yiwei, Liu, Huiwen, Li, Xuejing, Sun, Chunyan, Pang, Zhiqing, Zhang, Bo, and Hu, Yu
- Subjects
- *
CYTOKINE release syndrome , *HEMOPHAGOCYTIC lymphohistiocytosis , *MACROPHAGES , *MACROPHAGE activation , *PHAGOCYTOSIS , *CYTOKINES - Abstract
Hemophagocytic lymphohistiocytosis (HLH) is a life-threatening clinical syndrome characterized by a positive feedback loop between cytokine storm and macrophages and lymphocytes overactivation, which could serve as a valid therapeutic target for HLH treatment. In this study, the clinically extensively used JAK1/2 inhibitor ruxolitinib was encapsulated into macrophage membrane-coated nanoparticles (M@NP-R) with high drug-loading efficiency for targeted HLH treatment. In vitro and in vivo studies demonstrated that M@NP-R not only efficiently adsorbed extracellular proinflammation cytokines, like IFN-γ and IL-6 to alleviate the cytokine storm, but also effectively dampened macrophage activation and proliferation by intracellular JAK/STAT signaling pathway inhibition. M@NP-R treatment significantly ameliorated the clinical and laboratory manifestations of HLH in mouse models, including trilineage cytopenia, hypercytokinemia, organomegaly, hepatorenal dysfunction, and tissue inflammation. Importantly, M@NP-R significantly enhanced the survival of the lethal HLH mice. Altogether, M@NP-R successfully blocked the positive feedback loop between the cytokine storm and macrophage overactivation by depleting extracellular inflammatory cytokines and inhibiting the intracellular JAK/STAT signaling pathway, both of which worked synergistically in HLH treatment. As ruxolitinib has already been extensively used in clinics with favorable safety, and M@NP is biodegradable and highly biocompatible, M@NP-R has good prospects for clinical translation. [ABSTRACT FROM AUTHOR]
- Published
- 2024
- Full Text
- View/download PDF
227. Endotoxin-adsorbing macrophage-mimetic hybrid liposome for sepsis treatment.
- Author
-
Jiang, Lixian, Li, Ruixiang, Xu, Jiazhen, Luan, Pengwei, Cui, Qianfei, Pang, Zhiqing, Wang, Jianxin, Lin, Guoqiang, and Zhang, Jiange
- Subjects
- *
GRAM-negative bacterial diseases , *ENDOTOXINS , *SEPTIC shock , *THERAPEUTICS , *SEPSIS , *MYCOSES - Abstract
• Biomimetic strategy for LPS elimination and sepsis treatment. • Integration of nature cell membranes and artificial materials. • Enhanced stability of cell membranes by fusion with PEGylated-lipids. • Prolonged circulation of cell membranes by fusion with PEGylated-lipids. Sepsis is a life-threatening condition resulting from bacterial or fungal infections. Common treatments for sepsis, such as antibiotic therapy, are far from satisfactory. Because endotoxin (LPS) is a major pathogenic factor in sepsis caused by gram-negative bacterial infections, neutralization of LPS is a promising therapeutic target. To effectively eliminate LPS, in the current study, a macrophage-mimetic hybrid liposome (M-Lipo) was developed by fusing a macrophage membrane (M-membrane) with artificial lipids. The M-membrane could provide an intrinsic binding site for LPS. The artificial PEGylated lipid could stabilize the natural membrane and prolong the circulation of M-Lipo in the bloodstream. These two membranes could complement one another and extend their biofunction as they were integrated. The results showed that M-Lipo had surface proteins similar to those of macrophages and could substantially adsorb LPS. With the help of the PEGylated lipids, M-Lipo presented much better stability than the bare M-membrane vesicle. In addition, the pharmacokinetic results revealed that M-Lipo clearly had longer retention (∼16%) in blood (at 12 h) than the natural M-membrane (∼3.3%). By combining these properties, the hybrid M-Lipo not only reduced the toxicity of LPS in vitro but also protected the mouse against endotoxic shock in vivo. In conclusion, the hybrid liposome M-Lipo has the advantages of both natural membranes and artificial materials, eventually leading to the successful treatment of sepsis. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
228. On-demand CO release for amplification of chemotherapy by MOF functionalized magnetic carbon nanoparticles with NIR irradiation.
- Author
-
Yao, Jizong, Liu, Yao, Wang, Jiawen, Jiang, Qin, She, Dejun, Guo, Huishu, Sun, Nianrong, Pang, Zhiqing, Deng, Chunhui, Yang, Wuli, and Shen, Shun
- Subjects
- *
CARBON nanotubes , *THERAPEUTIC use of nanostructured materials , *THERAPEUTIC use of carbon monoxide , *TUMOR treatment , *COMPANION diagnostics , *NEAR infrared radiation - Abstract
Abstract Carbon monoxide (CO) gas therapy combined with chemotherapy and photothermal therapy (PTT) is a promising treatment mode for malignant tumor. Herein, we firstly reported doxorubicin (DOX) loaded Mn carbonyl modified Fe (Ⅲ)-based nanoMOFs (MIL-100) coated PEGylated magnetic carbon nanoparticles (denoted as MCM@PEG-CO-DOX NPs) as theranostics nanoplatforms for near-infrared (NIR)-responded CO-DOX combination therapy. MIL-100 as a good nanocarrier of DOX with high loading capacity can also chelate the Mn carbonyl after a smart modification. Meanwhile, magnetic carbon core possessed photothermal effect, which can convert the NIR light to heat by an 808 nm laser irradiation, resulting in the on-demand release of CO and DOX. As a result, combining with PTT, MCM@PEG-CO-DOX NPs killed tumor efficiently. Moreover, our synthesized MCM@PEG-CO-DOX NPs were capable of realizing tumor dual-mode imaging including magnetic resonance imaging (MRI) and photoacoustic imaging (PAI). [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
229. Erythrocyte-cancer hybrid membrane-camouflaged melanin nanoparticles for enhancing photothermal therapy efficacy in tumors.
- Author
-
Jiang, Qin, Liu, Yao, Guo, Ranran, Yao, Xianxian, Sung, Seunghyun, Pang, Zhiqing, and Yang, Wuli
- Subjects
- *
ERYTHROCYTES , *TUMOR treatment , *PHOTOTHERMAL effect - Abstract
Abstract Cell membrane coating has emerged as an intriguing biomimetic strategy to endow nanomaterials with functions and properties inherent to source cells for various biomedical applications. Hybrid membrane of different types of cells could be coated onto nanoparticle surface to achieve additional functions. Herein, we fused red blood cell (RBC) membrane together with MCF-7 cell membrane and fabricated an erythrocyte-cancer (RBC-M) hybrid membrane-camouflaged melanin nanoparticle (Melanin@RBC-M) platform for enhancing therapeutic efficacy of photothermal therapy (PTT). The fused RBC-M hybrid membrane vesicles retained both RBC and MCF-7 cell membrane proteins and the resultant Melanin@RBC-M exhibited prolonged blood circulation and homotypic targeting to source MCF-7 cells simultaneously. Interestingly, increasing MCF-7 membrane components in RBC-M significantly enhanced the homotypic targeting function of Melanin@RBC-M while increasing RBC membrane components in RBC-M effectively reduced the cellular uptake of Melanin@RBC-M by macrophages and improved their circulation time in the blood. After intravenous injection into MCF-7 tumor-bearing athymic nude mice, Melanin@RBC-M with 1:1 membrane protein weight ratio of RBC to MCF-7 exhibited significantly higher tumor accumulation and better PTT efficacy compared with other Melanin@RBC-M with different membrane protein weight ratios as well as pristine melanin nanoparticles, due to the optimal balance between prolonged blood circulation and homotypic targeting. In addition, in vitro photoacoustic results revealed that Melanin@RBC-M had a photoacoustic signal enhancement with the increase of nanoparticle size (64 → 148 nm) and the photoacoustic amplitudes increased linearly with nanoparticle concentration at the excitation wavelength ranged from 680 nm to 800 nm, which could be used for quantification of Melanin@RBC-M in vivo. Looking forward, coating hybrid membrane onto nanoparticles could add flexibility and controllability in enhancing nanoparticles functionality and offer new opportunities for biomedical applications. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
230. Ferroptosis-enhanced chemotherapy for triple-negative breast cancer with magnetic composite nanoparticles.
- Author
-
Zhang, Jiaxin, Zhou, Kaicheng, Lin, Jingbo, Yao, Xianxian, Ju, Dianwen, Zeng, Xian, Pang, Zhiqing, and Yang, Wuli
- Subjects
- *
IRON oxide nanoparticles , *IRON oxides , *TRIPLE-negative breast cancer , *MAGNETIC nanoparticles , *MAGNETIC nanoparticle hyperthermia - Abstract
Triple-negative breast cancer (TNBC) causes great suffering to patients because of its heterogeneity, poor prognosis, and chemotherapy resistance. Ferroptosis is characterized by iron-dependent oxidative damage by accumulating intracellular lipid peroxides to lethal levels, and plays a vital role in the treatment of TNBC based on its intrinsic characteristics. To identify the relationship between chemotherapy resistance and ferroptosis in TNBC, we analyzed the single cell RNA-sequencing public dataset of GSE205551. It was found that the expression of Gpx4 in DOX-resistant TNBC cells was significantly higher than that in DOX-sensitive TNBC cells. Based on this finding, we hypothesize that inducing ferroptosis by inhibiting the expression of Gpx4 can reduce the resistance of TNBC to DOX and enhance the therapeutic effect of chemotherapy on TNBC. Herein, dihydroartemisinin (DHA)-loaded polyglutamic acid-stabilized Fe 3 O 4 magnetic nanoparticles (Fe 3 O 4 -PGA-DHA) was combined with DOX-loaded polyaspartic acid-stabilized Fe 3 O 4 magnetic nanoparticles (Fe 3 O 4 -PASP-DOX) for ferroptosis-enhanced chemotherapy of TNBC. Compared with Fe 3 O 4 -PASP-DOX, Fe 3 O 4 -PGA-DHA + Fe 3 O 4 -PASP-DOX demonstrated significantly stronger cytotoxicity against different TNBC cell lines and achieved significantly more intracellular accumulation of reactive oxygen species and lipid peroxides. Furthermore, transcriptomic analyses demonstrated that Fe 3 O 4 -PASP-DOX-induced apoptosis could be enhanced by Fe 3 O 4 -PGA-DHA-induced ferroptosis and Fe 3 O 4 -PGA-DHA + Fe 3 O 4 -PASP-DOX might trigger ferroptosis in MDA-MB-231 cells by inhibiting the PI3K/AKT/mTOR/GPX4 pathway. Fe 3 O 4 -PGA-DHA + Fe 3 O 4 -PASP-DOX showed superior anti-tumor efficacy on MDA-MB-231 tumor-bearing mice, providing great potential for improving the therapeutic effect of TNBC. [ABSTRACT FROM AUTHOR]
- Published
- 2023
- Full Text
- View/download PDF
231. BQ123 selectively improved tumor perfusion and enhanced nanomedicine delivery for glioblastomas treatment.
- Author
-
Zhang, Bo, Wang, Honglan, Jin, Kai, Jiang, Ting, Shen, Shun, Luo, Zimiao, Tuo, Yanyan, Jiang, Xinguo, Liu, Xianping, Hu, Yu, and Pang, Zhiqing
- Subjects
- *
NANOMEDICINE , *ENDOTHELINS , *MYOBLASTS , *NANOPARTICLES , *TUMOR treatment , *IMMUNOFLUORESCENCE - Abstract
Blood perfusion was always lower in tumor tissues as compared with that in surrounding normal tissues which lead to inadequate nanomedicine delivery to tumors. Inspired by the upregulation of both endothelin-1 (ET1) and its ETA receptor in tumor tissues and the crucial contribution of ET1-ETA receptor signaling to maintain myogenic tone of tumor vessels, we supposed that inhibition of ET1-ETA receptor signaling might selectively improve tumor perfusion and help deliver nanomedicine to tumors. Using human U87 MG glioblastomas with abundant vessels as the tumor model, immunofluorescence staining demonstrated that ETA receptor was overexpressed by in glioblastomas tissues compared with normal brain tissues. A single administration of ETA receptor antagonist BQ123 at the dose of 0.5 mg/kg could effectively improve tumor perfusion which was evidenced by in vivo photoacoustic imaging. Additionally, a single treatment of BQ123 could significantly improve the accumulation of nanoparticles (NPs) around 115 nm in tumors with a more homogeneous distribution pattern by in vivo imaging, ex vivo imaging as well as in vivo distribution experiments. Furthermore, BQ123 successfully increased the therapeutic benefits of paclitaxel-loaded NPs and significantly elongated the survival time of orthotropic glioblastomas-bearing animal models. In summary, the present study provided a new strategy to selectively improve tumor perfusion and therefore benefit nanomedicine delivery for tumor therapy. As ET1-ETA receptor signaling was upregulated in a variety of tumors, this strategy might open a new avenue for tumor treatment. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
232. Captopril improves tumor nanomedicine delivery by increasing tumor blood perfusion and enlarging endothelial gaps in tumor blood vessels.
- Author
-
Zhang, Bo, Jiang, Ting, Tuo, Yanyan, Jin, Kai, Luo, Zimiao, Shi, Wei, Mei, Heng, Hu, Yu, Pang, Zhiqing, and Jiang, Xinguo
- Subjects
- *
NANOMEDICINE , *DRUG delivery systems , *BLOOD vessels , *CAPTOPRIL , *TUMOR treatment , *THERAPEUTICS , *ANIMALS , *ANTHROPOMETRY , *ANTINEOPLASTIC agents , *BLOOD circulation , *BRAIN tumors , *CAPILLARY permeability , *CELL lines , *ENDOTHELIUM , *GLIOMAS , *MICE , *NANOPARTICLES , *PACLITAXEL , *TIME , *VASODILATORS , *PHARMACODYNAMICS - Abstract
Poor tumor perfusion and unfavorable vessel permeability compromise nanomedicine drug delivery to tumors. Captopril dilates blood vessels, reducing blood pressure clinically and bradykinin, as the downstream signaling moiety of captopril, is capable of dilating blood vessels and effectively increasing vessel permeability. The hypothesis behind this study was that captopril can dilate tumor blood vessels, improving tumor perfusion and simultaneously enlarge the endothelial gaps of tumor vessels, therefore enhancing nanomedicine drug delivery for tumor therapy. Using the U87 tumor xenograft with abundant blood vessels as the tumor model, tumor perfusion experiments were carried out using laser Doppler imaging and lectin-labeling experiments. A single treatment of captopril at a dose of 100 mg/kg significantly increased the percentage of functional vessels in tumor tissues and improved tumor blood perfusion. Scanning electron microscopy of tumor vessels also indicated that the endothelial gaps of tumor vessels were enlarged after captopril treatment. Immunofluorescence-staining of tumor slices demonstrated that captopril significantly increased bradykinin expression, possibly explaining tumor perfusion improvements and endothelial gap enlargement. Additionally, imaging in vivo, imaging ex vivo and nanoparticle distribution in tumor slices indicated that after a single treatment with captopril, the accumulation of 115-nm nanoparticles in tumors had increased 2.81-fold with a more homogeneous distribution pattern in comparison to non-captopril treated controls. Finally, pharmacodynamics experiments demonstrated that captopril combined with paclitaxel-loaded nanoparticles resulted in the greatest tumor shrinkage and the most extensive necrosis in tumor tissues among all treatment groups. Taken together, the data from the present study suggest a novel strategy for improving tumor perfusion and enlarging blood vessel permeability simultaneously in order to improve nanomedicine delivery for tumor therapy. As captopril has already been extensively used clinically, such a strategy has great therapeutic potential. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
233. Enhanced photothermal therapy of biomimetic polypyrrole nanoparticles through improving blood flow perfusion.
- Author
-
Wang, Xuejun, Li, Haichun, Liu, Xianping, Tian, Ye, Guo, Huishu, Jiang, Ting, Luo, Zimiao, Jin, Kai, Kuai, Xinping, Liu, Yao, Pang, Zhiqing, Yang, Wuli, and Shen, Shun
- Subjects
- *
BIOMIMETIC chemicals , *PHOTOTHERAPY , *POLYPYRROLE , *BLOOD flow , *NANOMEDICINE , *DRUG delivery systems - Abstract
In this study, we reported a strategy to improve delivery efficiency of a long-circulation biomimetic photothermal nanoagent for enhanced photothermal therapy through selectively dilating tumor vasculature. By using a simply nanocoating technology, a biomimetic layer of natural red blood cell (RBC) membranes was camouflaged on the surface of photothermal polypyrrole nanoparticles (PPy@RBC NPs). The erythrocyte-mimicking PPy NPs inherited the immune evasion ability from natural RBC resulting in superior prolonged blood retention time. Additionally, excellent photothermal and photoacoustic imaging functionalities were all retained attributing to PPy NPs cores. To further improve the photothermal outcome, the endothelin A (ET A ) receptor antagonist BQ123 was jointly employed to regulate tumor microenvironment. The BQ123 could induce tumor vascular relaxation and increase blood flow perfusion through modulating an ET-1/ET A transduction pathway and blocking the ET A receptor, whereas the vessel perfusion of normal tissues was not altered. Through our well-designed tactic, the concentration of biomimetic PPy NPs in tumor site was significantly improved when administered systematically. The study documented that the antitumor efficiency of biomimetic PPy NPs combined with specific antagonist BQ123 was particularly prominent and was superior to biomimetic PPy NPs ( P < 0.05) and PEGylated PPy NPs with BQ123 ( P < 0.01), showing that the greatly enhanced photothermal treatment could be achieved with low-dose administration of photothermal agents. Our findings would provide a promising procedure for other similar enhanced photothermal treatment by blocking ET A receptor to dramatically increase the delivery of biomimetic photothermal nanomaterials. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
234. Red blood cell membrane-camouflaged melanin nanoparticles for enhanced photothermal therapy.
- Author
-
Jiang, Qin, Luo, Zimiao, Men, Yongzhi, Yang, Peng, Peng, Haibao, Guo, Ranran, Tian, Ye, Pang, Zhiqing, and Yang, Wuli
- Subjects
- *
NANOMEDICINE , *ERYTHROCYTES , *MELANINS , *PHOTOTHERAPY , *CANCER treatment , *ANTINEOPLASTIC agents , *ACOUSTIC imaging - Abstract
Photothermal therapy (PTT) has represented a promising noninvasive approach for cancer treatment in recent years. However, there still remain challenges in developing non-toxic and biodegradable biomaterials with high photothermal efficiency in vivo . Herein, we explored natural melanin nanoparticles extracted from living cuttlefish as effective photothermal agents and developed red blood cell (RBC) membrane-camouflaged melanin (Melanin@RBC) nanoparticles as a platform for in vivo antitumor PTT. The as-obtained natural melanin nanoparticles demonstrated strong absorption at NIR region, higher photothermal conversion efficiency (∼40%) than synthesized melanin-like polydopamine nanoparticles (∼29%), as well as favorable biocompatibility and biodegradability. It was shown that RBC membrane coating on melanin nanoparticles retained their excellent photothermal property, enhanced their blood retention and effectively improved their accumulation at tumor sites. With the guidance of their inherited photoacoustic imaging capability, optimal accumulation of Melanin@RBC at tumors was achieved around 4 h post intravenous injection. Upon irradiation by an 808-nm laser, the developed Melanin@RBC nanoparticles exhibited significantly higher PTT efficacy than that of bare melanin nanoparticles in A549 tumor-bearing mice. Given that both melanin nanoparticles and RBC membrane are native biomaterials, the developed Melanin@RBC platform could have great potential in clinics for anticancer PTT. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
235. Precise glioblastoma targeting by AS1411 aptamer-functionalized poly (l-γ-glutamylglutamine)–paclitaxel nanoconjugates.
- Author
-
Luo, Zimiao, Yan, Zhiqiang, Jin, Kai, Pang, Qiang, Jiang, Ting, Lu, Heng, Liu, Xianping, Pang, Zhiqing, Yu, Lei, and Jiang, Xinguo
- Subjects
- *
GLIOBLASTOMA multiforme treatment , *APTAMERS , *AMINES , *PACLITAXEL , *BIOCONJUGATES - Abstract
Chemotherapy is still the main adjuvant strategy after surgery in glioblastoma therapy. As the main obstacles of chemotherapeutic drugs for glioblastoma treatment, the blood brain barrier (BBB) and non-specific delivery to non-tumor tissues greatly limit the accumulation of drugs into tumor tissues and simultaneously cause serious toxicity to nearby normal tissues which altogether compromised the chemotherapeutic effect. In the present study, we established an aptamer AS1411-functionalized poly ( l -γ-glutamyl-glutamine)-paclitaxel (PGG-PTX) nanoconjugates drug delivery system (AS1411-PGG-PTX), providing an advantageous solution of combining the precisely active targeting and the optimized solubilization of paclitaxel. The receptor nucleolin, highly expressed in glioblastoma U87 MG cells as well as neo-vascular endothelial cells, mediated the binding and endocytosis of AS1411-PGG-PTX nanoconjugates, leading to significantly enhanced uptake of AS1411-PGG-PTX nanoconjugates by tumor cells and three-dimension tumor spheroids, and intensive pro-apoptosis effect of AS1411-PGG-PTX nanoconjugates. In vivo fluorescence imaging and tissue distribution further demonstrated the higher tumor distribution of AS1411-PGG-PTX as compared with PGG-PTX. As a result, the AS1411-PGG-PTX nanoconjugates presented the best anti-glioblastoma effect with prolonged median survival time and most tumor cell apoptosis in vivo as compared with other groups. In conclusion, the AS1411-PGG-PTX nanoconjugates exhibited a promising targeting delivery strategy for glioblastoma therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2017
- Full Text
- View/download PDF
236. Corrigendum to 'On-demand CO release for amplification of chemotherapy by MOF functionalized magnetic carbon nanoparticles with NIR irradiation' [Biomaterials 195 (2019) 51–62].
- Author
-
Yao, Jizong, Liu, Yao, Wang, Jiawen, Jiang, Qin, She, Dejun, Guo, Huishu, Sun, Nianrong, Pang, Zhiqing, Deng, Chunhui, Yang, Wuli, and Shen, Shun
- Subjects
- *
MAGNETIC nanoparticles , *BIOMATERIALS , *CANCER chemotherapy , *IRRADIATION , *PHOTOTHERMAL effect - Published
- 2023
- Full Text
- View/download PDF
237. Cyclopamine disrupts tumor extracellular matrix and improves the distribution and efficacy of nanotherapeutics in pancreatic cancer.
- Author
-
Zhang, Bo, Jiang, Ting, Shen, Shun, She, Xiaojian, Tuo, Yanyan, Hu, Yu, Pang, Zhiqing, and Jiang, Xinguo
- Subjects
- *
CYCLOPAMINE , *TUMORS , *EXTRACELLULAR matrix , *PANCREATIC cancer , *ADENOCARCINOMA - Abstract
The dense extracellular matrix in pancreatic ductal adenocarcinoma dramatically reduces the penetration and efficacy of nanotherapeutics. Disruption of the tumor extracellular matrix may help improve the distribution and efficacy of nanotherapeutics in pancreatic cancer. In this study, we tested whether cyclopamine, a special inhibitor of the hedgehog signaling pathway with powerful anti-fibrotic activity, could promote the penetration and efficacy of nanotherapeutics in pancreatic cancer. It was shown that cyclopamine disrupted tumor extracellular fibronectins, decompressed tumor blood vessels, and improved tumor perfusion. Furthermore, cyclopamine improved the accumulation and intratumoral distribution of i.v.-administered fluorescence indicator-labeled nanoparticles. Finally, cyclopamine also significantly improved the tumor growth inhibition effect of i.v.-injected nanotherapeutics in pancreatic tumor xenograft mouse models. Thus, cyclopamine may have great potential to improve the therapeutic effects of nanomedicine in patients with pancreatic cancer. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
238. Fibrin degradation by rtPA enhances the delivery of nanotherapeutics to A549 tumors in nude mice.
- Author
-
Zhang, Bo, Jiang, Ting, She, Xiaojian, Shen, Shun, Wang, Sheng, Deng, Jun, Shi, Wei, Mei, Heng, Hu, Yu, Pang, Zhiqing, and Jiang, Xinguo
- Subjects
- *
TUMOR treatment , *FIBRIN , *TISSUE plasminogen activator , *NANOMEDICINE , *LABORATORY mice , *EXTRACELLULAR matrix - Abstract
Effective drug delivery to a tumor depends on favorable blood perfusion within the tumor. As an important component of tumor extracellular matrix, fibrin is abundant near tumor vessels. Inspired by the distinct distribution pattern and vessel-dependent production of fibrin, we hypothesized that fibrin depletion in tumors decompresses tumor vessels to improve tumor blood perfusion and accordingly enhance drug delivery to tumors rich in vessels. In the present study, we attempted to employ a clinically used thrombolytic drug, recombinant tissue plasminogen activator (rtPA), to modulate fibrin deposition in tumors. We then combined this drug with a nanoparticle drug delivery system for tumor therapy. RtPA treatment (25 mg/kg/d i. p. administration for two weeks) successfully depleted fibrin deposition and enhanced blood perfusion within A549 tumor xenografts. Furthermore, rtPA treatment also improved the in vivo delivery of 115-nm nanoparticles to tumor tissues. Finally, rtPA combined with therapeutic agent-loaded nanoparticles resulted in the most effective shrinkage of A549 tumor xenografts compared with the control groups. Overall, the present study provides a new strategy to enhance the delivery of nanotherapeutics to tumors rich in vessels. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
239. Fibrin-targeting peptide CREKA-conjugated multi-walled carbon nanotubes for self-amplified photothermal therapy of tumor.
- Author
-
Zhang, Bo, Wang, Huafang, Shen, Shun, She, Xiaojian, Shi, Wei, Chen, Jun, Zhang, Qizhi, Hu, Yu, Pang, Zhiqing, and Jiang, Xinguo
- Subjects
- *
CANCER treatment , *FIBRIN , *MULTIWALLED carbon nanotubes , *PHOTOTHERMAL effect , *PEPTIDE analysis , *DRUG delivery systems - Abstract
Inability of nanomedicine to efficiently home to tumor site still poses great challenge in tumor drug delivery. Inspired by the amplified formation of fibrin in clotting cascade, a self-amplified drug delivery system was developed for tumor photothermal therapy (CMWNTs-PEG) using multi-walled carbon nanotubes (MWNTs) with favorable photothermal effect as the vector, polyethylene glycol as the shelter, CREKA peptide with special affinity for fibrin as the targeting moiety and NIR illumination as the external power. The self-amplified targeting property was carefully characterized. The in vivo temperature monitoring experiment demonstrated that CMWNTs-PEG could significantly elevate the temperature in the tumor region than its counterpart 24 h post an initial NIR illumination. The in vivo imaging and biodistribution experiment showed IR783-labeled CMWNTs-PEG with illumination could accumulate in tumors tissues about 6.4-fold higher than control group, much stronger than other treatment groups. In vivo distribution experiments revealed Cy3-labeled CMWNTs-PEG could deposit on the wall of tumor vessels, intravascular and extravascular spaces, far more extensive than its counterpart in tumor slices. The pharmacodynamics experiment revealed that after four times of illumination, the CMWNTs-PEG almost totally eradiated the tumor xenografts. Altogether, the self-amplified targeting system CMWNTs-PEG showed strong tumor targeting capacity and powerful photothermal therapeutic efficacy. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
240. A multimodal Pepstatin A peptide-based nanoagent for the molecular imaging of P-glycoprotein in the brains of epilepsy rats.
- Author
-
Yu, Xiangrong, Wang, Jianhong, Liu, Jiansheng, Shen, Shun, Cao, Zhonglian, Pan, Jiawei, Zhou, Shuyi, Pang, Zhiqing, Geng, Daoying, and Zhang, Jun
- Subjects
- *
PEPSTATIN , *P-glycoprotein , *TREATMENT of epilepsy , *LABORATORY rats , *GENETIC overexpression , *ANTICONVULSANTS , *COMBINED modality therapy - Abstract
Regional overexpression of the multidrug transporter P-glycoprotein (P-gp) in epileptic brain tissues may lower antiepileptic drugs concentrations at the target site and contribute to pharmacoresistance in refractory epilepsy. However, few techniques are available to quantitate the level of P-gp expression noninvasively in vivo. In this study, we developed a nanoagent by conjugating superparamagnetic iron oxide nanoparticles with a near infrared probe and the targeting element Pepstatin A, a peptide with specific affinity for P-gp. In a rat model of epilepsy, the nanoagent was readily and selectively accumulated within epileptogenic cerebral regions, which were detectable by both magnetic resonance imaging and optical imaging modalities. This P-gp-targeted nanoagent could be used not only in the molecular imaging of P-gp expression changes in seizure-induced regional, understanding the mechanisms of P-gp disorders, and the prediction of refractory epilepsy, but also in targeted therapies with P-gp modulators. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
241. Precisely co-delivery of protein and ROS scavenger with platesomes for enhanced endothelial barrier preservation against myocardial ischemia reperfusion injury.
- Author
-
Gao, Jinfeng, Song, Yanan, Wang, Qiaozi, Chen, Jing, Li, Qiyu, Tan, Haipeng, Yakufu, Wusiman, Zhang, Ning, Li, Su, Zhang, Jinyan, Yang, Hongbo, Wang, Zhengmin, Weng, Xueyi, Sun, Dili, Wang, Qibing, Li, Jia, Qian, Juying, Pang, Zhiqing, Huang, Zheyong, and Ge, Junbo
- Subjects
- *
MYOCARDIAL reperfusion , *THROMBIN receptors , *REPERFUSION injury , *MYOCARDIAL ischemia , *IRON oxides , *ENDOTHELIAL cells , *CELL receptors - Abstract
[Display omitted] • Intercellular VE-Cadherin maintanence and intracellular ROS scavenging are critical in endothelial barrier preservation. • Platelet membrane fused liposome facilitates targeted delivery. • Locally activated thrombin triggers extracellular release of ANGPTL4. • Neutral endothelial cytoplasm is suitable for Fe 3 O 4 to scavenge ROS. • Extracellular and intracellular drugs release are combined for enhanced endothelial barrier preservation. Following myocardial ischemia–reperfusion (MI/R), the endothelial barrier is heavily damaged. This is partially due to the accumulation of intracellular reactive oxygen species (ROS) in endothelial cells, leading to disruptive intercellular VE-Cadherin junctions and endothelial cells apoptosis. Systemic administration of drugs is limited by poor delivery and release at pathological sites, resulting in compromised therapeutic effect. Herein, by exploiting the inherent nature of platelets migrating to injured endothelium and local activation of thrombin, we fabricated functional platesomes (Fe@PLP-TR-A) to co-deliver angiopoietin-like 4 (ANGPTL4) and ROS scavenger Fe 3 O 4 to injured endothelium in heart affected by MI/R. Thanks to the capability for multidrug loading and easy-modification feature of platesomes, we conjugated ANGPTL4 on the surface of platesomes with a thrombin responsive peptide and encapsulated Fe 3 O 4 in the inner space. After systemic injection, Fe@PLP-TR-A successfully arrived at the endothelial cells at injured endothelium due to the platelet-mimetic adhesion effect of platesomes which was fabricated by fusing platelet membranes and lipid membranes. Following the cleavage of thrombin-responsive peptide, which was activated by injured endothelium, ANGPTL4 was released extracellularly and bound to the receptor on endothelial cells to prevent the disruption of VE-Cadherin junctions. Further, endo -lysosomal escape of Fe 3 O 4 to cytosol exerted ROS scavenging effect and protected endothelial cells from apoptosis. VE-Cadherin maintenance and cell rescue together preserved the endothelial barrier and subsequently decreased leukocytes extravasation and intracardial hemorrhage, alleviated cardiomyocyte apoptosis and eventually improved cardiac function. This work demonstrates a practical platform for the treatment of reperfusion injury by enhancing targeted release of drugs to injured endothelium. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
242. RGD functionalized polymeric nanoparticles targeting periodontitis epithelial cells for the enhanced treatment of periodontitis in dogs.
- Author
-
Yao, Wenxin, Xu, Peicheng, Zhao, Jingjing, Ling, Li, Li, Xiaoxia, Zhang, Bo, Cheng, Nengneng, and Pang, Zhiqing
- Subjects
- *
NANOPARTICLES , *PERIODONTITIS treatment , *EPITHELIAL cells , *DOG physiology , *TARGETED drug delivery , *GINGIVAL fluid - Abstract
Long term retention of antimicrobials with effective drug concentration in gingival crevicular fluid (GCF) is of vital importance for the treatment of chronic periodontitis. In this study, a novel epithelial cell-targeting nanoparticle drug delivery system by conjugating minocycline-loaded poly(ethylene glycol)–poly(lactic acid) (PEG–PLA) nanoparticles (NP-MIN) with RGD peptide were developed and administrated locally for targeting periodontitis epithelial cells and enhancing the treatment of periodontitis in dogs. Biodegradable NP-MIN was made with an emulsion/solvent evaporation technique. RGD peptide was conjugated to the surface of nanoparticles via Maleimide group reaction with hydrosulfide in RGD peptide (RGD-NP-MIN). Transmission electron microscopy examination and dynamic light scattering results revealed that RGD-NP-MIN had a sphere shape, with a mean diameter around 106 nm. In vitro release of minocycline from RGD-NP-MIN showed that RGD modification did not change the remarkable sustained releasing characteristic of NP-MIN. To elucidate the interaction of RGD-NP and epithelial cells, RGD-NP binding, uptake and cellular internalization mechanisms by calu-3 cells were investigated. It was shown RGD modification significantly enhanced nanoparticles binding and uptake by Calu-3 cells, and RGD-NP uptake was an energy-dependent process through receptor-mediated endocytosis. Both clathrin-associated endocytosis and caveolae-dependent endocytosis pathway were involved in the RGD-NP uptake, and the intracellular transport of RGD-NP was related to lysosome and Golgi apparatus. Finally, i n vivo pharmacokinetics of minocycline in the periodontal pockets and anti-periodontitis effects of RGD-NP-MIN on periodontitis-bearing dogs were evaluated. After local administration of RGD-NP-MIN, minocycline concentration in gingival crevicular fluid decreased slowly and maintained an effective drug concentration for a longer time than that of NP-MIN. Anti-periodontitis effects demonstrated that RGD-NP-MIN could significantly decrease symptoms of periodontitis, which was better than any other control group. These findings suggested that these epithelial cell-targeting nanoparticles offered a novel and effective local delivery system for the treatment of periodontitis. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
243. CREKA peptide-conjugated dendrimer nanoparticles for glioblastoma multiforme delivery.
- Author
-
Zhao, Jingjing, Zhang, Bo, Shen, Shun, Chen, Jun, Zhang, Qizhi, Jiang, Xinguo, and Pang, Zhiqing
- Subjects
- *
GLIOBLASTOMA multiforme , *CENTRAL nervous system cancer , *NANOMEDICINE , *DRUG carriers , *POLYAMIDOAMINE dendrimers , *IRON oxide nanoparticles ,MORTALITY risk factors - Abstract
Glioblastoma multiforme (GBM) is the most aggressive central nervous system (CNS) tumor because of its fast development, poor prognosis, difficult control and terrible mortality. Poor penetration and retention in the glioblastoma parenchyma were crucial challenges in GBM nanomedicine therapy. Nanoparticle diameter can significantly influence the delivery efficiency in tumor tissue. Decreasing nanoparticle size can improve the nanoparticle penetration in tumor tissue but decrease the nanoparticle retention effect. Therefore, small nanoparticles with high retention effect in tumor are urgently needed for effective GBM drug delivery. In present study, a small nanoparticle drug delivery system was developed by conjugating fibrin-binding peptide CREKA to Polyamidoamine (PAMAM) dendrimer, where PEGylated PAMAM is used as drug carrier due to its small size and good penetration in tumor and CREKA is used to target the abundant fibrin in GBM for enhanced retention in tumor. In vitro binding ability tests demonstrated that CREKA can significantly enhanced nanoparticle binding with fibrin. In vivo fluorescence imaging of GBM bearing nude mice, ex vivo brain imaging and frozen slices fluorescence imaging further revealed that the CREKA-modified PAMAM achieved higher accumulation and deeper penetration in GBM tissue than unmodified one. These results indicated that the CREKA-modified PAMAM could penetrate the GBM tissue deeply and enhance the retention effect, which was a promising strategy for brain tumor therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
244. UPA-sensitive ACPP-conjugated nanoparticles for multi-targeting therapy of brain glioma.
- Author
-
Zhang, Bo, Zhang, Yujie, Liao, Ziwei, Jiang, Ting, Zhao, Jingjing, Tuo, Yanyan, She, Xiaojian, Shen, Shun, Chen, Jun, Zhang, Qizhi, Jiang, Xinguo, Hu, Yu, and Pang, Zhiqing
- Subjects
- *
PLASMINOGEN activators , *NANOPARTICLES , *GLIOMA treatment , *DRUG delivery systems , *CELL-penetrating peptides , *SPHEROIDIZING (Heat treatment) , *PHARMACODYNAMICS , *THERAPEUTICS - Abstract
Now it is well evidenced that tumor growth is a comprehensive result of multiple pathways, and glioma parenchyma cells and stroma cells are closely associated and mutually compensatory. Therefore, drug delivery strategies targeting both of them simultaneously might obtain more promising therapeutic benefits. In the present study, we developed a multi-targeting drug delivery system modified with uPA-activated cell-penetrating peptide (ACPP) for the treatment of brain glioma (ANP). In vitro experiments demonstrated nanoparticles (NP) decorated with cell-penetrating peptide (CPP) or ACPP could significantly improve nanoparticles uptake by C6 glioma cells and nanoparticles penetration into glioma spheroids as compared with traditional NP and thus enhanced the therapeutic effects of its payload when paclitaxel (PTX) was loaded. In vivo imaging experiment revealed that ANP accumulated more specifically in brain glioma site than NP decorated with or without CPP. Brain slides further showed that ACPP contributed to more nanoparticles accumulation in glioma site, and ANP could co-localize not only with glioma parenchyma cells, but also with stroma cells including neo-vascular cells and tumor-associated macrophages. The pharmacodynamics results demonstrated ACPP could significantly improve the therapeutic benefits of nanoparticles by significantly prolonging the survival time of glioma-bearing mice. In conclusion, the results suggested that nanoparticles modified with uPA-sensitive ACPP could reach multiple types of cells in glioma tissues and provide a novel strategy for glioma targeted therapy. [ABSTRACT FROM AUTHOR]
- Published
- 2015
- Full Text
- View/download PDF
245. iNGR-modified PEG-PLGA nanoparticles that recognize tumor vasculature and penetrate gliomas.
- Author
-
Kang, Ting, Gao, Xiaoling, Hu, Quanyin, Jiang, Di, Feng, Xingye, Zhang, Xue, Song, Qingxiang, Yao, Lei, Huang, Meng, Jiang, Xinguo, Pang, Zhiqing, Chen, Hongzhuan, and Chen, Jun
- Subjects
- *
NANOMEDICINE , *POLYETHYLENE glycol , *GLYCOLIC acid , *GLIOMA treatment , *AMINOPEPTIDASES , *ENDOTHELIAL cells , *TUMORS - Abstract
Abstract: A major cross-cutting problem for glioma therapy is the poor extravasation and penetration of the payload drug in target glioma parenchyma. Here, to overcome these obstacles, a tumor vessel recognizing and tumor penetrating system is developed by functionalizating the poly (ethyleneglycol)-poly (l-lactic-co-glycolic acid) nanoparticles with an iNGR moiety (iNGR-NP). The nanoparticulate formulation is expected to achieve specific deep penetration in the tumor tissue by initially binding to aminopeptidase N, with iNGR proteolytically cleaved to CRNGR, and then bind with neuropilin-1 to mediate deep penetration in the tumor parenchyma. iNGR-NP exhibits significantly enhanced cellular uptake in human umbilical vein endothelial cells, improves the anti-proliferation and anti-tube formation abilities of paclitaxel in vitro. Following intravenous administration, iNGR-NP present favorable pharmacokinetic and tumor homing profiles. Glioma distribution and penetration assays confirm that iNGR-NP achieve the highest accumulation and deepest penetration at the glioma sites. The anti-glioma efficacy of paclitaxel-loaded iNGR-NP is verified by its improved anti-angiogenesis activity and the significantly prolonged survival time in mice bearing intracranial glioma. These evidences highlight the potential of iNGR-decorated nanoparticles in overcoming the leading edge problem in anti-glioma drug delivery. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
246. EGFP–EGF1-conjugated nanoparticles for targeting both neovascular and glioma cells in therapy of brain glioma.
- Author
-
Zhang, Bo, Wang, Huafang, Liao, Ziwei, Wang, Yu, Hu, Yue, Yang, Jiarong, Shen, Shun, Chen, Jun, Mei, Heng, Shi, Wei, Hu, Yu, Pang, Zhiqing, and Jiang, Xinguo
- Subjects
- *
EPIDERMAL growth factor , *BIOCONJUGATES , *NEOVASCULARIZATION , *GLIOMAS , *UMBILICAL veins , *ENDOTHELIAL cells - Abstract
Abstract: As neovascular and glioma cells were closely associated and might be mutually promoted in glioma growth, a dual-targeting strategy targeting to both neovascular and glioma cells would be more promising as compared with those targeting one of them. In this study, we reported a drug delivery system where nanoparticles were decorated with EGFP–EGF1 (ENP), a fusion protein derived from factor VII with special affinity for tissue factor (TF) over-expressed in glioma tissues, to facilitate anti-glioma delivery of paclitaxel (PTX) by targeting both neovascular and glioma cells. In vitro protein binding assay demonstrated that EGFP–EGF1 bound well to C6 cells and perturbed human umbilical vein endothelial cells (HUVEC) with a concentration-dependent manner but not to unperturbed HUVEC. EGFP–EGF1–TF interaction significantly enhanced nanoparticles uptake by perturbed HUVEC and glioma C6 cells as well as nanoparticles penetration in C6 glioma spheroids, and thus improved the cytotoxicity of their payload in both monolayer cells and glioma spheroids models. In vivo imaging of glioma-bearing mice demonstrated the specific accumulation of ENP in glioma tissues. In vivo distribution of nanoparticles intuitively showed ENP mainly sited in both extravascular glioma cells and neovascular cells. Pharmacodynamic results revealed that PTX-loaded ENP (ENP–PTX) significantly prolonged the median survival time of glioma-bearing mice compared with that of any other group. TUNEL assay and H&E staining showed that ENP–PTX treatment induced significantly more cell apoptosis and tumor necrosis compared with other treatments. In conclusion, the results of this contribution demonstrated the great potential of EGFP–EGF1-functionalized nanoparticles for dual-targeting therapy of brain glioma. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
247. Targeting fibronectins of glioma extracellular matrix by CLT1 peptide-conjugated nanoparticles.
- Author
-
Zhang, Bo, Shen, Shun, Liao, Ziwei, Shi, Wei, Wang, Yu, Zhao, Jingjing, Hu, Yue, Yang, Jiarong, Chen, Jun, Mei, Heng, Hu, Yu, Pang, Zhiqing, and Jiang, Xinguo
- Subjects
- *
FIBRONECTINS , *TARGETED drug delivery , *GLIOMAS , *EXTRACELLULAR matrix , *CELL-penetrating peptides , *DRUG delivery systems - Abstract
Abstract: The abundant extracellular matrix (ECM) in the glioma microenvironment play a critical role in the maintenance of glioma morphology, glioma cells differentiation and proliferation, but little has been done to understand the feasibility of ECM as the therapeutic target for glioma therapy. In this study, a drug delivery system targeting fibronectins (FNs), a prevailing component in the ECM of many solid tumors, was constructed for glioma therapy based on the interaction between the abundant FNs in glioma tissues and the FNs-targeting moiety CLT1 peptide. CLT1 peptide was successfully conjugated to PEG-PLA nanoparticles (CNP). FNs were demonstrated to be highly expressed in the ECM of glioma spheroids in vitro and glioma tissues in vivo. CLT1 modification favored targeting nanoparticles penetration into the core of glioma spheroids and consequently induced more severe inhibitive effects on glioma spheroids growth than traditional NP. In vivo imaging, ex vivo imaging and glioma tissue slides showed that CNP enhanced nanoparticles retention in glioma site, distributed more extensively and more deeply into glioma tissues than that of conventional NP, and mainly located in glioma cells rather than in extracellular matrix as conventional NP. Pharmacodynamics outcomes revealed that the median survival time of glioma-bearing mice models treated with paclitaxel-loaded CNP (CNP-PTX) was significantly prolonged when compared with that of any other group. TUNEL assay demonstrated that more extensive cell apoptosis was induced by CNP-PTX treatment compared with other treatments. Altogether, these promising results indicated that this ECM-targeting drug delivery system enhanced retention and glioma cell uptake of nanoparticles and might have a great potential for glioma therapy in clinical applications. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
248. Multimodal SPION-CREKA peptide based agents for molecular imaging of microthrombus in a rat myocardial ischemia-reperfusion model.
- Author
-
Song, Yanan, Huang, Zheyong, Xu, Jianfeng, Ren, Daoyuan, Wang, Yu, Zheng, Xinde, Shen, Yunli, Wang, Lili, Gao, Hongxiang, Hou, Jiayun, Pang, Zhiqing, Qian, Juying, and Ge, Junbo
- Subjects
- *
THROMBOSIS diagnosis , *CORONARY disease , *MYOCARDIAL reperfusion , *CARDIOVASCULAR diseases , *MOLECULAR diagnosis , *IRON oxide nanoparticles - Abstract
Abstract: Microthrombosis plays a key role in many cardiovascular diseases. Although it is not difficult to localize thrombus within large or middle-sized vessels, the noninvasive diagnostic regimen for the detection of microthrombus remains scarce. Here we developed a nanoagent by conjucting superparamagnetic iron-oxide nanoparticle with fluorophore and a targeting element, CREKA, a peptide with special affinity for fibrin. In a rat model of myocardial ischemia–reperfusion (MI/R), the multimodal nanoagents were readily and selectively accumulated within microthrombosis, which was detectable by both magnetic resonance and optical imaging modalities. The fibrin-targeted nanoagent could be expected to have utility not only in molecular imaging of fibrin, understanding the mechanisms of microcirculation disorders, but also in targeted therapy with fibrinolytic agents. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
249. Tumor cells and neovasculature dual targeting delivery for glioblastoma treatment.
- Author
-
Gao, Huile, Yang, Zhi, Cao, Shijie, Xiong, Yang, Zhang, Shuang, Pang, Zhiqing, and Jiang, Xinguo
- Subjects
- *
GLIOBLASTOMA multiforme treatment , *TARGETED drug delivery , *DRUG delivery systems , *BRAIN tumors , *CANCER cell proliferation , *NEOVASCULARIZATION , *LABORATORY mice - Abstract
Abstract: Glioblastoma multiforme (GBM), one of the most common primary malignant brain tumors, was characterized by angiogenesis and tumor cells proliferation. Antiangiogenesis and antitumor combination treatment gained much attention because of the potency in dual inhibition of both the tumor proliferation and the tumor invasion. In this study, a neovasculature and tumor cell dual targeting delivery system was developed through modification of nanoparticles with interleukin-13 peptide and RGD (IRNPs), in which interleukin-13 peptide was targeting GBM cells and RGD was targeting neovasculature. To evaluate the potency in GBM treatment, docetaxel was loaded into IRNPs. In vitro, interleukin-13 peptide and RGD could enhance the corresponding cells (C6 and human umbilical vein endothelial cells) uptake and cytotoxicity. In combination, IRNPs showed high uptake in both cells and increased the cytotoxicity on both cells. In vivo, IRNPs could effectively deliver cargoes to GBM with higher intensity than mono-modified nanoparticles. Correspondingly, docetaxel-IRNPs displayed best anti-tumor effect with a median survival time of 35 days, which was significantly longer than that of mono-modified and unmodified nanoparticles. Importantly, treatment with docetaxel-IRNPs could avoid the accumulation of HIF1α in GBM site, which was crucial for the tumor invasion. After the treatment, there was no obvious change in normal organs of mice. [Copyright &y& Elsevier]
- Published
- 2014
- Full Text
- View/download PDF
250. Targeted immunomodulation therapy for cardiac repair by platelet membrane engineering extracellular vesicles via hitching peripheral monocytes.
- Author
-
Li, Qiyu, Huang, Zheyong, Wang, Qiaozi, Gao, Jinfeng, Chen, Jing, Tan, Haipeng, Li, Su, Wang, Zhengmin, Weng, Xueyi, Yang, Hongbo, Pang, Zhiqing, Song, Yanan, Qian, Juying, and Ge, Junbo
- Subjects
- *
MYOCARDIAL reperfusion , *EXTRACELLULAR vesicles , *MONOCYTES , *IMMUNOREGULATION , *BLOOD platelets , *MESENCHYMAL stem cells , *MYOCARDIAL infarction - Abstract
Immune regulation therapies have been considered promising in the treatment of myocardial ischemia reperfusion (MI/R) injury. Mesenchymal stem cells derived extracellular vesicles (MSC-EVs) are of great potential for immune modulation by reprogramming macrophages but their therapeutic efficacy is hindered by insufficient targeting ability in vivo. Herein, we introduced the platelet membrane modified EVs (P-EVs) based on membrane fusion method to mimic the binding ability of platelets to monocytes. In the mouse model of MI/R injury, the intravenously injected P-EVs were mainly carried by circulating monocytes into the ischemic myocardium. In the inflammatory microenvironment, those monocytes subsequently differentiated into macrophages with enhanced phagocytosis, which probably promoted in-situ endocytosis of the superficial P-EVs by monocytes differentiated macrophages in large quantities. Then, the P-EVs successfully escaped from the macrophage lysosome and released the functional microRNAs (miRNAs) into the cytosol which facilitated the inflammatory macrophages (M1 phenotype) reprogramming to reparative macrophages (M2 phenotype). Finally, the immune microenvironment was regulated to realize cardiac repair. Thus, we supposed that the most likely delivery method was that monocytes mediated P-EVs migration into ischemic myocardium where P-EVs were mainly in-situ endocytosed by monocytes derived macrophages, which holds potential for immunoregulation on MI/R and other immune-related diseases in the future. [ABSTRACT FROM AUTHOR]
- Published
- 2022
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.