1
|
Lu C, Li C, Gu N, Yang F. Emerging Elastic Micro-Nano Materials for Diagnosis and Treatment of Thrombosis. RESEARCH (WASHINGTON, D.C.) 2025; 8:0614. [PMID: 40028043 PMCID: PMC11868703 DOI: 10.34133/research.0614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/14/2025] [Accepted: 01/24/2025] [Indexed: 03/05/2025]
Abstract
Thrombus is a blood clot that forms in a blood vessel at the point of flaking. Thrombosis is closely associated with cardiovascular diseases caused by different sources and factors. However, the current clinical methods of thrombus diagnosis and treatment still have problems with targeting, permeability, stability, and biosafety. Therefore, in recent years, based on the development of micro/nano technology, researchers have tried to develop some new strategies for the diagnosis and treatment of thrombosis. Due to the unique structural characteristics, the micro-nano materials in physiological environments show excellent transport and delivery properties such as better in vivo circulation, longer life span, better targeting ability, and controllable cellular internalization. Especially, elasticity and stiffness are inherent mechanical properties of some well-designed micro-nano materials, which can make them better adapted to the needs of thrombosis diagnosis and treatment. Herein, this review first introduces the thrombotic microenvironment to characterize the thrombus development process. Then, to fine-tune the pathological occurrence and development of thrombosis, the role of elastic micro-nano materials for thrombus diagnosis and treatment is summarized. The properties, preparation methods, and biological fate of these materials have been discussed in detail. Following, the applications of elastic micro-nano materials in biomedical imaging, drug delivery, and therapy of thrombosis are highlighted. Last, the shortcomings and future design strategies of elastic micro-nano materials in diagnosis and treatment of clinical thrombosis are discussed. This review will provide new ideas for the use of nanotechnology in clinical diagnosis and treatment of thrombus in the future.
Collapse
Affiliation(s)
- Chenxin Lu
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, P. R. China
| | - Chunjian Li
- Department of Cardiology,
The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, P. R. China
| | - Ning Gu
- Nanjing Key Laboratory for Cardiovascular Information and Health Engineering Medicine, Institute of Clinical Medicine, Nanjing Drum Tower Hospital, Medical School,
Nanjing University, Nanjing 210093, P. R. China
| | - Fang Yang
- State Key Laboratory of Digital Medical Engineering, Jiangsu Laboratory for Biomaterials and Devices, School of Biological Science and Medical Engineering,
Southeast University, Nanjing 210096, P. R. China
| |
Collapse
|
2
|
Peng B, Mohammed FS, Tang X, Liu J, Sheth KN, Zhou J. Nanotechnology approaches to drug delivery for the treatment of ischemic stroke. Bioact Mater 2025; 43:145-161. [PMID: 39386225 PMCID: PMC11462157 DOI: 10.1016/j.bioactmat.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/11/2024] [Accepted: 09/12/2024] [Indexed: 10/12/2024] Open
Abstract
Ischemic stroke is a major global public health concern that lacks effective treatment options. A significant challenge lies in delivering therapeutic agents to the brain due to the restrictive nature of the blood-brain barrier (BBB). The BBB's selectivity hampers the delivery of therapeutically relevant quantities of agents to the brain, resulting in a lack of FDA-approved pharmacotherapies for stroke. In this article, we review therapeutic agents that have been evaluated in clinical trials or are currently undergoing clinical trials. Subsequently, we survey strategies for synthesizing and engineering nanoparticles (NPs) for drug delivery to the ischemic brain. We then provide insights into the potential clinical translation of nanomedicine, offering a perspective on its transformative role in advancing stroke treatment strategies. In summary, existing literature suggests that drug delivery represents a major barrier for clinical translation of stroke pharmacotherapies. While nanotechnology has shown significant promise in addressing this challenge, further advancements aimed at improving delivery efficiency and simplifying formulations are necessary for successful clinical translation.
Collapse
Affiliation(s)
- Bin Peng
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurology, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, PR China
| | - Farrah S. Mohammed
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, New Haven, CT, 06510, USA
| | - Xiangjun Tang
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurosurgery, Taihe Hospital, Hubei, 442000, PR China
| | - Jia Liu
- Department of Neurosurgery, New Haven, CT, 06510, USA
| | - Kevin N. Sheth
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Neurology, Yale University, New Haven, CT, 06510, USA
| | - Jiangbing Zhou
- Department of Neurosurgery, New Haven, CT, 06510, USA
- Department of Biomedical Engineering, New Haven, CT, 06510, USA
| |
Collapse
|
3
|
Song M, Zeng Q, Ding X. Hypoxia-Responsive Self-Assembling Nanoparticles Based on an Amphiphilic Copolymer for Targeted Delivery of Tissue Plasminogen Activator in Acute Mesenteric Ischemia Therapy. ACS APPLIED NANO MATERIALS 2024; 7:27400-27407. [DOI: 10.1021/acsanm.4c05408] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2025]
Affiliation(s)
- Mingze Song
- Nanchang University Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, P. R. China
| | - Qiongrong Zeng
- Department of Gastroenterology, Guangxi Hospital Division of The First Affiliated Hospital, Sun Yat-sen University, Nanning 530028, P. R. China
| | - Xingwei Ding
- Nanchang University Queen Mary School, Jiangxi Medical College, Nanchang University, Nanchang 330031, P. R. China
- The National Engineering Research Center for Bioengineering Drugs and the Technologies, Institute of Translational Medicine, Jiangxi Medical College, Nanchang University, Nanchang 330031, P. R. China
| |
Collapse
|
4
|
He A, Huang Y, Cao C, Li X. Advances in drug delivery systems utilizing blood cells and their membrane-derived microvesicles. Drug Deliv 2024; 31:2425156. [PMID: 39520082 PMCID: PMC11552282 DOI: 10.1080/10717544.2024.2425156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 10/11/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
The advancement of drug delivery systems (DDSs) in recent decades has demonstrated significant potential in enhancing the efficacy of pharmacological agents. Despite the approval of certain DDSs for clinical use, challenges such as rapid clearance from circulation, toxic accumulation in the body, and ineffective targeted delivery persist as obstacles to successful clinical application. Blood cell-based DDSs have emerged as a popular strategy for drug administration, offering enhanced biocompatibility, stability, and prolonged circulation. These DDSs are well-suited for systemic drug delivery and have played a crucial role in formulating optimal drug combinations for treating a variety of diseases in both preclinical studies and clinical trials. This review focuses on recent advancements and applications of DDSs utilizing blood cells and their membrane-derived microvesicles. It addresses the current therapeutic applications of blood cell-based DDSs at the organ and tissue levels, highlighting their successful deployment at the cellular level. Furthermore, it explores the mechanisms of cellular uptake of drug delivery vectors at the subcellular level. Additionally, the review discusses the opportunities and challenges associated with these DDSs.
Collapse
Affiliation(s)
- Andong He
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital, Ningbo University School of Medicine, Ningbo, China
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| | - Yuye Huang
- Center for Medical and Engineering Innovation, Central Laboratory, The First Affiliated Hospital, Ningbo University School of Medicine, Ningbo, China
| | - Chao Cao
- Department of Respiratory and Critical Care Medicine, Key Laboratory of Respiratory Disease of Ningbo, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Xuejin Li
- Department of Engineering Mechanics, Zhejiang University, Hangzhou, China
| |
Collapse
|
5
|
Liu CH, Rethi L, Weng PW, Trung Nguyen H, Chuang AEY. Cutting-edge advances in nano/biomedicine: A review on transforming thrombolytic therapy. Biochem Pharmacol 2024; 229:116523. [PMID: 39251141 DOI: 10.1016/j.bcp.2024.116523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 09/03/2024] [Accepted: 09/05/2024] [Indexed: 09/11/2024]
Abstract
Thrombotic blockages within blood vessels give rise to critical cardiovascular disorders, including ischemic stroke, venous thromboembolism, and myocardial infarction. The current approach to the therapy of thrombolysis involves administering Plasminogen Activators (PA), but it is hindered by fast drug elimination, narrow treatment window, and the potential for bleeding complications. Leveraging nanomedicine to encapsulate and deliver PA offers a solution by improving the efficacy of therapy, safeguarding the medicine from proteinase biodegradation, and reducing unwanted effects in in vivo trials. In this review, we delve into the underlying venous as well as arterial thrombus pathophysiology and provide an overview of clinically approved PA used to address acute thrombotic conditions. We explore the existing challenges and potential directions within recent pivotal research on a variety of targeted nanocarriers, such as lipid, polymeric, inorganic, and biological carriers, designed for precise delivery of PA to specific sites. We also discuss the promising role of microbubbles and ultrasound-assisted Sono thrombolysis, which have exhibited enhanced thrombolysis in clinical studies. Furthermore, our review delves into approaches for the strategic development of nano-based carriers tailored for targeting thrombolytic action and efficient encapsulation of PA, considering the intricate interaction in biology systems as well as nanomaterials. In conclusion, the field of nanomedicine offers a valuable method for the exact and effective therapy of severe thrombus conditions, presenting a pathway toward improved patient outcomes and reduced complications.
Collapse
Affiliation(s)
- Chia-Hung Liu
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan; TMU Research Center of Urology and Kidney, Taipei Medical University, 250 Wu-Hsing Street, Taipei 11031, Taiwan; Department of Urology, Shuang Ho Hospital, Taipei Medical University, 291 Zhongzheng Road, Zhonghe District, New Taipei City 23561, Taiwan
| | - Lekshmi Rethi
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Pei-Wei Weng
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City 23561, Taiwan; Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Hieu Trung Nguyen
- Department of Orthopedics and Trauma, Faculty of Medicine, University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, 700000, Viet Nam
| | - Andrew E-Y Chuang
- International Ph.D. Program in Biomedical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan; Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei 11696, Taiwan.
| |
Collapse
|
6
|
Shan L, Wang J, Tu H, Zhang W, Li H, Slezak P, Lu F, Lee D, Hu E, Geng Z, Lan G, Xie R. Drug delivery under cover of erythrocytes extends drug half-life: A thrombolytic targeting therapy utilizing microenvironment-responsive artificial polysaccharide microvesicles. Carbohydr Polym 2024; 343:122505. [PMID: 39174110 DOI: 10.1016/j.carbpol.2024.122505] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 06/24/2024] [Accepted: 07/15/2024] [Indexed: 08/24/2024]
Abstract
The development of thrombolytic drug carriers capable of thrombus-targeting, prolonged circulation time, intelligent responsive release, and the ability to inhibit thrombotic recurrences remains a promising but significant challenge. To tackle this, an artificial polysaccharide microvesicle drug delivery system (uPA-CS/HS@RGD-ODE) was constructed. It is composed of cationic chitosan and anionic heparin assembled in a layer by layer structure, followed by surface modification using RGD peptide and 2-(N-oxide-N,N-diethylamino) ethylmethacrylate (ODE) before encapsulation of urokinase-type plasminogen activator (uPA). The effect of chitosan on the basic performances of uPA-CS/HS@RGD-ODE was estimated. The in vitro results suggest the uPA carrier, CS/HS@RGD-ODE, displayed outstanding targeting specific to activated platelets (61 %) and microenvironment-responsiveness at pH 6.5, facilitating thrombus-targeting and a controlled drug release, respectively. Most importantly, in vivo experiment suggests ODE from uPA-CS/HS@RGD-ODE substantially extends the half-life of uPA (120 min), as uPA-CS/HS@RGD-ODE can adhere onto erythrocytes and deliver uPA under cover of erythrocytes enabling a prolonged circulation time in the bloodstream. Further tail vein and abdominal aorta thrombosis models confirmed uPA-CS/HS@RGD-ODE exhibited superior targeting and thrombolysis capabilities compared to systemic administration of free uPA. To the knowledge of authors, this may be the first study to develop new drug carriers for delivery of thrombolytic drugs under the cover of erythrocytes for extended drug half-lives.
Collapse
Affiliation(s)
- Lianqi Shan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Junsu Wang
- Chongqing Customs, Chongqing 400044, China
| | - Hongyu Tu
- Chongqing Customs, Chongqing 400044, China
| | - Wenhan Zhang
- College of Computer and Information Science and College of Software, Southwest University, Chongqing 400715, China
| | - He Li
- Department of Geriatric Medicine, Wenzhou Ouhai District Chinese and Western Medical Association Hospital, Wenzhou 325000, China
| | - Paul Slezak
- Ludwig Boltzmann Institute for Traumatology, AUVA Research Center, 1200 Vienna, Austria
| | - Fei Lu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China
| | - Dongwon Lee
- Department of Bionanotechnology and Bioconvergence Engineering and Department of Polymer·Nano Science and Technology, Jeonbuk National University, Jeonju, Chonbuk 54896, Republic of Korea
| | - Enling Hu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; School of Fashion and Textiles, The Hong Kong Polytechnic University, Hong Kong.
| | - Zhen Geng
- Institute of Translational Medicine, Organoid Research Center, and National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, China.
| | - Guangqian Lan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China.
| | - Ruiqi Xie
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Department of Geriatric Medicine, Wenzhou Ouhai District Chinese and Western Medical Association Hospital, Wenzhou 325000, China.
| |
Collapse
|
7
|
Ahmaditabar P, Mahmoodi M, Taheri RA, Asefnejad A. Preparation and in vitro evaluation of tissue plasminogen activator-loaded nanoliposomes with anticoagulant coating. Biochim Biophys Acta Gen Subj 2024; 1868:130704. [PMID: 39178920 DOI: 10.1016/j.bbagen.2024.130704] [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: 04/10/2024] [Revised: 07/06/2024] [Accepted: 08/16/2024] [Indexed: 08/26/2024]
Abstract
The clinical efficacy of tissue plasminogen activator (tPA) is limited by its lack of specific delivery, requiring large therapeutic doses that increase the risk of intracerebral hemorrhage, bleeding at the surgical site, and patient mortality after angioplasty. To address these limitations, this study aimed to develop a chitosan polysulfate (CsPs)-coated liposomal formulation for the sustained release of tPA. The CsPs-coated liposomes containing tPA (Liposome-tPA/CsPs) were fabricated using the thin-film hydration technique and their properties were compared to tPA-encapsulated nanoliposomes without a coating layer (Liposome-tPA). Liposome-tPA/CsPs showed a quasi-spherical morphology with a hydrodynamic diameter of 110 nm, while Liposome-tPA had a diameter of 80 nm. The thermal analysis showed that the degradation temperature and glass transition temperature (Tg) of Liposome-tPA/CsPs were higher than that of tPA alone, indicating improved temperature stability. The in vitro release study demonstrated a slow and sustained release of tPA from the Liposome-tPA/CsPs, with a concentration of 0.02 mg/ml at 1 h and 0.23 mg/ml at 180 h. The CsPs coating layer enhanced the antibacterial and antioxidant activity of the nanoliposomes. Liposome-tPA/CsPs exhibited higher cell viability compared to Liposome-tPA. It also achieved a higher percentage of thrombolysis, with complete clot dissolution observed after 3 h of treatment. These findings suggest that the Liposome-tPA/CsPs can be a promising approach to overcome the limitations associated with the systemic administration of tPA, potentially enhancing its clinical efficacy while reducing the risk of adverse events.
Collapse
Affiliation(s)
- Parvin Ahmaditabar
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Mahboobeh Mahmoodi
- Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran; Joint Reconstruction Research Center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Ramezan Ali Taheri
- Department of Biology, Faculty of Sciences, University of Tehran, Tehran, Iran
| | - Azadeh Asefnejad
- Department of Biomedical Engineering, Science and Research Branch, Islamic Azad University, Tehran, Iran
| |
Collapse
|
8
|
Fan D, Liu X, Chen H. Endothelium-Mimicking Materials: A "Rising Star" for Antithrombosis. ACS APPLIED MATERIALS & INTERFACES 2024; 16:53343-53371. [PMID: 39344055 DOI: 10.1021/acsami.4c12117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/01/2024]
Abstract
The advancement of antithrombotic materials has significantly mitigated the thrombosis issue in clinical applications involving various medical implants. Extensive research has been dedicated over the past few decades to developing blood-contacting materials with complete resistance to thrombosis. However, despite these advancements, the risk of thrombosis and other complications persists when these materials are implanted in the human body. Consequently, the modification and enhancement of antithrombotic materials remain pivotal in 21st-century hemocompatibility studies. Previous research indicates that the healthy endothelial cells (ECs) layer is uniquely compatible with blood. Inspired by bionics, scientists have initiated the development of materials that emulate the hemocompatible properties of ECs by replicating their diverse antithrombotic mechanisms. This review elucidates the antithrombotic mechanisms of ECs and examines the endothelium-mimicking materials developed through single, dual-functional and multifunctional strategies, focusing on nitric oxide release, fibrinolytic function, glycosaminoglycan modification, and surface topography modification. These materials have demonstrated outstanding antithrombotic performance. Finally, the review outlines potential future research directions in this dynamic field, aiming to advance the development of antithrombotic materials.
Collapse
Affiliation(s)
- Duanqi Fan
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Xiaoli Liu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Hong Chen
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| |
Collapse
|
9
|
Weng PW, Liu CH, Jheng PR, Chiang CC, Chen YT, Rethi L, Hsieh YSY, Chuang AEY. Spermatozoon-propelled microcellular submarines combining innate magnetic hyperthermia with derived nanotherapies for thrombolysis and ischemia mitigation. J Nanobiotechnology 2024; 22:470. [PMID: 39118029 PMCID: PMC11308583 DOI: 10.1186/s12951-024-02716-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2023] [Accepted: 07/09/2024] [Indexed: 08/10/2024] Open
Abstract
Thrombotic cardiovascular diseases are a prevalent factor contributing to both physical impairment and mortality. Thrombolysis and ischemic mitigation have emerged as leading contemporary therapeutic approaches for addressing the consequences of ischemic injury and reperfusion damage. Herein, an innovative cellular-cloaked spermatozoon-driven microcellular submarine (SPCS), comprised of multimodal motifs, was designed to integrate nano-assembly thrombolytics with an immunomodulatory ability derived from innate magnetic hyperthermia. Rheotaxis-based navigation was utilized to home to and cross the clot barrier, and finally accumulate in ischemic vascular organs, where the thrombolytic motif was "switched-on" by the action of thrombus magnetic red blood cell-driven magnetic hyperthermia. In a murine model, the SPCS system combining innate magnetic hyperthermia demonstrated the capacity to augment delivery efficacy, produce nanotherapeutic outcomes, exhibit potent thrombolytic activity, and ameliorate ischemic tissue damage. These findings underscore the multifaceted potential of our designed approach, offering both thrombolytic and ischemia-mitigating effects. Given its extended therapeutic effects and thrombus-targeting capability, this biocompatible SPCS system holds promise as an innovative therapeutic agent for enhancing efficacy and preventing risks after managing thrombosis.
Collapse
Affiliation(s)
- Pei-Wei Weng
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering, Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, 23561, Taiwan
- Department of Orthopedics, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
- Department of Orthopedics, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
| | - Chia-Hung Liu
- Department of Urology, School of Medicine, College of Medicine, Taipei Medical University, Taipei, 11031, Taiwan
- TMU Research Center of Urology and Kidney, Taipei Medical University, Taipei, 11031, Taiwan
- Department of Urology, Shuang Ho Hospital, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Pei-Ru Jheng
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering, Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Chia-Che Chiang
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering, Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Yan-Ting Chen
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering, Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Lekshmi Rethi
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering, Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, 23561, Taiwan
| | - Yves S Y Hsieh
- School of Pharmacy, College of Pharmacy, Taipei Medical University, Taipei, 11031, Taiwan
- Division of Glycoscience, Department of Chemistry, School of Engineering Sciences in Chemistry, Biotechnology, and Health, KTH Royal Institute of Technology, Alba Nova University Centre, Stockholm, SE106 91, Sweden
| | - Andrew E-Y Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, International Ph.D. Program in Biomedical Engineering, Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, New Taipei City, 23561, Taiwan.
- Cell Physiology and Molecular Image Research Center, Taipei Medical University-Wan Fang Hospital, Taipei, 11696, Taiwan.
- Precision Medicine and Translational Cancer Research Center, Taipei Medical University Hospital, Taipei, 11031, Taiwan.
| |
Collapse
|
10
|
Huang Y, Wang J, Guo Y, Shen L, Li Y. Fibrinogen binding to activated platelets and its biomimetic thrombus-targeted thrombolytic strategies. Int J Biol Macromol 2024; 274:133286. [PMID: 38908635 DOI: 10.1016/j.ijbiomac.2024.133286] [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: 11/24/2023] [Revised: 06/18/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
Thrombosis is associated with various fatal arteriovenous syndromes including ischemic stroke, myocardial infarction, and pulmonary embolism. However, current clinical thrombolytic treatment strategies still have many problems in targeting and safety to meet the thrombolytic therapy needs. Understanding the molecular mechanism that underlies thrombosis is critical in developing effective thrombolytic strategies. It is well known that platelets play a central role in thrombosis and the binding of fibrinogen to activated platelets is a common pathway in the process of clot formation. Based on this, a concept of biomimetic thrombus-targeted thrombolytic strategy inspired from fibrinogen binding to activated platelets in thrombosis was proposed, which could selectively bind to activated platelets at a thrombus site, thus enabling targeted delivery and local release of thrombolytic agents for effective thrombolysis. In this review, we first summarized the main characteristics of platelets and fibrinogen, and then introduced the classical molecular mechanisms of thrombosis, including platelet adhesion, platelet activation and platelet aggregation through the interactions of activated platelets with fibrinogen. In addition, we highlighted the recent advances in biomimetic thrombus-targeted thrombolytic strategies which inspired from fibrinogen binding to activated platelets in thrombosis. The possible future directions and perspectives in this emerging area are briefly discussed.
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.
| | - 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
| | - Lingyue Shen
- Department of Oral & Maxillofacial-Head & Neck Oncology, Department of Laser and Aesthetic Medicine, Shanghai Ninth People's Hospital, College of Stomatology, Shanghai Jiao Tong University School of Medicine, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stoma-tology & Shanghai Research Institute of Stomatology, 639 Zhizaoju Road, Shanghai 200011, PR China.
| | - 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.
| |
Collapse
|
11
|
Ye Y, Chen Z, Zhang S, Slezak P, Lu F, Xie R, Lee D, Lan G, Hu E. pH-Responsive Theranostic Colloidosome Drug Carriers Enable Real-Time Imaging of Targeted Thrombolytic Process with Near-Infrared-II for Deep Venous Thrombosis. RESEARCH (WASHINGTON, D.C.) 2024; 7:0388. [PMID: 38812529 PMCID: PMC11136571 DOI: 10.34133/research.0388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2024] [Accepted: 04/21/2024] [Indexed: 05/31/2024]
Abstract
Thrombosis can cause life-threatening disorders. Unfortunately, current therapeutic methods for thrombosis using injecting thrombolytic medicines systemically resulted in unexpected bleeding complications. Moreover, the absence of practical imaging tools for thrombi raised dangers of undertreatment and overtreatment. This study develops a theranostic drug carrier, Pkr(IR-Ca/Pda-uPA)-cRGD, that enables real-time monitoring of the targeted thrombolytic process of deep vein thrombosis (DVT). Pkr(IR-Ca/Pda-uPA)-cRGD, which is prepared from a Pickering-emulsion-like system, encapsulates both near-infrared-II (NIR-II) contrast agent (IR-1048 dye, loading capacity: 28%) and urokinase plasminogen activators (uPAs, encapsulation efficiency: 89%), pioneering the loading of multiple drugs with contrasting hydrophilicity into one single-drug carrier. Upon intravenous injection, Pkr(IR-Ca/Pda-uPA)-cRGD considerably targets to thrombi selectively (targeting rate: 91%) and disintegrates in response to acidic thrombi to release IR-1048 dye and uPA for imaging and thrombolysis, respectively. Investigations indicate that Pkr(IR-Ca/Pda-uPA)-cRGD enabled real-time visualization of targeted thrombolysis using NIR-II imaging in DVT models, in which thrombi were eliminated (120 min after drug injection) without bleeding complications. This may be the first study using convenient NIR-II imaging for real-time visualization of targeted thrombolysis. It represents the precision medicine that enables rapid response to acquire instantaneous medical images and make necessary real-time adjustments to diagnostic and therapeutic protocols during treatment.
Collapse
Affiliation(s)
- Yaxin Ye
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
| | - Zhechang Chen
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
| | - Shengzhang Zhang
- Department of Cardiovascular Medicine,
Yueqing People's Hospital, Wenzhou 325699, China
| | - Paul Slezak
- Ludwig Boltzmann Institute for Traumatology,
AUVA Research Center, 1200 Vienna, Austria
| | - Fei Lu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
| | - Ruiqi Xie
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
- Ludwig Boltzmann Institute for Traumatology,
AUVA Research Center, 1200 Vienna, Austria
| | - Dongwon Lee
- Department of Bionanotechnology and Bioconvergence Engineering and Department of Polymer·Nano Science and Technology,
Jeonbuk National University, Jeonju, Chonbuk 54896, Republic of Korea
| | - Guangqian Lan
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
| | - Enling Hu
- State Key Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences,
Southwest University, Chongqing 400715, China
- School of Fashion and Textiles,
The Hong Kong Polytechnic University, Hong Kong
| |
Collapse
|
12
|
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
|
13
|
Chen Z, Yuan C, Ye Y, Lu B, Hu E, Lu F, Yu K, Xie R, Lan G. Dual-targeting fucoidan-based microvesicle for arterial thrombolysis and re-occlusion inhibition. Carbohydr Polym 2024; 328:121703. [PMID: 38220339 DOI: 10.1016/j.carbpol.2023.121703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 12/13/2023] [Accepted: 12/14/2023] [Indexed: 01/16/2024]
Abstract
Arterial thrombosis is a critical thrombotic disease that poses a significant threat to human health. However, the existing clinical treatment of arterial thrombosis lacks effective targeting and precise drug release capability. In this study, we developed a system for targeted delivery and on-demand release in arterial thrombosis treatment. The carrier was constructed using chitosan (CS) and fucoidan (Fu) through layer-by-layer assembly, with subsequent surface modification using cRGD peptide. Upon encapsulation of urokinase-type plasminogen activator (uPA), the resulting therapeutic drug delivery system, uPA-CS/Fu@cRGD, demonstrated dual-targeting abilities towards P-selectin and αIIbβ3, as well as pH and platelet-responsive release properties. Importantly, we have demonstrated that the dual targeting effect exhibits higher targeting efficiency at shear rates simulating thrombosed arterial conditions (1800 s-1) compared to single targeting for the first time. In the mouse common iliac artery model, uPA-CS/Fu@cRGD exhibited great thrombolytic capability while promoting the down-regulation of coagulation factors (FXa and PAI-1) and inflammatory factors (TNF-α and IL-6), thus improving the thrombus microenvironment and exerting potential in preventing re-occlusion. Our dual-target and dual-responsive, fucoidan-based macrovesicle represent a promising platform for advanced drug target delivery applications, with potential to prevent coagulation tendencies as well as improving thrombolytic and reducing the risk of re-occlusion.
Collapse
Affiliation(s)
- Zhechang Chen
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Caijie Yuan
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Yaxin Ye
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Bitao Lu
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Enling Hu
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Fei Lu
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Kun Yu
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China
| | - Ruiqi Xie
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Ludwig Boltzmann Institute for Experimental and Clinical Traumatology, AUVA Research Center, 1200 Vienna, Austria.
| | - Guangqian Lan
- State Key Laboratory of Laboratory of Resource Insects, College of Sericulture, Textile and Biomass Sciences, Southwest University, Chongqing 400715, China; Chongqing Engineering Research Center of Biomaterial Fiber and Modern Textile, Chongqing 400715, China.
| |
Collapse
|
14
|
Han X, Qin Y, Mei C, Jiao F, Khademolqorani S, Nooshin Banitaba S. Current trends and future perspectives of stroke management through integrating health care team and nanodrug delivery strategy. Front Cell Neurosci 2023; 17:1266660. [PMID: 38034591 PMCID: PMC10685387 DOI: 10.3389/fncel.2023.1266660] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/25/2023] [Indexed: 12/02/2023] Open
Abstract
Stroke is accounted as the second-most mortality and adult disability factor in worldwide, while causes the bleeding promptly and lifetime consequences. The employed functional recovery after stroke is highly variable, allowing to deliver proper interventions to the right stroke patient at a specific time. Accordingly, the multidisciplinary nursing team, and the administrated drugs are major key-building-blocks to enhance stroke treatment efficiency. Regarding the healthcare team, adequate continuum of care have been declared as an integral part of the treatment process from the pre-hospital, in-hospital, to acute post-discharge phases. As a curative perspective, drugs administration is also vital in surviving at the early step and reducing the probability of disabilities in later. In this regard, nanotechnology-based medicinal strategy is exorbitantly burgeoning. In this review, we have highlighted the effectiveness of current clinical care considered by nursing teams to treat stroke. Also, the advancement of drugs through synthesis of miniaturized nanodrug formations relating stroke treatment is remarked. Finally, the remained challenges toward standardizing the healthcare team and minimizing the nanodrugs downsides are discussed. The findings ensure that future works on normalizing the healthcare nursing teams integrated with artificial intelligence technology, as well as advancing the operative nanodrugs can provide value-based stroke cares.
Collapse
Affiliation(s)
- Xuelu Han
- Nursing Clinic, Affiliated Hospital of Jilin Medical University, Jilin, China
| | - Yingxin Qin
- Department of Nursing, Affiliated Hospital of Jilin Medical University, Jilin, China
| | - Chunli Mei
- Nursing College, Beihua University, Jilin, China
| | - Feitong Jiao
- Nursing Training Center, School of Nursing, Jilin Medical University, Jilin, China
| | - Sanaz Khademolqorani
- Department of Textile Engineering, Isfahan University of Technology, Isfahan, Iran
- Emerald Experts Laboratory, Isfahan Science and Technology Town, Isfahan, Iran
| | - Seyedeh Nooshin Banitaba
- Emerald Experts Laboratory, Isfahan Science and Technology Town, Isfahan, Iran
- Department of Textile Engineering, Amirkabir University of Technology, Tehran, Iran
| |
Collapse
|
15
|
Omidian H, Babanejad N, Cubeddu LX. Nanosystems in Cardiovascular Medicine: Advancements, Applications, and Future Perspectives. Pharmaceutics 2023; 15:1935. [PMID: 37514121 PMCID: PMC10386572 DOI: 10.3390/pharmaceutics15071935] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 07/02/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Cardiovascular diseases (CVDs) remain a leading cause of morbidity and mortality globally. Despite significant advancements in the development of pharmacological therapies, the challenges of targeted drug delivery to the cardiovascular system persist. Innovative drug-delivery systems have been developed to address these challenges and improve therapeutic outcomes in CVDs. This comprehensive review examines various drug delivery strategies and their efficacy in addressing CVDs. Polymeric nanoparticles, liposomes, microparticles, and dendrimers are among the drug-delivery systems investigated in preclinical and clinical studies. Specific strategies for targeted drug delivery, such as magnetic nanoparticles and porous stent surfaces, are also discussed. This review highlights the potential of innovative drug-delivery systems as effective strategies for the treatment of CVDs.
Collapse
Affiliation(s)
- Hossein Omidian
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Niloofar Babanejad
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| | - Luigi X Cubeddu
- Barry and Judy Silverman College of Pharmacy, Nova Southeastern University, Fort Lauderdale, FL 33328, USA
| |
Collapse
|
16
|
Zhou S, Zhao W, Hu J, Mao C, Zhou M. Application of Nanotechnology in Thrombus Therapy. Adv Healthc Mater 2023; 12:e2202578. [PMID: 36507827 DOI: 10.1002/adhm.202202578] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 11/26/2022] [Indexed: 12/14/2022]
Abstract
A thrombus is a blood clot that forms in the lumen of an artery or vein, restricting blood flow and causing clinical symptoms. Thrombosis is associated with many life-threatening cardiovascular diseases. However, current clinical therapeutic technologies still have many problems in targeting, enrichment, penetration, and safety to meet the thrombosis treatment needs. Therefore, researchers devote themselves to developing nanosystems loaded with antithrombotic drugs to address this paradox in recent years. Herein, the existing thrombosis treatment technologies are first reviewed; and then, their advantages and disadvantages are outlined based on a brief discussion of thrombosis's definition and formation mechanism. Furthermore, the need and application cases for introducing nanotechnology are discussed, focusing on thrombus-specific targeted ligand modification technology and microenvironment-triggered responsive drug release technology. Then, nanomaterials that can be used to design antithrombotic nanotherapeutic systems are summarized. Moreover, a variety of drug delivery technologies driven by nanomotors in thrombosis therapy is also introduced. Last of all, a prospective discussion on the future development of nanotechnology for thrombosis therapy is highlighted.
Collapse
Affiliation(s)
- Shuyin Zhou
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China.,Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| | - Wenbo Zhao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Jinglei Hu
- Kuang Yaming Honors School, Nanjing University, Nanjing, 210023, China
| | - Chun Mao
- National and Local Joint Engineering Research Center of Biomedical Functional Materials, School of Chemistry and Materials Science, Nanjing Normal University, Nanjing, 210023, China
| | - Min Zhou
- Department of Vascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, 210008, China
| |
Collapse
|
17
|
Russell P, Esser L, Hagemeyer CE, Voelcker NH. The potential impact of nanomedicine on COVID-19-induced thrombosis. NATURE NANOTECHNOLOGY 2023; 18:11-22. [PMID: 36536042 DOI: 10.1038/s41565-022-01270-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Accepted: 10/12/2022] [Indexed: 06/17/2023]
Abstract
Extensive reports of pulmonary embolisms, ischaemic stroke and myocardial infarctions caused by coronavirus disease 2019 (COVID-19), as well as a significantly increased long-term risk of cardiovascular diseases in COVID-19 survivors, have highlighted severe deficiencies in our understanding of thromboinflammation and the need for new therapeutic options. Due to the complexity of the immunothrombosis pathophysiology, the efficacy of treatment with conventional anti-thrombotic medication is questioned. Thrombolytics do appear efficacious, but are hindered by severe bleeding risks, limiting their use. Nanomedicine can have profound impact in this context, protecting delicate (bio)pharmaceuticals from degradation en route and enabling delivery in a targeted and on demand manner. We provide an overview of the most promising nanocarrier systems and design strategies that may be adapted to develop nanomedicine for COVID-19-induced thromboinflammation, including dual-therapeutic approaches with antiviral and immunosuppressants. Resultant targeted and side-effect-free treatment may aid greatly in the fight against the ongoing COVID-19 pandemic.
Collapse
Affiliation(s)
- Peije Russell
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, Australia
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia
| | - Lars Esser
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Commonwealth Scientific and Industrial Research Organisation (CSIRO), Manufacturing, Clayton, Victoria, Australia
| | - Christoph E Hagemeyer
- Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria, Australia.
| | - Nicolas H Voelcker
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia.
- Melbourne Centre for Nanofabrication, Victorian Node of Australian National Fabrication Facility, Clayton, Victoria, Australia.
- Department of Materials Science and Engineering, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
18
|
Laser thrombolysis and in vitro release kinetics of tPA encapsulated in chitosan polysulfate-coated nanoliposome. Carbohydr Polym 2023; 299:120225. [PMID: 36876826 DOI: 10.1016/j.carbpol.2022.120225] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2022] [Revised: 10/05/2022] [Accepted: 10/11/2022] [Indexed: 11/09/2022]
Abstract
A major challenge in managing coronary artery disease is to find an effective thrombolytic therapy with minimal side effects. Laser thrombolysis is a practical procedure to remove the thrombus from inside blocked arteries, although it can cause embolism and re-occlusion of the vessel. The present study aimed to design a liposome drug delivery system for the controlled release of tissue plasminogen activator (tPA) and delivery of drug system into the thrombus by Nd:YAG laser at a wavelength of 532 nm for the treatment of arterial occlusive diseases. In this study, tPA encapsulated into the chitosan polysulfate-coated liposome (Lip/PSCS-tPA) was fabricated by a thin-film hydration technique. The particle size of Lip/tPA and Lip/PSCS-tPA was 88 and 100 nm, respectively. The release rate of tPA from Lip/PSCS-tPA was measured to be 35 % and 66 % after 24 h and 72 h, respectively. Thrombolysis through the delivery of Lip/PSCS-tPA into the thrombus during the laser irradiation was higher compared to irradiated thrombus without the nanoliposomes. The expression of IL-10 and TNF-α genes was studied by RT-PCR. The level of TNF-α for Lip/PSCS-tPA was lower than that of tPA, which can lead to improved cardiac function. Also, in this study, the thrombus dissolution process was studied using a rat model. After 4 h, the thrombus area in the femoral vein was significantly lower for groups treated with Lip/PSCS-tPA (5 %) compared to the groups treated with tPA alone (45 %). Thus, according to our results, the combination of Lip/PSCS-tPA and laser thrombolysis can be introduced as an appropriate technique for accelerating thrombolysis.
Collapse
|
19
|
Hale MM, Medina SH. Biomaterials-Enabled Antithrombotics: Recent Advances and Emerging Strategies. Mol Pharm 2022; 19:4453-4465. [PMID: 36149250 PMCID: PMC9728464 DOI: 10.1021/acs.molpharmaceut.2c00626] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2022] [Revised: 09/13/2022] [Accepted: 09/14/2022] [Indexed: 12/13/2022]
Abstract
Antithrombotic and thrombolytic therapies are used to prevent, treat, and remove blood clots in various clinical settings, from emergent to prophylactic. While ubiquitous in their healthcare application, short half-lives, off-target effects, overdosing complications, and patient compliance continue to be major liabilities to the utility of these agents. Biomaterials-enabled strategies have the potential to comprehensively address these limitations by creating technologies that are more precise, durable, and safe in their antithrombotic action. In this review, we discuss the state of the art in anticoagulant and thrombolytic biomaterials, covering the nano to macro length scales. We emphasize current methods of formulation, discuss how material properties affect controlled release kinetics, and summarize modern mechanisms of clot-specific drug targeting. The preclinical efficacy of these technologies in an array of cardiovascular applications, including stroke, pulmonary embolism, myocardial infarction, and blood contacting devices, is summarized and performance contrasted. While significant advances have already been made, ongoing development efforts look to deliver bioresponsive "smart" biomaterials that will open new precision medicine opportunities in cardiology.
Collapse
Affiliation(s)
- Macy M. Hale
- Department
of Biomedical Engineering, Pennsylvania
State University, University
Park, Pennsylvania 16802-4400, United States
| | - Scott H. Medina
- Department
of Biomedical Engineering, Pennsylvania
State University, University
Park, Pennsylvania 16802-4400, United States
- Huck
Institutes of the Life Sciences, Pennsylvania
State University, University Park, Pennsylvania 16802-4400, United States
| |
Collapse
|
20
|
Hu L, Xu J, Zhang W, Wang J, Fang N, Luo Y, Xu L, Liu J, Zhang Y, Ran H, Guo D, Zhou J. A Synergistic and Efficient Thrombolytic Nanoplatform: A Mechanical Method of Blasting Combined with Thrombolytic Drugs. Int J Nanomedicine 2022; 17:5229-5246. [PMID: 36388875 PMCID: PMC9662339 DOI: 10.2147/ijn.s382964] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/07/2022] [Indexed: 09/29/2023] Open
Abstract
BACKGROUND AND OBJECTIVE Thrombosis is a common disease that poses a great threat to life and health. Most thrombolytic effects of traditional treatments or nanomedicine are not efficient or safe enough. Therefore, we designed a nanoparticle (NP) with a combination of a phase transition material and thrombolytic drugs for efficient and safe thrombolysis. METHODS A thrombus fibrin-targeted and phase transition NP was designed and contained perfluorohexane (PFH) and the thrombolytic drug rtPA core, with CREKA polypeptides attached to the shell of the PLGA NPs. Characterization of the phase transition and ultrasound imaging of the NPs was carried out under low-intensity focused ultrasound (LIFU). LIFU-responsive drug release in vitro was also explored. Under the synergistic effect of PFH and rtPA, the efficient thrombolysis ability of the NPs was studied in vitro and in vivo. In vivo monitoring of thrombosis and biosafety were also verified. RESULTS The PPrC NPs had good ultrasound imaging ability under LIFU irradiation and were related to the phase transition characteristics of the NPs. CREKA polypeptides can effectively increase the aggregation of the NPs on thrombi. Under static and dynamic conditions in vitro, the "liquid to gas" transformation effect of PFH can perform the destruction function of the excavator at the thrombus site and promote the specific release of rtPA, and the subsequent rtPA drug thrombolysis can further fully dissolve the thrombus. In vivo experiments showed that the NPs can monitor the formation of thrombi and have good thrombolytic effects, with significantly reduced bleeding side effects. The biochemical indexes of the rats were within normal limits after treatment. CONCLUSION PPrC NPs loaded with PFH and rtPA combining a mechanical way of blasting with thrombolytic drugs may be a promising new and reliable approach for thrombus monitoring and treatment.
Collapse
Affiliation(s)
- Liu Hu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jie Xu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Wenli Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Junrui Wang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ni Fang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Ying Luo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Lian Xu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jia Liu
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Yu Zhang
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Haitao Ran
- Department of Ultrasound, Institute of Ultrasound Imaging, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Dajing Guo
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| | - Jun Zhou
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, People’s Republic of China
| |
Collapse
|
21
|
Huang Y, Jiang J, Ren J, Guo Y, Zhao Q, Zhou J, Li Y, Chen R. A Fibrinogen-Mimicking, Activated-Platelet-Sensitive Nanocoacervate Enhances Thrombus Targeting and Penetration of Tissue Plasminogen Activator for Effective Thrombolytic Therapy. Adv Healthc Mater 2022; 11:e2201265. [PMID: 35864062 PMCID: PMC11468879 DOI: 10.1002/adhm.202201265] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/01/2022] [Indexed: 01/27/2023]
Abstract
The development of a fibrinolytic system with long circulation time, high thrombus targeting, efficient thrombus penetration, effective thrombolysis, and minimal hemorrhagic risk remains a major challenge. Herein, inspired by fibrinogen binding to activated platelets in thrombosis, this article reports a fibrinogen-mimicking, activated-platelet-sensitive nanocoacervate to enhance thrombus penetration of tissue plasminogen activator (tPA) for targeted thrombolytic therapy. This biomimetic nanothrombolytic system, denoted as RGD-Chi@tPA, is constructed by "one-pot" coacervation through electrostatic interactions between positively charged arginine-glycine-aspartic acid (RGD)-grafted chitosan (RGD-Chi) and negatively charged tPA. Flow cytometry and confocal laser scanning microscopy measurements show targeting of RGD-Chi@tPA to activated platelets. Controlled tPA release triggered by activated platelets at a thrombus site is demonstrated. Its targeted fibrinolytic and thrombolytic activities are measured in in vitro models. The pharmacokinetic profiles show that RGD-Chi@tPA can significantly prolong circulation time compared to free tPA. In a mouse tail thrombus model, RGD-Chi@tPA displays efficient thrombus targeting and penetration, enabling a complete vascular recanalization as confirmed by the fluorescence imaging, histochemical assay, and laser speckle contrast imager. Consequently, RGD-Chi@tPA induces a substantial enhancement in thrombolysis with minimal hemorrhagic risk compared to free tPA. This simple, effective, and safe platform holds great promise for the development of thrombolytic nanomedicines.
Collapse
Affiliation(s)
- Yu Huang
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai, 200233, P. R. China
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Jingxuan Jiang
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Jie Ren
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| | - Yuanyuan Guo
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Qianqian Zhao
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Jia Zhou
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Yuehua Li
- Department of Radiology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, 600 Yi Shan Road, Shanghai, 200233, P. R. China
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, SW7 2AZ, UK
| |
Collapse
|
22
|
Tapeinos C, Gao H, Bauleth-Ramos T, Santos HA. Progress in Stimuli-Responsive Biomaterials for Treating Cardiovascular and Cerebrovascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200291. [PMID: 35306751 DOI: 10.1002/smll.202200291] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs) describe abnormal vascular system conditions affecting the brain and heart. Among these, ischemic heart disease and ischemic stroke are the leading causes of death worldwide, resulting in 16% and 11% of deaths globally. Although several therapeutic approaches are presented over the years, the continuously increasing mortality rates suggest the need for more advanced strategies for their treatment. One of these strategies lies in the use of stimuli-responsive biomaterials. These "smart" biomaterials can specifically target the diseased tissue, and after "reading" the altered environmental cues, they can respond by altering their physicochemical properties and/or their morphology. In this review, the progress in the field of stimuli-responsive biomaterials for CCVDs in the last five years, aiming at highlighting their potential as early-stage therapeutics in the preclinical scenery, is described.
Collapse
Affiliation(s)
- Christos Tapeinos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Han Gao
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Tomás Bauleth-Ramos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| |
Collapse
|
23
|
Li YX, Wang HB, Li J, Jin JB, Hu JB, Yang CL. Targeting pulmonary vascular endothelial cells for the treatment of respiratory diseases. Front Pharmacol 2022; 13:983816. [PMID: 36110525 PMCID: PMC9468609 DOI: 10.3389/fphar.2022.983816] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
Pulmonary vascular endothelial cells (VECs) are the main damaged cells in the pathogenesis of various respiratory diseases and they mediate the development and regulation of the diseases. Effective intervention targeting pulmonary VECs is of great significance for the treatment of respiratory diseases. A variety of cell markers are expressed on the surface of VECs, some of which can be specifically combined with the drugs or carriers modified by corresponding ligands such as ICAM-1, PECAM-1, and P-selectin, to achieve effective delivery of drugs in lung tissues. In addition, the great endothelial surface area of the pulmonary vessels, the “first pass effect” of venous blood in lung tissues, and the high volume and relatively slow blood perfusion rate of pulmonary capillaries further promote the drug distribution in lung tissues. This review summarizes the representative markers at the onset of respiratory diseases, drug delivery systems designed to target these markers and their therapeutic effects.
Collapse
Affiliation(s)
- Yi-Xuan Li
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
| | - Hong-Bo Wang
- Department of Pharmacy, Yuyao People’s Hospital, Yuyao, China
| | - Jing Li
- Department of Pharmacy, Yuyao People’s Hospital, Yuyao, China
| | - Jian-Bo Jin
- Department of Pharmacy, Yuyao People’s Hospital, Yuyao, China
| | - Jing-Bo Hu
- School of Materials Science and Chemical Engineering, Ningbo University, Ningbo, China
- *Correspondence: Jing-Bo Hu, ; Chun-Lin Yang,
| | - Chun-Lin Yang
- Department of Pharmacy, Yuyao People’s Hospital, Yuyao, China
- *Correspondence: Jing-Bo Hu, ; Chun-Lin Yang,
| |
Collapse
|
24
|
Zhang S, Li J, Ren J, Xue Z, Qi X, Si Q. Cyclic RGD functionalized PLGA nanoparticles loaded with noncovalent complex of indocyanine green with urokinase for synergistic thrombolysis. Front Bioeng Biotechnol 2022; 10:945531. [PMID: 36032719 PMCID: PMC9399888 DOI: 10.3389/fbioe.2022.945531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022] Open
Abstract
Thrombotic diseases have the characteristics of long latency period, rapid onset, and high mortality rate, which seriously threaten people's life and health. The aim of this research is to fabricate a novel indocyanine green complex of urokinase (ICG@uPA) and employ the amphiphilic PEG-PLGA polymer to deliver the complex as an enzyme-phototherapeutic synergistic thrombolysis platform. The noncovalent indocyanine green (ICG) complex of urokinase (ICG@uPA) was prepared via supramolecular self-assembly and then encapsulated into cRGD decorated polymeric nanoparticles (cRGD-ICG-uPA NPs) by double-emulsion solvent evaporation method. Then the nanoparticles (NPs) were characterized in terms of particle size, optical properties, in vitro release, etc. The targeting and thrombolytic effect of the nanoparticles were studied both in vitro and in vivo. ICG@uPA and cRGD-ICG-uPA NPs displayed significantly higher photostability and laser energy conversion efficiency than free ICG. Concomitantly, the NPs exhibited selective binding affinity to the activated platelets and specific accumulation in the mouse mesenteric vessel thrombus. Significant thrombolysis was achieved in vivo by photo-assisted synergistic therapy with reduced dose and systemic bleeding risk of uPA. Our results prove that the functional PLGA nanoparticle loaded with the ICG@uPA offers a novel option for effective and safe thrombolytic treatment.
Collapse
Affiliation(s)
- Sha Zhang
- Department of Geriatric Cardiology, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Jinjie Li
- Centre of Sport Nutrition and Health, Zhengzhou University, Zhengzhou, China
| | - Jiefeng Ren
- Department of Geriatric Cardiology, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Zaiyao Xue
- Department of Geriatric Cardiology, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Xinlian Qi
- Department of Geriatric Cardiology, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
- Medical School of Chinese PLA, Beijing, China
| | - Quanjin Si
- Department of the Third Health Care, Second Medical Center of Chinese PLA General Hospital, National Clinical Research Center for Geriatric Diseases, Beijing, China
| |
Collapse
|
25
|
Quan X, Han Y, Lu P, Ding Y, Wang Q, Li Y, Wei J, Huang Q, Wang R, Zhao Y. Annexin V-Modified Platelet-Biomimetic Nanomedicine for Targeted Therapy of Acute Ischemic Stroke. Adv Healthc Mater 2022; 11:e2200416. [PMID: 35708176 DOI: 10.1002/adhm.202200416] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Revised: 05/18/2022] [Indexed: 01/24/2023]
Abstract
Thromboembolic stroke is typically characterized by the activation of platelets, resulting in thrombus in the cerebral vascular system, leading to high morbidity and mortality globally. Intravenous thrombolysis by tissue plasminogen activator (tPA) administration within 4.5 h from the onset of symptoms is providing a standard therapeutic strategy for ischemic stroke, but this reagent simultaneously shows potential serious adverse effects, e.g., hemorrhagic transformation. Herein, a novel delivery platform based on Annexin V and platelet membrane is developed for tPA (APLT-PA) to enhance targeting efficiency, therapeutic effects, and reduce the risk of intracerebral hemorrhage in acute ischemic stroke. After preparation by extrusion of platelet membrane and subsequent insertion of Annexin V to liposomes, APLT-PA exhibits a high targeting efficiency to activated platelet in vitro and thrombosis site in vivo, due to the binding to phosphatidylserine (PS) and activated platelet membrane proteins. One dose of APLT-PA leads to obvious thrombolysis and significant improvement of neurological function within 7 days in mice with photochemically induced acute ischemic stroke. This study provides a novel, safe platelet-biomimetic nanomedicine for precise thrombolytic treatment of acute ischemic stroke, and offers new theories for the design and exploitation of cell-mimetic nanomedicine for diverse biomedical applications.
Collapse
Affiliation(s)
- Xingping Quan
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Yan Han
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Pengde Lu
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Yuanfu Ding
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Qingfu Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Yiyang Li
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Jianwen Wei
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Qiaoxian Huang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Ruibing Wang
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China.,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| | - Yonghua Zhao
- Institute of Chinese Medical Sciences, State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Taipa, Macau SAR, 999078, P. R. China.,Department of Pharmaceutical Sciences, Faculty of Health Sciences, University of Macau, Taipa, Macau SAR, 999078, P. R. China
| |
Collapse
|
26
|
Lin X, Li N, Tang H. Recent Advances in Nanomaterials for Diagnosis, Treatments, and Neurorestoration in Ischemic Stroke. Front Cell Neurosci 2022; 16:885190. [PMID: 35836741 PMCID: PMC9274459 DOI: 10.3389/fncel.2022.885190] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Accepted: 04/11/2022] [Indexed: 11/13/2022] Open
Abstract
Stroke is a major public health issue, corresponding to the second cause of mortality and the first cause of severe disability. Ischemic stroke is the most common type of stroke, accounting for 87% of all strokes, where early detection and clinical intervention are well known to decrease its morbidity and mortality. However, the diagnosis of ischemic stroke has been limited to the late stages, and its therapeutic window is too narrow to provide rational and effective treatment. In addition, clinical thrombolytics suffer from a short half-life, inactivation, allergic reactions, and non-specific tissue targeting. Another problem is the limited ability of current neuroprotective agents to promote recovery of the ischemic brain tissue after stroke, which contributes to the progressive and irreversible nature of ischemic stroke and also the severity of the outcome. Fortunately, because of biomaterials’ inherent biochemical and biophysical properties, including biocompatibility, biodegradability, renewability, nontoxicity, long blood circulation time, and targeting ability. Utilization of them has been pursued as an innovative and promising strategy to tackle these challenges. In this review, special emphasis will be placed on the recent advances in the study of nanomaterials for the diagnosis and therapy of ischemic stroke. Meanwhile, nanomaterials provide much promise for neural tissue salvage and regeneration in brain ischemia, which is also highlighted.
Collapse
Affiliation(s)
- Xinru Lin
- Department of Anesthesiology, Wenzhou Key Laboratory of Perioperative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Na Li
- Oujiang Laboratory, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, China
- *Correspondence: Na Li Hongli Tang
| | - Hongli Tang
- Department of Anesthesiology, Wenzhou Key Laboratory of Perioperative Medicine, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Na Li Hongli Tang
| |
Collapse
|
27
|
Lv J, Zhang L, Du W, Ling G, Zhang P. Functional gold nanoparticles for diagnosis, treatment and prevention of thrombus. J Control Release 2022; 345:572-585. [DOI: 10.1016/j.jconrel.2022.03.044] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 03/21/2022] [Accepted: 03/22/2022] [Indexed: 12/23/2022]
|
28
|
Zhang W, Wang J, Xie Z, Zou H, Chen Q, Xu L, Hu L, Fang N, Xu J, Zhou J, Liu J, Ran H, Wang Z, Zhang Y, Guo D. Antithrombotic Therapy by Regulating the ROS-Mediated Thrombosis Microenvironment and Specific Nonpharmaceutical Thrombolysis Using Prussian Blue Nanodroplets. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2106252. [PMID: 35246943 DOI: 10.1002/smll.202106252] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 01/30/2022] [Indexed: 06/14/2023]
Abstract
In thrombotic diseases, the effects of reactive oxygen species (ROS)-mediated oxidative stress as a "perpetrator" in thrombosis must be resolved. Accordingly, an insufficient understanding of thrombus therapy prompted the authors to pursue a more comprehensive and efficient antithrombotic treatment strategy. A Prussian blue (PB)-based nanodroplet system (PB-PFP@PC) is designed using PB and perfluorinated pentane (PFP) in the core, and a targeting peptide (CREKA, Cys-Arg-Glu-Lys-Ala) is attached to poly(lactic-coglycolic acid) (PLGA) as the delivery carrier shell. Upon near-infrared (NIR) laser irradiation, PB and PFP jointly achieve an unprecedented dual strategy for drug-free thrombolysis: photothermal therapy (PTT) combined with optical droplet vaporization (ODV). PB, a nanoenzyme, also regulates the vascular microenvironment via its antioxidant activity to continuously scavenge abnormally elevated ROS and correspondingly reduce inflammatory factors in the thrombus site. This study provides a demonstration of not only the potential of ODV in thrombus therapy but also the mechanism underlying PTT thrombolysis due to thermal ablation-induced fibrin network structural damage. Moreover, PB catalyzes ROS to generate oxygen (O2 ), which combines with the ODV effect, enhancing the ultrasound signal. Thus, regulation of the thrombosis microenvironment combined with specific nonpharmaceutical thrombolysis by PB nanodroplets provides a more comprehensive and efficient antithrombotic therapeutic strategy.
Collapse
Affiliation(s)
- Wenli Zhang
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Junrui Wang
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Zhuoyan Xie
- Department of Ultrasound, Chongqing General Hospital of Chinese Academy of Sciences, Chongqing, 401121, China
| | - Hongmi Zou
- Department of Ophthalmology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Qiaoqi Chen
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Lian Xu
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Liu Hu
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Ni Fang
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Jie Xu
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Jun Zhou
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Jia Liu
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Haitao Ran
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Zhigang Wang
- Chongqing Key Laboratory of Ultrasound Molecular Imaging & Department of Ultrasound, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Yu Zhang
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| | - Dajing Guo
- Department of Radiology, The Second Clinical Medical College, Chongqing Medical University, Chongqing, 400010, China
| |
Collapse
|
29
|
Xie S, Mo C, Cao W, Xie S, Li S, Zhang Z, Li X. Bacteria-propelled microtubular motors for efficient penetration and targeting delivery of thrombolytic agents. Acta Biomater 2022; 142:49-59. [PMID: 35158079 DOI: 10.1016/j.actbio.2022.02.008] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Revised: 01/12/2022] [Accepted: 02/07/2022] [Indexed: 11/01/2022]
Abstract
Effective thrombolysis is critical to rapidly rebuild blood flow for thrombosis patients. Drug delivery systems have been developed to address inadequate pharmacokinetics of thrombolytic agents, but challenges still remain in the timely removal of blood clots regarding the dense fibrin networks. Herein, rod-shaped tubular micromotors were developed to achieve efficient penetration and thorough destruction of thrombi. By using electrospun fiber fragments as the template, urokinase (uPA)-loaded polydopamine (PDA) microtubes with surface decorated fucoidan (FuPDAuPA) were prepared at the aspect ratio of around 2. One E. coli Nissle 1917 (EcN) was assembled into one microtube to construct a FuPDAuPA@EcN hybrid micromotor through PDA adhesion and L-aspartate induction. The pharmacokinetic analysis indicates that the encapsulation of uPA into micromotors extends the half-life from 0.4 to 5.6 h and increases the bioavailability over 10 times. EcN-propelled motion elevates adsorption capacities of FuPDAuPA@EcN for more than four times compared with that of FuPDAuPA. The fucoidan-mediated targeting causes 2-fold higher thrombolysis capacity in vitro and over 10-fold higher uPA accumulation in thrombi in vivo. In the treatment of venous thrombi at mouse hindlimbs, intravenous administration of FuPDAuPA@EcN completely removed blood clots with almost full recovery of blood flows and apparently alleviated tail bleeding. It should be noted that FuPDAuPA@EcN treatment at a reduced uPA dose caused no significant difference in the blood flow rate compared with those of FuPDAuPA. The synergistic action of fucoidan-induced targeting and EcN-driven motion provides a prerequisite for promoting thrombolytic efficacy and reducing uPA dose and bleeding side effect. STATEMENT OF SIGNIFICANCE: The standard treatment to thrombosis patient is intravenous infusion of thrombolytic agents, but the associated bleeding complications and impairment of normal haemostasis greatly offset the therapeutic benefits. Drug delivery systems have been developed to address the limitations of inadequate pharmacokinetics of thrombolytic agents, but challenges still exist in less efficient penetration into dense networks for thorough destruction of thrombi. Up to now only few attempts have been made to construct nano-/micromotors for combating thrombosis and there is no single case that antithrombosis is assisted by bacteria or cells-propelled motors. Herein, bacteria-propelled microtubes were developed to carry urokinase for efficient penetration into blood clots and effective thrombolysis. The synergistic action of bacteria-driven motion and specific ligand-induced targeting holds a promising treatment strategy for life-threatening cardiovascular diseases such as thrombosis and atherosclerosis.
Collapse
|
30
|
Li Y, Jin Y, He X, Tang Y, Zhou M, Guo W, Miao W. Cyclo(RGD) peptide-decorated silver nanoparticles with anti-platelet potential for active platelet-rich thrombus targeting. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2022; 41:102520. [PMID: 35038589 DOI: 10.1016/j.nano.2022.102520] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2021] [Revised: 11/15/2021] [Accepted: 12/29/2021] [Indexed: 06/14/2023]
Abstract
The development of integrated nanomedicine for prevention and early diagnosis of thrombosis is highly significant. Platelet plays a vital role in thrombotic disorders, offering an ideal target for thromboprophylaxis and imaging of thrombi. We herein fabricated cyclo(RGD) peptide-decorated AgNPs (designated cRGD-AgNPs) for active targeting platelet-rich thrombi. In vitro cytotoxicity and hemolysis assays demonstrated that cRGD-AgNPs have acceptable biocompatibility pattern. Both PEG-AgNPs (non-targeted version) and cRGD-AgNPs can inhibit agonist-mediated platelet aggregation, whereas the latter exhibited significant attenuation on platelet activation and adhesion onto collagen and fibrinogen matrix. Furthermore, the superior binding ability of cRGD-AgNPs with platelet-rich thrombus was demonstrated in static/dynamic condition in vitro. In vivo studies revealed that cRGD-AgNPs could actively target thrombi in a mouse model of carotid artery thrombi with favorable safety. Our results here suggest that cRGD-AgNPs with intrinsic anti-platelet potential might be promising nano theranostics for thromboprophylaxis and active thrombus targeting.
Collapse
Affiliation(s)
- Yuanyuan Li
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, PR China
| | - Yangye Jin
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, PR China
| | - Xiaofeng He
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, PR China
| | - Yonghui Tang
- Department of General Surgery, Children's Hospital of Nanjing Medical University, Nanjing, PR China.
| | - Min Zhou
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, PR China
| | - Wenjing Guo
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, PR China
| | - Wenjun Miao
- School of Pharmaceutical Sciences, Nanjing Tech University, Nanjing, PR China.
| |
Collapse
|
31
|
Yu W, Yin N, Yang Y, Xuan C, Liu X, Liu W, Zhang Z, Zhang K, Liu J, Shi J. Rescuing ischemic stroke by biomimetic nanovesicles through accelerated thrombolysis and sequential ischemia-reperfusion protection. Acta Biomater 2022; 140:625-640. [PMID: 34902617 DOI: 10.1016/j.actbio.2021.12.009] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/03/2021] [Accepted: 12/07/2021] [Indexed: 12/11/2022]
Abstract
Rational design of nanomedicine to accelerate thrombolysis and sequentially avoid thrombolysis-mediated reperfusion injury is still a challenge. Here, we develop a biomimetic nanovesicle (tPA/MNP@PM, tMP) by simple encapsulating melanin nanoparticles (MNP) and tPA with a platelet membrane vesicle (PM), which integrates the thrombus targeting property of PM, the photothermal conversion performance and free radical scavenging property of natural melanin for cascaded ischemic stroke treatment. Benefiting from natural thrombus-targeted adhesion capability of PM, nanovesicles could efficiently target thrombus site. Then near-infrared (NIR) mediated photothermal of MNP could lead to rupture of nanovesicles, thus achieving precise release of tPA in thrombus. Interestingly, local hyperthermia also increases the activity of tPA for accelerating thrombolysis. Afterwards, site specific released MNP (4.5 nm) accompanied by hemoperfusion can cross the BBB and accumulate in cerebral ischemia site, scavenging various free radicals and suppressing inflammation- and immune response-induced injury to achieve neuroprotection after thrombolysis. In addition, the biomimetic nanovesicle could block tPA-induced brain hemorrhage after stroke to improve thrombolytic therapy. The evaluation in ischemic stroke mice confirmed that the simple-prepared nanomedicine with cascaded thrombus targeting, precise thrombolysis and ischemia-reperfusion protection properties can significantly enhance the treatment effect of ischemic stroke. STATEMENT OF SIGNIFICANCE: Ischemic stroke is recognized as a leading cause of death and disability in the world. Rational design of nanomedicine to accelerate thrombolysis and sequentially avoid thrombolysis-mediated reperfusion injury is still a challenge. Herein, a biomimetic nanovesicle (tMP) was developed for sequential ischemic stroke treatment. It could overcome the drawbacks of free tPA for safe thrombolysis: i) platelet membrane biomimetic coating significantly increases thrombus targeting; ii) NIR-mediated photothermal of natural melanin precise controlled release of tPA in thrombus in situ, and local hyperthermia also increases the thrombolytic activity of tPA. Notably, released melanin nanoparticles (4.5 nm) accompanied by hemoperfusion can across BBB and avoid ischemia-reperfusion injury through free radical scavenging and inflammation/immune response suppression.
Collapse
Affiliation(s)
- Wenyan Yu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Na Yin
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Yue Yang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Cuiping Xuan
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Xin Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China
| | - Wei Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China
| | - Zhenzhong Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450001, China
| | - Kaixiang Zhang
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450001, China.
| | - Junjie Liu
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450001, China.
| | - Jinjin Shi
- School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou 450001, China; Key Laboratory of Targeting Therapy and Diagnosis for Critical Diseases, Zhengzhou 450001, China; State Key Laboratory of Esophageal Cancer Prevention and Treatment, Zhengzhou 450001, China.
| |
Collapse
|
32
|
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]
|
33
|
Fukuta T, Oku N, Kogure K. Application and Utility of Liposomal Neuroprotective Agents and Biomimetic Nanoparticles for the Treatment of Ischemic Stroke. Pharmaceutics 2022; 14:pharmaceutics14020361. [PMID: 35214092 PMCID: PMC8877231 DOI: 10.3390/pharmaceutics14020361] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/27/2022] [Accepted: 02/02/2022] [Indexed: 12/04/2022] Open
Abstract
Ischemic stroke is still one of the leading causes of high mortality and severe disability worldwide. Therapeutic options for ischemic stroke and subsequent cerebral ischemia/reperfusion injury remain limited due to challenges associated with drug permeability through the blood-brain barrier (BBB). Neuroprotectant delivery with nanoparticles, including liposomes, offers a promising solution to address this problem, as BBB disruption following ischemic stroke allows nanoparticles to pass through the intercellular gaps between endothelial cells. To ameliorate ischemic brain damage, a number of nanotherapeutics encapsulating neuroprotective agents, as well as surface-modified nanoparticles with specific ligands targeting the injured brain regions, have been developed. Combination therapy with nanoparticles encapsulating neuroprotectants and tissue plasminogen activator (t-PA), a globally approved thrombolytic agent, has been demonstrated to extend the narrow therapeutic time window of t-PA. In addition, the design of biomimetic drug delivery systems (DDS) employing circulating cells (e.g., leukocytes, platelets) with unique properties has recently been investigated to overcome the injured BBB, utilizing these cells’ inherent capability to penetrate the ischemic brain. Herein, we review recent findings on the application and utility of nanoparticle DDS, particularly liposomes, and various approaches to developing biomimetic DDS functionalized with cellular membranes/membrane proteins for the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Tatsuya Fukuta
- Department of Physical Pharmaceutics, School of Pharmaceutical Sciences, Wakayama Medical University, 25-1 Shichiban-cho, Wakayama 640-8156, Japan
| | - Naoto Oku
- Faculty of Pharma-Science, Teikyo University, 2-11-1 Kaga, Itabashi-ku, Tokyo 173-8605, Japan
- Department of Medical Biochemistry, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Kentaro Kogure
- Department of Pharmaceutical Health Chemistry, Graduate School of Biomedical Sciences, Tokushima University, Shomachi 1, Tokushima 770-8505, Japan
| |
Collapse
|
34
|
Yu H, Palazzolo JS, Zhou J, Hu Y, Niego B, Pan S, Ju Y, Wang TY, Lin Z, Hagemeyer CE, Caruso F. Bioresponsive Polyphenol-Based Nanoparticles as Thrombolytic Drug Carriers. ACS APPLIED MATERIALS & INTERFACES 2022; 14:3740-3751. [PMID: 35019268 DOI: 10.1021/acsami.1c19820] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Thrombolytic (clot-busting) therapies with plasminogen activators (PAs) are first-line treatments against acute thrombosis and ischemic stroke. However, limitations such as narrow therapeutic windows, low success rates, and bleeding complications hinder their clinical use. Drug-loaded polyphenol-based nanoparticles (NPs) could address these shortfalls by delivering a more targeted and safer thrombolysis, coupled with advantages such as improved biocompatibility and higher stability in vivo. Herein, a template-mediated polyphenol-based supramolecular assembly strategy is used to prepare nanocarriers of thrombolytic drugs. A thrombin-dependent drug release mechanism is integrated using tannic acid (TA) to cross-link urokinase-type PA (uPA) and a thrombin-cleavable peptide on a sacrificial mesoporous silica template via noncovalent interactions. Following drug loading and template removal, the resulting NPs retain active uPA and demonstrate enhanced plasminogen activation in the presence of thrombin (1.14-fold; p < 0.05). Additionally, they display lower association with macrophage (RAW 264.7) and monocytic (THP-1) cell lines (43 and 7% reduction, respectively), reduced hepatic accumulation, and delayed blood clearance in vivo (90% clearance at 60 min vs 5 min) compared with the template-containing NPs. Our thrombin-responsive, polyphenol-based NPs represent a promising platform for advanced drug delivery applications, with potential to improve thrombolytic therapies.
Collapse
Affiliation(s)
- Haitao Yu
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Jason S Palazzolo
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Jiajing Zhou
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yingjie Hu
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Be'eri Niego
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Shuaijun Pan
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Yi Ju
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Ting-Yi Wang
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Zhixing Lin
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Christoph E Hagemeyer
- NanoBiotechnology Laboratory, Australian Centre for Blood Diseases, Central Clinical School, Monash University, Melbourne, Victoria 3004, Australia
| | - Frank Caruso
- Department of Chemical Engineering, The University of Melbourne, Parkville, Victoria 3010, Australia
| |
Collapse
|
35
|
Li S, Li L, Lin X, Chen C, Luo C, Huang Y. Targeted Inhibition of Tumor Inflammation and Tumor-Platelet Crosstalk by Nanoparticle-Mediated Drug Delivery Mitigates Cancer Metastasis. ACS NANO 2022; 16:50-67. [PMID: 34873906 DOI: 10.1021/acsnano.1c06022] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Sowing malignant cells (the "seeds" of metastasis) to engraft secondary sites requires a conducive premetastatic niche (PMN, the "soil" of metastasis). Inflammation and tumor associated platelet (TAP) has been hijacked by primary tumors to induce PMN "soil" in distant organs, as well as facilitate the dissemination of "seeds". This study reports a combinatory strategy with activated platelet-targeting nanoparticles to aim at the dynamic process of entire cancer metastasis, which exerts robust antimetastasis efficacy by simultaneously inhibiting tumor inflammation and tumor-platelet crosstalk. Our results reveals that the PSN peptide (a P-selectin-targeting peptide) modification enriched the accumulation of nanoparticles in primary tumor, pulmonary PMN, and metastases via capturing activated platelet. Such characteristics contribute to the efficient inhibition on almost every crucial and consecutive step of the metastasis cascade by retarding epithelial-mesenchymal transition (EMT) progression within tumors, specifically blocking the tumor-platelet crosstalk to remove the platelets "protective shield" around disseminated "seeds", and reversing the inflammatory microenvironment to interfere with the "soil" formation. Consisting of inflammation inhibiting and TAP impeding nanoparticles, this approach prominently reduces various metastasis in abscopal lung, including spontaneous metastasis, disseminated tumor cells metastasis, and post-operative metastasis. This work provides a generalizable nanoplatform of parallel inflammation disturbance and tumor-TAP crosstalk blockade to resist metastatic tumors.
Collapse
Affiliation(s)
- Shujie Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, People's Republic of China
| | - Lian Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, People's Republic of China
| | - Xi Lin
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, People's Republic of China
| | - Cheng Chen
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, People's Republic of China
| | - Chaohui Luo
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, People's Republic of China
| | - Yuan Huang
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry and Sichuan Province, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, No. 17, Block 3, South Renmin Road, Chengdu 610041, People's Republic of China
| |
Collapse
|
36
|
Multiphysics Modelling and Simulation of Thrombolysis via Activated Platelet-Targeted Nanomedicine. Pharm Res 2022; 39:41-56. [PMID: 35044591 PMCID: PMC8837543 DOI: 10.1007/s11095-021-03161-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2021] [Accepted: 12/22/2021] [Indexed: 11/01/2022]
Abstract
PURPOSE This study establishes a multiphysics simulation platform for both conventional and targeted thrombolysis using tissue plasminogen activator (tPA). Based on our computational results, the effects of therapeutic parameters on the dynamics of thrombolysis and the risk of side effects are investigated. METHODS The model extends our previously developed one-dimensional(1D) mathematical models for fibrinolysis by incorporating targeted thrombolysis. It consists of two parts: (i) a coupled mathematical model of systemic pharmacokinetics (PK) and pharmacodynamics (PD) and local PD in a 1D occluded artery, and (ii) a mechanistic model for a targeted thrombolytic system via activated platelet-targeted tPA-loaded nanovesicles (tPA-NV), with model parameters derived from our in vitro experiments. A total of 16 therapeutic scenarios are simulated by varying the clot location and composition as well as the dosing regimen with free tPA or tPA-NV. RESULTS Our simulation results indicate that tPA-NV offers several advantages over free tPA for thrombolysis. It reduces systemic exposure of tPA, thereby minimising the risk of bleeding complications. Simulations with different tPA-NV doses reveal that tPA-NV at 10% of the recommended dose can be as effective as the standard regimen with the full recommended dose of free tPA, demonstrating the potential of our tPA-NV as a new thrombolytic strategy with a reduced tPA dose. Moreover, faster recanalisation can be achieved with tPA-NV, especially for platelet-rich(or fibrin-poor) clots. CONCLUSIONS Our simulation platform for thrombolysis with well-tuned model parameters can be used to evaluate and optimise treatment regimens of existing and new thrombolytic therapies via benefit/risk assessment under various therapeutic scenarios.
Collapse
|
37
|
Guo R, Deng M, He X, Li M, Li J, He P, Liu H, Li M, Zhang Z, He Q. Fucoidan-functionalized activated platelet-hitchhiking micelles simultaneously track tumor cells and remodel the immunosuppressive microenvironment for efficient metastatic cancer treatment. Acta Pharm Sin B 2022; 12:467-482. [PMID: 35127399 PMCID: PMC8799858 DOI: 10.1016/j.apsb.2021.05.012] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2021] [Revised: 03/21/2021] [Accepted: 04/19/2021] [Indexed: 12/21/2022] Open
Abstract
Tumor metastasis is responsible for most mortality in cancer patients, and remains a challenge in clinical cancer treatment. Platelets can be recruited and activated by tumor cells, then adhere to circulating tumor cells (CTCs) and assist tumor cells extravasate in distant organs. Therefore, nanoparticles specially hitchhiking on activated platelets are considered to have excellent targeting ability for primary tumor, CTCs and metastasis in distant organs. However, the activated tumor-homing platelets will release transforming growth factor-β (TGF-β), which promotes tumor metastasis and forms immunosuppressive microenvironment. Therefore, a multitalent strategy is needed to balance the accurate tumor tracking and alleviate the immunosuppressive signals. In this study, a fucoidan-functionalized micelle (FD/DOX) was constructed, which could efficiently adhere to activated platelets through P-selectin. Compared with the micelle without P-selectin targeting effect, FD/DOX had increased distribution in both tumor tissue and metastasis niche, and exhibited excellent anti-tumor and anti-metastasis efficacy on 4T1 spontaneous metastasis model. In addition, due to the contribution of fucoidan, FD/DOX treatment was confirmed to inhibit the expression of TGF-β, thereby stimulating anti-tumor immune response and reversing the immunosuppressive microenvironment. The fucoidan-functionalized activated platelets-hitchhiking micelle was promising for the metastatic cancer treatment.
Collapse
|
38
|
|
39
|
Marson D, Aulic S, Fermeglia A, Laurini E, Pricl S. Nanovesicles for the delivery of cardiovascular drugs. APPLICATIONS OF NANOVESICULAR DRUG DELIVERY 2022:341-369. [DOI: 10.1016/b978-0-323-91865-7.00009-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
40
|
Patel D, Wairkar S. Biotechnology-based therapeutics for management of cerebral stroke. Eur J Pharmacol 2021; 913:174638. [PMID: 34801531 DOI: 10.1016/j.ejphar.2021.174638] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 11/05/2021] [Accepted: 11/11/2021] [Indexed: 02/07/2023]
Abstract
Cerebral stroke, commonly caused due to hindrance in blood flow, is broadly classified into two categories-ischemic and haemorrhagic strokes. The onset of stroke triggers multiple mechanisms causing inflammation, generation of free radicals and protein damage leading to apoptosis of neuronal cells. The current therapies available for cerebral strokes involve use of complex surgical treatments and tissue plasminogen activator which increases the risk of internal bleeding, brain edema and cerebral damage, thereby restricting their use in clinical setting. The alarming need to develop safe, effective, target specific systems which, promote neuronal growth and reduce cerebral inflammation can be accomplished with use of biotechnological approaches. The article gives an insight to biotechnology-based advancements for tissue plasminogen activators, cell penetrating peptides, growth factors, ribonucleic acid systems and monoclonal antibodies for cerebral stroke. We also emphasis on challenges and future perspective of biotechnology-based therapeutics for better management of stroke.
Collapse
Affiliation(s)
- Dhrumi Patel
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India
| | - Sarika Wairkar
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKMs NMIMS, V.L.Mehta Road, Vile Parle (W), Mumbai, Maharashtra, 400056, India.
| |
Collapse
|
41
|
Cao W, Liu Y, Ran P, He J, Xie S, Weng J, Li X. Ultrasound-Propelled Janus Rod-Shaped Micromotors for Site-Specific Sonodynamic Thrombolysis. ACS APPLIED MATERIALS & INTERFACES 2021; 13:58411-58421. [PMID: 34846117 DOI: 10.1021/acsami.1c19288] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Antithrombosis therapy is confronted with short half-lives of thrombolytic agents, limited therapeutic effects, and bleeding complications. Drug delivery systems of thrombolytic agents face challenges in effective penetration into thrombi, which are characterized by well-organized fibrin filled with abundant activated platelets. Herein, Janus rod (JR)-shaped micromotors are constructed by side-by-side electrospinning and cryosection, possessing advantages in controlling the Janus structure and aspect ratio of microrods. Silicon phthalocyanine (Pc) and CaO2 nanoparticles (NPs) are loaded into the separate sides of JRs, and Arg-Gly-Asp (RGD) peptides are grafted on the surface to obtain Pc/Ca@r-JRs for the sonodynamic therapy (SDT) of thrombosis without using any thrombolytic agents. Decomposition of CaO2 NPs ejects O2 bubbles from one side of JRs, and ultrasonication of O2 bubbles produces the cavitation effect, both generating mechanical force to drive the thrombus penetration. The integration of ultrasonication-propelled motion and RGD mediation effectively increases the targeting capabilities of r-JRs to activated platelets. In addition to mechanical thrombolysis, ultrasonication of the released Pc produces 1O2 to destruct fibrin networks of clots. In vitro thrombolysis of whole blood clots shows that ultrasonication of Pc/Ca@r-JRs has a significantly higher thrombolysis rate (73.6%) than those without propelled motion or RGD-mediated clot targeting. In a lower limb thrombosis model, intravenous administration of Pc/Ca@r-JRs indicates 3.4-fold higher accumulations at the clot site than those of JRs, and ultrasonication-propelled motion further increases thrombus retention 2.1 times. Treatment with Pc/Ca@r-JRs and ultrasonication fully removes thrombi and significantly prolongs tail bleeding time. Thus, this study has achieved precise and prompt thrombolysis through selective targeting to clots, efficient penetration into dense networks of thrombi, and SDT-executed thrombolysis.
Collapse
Affiliation(s)
- Wenxiong Cao
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Yuan Liu
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Pan Ran
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Jie He
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Shuang Xie
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Jie Weng
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| | - Xiaohong Li
- Key Laboratory of Advanced Technologies of Materials, Ministry of Education, School of Materials Science and Engineering, Southwest Jiaotong University, Chengdu 610031, P. R. China
| |
Collapse
|
42
|
Tian H, Lin L, Ba Z, Xue F, Li Y, Zeng W. Nanotechnology combining photoacoustic kinetics and chemical kinetics for thrombosis diagnosis and treatment. CHINESE CHEM LETT 2021. [DOI: 10.1016/j.cclet.2021.05.070] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
43
|
Guan Q, Dou H. Thrombus-Targeting Polymeric Nanocarriers and Their Biomedical Applications in Thrombolytic Therapy. Front Physiol 2021; 12:763085. [PMID: 34916956 PMCID: PMC8669757 DOI: 10.3389/fphys.2021.763085] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 11/08/2021] [Indexed: 01/08/2023] Open
Abstract
Due to the high morbidity and mortality of cardiovascular diseases, there is an urgent need for research on antithrombotic strategies. In view of the short half-life, insufficient drug penetration, poor targeting capabilities, and hemorrhagic side-effects of traditional thrombus treatment methods, the combination of thrombolytic therapy and nanocarriers brought by the development of nanotechnology in recent years may provide effective solutions for these undesirable side-effects caused by insufficient targeting. Polymeric nanocarriers, based on macromolecules and various functional groups, can connect specific targeting molecules together through chemical modification to achieve the protection and targeted delivery of thrombolytic drugs. However, simple chemical molecular modifications may be easily affected by the physiological environment encountered in the circulatory system. Therefore, the modification of nanocarriers with cell membranes can provide camouflage to these platforms and help to extend their circulation time while also imparting them with the biological functions of cell membranes, thus providing them with precise targeting capabilities, among which the most important is the biological modification of platelet membranes. In addition, some nanoparticles with their own therapeutic functions have also been developed, such as polypyrrole, which can exhibit a photothermal effect to induce thrombolysis. Herein, combined with the mechanism of thrombosis and thrombolysis, we outline the recent advances achieved with thrombus-targeting nanocarriers with regard to thrombosis treatment. On this basis, the design considerations, advantages, and challenges of these thrombolytic therapies in clinical transformation are discussed.
Collapse
Affiliation(s)
| | - Hongjing Dou
- State Key Laboratory of Metal Matrix Composites, School of Materials Science and Engineering, Shanghai Jiao Tong University, Shanghai, China
| |
Collapse
|
44
|
Adzerikho IE, Vladimirskaya TE, Lutsik IL, Dubatouka KI, Agabekov VE, Branovitskaya ES, Chernyavsky EA, Lugovska N. Fibrinspecific liposomes as a potential method of delivery of the thrombolytic preparation streptokinase. J Thromb Thrombolysis 2021; 53:313-320. [PMID: 34816379 DOI: 10.1007/s11239-021-02614-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/14/2021] [Indexed: 11/26/2022]
Abstract
The use of streptokinase (SK) in the clinic is limited by the lack of fibrin-specificity and the short half-life of the drug. We have developed a new dosage form of streptokinase (immunoliposome), which consists of "free" native streptokinase and "bound" encapsulated in liposomes conjugated through carboxylated dextran with fibrin-specific monoclonal antibodies FnI-3C (IgG2 class), in a ratio of 60 and 40%, respectively, and studied their physicochemical properties, pharmacokinetic parameters, and the ability of fibrin-specific liposomes with SK for targeted delivery to fibrin in an in vivo experiment. The obtained immunoliposomes had a hydrodynamic diameter of ~ 140 nm, a zeta potential of - 19.6 mV, and entrapment efficiency of 14.1%. Fluorescent labels bound to immunoliposomes with streptokinase selectively accumulated in model rat vein thrombi at sites containing fibrin in 30 min after injection. Studies of pharmacokinetic parameters showed that the administration of immunoliposomes with streptokinase to rats was accompanied by an increase in the half-life from 1.8 to 24.1 min, the time to reach the maximum concentration from 15 to 30 min, and a decrease in the elimination constant by about 13 times compared with the native streptokinase preparation. Further studies are needed to evaluate the thrombolytic efficacy a new dosage form of streptokinase in experiment in vivo.
Collapse
Affiliation(s)
- I E Adzerikho
- Belarusian Medical Academy of Postgraduate Education, Minsk, Belarus
| | - T E Vladimirskaya
- Belarusian Medical Academy of Postgraduate Education, Minsk, Belarus
| | - I L Lutsik
- Belarusian Medical Academy of Postgraduate Education, Minsk, Belarus
| | - K I Dubatouka
- Institute of Chemistry of New Materials of the National Academy of Sciences of Belarus, Minsk, Belarus.
| | - V E Agabekov
- Institute of Chemistry of New Materials of the National Academy of Sciences of Belarus, Minsk, Belarus
| | - E S Branovitskaya
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus
| | - E A Chernyavsky
- Research Institute for Physical Chemical Problems of the Belarusian State University, Minsk, Belarus
| | - N Lugovska
- Palladin Institute of Biochemistry of the National Academy of Sciences of Ukraine, Kyiv, Ukraine
| |
Collapse
|
45
|
Chang LH, Chuang EY, Cheng TM, Lin C, Shih CM, Wu AT, Jheng PR, Lu HY, Shih CC, Mi FL. Thrombus-specific theranostic nanocomposite for codelivery of thrombolytic drug, algae-derived anticoagulant and NIR fluorescent contrast agent. Acta Biomater 2021; 134:686-701. [PMID: 34358695 DOI: 10.1016/j.actbio.2021.07.072] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/13/2021] [Accepted: 07/29/2021] [Indexed: 12/12/2022]
Abstract
Thrombolysis is a standard treatment for rapidly restoring blood flow. However, the application of urokinase-type plasminogen activator (Uk) in clinical therapy is limited due to its nonspecific distribution and inadequate therapeutic accumulation. Precise thrombus imaging and site-specific drug delivery can enhance the diagnostic and therapeutic efficacy for thrombosis. Accordingly, we developed a P-selectin-specific, photothermal theranostic nanocomposite for thrombus-targeted codelivery of Uk and indocyanine green (ICG, a contrast agent for near-infrared (NIR) fluorescence imaging). We evaluated its capabilities for thrombus imaging and enzyme/hyperthermia combined thrombolytic therapy. Mesoporous silica-coated gold nanorods (Si-AuNRs) were functionalized with an arginine-rich peptide to create an organic template for the adsorption of ICG and fucoidan (Fu), an algae-derived anticoagulant. Uk was loaded into the SiO2 pores of the Si-AuNRs through the formation of a Fu-Uk-ICG complex on the peptide-functionalized template. The Fu-Uk/ICG@SiAu NRs nanocomposite increased the photostability of ICG and improved its targeting/accumulation at blood clot sites with a strong NIR fluorescence intensity for precise thrombus imaging. Furthermore, ICG incorporated into the nanocomposite enhanced the photothermal effect of Si-AuNRs. Fu, as a P-selectin-targeting ligand, enabled the nanocomposite to target a thrombus site where platelets were activated. The nanocomposite enabled a faster release of Uk for rapid clearing of blood clots and a slower release of Fu for longer lasting prevention of thrombosis regeneration. The nanocomposite with multiple functions, including thrombus-targeting drug delivery, photothermal thrombolysis, and NIR fluorescence imaging, is thus an advanced theranostic platform for thrombolytic therapy with reduced hemorrhaging risk and enhanced imaging/thrombolysis efficiency. STATEMENT OF SIGNIFICANCE: Herein, for the first time, a P-selectin specific, photothermal theranostic nanocomposite for thrombus-targeted co-delivery of urokinase and NIR fluorescence contrast agent indocyanine green (ICG) was developed. We evaluated the potential of this theranostic nanocomposite for thrombus imaging and enzyme/hyperthermia combined thrombolytic therapy. The nanocomposite showed multiple functions including thrombus targeting and imaging, and photothermal thrombolysis. Besides, it allowed faster release of the thrombolytic urokinase for rapidly clearing blood clots and slower release of a brown algae-derived anticoagulant fucoidan (also acting as a P-selectin ligand) for prevention of thrombosis regeneration. The nanocomposite is thus a new and advanced theranostic platform for targeted thrombolytic therapy.
Collapse
Affiliation(s)
- Lee-Hsin Chang
- School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Er-Yuan Chuang
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Tsai-Mu Cheng
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Chi Lin
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Chun-Ming Shih
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan; Division of Cardiology, Department of Internal Medicine, Taipei Medical University Hospital, Taipei 11031, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan
| | - Alexander Th Wu
- The PhD Program for Translational Medicine, College of Medical Science and Technology, Taipei Medical University, Taipei 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan
| | - Pei-Ru Jheng
- Graduate Institute of Biomedical Materials and Tissue Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan
| | - Hsin-Ying Lu
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan; Institute of Clinical Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan
| | - Chun-Che Shih
- Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan; Division of Cardiovascular Surgery, Department of Surgery, Wan Fang Hospital, Taipei Medical University, Taipei 11031, Taiwan; Institute of Clinical Medicine, School of Medicine, National Yang-Ming Chiao Tung University, Taipei, Taiwan; Department of Surgery, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan.
| | - Fwu-Long Mi
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Taipei Heart Institute, Taipei Medical University, Taipei 11031, Taiwan; Department of Biochemistry and Molecular Cell Biology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Graduate Institute of Nanomedicine and Medical Engineering, College of Biomedical Engineering, Taipei Medical University, Taipei 11031, Taiwan.
| |
Collapse
|
46
|
Pan Q, Xu J, Wen CJ, Xiong YY, Gong ZT, Yang YJ. Nanoparticles: Promising Tools for the Treatment and Prevention of Myocardial Infarction. Int J Nanomedicine 2021; 16:6719-6747. [PMID: 34621124 PMCID: PMC8491866 DOI: 10.2147/ijn.s328723] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
Despite several recent advances, current therapy and prevention strategies for myocardial infarction are far from satisfactory, owing to limitations in their applicability and treatment effects. Nanoparticles (NPs) enable the targeted and stable delivery of therapeutic compounds, enhance tissue engineering processes, and regulate the behaviour of transplants such as stem cells. Thus, NPs may be more effective than other mechanisms, and may minimize potential adverse effects. This review provides evidence for the view that function-oriented systems are more practical than traditional material-based systems; it also summarizes the latest advances in NP-based strategies for the treatment and prevention of myocardial infarction.
Collapse
Affiliation(s)
- Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Cen-Jin Wen
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhao-Ting Gong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
47
|
Near-infrared light-responsive liposomes for protein delivery: Towards bleeding-free photothermally-assisted thrombolysis. J Control Release 2021; 337:212-223. [PMID: 34284049 DOI: 10.1016/j.jconrel.2021.07.024] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 07/11/2021] [Accepted: 07/14/2021] [Indexed: 11/23/2022]
Abstract
Smart drug delivery systems represent state-of-the-art approaches for targeted therapy of life-threatening diseases such as cancer and cardiovascular diseases. Stimuli-responsive on-demand release of therapeutic agents at the diseased site can significantly limit serious adverse effects. In this study, we engineered a near-infrared (NIR) light-responsive liposomal gold nanorod-containing platform for on-demand delivery of proteins using a hybrid formulation of ultrasmall gold nanorods (AuNRs), thermosensitive phospholipid (DPPC) and non-ionic surfactant (Brij58). In light-triggered release optimization studies, 55.6% (± 4.8) of a FITC-labelled model protein, ovalbumin (MW 45 kDa) was released in 15 min upon NIR irradiation (785 nm, 1.35 W/cm2 for 5 min). This platform was then utilized to test on-demand delivery of urokinase-plasminogen activator (uPA) for bleeding-free photothermally-assisted thrombolysis, where the photothermal effect of AuNRs would synergize with the released uPA in clot lysis. Urokinase light-responsive liposomes showed 80.7% (± 4.5) lysis of an in vitro halo-clot model in 30 min following NIR irradiation (785 nm, 1.35 W/cm2 for 5 min) compared to 36.3% (± 4.4) and 15.5% (± 5.5) clot lysis from equivalent free uPA and non-irradiated liposomes respectively. These results show the potential of low-dose, site-specific thrombolysis via the combination of light-triggered delivery/release of uPA from liposomes combined with photothermal thrombolytic effects from gold nanorods. In conclusion, newly engineered, gold nanorod-based, NIR light-responsive liposomes represent a promising drug delivery system for site-directed, photothermally-stimulated therapeutic protein release.
Collapse
|
48
|
Huang Y, Gu B, Salles-Crawley II, Taylor KA, Yu L, Ren J, Liu X, Emerson M, Longstaff C, Hughes AD, Thom SA, Xu XY, Chen R. Fibrinogen-mimicking, multiarm nanovesicles for human thrombus-specific delivery of tissue plasminogen activator and targeted thrombolytic therapy. SCIENCE ADVANCES 2021; 7:7/23/eabf9033. [PMID: 34078604 PMCID: PMC8172176 DOI: 10.1126/sciadv.abf9033] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Accepted: 04/14/2021] [Indexed: 05/03/2023]
Abstract
Clinical use of tissue plasminogen activator (tPA) in thrombolytic therapy is limited by its short circulation time and hemorrhagic side effects. Inspired by fibrinogen binding to activated platelets, we report a fibrinogen-mimicking, multiarm nanovesicle for thrombus-specific tPA delivery and targeted thrombolysis. This biomimetic system is based on the lipid nanovesicle coated with polyethylene glycol (PEG) terminally conjugated with a cyclic RGD (cRGD) peptide. Our experiments with human blood demonstrated its highly selective binding to activated platelets and efficient tPA release at a thrombus site under both static and physiological flow conditions. Its clot dissolution time in a microfluidic system was comparable to that of free tPA. Furthermore, we report a purpose-built computational model capable of simulating targeted thrombolysis of the tPA-loaded nanovesicle and with a potential in predicting the dynamics of thrombolysis in physiologically realistic scenarios. This combined experimental and computational work presents a promising platform for development of thrombolytic nanomedicines.
Collapse
Affiliation(s)
- Yu Huang
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Boram Gu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
- School of Chemical Engineering, Chonnam National University, Gwangju, Republic of Korea
| | - Isabelle I Salles-Crawley
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, Hammersmith Hospital Campus, London, UK
| | - Kirk A Taylor
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Li Yu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Jie Ren
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Xuhan Liu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK
| | - Michael Emerson
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Colin Longstaff
- Biotherapeutics Section, National Institute for Biological Standards and Control, South Mimms, Herts, UK
| | - Alun D Hughes
- Institute of Cardiovascular Science, University College London, London, UK
- MRC Unit for Lifelong Health and Ageing at University College London, London, UK
| | - Simon A Thom
- National Heart and Lung Institute, Imperial College London, London, UK
| | - Xiao Yun Xu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK.
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London, UK.
| |
Collapse
|
49
|
Refaat A, del Rosal B, Palasubramaniam J, Pietersz G, Wang X, Peter K, Moulton SE. Smart Delivery of Plasminogen Activators for Efficient Thrombolysis; Recent Trends and Future Perspectives. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100047] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Ahmed Refaat
- Department of Telecommunications, Electrical, Robotics and Biomedical Engineering, Faculty of Science, Engineering and Technology Swinburne University of Technology John St Melbourne VIC 3122 Australia
- Atherothrombosis and Vascular Biology Laboratory Baker Heart and Diabetes Institute 75 Commercial Road Melbourne VIC 3004 Australia
- Molecular Imaging and Theranostics Laboratory Baker Heart and Diabetes Institute 75 Commercial Road Melbourne VIC 3004 Australia
- Pharmaceutics Department Faculty of Pharmacy ‐ Alexandria University 1 El‐Khartoum Square Azarita Alexandria 21521 Egypt
| | - Blanca del Rosal
- ARC Centre of Excellence for Nanoscale BioPhotonics School of Science RMIT University 124 La Trobe St Melbourne VIC 3000 Australia
| | - Jathushan Palasubramaniam
- Atherothrombosis and Vascular Biology Laboratory Baker Heart and Diabetes Institute 75 Commercial Road Melbourne VIC 3004 Australia
- Molecular Imaging and Theranostics Laboratory Baker Heart and Diabetes Institute 75 Commercial Road Melbourne VIC 3004 Australia
- Department of Medicine Monash University 27 Rainforest Walk Melbourne VIC 3800 Australia
- Department of Cardiology Alfred Hospital 55 Commercial Rd Melbourne VIC 3004 Australia
| | - Geoffrey Pietersz
- Atherothrombosis and Vascular Biology Laboratory Baker Heart and Diabetes Institute 75 Commercial Road Melbourne VIC 3004 Australia
- Burnet Institute 85 Commercial Road Melbourne VIC 3004 Australia
| | - Xiaowei Wang
- Atherothrombosis and Vascular Biology Laboratory Baker Heart and Diabetes Institute 75 Commercial Road Melbourne VIC 3004 Australia
- Molecular Imaging and Theranostics Laboratory Baker Heart and Diabetes Institute 75 Commercial Road Melbourne VIC 3004 Australia
- Department of Medicine Monash University 27 Rainforest Walk Melbourne VIC 3800 Australia
- Department of Cardiometabolic Health University of Melbourne Melbourne VIC 3010 Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory Baker Heart and Diabetes Institute 75 Commercial Road Melbourne VIC 3004 Australia
- Department of Medicine Monash University 27 Rainforest Walk Melbourne VIC 3800 Australia
- Department of Cardiology Alfred Hospital 55 Commercial Rd Melbourne VIC 3004 Australia
- Department of Cardiometabolic Health University of Melbourne Melbourne VIC 3010 Australia
| | - Simon E. Moulton
- Department of Telecommunications, Electrical, Robotics and Biomedical Engineering, Faculty of Science, Engineering and Technology Swinburne University of Technology John St Melbourne VIC 3122 Australia
- ARC Centre of Excellence for Electromaterials Science Swinburne University of Technology John St Melbourne VIC 3122 Australia
- Aikenhead Centre for Medical Discovery (ACMD) St Vincent's Hospital Melbourne VIC 3065 Australia
- Iverson Health Innovation Research Institute Swinburne University of Technology John St Melbourne VIC 3122 Australia
- Australian Institute for Innovative Materials, Intelligent Polymer Research Institute University of Wollongong Wollongong NSW 2500 Australia
| |
Collapse
|
50
|
Zhao Y, Xie R, Yodsanit N, Ye M, Wang Y, Gong S. Biomimetic fibrin-targeted and H 2O 2-responsive nanocarriers for thrombus therapy. NANO TODAY 2020; 35:100986. [PMID: 33072177 PMCID: PMC7561002 DOI: 10.1016/j.nantod.2020.100986] [Citation(s) in RCA: 74] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Thrombosis is a principle cause of various life-threatening cardiovascular diseases. However, current antithrombotic treatments using drugs only offer limited efficacy due to short half-life, low targeting ability to the thrombus site, and unexpected bleeding complications. Taking into account of the biological characteristics of thrombus including upregulation of hydrogen peroxide (H2O2) and abundance of fibrin, we engineered a H2O2-responsive nanocarrier for thrombus-targeting delivery of an antithrombotic agent (i.e., tirofiban). The nanocarrier was composed of a drug-conjugated dextran nanocore and a red blood cell (RBC) membrane shell, and its surface was functionalized with a fibrin-targeting peptide, CREKA. Tirofiban was conjugated to dextran through a H2O2-cleavable phenylboronic ester linkage. The fibrin-targeting RBC membrane-cloaked dextran-tirofiban conjugate nanoparticles (i.e., T-RBC-DTC NPs) can scavenge H2O2 and provide controlled release of tirofiban to achieve site-specific antithrombotic effects. In RAW 264.7 cells and HUVECs, the T-RBC-DTC NPs effectively scavenged H2O2 and protected cells from H2O2-induced cytotoxicity. In the ferric chloride-induced carotid thrombosis mouse model, the T-RBC-DTC NPs efficiently accumulated at the injured carotid artery and exhibited significantly enhanced antithrombotic activity compared to free drug. The T-RBC-DTC NPs also exhibited good biocompatibility according to histology analysis. Overall, our results indicated that this bioengineered nanocarrier offers a promising therapeutic strategy for thrombotic disorders.
Collapse
Affiliation(s)
- Yi Zhao
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Ruosen Xie
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Nisakorn Yodsanit
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Mingzhou Ye
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yuyuan Wang
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Shaoqin Gong
- Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Material Science and Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53715, USA
- Corresponding author. Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA. (S. Gong)
| |
Collapse
|