1
|
Zuberi HA, Lal M, Verma S, Chamkha AJ, Zainal NA. Impact of gold and silver nanoparticles injected in blood with viscous dissipation. Comput Methods Biomech Biomed Engin 2025:1-25. [PMID: 40312656 DOI: 10.1080/10255842.2025.2495893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 02/14/2025] [Accepted: 04/13/2025] [Indexed: 05/03/2025]
Abstract
A nano-blood model is developed to study the flow of gold- and silver-infused blood through a porous, stenotic artery under Newtonian assumptions. Wall curvature, convective heating, wall motion, and viscous dissipation are considered. Darcy's model simulates porous resistance, and the Tiwari-Das model captures nanoparticle effects. Governing equations are reduced via similarity transformations and solved using MATLAB's bvp4c solver. Validation against existing studies is provided. Results show gold-blood nanofluid achieves higher velocities than silver-blood. Increasing the Biot number enhances cooling at the arterial wall. Detailed graphs and 3D contour plots illustrate the effects on temperature, velocity, skin friction, and Nusselt number.
Collapse
Affiliation(s)
- Haris Alam Zuberi
- Department of Applied Mathematics, M. J. P. Rohilkhand University, Bareilly, India
| | - Madan Lal
- Department of Applied Mathematics, M. J. P. Rohilkhand University, Bareilly, India
| | - Shivangi Verma
- Department of Applied Mathematics, M. J. P. Rohilkhand University, Bareilly, India
| | - Ali J Chamkha
- Faculty of Engineering, Kuwait College of Science and Technology, Doha District, Kuwait
| | - Nurul Amira Zainal
- Fakulti Teknologi dan Kejuruteraan Mekanikal, Universiti Teknikal Malaysia Melaka, Hang Tuah Jaya, 76100 Durian Tunggal, Melaka, Malaysia
| |
Collapse
|
2
|
Naskar A, Kilari S, Baranwal G, Kane J, Misra S. Nanoparticle-Based Drug Delivery for Vascular Applications. Bioengineering (Basel) 2024; 11:1222. [PMID: 39768040 PMCID: PMC11673055 DOI: 10.3390/bioengineering11121222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 01/05/2025] Open
Abstract
Nanoparticle (NP)-based drug delivery systems have received widespread attention due to the excellent physicochemical properties of nanomaterials. Different types of NPs such as lipid NPs, poly(lactic-co-glycolic) acid (PLGA) NPs, inorganic NPs (e.g., iron oxide and Au), carbon NPs (graphene and carbon nanodots), 2D nanomaterials, and biomimetic NPs have found favor as drug delivery vehicles. In this review, we discuss the different types of customized NPs for intravascular drug delivery, nanoparticle behaviors (margination, adhesion, and endothelium uptake) in blood vessels, and nanomaterial compatibility for successful drug delivery. Additionally, cell surface protein targets play an important role in targeted drug delivery, and various vascular drug delivery studies using nanoparticles conjugated to these proteins are reviewed. Finally, limitations, challenges, and potential solutions for translational research regarding NP-based vascular drug delivery are discussed.
Collapse
Affiliation(s)
| | | | | | | | - Sanjay Misra
- Vascular and Interventional Radiology Translational Laboratory, Department of Radiology, Mayo Clinic, Rochester, MN 55905, USA; (A.N.); (S.K.); (G.B.); (J.K.)
| |
Collapse
|
3
|
Zhao L, Feng L, Shan R, Huang Y, Shen L, Fan M, Wang Y. Nanoparticle-based approaches for treating restenosis after vascular injury. Front Pharmacol 2024; 15:1427651. [PMID: 39512830 PMCID: PMC11540800 DOI: 10.3389/fphar.2024.1427651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2024] [Accepted: 10/14/2024] [Indexed: 11/15/2024] Open
Abstract
Percutaneous coronary intervention (PCI) is currently the main method for treating coronary artery stenosis, but the incidence of restenosis after PCI is relatively high. Restenosis, the narrowing of blood vessels by more than 50% of the normal diameter after PCI, severely compromises the therapeutic efficacy. Therefore, preventing postinterventional restenosis is important. Vascular restenosis is mainly associated with endothelial injury, the inflammatory response, the proliferation and migration of vascular smooth muscle cells (VSMCs), excessive deposition of extracellular matrix (ECM) and intimal hyperplasia (IH) and is usually prevented by administering antiproliferative or anti-inflammatory drugs through drug-eluting stents (DESs); however, DESs can lead to uncontrolled drug release. In addition, as extracorporeal implants, they can cause inflammation and thrombosis, resulting in suboptimal treatment. Therefore, there is an urgent need for a drug carrier with controlled drug release and high biocompatibility for in vivo drug delivery to prevent restenosis. The development of nanotechnology has enabled the preparation of nanoparticle drug carriers with low toxicity, high drug loading, high biocompatibility, precise targeting, controlled drug release and excellent intracellular delivery ability. This review summarizes the advantages of nanoparticle drug carriers for treating vascular restenosis, as well as how nanoparticles have improved targeting, slowed the release of therapeutic agents, and prolonged circulation in vivo to prevent vascular restenosis more effectively. The overall purpose of this review is to present an overview of nanoparticle therapy for vascular restenosis. We expect these findings to provide insight into nanoparticle-based therapeutic approaches for vascular restenosis.
Collapse
Affiliation(s)
- Liangfeng Zhao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Liuliu Feng
- Department of Cardiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Rong Shan
- Department of Cardiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| | - Yue Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Li Shen
- Department of Cardiology, Zhongshan Hospital, Shanghai Institute of Cardiovascular Diseases, Fudan University, Shanghai, China
| | - Mingliang Fan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Yu Wang
- Department of Cardiology, Shidong Hospital Affiliated to University of Shanghai for Science and Technology, Shanghai, China
| |
Collapse
|
4
|
Makharadze D, Kantaria T, Yousef I, del Valle LJ, Katsarava R, Puiggalí J. PEGylated Micro/Nanoparticles Based on Biodegradable Poly(Ester Amides): Preparation and Study of the Core-Shell Structure by Synchrotron Radiation-Based FTIR Microspectroscopy and Electron Microscopy. Int J Mol Sci 2024; 25:6999. [PMID: 39000109 PMCID: PMC11241343 DOI: 10.3390/ijms25136999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 06/18/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Surface modification of drug-loaded particles with polyethylene glycol (PEG) chains is a powerful tool that promotes better transport of therapeutic agents, provides stability, and avoids their detection by the immune system. In this study, we used a new approach to synthesize a biodegradable poly(ester amide) (PEA) and PEGylating surfactant. These were employed to fabricate micro/nanoparticles with a core-shell structure. Nanoparticle (NP)-protein interactions and self-assembling were subsequently studied by synchrotron radiation-based FTIR microspectroscopy (SR-FTIRM) and transmission electron microscopy (TEM) techniques. The core-shell structure was identified using IR absorption bands of characteristic chemical groups. Specifically, the stretching absorption band of the secondary amino group (3300 cm-1) allowed us to identify the poly(ester amide) core, while the band at 1105 cm-1 (C-O-C vibration) was useful to demonstrate the shell structure based on PEG chains. By integration of absorption bands, a 2D intensity map of the particle was built to show a core-shell structure, which was further supported by TEM images.
Collapse
Affiliation(s)
- Davit Makharadze
- Departament de Enginyeria Química, Universitat Politècnica de Catalunya, EEBE, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain; (D.M.); (L.J.d.V.)
| | - Temur Kantaria
- Institute of Chemistry and Molecular Engineering, Agricultural University of Georgia, Tbilisi 0159, Georgia; (T.K.); (R.K.)
| | - Ibraheem Yousef
- ALBA Synchrotron Light Facility, Carrer de la Llum 2-26, Cerdanyola del Vallès, 08290 Barcelona, Spain;
| | - Luis J. del Valle
- Departament de Enginyeria Química, Universitat Politècnica de Catalunya, EEBE, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain; (D.M.); (L.J.d.V.)
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Campus Diagonal-Besòs, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain
| | - Ramaz Katsarava
- Institute of Chemistry and Molecular Engineering, Agricultural University of Georgia, Tbilisi 0159, Georgia; (T.K.); (R.K.)
| | - Jordi Puiggalí
- Departament de Enginyeria Química, Universitat Politècnica de Catalunya, EEBE, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain; (D.M.); (L.J.d.V.)
- Barcelona Research Center in Multiscale Science and Engineering, Universitat Politècnica de Catalunya, Campus Diagonal-Besòs, Av. Eduard Maristany 10-14, 08019 Barcelona, Spain
| |
Collapse
|
5
|
Pang ASR, Dinesh T, Pang NYL, Dinesh V, Pang KYL, Yong CL, Lee SJJ, Yip GW, Bay BH, Srinivasan DK. Nanoparticles as Drug Delivery Systems for the Targeted Treatment of Atherosclerosis. Molecules 2024; 29:2873. [PMID: 38930939 PMCID: PMC11206617 DOI: 10.3390/molecules29122873] [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: 04/11/2024] [Revised: 06/03/2024] [Accepted: 06/10/2024] [Indexed: 06/28/2024] Open
Abstract
Atherosclerosis continues to be a leading cause of morbidity and mortality globally. The precise evaluation of the extent of an atherosclerotic plaque is essential for forecasting its likelihood of causing health concerns and tracking treatment outcomes. When compared to conventional methods used, nanoparticles offer clear benefits and excellent development opportunities for the detection and characterisation of susceptible atherosclerotic plaques. In this review, we analyse the recent advancements of nanoparticles as theranostics in the management of atherosclerosis, with an emphasis on applications in drug delivery. Furthermore, the main issues that must be resolved in order to advance clinical utility and future developments of NP research are discussed. It is anticipated that medical NPs will develop into complex and advanced next-generation nanobotics that can carry out a variety of functions in the bloodstream.
Collapse
Affiliation(s)
- Alexander Shao-Rong Pang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Tarini Dinesh
- Department of Medicine, Government Kilpauk Medical College, Chennai 600010, Tamilnadu, India;
| | - Natalie Yan-Lin Pang
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Vishalli Dinesh
- Department of Pathology, Dhanalakshmi Srinivasan Medical College Hospital, Perambalur 621113, Tamilnadu, India;
| | - Kimberley Yun-Lin Pang
- Division of Medicine, South Australia Health, Northern Adelaide Local Health Network, Adelaide, SA 5112, Australia; (K.Y.-L.P.); (S.J.J.L.)
| | - Cai Ling Yong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117597, Singapore; (A.S.-R.P.); (N.Y.-L.P.); (C.L.Y.)
| | - Shawn Jia Jun Lee
- Division of Medicine, South Australia Health, Northern Adelaide Local Health Network, Adelaide, SA 5112, Australia; (K.Y.-L.P.); (S.J.J.L.)
| | - George W. Yip
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| | - Boon Huat Bay
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| | - Dinesh Kumar Srinivasan
- Department of Anatomy, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117594, Singapore; (G.W.Y.); (B.H.B.)
| |
Collapse
|
6
|
Tripathi S, Rani K, Raj VS, Ambasta RK. Drug repurposing: A multi targetted approach to treat cardiac disease from existing classical drugs to modern drug discovery. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2024; 207:151-192. [PMID: 38942536 DOI: 10.1016/bs.pmbts.2024.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Cardiovascular diseases (CVDs) are characterized by abnormalities in the heart, blood vessels, and blood flow. CVDs comprise a diverse set of health issues. There are several types of CVDs like stroke, endothelial dysfunction, thrombosis, atherosclerosis, plaque instability and heart failure. Identification of a new drug for heart disease takes longer duration and its safety efficacy test takes even longer duration of research and approval. This chapter explores drug repurposing, nano-therapy, and plant-based treatments for managing CVDs from existing drugs which saves time and safety issues with testing new drugs. Existing drugs like statins, ACE inhibitor, warfarin, beta blockers, aspirin and metformin have been found to be useful in treating cardiac disease. For better drug delivery, nano therapy is opening new avenues for cardiac research by targeting interleukin (IL), TNF and other proteins by proteome interactome analysis. Nanoparticles enable precise delivery to atherosclerotic plaques, inflammation areas, and damaged cardiac tissues. Advancements in nano therapeutic agents, such as drug-eluting stents and drug-loaded nanoparticles are transforming CVDs management. Plant-based treatments, containing phytochemicals from Botanical sources, have potential cardiovascular benefits. These phytochemicals can mitigate risk factors associated with CVDs. The integration of these strategies opens new avenues for personalized, effective, and minimally invasive cardiovascular care. Altogether, traditional drugs, phytochemicals along with nanoparticles can revolutionize the future cardiac health care by identifying their signaling pathway, mechanism and interactome analysis.
Collapse
Affiliation(s)
- Shyam Tripathi
- Centre for Drug Design Discovery and Development (C4D), Department of Biotechnology and Microbiology, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
| | - Kusum Rani
- Centre for Drug Design Discovery and Development (C4D), Department of Biotechnology and Microbiology, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India
| | - V Samuel Raj
- Centre for Drug Design Discovery and Development (C4D), Department of Biotechnology and Microbiology, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India.
| | - Rashmi K Ambasta
- Centre for Drug Design Discovery and Development (C4D), Department of Biotechnology and Microbiology, SRM University, Delhi-NCR, Rajiv Gandhi Education City, Sonepat, Haryana, India.
| |
Collapse
|
7
|
Ullah A, Ullah M, Lim SI. Recent advancements in nanotechnology based drug delivery for the management of cardiovascular disease. Curr Probl Cardiol 2024; 49:102396. [PMID: 38266693 DOI: 10.1016/j.cpcardiol.2024.102396] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Cardiovascular diseases (CVDs) constitute a predominant cause of both global mortality and morbidity. To address the challenges in the early diagnosis and management of CVDs, there is growing interest in the field of nanotechnology and nanomaterials to develop innovative diagnostic and therapeutic approaches. This review focuses on the recent advancements in nanotechnology-based diagnostic techniques, including cardiac immunoassays (CIA), cardiac circulating biomarkers, cardiac exosomal biomarkers, and molecular Imaging (MOI). Moreover, the article delves into the exciting developments in nanoparticles (NPs), biomimetic NPs, nanofibers, nanogels, and nanopatchs for cardiovascular applications. And discuss how these nanoscale technologies can improve the precision, sensitivity, and speed of CVD diagnosis and management. While highlighting their vast potential, we also address the limitations and challenges that must be overcome to harness these innovations successfully. Furthermore, this review focuses on the emerging opportunities for personalized and effective cardiovascular care through the integration of nanotechnology, ultimately aiming to reduce the global burden of CVDs.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea.
| |
Collapse
|
8
|
Islam P, Schaly S, Abosalha AK, Boyajian J, Thareja R, Ahmad W, Shum-Tim D, Prakash S. Nanotechnology in development of next generation of stent and related medical devices: Current and future aspects. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1941. [PMID: 38528392 DOI: 10.1002/wnan.1941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 12/08/2023] [Accepted: 01/03/2024] [Indexed: 03/27/2024]
Abstract
Coronary stents have saved millions of lives in the last three decades by treating atherosclerosis especially, by preventing plaque protrusion and subsequent aneurysms. They attenuate the vascular SMC proliferation and promote reconstruction of the endothelial bed to ensure superior revascularization. With the evolution of modern stent types, nanotechnology has become an integral part of stent technology. Nanocoating and nanosurface fabrication on metallic and polymeric stents have improved their drug loading capacity as well as other mechanical, physico-chemical, and biological properties. Nanofeatures can mimic the natural nanofeatures of vascular tissue and control drug-delivery. This review will highlight the role of nanotechnology in addressing the challenges of coronary stents and the recent advancements in the field of related medical devices. Different generations of stents carrying nanoparticle-based formulations like liposomes, lipid-polymer hybrid NPs, polymeric micelles, and dendrimers are discussed highlighting their roles in local drug delivery and anti-restenotic properties. Drug nanoparticles like Paclitaxel embedded in metal stents are discussed as a feature of first-generation drug-eluting stents. Customized precision stents ensure safe delivery of nanoparticle-mediated genes or concerted transfer of gene, drug, and/or bioactive molecules like antibodies, gene mimics via nanofabricated stents. Nanotechnology can aid such therapies for drug delivery successfully due to its easy scale-up possibilities. However, limitations of this technology such as their potential cytotoxic effects associated with nanoparticle delivery that can trigger hypersensitivity reactions have also been discussed in this review. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Cardiovascular Disease Therapeutic Approaches and Drug Discovery > Emerging Technologies.
Collapse
Affiliation(s)
- Paromita Islam
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Sabrina Schaly
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Ahmed Kh Abosalha
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
- Pharmaceutical Technology Department, Faculty of Pharmacy, Tanta University, Tanta, Egypt
| | - Jacqueline Boyajian
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Rahul Thareja
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Waqar Ahmad
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| | - Dominique Shum-Tim
- Division of Cardiac Surgery, Royal Victoria Hospital, McGill University Health Centre, McGill University, Faculty of Medicine and Health Sciences, Montreal, Quebec, Canada
| | - Satya Prakash
- Biomedical Technology and Cell Therapy Research Laboratory, Department of Biomedical Engineering, Faculty of Medicine and Health Sciences, McGill University, Montreal, Quebec, Canada
| |
Collapse
|
9
|
Jain S, Kumar M, Kumar P, Verma J, Rosenholm JM, Bansal KK, Vaidya A. Lipid-Polymer Hybrid Nanosystems: A Rational Fusion for Advanced Therapeutic Delivery. J Funct Biomater 2023; 14:437. [PMID: 37754852 PMCID: PMC10531762 DOI: 10.3390/jfb14090437] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Revised: 08/17/2023] [Accepted: 08/21/2023] [Indexed: 09/28/2023] Open
Abstract
Lipid nanoparticles (LNPs) are spherical vesicles composed of ionizable lipids that are neutral at physiological pH. Despite their benefits, unmodified LNP drug delivery systems have substantial drawbacks, including a lack of targeted selectivity, a short blood circulation period, and in vivo instability. lipid-polymer hybrid nanoparticles (LPHNPs) are the next generation of nanoparticles, having the combined benefits of polymeric nanoparticles and liposomes. LPHNPs are being prepared from both natural and synthetic polymers with various techniques, including one- or two-step methods, emulsification solvent evaporation (ESE) method, and the nanoprecipitation method. Varieties of LPHNPs, including monolithic hybrid nanoparticles, core-shell nanoparticles, hollow core-shell nanoparticles, biomimetic lipid-polymer hybrid nanoparticles, and polymer-caged liposomes, have been investigated for various drug delivery applications. However, core-shell nanoparticles having a polymeric core surrounded by a highly biocompatible lipid shell are the most commonly explored LPHNPs for the treatment of various diseases. In this review, we will shed light on the composition, methods of preparation, classification, surface functionalization, release mechanism, advantages and disadvantages, patents, and clinical trials of LPHNPs, with an emphasis on core-shell-structured LPHNPs.
Collapse
Affiliation(s)
- Shweta Jain
- Sir Madan Lal Institute of Pharmacy, Etawah 206310, India;
| | - Mudit Kumar
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India; (M.K.); (P.K.)
| | - Pushpendra Kumar
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India; (M.K.); (P.K.)
| | - Jyoti Verma
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland; (J.V.); (J.M.R.)
| | - Jessica M. Rosenholm
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland; (J.V.); (J.M.R.)
| | - Kuldeep K. Bansal
- Pharmaceutical Sciences Laboratory, Faculty of Science and Engineering, Åbo Akademi University, 20520 Turku, Finland; (J.V.); (J.M.R.)
| | - Ankur Vaidya
- Faculty of Pharmacy, Uttar Pradesh University of Medical Sciences, Saifai, Etawah 206130, India; (M.K.); (P.K.)
| |
Collapse
|
10
|
Uruski P, Matuszewska J, Leśniewska A, Rychlewski D, Niklas A, Mikuła-Pietrasik J, Tykarski A, Książek K. An integrative review of nonobvious puzzles of cellular and molecular cardiooncology. Cell Mol Biol Lett 2023; 28:44. [PMID: 37221467 DOI: 10.1186/s11658-023-00451-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/17/2023] [Indexed: 05/25/2023] Open
Abstract
Oncologic patients are subjected to four major treatment types: surgery, radiotherapy, chemotherapy, and immunotherapy. All nonsurgical forms of cancer management are known to potentially violate the structural and functional integrity of the cardiovascular system. The prevalence and severity of cardiotoxicity and vascular abnormalities led to the emergence of a clinical subdiscipline, called cardiooncology. This relatively new, but rapidly expanding area of knowledge, primarily focuses on clinical observations linking the adverse effects of cancer therapy with deteriorated quality of life of cancer survivors and their increased morbidity and mortality. Cellular and molecular determinants of these relations are far less understood, mainly because of several unsolved paths and contradicting findings in the literature. In this article, we provide a comprehensive view of the cellular and molecular etiology of cardiooncology. We pay particular attention to various intracellular processes that arise in cardiomyocytes, vascular endothelial cells, and smooth muscle cells treated in experimentally-controlled conditions in vitro and in vivo with ionizing radiation and drugs representing diverse modes of anti-cancer activity.
Collapse
Affiliation(s)
- Paweł Uruski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Julia Matuszewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Aleksandra Leśniewska
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Daniel Rychlewski
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Arkadiusz Niklas
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Justyna Mikuła-Pietrasik
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Andrzej Tykarski
- Department of Hypertensiology, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland
| | - Krzysztof Książek
- Department of Pathophysiology of Ageing and Civilization Diseases, Poznań University of Medical Sciences, Długa ½ Str., 61-848, Poznan, Poland.
| |
Collapse
|
11
|
Smith BR, Edelman ER. Nanomedicines for cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:351-367. [PMID: 39195953 DOI: 10.1038/s44161-023-00232-y] [Citation(s) in RCA: 48] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 01/25/2023] [Indexed: 08/29/2024]
Abstract
The leading cause of death in the world, cardiovascular disease (CVD), remains a formidable condition for researchers, clinicians and patients alike. CVD comprises a broad collection of diseases spanning the heart, the vasculature and the blood that runs through and interconnects them. Limitations in CVD therapeutic and diagnostic landscapes have generated excitement for advances in nanomedicine, a field focused on improving patient outcomes through transformative therapies, imaging agents and ex vivo diagnostics. CVD nanomedicines are fundamentally shaped by their intended clinical application, including (1) cardiac or heart-related biomaterials, which can be functionally (for example, mechanically, immunologically, electrically) improved by incorporating nanomaterials; (2) the vasculature, involving systemically injected nanotherapeutics and imaging nanodiagnostics, nano-enabled biomaterials or tissue-nanoengineered solutions; and (3) improving the sensitivity and/or specificity of ex vivo diagnostic devices for patient samples. While immunotherapy has developed into a key pillar of oncology in the past dozen years, CVD immunotherapy and immunoimaging are recently emergent and likely to factor substantially in CVD management in the coming decade. The nanomaterials in CVD-related clinical trials and many promising preclinical strategies indicate that nanomedicine is on the cusp of greatly impacting patients with CVD. Here we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD nanomedicine.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
12
|
Raghunathan S, Rayes J, Sen Gupta A. Platelet-inspired nanomedicine in hemostasis thrombosis and thromboinflammation. J Thromb Haemost 2022; 20:1535-1549. [PMID: 35435322 PMCID: PMC9323419 DOI: 10.1111/jth.15734] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 03/30/2022] [Accepted: 04/01/2022] [Indexed: 12/01/2022]
Abstract
Platelets are anucleate cell-fragments derived predominantly from megakaryocytes in the bone marrow and released in the blood circulation, with a normal count of 150 000-40 000 per μl and a lifespan of approximately 10 days in humans. A primary role of platelets is to aid in vascular injury site-specific clot formation to stanch bleeding, termed hemostasis. Platelets render hemostasis by a complex concert of mechanisms involving platelet adhesion, activation and aggregation, coagulation amplification, and clot retraction. Additionally, platelet secretome can influence coagulation kinetics and clot morphology. Therefore, platelet defects and dysfunctions result in bleeding complications. Current treatment for such complications involve prophylactic or emergency transfusion of platelets. However, platelet transfusion logistics constantly suffer from limited donor availability, challenges in portability and storage, high bacterial contamination risks, and very short shelf life (~5 days). To address these issues, an exciting area of research is focusing on the development of microparticle- and nanoparticle-based platelet surrogate technologies that can mimic various hemostatic mechanisms of platelets. On the other hand, aberrant occurrence of the platelet mechanisms lead to the pathological manifestation of thrombosis and thromboinflammation. The treatments for this are focused on inhibiting the mechanisms or resolving the formed clots. Here, platelet-inspired technologies can provide unique platforms for disease-targeted drug delivery to achieve high therapeutic efficacy while avoiding systemic side-effects. This review will provide brief mechanistic insight into the role of platelets in hemostasis, thrombosis and thromboinflammation, and present the current state-of-art in the design of platelet-inspired nanomedicine for applications in these areas.
Collapse
Affiliation(s)
- Shruti Raghunathan
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| | - Julie Rayes
- Institute of Cardiovascular SciencesCollege of Medical and Dental SciencesUniversity of BirminghamBirminghamUK
| | - Anirban Sen Gupta
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOhioUSA
| |
Collapse
|
13
|
Shah P, Chandra S. Review on emergence of nanomaterial coatings in bio-engineered cardiovascular stents. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103224] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
14
|
Yang B, Wang Z, Jia Y, Fang D, Li R, Liu Y. Paclitaxel and its derivative facilitate the transmission of plasmid-mediated antibiotic resistance genes through conjugative transfer. THE SCIENCE OF THE TOTAL ENVIRONMENT 2022; 810:152245. [PMID: 34896514 DOI: 10.1016/j.scitotenv.2021.152245] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 11/24/2021] [Accepted: 12/04/2021] [Indexed: 06/14/2023]
Abstract
The rapid dissemination of antibiotic resistance by horizontal gene transfer (HGT) renders the global resistance crisis more tense and urgent as few effective antimicrobials are available to combat multidrug-resistant (MDR) pathogens at present. Conjugation is one of the most dominant and representative pathways of HGT. Antibiotic residue in environment is recognized as an important accelerator for conjugal transfer, whereas the roles of non-antibiotic pharmaceuticals in this process are not fully understood. Here we found that environmentally relevant concentrations of paclitaxel as well as its derivative docetaxel, two commonly used anticancer drugs, remarkably facilitated the conjugative transfer of resistance plasmids carrying multiple antibiotic resistance genes (ARGs). The underlying mechanisms accounting for the enhanced conjugation were investigated by detecting the activity of RpoS regulon, membrane permeability, SOS response and gene expression of conjugative transfer systems. Our results showed that paclitaxel induced a series of cellular responses, including up-regulation of rpoS expression, activated SOS response, increased cell membrane permeability, enhanced plasmid replication and mating pilus formation. Collectively, our data provide new insight on the roles of paclitaxel and its derivative in promoting the conjugal transfer of ARGs, highlighting the importance of good antimicrobial stewardship.
Collapse
Affiliation(s)
- Bingqing Yang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Zhiqiang Wang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuqian Jia
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Dan Fang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Ruichao Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Yuan Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, Jiangsu, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Joint International Research Laboratory of Agriculture and Agri-Product Safety, the Ministry of Education of China, Yangzhou University, Yangzhou, Jiangsu, China; Institute of Comparative Medicine, Yangzhou University, Yangzhou, Jiangsu, China.
| |
Collapse
|
15
|
Zhu H, Kong L, Zhu X, Ran T, Ji X. pH-Responsive Nanoparticles for Delivery of Paclitaxel to the Injury Site for Inhibiting Vascular Restenosis. Pharmaceutics 2022; 14:pharmaceutics14030535. [PMID: 35335910 PMCID: PMC8949492 DOI: 10.3390/pharmaceutics14030535] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/23/2022] [Accepted: 02/24/2022] [Indexed: 02/04/2023] Open
Abstract
A high incidence of restenosis has been reported at the site of inflammation following angioplasty and stent implantation. The anti-proliferative drug paclitaxel (PTX) could help to reduce inflammation and restenosis; however, it has poor water solubility and serious adverse side effects at high doses. Given the presence of metabolic acidosis at the site of inflammation, we hypothesized that nanoparticles that are responsive to low pH could precisely release the loaded drug at the target site. We successfully constructed pH-responsive poly(D, L-lactic-co-glycolic acid) (PLGA) nanoparticles loaded with PTX and NaHCO3 as a pH-sensitive therapeutic agent (PTX-NaHCO3-PLGA NPs). The NPs exhibited remarkable pH sensitivity and a good safety profile both in vitro in rat vascular smooth muscle cells and in vivo in Sprague Dawley rats after tail vein injection. In the rat model, the PTX-NaHCO3-PLGA NPs treatment group showed suppressed intimal proliferation following balloon-induced carotid artery injury compared with that of the saline-treated control. Overall, these results demonstrate that our newly developed pH-responsive nanodrug delivery platform has the potential to effectively inhibit restenosis.
Collapse
Affiliation(s)
- Huiru Zhu
- Department of Ultrasound Imaging, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China; (H.Z.); (L.K.); (X.Z.); (T.R.)
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Li Kong
- Department of Ultrasound Imaging, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China; (H.Z.); (L.K.); (X.Z.); (T.R.)
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- State Key Laboratory of Ultrasound in Medicine and Engineering, College of Biomedical Engineering, Chongqing Medical University, Chongqing 400016, China
| | - Xu Zhu
- Department of Ultrasound Imaging, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China; (H.Z.); (L.K.); (X.Z.); (T.R.)
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Tingting Ran
- Department of Ultrasound Imaging, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China; (H.Z.); (L.K.); (X.Z.); (T.R.)
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
| | - Xiaojuan Ji
- Department of Ultrasound Imaging, Children’s Hospital of Chongqing Medical University, Chongqing 400014, China; (H.Z.); (L.K.); (X.Z.); (T.R.)
- National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Pediatrics, Chongqing 400014, China
- Correspondence:
| |
Collapse
|
16
|
Ng JCK, Toong DWY, Ow V, Chaw SY, Toh H, Wong PEH, Venkatraman S, Chong TT, Tan LP, Huang YY, Ang HY. Progress in drug-delivery systems in cardiovascular applications: stents, balloons and nanoencapsulation. Nanomedicine (Lond) 2022; 17:325-347. [PMID: 35060758 DOI: 10.2217/nnm-2021-0288] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Drug-delivery systems in cardiovascular applications regularly include the use of drug-eluting stents and drug-coated balloons to ensure sufficient drug transfer and efficacy in the treatment of cardiovascular diseases. In addition to the delivery of antiproliferative drugs, the use of growth factors, genetic materials, hormones and signaling molecules has led to the development of different nanoencapsulation techniques for targeted drug delivery. The review will cover drug delivery and coating mechanisms in current drug-eluting stents and drug-coated balloons, novel innovations in drug-eluting stent technologies and drug encapsulation in nanocarriers for delivery in vascular diseases. Newer technologies and advances in nanoencapsulation techniques, such as the use of liposomes, nanogels and layer-by-layer coating to deliver therapeutics in the cardiovascular space, will be highlighted.
Collapse
Affiliation(s)
- Jaryl Chen Koon Ng
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore.,Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore, 117583, Singapore
| | - Daniel Wee Yee Toong
- Department of Materials Science and Engineering, Nanyang Technological University, Nanyang Avenue, Singapore, 639798, Singapore
| | - Valerie Ow
- Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore, 117583, Singapore
| | - Su Yin Chaw
- Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Hanwei Toh
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore.,Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore, 117583, Singapore
| | - Philip En Hou Wong
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore.,Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore
| | - Subbu Venkatraman
- Department of Material Science Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore, 117575, Singapore
| | - Tze Tec Chong
- Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,Department of Vascular Surgery, Singapore General Hospital, Outram Road, Singapore, 169608, Singapore
| | - Lay Poh Tan
- Department of Materials Science and Engineering, Nanyang Technological University, Nanyang Avenue, Singapore, 639798, Singapore
| | - Ying Ying Huang
- Department of Materials Science and Engineering, Nanyang Technological University, Nanyang Avenue, Singapore, 639798, Singapore
| | - Hui Ying Ang
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore, 169609, Singapore.,Department of Biomedical Engineering, National University of Singapore, 4 Engineering Drive 3, Singapore, 117583, Singapore.,Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore
| |
Collapse
|
17
|
Zhao Y, Shirasu T, Yodsanit N, Kent E, Ye M, Wang Y, Xie R, Gregg AC, Huang Y, Kent KC, Guo LW, Gong S, Wang B. Biomimetic, ROS-detonable nanoclusters - A multimodal nanoplatform for anti-restenotic therapy. J Control Release 2021; 338:295-306. [PMID: 34416322 PMCID: PMC12126213 DOI: 10.1016/j.jconrel.2021.08.025] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Revised: 07/14/2021] [Accepted: 08/15/2021] [Indexed: 12/18/2022]
Abstract
The long-term success of endovascular intervention has long been overshadowed by vessel re-occlusion, also known as restenosis. Mainstream anti-restenotic devices, such as drug-eluting stent (DES) and drug-coated balloon (DCB), were recently shown with suboptimal performances and life-threatening complications, thereby underpinning the urgent need for alternative strategies with enhanced efficacy and safety profile. In our current study, we engineered a multimodal nanocluster formed by self-assembly of unimolecular nanoparticles and surface coated with platelet membrane, specifically tailored for precision drug delivery in endovascular applications. More specifically, it incorporates the combined merits of platelet membrane coating (lesion targetability and biocompatibility), reactive oxygen species (ROS)-detonable "cluster-bomb" chemistry (to trigger the large-to-small size transition at the target site, thereby achieving longer circulation time and higher tissue penetration), and sustained drug release. Using RVX-208 (an emerging anti-restenotic drug under clinical trials) as the model payload, we demonstrated the superior performances of our nanocluster over conventional poly(lactic-co-glycolic acid) (PLGA) nanoparticle. In cultured vascular smooth muscle cell (VSMC), the drug-loaded nanocluster induced effective inhibition of proliferation and protective gene expression (e.g., APOA-I) with a significantly reduced dosage of RVX-208 (1 μM). In a rat model of balloon angioplasty, intravenous injection of Cy5.5-tagged nanocluster led to greater lesion targetability, improved biodistribution, and deeper penetration into injured vessel walls featuring enriched ROS. Moreover, in contrast to either free drug solution or drug-loaded PLGA nanoparticle formulation, a single injection with the drug-loaded nanocluster (10 mg/kg of RVX-208) was sufficient to substantially mitigate restenosis. Additionally, this nanocluster also demonstrated biocompatibility according to in vitro cytotoxicity assay and in vivo histological and tissue qPCR analysis. Overall, our multimodal nanocluster offers improved targetability, tissue penetration, and ROS-responsive release over conventional nanoparticles, therefore making it a highly promising platform for development of next-generation endovascular therapies.
Collapse
Affiliation(s)
- Yi Zhao
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA.; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Takuro Shirasu
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Nisakorn Yodsanit
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA.; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Eric Kent
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - Mingzhou Ye
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA.; Wisconsin Institute for Discovery, University of Wisconsin-Madison, Madison, WI, 53715, USA
| | - Yuyuan Wang
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA.; Wisconsin Institute for Discovery, 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
| | | | - Yitao Huang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA.; The Biomedical Sciences Graduate Program, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA
| | - K Craig Kent
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA..
| | - Lian-Wang Guo
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA..
| | - Shaoqin Gong
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, 53715, USA.; 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.; Department of Chemistry, University of Wisconsin-Madison, Madison, WI, 53715, USA..
| | - Bowen Wang
- Department of Surgery, School of Medicine, University of Virginia, Charlottesville, VA, 22908, USA..
| |
Collapse
|
18
|
Applications of Nanosized-Lipid-Based Drug Delivery Systems in Wound Care. APPLIED SCIENCES-BASEL 2021. [DOI: 10.3390/app11114915] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Impaired wound healing is an encumbering public health issue that increases the demand for developing new therapies in order to minimize health costs and enhance treatment efficacy. Available conventional therapies are still unable to maximize their potential in penetrating the skin at the target site and accelerating the healing process. Nanotechnology exhibits an excellent opportunity to enrich currently available medical treatments, enhance standard care and manage wounds. It is a promising approach, able to address issues such as the permeability and bioavailability of drugs with reduced stability or low water solubility. This paper focuses on nanosized-lipid-based drug delivery systems, describing their numerous applications in managing skin wounds. We also highlight the relationship between the physicochemical characteristics of nanosized, lipid-based drug delivery systems and their impact on the wound-healing process. Different types of nanosized-lipid-based drug delivery systems, such as vesicular systems and lipid nanoparticles, demonstrated better applicability and enhanced skin penetration in wound healing therapy compared with conventional treatments. Moreover, an improved chemically and physically stable drug delivery system, with increased drug loading capacity and enhanced bioavailability, has been shown in drugs encapsulated in lipid nanoparticles. Their applications in wound care show potential for overcoming impediments, such as the inadequate bioavailability of active agents with low solubility. Future research in nanosized-lipid-based drug delivery systems will allow the achievement of increased bioavailability and better control of drug release, providing the clinician with more effective therapies for wound care.
Collapse
|
19
|
Groner J, Goepferich A, Breunig M. Atherosclerosis: Conventional intake of cardiovascular drugs versus delivery using nanotechnology - A new chance for causative therapy? J Control Release 2021; 333:536-559. [PMID: 33794270 DOI: 10.1016/j.jconrel.2021.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 02/06/2023]
Abstract
Atherosclerosis is the leading cause of death in developed countries. The pathogenetic mechanism relies on a macrophage-based immune reaction to low density lipoprotein (LDL) deposition in blood vessels with dysfunctional endothelia. Thus, atherosclerosis is defined as a chronic inflammatory disease. A plethora of cardiovascular drugs have been developed and are on the market, but the major shortcoming of standard medications is that they do not address the root cause of the disease. Statins and thiazolidinediones that have recently been recognized to exert specific anti-atherosclerotic effects represent a potential breakthrough on the horizon. But their whole potential cannot be realized due to insufficient availability at the pathological site and severe off-target effects. The focus of this review will be to elaborate how both groups of drugs could immensely profit from nanoparticulate carriers. This delivery principle would allow for their accumulation in target macrophages and endothelial cells of the atherosclerotic plaque, increasing bioavailability where it is needed most. Based on the analyzed literature we conclude design criteria for the delivery of statins and thiazolidinediones with nanoparticles for anti-atherosclerotic therapy. Nanoparticles need to be below a diameter of 100 nm to accumulate in the atherosclerotic plaque and should be fabricated using biodegradable materials. Further, the thiazolidinediones or statins must be encapsulated into the particle core, because especially for thiazolidindiones the uptake into cells is prerequisite for their mechanism of action. For optimal uptake into targeted macrophages and endothelial cells, the ideal particle should present ligands on its surface which bind specifically to scavenger receptors. The impact of statins on the lectin-type oxidized LDL receptor 1 (LOX1) seems particularly promising because of its outstanding role in the inflammatory process. Using this pioneering concept, it will be possible to promote the impact of statins and thiazolidinediones on macrophages and endothelial cells and significantly enhance their anti-atherosclerotic therapeutic potential.
Collapse
Affiliation(s)
- Jonas Groner
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany
| | - Miriam Breunig
- Department of Pharmaceutical Technology, University of Regensburg, Universitätsstraße 31, 93053 Regensburg, Germany.
| |
Collapse
|
20
|
Santiago FS, Li Y, Zhong L, Raftery MJ, Lins L, Khachigian LM. Truncated YY1 interacts with BASP1 through a 339KLK341 motif in YY1 and suppresses vascular smooth muscle cell growth and intimal hyperplasia after vascular injury. Cardiovasc Res 2021; 117:2395-2406. [PMID: 33508088 DOI: 10.1093/cvr/cvab021] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 10/27/2020] [Accepted: 01/19/2021] [Indexed: 11/12/2022] Open
Abstract
AIMS In-stent restenosis and late stent thrombosis are complications associated with the use of metallic and drug-coated stents. Strategies that inhibit vascular smooth muscle cell (SMC) proliferation without affecting endothelial cell (EC) growth would be helpful in reducing complications arising from percutaneous interventions. Our group previously showed that the forced expression of the injury-inducible zinc finger (ZNF) transcription factor, yin yang-1 (YY1) comprising 414 residues inhibits neointima formation in carotid arteries of rabbits and rats. YY1 inhibits SMC proliferation without affecting EC growth. Identifying a shorter version of YY1 retaining cell-selective inhibition would make it more amenable for potential use as a gene therapeutic agent. METHODS AND RESULTS We dissected YY1 into a range of shorter fragments (YY1A-D, YY1Δ) and found that the first two ZNFs in YY1 (construct YY1B, spanning 52 residues) repressed SMC proliferation. Receptor Binding Domain analysis predicts a three residue (339KLK341) interaction domain. Mutation of 339KLK341 to 339AAA341 in YY1B (called YY1Bm) abrogated YY1B's ability to inhibit SMC but not EC proliferation and migration. Incubation of recombinant GST-YY1B and GST-YY1Bm with SMC lysates followed by precipitation with glutathione-agarose beads and mass spectrometric analysis identified a novel interaction between YY1B and BASP1. Overexpression of BASP1, like YY1, inhibited SMC but not EC proliferation and migration. BASP1 siRNA partially rescued SMC from growth inhibition by YY1B. In the rat carotid balloon injury model, adenoviral overexpression of YY1B, like full-length YY1, reduced neointima formation, whereas YY1Bm had no such effect. CD31 immunostaining suggested YY1B could increase re-endothelialization in a 339KLK341-dependent manner. CONCLUSIONS These studies identify a truncated form of YY1 (YY1B) that can interact with BASP1 and inhibits SMC proliferation, migration and intimal hyperplasia after balloon injury of rat carotid arteries as effectively as full length YY1. We demonstrate the therapeutic potential of YY1B in vascular proliferative disease.
Collapse
Affiliation(s)
- Fernando S Santiago
- Vascular Biology and Translational Research Laboratory, School of Medical Sciences, UNSW Medicine, University of New South Wales, Sydney NSW 2052, Australia
| | - Yue Li
- Vascular Biology and Translational Research Laboratory, School of Medical Sciences, UNSW Medicine, University of New South Wales, Sydney NSW 2052, Australia
| | - Ling Zhong
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney NSW 2052, Australia
| | - Mark J Raftery
- Bioanalytical Mass Spectrometry Facility, Mark Wainwright Analytical Centre, University of New South Wales, Sydney NSW 2052, Australia
| | - Laurence Lins
- Molecular Biophysics at Interface Lab, University of Liège-Gembloux Agro Bio Tech, Passage des Déportés, 2-5030 Gembloux-Belgium
| | - Levon M Khachigian
- Vascular Biology and Translational Research Laboratory, School of Medical Sciences, UNSW Medicine, University of New South Wales, Sydney NSW 2052, Australia
| |
Collapse
|
21
|
Poly (Lactic- co-Glycolic Acid) Nanoparticles and Nanoliposomes for Protein Delivery in Targeted Therapy: A Comparative In Vitro Study. Polymers (Basel) 2020; 12:polym12112566. [PMID: 33139610 PMCID: PMC7692461 DOI: 10.3390/polym12112566] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 10/27/2020] [Accepted: 10/29/2020] [Indexed: 12/12/2022] Open
Abstract
Over the previous years, the design, development, and potential application of nanocarriers in the medical field have been intensively studied for their ability to preserve drug properties, especially their pharmacological activity, and to improve their bioavailability. This work is a comparative study between two different types of nanocarriers, poly (lactic-co-glycolic acid)-based nanoparticles and phosphatidylcholine-based nanoliposomes, both prepared for the encapsulation of bovine serum albumin as a model protein. Polymeric nanoparticles were produced using the double emulsion water-oil-water evaporation method, whereas nanoliposomes were obtained by the thin-film hydration method. Both nanocarriers were characterized by morphological analysis, particle mean size, particle size distribution, and protein entrapment efficiency. Invitro release studies were performed for 12 days at 37 °C. In order to explore a possible application of these nanocarriers for a targeted therapy in the cardiovascular field, hemolytic activity and biocompatibility, in terms of cell viability, were performed by using human red blood cells and EA.hy926 human endothelial cell line, respectively.
Collapse
|
22
|
Wang S, Guo X, Ren L, Wang B, Hou L, Zhou H, Gao Q, Gao Y, Wang L. Targeting and deep-penetrating delivery strategy for stented coronary artery by magnetic guidance and ultrasound stimulation. ULTRASONICS SONOCHEMISTRY 2020; 67:105188. [PMID: 32473543 DOI: 10.1016/j.ultsonch.2020.105188] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Revised: 04/30/2020] [Accepted: 05/24/2020] [Indexed: 05/13/2023]
Abstract
Stent placement is an effective treatment for atherosclerosis, but is suffered from in-stent restenosis (ISR) caused by stent mechanical damage. Conventional ISR treatment such as drug-eluting stents (DES) is challenged by the low therapeutic efficacy and severe complications, unchangeable drug dosage for individuals, and limited drug penetration in the vascular tissue. We hypothesize that magnetic targeting and deep-penetrating delivery strategy by magnetic guidance and ultrasound stimulation might be an effective approach for ISR treatment. In the present study, antiproliferative drug (paclitaxel, PTX) loaded poly (lactide-co-glycolide) (PLGA) nanoparticles (PLGA-PTX) were embedded within the shells of the magnetic nanoparticle coated microbubbles (MMB-PLGA-PTX). Once being targeted to the stent under a magnetic field, a low intensity focused ultrasound (LIFU) is applied to activate stable microbubble oscillations, thereby triggering the release of PLGA-PTX. The generated mechanical force and microstreaming facilitate the penetration of released PLGA-PTX into the thickened vascular tissue and enhance their internalization by smooth muscle cells (SMCs), thereby reducing the clearance by blood flow. In an ex vivo experiment, magnetic targeting improved the accumulation amount of MMB-PLGA-PTX by 10 folds, while the LIFU facilitated the penetration of released PLGA-PTX into the tunica media region of the porcine coronary artery, resulting in prolonged retention time at the stented vascular tissue. With the combination effects, this strategy holds great promise in the precision delivery of antiproliferative drugs to the stented vascular tissue for ISR treatment.
Collapse
Affiliation(s)
- Siyu Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Xixi Guo
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Lili Ren
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Bo Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Lixin Hou
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Hao Zhou
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Qinchang Gao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China
| | - Yu Gao
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| | - Lianhui Wang
- Key Laboratory for Organic Electronics and Information Displays & Jiangsu Key Laboratory for Biosensors, Institute of Advanced Materials (IAM), Jiangsu National Synergistic Innovation Center for Advanced Materials (SICAM), Nanjing University of Posts & Telecommunications, Nanjing 210023, China.
| |
Collapse
|
23
|
Hu B, Boakye‐Yiadom KO, Yu W, Yuan Z, Ho W, Xu X, Zhang X. Nanomedicine Approaches for Advanced Diagnosis and Treatment of Atherosclerosis and Related Ischemic Diseases. Adv Healthc Mater 2020; 9:e2000336. [PMID: 32597562 DOI: 10.1002/adhm.202000336] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Revised: 04/30/2020] [Indexed: 12/16/2022]
Abstract
Cardiovascular diseases (CVDs) remain one of the major causes of mortality worldwide. In response to this and other worldwide health epidemics, nanomedicine has emerged as a rapidly evolving discipline that involves the development of innovative nanomaterials and nanotechnologies and their applications in therapy and diagnosis. Nanomedicine presents unique advantages over conventional medicines due to the superior properties intrinsic to nanoscopic therapies. Once used mainly for cancer therapies, recently, tremendous progress has been made in nanomedicine that has led to an overall improvement in the treatment and diagnosis of CVDs. This review elucidates the pathophysiology and potential targets of atherosclerosis and associated ischemic diseases. It may be fruitful to pursue future work in the nanomedicine-mediated treatment of CVDs based on these targets. A comprehensive overview is then provided featuring the latest preclinical and clinical outcomes in cardiovascular imaging, biomarker detection, tissue engineering, and nanoscale delivery, with specific emphasis on nanoparticles, nanostructured scaffolds, and nanosensors. Finally, the challenges and opportunities regarding the future development and clinical translation of nanomedicine in related fields are discussed. Overall, this review aims to provide a deep and thorough understanding of the design, application, and future development of nanomedicine for atherosclerosis and related ischemic diseases.
Collapse
Affiliation(s)
- Bin Hu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Kofi Oti Boakye‐Yiadom
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Wei Yu
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - Zi‐Wei Yuan
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| | - William Ho
- Department of Chemical and Materials EngineeringNew Jersey Institute of Technology Newark NJ 07102 USA
| | - Xiaoyang Xu
- Department of Chemical and Materials EngineeringNew Jersey Institute of Technology Newark NJ 07102 USA
| | - Xue‐Qing Zhang
- Engineering Research Center of Cell & Therapeutic Antibody, Ministry of Education, and School of PharmacyShanghai Jiao Tong University 800 Dongchuan Road Shanghai 200240 P. R. China
| |
Collapse
|
24
|
Abstract
The term "nanotechnology" was coined by Norio Taniguchi in the 1970s to describe the manipulation of materials at the nano (10-9) scale, and the term "nanomedicine" was put forward by Eric Drexler and Robert Freitas Jr. in the 1990s to signify the application of nanotechnology in medicine. Nanomedicine encompasses a variety of systems including nanoparticles, nanofibers, surface nano-patterning, nanoporous matrices, and nanoscale coatings. Of these, nanoparticle-based applications in drug formulations and delivery have emerged as the most utilized nanomedicine system. This review aims to present a comprehensive assessment of nanomedicine approaches in vascular diseases, emphasizing particle designs, therapeutic effects, and current state-of-the-art. The expected advantages of utilizing nanoparticles for drug delivery stem from the particle's ability to (1) protect the drug from plasma-induced deactivation; (2) optimize drug pharmacokinetics and biodistribution; (3) enhance drug delivery to the disease site via passive and active mechanisms; (4) modulate drug release mechanisms via diffusion, degradation, and other unique stimuli-triggered processes; and (5) biodegrade or get eliminated safely from the body. Several nanoparticle systems encapsulating a variety of payloads have shown these advantages in vascular drug delivery applications in preclinical evaluation. At the same time, new challenges have emerged regarding discrepancy between expected and actual fate of nanoparticles in vivo, manufacturing barriers of complex nanoparticle designs, and issues of toxicity and immune response, which have limited successful clinical translation of vascular nanomedicine systems. In this context, this review will discuss challenges and opportunities to advance the field of vascular nanomedicine.
Collapse
Affiliation(s)
- Michael Sun
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| | - Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio
| |
Collapse
|
25
|
Yan H, Mi X, Midgley AC, Du X, Huang Z, Wei T, Liu R, Ma T, Zhi D, Zhu D, Wang T, Feng G, Zhao Y, Zhang W, He J, Zhu M, Kong D, Wang K. Targeted Repair of Vascular Injury by Adipose-Derived Stem Cells Modified with P-Selectin Binding Peptide. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2020; 7:1903516. [PMID: 32537407 PMCID: PMC7284211 DOI: 10.1002/advs.201903516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/26/2020] [Accepted: 03/12/2020] [Indexed: 05/18/2023]
Abstract
Percutaneous coronary intervention for coronary artery disease treatment often results in pathological vascular injury, characterized by P-selectin overexpression. Adipose-derived stem cells (ADSCs) therapeutic efficacy remains elusive due to poor ADSCs targeting and retention in injured vessels. Here, conjugated P-selectin binding peptide (PBP) to polyethylene glycol-conjugated phospholipid derivative (DMPE-PEG) linkers (DMPE-PEG-PBP; DPP) are used to facilitate the modification of PBP onto ADSCs cell surfaces via hydrophobic interactions between DMPE-PEG and the phospholipid bilayer. DPP modification neither has influence on ADSCs proliferation nor apoptosis/paracrine factor gene expression. A total of 5 × 10-6 m DPP-modified ADSCs (DPP-ADSCs) strongly binds to P-selectin-displaying activated platelets and endothelial cells (ECs) in vitro and to wire-injured rat femoral arteries when administered by intra-arterial injection. Targeted binding of ADSCs shields injury sites from platelet and leukocyte adhesion, thereby decreasing inflammation at injury sites. Furthermore, targeted binding of ADSCs recovers injured ECs functionality and reduces platelet-initiated vascular smooth muscle cells (VSMCs) chemotactic migration. Targeted binding of DPP-human ADSCs to balloon-injured human femoral arteries is also demonstrated in ex vivo experiments. Overall, DPP-ADSCs promote vascular repair, inhibit neointimal hyperplasia, increase endothelium functionality, and maintain normal VSMCs alignment, supporting preclinical noninvasive utilization of DPP-ADSCs for vascular injury.
Collapse
Affiliation(s)
- Hongyu Yan
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xingyan Mi
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Adam C. Midgley
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Xinchen Du
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Ziqi Huang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Tingting Wei
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Ruihua Liu
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Tengzhi Ma
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Dengke Zhi
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Dashuai Zhu
- School of MedicineNankai UniversityTianjin300071China
| | - Ting Wang
- Urban Transport Emission Control Research CentreCollege of Environmental Science and EngineeringNankai UniversityTianjin300071China
| | - Guowei Feng
- Department of Genitourinary OncologyTianjin Medical University Cancer Institute and HospitalNational Clinical Research Center for CancerKey Laboratory of Cancer Prevention and TherapyTianjin300060China
| | - Ying Zhao
- Donation ServicesTianjin First Central HospitalTianjin300192China
| | - Weiye Zhang
- Donation ServicesTianjin First Central HospitalTianjin300192China
| | - Ju He
- Department of Vascular SurgeryTianjin First Central HospitalTianjin300192China
| | - Meifeng Zhu
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Deling Kong
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| | - Kai Wang
- Key Laboratory of Bioactive MaterialsMinistry of EducationCollege of Life SciencesNankai UniversityTianjin300071China
| |
Collapse
|
26
|
Recent advances in theranostic polymeric nanoparticles for cancer treatment: A review. Int J Pharm 2020; 582:119314. [PMID: 32283197 DOI: 10.1016/j.ijpharm.2020.119314] [Citation(s) in RCA: 98] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 04/03/2020] [Accepted: 04/06/2020] [Indexed: 12/16/2022]
Abstract
Nanotheranostics is fast-growing pharmaceutical technology for simultaneously monitoring drug release and its distribution, and to evaluate the real time therapeutic efficacy through a single nanoscale for treatment and diagnosis of deadly disease such as cancers. In recent two decades, biodegradable polymers have been discovered as important carriers to accommodate therapeutic and medical imaging agents to facilitate construction of multi-modal formulations. In this review, we summarize various multifunctional polymeric nano-sized formulations such as polymer-based super paramagnetic nanoparticles, ultrasound-triggered polymeric nanoparticles, polymeric nanoparticles bearing radionuclides, and fluorescent polymeric nano-sized formulations for purpose of theranostics. The use of such multi-modal nano-sized formulations for near future clinical trials can assist clinicians to predict therapeutic properties (for instance, depending upon the quantity of drug accumulated at the cancerous site) and observed the progress of tumor growth in patients, thus improving tailored medicines.
Collapse
|
27
|
Mi P, Cabral H, Kataoka K. Ligand-Installed Nanocarriers toward Precision Therapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2020; 32:e1902604. [PMID: 31353770 DOI: 10.1002/adma.201902604] [Citation(s) in RCA: 187] [Impact Index Per Article: 37.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/24/2019] [Revised: 06/04/2019] [Indexed: 05/20/2023]
Abstract
Development of drug-delivery systems that selectively target neoplastic cells has been a major goal of nanomedicine. One major strategy for achieving this milestone is to install ligands on the surface of nanocarriers to enhance delivery to target tissues, as well as to enhance internalization of nanocarriers by target cells, which improves accuracy, efficacy, and ultimately enhances patient outcomes. Herein, recent advances regarding the development of ligand-installed nanocarriers are introduced and the effect of their design on biological performance is discussed. Besides academic achievements, progress on ligand-installed nanocarriers in clinical trials is presented, along with the challenges faced by these formulations. Lastly, the future perspectives of ligand-installed nanocarriers are discussed, with particular emphasis on their potential for emerging precision therapies.
Collapse
Affiliation(s)
- Peng Mi
- Department of Radiology, Center for Medical Imaging, and State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University, No.17 People's South Road, Chengdu, 610041, China
| | - Horacio Cabral
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Kazunori Kataoka
- Innovation Center of Nanomedicine (iCONM), Kawasaki Institute of Industrial Promotion, 3-25-14 Tonomachi, Kawasaki-ku, Kawasaki, 210-0821, Japan
- Institute for Future Initiatives, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-0033, Japan
| |
Collapse
|
28
|
Zhang R, Liu R, Liu C, Pan L, Qi Y, Cheng J, Guo J, Jia Y, Ding J, Zhang J, Hu H. A pH/ROS dual-responsive and targeting nanotherapy for vascular inflammatory diseases. Biomaterials 2019; 230:119605. [PMID: 31740099 DOI: 10.1016/j.biomaterials.2019.119605] [Citation(s) in RCA: 86] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/14/2019] [Accepted: 11/05/2019] [Indexed: 01/11/2023]
Abstract
Cardiovascular diseases (CVDs) remain the leading cause of morbidity and mortality worldwide. Vascular inflammation is closely related to the pathogenesis of a diverse group of CVDs. Currently, it remains a great challenge to achieve site-specific delivery and controlled release of therapeutics at vascular inflammatory sites. Herein we hypothesize that active targeting nanoparticles (NPs) simultaneously responsive to low pH and high levels of reactive oxygen species (ROS) can serve as an effective nanoplatform for precision delivery of therapeutic cargoes to the sites of vascular inflammation, in view of acidosis and oxidative stress at inflamed sites. The pH/ROS dual-responsive NPs were constructed by combination of a pH-sensitive material (ACD) and an oxidation-responsive material (OCD) that can be facilely synthesized by chemical functionalization of β-cyclodextrin, a cyclic oligosaccharide. Simply by regulating the weight ratio of ACD and OCD, the pH/ROS responsive capacity can be easily modulated, affording NPs with varied hydrolysis profiles under inflammatory microenvironment. Using rapamycin (RAP) as a candidate drug, we first demonstrated in vitro therapeutic advantages of RAP-containing NPs with optimal dual-responsive capability, i.e. RAP/AOCD NP, and a non-responsive nanotherapy (RAP/PLGA NP) and two single-responsive nanotherapies (RAP/ACD NP and RAP/OCD NP) were used as controls. In an animal model of vascular inflammation in rats subjected to balloon injury in carotid arteries, AOCD NP could accumulate at the diseased site after intravenous (i.v.) injection. Consistently, i. v. treatment with RAP/AOCD NP more effectively inhibited neointimal hyperplasia in rats with induced arterial injuries, compared to RAP/PLGA NP, RAP/ACD NP, and RAP/OCD NP. By surface decoration of AOCD NP with a peptide (KLWVLPKGGGC) targeting type IV collagen (Col-IV), the obtained Col-IV targeting, dual-responsive nanocarrier TAOCD NP showed dramatically increased accumulation at injured carotid arteries. Furthermore, RAP/TAOCD NP exhibited significantly potentiated in vivo efficacy in comparison to the passive targeting nanotherapy RAP/AOCD NP. Importantly, in vitro cell culture experiments and in vivo animal studies in both mice and rats revealed good safety for AOCD NP and RAP/AOCD NP, even after long-term treatment via i. v. injection. Consequently, our results demonstrated that the newly developed Col-IV targeting, pH/ROS dual-responsive NPs may serve as an effective and safe nanovehicle for precision therapy of arterial restenosis and other vascular inflammatory diseases.
Collapse
Affiliation(s)
- Runjun Zhang
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Renfeng Liu
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Chao Liu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Lina Pan
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Yuantong Qi
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Juan Cheng
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Jiawei Guo
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China; Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Yi Jia
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China
| | - Jun Ding
- Department of Ultrasound, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China
| | - Jianxiang Zhang
- Department of Pharmaceutics, College of Pharmacy, Third Military Medical University, Chongqing, 400038, China.
| | - Houyuan Hu
- Department of Cardiology, Southwest Hospital, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
29
|
Drug repurposing for breast cancer therapy: Old weapon for new battle. Semin Cancer Biol 2019; 68:8-20. [PMID: 31550502 PMCID: PMC7128772 DOI: 10.1016/j.semcancer.2019.09.012] [Citation(s) in RCA: 82] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2019] [Revised: 09/17/2019] [Accepted: 09/18/2019] [Indexed: 12/24/2022]
Abstract
Despite tremendous resources being invested in prevention and treatment, breast cancer remains a leading cause of cancer deaths in women globally. The available treatment modalities are very costly and produces severe side effects. Drug repurposing that relate to new uses for old drugs has emerged as a novel approach for drug development. Repositioning of old, clinically approved, off patent non-cancer drugs with known targets, into newer indication is like using old weapons for new battle. The advances in genomics, proteomics and information computational biology has facilitated the process of drug repurposing. Repositioning approach not only fastens the process of drug development but also offers more effective, cheaper, safer drugs with lesser/known side effects. During the last decade, drugs such as alkylating agents, anthracyclins, antimetabolite, CDK4/6 inhibitor, aromatase inhibitor, mTOR inhibitor and mitotic inhibitors has been repositioned for breast cancer treatment. The repositioned drugs have been successfully used for the treatment of most aggressive triple negative breast cancer. The literature review suggest that serendipity plays a major role in the drug development. This article describes the comprehensive overview of the current scenario of drug repurposing for the breast cancer treatment. The strategies as well as several examples of repurposed drugs are provided. The challenges associated with drug repurposing are discussed.
Collapse
|
30
|
Nanotherapies for Treatment of Cardiovascular Disease: A Case for Antioxidant Targeted Delivery. CURRENT PATHOBIOLOGY REPORTS 2019; 7:47-60. [PMID: 31396435 DOI: 10.1007/s40139-019-00196-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Purpose of Review Cardiovascular disease (CVD) involves a broad range of clinical manifestations resulting from a dysfunctional vascular system. Overproduction of reactive oxygen and nitrogen species are causally implicated in the severity of vascular dysfunction and CVD. Antioxidant therapy is an attractive avenue for treatment of CVD associated pathologies. Implementation of targeted nano-antioxidant therapies has the potential to overcome hurdles associated with systemic delivery of antioxidants. This review examines the currently available options for nanotherapeutic targeting CVD, and explores successful studies showcasing targeted nano-antioxidant therapy. Recent Findings Active targeting strategies in the context of CVD heavily focus on immunotargeting to inflammatory markers like cell adhesion molecules, or to exposed extracellular matrix components. Targeted antioxidant nanotherapies have found success in pre-clinical studies. Summary This review underscores the potential of targeted nanocarriers as means of finding success translating antioxidant therapies to the clinic, all with a focus on CVD.
Collapse
|
31
|
Zong Z, Hua L, Wang Z, Xu H, Ye C, Pan B, Zhao Z, Zhang L, Lu J, Mei LH, Rutong Y. Self-assembled angiopep-2 modified lipid-poly (hypoxic radiosensitized polyprodrug) nanoparticles delivery TMZ for glioma synergistic TMZ and RT therapy. Drug Deliv 2019; 26:34-44. [PMID: 30744436 PMCID: PMC6394306 DOI: 10.1080/10717544.2018.1534897] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
The addition of temozolomide (TMZ) to radiotherapy (RT) improves survival of patients with glioblastoma (GBM). However, TMZ + RT causes excess toxicity in patients. In this study, we prepared angiopep-2 (A2) modified lipid-poly (hypoxic radiosensitized polyprodrug) nanoparticles for TMZ delivery (A2-P(MIs)25/TMZ) to achieve synergistic effects against glioma. This A2-P(MIs)25/TMZ display highly promising advantages: (1) a hydrophobic P-(MIs)25 core where poorly water-soluble TMZ can be encapsulated; (2) nitro groups of the hydrophobic P-(MIs)25 core that are converted into hydrophilic amino groups (P(NH2s)25) under low oxygen conditions to mimic the oxygen-increased sensitization to RT; (3) a lipid monolayer at the interface of the core and the shell to modify the A2 (a specific ligand for low-density lipoprotein receptor-related protein-1 (LRP-1), which are expressed in the blood-brain barrier (BBB) and human glioma cells), thereby enhancing the drug encapsulation efficiency in glioma. These nanoparticles appear as a promising and robust nanoplatforms for TMZ and hypoxic cell radiosensitization delivery.
Collapse
Affiliation(s)
- Zhenkun Zong
- a Nanjing Medical University , Nanjing , China.,b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China.,c Department of Neurosurgery , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Lei Hua
- a Nanjing Medical University , Nanjing , China.,b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China.,c Department of Neurosurgery , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Zhen Wang
- b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China
| | - Haoyue Xu
- b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China
| | - Chengkun Ye
- b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China
| | - Bomin Pan
- b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China
| | - Zongren Zhao
- b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China
| | - Longzhen Zhang
- d Department of Radiation Oncology , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China
| | - Jun Lu
- e Key Laboratory for Biotechnology on Medicinal Plants of Jiangsu Province School of Life Science , Jiangsu Normal University , Xuzhou , China
| | - Liu Hong Mei
- b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China.,c Department of Neurosurgery , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China.,f Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy , Cancer Institute, Xuzhou Medical University , Xuzhou , China
| | - Yu Rutong
- a Nanjing Medical University , Nanjing , China.,b Institute of Nervous System Diseases , Xuzhou Medical University , Xuzhou , China.,c Department of Neurosurgery , Affiliated Hospital of Xuzhou Medical University , Xuzhou , China.,f Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy , Cancer Institute, Xuzhou Medical University , Xuzhou , China
| |
Collapse
|
32
|
Dave V, Tak K, Sohgaura A, Gupta A, Sadhu V, Reddy KR. Lipid-polymer hybrid nanoparticles: Synthesis strategies and biomedical applications. J Microbiol Methods 2019; 160:130-142. [DOI: 10.1016/j.mimet.2019.03.017] [Citation(s) in RCA: 88] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2019] [Revised: 03/09/2019] [Accepted: 03/17/2019] [Indexed: 11/28/2022]
|
33
|
Flores AM, Ye J, Jarr KU, Hosseini-Nassab N, Smith BR, Leeper NJ. Nanoparticle Therapy for Vascular Diseases. Arterioscler Thromb Vasc Biol 2019; 39:635-646. [PMID: 30786744 PMCID: PMC6436996 DOI: 10.1161/atvbaha.118.311569] [Citation(s) in RCA: 96] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Nanoparticles promise to advance strategies to treat vascular disease. Since being harnessed by the cancer field to deliver safer and more effective chemotherapeutics, nanoparticles have been translated into applications for cardiovascular disease. Systemic exposure and drug-drug interactions remain a concern for nearly all cardiovascular therapies, including statins, antithrombotic, and thrombolytic agents. Moreover, off-target effects and poor bioavailability have limited the development of completely new approaches to treat vascular disease. Through the rational design of nanoparticles, nano-based delivery systems enable more efficient delivery of a drug to its therapeutic target or even directly to the diseased site, overcoming biological barriers and enhancing a drug's therapeutic index. In addition, advances in molecular imaging have led to the development of theranostic nanoparticles that may simultaneously act as carriers of both therapeutic and imaging payloads. The following is a summary of nanoparticle therapy for atherosclerosis, thrombosis, and restenosis and an overview of recent major advances in the targeted treatment of vascular disease.
Collapse
Affiliation(s)
- Alyssa M. Flores
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
- Geisel School of Medicine at Dartmouth, Hanover, NH
| | - Jianqin Ye
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
| | - Kai-Uwe Jarr
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
| | - Niloufar Hosseini-Nassab
- Department of Radiology, Stanford University School of Medicine, Michigan State University, East Lansing, MI, USA
| | - Bryan R. Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI, USA
| | - Nicholas J. Leeper
- Department of Surgery, Division of Vascular Surgery, Stanford University School of Medicine., Hanover, NH
- Department of Medicine, Division of Cardiovascular Medicine, Stanford University School of Medicine, Stanford, CA
- Stanford Cardiovascular Institute, Stanford, CA 94305, USA
| |
Collapse
|
34
|
Mo X, Yan F, Zhang B. Molecular Ultrasound Monitoring of Early Artery Injury After Carotid Balloon Angioplasty. Front Pharmacol 2019; 9:1569. [PMID: 30740054 PMCID: PMC6355681 DOI: 10.3389/fphar.2018.01569] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Accepted: 12/24/2018] [Indexed: 12/26/2022] Open
Abstract
Cardiovascular intervention is a common treatment procedure for many cardiovascular diseases. But restenosis often occurs after these procedures, greatly discounting their long-term therapeutic effects. Early detection of endothelial denudation is helpful for the diagnosis and prevention of restenosis. Here, we fabricated targeted microbubbles by conjugating anti-collagen IV antibodies to the surface of biotinylated microbubbles (MBColIV) and applied them for ultrasound molecular imaging of endothelial injury at early stage. Our results showed that the MBColIV, with a typical multi-peak particle distribution, was successfully constructed, which was confirmed by Alexa Fluor® 555-labeled secondary antibody. Ex vivo adhesion of microbubbles revealed that MBColIV can effectively and specially bind to the surface of balloon-injured carotid artery. The in vivo animal experiments showed ultrasound molecular imaging signals from carotid artery-injured rats administrated with MBColIV were significantly higher than those administrated with isotype control microbubbles. Histological staining of the left carotid common artery revealed that collagen IV was obviously exposed after endothelium denudation in balloon-injured artery. In conclusion, our current study provides an effective approach to detect vascular injury at the early stage and a potential platform for image-guided therapy to vascular injury.
Collapse
Affiliation(s)
- Xinhai Mo
- Department of Ultrasound in Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| | - Fei Yan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Bo Zhang
- Department of Ultrasound in Medicine, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, China
| |
Collapse
|
35
|
Cervadoro A, Palomba R, Vergaro G, Cecchi R, Menichetti L, Decuzzi P, Emdin M, Luin S. Targeting Inflammation With Nanosized Drug Delivery Platforms in Cardiovascular Diseases: Immune Cell Modulation in Atherosclerosis. Front Bioeng Biotechnol 2018; 6:177. [PMID: 30542650 PMCID: PMC6277804 DOI: 10.3389/fbioe.2018.00177] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Accepted: 11/06/2018] [Indexed: 12/18/2022] Open
Abstract
Atherosclerosis (AS) is a disorder of large and medium-sized arteries; it consists in the formation of lipid-rich plaques in the intima and inner media, whose pathophysiology is mostly driven by inflammation. Currently available interventions and therapies for treating atherosclerosis are not always completely effective; side effects associated with treatments, mainly caused by immunodepression for anti-inflammatory molecules, limit the systemic administration of these and other drugs. Given the high degree of freedom in the design of nanoconstructs, in the last decades researchers have put high effort in the development of nanoparticles (NPs) formulations specifically designed for either drug delivery, visualization of atherosclerotic plaques, or possibly the combination of both these and other functionalities. Here we will present the state of the art of these subjects, the knowledge of which is necessary to rationally address the use of NPs for prevention, diagnosis, and/or treatment of AS. We will analyse the work that has been done on: (a) understanding the role of the immune system and inflammation in cardiovascular diseases, (b) the pathological and biochemical principles in atherosclerotic plaque formation, (c) the latest advances in the use of NPs for the recognition and treatment of cardiovascular diseases, (d) the cellular and animal models useful to study the interactions of NPs with the immune system cells.
Collapse
Affiliation(s)
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Giuseppe Vergaro
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Roberta Cecchi
- NEST Laboratory, Scuola Normale Superiore, Pisa, Italy.,Center for Nanotechnology Innovation (CNI@NEST), Istituto Italiano di Tecnologia, Pisa, Italy
| | | | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genova, Italy
| | - Michele Emdin
- Division of Cardiology and Cardiovascular Medicine, Fondazione Toscana Gabriele Monasterio, Pisa, Italy.,Institute of Life Sciences, Scuola Superiore Sant'Anna, Pisa, Italy
| | - Stefano Luin
- NEST Laboratory, Scuola Normale Superiore, Pisa, Italy.,NEST Laboratory, Istituto Nanoscienze, CNR, Pisa, Italy
| |
Collapse
|
36
|
Taneja G, Sud A, Pendse N, Panigrahi B, Kumar A, Sharma AK. Nano-medicine and Vascular Endothelial Dysfunction: Options and Delivery Strategies. Cardiovasc Toxicol 2018; 19:1-12. [DOI: 10.1007/s12012-018-9491-x] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
37
|
Li T, Liang W, Xiao X, Qian Y. Nanotechnology, an alternative with promising prospects and advantages for the treatment of cardiovascular diseases. Int J Nanomedicine 2018; 13:7349-7362. [PMID: 30519019 PMCID: PMC6233477 DOI: 10.2147/ijn.s179678] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Cardiovascular diseases (CVDs) are one of the most important causes of mortality and affecting the health status of patients. At the same time, CVDs cause a huge health and economic burden to the whole world. Although a variety of therapeutic drugs and measures have been produced to delay the progress of the disease and improve the quality of life of patients, most of the traditional therapeutic strategies can only cure the symptoms and cannot repair or regenerate the damaged ischemic myocardium. In addition, they may bring some unpleasant side effects. Therefore, it is vital to find and explore new technologies and drugs to solve the shortcomings of conventional treatments. Nanotechnology is a new way of using and manipulating the matter at the molecular scale, whose functional organization is measured in nanometers. Because nanoscale phenomena play an important role in cell signal transduction, enzyme action and cell cycle, nanotechnology is closely related to medical research. The application of nanotechnology in the field of medicine provides an alternative and novel direction for the treatment of CVDs, and shows excellent performance in the field of targeted drug therapy and the development of biomaterials. This review will briefly introduce the latest applications of nanotechnology in the diagnosis and treatment of common CVDs.
Collapse
Affiliation(s)
- Tao Li
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China,
| | - Weitao Liang
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China,
| | - Xijun Xiao
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China,
| | - Yongjun Qian
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, China,
| |
Collapse
|
38
|
Grad E, Zolotarevsky K, Danenberg HD, Nordling-David MM, Gutman D, Golomb G. The role of monocyte subpopulations in vascular injury following partial and transient depletion. Drug Deliv Transl Res 2018; 8:945-953. [PMID: 28656488 DOI: 10.1007/s13346-017-0404-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The innate immunity system plays a critical role in vascular repair and restenosis development. Liposomes encapsulating bisphosphonates (LipBPs), but not free BPs, suppress neointima formation following vascular injury mediated in part by monocytes. The objective of this study was to elucidate the role of monocyte subpopulations on vascular healing following LipBP treatment. The potency- and dose-dependent treatment effect of clodronate (CLOD) and alendronate (ALN) liposomes on restenosis inhibition, total monocyte depletion, and monocytes subpopulation was studied. Rats subjected to carotid injury were treated by a single IV injection of LipBPs at the time of injury. Low- and high-dose LipALN treatment (3 and 10 mg/kg, respectively) resulted in a dose-dependent effect on restenosis development after 30 days. Both doses of LipALN resulted in a dose-dependent inhibition of restenosis, but only high dose of LipALN depleted monocytes (-60.1 ± 4.4%, 48 h post injury). Although LipCLOD treatment (at an equivalent potency to 3 mg/kg alendronate) significantly reduced monocyte levels (72.1 ± 6%), no restenosis inhibition was observed. The major finding of this study is the correlation found between monocyte subclasses and restenosis inhibition. Non-classical monocyte (NCM) levels were found higher in LipALN-treated rats, but lower in LipCLOD-treated rats, 24 h after injury and treatment. We suggest that the inhibition of circulating monocyte subpopulations is the predominant mechanism by which LipBPs prevent restenosis. The effect of LipBP treatment on the monocyte subpopulation correlates with the dose and potency of LipBPs.
Collapse
Affiliation(s)
- Etty Grad
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel
| | - Ksenia Zolotarevsky
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel
| | - Haim D Danenberg
- Cardiovascular Research Center, Hadassah Hebrew University Medical Center, The Hebrew University of Jerusalem, 91120, Jerusalem, Israel
| | - Mirjam M Nordling-David
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel
| | - Dikla Gutman
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel
| | - Gershon Golomb
- Institute for Drug Research, School of Pharmacy, Faculty of Medicine, The Hebrew University of Jerusalem, 12065 Ein Kerem Medical Cenre, 91120, Jerusalem, Israel.
| |
Collapse
|
39
|
Garg NK, Tandel N, Jadon RS, Tyagi RK, Katare OP. Lipid-polymer hybrid nanocarrier-mediated cancer therapeutics: current status and future directions. Drug Discov Today 2018; 23:1610-1621. [PMID: 29857164 DOI: 10.1016/j.drudis.2018.05.033] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Revised: 04/23/2018] [Accepted: 05/23/2018] [Indexed: 02/07/2023]
Abstract
The new generation of nanoparticles (NPs) encompass attributes of lipids and polymers and are referred to as 'lipid-polymer hybrid nanoparticles' (LPHNPs). LPHNPs have helped shed light on the mechanisms involved in targeted and non-specific drug delivery. Research has also highlighted the opportunities and challenges faced by the use of nanomedicine as personalized therapies in oncology. Here, we review the development of LPHNPs as cancer therapeutics, focusing on the methods deployed for enhancing the targeting efficiency and applications of LPHNPs.
Collapse
Affiliation(s)
- Neeraj K Garg
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India; Department of Endocrinology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh 160012, India.
| | - Nikunj Tandel
- Institute of Science, Nirma University Ahmedabad, Gujarat 382481, India
| | - Rajesh S Jadon
- School of Studies, Jiwaji University, Gwalior 474002, India
| | - Rajeev K Tyagi
- Biomedical Parasitology and Nano-immunology Lab, Amity Institute of Microbial Technology, Amity University, Noida, India; Department of Periodontics, College of Dental Medicine, Augusta University, Augusta, GA 30912, USA.
| | - Om P Katare
- University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh 160014, India
| |
Collapse
|
40
|
Gubala V, Johnston LJ, Krug HF, Moore CJ, Ober CK, Schwenk M, Vert M. Engineered nanomaterials and human health: Part 2. Applications and nanotoxicology (IUPAC Technical Report). PURE APPL CHEM 2018. [DOI: 10.1515/pac-2017-0102] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
AbstractResearch on engineered nanomaterials (ENM) has progressed rapidly from the very early stages of studying their unique, size-dependent physicochemical properties and commercial exploration to the development of products that influence our everyday lives. We have previously reviewed various methods for synthesis, surface functionalization, and analytical characterization of ENM in a publication titled ‘Engineered Nanomaterials: Preparation, Functionalization and Characterization’. In this second, inter-linked document, we first provide an overview of important applications of ENM in products relevant to human healthcare and consumer goods, such as food, textiles, and cosmetics. We then highlight the challenges for the design and development of new ENM for bio-applications, particularly in the rapidly developing nanomedicine sector. The second part of this document is dedicated to nanotoxicology studies of ENM in consumer products. We describe the various biological targets where toxicity may occur, summarize the four nanotoxicology principles, and discuss the need for careful consideration of the biodistribution, degradation, and elimination routes of nanosized materials before they can be safely used. Finally, we review expert opinions on the risk, regulation, and ethical aspects of using engineered nanomaterials in applications that may have direct or indirect impact on human health or our environment.
Collapse
|
41
|
Kandaswamy E, Zuo L. Recent Advances in Treatment of Coronary Artery Disease: Role of Science and Technology. Int J Mol Sci 2018; 19:ijms19020424. [PMID: 29385089 PMCID: PMC5855646 DOI: 10.3390/ijms19020424] [Citation(s) in RCA: 77] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2017] [Revised: 01/06/2018] [Accepted: 01/15/2018] [Indexed: 12/11/2022] Open
Abstract
Coronary artery disease (CAD) is one of the most common causes of death worldwide. In the last decade, significant advancements in CAD treatment have been made. The existing treatment is medical, surgical or a combination of both depending on the extent, severity and clinical presentation of CAD. The collaboration between different science disciplines such as biotechnology and tissue engineering has led to the development of novel therapeutic strategies such as stem cells, nanotechnology, robotic surgery and other advancements (3-D printing and drugs). These treatment modalities show promising effects in managing CAD and associated conditions. Research on stem cells focuses on studying the potential for cardiac regeneration, while nanotechnology research investigates nano-drug delivery and percutaneous coronary interventions including stent modifications and coatings. This article aims to provide an update on the literature (in vitro, translational, animal and clinical) related to these novel strategies and to elucidate the rationale behind their potential treatment of CAD. Through the extensive and continued efforts of researchers and clinicians worldwide, these novel strategies hold the promise to be effective alternatives to existing treatment modalities.
Collapse
Affiliation(s)
- Eswar Kandaswamy
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| | - Li Zuo
- Radiologic Sciences and Respiratory Therapy Division, School of Health and Rehabilitation Sciences, The Ohio State University College of Medicine, Columbus, OH 43210, USA.
| |
Collapse
|
42
|
Chan CKW, Zhang L, Cheng CK, Yang H, Huang Y, Tian XY, Choi CHJ. Recent Advances in Managing Atherosclerosis via Nanomedicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:1702793. [PMID: 29239134 DOI: 10.1002/smll.201702793] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2017] [Revised: 10/15/2017] [Indexed: 06/07/2023]
Abstract
Atherosclerosis, driven by chronic inflammation of the arteries and lipid accumulation on the blood vessel wall, underpins many cardiovascular diseases with high mortality rates globally, such as stroke and ischemic heart disease. Engineered bio-nanomaterials are now under active investigation as carriers of therapeutic and/or imaging agents to atherosclerotic plaques. This Review summarizes the latest bio-nanomaterial-based strategies for managing atherosclerosis published over the past five years, a period marked by a rapid surge in preclinical applications of bio-nanomaterials for imaging and/or treating atherosclerosis. To start, the biomarkers exploited by emerging bio-nanomaterials for targeting various components of atherosclerotic plaques are outlined. In addition, recent efforts to rationally design and screen for bio-nanomaterials with the optimal physicochemical properties for targeting plaques are presented. Moreover, the latest preclinical applications of bio-nanomaterials as carriers of imaging, therapeutic, or theranostic agents to atherosclerotic plaques are discussed. Finally, a mechanistic understanding of the interactions between bio-nanomaterials and the plaque ("athero-nano" interactions) is suggested, the opportunities and challenges in the clinical translation of bio-nanomaterials for managing atherosclerosis are discussed, and recent clinical trials for atherosclerotic nanomedicines are introduced.
Collapse
Affiliation(s)
- Cecilia Ka Wing Chan
- Department of Surgery, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Lei Zhang
- Department of Biomedical Engineering, Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chak Kwong Cheng
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Hongrong Yang
- Department of Biomedical Engineering, Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Yu Huang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| | - Chung Hang Jonathan Choi
- Department of Biomedical Engineering, Shun Hing Institute of Advanced Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong, China
| |
Collapse
|
43
|
Ahmaditabar P, Momtazi-Borojeni AA, Rezayan AH, Mahmoodi M, Sahebkar A, Mellat M. Enhanced Entrapment and Improved in Vitro Controlled Release of N-Acetyl Cysteine in Hybrid PLGA/Lecithin Nanoparticles Prepared Using a Nanoprecipitation/Self-Assembly Method. J Cell Biochem 2017; 118:4203-4209. [PMID: 28419535 DOI: 10.1002/jcb.26070] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Accepted: 04/14/2017] [Indexed: 12/22/2022]
Abstract
To enhance the in vitro controlled release of N-acetyl cysteine (NAC), hybrid nanoparticles (NPs) consisting of a poly(lactide-co-glycolide) (PLGA) hydrophobic core and a soybean lecithin mono-layer coat were prepared. Hybrid NPs were synthesized using a nanoprecipitation combined with self-assembly method. To characterize prepared NPs, zeta potential, diameter size, surface morphology, disparity, and lipid coating of hybrid NPs were detrmined using dynamic light scattering, scanning electron microscope and Fourier transform infrared spectroscopy techniques. High-performance liquid chromatography was employed to evaluate drug loading yield and encapsulation efficiency and in vitro drug release of prepared NPs. The cytotoxicity of hybrid NPs was assayed on normal L929 alveolar epithelial cells using MTT method. Prepared NPs were found to disperse as individual NPs with a well-defined spherical shape. The hydrodynamic diameter and surface charge of NAC-loaded hybrid NPs were 81.8 ± 1.3 nm and -33.1 ± 2.1 mV, respectively. Drug loading yield and encapsulation efficiency of NAC-loaded hybrid NPs were found to be 38 ± 2.1% and 67 ± 5.7%, respectively. Prepared hybrid NPs showed no significant cytotoxicity against normal alveolar cells. Our data suggest that the hybrid PLGA-lecithin NPs may be An efficient controlled release drug delivery system for NAC. J. Cell. Biochem. 118: 4203-4209, 2017. © 2017 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Parvin Ahmaditabar
- Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Amir A Momtazi-Borojeni
- Nanotechnology Research Center, Student Research Committee, Department of Medical Biotechnology, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ali H Rezayan
- Department of Life Science Engineering, Faculty of New Sciences and Technologies, University of Tehran, Tehran, Iran
| | - Mahboobeh Mahmoodi
- Department of Biomedical Engineering, Yazd Branch, Islamic Azad University, Yazd, Iran
| | - Amirhossein Sahebkar
- Biotechnology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Mostafa Mellat
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| |
Collapse
|
44
|
Wu B, Lu ST, Zhang LJ, Zhuo RX, Xu HB, Huang SW. Codelivery of doxorubicin and triptolide with reduction-sensitive lipid-polymer hybrid nanoparticles for in vitro and in vivo synergistic cancer treatment. Int J Nanomedicine 2017; 12:1853-1862. [PMID: 28331310 PMCID: PMC5352248 DOI: 10.2147/ijn.s131235] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
Codelivery is a promising strategy to overcome the limitations of single chemotherapeutic agents in cancer treatment. Despite progress, codelivery of two or more different functional drugs to increase anticancer efficiency still remains a challenge. Here, reduction-sensitive lipid–polymer hybrid nanoparticles (LPNPs) drug delivery system composed of monomethoxy-poly(ethylene glycol)-S-S-hexadecyl (mPEG-S-S-C16), soybean lecithin, and poly(D,L-lactide-co-glycolide) (PLGA) was used for codelivery of doxorubicin (DOX) and a Chinese herb extract triptolide (TPL). Hydrophobic DOX and TPL could be successfully loaded in LPNPs by self-assembly. More importantly, drug release and cellular uptake experiments demonstrated that the two drugs were reduction sensitive, released simultaneously from LPNPs, and taken up effectively by the tumor cells. DOX/TPL-coloaded LPNPs (DOX/TPL-LPNPs) exhibited a high level of synergistic activation with low combination index (CI) in vitro and in vivo. Moreover, the highest synergistic therapeutic effect was achieved at the ratio of 1:0.2 DOX/TPL. Further experiments showed that TPL enhanced the uptake of DOX by human oral cavity squamous cell carcinoma cells (KB cells). Overall, DOX/TPL-coencapsulated reduction-sensitive nanoparticles will be a promising strategy for cancer treatment.
Collapse
Affiliation(s)
- Bo Wu
- Department of Radiology, Zhongnan Hospital of Wuhan University; Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, People's Republic of China
| | - Shu-Ting Lu
- Department of Radiology, Zhongnan Hospital of Wuhan University
| | - Liu-Jie Zhang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, People's Republic of China
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, People's Republic of China
| | - Hai-Bo Xu
- Department of Radiology, Zhongnan Hospital of Wuhan University
| | - Shi-Wen Huang
- Key Laboratory of Biomedical Polymers of Ministry of Education, Department of Chemistry, Wuhan University, Wuhan, People's Republic of China
| |
Collapse
|
45
|
Ambesh P, Campia U, Obiagwu C, Bansal R, Shetty V, Hollander G, Shani J. Nanomedicine in coronary artery disease. Indian Heart J 2017; 69:244-251. [PMID: 28460774 PMCID: PMC5414944 DOI: 10.1016/j.ihj.2017.02.007] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2016] [Revised: 01/21/2017] [Accepted: 02/10/2017] [Indexed: 12/25/2022] Open
Abstract
Nanomedicine is one of the most promising therapeutic modalities researchers are working on. It involves development of drugs and devices that work at the nanoscale (10-9m). Coronary artery disease (CAD) is responsible for more than a third of all deaths in age group >35 years. With such a huge burden of mortality, CAD is one of the diseases where nanomedicine is being employed for preventive and therapeutic interventions. Nanomedicine can effectively deliver focused drug payload at sites of local plaque formation. Non-invasive strategies include thwarting angiogenesis, intra-arterial thrombosis and local inflammation. Invasive strategies following percutaneous coronary intervention (PCI) include anti-restenosis and healing enhancement. However, before practical application becomes widespread, many challenges need to be dealt with. These include manufacturing at the nanoscale, direct nanomaterial cellular toxicity and visualization.
Collapse
Affiliation(s)
- Paurush Ambesh
- Department of Internal Medicine, Maimonides Medical Center, New York City, USA.
| | - Umberto Campia
- Department of Cardiology, Brigham and Women's Hospital, Boston, USA
| | - Chukwudi Obiagwu
- Department of Cardiology, Maimonides Medical Center, New York City, USA
| | - Rashika Bansal
- Department of Internal Medicine, St. Joseph Regional Medical Center, NJ, USA
| | - Vijay Shetty
- Department of Cardiology, Maimonides Medical Center, New York City, USA
| | - Gerald Hollander
- Department of Cardiology, Maimonides Medical Center, New York City, USA
| | - Jacob Shani
- Department of Cardiology, Maimonides Medical Center, New York City, USA
| |
Collapse
|
46
|
Meyers MW, Rink JS, Jiang Q, Kelly ME, Vercammen JM, Thaxton CS, Kibbe MR. Systemically administered collagen-targeted gold nanoparticles bind to arterial injury following vascular interventions. Physiol Rep 2017; 5:e13128. [PMID: 28242820 PMCID: PMC5328771 DOI: 10.14814/phy2.13128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/14/2016] [Accepted: 12/17/2016] [Indexed: 11/24/2022] Open
Abstract
Surgical and endovascular therapies for severe atherosclerosis often fail due to the development of neointimal hyperplasia and arterial restenosis. Our objective was to synthesize, characterize, and evaluate the targeting specificity and biocompatibility of a novel systemically injected nanoparticle. We hypothesize that surface-functionalization of gold nanoparticles (AuNPs) with a collagen-targeting peptide will be biocompatible and target specifically to vascular injury. 13 nm AuNPs were surface functionalized with a peptide-molecular fluorophore and targeted to collagen (T-AuNP) or a scrambled peptide sequence (S-AuNP). After rat carotid artery balloon injury and systemic injection of T-AuNP or S-AuNP, arteries and organs were harvested and assessed for binding specificity and biocompatibility. The T-AuNP bound with specificity to vascular injury for a minimum of 24 h. No significant inflammation was evident locally at arterial injury or systemically in major organs. The T-AuNP did not impact endothelial cell viability or induce apoptosis at the site of injury in vivo. No major changes were evident in hepatic or renal blood chemistry profiles. Herein, we synthesized a biocompatible nanoparticle that targets to vascular injury following systemic administration. These studies demonstrate proof-of-principle and serve as the foundation for further T-AuNP optimization to realize systemic, targeted delivery of therapeutics to the sites of vascular injury.
Collapse
Affiliation(s)
- Molly Wasserman Meyers
- Department of Surgery, Northwestern University, Chicago, Illinois
- Simpson Querrey Institute for BioNanotechnology, Chicago, Illinois
| | - Jonathan S Rink
- Simpson Querrey Institute for BioNanotechnology, Chicago, Illinois
- Department of Urology, Northwestern University, Chicago, Illinois
| | - Qun Jiang
- Department of Surgery, Northwestern University, Chicago, Illinois
- Simpson Querrey Institute for BioNanotechnology, Chicago, Illinois
| | - Megan E Kelly
- Department of Surgery, Northwestern University, Chicago, Illinois
- Simpson Querrey Institute for BioNanotechnology, Chicago, Illinois
| | - Janet M Vercammen
- Department of Surgery, Northwestern University, Chicago, Illinois
- Simpson Querrey Institute for BioNanotechnology, Chicago, Illinois
| | - Colby S Thaxton
- Simpson Querrey Institute for BioNanotechnology, Chicago, Illinois
- Department of Urology, Northwestern University, Chicago, Illinois
| | - Melina R Kibbe
- Department of Surgery, Northwestern University, Chicago, Illinois
- Simpson Querrey Institute for BioNanotechnology, Chicago, Illinois
- Department of Surgery, University of North Carolina, Chapel Hill, North Carolina
| |
Collapse
|
47
|
Zhang RX, Ahmed T, Li LY, Li J, Abbasi AZ, Wu XY. Design of nanocarriers for nanoscale drug delivery to enhance cancer treatment using hybrid polymer and lipid building blocks. NANOSCALE 2017; 9:1334-1355. [PMID: 27973629 DOI: 10.1039/c6nr08486a] [Citation(s) in RCA: 97] [Impact Index Per Article: 12.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Polymer-lipid hybrid nanoparticles (PLN) are an emerging nanocarrier platform made from building blocks of polymers and lipids. PLN integrate the advantages of biomimetic lipid-based nanoparticles (i.e. solid lipid nanoparticles and liposomes) and biocompatible polymeric nanoparticles. PLN are constructed from diverse polymers and lipids and their numerous combinations, which imparts PLN with great versatility for delivering drugs of various properties to their nanoscale targets. PLN can be classified into two types based on their hybrid nanoscopic structure and assembly methods: Type-I monolithic matrix and Type-II core-shell systems. This article reviews the history of PLN development, types of PLN, lipid and polymer candidates, fabrication methods, and unique properties of PLN. The applications of PLN in delivery of therapeutic or imaging agents alone or in combination for cancer treatment are summarized and illustrated with examples. Important considerations for the rational design of PLN for advanced nanoscale drug delivery are discussed, including selection of excipients, synthesis processes governing formulation parameters, optimization of nanoparticle properties, improvement of particle surface functionality to overcome macroscopic, microscopic and cellular biological barriers. Future directions and potential clinical translation of PLN are also suggested.
Collapse
Affiliation(s)
- Rui Xue Zhang
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Taksim Ahmed
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Lily Yi Li
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Jason Li
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Azhar Z Abbasi
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| | - Xiao Yu Wu
- Advanced Pharmaceutics and Drug Delivery Laboratory, Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON, CanadaM5S 3M2.
| |
Collapse
|
48
|
Recent advances in the design, development, and targeting mechanisms of polymeric micelles for delivery of siRNA in cancer therapy. Prog Polym Sci 2017. [DOI: 10.1016/j.progpolymsci.2016.09.008] [Citation(s) in RCA: 127] [Impact Index Per Article: 15.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
49
|
Quinteros DA, Bermúdez JM, Ravetti S, Cid A, Allemandi DA, Palma SD. Therapeutic use of monoclonal antibodies: general aspects and challenges for drug delivery. NANOSTRUCTURES FOR DRUG DELIVERY 2017. [PMCID: PMC7151974 DOI: 10.1016/b978-0-323-46143-6.00025-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Monoclonal antibodies are routinely used in several fields but the great challenge has been their use as therapeutic agents for the treatment of diseases, such as breast cancer, leukemia, asthma, macular degeneration, arthritis, Crohn’s disease, and transplants, among others. Monoclonal antibodies are protein molecules made in the laboratory from hybridoma cells by recombinant DNA technology. Important advances have been made over the past decade to improve some critical points, such as safety and efficacy of the first generation of therapeutic antibodies. This type of molecules presents a significant challenge from the pharmaceutical point of view due to their characteristics, such as molecular size, stability, and solubility. In this chapter we have attempted to identify the major issues associated with therapeutic approaches, formulating drawbacks and delivering antibody drugs, particularly focused on the challenges and opportunities that these present for the future.
Collapse
Affiliation(s)
| | | | | | - Alicia Cid
- National University of Córdoba, Córdoba, Argentina
| | | | | |
Collapse
|
50
|
Zhang LJ, Wu B, Zhou W, Wang CX, Wang Q, Yu H, Zhuo RX, Liu ZL, Huang SW. Two-component reduction-sensitive lipid–polymer hybrid nanoparticles for triggered drug release and enhanced in vitro and in vivo anti-tumor efficacy. Biomater Sci 2017; 5:98-110. [DOI: 10.1039/c6bm00662k] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Two-component reduction-sensitive lipid–polymer hybrid nanoparticles composed of DLPE-S-S-MPEG and PCL were developed for intracellular reduction triggered delivery of DOX.
Collapse
Affiliation(s)
- Liu-Jie Zhang
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| | - Bo Wu
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| | - Wei Zhou
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| | - Cai-Xia Wang
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| | - Qian Wang
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| | - Hui Yu
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| | - Ren-Xi Zhuo
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| | - Zhi-Lan Liu
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| | - Shi-Wen Huang
- Key Laboratory of Biomedical Polymers
- Ministry of Education
- College of Chemistry and Molecular Sciences
- Wuhan University
- Wuhan 430072
| |
Collapse
|