1
|
Taledaohan A, Tuohan MM, Jia R, Wang K, Chan L, Jia Y, Wang F, Wang Y. An RGD-Conjugated Prodrug Nanoparticle with Blood-Brain-Barrier Penetrability for Neuroprotection Against Cerebral Ischemia-Reperfusion Injury. Antioxidants (Basel) 2024; 13:1339. [PMID: 39594481 PMCID: PMC11591307 DOI: 10.3390/antiox13111339] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/26/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024] Open
Abstract
Cerebral ischemia-reperfusion injury significantly contributes to global morbidity and mortality. Loganin is a natural product with various neuroprotective effects; however, it lacks targeted specificity for particular cells or receptors, which may result in reduced therapeutic efficacy and an increased risk of side effects. To address the limitations of loganin, we developed LA-1, a novel compound incorporating an Arg-Gly-Asp (RGD) peptide to target integrin receptor αvβ3, enhancing brain-targeting efficacy. LA-1 exhibited optimal nanoscale properties, significantly improved cell viability, reduced ROS production, and enhanced survival rates in vitro. In vivo, LA-1 decreased infarct sizes, improved neurological function, and reduced oxidative stress and neuroinflammation. Proteomic analysis showed LA-1 modulates PI3K/Akt and Nrf2/HO-1 pathways, providing targeted neuroprotection. These findings suggest LA-1's potential for clinical applications in treating cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Ayijiang Taledaohan
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (A.T.); (M.M.T.); (R.J.); (K.W.); (L.C.); (Y.J.)
- Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, China
| | - Maer Maer Tuohan
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (A.T.); (M.M.T.); (R.J.); (K.W.); (L.C.); (Y.J.)
- Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, China
| | - Renbo Jia
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (A.T.); (M.M.T.); (R.J.); (K.W.); (L.C.); (Y.J.)
- Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, China
| | - Kai Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (A.T.); (M.M.T.); (R.J.); (K.W.); (L.C.); (Y.J.)
- Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, China
| | - Liujia Chan
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (A.T.); (M.M.T.); (R.J.); (K.W.); (L.C.); (Y.J.)
- Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, China
| | - Yijiang Jia
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (A.T.); (M.M.T.); (R.J.); (K.W.); (L.C.); (Y.J.)
- Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, China
| | - Feng Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (A.T.); (M.M.T.); (R.J.); (K.W.); (L.C.); (Y.J.)
- Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, China
| | - Yuji Wang
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences of Capital Medical University, Beijing 100069, China; (A.T.); (M.M.T.); (R.J.); (K.W.); (L.C.); (Y.J.)
- Department of Medicinal Chemistry, Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, 10 Xi Tou Tiao, You An Men, Beijing 100069, China
| |
Collapse
|
2
|
Xie Q, Zhou J, He C, Xu Y, Tao F, Hu M. Unlocking the intricacies: Exploring the complex interplay between platelets and ovarian cancer. Crit Rev Oncol Hematol 2024; 202:104465. [PMID: 39097249 DOI: 10.1016/j.critrevonc.2024.104465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 07/24/2024] [Accepted: 07/28/2024] [Indexed: 08/05/2024] Open
Abstract
Ovarian cancer, an aggressive malignancy of the female reproductive tract, is frequently linked to an elevated risk of thrombotic events. This association is manifested by a pronounced rise in platelet counts and activation levels. Current research firmly supports the pivotal role of platelets in the oncogenic processes of ovarian cancer, influencing tumor cell proliferation and metastasis. Platelets influence these processes through direct interactions with tumor cells or by secreting cytokines and growth factors that enhance tumor growth, angiogenesis, and metastasis. This review aims to thoroughly dissect the interactions between platelets and ovarian cancer cells, emphasizing their combined role in tumor progression and associated thrombotic events. Additionally, it summarizes therapeutic strategies targeting platelet-cancer interface which show significant promise. Such approaches could not only be effective in managing the primary ovarian tumor but also play a pivotal role in preventing metastasis and attenuating thrombotic complications associated with ovarian cancer.
Collapse
Affiliation(s)
- Qianxin Xie
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Jie Zhou
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chaonan He
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Ye Xu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fangfang Tao
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| | - Mengjiao Hu
- Department of Immunology and Microbiology, School of Basic Medical Sciences, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
3
|
Huang Y, Wang J, Guo Y, Park SY, Yang H, Lu A, Li Y, Chen R. Selective binding of cationic fibrinogen-mimicking chitosan nanoparticles to activated platelets and efficient drug release for antithrombotic therapy. Int J Biol Macromol 2024; 268:131742. [PMID: 38653430 DOI: 10.1016/j.ijbiomac.2024.131742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 02/29/2024] [Accepted: 04/19/2024] [Indexed: 04/25/2024]
Abstract
Thrombosis is the main cause of catastrophic events including ischemic stroke, myocardial infarction and pulmonary embolism. Acetylsalicylic acid (ASA) therapy offers a desirable approach to antithrombosis through a reduction of platelet reactivity. However, major bleeding complications, severe off-target side effects, and resistance or nonresponse to ASA greatly attenuate its clinical outcomes. Herein, we report a cationic fibrinogen-mimicking nanoparticle, denoted as ASA-RGD-CS@TPP, to achieve activated-platelet-targeted delivery and efficient release of ASA for safer and more effective antithrombotic therapy. This biomimetic antithrombotic system was prepared by one-pot ionic gelation between cationic arginine-glycine-aspartic acid (RGD)-grafted chitosan (RGD-CS) and anionic tripolyphosphate (TPP). The platform exhibited selective binding to activated platelets, leading to efficient release of ASA and subsequent attenuation of platelet functions, including the remarkable inhibition of platelet aggregation through a potent blockage of cyclooxygenase-1 (COX-1). After intravenous administration, ASA-RGD-CS@TPP displayed significantly prolonged circulation time and successful prevention of thrombosis in a mouse model. ASA-RGD-CS@TPP was demonstrated to significantly enhance antithrombotic therapy while showing minimal coagulation and hemorrhagic risks and excellent biocompatibility in vivo as compared to free ASA. This platform provides a simple, safe, effective and targeted strategy for the development of antithrombotic nanomedicines.
Collapse
Affiliation(s)
- Yu Huang
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China; Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom.
| | - Jiahua Wang
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China
| | - Yuanyuan Guo
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China
| | - Seun Young Park
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Hongtian Yang
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Annabelle Lu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom
| | - Yuehua Li
- Department of Radiology, Shanghai Jiao Tong University School of Medicine Affiliated Shanghai Sixth People's Hospital, 600 Yi Shan Road, Shanghai 200233, PR China.
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, United Kingdom.
| |
Collapse
|
4
|
Abstract
In order to improve bioavailability, stability, control release, and target delivery of active pharmaceutical ingredients (APIs), as well as to mask their bitter taste, to increase their efficacy, and to minimize their side effects, a variety of microencapsulation (including nanoencapsulation, particle size <100 nm) technologies have been widely used in the pharmaceutical industry. Commonly used microencapsulation technologies are emulsion, coacervation, extrusion, spray drying, freeze-drying, molecular inclusion, microbubbles and microsponge, fluidized bed coating, supercritical fluid encapsulation, electro spinning/spray, and polymerization. In this review, APIs are categorized by their molecular complexity: small APIs (compounds with low molecular weight, like Aspirin, Ibuprofen, and Cannabidiol), medium APIs (compounds with medium molecular weight like insulin, peptides, and nucleic acids), and living microorganisms (such as probiotics, bacteria, and bacteriophages). This article provides an overview of these microencapsulation technologies including their processes, matrix, and their recent applications in microencapsulation of APIs. Furthermore, the advantages and disadvantages of these common microencapsulation technologies in terms of improving the efficacy of APIs for pharmaceutical treatments are comprehensively analyzed. The objective is to summarize the most recent progresses on microencapsulation of APIs for enhancing their bioavailability, control release, target delivery, masking their bitter taste and stability, and thus increasing their efficacy and minimizing their side effects. At the end, future perspectives on microencapsulation for pharmaceutical applications are highlighted.
Collapse
Affiliation(s)
- Cuie Yan
- Division of Encapsulation, Blue California, Rancho Santa Margarita, California 92688, United States
| | - Sang-Ryoung Kim
- Division of Encapsulation, Blue California, Rancho Santa Margarita, California 92688, United States
| |
Collapse
|
5
|
Liu L, Ding W, He L, Yang Y, Guan F, Sun X, Peng Y, Chen X, Zhao W, Xiao Y, Luo P. RGD and Scutellarin Conjugate (WK001) Targeting Platelet Glycoprotein IIb/IIIa Receptor Protects from Myocardial Ischemia/Reperfusion Injury: Synthesis, Characterization, and Bioactivity Evaluation. Bioconjug Chem 2023; 34:477-488. [PMID: 36740781 DOI: 10.1021/acs.bioconjchem.2c00439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Myocardial ischemia/reperfusion (MI/R) injury is an unresolved clinical challenge. The blockade of binding fibrinogen by glycoprotein IIb/IIIa (GPIIb-IIIa) inhibitors has become a new therapeutic approach against MI/R injury. In this study, we modified the RGD structure to combine with scutellarin and synthesized a novel peptide, scutellarin-HomoArg-Gly-Asp-Trp-NH2 (WK001). Herein, reported experimental and docking evidence indicates that WK001 provides immediate and potent platelet inhibition, with stronger inhibition of platelet aggregation than eptifibatide and scutellarin. In particular, it is administered intravenously to prevent thrombus formation and attenuate myocardial fibrosis progression in vivo. Therefore, WK001 could be developed as an antiplatelet drug to treat thrombosis-associated diseases, such as stroke and myocardial infarction.
Collapse
Affiliation(s)
- Lancong Liu
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau999078, China
| | - Wenfeng Ding
- Shenzhen Winkey Technology Co., Ltd., Shenzhen518000, China
| | - Lili He
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau999078, China
| | - Yi Yang
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau999078, China
| | - Fuyi Guan
- Shenzhen Winkey Technology Co., Ltd., Shenzhen518000, China
| | - Xinlin Sun
- Shenzhen Winkey Technology Co., Ltd., Shenzhen518000, China
| | - Yan Peng
- Shenzhen Winkey Technology Co., Ltd., Shenzhen518000, China
| | - Xue Chen
- Shenzhen Winkey Technology Co., Ltd., Shenzhen518000, China
| | - Wenhao Zhao
- Shenzhen Winkey Technology Co., Ltd., Shenzhen518000, China
| | - Yu Xiao
- Shenzhen Winkey Technology Co., Ltd., Shenzhen518000, China
| | - Pei Luo
- State Key Laboratories for Quality Research in Chinese Medicines, Macau University of Science and Technology, Macau999078, China
| |
Collapse
|
6
|
Advanced drug delivery system against ischemic stroke. J Control Release 2022; 344:173-201. [PMID: 35248645 DOI: 10.1016/j.jconrel.2022.02.036] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2021] [Revised: 02/28/2022] [Accepted: 02/28/2022] [Indexed: 02/06/2023]
|
7
|
Liu H, Pietersz G, Peter K, Wang X. Nanobiotechnology approaches for cardiovascular diseases: site-specific targeting of drugs and nanoparticles for atherothrombosis. J Nanobiotechnology 2022; 20:75. [PMID: 35135581 PMCID: PMC8822797 DOI: 10.1186/s12951-022-01279-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 01/21/2022] [Indexed: 02/18/2023] Open
Abstract
Atherosclerosis and atherothrombosis, the major contributors to cardiovascular diseases (CVDs), represent the leading cause of death worldwide. Current pharmacological therapies have been associated with side effects or are insufficient at halting atherosclerotic progression effectively. Pioneering work harnessing the passive diffusion or endocytosis properties of nanoparticles and advanced biotechnologies in creating recombinant proteins for site-specific delivery have been utilized to overcome these limitations. Since CVDs are complex diseases, the most challenging aspect of developing site-specific therapies is the identification of an individual and unique antigenic epitope that is only expressed in lesions or diseased areas. This review focuses on the pathological mechanism of atherothrombosis and discusses the unique targets that are important during disease progression. We review recent advances in site-specific therapy using novel targeted drug-delivery and nanoparticle-carrier systems. Furthermore, we explore the limitations and future perspectives of site-specific therapy for CVDs.
Collapse
Affiliation(s)
- Haikun Liu
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia
| | - Geoffrey Pietersz
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Burnet Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia.,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia.,Department of Medicine, Monash University, Melbourne, VIC, Australia.,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC, 3004, Australia. .,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia. .,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia. .,Department of Medicine, Monash University, Melbourne, VIC, Australia. .,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia.
| |
Collapse
|
8
|
|
9
|
Yu L, Guo Y, Chang Z, Zhang D, Zhang S, Pei H, Pang J, Zhao ZJ, Chen Y. Bidirectional Interaction Between Cancer Cells and Platelets Provides Potential Strategies for Cancer Therapies. Front Oncol 2021; 11:764119. [PMID: 34722319 PMCID: PMC8551800 DOI: 10.3389/fonc.2021.764119] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 09/28/2021] [Indexed: 12/13/2022] Open
Abstract
Platelets are essential components in the tumor microenvironment. For decades, clinical data have demonstrated that cancer patients have a high risk of thrombosis that is associated with adverse prognosis and decreased survival, indicating the involvement of platelets in cancer progression. Increasing evidence confirms that cancer cells are able to induce production and activation of platelets. Once activated, platelets serve as allies of cancer cells in tumor growth and metastasis. They can protect circulating tumor cells (CTCs) against the immune system and detachment-induced apoptosis while facilitating angiogenesis and tumor cell adhesion and invasion. Therefore, antiplatelet agents and platelet-based therapies should be developed for cancer treatment. Here, we discuss the mechanisms underlying the bidirectional cancer-platelet crosstalk and platelet-based therapeutic approaches.
Collapse
Affiliation(s)
- Liuting Yu
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Yao Guo
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Zhiguang Chang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Dengyang Zhang
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Shiqiang Zhang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Hanzhong Pei
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| | - Jun Pang
- Department of Urology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Zhizhuang Joe Zhao
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK, United States
| | - Yun Chen
- Edmond H. Fischer Translational Medical Research Laboratory, Scientific Research Center, The Seventh Affiliated Hospital, Sun Yat-Sen University, Shenzhen, China
| |
Collapse
|
10
|
Ichikawa M, Goto H. Synthesis of a polyacetylene derivative bearing aspirin with chiral charge carrier “chiralions”. JOURNAL OF MACROMOLECULAR SCIENCE PART A-PURE AND APPLIED CHEMISTRY 2021. [DOI: 10.1080/10601325.2021.1981765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Affiliation(s)
- Mai Ichikawa
- Department of Materials Science, Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| | - Hiromasa Goto
- Department of Materials Science, Faculty of Pure and Applied Sciences, University of Tsukuba, Tsukuba, Japan
| |
Collapse
|
11
|
Friedrich RP, Janko C, Unterweger H, Lyer S, Alexiou C. SPIONs and magnetic hybrid materials: Synthesis, toxicology and biomedical applications. PHYSICAL SCIENCES REVIEWS 2021. [DOI: 10.1515/psr-2019-0093] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Abstract
In the past decades, a wide variety of different superparamagnetic iron oxide nanoparticles (SPIONs) have been synthesized. Due to their unique properties, such as big surface-to-volume ratio, superparamagnetism and comparatively low toxicity, they are principally well suited for many different technical and biomedical applications. Meanwhile, there are a numerous synthesis methods for SPIONs, but high requirements for biocompatibility have so far delayed a successful translation into the clinic. Moreover, depending on the planned application, such as for imaging, magnetic drug targeting, hyperthermia or for hybrid materials intended for regenerative medicine, specific physicochemical and biological properties are inevitable. Since a summary of all existing SPION systems, their properties and application is far too extensive, this review reports on selected methods for SPION synthesis, their biocompatibility and biomedical applications.
Collapse
Affiliation(s)
- Ralf P. Friedrich
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| | - Christina Janko
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| | - Harald Unterweger
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| | - Stefan Lyer
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| | - Christoph Alexiou
- Department of Otorhinolaryngology, Head and Neck Surgery , Section of Experimental Oncology and Nanomedicine (SEON), Else Kröner-Fresenius-Stiftung-Professorship Universitätsklinikum , Erlangen , Germany
| |
Collapse
|
12
|
Zhang L, Li Z, Ye X, Chen Z, Chen ZS. Mechanisms of thrombosis and research progress on targeted antithrombotic drugs. Drug Discov Today 2021; 26:2282-2302. [PMID: 33895314 DOI: 10.1016/j.drudis.2021.04.023] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Revised: 04/14/2021] [Accepted: 04/15/2021] [Indexed: 12/26/2022]
Abstract
Globally, the incidence of thromboembolic diseases has increased in recent years, accompanied by an increase in patient mortality. Currently, several targeting delivery strategies have been developed to treat thromboembolic diseases. In this review, we discuss the mechanisms of thrombolysis and current anticoagulant drugs, particularly those with targeting capability, highlighting advances in the accurate treatment of thrombolysis with fewer adverse effects. Such approaches include magnetic drug-loading systems combined with molecular imaging to recanalize blood vessels and systems based on chimeric Arg-Gly-Asp (RGD) sequences that can target platelet glycoprotein receptor. With such progress in targeted antithrombotic drugs, targeted thrombolysis treatment shows significant potential benefit for patients.
Collapse
Affiliation(s)
- Lei Zhang
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Zhen Li
- Fujian Cancer Hospital, Fujian Provincial Cancer Hospital of Fujian Medical University, Fuzhou 350014, China
| | - Xianren Ye
- Fujian Cancer Hospital, Fujian Provincial Cancer Hospital of Fujian Medical University, Fuzhou 350014, China.
| | - Zhuo Chen
- State Key Laboratory of Structural Chemistry, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou 350002, China; University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, NY 11439, USA.
| |
Collapse
|
13
|
Cirillo M, Giacomini D. Molecular Delivery of Cytotoxic Agents via Integrin Activation. Cancers (Basel) 2021; 13:299. [PMID: 33467465 PMCID: PMC7830197 DOI: 10.3390/cancers13020299] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/16/2022] Open
Abstract
Integrins are cell adhesion receptors overexpressed in tumor cells. A direct inhibition of integrins was investigated, but the best inhibitors performed poorly in clinical trials. A gained attention towards these receptors arouse because they could be target for a selective transport of cytotoxic agents. Several active-targeting systems have been developed to use integrins as a selective cell entrance for some antitumor agents. The aim of this review paper is to report on the most recent results on covalent conjugates between integrin ligands and antitumor drugs. Cytotoxic drugs thus conjugated through specific linker to integrin ligands, mainly RGD peptides, demonstrated that the covalent conjugates were more selective against tumor cells and hopefully with fewer side effects than the free drugs.
Collapse
Affiliation(s)
| | - Daria Giacomini
- Department of Chemistry “Giacomo Ciamician”, Alma Mater Studiorum University of Bologna, Via Selmi 2, 40126 Bologna, Italy;
| |
Collapse
|
14
|
Xu J, Zhang Y, Nie G. Intelligent antithrombotic nanomedicines: Progress, opportunities, and challenges. VIEW 2021. [DOI: 10.1002/viw.20200145] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Junchao Xu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing China
| | - Yinlong Zhang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing China
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials & Nanosafety CAS Center for Excellence in Nanoscience National Center for Nanoscience and Technology Beijing China
- Center of Materials Science and Optoelectronics Engineering University of Chinese Academy of Sciences Beijing China
- GBA Research Innovation Institute for Nanotechnology Guangdong China
- Henan Institute of Advanced Technology Zhengzhou University Zhengzhou China
| |
Collapse
|
15
|
He H, Adili R, Liu L, Hong K, Holinstat M, Schwendeman A. Synthetic high-density lipoproteins loaded with an antiplatelet drug for efficient inhibition of thrombosis in mice. SCIENCE ADVANCES 2020; 6:6/49/eabd0130. [PMID: 33277254 PMCID: PMC7821904 DOI: 10.1126/sciadv.abd0130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2020] [Accepted: 10/21/2020] [Indexed: 05/12/2023]
Abstract
Antiplatelet agents offer a desirable approach to thrombosis prevention through the reduction of platelet reactivity. However, major bleeding events greatly attenuate the clinical outcomes of most antithrombotic agents. Therefore, the development of safer and more effective strategies to prevent vascular occlusion and avoid bleeding is urgently needed. A reconstituted nanoparticle, synthetic high-density lipoprotein (sHDL), which mimics the native HDL, has been established as clinically safe and is easily manufactured on a large scale. In this study, we propose that the delivery of the antiplatelet drug ML355, a selective inhibitor of 12(S)-lipoxygenase (12-LOX), by sHDL will efficiently inhibit thrombosis by targeting ML355 to the intended site of action, improving the pharmaceutical profile and harnessing the innate antithrombotic efficacy of the sHDL carrier. Our data show that ML355-sHDL exhibits more potent inhibition of thrombus formation in both small arterioles and larger arteries in mice without impairing the normal hemostasis in vivo.
Collapse
Affiliation(s)
- Hongliang He
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Reheman Adili
- Department of Pharmacology, University of Michigan Medical School, 1150 W. Medical Center Dr., Room 2220D, Medical Sciences Research Building III, Ann Arbor, MI 48109, USA
| | - Lisha Liu
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Kristen Hong
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA
| | - Michael Holinstat
- Department of Pharmacology, University of Michigan Medical School, 1150 W. Medical Center Dr., Room 2220D, Medical Sciences Research Building III, Ann Arbor, MI 48109, USA
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of Michigan Medical School, 1150 W. Medical Center Dr., Room 2220D, Medical Sciences Research Building III, Ann Arbor, MI 48109, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Michigan, 428 Church St., Ann Arbor, MI 48109, USA.
- Biointerfaces Institute, NCRC, 2800 Plymouth Rd., Ann Arbor, MI 48109, USA
| |
Collapse
|
16
|
Abstract
Further complications associated with infection by severe acute respiratory syndrome coronavirus 2 (a.k.a. SARS-CoV-2) continue to be reported. Very recent findings reveal that 20-30% of patients at high risk of mortality from COVID-19 infection experience blood clotting that leads to stroke and sudden death. Timely assessment of the severity of blood clotting will be of enormous help to clinicians in determining the right blood-thinning medications to prevent stroke or other life-threatening consequences. Therefore, rapid identification of blood-clotting-related proteins in the plasma of COVID-19 patients would save many lives. Several nanotechnology-based approaches are being developed to diagnose patients at high risk of death due to complications from COVID-19 infections, including blood clots. This Perspective outlines (i) the significant potential of nanomedicine in assessing the risk of blood clotting and its severity in SARS-CoV-2 infected patients and (ii) its synergistic roles with advanced mass-spectrometry-based proteomics approaches in identifying the important protein patterns that are involved in the occurrence and progression of this disease. The combination of such powerful tools might help us understand the clotting phenomenon and pave the way for development of new diagnostics and therapeutics in the fight against COVID-19.
Collapse
Affiliation(s)
- Amir Ata Saei
- Division
of Physiological Chemistry I, Department of Medical Biochemistry and
Biophysics, Karolinska Institutet, 171 65 Stockholm, Sweden
| | - Shahriar Sharifi
- Precision
Health Program and Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Morteza Mahmoudi
- Precision
Health Program and Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
17
|
Zhang X, Zhang Y, Wang Y, Wu J, Chen H, Zhao M, Peng S. Modifying ICCA with Trp-Phe-Phe to Enhance in vivo Activity and Form Nano-Medicine. Int J Nanomedicine 2020; 15:465-481. [PMID: 32021191 PMCID: PMC6982437 DOI: 10.2147/ijn.s229856] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 12/15/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND 1-(4-isopropylphenyl)-β-carboline-3-carboxylic acid (ICCA) was modified by Trp-Phe-Phe to form 1-(4-isopropylphenyl)-β-carboline-3-carbonyl-Trp-Phe-Phe (ICCA-WFF). PURPOSE The object of preparing ICCA-WFF was to enhance the in vivo efficacy of ICCA, to explore the possible targeting action, and to visualize the nano-feature. METHODS The advantages of ICCA-WFF over ICCA were demonstrated by a series of in vivo assays, such as anti-tumor assay, anti-arterial thrombosis assay, anti-venous thrombosis assay, P-selectin expression assay, and GPIIb/IIIa expression assay. The nano-features of ICCA-WFF were visualized by TEM, SEM and AFM images. The thrombus targeting and tumor-targeting actions were evidenced by FT-MS spectrum analysis. RESULTS The minimal effective dose of ICCA-WFF slowing tumor growth and inhibiting thrombosis was 10-fold lower than that of ICCA. ICCA-WFF, but not ICCA, formed nano-particles capable of safe delivery in blood circulation. In vivo ICCA-WFF, but not ICCA, can target thrombus and tumor. In thrombus and tumor, ICCA-WFF released Trp-Phe-Phe and/or ICCA. CONCLUSION Modifying ICCA with Trp-Phe-Phe successfully enhanced the anti-tumor activity, improved the anti-thrombotic action, formed nano-particles, targeted tumor tissue and thrombus, and provided an oligopeptide modification strategy for heterocyclic compounds.
Collapse
Affiliation(s)
- Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Yixin Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Haiyan Chen
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Beijing Laboratory of Biomedical Materials and Key Laboratory of Biomedical Materials of Natural Macromolecules, Department of Biomaterials, College of Materials Science and Engineering, Beijing University of Chemical Technology, Beijing100026, People’s Republic of China
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| |
Collapse
|
18
|
Synthesis and biological evaluation of RGD conjugated with Ketoprofen/Naproxen and radiolabeled with [ 99mTc] via N4(GGAG) for α Vβ 3 integrin-targeted drug delivery. ACTA ACUST UNITED AC 2019; 28:87-96. [PMID: 31845157 DOI: 10.1007/s40199-019-00318-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Accepted: 11/14/2019] [Indexed: 01/03/2023]
Abstract
BACKGROUND Integrins are interesting targets in oncology. RGD sequence has high affinity for αVβ3 integrin receptors. Diagnostic/therapeutic agents can be selectively delivered into cancer cells overexpressing αVβ3 integrin by using RGD as a carrier. Nonsteroidal anti-inflammatory drugs (NSAIDs) have shown anticancer properties in in vitro and in vivo studies. The anti-cancer properties of NSAIDs occur though COX-2 inhibition. Regarding the anti-cancer properties of NSAIDs and overexpression of COX-2 enzyme in cancer cells, targeted delivery of NSAIDs into cancer cells to maximize their efficiency and minimize their side effects may gain increased clinical interest. OBJECTIVES In this study, RGD was conjugated to ketoprofen/Naproxen to selectively transfer these non-selective COX inhibitors into cancer cells. METHODS Keto/Nap-RGD-N4 peptides were synthesized based on solid phase fmoc peptide synthesis. Radiolabeling with [99mTc] via N4 (GGAG) ligand was done for biological evaluation. Affinity and specificity of Keto/Nap-RGD-N4 to integrin was determined using A2780, OVCAR-3, SKOV-3 and HT-1080 cell lines. Percentage of Intenalization was measured in A2780 cells. Biodistriburion was studied in normal and tumor model mice. RESULTS Radiolabeled compounds showed high affinity to cells expressing αVβ3 integrin in comparison to cells not expressing αVβ3. The affinity to A2780 was significantly higher than OVCAR-3 cells. The %internalization into A2780 cells was quite low. Compounds showed more than 50% inhibition on A2780 and OVCAR-3 cells, less than 10% on MCF-7 and HT-1080 cells and no cytotoxicity on fibroblast cells after 48 h incubation. Although uptake of radiolabeled compounds in tumor was high at 1 h post-injection, the tumor/blood ratio was less than 1.5 which made SPECT imaging impossible. CONCLUSION Provided that NSAID drugs are conjugated to RGD, there will be a selective delivery to target tissues as well as synergetic anti-tumor effects which reduce systemic doses and toxicity. Graphical abstract.
Collapse
|
19
|
Liu W, Mao Y, Zhang X, Wang Y, Wu J, Zhao S, Peng S, Zhao M. RGDV-modified gemcitabine: a nano-medicine capable of prolonging half-life, overcoming resistance and eliminating bone marrow toxicity of gemcitabine. Int J Nanomedicine 2019; 14:7263-7279. [PMID: 31686807 PMCID: PMC6737205 DOI: 10.2147/ijn.s212978] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2019] [Accepted: 08/15/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Gemcitabine has been widely used as a chemotherapeutic drug. However, drug resistance, short half-life and side effects seriously decrease its chemotherapeutic efficacy. PURPOSE The object of preparing RGDV-gemcitabine was to prolong the half-life, to overcome drug resistance and to eliminate bone marrow toxicity of gemcitabine. METHODS Arg-Gly-Asp-Val was coupled with gemcitabine, forming 4-(Arg-Gly-Asp-Val-amino)-1-[3,3-difluoro-4-hydroxy-5-(hydroxylmethyl)oxo-lan-2-yl]pyrimidin-2-one (RGDV-gemcitabine) involving 9-step reactions. The advantages of RGDV-gemcitabine to gemcitabine were demonstrated by a series of assays, such as in vitro half-life assay, in vitro drug resistance assay, in vivo anti-tumor assay, in vivo kidney toxicity assay, in vivo liver toxicity assay and in vivo marrow toxicity assay. The nano-features of RGDV-gemcitabine were visualized by TEM, SEM and AFM images. The tumor-targeting action and release of RGDV-gemcitabine were evidenced by FT-MS spectra. RESULTS Half-life and anti-tumor activity of RGDV-gemcitabine were 17-fold longer and 10-fold higher than that of gemcitabine, respectively. RGDV-gemcitabine, but not gemcitabine, showed no kidney toxicity, no liver toxicity, no marrow toxicity and no drug resistance. The advantages attributed to the nanofeatures of RGDV-gemcitabine were targeting tumor tissue and releasing gemcitabine in tumor tissue. CONCLUSION RGDV-gemcitabine successively overcame the defects of gemcitabine and provided a practical strategy of nano-medicine.
Collapse
Affiliation(s)
- Wenchao Liu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Yujia Mao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Department of Medicinal Chemistry, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Department of Biomaterials, Beijing Laboratory of Biomedical Materials and Key Laboratory of Biomedical Materials of Natural Macromolecules, Beijing University of Chemical Technology, Beijing100026, People’s Republic of China
| |
Collapse
|
20
|
Wu J, Cui Y, Zhang X, Gui L, Wang Y, Peng S, Zhao M. BCESA: a nano-scaled intercalator capable of targeting tumor tissue and releasing anti-tumoral β-carboline-3-carboxylic acid. Int J Nanomedicine 2019; 14:3027-3041. [PMID: 31118620 PMCID: PMC6508158 DOI: 10.2147/ijn.s187600] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 03/20/2019] [Indexed: 12/12/2022] Open
Abstract
Background: In the discovery of DNA intercalators, β-carbolines compose one member of the most interesting alkaloid family and are of clinical importance. In the efforts, N-(3-benzyloxycarbonyl-β-carboline-1-yl)ethyl-Ser-Ala-OBzl (BCESA) was designed as a nano-scaled DNA intercalator without Dox-like toxicity. Methods: Based on the structural analysis and CDOCKER energy comparison, BCESA was rationally designed as such a nano-scaled intercalator. The anti-tumor activity, the toxicity and the tumor targeting action of BCESA were evaluated on mouse models. Results: The in vitro proliferation of cancer cells, but not non-cancer cells, was effectively inhibited by BCESA. On S180 mouse model BCESA dose-dependently slowed the tumor growth, and 0.01 μmol/kg/day was found as a minimal effective dose. Both BCESA and its moiety were found in the tumor tissue, but not in the organs and the blood, of S180 mice. Conclusion: BCESA should be a nano-scaled intercalator capable of targeting tumor tissue to release anti-tumoral β-carboline-3-carboxylic acid and its 1-methyl derivative, while Ser-Ala-OBzl is a simple and desirable carrier.
Collapse
Affiliation(s)
- Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Yue Cui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Lin Gui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, School of Pharmaceutical Sciences, Capital Medical University, Beijing100069, People’s Republic of China
- Beijing Laboratory of Biomedical Materials and Key Laboratory of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology), Ministry of Education, Beijing, People’s Republic of China
| |
Collapse
|
21
|
Huang Y, Yu L, Ren J, Gu B, Longstaff C, Hughes AD, Thom SA, Xu XY, Chen R. An activated-platelet-sensitive nanocarrier enables targeted delivery of tissue plasminogen activator for effective thrombolytic therapy. J Control Release 2019; 300:1-12. [PMID: 30807804 DOI: 10.1016/j.jconrel.2019.02.033] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/17/2019] [Accepted: 02/21/2019] [Indexed: 11/28/2022]
Abstract
It remains a major challenge to develop a selective and effective fibrinolytic system for thrombolysis with minimal undesirable side effects. Herein, we report a multifunctional liposomal system (164.6 ± 5.3 nm in diameter) which can address this challenge through targeted delivery and controlled release of tissue plasminogen activator (tPA) at the thrombus site. The tPA-loaded liposomes were PEGylated to improve their stability, and surface coated with a conformationally-constrained, cyclic arginine-glycine-aspartic acid (cRGD) to enable highly selective binding to activated platelets. The in vitro drug release profiles at 37 °C showed that over 90% of tPA was released through liposomal membrane destabilization involving membrane fusion upon incubation with activated platelets within 1 h, whereas passive release of the encapsulated tPA in pH 7.4 PBS buffer was 10% after 6 h. The release of tPA could be readily manipulated by changing the concentration of activated platelets. The presence of activated platelets enabled the tPA-loaded, cRGD-coated, PEGylated liposomes to induce efficient fibrin clot lysis in a fibrin-agar plate model and the encapsulated tPA retained 97.4 ± 1.7% of fibrinolytic activity as compared with that of native tPA. Furthermore, almost complete blood clot lysis was achieved in 75 min, showing considerably higher and quicker thrombolytic activity compared to the tPA-loaded liposomes without cRGD labelling. These results suggest that the nano-sized, activated-platelet-sensitive, multifunctional liposomes could facilitate selective delivery and effective release of tPA at the site of thrombus, thus achieving efficient clot dissolution whilst minimising undesirable side effects.
Collapse
Affiliation(s)
- Yu Huang
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Li Yu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Jie Ren
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Boram Gu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Colin Longstaff
- Biotherapeutics Section, National Institute for Biological Standards and Control, South Mimms, Herts, United Kingdom
| | - Alun D Hughes
- Institute of Cardiovascular Science, University College London, London, United Kingdom; MRC Unit for Lifelong Health and Ageing at University College London, London, United Kingdom
| | - Simon A Thom
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, London, United Kingdom
| | - Xiao Yun Xu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, United Kingdom
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, United Kingdom.
| |
Collapse
|
22
|
Kim KS, Song CG, Kang PM. Targeting Oxidative Stress Using Nanoparticles as a Theranostic Strategy for Cardiovascular Diseases. Antioxid Redox Signal 2019; 30:733-746. [PMID: 29228781 PMCID: PMC6350062 DOI: 10.1089/ars.2017.7428] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
SIGNIFICANCE Nanomedicine is an application of nanotechnology that provides solutions to unmet medical challenges. The unique features of nanoparticles, such as their small size, modifiable components, and diverse functionality, make them attractive and suitable materials for novel diagnostic, therapeutic, or theranostic applications. Cardiovascular diseases (CVDs) are the major cause of noncommunicable illness in both developing and developed countries. Nanomedicine offers novel theranostic options for the treatment of CVDs. Recent Advances: Many innovative nanoparticles to target reactive oxygen species (ROS) have been developed. In this article, we review the characteristics of nanoparticles that are responsive to ROS, their limitations, and their potential clinical uses. Significant advances made in diagnosis of atherosclerosis and treatment of acute coronary syndrome using nanoparticles are discussed. CRITICAL ISSUES Although there is a tremendous potential for the nanoparticle applications in medicine, their safety should be considered while using in humans. We discuss the challenges that may be encountered with some of the innovative nanoparticles used in CVDs. FUTURE DIRECTIONS The unique properties of nanoparticles offer novel diagnostic tool and potential therapeutic strategies. However, nanomedicine is still in its infancy, and further in-depth studies are needed before wide clinical application is achieved.
Collapse
Affiliation(s)
- Kye S Kim
- 1 Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,2 Harvard Medical School, Boston, Massachusetts
| | - Chul Gyu Song
- 3 Department of Electronic Engineering, Chonbuk National University, Jeonju, South Korea
| | - Peter M Kang
- 1 Cardiovascular Institute, Beth Israel Deaconess Medical Center, Boston, Massachusetts.,2 Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
23
|
Lu Y, Hu Q, Jiang C, Gu Z. Platelet for drug delivery. Curr Opin Biotechnol 2018; 58:81-91. [PMID: 30529814 DOI: 10.1016/j.copbio.2018.11.010] [Citation(s) in RCA: 126] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Accepted: 11/14/2018] [Indexed: 01/09/2023]
Abstract
Platelets play a vital physiological role in hemostasis, inflammation and tissue regeneration, which are associated with wound healing as well as cancer development and metastasis. These years, a variety of platelet-mediated drug delivery approaches have been developed due to their unique properties, such as quick replenishment and site-specific activation/adhesion. In this Current Opinion, focuses are put on strategies leveraging the physiological functions of platelets for the design of drug delivery systems, including platelet engineering, platelet hitchhiking, membrane coating, synthetic platelet fabrication and platelet-triggered drug release for different applications.
Collapse
Affiliation(s)
- Yifei Lu
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China
| | - Quanyin Hu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, United States
| | - Chen Jiang
- Key Laboratory of Smart Drug Delivery, Ministry of Education, State Key Laboratory of Medical Neurobiology, Department of Pharmaceutics, School of Pharmacy, Fudan University, Shanghai 201203, China.
| | - Zhen Gu
- Department of Bioengineering, University of California, Los Angeles, CA 90095, United States; California NanoSystems Institute, Jonsson Comprehensive Cancer Center, and Center for Minimally Invasive Therapeutics (C-MIT), University of California, Los Angeles, CA 90095, United States.
| |
Collapse
|
24
|
Heptapeptide-based modification leading to enhancing the action of MTCA on activated platelets, P-selectin, GPIIb/IIIa. Future Med Chem 2018; 10:1957-1970. [PMID: 29973078 DOI: 10.4155/fmc-2018-0055] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
AIM The modification of platelet inhibitor to enhance its targeting capacity toward platelets is of clinical importance. Thus, (1R, 3S)-1-methyl-1, 2, 3, 4-tetrahydro-β-carboline-3-carboxylic acid (MTCA), a platelet inhibitor, was modified with Lys(Pro-Ala-Lys)-Arg-Gly-Asp-Val (KKV), platelet targeting peptide, to form MTCA-KKV. MATERIALS & METHODS MTCA and MTCA-KKV were synthesized to identify the effect of KKV modification on MTCA and platelets. RESULTS Atomic force microscopy imaged MTCA-KKV effectively accumulated on activated platelets. UV spectra showed that MTCA-KKV concentration dependently changed P-selectin and GPIIb/IIIa conformations. For platelet aggregation, the IC50 of MTCA-KKV was approximately 1/10 folds of MTCA. CONCLUSION KKV modification led to forming MTCA-KKV that is superior to MTCA in terms of accumulating on activated platelets, targeting P-selectin and GPIIb/IIIa and inhibiting platelet aggregation. MTCA-KKV could be a promising lead for further investigation.
Collapse
|
25
|
Chen H, Lu A, Zhang X, Gui L, Wang Y, Wu J, Feng H, Peng S, Zhao M. Design and development of ICCA as a dual inhibitor of GPIIb/IIIa and P-selectin receptors. Drug Des Devel Ther 2018; 12:2097-2110. [PMID: 30022809 PMCID: PMC6042529 DOI: 10.2147/dddt.s169238] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
BACKGROUND The impact of upregulation of platelet membrane glycoprotein (GP)IIb/IIIa and P-selectin on the onset of arterial thrombosis, venous thrombosis, and cancer encourages to hypothesize that dual inhibitor of GPIIb/IIIa and P-selectin receptors should simultaneously inhibit arterial thrombosis, block venous thrombosis, and slow tumor growth. METHODS For this reason, the structural characteristics and the CDOCKER interaction energies of 12 carbolines were analyzed. This led to the design of 1-(4-isopropyl-phenyl)-β-carboline-3-carboxylic acid (ICCA) as a promising inhibitor of GPIIb/IIIa and P-selectin receptors. RESULTS The synthetic route provided ICCA in 48% total yield and 99.6% high-performance liquid chromatography purity. In vivo 5 μmol/kg oral ICCA downregulated GPIIb/IIIa and P-selectin expression thereby inhibited arterial thrombosis, blocked venous thrombosis, and slowed down tumor growth, but did not damage the kidney and the liver. CONCLUSION Therefore, ICCA could be a promising candidate capable of downregulating GPIIb/IIIa and P-selectin receptors, inhibiting arterial thrombosis, blocking venous thrombosis, and slowing down tumor growth.
Collapse
MESH Headings
- Animals
- Antibiotics, Antineoplastic/chemical synthesis
- Antibiotics, Antineoplastic/chemistry
- Antibiotics, Antineoplastic/pharmacology
- Carbolines/chemical synthesis
- Carbolines/chemistry
- Carbolines/pharmacology
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Dose-Response Relationship, Drug
- Down-Regulation/drug effects
- Doxorubicin/chemical synthesis
- Doxorubicin/chemistry
- Doxorubicin/pharmacology
- Drug Design
- Drug Screening Assays, Antitumor
- Humans
- Male
- Mice
- Mice, Inbred ICR
- Models, Molecular
- Molecular Structure
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/metabolism
- Neoplasms, Experimental/pathology
- P-Selectin/antagonists & inhibitors
- P-Selectin/metabolism
- Platelet Aggregation/drug effects
- Platelet Aggregation Inhibitors/chemical synthesis
- Platelet Aggregation Inhibitors/chemistry
- Platelet Aggregation Inhibitors/pharmacology
- Platelet Glycoprotein GPIIb-IIIa Complex/antagonists & inhibitors
- Platelet Glycoprotein GPIIb-IIIa Complex/metabolism
- Rats
- Rats, Sprague-Dawley
- Structure-Activity Relationship
Collapse
Affiliation(s)
- Haiyan Chen
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - An Lu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Lin Gui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Hua Feng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China, ;
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan, ;
| |
Collapse
|
26
|
Griffin MT, Zhu Y, Liu Z, Aidun CK, Ku DN. Inhibition of high shear arterial thrombosis by charged nanoparticles. BIOMICROFLUIDICS 2018; 12:042210. [PMID: 29887934 PMCID: PMC5973895 DOI: 10.1063/1.5025349] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Accepted: 05/07/2018] [Indexed: 05/02/2023]
Abstract
Platelet accumulation under high shear rates at the site of atherosclerotic plaque rupture leads to myocardial infarction and stroke. Current antiplatelet therapies remain ineffective within a large percentage of the population, while presenting significant risks for bleeding. We explore a novel way to inhibit arterial thrombus formation by biophysical means without the use of platelet inactivating drugs. Our computational multi-scale dynamics model has predicted that charged particles of a specific size may entangle von Willebrand Factor (vWF) polymers and reduce the amount of elongation at high shear rates. We tested this hypothesis experimentally for negatively charged nanoparticles (CNP) to inhibit arterial thrombus formation. CNP of a particular size and charge inhibited thrombus formation, with a 10-fold peak inhibition over control conditions of thrombotic occlusion. Particles of differing material composition, size, and charge had little effect as predicted by computational studies. Surprisingly, the dose response curve was not sigmoidal, but exhibited a peak at 1.5 CNP:vWF proteins, which was not predicted by the model. This study describes a new antithrombotic agent that may have a different mechanism of action than current pharmaceutical therapies.
Collapse
Affiliation(s)
| | - Yuanzheng Zhu
- G.W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | - Zixiang Liu
- G.W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, Georgia 30332, USA
| | | | - David N. Ku
- Author to whom correspondence should be addressed:
| |
Collapse
|
27
|
Wu J, Zhu H, Yang G, He J, Wang Y, Zhao S, Zhang X, Gui L, Zhao M, Peng S. Design and synthesis of nanoscaled IQCA-TAVV as a delivery system capable of antiplatelet activation, targeting arterial thrombus and releasing IQCA. Int J Nanomedicine 2018; 13:1139-1158. [PMID: 29520141 PMCID: PMC5833776 DOI: 10.2147/ijn.s150205] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
BACKGROUND Arterial thrombosis has been associated with a series of pathological conditions, and the discovery of arterial thrombosis inhibitor is of clinical importance. METHODS By analyzing the pharmacophores of anti-platelet agents, thrombus targeting peptide and anti-thrombotic nano-systems 3S-1,2,3,4-tetrahydroisoquino-line-3-carbonyl-Thr-Ala-Arg-Gly-Asp(Val)-Val (IQCA-TAVV) was designed and prepared as a nano-scaled arterial thrombosis inhibitor. RESULTS In vitro the nanoparticles of IQCA-TAVV were able to adhere onto the surface of activated platelets, attenuate activated platelets to extend pseudopodia and inhibit activated platelets to form aggregators. In vivo IQCA-TAVV targeted arterial thrombus, dose dependently inhibited arterial thrombosis with a 1 nmol/kg of minimal effective dose, and the activity waŝ1670 folds of that of aspirin. CONCLUSION IQCA-TAVV represented the design, preparation and application of nanomedicine capable of adhering on the surface of activated platelets, attenuating platelet activation, targeting arterial thrombus and inhibiting arterial thrombosis.
Collapse
Affiliation(s)
- Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Guodong Yang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
| | - Jianhong He
- Department of Internal Medicine of TCM, The First Affiliated Hospital of Guanxi University of Chinese Medicine, Nanning, China
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Lin Gui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing, China
- Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, China
| |
Collapse
|
28
|
Wu J, Zhu H, Yang G, Wang Y, Wang Y, Zhao S, Zhao M, Peng S. IQCA-TAVV: To explore the effect of P-selectin, GPIIb/IIIa, IL-2, IL-6 and IL-8 on deep venous thrombosis. Oncotarget 2017; 8:91391-91401. [PMID: 29207652 PMCID: PMC5710932 DOI: 10.18632/oncotarget.20588] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2017] [Accepted: 08/04/2017] [Indexed: 11/25/2022] Open
Abstract
Deep vein thrombosis (DVT) associates with considerable morbidity, functional disability and mortality. Due to the lack of suitable inhibitor the correlation of various factors in DVT onset remains unknown. In this context we analyzed the structure of anti-platelet aggregation agent, P-selectin down-regulator, GPIIb/IIIa down-regulator and anti-inflammatory agent, thereby designed N-(3S-1,2,3,4-tetrahydroisoquinoline-3-carbonyl)- Thr-Ala-Arg-Gly-Asp(Val)-Val (IQCA-TAVV) as an inhibitor of DVT to receive evaluations. The docking predicted that IQCA-TAVV can target P-selectin and GPIIb/IIIa. The UV showed that IQCA-TAVV can act on P-selectin and GPIIb/IIIa. ELISA indicated that IQCA-TAVV concentration dependently inhibited activated platelets to express P-selectin and GPIIb/IIIa, and the minimal effective concentration was 1 nM. IC50 of IQCA-TAVV against platelet aggregation induced by arachidonic acid, adenosine diphosphate and platelet activating factor fell within a range of 0.13 nM to 0.30 nM. In vivo IQCA-TAVV dose-dependently inhibited venous thrombosis and the minimal effective dose was 1 nmol/kg. On ear edema model the anti-inflammation activity of 10 nmol/kg IQCA-TAVV equaled that of 1.1mmol/kg aspirin. The concentration of IL-2, IL-6 and IL-8 in the serum of the ear edema mice were also significantly decreased by 10 nmol/kg IQCA-TAVV. Even at 1 μmol/kg of dose IQCA-TAVV still did not injure the kidney, the liver, and the nerves of healthy mice. Thereby IQCA-TAVV depicts a relationship of three levels (inhibiting platelet activation, targeting externalized membrane receptor, decreasing serum inflammatory factor) for the down-regulation of P-selectin, GPIIb/IIIa, IL-2, IL-6 and IL-8 in DVT.
Collapse
Affiliation(s)
- Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences of Capital Medical University, Beijing, PR China
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences of Capital Medical University, Beijing, PR China
| | - Guodong Yang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences of Capital Medical University, Beijing, PR China
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences of Capital Medical University, Beijing, PR China
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences of Capital Medical University, Beijing, PR China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences of Capital Medical University, Beijing, PR China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences of Capital Medical University, Beijing, PR China
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences of Capital Medical University, Beijing, PR China
| |
Collapse
|
29
|
Xu X, Wang Y, Wu J, Hu X, Zhu H, Zhang X, Wang Y, Gui L, Zhao M, Peng S. ATIQCTPC: a nanomedicine capable of targeting tumor and blocking thrombosis in vivo. Int J Nanomedicine 2017; 12:4415-4431. [PMID: 28652742 PMCID: PMC5476604 DOI: 10.2147/ijn.s129989] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
To overcome the harmful side effects, low tolerance, and undesirable outcomes of the anticancer drugs, we used ethane-1,2-diamine to bridge antitumoral (S)-3-acetyl-4-oxo-tetrahydroindolo[2,3-a]quinolizine-6-carboxylic acid (ATIQC) and tumor-targeting d-glucuronic acid, thereby providing (6S)-3-acetyl-4-oxo-N-(2-(3,4,5,6-tetrahydroxytetrahydro-2H-pyran-2-carboxamido)ethyl)-4,6,7,12-tetrahydroindolo[2,3-a]quinolizine-6-carboxamide (ATIQCTPC). Atomic force microscopy images visualized, that in serum, ATIQCTPC formed particles of height <81 nm. These particles effectively avoided phagocytosis of macrophages and were stable in blood circulation. Distribution analysis indicated that ATIQCTPC accumulated and released ATIQC in the tumor tissue through a targeting manner. Thus, the antitumor and the anti-thrombotic activities of ATIQCTPC were 100-fold higher than those of ATIQC, and ATIQCTPC was able to prevent cancer patients from suffering from thrombosis. Based on the observation that ATIQCTPC decreased serum tumor necrosis factor-α (TNF-α) and interleukin-8 (IL-8) in S180 mice, we hypothesized that this is the mechanism that ATIQCTPC utilized to slow tumor growth. Additionally, we observed that ATIQCTPC inhibited thrombosis by decreasing serum P-selectin of thrombotic rats. The intermolecular association and the hexamerization manner of ATIQCTPC were experimentally evidenced and correlated with the formation of the nanoparticles.
Collapse
Affiliation(s)
- Xinyi Xu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Xi Hu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Lin Gui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
30
|
Wu J, Zhao M, Wang Y, Wang Y, Zhu H, Zhao S, Gui L, Zhang X, Peng S. N-(3-hydroxymethyl-β-carboline-1-yl-ethyl- 2-yl)-l-Phe: development toward a nanoscaled antitumor drug capable of treating complicated thrombosis and inflammation. DRUG DESIGN DEVELOPMENT AND THERAPY 2017; 11:225-239. [PMID: 28176928 PMCID: PMC5265142 DOI: 10.2147/dddt.s123919] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
It is well documented that the surfaces of cancer cells, activated platelets and inflammatory cells are rich in P-selectin. N-(3-hydroxymethyl-β-carboline-1-yl-ethyl-2-yl)-l-Phe (HMCEF) is a P-selectin inhibitor capable of simultaneously inhibiting thrombosis and inflammation. Based on the knowledge that P-selectin is a common target for antithrombotic, anti-inflammatory and antitumor drugs, the aim of this study article was to estimate the possibility of HMCEF as a nanoscaled antitumor drug. Images of transmission electron micro scopy, scanning electron microscopy and atomic force microscopy proved that HMCEF forms nanoparticles with a diameter of <120 nm that promote delivery in blood circulation. In vitro HMCEF intercalates into calf thymus DNA, cuts off DNA pBR22 and inhibits the proliferation of cancer cells. In vivo HMCEF dose dependently (0.2, 2 and 200 nmol/kg per day) slows tumor growth in treated S180 mice, and has a minimal effective dose of 2 nmol/kg per day. At 200 nmol/kg per day, HMCEF does not affect the liver and the kidney of the treated S180 mice, and at 20,000 nmol/kg HMCEF does not affect the liver and the kidney of the treated healthy ICR mice. HMCEF is a promising antitumor drug, which is characterized by its high safety and efficacy in the prevention of the complications of thrombosis and inflammation in patients.
Collapse
Affiliation(s)
- Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan, Republic of China
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Lin Gui
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs; Engineering Research Center of Endogenous Prophylactic, Ministry of Education of China; Beijing Laboratory of Biomedical Materials; College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
31
|
Li Y, Li J, Liu T, Wang Y, Zhou Z, Cheng F, Feng C, Cheng X, Liu H, Chen X. Preparation and antithrombotic activity identification of Perinereis aibuhitensis extract: a high temperature and wide pH range stable biological agent. Food Funct 2017; 8:3533-3541. [DOI: 10.1039/c7fo00987a] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
In this work, a thermal and wide pH range stable biological agent was extracted from Perinereis aibuhitensis, whose antithrombotic activity was investigated.
Collapse
Affiliation(s)
- Yang Li
- The Research Center for Processing and Quality Control of Aquaculture Production
- Marine Biology Institute of Shandong Province
- Qingdao
- P.R. China
- College of Marine Life Science
| | - Jing Li
- College of Marine Life Science
- Ocean University of China
- Qingdao
- P.R. China
| | - Tianhong Liu
- The Research Center for Processing and Quality Control of Aquaculture Production
- Marine Biology Institute of Shandong Province
- Qingdao
- P.R. China
| | - Ying Wang
- The Research Center for Processing and Quality Control of Aquaculture Production
- Marine Biology Institute of Shandong Province
- Qingdao
- P.R. China
| | - Zhongzheng Zhou
- College of Marine Life Science
- Ocean University of China
- Qingdao
- P.R. China
| | - Feng Cheng
- Center Blood Station of Qingdao
- Qingdao
- P.R. China
| | - Chao Feng
- College of Marine Life Science
- Ocean University of China
- Qingdao
- P.R. China
| | - Xiaojie Cheng
- College of Marine Life Science
- Ocean University of China
- Qingdao
- P.R. China
| | - Hongjun Liu
- The Research Center for Processing and Quality Control of Aquaculture Production
- Marine Biology Institute of Shandong Province
- Qingdao
- P.R. China
| | - Xiguang Chen
- College of Marine Life Science
- Ocean University of China
- Qingdao
- P.R. China
| |
Collapse
|
32
|
Bi Y, Zhang Y, Cui C, Ren L, Jiang X. Gene-silencing effects of anti-survivin siRNA delivered by RGDV-functionalized nanodiamond carrier in the breast carcinoma cell line MCF-7. Int J Nanomedicine 2016; 11:5771-5787. [PMID: 27853365 PMCID: PMC5104303 DOI: 10.2147/ijn.s117611] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Nanodiamond (ND) is a renowned material in nonviral small interfering RNA (siRNA) carrier field due to its unique physical, chemical, and biological properties. In our previous work, it was proven that ND could deliver siRNA into cells efficiently and downregulate the expression of desired protein. However, synthesizing a high-efficient tumor-targeting carrier using ND is still a challenge. In this study, a novel carrier, NDCONH(CH2)2NH-VDGR, was synthesized for siRNA delivery, and its properties were characterized with methods including Fourier transform infrared spectrometry, transmission electron microscopy, scanning electron microscopy, gel retardation assay, differential scanning calorimetry, confocal microscopy, releasing test, real-time polymerase chain reaction (PCR) assay, enzyme-linked immunosorbent assay (ELISA), flow cytometry, cytotoxicity assay, and gene-silencing efficacy assay in vitro and in vivo. The mechanism of NDCONH(CH2)2NH-VDGR/survivin-siRNA-induced tumor apoptosis was evaluated via flow cytometer assay using Annexin V–fluorescein isothiocyanate/propidium iodide staining method. The NDCONH(CH2)2NH-VDGR/survivin-siRNA nanoparticle with 60–110 nm diameter and 35.65±3.90 mV zeta potential was prepared. For real-time PCR assay, the results showed that the expression of survivin mRNA was reduced to 46.77%±6.3%. The expression of survivin protein was downregulated to 48.49%±2.25%, as evaluated by ELISA assay. MTT assay showed that NDCONH(CH2)2NH-VDGR/survivin-siRNA had an inhibitory effect on MCF-7 cell proliferation. According to these results, the survivin-siRNA could be delivered, transported, and released stably, which benefits in increasing the gene-silencing effect. Therefore, as an siRNA carrier, NDCONH(CH2)2NH-VDGR was suggested to be used in siRNA delivery system and in cancer treatments.
Collapse
Affiliation(s)
- Yanzhao Bi
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yifan Zhang
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Chunying Cui
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Lulu Ren
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Xueyun Jiang
- School of Chemical Biology and Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
33
|
Jiang X, Zhao M, Wang Y, Zhu H, Zhao S, Wu J, Song Y, Peng S. RGD(F/S/V)-Dex: towards the development of novel, effective, and safe glucocorticoids. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:1059-76. [PMID: 27022245 PMCID: PMC4789840 DOI: 10.2147/dddt.s99568] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dexamethasone (Dex) is an effective glucocorticoid in treating inflammation and preventing rejection reaction. However, the side effects limit its clinical application. To improve its druggable profile, the conjugates of RGD-peptide-modified Dex were presented and their enhanced anti-inflammation activity, minimized osteoporotic action, and nanoscaled assembly were explored. (RGD stands for Arg-Gly-Asp. Standard single letter biochemical abbreviations for amino acids have been used throughout this paper.) In respect of the rejection reaction, the survival time of the implanted myocardium of the mice treated with 1.43 µmol/kg/d of the conjugates for 15 consecutive days was significantly longer than that of the mice treated with 2.5 µmol/kg/d of Dex, and the conjugates, but not Dex, exhibited no toxic action. At a single dose of 14.3 µmol/kg (100 times minimal effective dose, 0.143 µmol/kg), the conjugates induced no liver, kidney, or systemic toxicity. At the dose of 1.43 µmol/kg, the conjugates, but not Dex, prolonged the bleeding time of the mice, and inhibited the thrombosis of the rats. In water and rat plasma, the conjugates formed nanoparticles of 14-250 and 101-166 nm in diameter, respectively. Since the nanoparticles of ~100 nm in size cannot be entrapped by macrophages in the circulation, RGDF-Dex would particularly be worthy of development, since its nanoparticle diameter is 101 nm.
Collapse
Affiliation(s)
- Xueyun Jiang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China; Faculty of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| | - Yuanbo Song
- Guangxi Pusen Biotechnology Co. Ltd., Nanning, Guangxi, People's Republic of China
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
34
|
Wang Y, Tang J, Zhu H, Jiang X, Liu J, Xu W, Ma H, Feng Q, Wu J, Zhao M, Peng S. Aqueous extract of Rabdosia rubescens leaves: forming nanoparticles, targeting P-selectin, and inhibiting thrombosis. Int J Nanomedicine 2015; 10:6905-18. [PMID: 26604756 PMCID: PMC4639563 DOI: 10.2147/ijn.s91316] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The hot water extract of Rabdosia rubescens was traditionally used as an antithrombotic medicine. To explore its antithrombotic utility and mechanism, we carried out a series of in vitro and in vivo assays in this study. In vitro platelet aggregation assay showed that the half maximal inhibitory concentration values of aqueous extract of R. rubescens leaves (AERL) inhibiting platelet aggregation induced by thrombin, arachidonic acid, adenosine diphosphate, and platelet-activating factor ranged from 0.12 mg/mL to 1.43 mg/mL. The minimal effective oral dose of AERL inhibiting the rats from forming thrombus was 25 mg/kg. Both in vitro and in vivo actions were correlated with AERL concentration-dependently inhibiting sP-selectin release. In water, AERL formed nanoparticles, and their size depended on the concentration. Docking the five nucleotides, 21 phenolic acids, and four diterpenoids identified by high-performance liquid chromatography-photodiode array detector/(-)electrospray ionization-tandem mass spectrometry analysis into the active site of P-selectin, rosmarinic acid was predicted to be the antithrombotic ingredient of AERL. In flow cytometry analysis, 1 μM of rosmarinic acid effectively inhibited sP-selectin release in arachidonic acid-activated platelets. In a rat model, 5 mg/kg of oral rosmarinic acid effectively inhibited thrombosis.
Collapse
Affiliation(s)
- Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Jingcheng Tang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Xueyun Jiang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Jiawang Liu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Wenyun Xu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Haiping Ma
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Qiqi Feng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
- Faculty of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, Beijing Laboratory of Biomedical Materials, College of Pharmaceutical Sciences, Capital Medical University, Beijing, People’s Republic of China
| |
Collapse
|
35
|
Cicha I. Thrombosis: Novel nanomedical concepts of diagnosis and treatment. World J Cardiol 2015; 7:434-441. [PMID: 26322182 PMCID: PMC4549776 DOI: 10.4330/wjc.v7.i8.434] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Revised: 03/23/2015] [Accepted: 05/18/2015] [Indexed: 02/06/2023] Open
Abstract
Intravascular thrombosis, a critical pathophysiological feature of many cardiovascular disorders, leads to the formation of life-threatening obstructive blood clots within the vessels. Rapid recanalization of occluded vessels is essential for the patients’ outcome, but the currently available systemic fibrinolytic therapy is associated with low efficacy and tremendous side effects. Additionally, many patients are ineligible for systemic thrombolytic therapy, either due to delayed admission to the hospital after symptom onset, or because of recent surgery, or bleeding. In order to improve the treatment efficacy and to limit the risk of hemorrhagic complications, both precise imaging of the affected vascular regions, and the localized application of fibrinolytic agents, are required. Recent years have brought about considerable advances in nanomedical approaches to thrombosis. Although these thrombus-targeting imaging agents and nanotherapies are not yet implemented in humans, substantial amount of successful in vivo applications have been reported, including animal models of stroke, acute arterial thrombosis, and pulmonary embolism. It is evident that the future progress in diagnosis and treatment of thrombosis will be closely bound with the development of novel nanotechnology-based strategies. This Editorial focuses on the recently reported approaches, which hold a great promise for personalized, disease-targeted treatment and reduced side effects in the patients suffering from this life-threatening condition.
Collapse
|
36
|
Li S, Wang Y, Wang F, Wang Y, Zhang X, Zhao M, Feng Q, Wu J, Zhao S, Wu W, Peng S. Small molecule PZL318: forming fluorescent nanoparticles capable of tracing their interactions with cancer cells and activated platelets, slowing tumor growth and inhibiting thrombosis. Int J Nanomedicine 2015; 10:5273-92. [PMID: 26345234 PMCID: PMC4554418 DOI: 10.2147/ijn.s88052] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Low selectivity of chemotherapy correlates with poor outcomes of cancer patients. To improve this issue, a novel agent, N-(1-[3-methoxycarbonyl-4-hydroxyphenyl]-β-carboline-3-carbonyl)-Trp-Lys-OBzl (PZL318), was reported here. The transmission electron microscopy, scanning electron microscopy, and atomic force microscopy images demonstrated that PZL318 can form nanoparticles. Fluorescent and confocal images visualized that PZL318 formed fluorescent nanoparticles capable of targeting cancer cells and tracing their interactions with cancer cells. In vitro, 40 μM of PZL318 inhibited the proliferation of tumorigenic cells, but not nontumorigenic cells. In vivo, 10 nmol/kg of PZL318 slowed the tumor growth of S180 mice and alleviated the thrombosis of ferric chloride-treated ICR mice, while 100 μmol/kg of PZL318 did not injure healthy mice and they exhibited no liver toxicity. By analyzing Fourier transform–mass spectrometry and rotating-frame Overhauser spectroscopy (ROESY) two-dimensional nuclear magnetic resonance spectra, the chemical mechanism of PZL318-forming trimers and nanoparticles was explored. By using mesoscale simulation, a nanoparticle of 3.01 nm in diameter was predicted containing 13 trimers. Scavenging free radicals, downregulating sP-selectin expression and intercalating toward DNA were correlated with the antitumor mechanism of PZL318.
Collapse
Affiliation(s)
- Shan Li
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| | - Feng Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| | - Yaonan Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| | - Xiaoyi Zhang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China ; Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Qiqi Feng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| | - Shurui Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| | - Wei Wu
- College of Basic Medicine of Capital Medical University, Beijing, People's Republic of China
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs, Beijing Laboratory of Biomedical Materials, Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China, College of Pharmaceutical Sciences of Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
37
|
Wang Z, Wang W, Bu X, Wei Z, Geng L, Wu Y, Dong C, Li L, Zhang D, Yang S, Wang F, Lausted C, Hood L, Hu Z. Microarray based screening of peptide nano probes for HER2 positive tumor. Anal Chem 2015. [PMID: 26218790 DOI: 10.1021/acs.analchem.5b01588] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Peptides are excellent biointerface molecules and diagnostic probes with many advantages such as good penetration, short turnover time, and low cost. We report here an efficient peptide screening strategy based on in situ single bead sequencing on a microarray. Two novel peptides YLFFVFER (H6) and KLRLEWNR (H10) specifically binding to the tumor biomarker human epidermal growth factor receptor 2 (HER2) with aKD of 10(-8) M were obtained from a 10(5) library. Conjugated to nanoparticles, both the H6 and H10 probes showed specific accumulation in HER2-positive tumor tissues in xenografted mice by in vivo imaging.
Collapse
Affiliation(s)
| | | | | | | | | | - Yue Wu
- ‡Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing China, 100191
| | - Chengyan Dong
- ‡Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing China, 100191
| | - Liqiang Li
- ‡Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing China, 100191
| | | | | | - Fan Wang
- ‡Medical Isotopes Research Center, Department of Radiation Medicine, School of Basic Medical Sciences, Peking University, Beijing China, 100191
| | - Christopher Lausted
- §Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109, United States
| | - Leroy Hood
- §Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109, United States
| | - Zhiyuan Hu
- §Institute for Systems Biology, 401 Terry Avenue North, Seattle, Washington 98109, United States.,∥Beijing Proteome Research Center, Beijing Institute of Radiation Medicine, Beijing China, 102206
| |
Collapse
|
38
|
Chen S, Wang Y, Li S, Wang Y, Zhao M, Zhu H, Wu J, Peng S. Poly-α,β-aspartyl-Arg-Gly-Asp-Phe: a novel polymeric nanomedicine. MEDCHEMCOMM 2015. [DOI: 10.1039/c4md00331d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
High anti-thrombotic efficacy, action target and nano-structure of a novel nanomedicine were described.
Collapse
Affiliation(s)
- Shuangling Chen
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Beijing Laboratory of Biomedical Materials
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- College of Pharmaceutical Sciences of Capital Medical University
- Beijing 100069
| | - Yuji Wang
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Beijing Laboratory of Biomedical Materials
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- College of Pharmaceutical Sciences of Capital Medical University
- Beijing 100069
| | - Shan Li
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Beijing Laboratory of Biomedical Materials
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- College of Pharmaceutical Sciences of Capital Medical University
- Beijing 100069
| | - Yaonan Wang
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- Medical Experiment and Test Center of Capital Medical University
- Beijing 100069
- P. R. China
| | - Ming Zhao
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Beijing Laboratory of Biomedical Materials
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- College of Pharmaceutical Sciences of Capital Medical University
- Beijing 100069
| | - Haimei Zhu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Beijing Laboratory of Biomedical Materials
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- College of Pharmaceutical Sciences of Capital Medical University
- Beijing 100069
| | - Jianhui Wu
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Beijing Laboratory of Biomedical Materials
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- College of Pharmaceutical Sciences of Capital Medical University
- Beijing 100069
| | - Shiqi Peng
- Beijing Area Major Laboratory of Peptide and Small Molecular Drugs
- Beijing Laboratory of Biomedical Materials
- Engineering Research Center of Endogenous Prophylactic of Ministry of Education of China
- College of Pharmaceutical Sciences of Capital Medical University
- Beijing 100069
| |
Collapse
|
39
|
Du F, Zhang X, Li S, Wang Y, Zheng M, Wang Y, Zhao S, Wu J, Gui L, Zhao M, Peng S. Mechanism of forming trimer, self-assembling nano-particle and inhibiting tumor growth of small molecule CIPPCT. MEDCHEMCOMM 2014. [DOI: 10.1039/c4md00158c] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The mechanism whereby CIPPCT forms nanoparticles capable of delivery in circulation and adhering on cancer cells is presented.
Collapse
Affiliation(s)
- Fengxiang Du
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Xiaoyi Zhang
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Shan Li
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Yaonan Wang
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Meiqing Zheng
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Yuji Wang
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Shurui Zhao
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Jianhui Wu
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Lin Gui
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| | - Ming Zhao
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
- Department of Biomedical Science and Environmental Biology
- Kaohsiung Medical University
| | - Shiqi Peng
- College of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069, P.R. China
| |
Collapse
|