1
|
Zong Q, He C, Long B, Huang Q, Chen Y, Li Y, Dang Y, Cai C. Targeted Delivery of Nanoparticles to Blood Vessels for the Treatment of Atherosclerosis. Biomedicines 2024; 12:1504. [PMID: 39062077 PMCID: PMC11275173 DOI: 10.3390/biomedicines12071504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 06/21/2024] [Accepted: 07/04/2024] [Indexed: 07/28/2024] Open
Abstract
Atherosclerosis is a common form of cardiovascular disease, which is one of the most prevalent causes of death worldwide, particularly among older individuals. Surgery is the mainstay of treatment for severe stenotic lesions, though the rate of restenosis remains relatively high. Current medication therapy for atherosclerosis has limited efficacy in reversing the formation of atherosclerotic plaques. The search for new drug treatment options is imminent. Some potent medications have shown surprising therapeutic benefits in inhibiting inflammation and endothelial proliferation in plaques. Unfortunately, their use is restricted due to notable dose-dependent systemic side effects or degradation. Nevertheless, with advances in nanotechnology, an increasing number of nano-related medical applications are emerging, such as nano-drug delivery, nano-imaging, nanorobots, and so forth, which allow for restrictions on the use of novel atherosclerotic drugs to be lifted. This paper reviews new perspectives on the targeted delivery of nanoparticles to blood vessels for the treatment of atherosclerosis in both systemic and local drug delivery. In systemic drug delivery, nanoparticles inhibit drug degradation and reduce systemic toxicity through passive and active pathways. To further enhance the precise release of drugs, the localized delivery of nanoparticles can also be accomplished through blood vessel wall injection or using endovascular interventional devices coated with nanoparticles. Overall, nanotechnology holds boundless potential for the diagnosis and treatment of atherosclerotic diseases in the future.
Collapse
Affiliation(s)
- Qiushuo Zong
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chengyi He
- Department of Vascular Surgery, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China;
| | - Binbin Long
- Department of General Surgery, Taihe Hospital Affiliated to Hubei University of Medicine, Shiyan 442099, China;
| | - Qingyun Huang
- Department of Cardiothoracic Surgery, The First Hospital of Putian Affiliated to Fujian Medical University, Putian 351106, China;
| | - Yunfei Chen
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiqing Li
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Yiping Dang
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| | - Chuanqi Cai
- Department of Vascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (Q.Z.); (Y.C.); (Y.L.)
| |
Collapse
|
2
|
Wang S, He H, Mao Y, Zhang Y, Gu N. Advances in Atherosclerosis Theranostics Harnessing Iron Oxide-Based Nanoparticles. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308298. [PMID: 38368274 PMCID: PMC11077671 DOI: 10.1002/advs.202308298] [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: 11/01/2023] [Revised: 02/06/2024] [Indexed: 02/19/2024]
Abstract
Atherosclerosis, a multifaceted chronic inflammatory disease, has a profound impact on cardiovascular health. However, the critical limitations of atherosclerosis management include the delayed detection of advanced stages, the intricate assessment of plaque stability, and the absence of efficacious therapeutic strategies. Nanotheranostic based on nanotechnology offers a novel paradigm for addressing these challenges by amalgamating advanced imaging capabilities with targeted therapeutic interventions. Meanwhile, iron oxide nanoparticles have emerged as compelling candidates for theranostic applications in atherosclerosis due to their magnetic resonance imaging capability and biosafety. This review delineates the current state and prospects of iron oxide nanoparticle-based nanotheranostics in the realm of atherosclerosis, including pivotal aspects of atherosclerosis development, the pertinent targeting strategies involved in disease pathogenesis, and the diagnostic and therapeutic roles of iron oxide nanoparticles. Furthermore, this review provides a comprehensive overview of theranostic nanomedicine approaches employing iron oxide nanoparticles, encompassing chemical therapy, physical stimulation therapy, and biological therapy. Finally, this review proposes and discusses the challenges and prospects associated with translating these innovative strategies into clinically viable anti-atherosclerosis interventions. In conclusion, this review offers new insights into the future of atherosclerosis theranostic, showcasing the remarkable potential of iron oxide-based nanoparticles as versatile tools in the battle against atherosclerosis.
Collapse
Affiliation(s)
- Shi Wang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Hongliang He
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Yu Mao
- School of MedicineNanjing UniversityNanjing210093P. R. China
| | - Yu Zhang
- State Key Laboratory of Digital Medical EngineeringJiangsu Key Laboratory for Biomaterials and DevicesSchool of Biological Sciences & Medical EngineeringSoutheast UniversityNanjing210009P. R. China
| | - Ning Gu
- School of MedicineNanjing UniversityNanjing210093P. R. China
| |
Collapse
|
3
|
Lungu CN, Creteanu A, Mehedinti MC. Endovascular Drug Delivery. Life (Basel) 2024; 14:451. [PMID: 38672722 PMCID: PMC11051410 DOI: 10.3390/life14040451] [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: 02/06/2024] [Revised: 03/12/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024] Open
Abstract
Drug-eluting stents (DES) and balloons revolutionize atherosclerosis treatment by targeting hyperplastic tissue responses through effective local drug delivery strategies. This review examines approved and emerging endovascular devices, discussing drug release mechanisms and their impacts on arterial drug distribution. It emphasizes the crucial role of drug delivery in modern cardiovascular care and highlights how device technologies influence vascular behavior based on lesion morphology. The future holds promise for lesion-specific treatments, particularly in the superficial femoral artery, with recent CE-marked devices showing encouraging results. Exciting strategies and new patents focus on local drug delivery to prevent restenosis, shaping the future of interventional outcomes. In summary, as we navigate the ever-evolving landscape of cardiovascular intervention, it becomes increasingly evident that the future lies in tailoring treatments to the specific characteristics of each lesion. By leveraging cutting-edge technologies and harnessing the potential of localized drug delivery, we stand poised to usher in a new era of precision medicine in vascular intervention.
Collapse
Affiliation(s)
- Claudiu N. Lungu
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania;
| | - Andreea Creteanu
- Department of Pharmaceutical Technology, University of Medicine and Pharmacy Grigore T Popa, 700115 Iași, Romania
| | - Mihaela C. Mehedinti
- Department of Functional and Morphological Science, Faculty of Medicine and Pharmacy, Dunarea de Jos University, 800010 Galati, Romania;
| |
Collapse
|
4
|
de Oliveira Laterza Ribeiro M, Correia VM, Herling de Oliveira LL, Soares PR, Scudeler TL. Evolving Diagnostic and Management Advances in Coronary Heart Disease. Life (Basel) 2023; 13:951. [PMID: 37109480 PMCID: PMC10143565 DOI: 10.3390/life13040951] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2023] [Revised: 04/02/2023] [Accepted: 04/03/2023] [Indexed: 04/29/2023] Open
Abstract
Despite considerable improvement in diagnostic modalities and therapeutic options over the last few decades, the global burden of ischemic heart disease is steadily rising, remaining a major cause of death worldwide. Thus, new strategies are needed to lessen cardiovascular events. Researchers in different areas such as biotechnology and tissue engineering have developed novel therapeutic strategies such as stem cells, nanotechnology, and robotic surgery, among others (3D printing and drugs). In addition, advances in bioengineering have led to the emergence of new diagnostic and prognostic techniques, such as quantitative flow ratio (QFR), and biomarkers for atherosclerosis. In this review, we explore novel diagnostic invasive and noninvasive modalities that allow a more detailed characterization of coronary disease. We delve into new technological revascularization procedures and pharmacological agents that target several residual cardiovascular risks, including inflammatory, thrombotic, and metabolic pathways.
Collapse
Affiliation(s)
| | | | | | | | - Thiago Luis Scudeler
- Instituto do Coração (InCor), Hospital das Clínicas da Faculdade de Medicina da Universidade de São Paulo, São Paulo 05403-010, Brazil
| |
Collapse
|
5
|
Mishra S, Bhatt T, Kumar H, Jain R, Shilpi S, Jain V. Nanoconstructs for theranostic application in cancer: Challenges and strategies to enhance the delivery. Front Pharmacol 2023; 14:1101320. [PMID: 37007005 PMCID: PMC10050349 DOI: 10.3389/fphar.2023.1101320] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/06/2023] [Indexed: 03/17/2023] Open
Abstract
Nanoconstructs are made up of nanoparticles and ligands, which can deliver the loaded cargo at the desired site of action. Various nanoparticulate platforms have been utilized for the preparation of nanoconstructs, which may serve both diagnostic as well as therapeutic purposes. Nanoconstructs are mostly used to overcome the limitations of cancer therapies, such as toxicity, nonspecific distribution of the drug, and uncontrolled release rate. The strategies employed during the design of nanoconstructs help improve the efficiency and specificity of loaded theranostic agents and make them a successful approach for cancer therapy. Nanoconstructs are designed with a sole purpose of targeting the requisite site, overcoming the barriers which hinders its right placement for desired benefit. Therefore, instead of classifying modes for delivery of nanoconstructs as actively or passively targeted systems, they are suitably classified as autonomous and nonautonomous types. At large, nanoconstructs offer numerous benefits, however they suffer from multiple challenges, too. Hence, to overcome such challenges computational modelling methods and artificial intelligence/machine learning processes are being explored. The current review provides an overview on attributes and applications offered by nanoconstructs as theranostic agent in cancer.
Collapse
Affiliation(s)
- Shivani Mishra
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
| | - Tanvi Bhatt
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
| | - Hitesh Kumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
| | - Rupshee Jain
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
| | - Satish Shilpi
- Department of Pharmaceutics, School of Pharmaceutical and Populations Health Informatics, DIT University, Dehradun, India
| | - Vikas Jain
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research, Mysuru, India
- *Correspondence: Vikas Jain,
| |
Collapse
|
6
|
Montelione N, Loreni F, Nenna A, Catanese V, Scurto L, Ferrisi C, Jawabra M, Gabellini T, Codispoti FA, Spinelli F, Chello M, Stilo F. Tissue Engineering and Targeted Drug Delivery in Cardiovascular Disease: The Role of Polymer Nanocarrier for Statin Therapy. Biomedicines 2023; 11:798. [PMID: 36979777 PMCID: PMC10045667 DOI: 10.3390/biomedicines11030798] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 02/21/2023] [Accepted: 02/24/2023] [Indexed: 03/09/2023] Open
Abstract
Atherosclerosis-related coronary artery disease (CAD) is the leading cause of mortality and morbidity worldwide. This requires effective primary and secondary prevention in reducing the complications related to CAD; the regression or stabilization of the pathology remains the mainstay of treatment. Statins have proved to be the most effective treatment in reducing adverse effects, but there are limitations related to the administration and achievement of effective doses as well as side effects due to the lack of target-related molecular specificity. The implemented technological steps are polymers and nanoparticles for the administration of statins, as it has been seen how the conjugation of drug delivery systems (DDSs) with statins increases bioavailability by circumventing the hepatic-renal filter and increases the related target specificity, enhancing their action and decreasing side effects. Reduction of endothelial dysfunction, reduced intimal hyperplasia, reduced ischemia-reperfusion injury, cardiac regeneration, positive remodeling in the extracellular matrix, reduced neointimal growth, and increased reendothelialization are all drug-related effects of statins enhanced by binding with DDSs. Recent preclinical studies demonstrate how the effect of statins stimulates the differentiation of endogenous cardiac stem cells. Poly-lactic-co-glycolic acid (PLGA) seems to be the most promising DDS as it succeeds more than the others in enhancing the effect of the bound drug. This review intends to summarize the current evidence on polymers and nanoparticles for statin delivery in the field of cardiovascular disease, trying to shed light on this topic and identify new avenues for future studies.
Collapse
Affiliation(s)
- Nunzio Montelione
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Francesco Loreni
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Antonio Nenna
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Vincenzo Catanese
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Lucia Scurto
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Chiara Ferrisi
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Mohamad Jawabra
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Teresa Gabellini
- Residency Program of Vascular and Endovascular Surgery, University of Ferrara, 44121 Ferrara, Italy
| | | | - Francesco Spinelli
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Massimo Chello
- Unit of Cardiac Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| | - Francesco Stilo
- Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
- Head of Research Unit of Vascular Surgery, Università Campus Bio-Medico di Roma, 00128 Rome, Italy
| |
Collapse
|
7
|
The Association of Patent Ductus Arteriosus with Inflammation: A Narrative Review of the Role of Inflammatory Biomarkers and Treatment Strategy in Premature Infants. Int J Mol Sci 2022; 23:ijms232213877. [PMID: 36430355 PMCID: PMC9699120 DOI: 10.3390/ijms232213877] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/27/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
Patent ductus arteriosus (PDA) is a common cardiovascular complication that complicates clinical care in the intensive care of premature infants. Prenatal and postnatal infections and the inflammation process can contribute to PDA, and intrauterine inflammation is a known risk factor of PDA. A variety of inflammatory biomarkers have been reported to be associated with PDA. Chorioamnionitis induces the fetal inflammatory process via several cytokines that have been reported to be associated with the presence of PDA and may have a role in the vascular remodeling process or vessel dilation of the ductus. On the other hand, anti-inflammatory agents, such as antenatal steroids, decrease PDA incidence and severity in patients born to those with chorioamnionitis. Proinflammatory cytokines, which are expressed more significantly in preterm neonates and chorioamnionitis, are associated with the presence of PDA. In this review, we focus on the pathogenesis of PDA in preterm infants and the role of biomarkers associated with the perinatal inflammatory process.
Collapse
|
8
|
Emerging trends in the nanomedicine applications of functionalized magnetic nanoparticles as novel therapies for acute and chronic diseases. J Nanobiotechnology 2022; 20:393. [PMID: 36045375 PMCID: PMC9428876 DOI: 10.1186/s12951-022-01595-3] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Accepted: 08/13/2022] [Indexed: 11/10/2022] Open
Abstract
High-quality point-of-care is critical for timely decision of disease diagnosis and healthcare management. In this regard, biosensors have revolutionized the field of rapid testing and screening, however, are confounded by several technical challenges including material cost, half-life, stability, site-specific targeting, analytes specificity, and detection sensitivity that affect the overall diagnostic potential and therapeutic profile. Despite their advances in point-of-care testing, very few classical biosensors have proven effective and commercially viable in situations of healthcare emergency including the recent COVID-19 pandemic. To overcome these challenges functionalized magnetic nanoparticles (MNPs) have emerged as key players in advancing the biomedical and healthcare sector with promising applications during the ongoing healthcare crises. This critical review focus on understanding recent developments in theranostic applications of functionalized magnetic nanoparticles (MNPs). Given the profound global economic and health burden, we discuss the therapeutic impact of functionalized MNPs in acute and chronic diseases like small RNA therapeutics, vascular diseases, neurological disorders, and cancer, as well as for COVID-19 testing. Lastly, we culminate with a futuristic perspective on the scope of this field and provide an insight into the emerging opportunities whose impact is anticipated to disrupt the healthcare industry.
Collapse
|
9
|
Botts SR, Fish JE, Howe KL. Dysfunctional Vascular Endothelium as a Driver of Atherosclerosis: Emerging Insights Into Pathogenesis and Treatment. Front Pharmacol 2021; 12:787541. [PMID: 35002720 PMCID: PMC8727904 DOI: 10.3389/fphar.2021.787541] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Accepted: 12/06/2021] [Indexed: 12/28/2022] Open
Abstract
Atherosclerosis, the chronic accumulation of cholesterol-rich plaque within arteries, is associated with a broad spectrum of cardiovascular diseases including myocardial infarction, aortic aneurysm, peripheral vascular disease, and stroke. Atherosclerotic cardiovascular disease remains a leading cause of mortality in high-income countries and recent years have witnessed a notable increase in prevalence within low- and middle-income regions of the world. Considering this prominent and evolving global burden, there is a need to identify the cellular mechanisms that underlie the pathogenesis of atherosclerosis to discover novel therapeutic targets for preventing or mitigating its clinical sequelae. Despite decades of research, we still do not fully understand the complex cell-cell interactions that drive atherosclerosis, but new investigative approaches are rapidly shedding light on these essential mechanisms. The vascular endothelium resides at the interface of systemic circulation and the underlying vessel wall and plays an essential role in governing pathophysiological processes during atherogenesis. In this review, we present emerging evidence that implicates the activated endothelium as a driver of atherosclerosis by directing site-specificity of plaque formation and by promoting plaque development through intracellular processes, which regulate endothelial cell proliferation and turnover, metabolism, permeability, and plasticity. Moreover, we highlight novel mechanisms of intercellular communication by which endothelial cells modulate the activity of key vascular cell populations involved in atherogenesis, and discuss how endothelial cells contribute to resolution biology - a process that is dysregulated in advanced plaques. Finally, we describe important future directions for preclinical atherosclerosis research, including epigenetic and targeted therapies, to limit the progression of atherosclerosis in at-risk or affected patients.
Collapse
Affiliation(s)
- Steven R. Botts
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Temerty Faculty of Medicine, University of Toronto, Toronto, ON, Canada
| | - Jason E. Fish
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
| | - Kathryn L. Howe
- Toronto General Hospital Research Institute, University Health Network, Toronto, ON, Canada
- Institute of Medical Science, University of Toronto, Toronto, ON, Canada
- Peter Munk Cardiac Centre, University Health Network, Toronto, ON, Canada
- Division of Vascular Surgery, Department of Surgery, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
10
|
Pan Q, Xu J, Wen CJ, Xiong YY, Gong ZT, Yang YJ. Nanoparticles: Promising Tools for the Treatment and Prevention of Myocardial Infarction. Int J Nanomedicine 2021; 16:6719-6747. [PMID: 34621124 PMCID: PMC8491866 DOI: 10.2147/ijn.s328723] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022] Open
Abstract
Despite several recent advances, current therapy and prevention strategies for myocardial infarction are far from satisfactory, owing to limitations in their applicability and treatment effects. Nanoparticles (NPs) enable the targeted and stable delivery of therapeutic compounds, enhance tissue engineering processes, and regulate the behaviour of transplants such as stem cells. Thus, NPs may be more effective than other mechanisms, and may minimize potential adverse effects. This review provides evidence for the view that function-oriented systems are more practical than traditional material-based systems; it also summarizes the latest advances in NP-based strategies for the treatment and prevention of myocardial infarction.
Collapse
Affiliation(s)
- Qi Pan
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Jing Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Cen-Jin Wen
- Department of Cardiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zhao-Ting Gong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
11
|
MacRitchie N, Di Francesco V, Ferreira MFMM, Guzik TJ, Decuzzi P, Maffia P. Nanoparticle theranostics in cardiovascular inflammation. Semin Immunol 2021; 56:101536. [PMID: 34862118 PMCID: PMC8811479 DOI: 10.1016/j.smim.2021.101536] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 11/17/2021] [Accepted: 11/18/2021] [Indexed: 12/30/2022]
Abstract
Theranostics, literally derived from the combination of the words diagnostics and therapy, is an emerging field of clinical and preclinical research, where contrast agents, drugs and diagnostic techniques are combined to simultaneously diagnose and treat pathologies. Nanoparticles are extensively employed in theranostics due to their potential to target specific organs and their multifunctional capacity. In this review, we will discuss the current state of theranostic nanomedicine, providing key examples of its application in the imaging and treatment of cardiovascular inflammation.
Collapse
Affiliation(s)
- Neil MacRitchie
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom.
| | - Valentina Di Francesco
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | | | - Tomasz J Guzik
- Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Internal Medicine, Jagiellonian University, Collegium Medicum, Kraków, Poland
| | - Paolo Decuzzi
- Laboratory of Nanotechnology for Precision Medicine, Fondazione Istituto Italiano di Tecnologia, Genoa, Italy
| | - Pasquale Maffia
- Centre for Immunobiology, Institute of Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Institute of Cardiovascular and Medical Sciences, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom; Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, Naples, Italy.
| |
Collapse
|
12
|
Xu X, Hua Y, Liu B, Zhou F, Wang L, Hou W. Correlation Between Calcification Characteristics of Carotid Atherosclerotic Plaque and Plaque Vulnerability. Ther Clin Risk Manag 2021; 17:679-690. [PMID: 34234444 PMCID: PMC8257076 DOI: 10.2147/tcrm.s303485] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Accepted: 05/17/2021] [Indexed: 11/25/2022] Open
Abstract
Purpose To investigate the relationship between calcification characteristics of carotid atherosclerotic plaque and lipid rich necrotic core (LRNC) and intraplaque hemorrhage (IPH). Methods Patients with severe carotid stenosis undergoing carotid endarterectomy (CEA) were selected. Ultrasound and CT angiography (CTA) were performed to evaluate the calcification characteristics of the plaque before the surgery. Results A total of 142 patients were included and 142 pathological specimens of postoperative plaque were obtained accordingly. There were 78 plaques (54.9%) with LRNC and 41 (28.9%) with IPH. The plaque with LRNC had higher calcification rate (93.6%) compared with the plaque with IPH (87.8%). LRNC was often found in multiple calcification (P = 0.003) and mixed type calcification (P = 0.001). Multiple calcification was more likely to combine with IPH (P = 0.008), while simple basal calcification was not likely to combine IPH (P = 0.002). Smaller granular calcification was more likely to be associated with IPH (P < 0.05). In multivariate regression analysis of IPH and calcification characteristics, simple basal calcification was still a protective factor for IPH (OR, 0.25; 95% CI, 0.09–0.66; P = 0.005), while multiple calcification was closely related to the occurrence of IPH (OR, 3.58; 95% CI, 1.49–8.61; P = 0.004). Conclusion Calcification characteristics of carotid atherosclerotic plaques are closely related to the vulnerability of plaques, especially multiple calcification and mixed type calcification.
Collapse
Affiliation(s)
- Xiangli Xu
- Department of Ultrasound, the Second Hospital of Harbin, Harbin, People's Republic of China
| | - Yang Hua
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Beibei Liu
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Fubo Zhou
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Lili Wang
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| | - Weihong Hou
- Department of Vascular Ultrasonography, Xuanwu Hospital, Capital Medical University, Beijing, People's Republic of China
| |
Collapse
|
13
|
Fleischmann D, Goepferich A. General sites of nanoparticle biodistribution as a novel opportunity for nanomedicine. Eur J Pharm Biopharm 2021; 166:44-60. [PMID: 34087354 DOI: 10.1016/j.ejpb.2021.05.027] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/22/2021] [Accepted: 05/27/2021] [Indexed: 02/07/2023]
Abstract
The development of nanomedical devices has led to a considerable number of clinically applied nanotherapeutics. Yet, the overall poor translation of nanoparticular concepts into marketable systems has not met the initial expectations and led to increasing criticism in recent years. Most novel nano approaches thereby use highly refined formulations including a plethora of active targeting sequences, but ultimately fail to reach their target due to a generally high off-target deposition in organs such as the liver or kidney. In this context, we argue that initial nanoparticle (NP) development should not entirely become set on conventional formulation aspects. In contrast, we propose a change of focus towards a prior analysis of general sites of NP in vivo deposition and an assessment of how accumulation in these organs or tissues can be harnessed to develop therapies for site-related pathologies. We therefore give a comprehensive overview of existing nanotherapeutic targeting strategies for specific cell types within three of the usual suspects, i.e. the liver, kidney and the vascular system. We discuss the physiological surroundings and relevant pathologies of described tissues as well as the implications for NP-mediated drug delivery. Additionally, successful cell-selective NP concepts using active targeting strategies are assessed. By bringing together both (patho)physiological aspects and concepts for cell-selective NP formulations, we hope to show a novel opportunity for the development of more promising nanotherapeutic devices.
Collapse
Affiliation(s)
- Daniel Fleischmann
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany
| | - Achim Goepferich
- Department of Pharmaceutical Technology, University of Regensburg, Universitaetsstrasse 31, 93053 Regensburg, Germany.
| |
Collapse
|
14
|
Makowski MR, Rischpler C, Ebersberger U, Keithahn A, Kasel M, Hoffmann E, Rassaf T, Kessler H, Wester HJ, Nekolla SG, Schwaiger M, Beer AJ. Multiparametric PET and MRI of myocardial damage after myocardial infarction: correlation of integrin αvβ3 expression and myocardial blood flow. Eur J Nucl Med Mol Imaging 2021; 48:1070-1080. [PMID: 32970218 PMCID: PMC8041712 DOI: 10.1007/s00259-020-05034-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Accepted: 09/08/2020] [Indexed: 12/21/2022]
Abstract
PURPOSE Increased angiogenesis after myocardial infarction is considered an important favorable prognostic parameter. The αvβ3 integrin is a key mediator of cell-cell and cell-matrix interactions and an important molecular target for imaging of neovasculature and repair processes after MI. Thus, imaging of αvβ3 expression might provide a novel biomarker for assessment of myocardial angiogenesis as a prognostic marker of left ventricular remodeling after MI. Currently, there is limited data available regarding the association of myocardial blood flow and αvβ3 integrin expression after myocardial infarction in humans. METHODS Twelve patients were examined 31 ± 14 days after MI with PET/CT using [18F]Galacto-RGD and [13N]NH3 and with cardiac MRI including late enhancement on the same day. Normal myocardium (remote) and areas of infarction (lesion) were identified on the [18F]Galacto-RGD PET/CT images by correlation with [13N]NH3 PET and cardiac MRI. Lesion/liver-, lesion/blood-, and lesion/remote ratios were calculated. Blood flow and [18F]Galacto-RGD uptake were quantified and correlated for each myocardial segment (AHA 17-segment model). RESULTS In 5 patients, increased [18F]Galacto-RGD uptake was notable within or adjacent to the infarction areas with a lesion/remote ratio of 46% (26-83%; lesion/blood 1.15 ± 0.06; lesion/liver 0.61 ± 0.18). [18F]Galacto-RGD uptake correlated significantly with infarct size (R = 0.73; p = 0.016). Moreover, it correlated significantly with restricted blood flow for all myocardial segments (R = - 0.39; p < 0.0001) and even stronger in severely hypoperfused areas (R = - 0.75; p < 0.0001). CONCLUSION [18F]Galacto-RGD PET/CT allows the visualization and quantification of myocardial αvβ3 expression as a key player in angiogenesis in a subset of patients after MI. αvβ3 expression was more pronounced in patients with larger infarcts and was generally more intense but not restricted to areas with more impaired blood flow, proving that tracer uptake was largely independent of unspecific perfusion effects. Based on these promising results, larger prospective studies are warranted to evaluate the potential of αvβ3 imaging for assessment of myocardial angiogenesis and prediction of ventricular remodeling.
Collapse
Affiliation(s)
- Marcus R Makowski
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Diagnostic and Interventional Radiology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Christoph Rischpler
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany.
- Clinic for Nuclear Medicine, University Hospital Essen, University of Duisburg-Essen, Hufelandstrasse 55, 45147, Essen, Germany.
| | | | - Alexandra Keithahn
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Markus Kasel
- Department of Cardiology, Klinikum Bogenhausen, Munich, Germany
| | - Ellen Hoffmann
- Department of Cardiology, Klinikum Bogenhausen, Munich, Germany
| | - Tienush Rassaf
- Department of Cardiology and Vascular Medicine, West German Heart and Vascular Center, University Hospital Essen, University of Duisburg-Essen, Essen, Germany
| | - Horst Kessler
- Department of Chemistry, Institute for Advanced Study and Center of Integrated Protein Science, Technical University of Munich, Garching, Germany
| | - Hans-Jürgen Wester
- Pharmaceutical Radiochemistry, Technical University of Munich, Klinikum rechts der Isar, Munich, Germany
| | - Stephan G Nekolla
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Markus Schwaiger
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
| | - Ambros J Beer
- Department of Nuclear Medicine, School of Medicine, Technical University of Munich, Munich, Germany
- Department of Nuclear Medicine, University Ulm, Ulm, Germany
| |
Collapse
|
15
|
Nanotechnology applications for cardiovascular disease treatment: Current and future perspectives. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2021; 34:102387. [PMID: 33753283 DOI: 10.1016/j.nano.2021.102387] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 02/24/2021] [Accepted: 03/03/2021] [Indexed: 11/22/2022]
Abstract
A large majority of cardiovascular nanomedicine research has focused on fabricating designer nanoparticles for improved targeting as a means to overcome biological barriers. For cardiac related disorders, such as atherosclerosis, hypertension, and myocardial infarction, designer micro or nanoparticles are often administered into the vasculature or targeted vessel with the hope to circumvent problems associated with conventional drug delivery, including negative systemic side effects. Additionally, novel nano-drug carriers that enter circulation can be selectively uptaken by immune cells with the intended purpose that they modulate inflammatory processes and migrate locally to plaque for therapeutic payload delivery. Indeed, innovative design in nanoparticle composition, formulation, and functionalization has advanced the field as a means to achieve therapeutic efficacy for a variety of cardiac disease indications. This perspective aims to discuss these advances and provide new interpretations of how nanotechnology can be best applied to aid in cardiovascular disease treatment. In an effort to spark discussions on where the field of research should go, we share our outlook in new areas of nanotechnological inclusion and integration, such as in vascular, implantable, or wearable device technologies as well as nanocomposites and nanocoatings. Further, as cardiovascular diseases (CVD) increasingly claim a number of lives globally, we propose more attention should be placed by researchers on nanotechnological approaches for risk factor treatment to aid in early prevention and treatment of CVD.
Collapse
|
16
|
Ou LC, Zhong S, Ou JS, Tian JW. Application of targeted therapy strategies with nanomedicine delivery for atherosclerosis. Acta Pharmacol Sin 2021; 42:10-17. [PMID: 32457416 DOI: 10.1038/s41401-020-0436-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2020] [Accepted: 05/09/2020] [Indexed: 12/20/2022] Open
Abstract
Atherosclerosis (AS) is the main pathological cause of coronary heart disease (CHD). Current clinical interventions including statin drugs can effectively reduce acute myocardial infarction and stroke to some extent, but residual risk remains high. The current clinical treatment regimens are relatively effective for early atherosclerotic plaques and can even reverse their progression. However, the effectiveness of these treatments for advanced AS is not ideal, and advanced atherosclerotic plaques-the pathological basis of residual risk-can still cause a recurrence of acute cardiovascular and cerebrovascular events. Recently, nanomedicine-based treatment strategies have been extensively used in antitumor therapy, and also shown great potential in anti-AS therapy. There are many microstructures in late-stage atherosclerotic plaques, such as neovascularization, micro-calcification, and cholesterol crystals, and these have become important foci for targeted nanomedicine delivery. The use of targeted nanoparticles has become an important strategy for the treatment of advanced AS to further reduce the residual risk of cardiovascular events. Furthermore, the feasibility and safety of nanotechnology in clinical treatment have been preliminarily confirmed. In this review, we summarize the application of nanomedicine delivery in the treatment of advanced AS and the clinical value of several promising nanodrugs.
Collapse
|
17
|
Sun M, Lee J, Chen Y, Hoshino K. Studies of nanoparticle delivery with in vitro bio-engineered microtissues. Bioact Mater 2020; 5:924-937. [PMID: 32637755 PMCID: PMC7330434 DOI: 10.1016/j.bioactmat.2020.06.016] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Revised: 06/12/2020] [Accepted: 06/22/2020] [Indexed: 01/04/2023] Open
Abstract
A variety of engineered nanoparticles, including lipid nanoparticles, polymer nanoparticles, gold nanoparticles, and biomimetic nanoparticles, have been studied as delivery vehicles for biomedical applications. When assessing the efficacy of a nanoparticle-based delivery system, in vitro testing with a model delivery system is crucial because it allows for real-time, in situ quantitative transport analysis, which is often difficult with in vivo animal models. The advent of tissue engineering has offered methods to create experimental models that can closely mimic the 3D microenvironment in the human body. This review paper overviews the types of nanoparticle vehicles, their application areas, and the design strategies to improve delivery efficiency, followed by the uses of engineered microtissues and methods of analysis. In particular, this review highlights studies on multicellular spheroids and other 3D tissue engineering approaches for cancer drug development. The use of bio-engineered tissues can potentially provide low-cost, high-throughput, and quantitative experimental platforms for the development of nanoparticle-based delivery systems.
Collapse
Affiliation(s)
- Mingze Sun
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Rd, Storrs, CT, 06269, USA
| | - Jinhyung Lee
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Rd, Storrs, CT, 06269, USA
| | - Yupeng Chen
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Rd, Storrs, CT, 06269, USA
| | - Kazunori Hoshino
- Department of Biomedical Engineering, University of Connecticut, 260 Glenbrook Rd, Storrs, CT, 06269, USA
| |
Collapse
|
18
|
Farber G, Boczar KE, Wiefels CC, Zelt JG, Guler EC, deKemp RA, Beanlands RS, Rotstein BH. The Future of Cardiac Molecular Imaging. Semin Nucl Med 2020; 50:367-385. [DOI: 10.1053/j.semnuclmed.2020.02.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
19
|
Deng Y, Zhang X, Shen H, He Q, Wu Z, Liao W, Yuan M. Application of the Nano-Drug Delivery System in Treatment of Cardiovascular Diseases. Front Bioeng Biotechnol 2020; 7:489. [PMID: 32083068 PMCID: PMC7005934 DOI: 10.3389/fbioe.2019.00489] [Citation(s) in RCA: 88] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 12/31/2019] [Indexed: 12/13/2022] Open
Abstract
Cardiovascular diseases (CVDs) have become a serious threat to human life and health. Though many drugs acting via different mechanism of action are available in the market as conventional formulations for the treatment of CVDs, they are still far from satisfactory due to poor water solubility, low biological efficacy, non-targeting, and drug resistance. Nano-drug delivery systems (NDDSs) provide a new drug delivery method for the treatment of CVDs with the development of nanotechnology, demonstrating great advantages in solving the above problems. Nevertheless, there are some problems about NDDSs need to be addressed, such as cytotoxicity. In this review, the types and targeting strategies of NDDSs were summarized, and the new research progress in the diagnosis and therapy of CVDs in recent years was reviewed. Future prospective for nano-carriers in drug delivery for CVDs includes gene therapy, in order to provide more ideas for the improvement of cardiovascular drugs. In addition, its safety was also discussed in the review.
Collapse
Affiliation(s)
- Yudi Deng
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Xudong Zhang
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Haibin Shen
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Qiangnan He
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Zijian Wu
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Wenzhen Liao
- Guangdong Provincial Key Laboratory of Tropical Disease Research, Department of Nutrition and Food Hygiene, School of Public Health, Southern Medical University, Guangzhou, China
| | - Miaomiao Yuan
- The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| |
Collapse
|
20
|
Gorabi AM, Kiaie N, Reiner Ž, Carbone F, Montecucco F, Sahebkar A. The Therapeutic Potential of Nanoparticles to Reduce Inflammation in Atherosclerosis. Biomolecules 2019; 9:416. [PMID: 31455044 PMCID: PMC6769786 DOI: 10.3390/biom9090416] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Revised: 08/14/2019] [Accepted: 08/23/2019] [Indexed: 01/08/2023] Open
Abstract
Chronic inflammation is one of the main determinants of atherogenesis. The traditional medications for treatment of atherosclerosis are not very efficient in targeting atherosclerotic inflammation. Most of these drugs are non-selective, anti-inflammatory and immunosuppressive agents that have adverse effects and very limited anti-atherosclerotic effects, which limits their systemic administration. New approaches using nanoparticles have been investigated to specifically deliver therapeutic agents directly on atherosclerotic lesions. The use of drug delivery systems, such as polymeric nanoparticles, liposomes, and carbon nanotubes are attractive strategies, but some limitations exist. For instance, nanoparticles may alter the drug kinetics, based on the pathophysiological mechanisms of the diseases. In this review, we will update pathophysiological evidence for the use of nanoparticles to reduce inflammation and potentially prevent atherogenesis in different experimental models.
Collapse
Affiliation(s)
- Armita Mahdavi Gorabi
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran 1411713138, Iran
| | - Nasim Kiaie
- Research Center for Advanced Technologies in Cardiovascular Medicine, Tehran Heart Center, Tehran University of Medical Sciences, Tehran 1411713138, Iran
| | - Željko Reiner
- University Hospital Centre Zagreb, School of Medicine University of Zagreb, Department of Internal Medicine, 1000 Zagreb, Croatia
| | - Federico Carbone
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 16132 Genoa, Italy
| | - Fabrizio Montecucco
- IRCCS Ospedale Policlinico San Martino Genoa - Italian Cardiovascular Network, 16132 Genoa, Italy
- First Clinic of Internal Medicine, Department of Internal Medicine, University of Genoa, 16132 Genoa, Italy and Centre of Excellence for Biomedical Research (CEBR), University of Genoa, 16132 Genoa, Italy
| | - Amirhossein Sahebkar
- Neurogenic Inflammation Research Center, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
- Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
- School of Pharmacy, Mashhad University of Medical Sciences, Mashhad 9177948564, Iran.
| |
Collapse
|
21
|
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: 0.8] [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
|
22
|
Abstract
Noninvasive imaging has played an increasing role in the process of cardiovascular drug development. This review focuses specifically on the use of molecular imaging, which has been increasingly applied to improve and accelerate certain preclinical steps in drug development, including the identification of appropriate therapeutic targets, evaluation of on-target and off-target effects of candidate therapies, assessment of dose response, and the evaluation of drug or biological biodistribution and pharmacodynamics. Unlike the case in cancer medicine, in cardiovascular medicine, molecular imaging has not been used as a primary surrogate clinical end point for drug approval. However, molecular imaging has been applied in early clinical trials, particularly in phase 0 studies, to demonstrate proof-of-concept or to explain variation in treatment effect. Many of these applications where molecular imaging has been used in drug development have involved the retasking of technologies that were originally intended as clinical diagnostics. With greater experience and recognition of the rich information provided by in vivo molecular imaging, it is anticipated that it will increasingly be used to address the enormous time and costs associated with bringing a new drug to clinical launch.
Collapse
Affiliation(s)
- Jonathan R Lindner
- From the Knight Cardiovascular Institute (J.R.L.), Oregon National Primate Research Center (J.R.L.), and Center for Radiologic Research (J.L.), Oregon Health and Science University, Portland.
| | - Jeanne Link
- From the Knight Cardiovascular Institute (J.R.L.), Oregon National Primate Research Center (J.R.L.), and Center for Radiologic Research (J.L.), Oregon Health and Science University, Portland
| |
Collapse
|
23
|
Zhang Y, Koradia A, Kamato D, Popat A, Little PJ, Ta HT. Treatment of atherosclerotic plaque: perspectives on theranostics. ACTA ACUST UNITED AC 2019; 71:1029-1043. [PMID: 31025381 DOI: 10.1111/jphp.13092] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Accepted: 03/16/2019] [Indexed: 01/20/2023]
Abstract
OBJECTIVES Atherosclerosis, a progressive condition characterised by the build-up of plaque due to the accumulation of low-density lipoprotein and fibrous substances in the damaged arteries, is the major underlying pathology of most cardiovascular diseases. Despite the evidence of the efficacy of the present treatments for atherosclerosis, the complex and poorly understood underlying mechanisms of atherosclerosis development and progression have prevented them from reaching their full potential. Novel alternative treatments like usage of nanomedicines and theranostics are gaining attention of the researchers worldwide. This review will briefly discuss the current medications for the disease and explore potential future developments based on theranostics nanomaterials that may help resolve atherosclerotic cardiovascular disease. KEY FINDINGS Various drugs can slow the effects of atherosclerosis. They include hyperlipidaemia medications, anti-platelet drugs, hypertension and hyperglycaemia medications. Most of the theranostic agents developed for atherosclerosis have shown the feasibility of rapid and noninvasive diagnosis, as well as effective and specific treatment in animal models. However, there are still some limitation exist in their structure design, stability, targeting efficacy, toxicity and production, which should be optimized in order to develop clinically acceptable nanoparticle based theronostics for atherosclerosis. SUMMARY Current medications for atherosclerosis and potential theranostic nanomaterials developed for the disease are discussed in the current review. Further investigations remain to be carried out to achieve clinical translation of theranostic agents for atherosclerosis.
Collapse
Affiliation(s)
- Yicong Zhang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld, Australia
| | - Aayushi Koradia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Qld, Australia
| | - Danielle Kamato
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Qld, Australia
| | - Amirali Popat
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Qld, Australia
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Qld, Australia.,Department of Pharmacy, Xinhua College of Sun Yat-sen University, Guangzhou, China
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, Qld, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, Qld, Australia
| |
Collapse
|
24
|
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: 78] [Impact Index Per Article: 15.6] [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
|
25
|
Tan KX, Pan S, Jeevanandam J, Danquah MK. Cardiovascular therapies utilizing targeted delivery of nanomedicines and aptamers. Int J Pharm 2019; 558:413-425. [PMID: 30660748 DOI: 10.1016/j.ijpharm.2019.01.023] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2018] [Revised: 01/03/2019] [Accepted: 01/05/2019] [Indexed: 01/01/2023]
Abstract
Cardiovascular ailments are the foremost trigger of death in the world today, including myocardial infarction and ischemic heart diseases. To date, extraordinary measures have been prescribed, from the perspectives of both conventional medical therapies and surgeries, to enforce cardiac cell regeneration post cardiac traumas, albeit with limited long-term success. The prospects of successful heart transplants are also grim, considering exorbitant costs and unavailability of suitable donors in most cases. From the perspective of cardiac revascularization, use of nanoparticles and nanoparticle mediated targeted drug delivery have garnered substantial attention, attributing to both active and passive heart targeting, with enhanced target specificity and sensitivity. This review focuses on this aspect, while outlining the progress in targeted delivery of nanomedicines in the prognosis and subsequent therapy of cardiovascular disorders, and recapitulating the benefits and intrinsic challenges associated with the incorporation of nanoparticles. This article categorically provides an overview of nanoparticle-mediated targeted delivery systems and their implications in handling cardiovascular diseases, including their intrinsic benefits and encountered procedural trials and challenges. Additionally, the solicitations of aptamers in targeted drug delivery with identical objectives, are presented. This includes a detailed appraisal on various aptamer-navigated nanoparticle targeted delivery platforms in the diagnosis and treatment of cardiovascular maladies. Despite a few impending challenges, subject to additional investigations, both nanoparticles as well as aptamers show a high degree of promise, and pose as the next generation of drug delivery vehicles, in targeted cardiovascular therapy.
Collapse
Affiliation(s)
- Kei Xian Tan
- Department of Chemical Engineering, Curtin University of Technology, 98009 Sarawak, Malaysia.
| | - Sharadwata Pan
- School of Life Sciences Weihenstephan, Technical University of Munich, 85354 Freising, Germany.
| | - Jaison Jeevanandam
- Department of Chemical Engineering, Curtin University of Technology, 98009 Sarawak, Malaysia.
| | - Michael K Danquah
- Department of Chemical Engineering, University of Tennessee, Chattanooga, TN 37403, United States.
| |
Collapse
|
26
|
Strobel HA, Qendro EI, Alsberg E, Rolle MW. Targeted Delivery of Bioactive Molecules for Vascular Intervention and Tissue Engineering. Front Pharmacol 2018; 9:1329. [PMID: 30519186 PMCID: PMC6259603 DOI: 10.3389/fphar.2018.01329] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2018] [Accepted: 10/29/2018] [Indexed: 01/25/2023] Open
Abstract
Cardiovascular diseases are the leading cause of death in the United States. Treatment often requires surgical interventions to re-open occluded vessels, bypass severe occlusions, or stabilize aneurysms. Despite the short-term success of such interventions, many ultimately fail due to thrombosis or restenosis (following stent placement), or incomplete healing (such as after aneurysm coil placement). Bioactive molecules capable of modulating host tissue responses and preventing these complications have been identified, but systemic delivery is often harmful or ineffective. This review discusses the use of localized bioactive molecule delivery methods to enhance the long-term success of vascular interventions, such as drug-eluting stents and aneurysm coils, as well as nanoparticles for targeted molecule delivery. Vascular grafts in particular have poor patency in small diameter, high flow applications, such as coronary artery bypass grafting (CABG). Grafts fabricated from a variety of approaches may benefit from bioactive molecule incorporation to improve patency. Tissue engineering is an especially promising approach for vascular graft fabrication that may be conducive to incorporation of drugs or growth factors. Overall, localized and targeted delivery of bioactive molecules has shown promise for improving the outcomes of vascular interventions, with technologies such as drug-eluting stents showing excellent clinical success. However, many targeted vascular drug delivery systems have yet to reach the clinic. There is still a need to better optimize bioactive molecule release kinetics and identify synergistic biomolecule combinations before the clinical impact of these technologies can be realized.
Collapse
Affiliation(s)
- Hannah A. Strobel
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States
| | - Elisabet I. Qendro
- Graduate School of Biomedical Sciences, University of Massachusetts Medical School, Worcester, MA, United States
| | - Eben Alsberg
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, United States
| | - Marsha W. Rolle
- Department of Biomedical Engineering, Worcester Polytechnic Institute, Worcester, MA, United States
| |
Collapse
|
27
|
Noukeu LC, Wolf J, Yuan B, Banerjee S, Nguyen KT. Nanoparticles for Detection and Treatment of Peripheral Arterial Disease. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2018; 14:e1800644. [PMID: 29952061 DOI: 10.1002/smll.201800644] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/14/2018] [Indexed: 06/08/2023]
Abstract
Peripheral arterial disease (PAD) is defined as a slow, progressive disorder of the lower extremity arterial vessels characterized by chronic narrowing that often results in occlusion and is associated with loss of functional capacity. Although the PAD occurrence rate is increasing in the elderly population, outcomes with current treatment strategies are suboptimal. Hence, there is an urgent need to develop new technologies that overcome limitations of traditional modalities for PAD detection and therapy. In this Review, the application of nanotechnology as a tool that bridges the gap in PAD diagnosis and therapy is in focus. Several materials including synthetic, natural, biodegradable, and biocompatible materials are used to develop nanoparticles for PAD diagnostic and/or therapeutic applications. Moreover, various recent research approaches are being explored to diagnose PAD through multimodality imaging with different nanoplatforms. Further efforts include targeted delivery of various therapeutic agents using nanostructures as carriers to treat PAD. Last, but not least, despite being a fairly new field, researchers are exploring the use of nanotheranostics for PAD detection and therapy.
Collapse
Affiliation(s)
- Linda C Noukeu
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
- Joint Biomedical Engineering Program, University of Texas Southwestern, Dallas, TX, 75235, USA
| | - Joseph Wolf
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
- Joint Biomedical Engineering Program, University of Texas Southwestern, Dallas, TX, 75235, USA
| | - Baohong Yuan
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
- Joint Biomedical Engineering Program, University of Texas Southwestern, Dallas, TX, 75235, USA
| | - Subhash Banerjee
- Division of Cardiology, University of Texas Southwestern Medical Center, Dallas, TX, 75235, USA
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76010, USA
- Joint Biomedical Engineering Program, University of Texas Southwestern, Dallas, TX, 75235, USA
| |
Collapse
|
28
|
Maji D, Lu J, Sarder P, Schmieder AH, Cui G, Yang X, Pan D, Lew MD, Achilefu S, Lanza GM. Cellular Trafficking of Sn-2 Phosphatidylcholine Prodrugs Studied with Fluorescence Lifetime Imaging and Super-resolution Microscopy. PRECISION NANOMEDICINE 2018; 1:128-145. [PMID: 31249994 PMCID: PMC6597004 DOI: 10.33218/prnano1(2).180724.1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
While the in vivo efficacy of Sn-2 phosphatidylcholine prodrugs incorporated into targeted, non-pegylated lipid-encapsulated nanoparticles was demonstrated in prior preclinical studies, the microscopic details of cell prodrug internalization and trafficking events are unknown. Classic fluorescence microscopy, fluorescence lifetime imaging microscopy, and single-molecule super-resolution microscopy were used to investigate the cellular handling of doxorubicin-prodrug and AlexaFluor™-488-prodrug. Sn-2 phosphatidylcholine prodrugs delivered by hemifusion of nanoparticle and cell phospholipid membranes functioned as phosphatidylcholine mimics, circumventing the challenges of endosome sequestration and release. Phosphatidylcholine prodrugs in the outer cell membrane leaflet translocated to the inner membrane leaflet by ATP-dependent and ATP-independent mechanisms and distributed broadly within the cytosolic membranes over the next 12 h. A portion of the phosphatidylcholine prodrug populated vesicle membranes trafficked to the perinuclear Golgi/ER region, where the drug was enzymatically liberated and activated. Native doxorubicin entered the cells, passed rapidly to the nucleus, and bound to dsDNA, whereas DOX was first enzymatically liberated from DOX-prodrug within the cytosol, particularly in the perinuclear region, before binding nuclear dsDNA. Much of DOX-prodrug was initially retained within intracellular membranes. In vitro anti-proliferation effectiveness of the two drug delivery approaches was equivalent at 48 h, suggesting that residual intracellular DOX-prodrug may constitute a slow-release drug reservoir that enhances effectiveness. We have demonstrated that Sn-2 phosphatidylcholine prodrugs function as phosphatidylcholine mimics following reported pathways of phosphatidylcholine distribution and metabolism. Drug complexed to the Sn-2 fatty acid is enzymatically liberated and reactivated over many hours, which may enhance efficacy overtime.
Collapse
Affiliation(s)
- Dolonchampa Maji
- Optical Radiology Lab, Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Jin Lu
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University of Buffalo, Buffalo, NY 14203
| | - Anne H Schmieder
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Grace Cui
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xiaoxia Yang
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dipanjan Pan
- Department of Bioengineering, University of Illinois at Urbana-Champaign, Champaign, IL, USA
| | - Matthew D Lew
- Department of Electrical and Systems Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Samuel Achilefu
- Optical Radiology Lab, Department of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.,Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA
| | - Gregory M Lanza
- Department of Biomedical Engineering, Washington University in St. Louis, MO 63130, USA.,Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
29
|
Carvalho PM, Felício MR, Santos NC, Gonçalves S, Domingues MM. Application of Light Scattering Techniques to Nanoparticle Characterization and Development. Front Chem 2018; 6:237. [PMID: 29988578 PMCID: PMC6026678 DOI: 10.3389/fchem.2018.00237] [Citation(s) in RCA: 136] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2018] [Accepted: 06/04/2018] [Indexed: 01/07/2023] Open
Abstract
Over the years, the scientific importance of nanoparticles for biomedical applications has increased. The high stability and biocompatibility, together with the low toxicity of the nanoparticles developed lead to their use as targeted drug delivery systems, bioimaging systems, and biosensors. The wide range of nanoparticles size, from 10 nm to 1 μm, as well as their optical properties, allow them to be studied using microscopy and spectroscopy techniques. In order to be effectively used, the physicochemical properties of nanoparticle formulations need to be taken into account, namely, particle size, surface charge distribution, surface derivatization and/or loading capacity, and related interactions. These properties need to be optimized considering the final nanoparticle intended biodistribution and target. In this review, we cover light scattering based techniques, namely dynamic light scattering and zeta-potential, used for the physicochemical characterization of nanoparticles. Dynamic light scattering is used to measure nanoparticles size, but also to evaluate their stability over time in suspension, at different pH and temperature conditions. Zeta-potential is used to characterize nanoparticles surface charge, obtaining information about their stability and surface interaction with other molecules. In this review, we focus on nanoparticle characterization and application in infection, cancer and cardiovascular diseases.
Collapse
Affiliation(s)
- Patrícia M Carvalho
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Mário R Felício
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Nuno C Santos
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Sónia Gonçalves
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| | - Marco M Domingues
- Instituto de Medicina Molecular, Faculdade de Medicina, Universidade de Lisboa, Lisbon, Portugal
| |
Collapse
|
30
|
Ma S, Motevalli SM, Chen J, Xu MQ, Wang Y, Feng J, Qiu Y, Han D, Fan M, Ding M, Fan L, Guo W, Liang XJ, Cao F. Precise theranostic nanomedicines for inhibiting vulnerable atherosclerotic plaque progression through regulation of vascular smooth muscle cell phenotype switching. Theranostics 2018; 8:3693-3706. [PMID: 30026877 PMCID: PMC6037040 DOI: 10.7150/thno.24364] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Accepted: 05/22/2018] [Indexed: 02/06/2023] Open
Abstract
Coronary heart disease is a prevalent and fatal killer caused by vulnerable atherosclerotic plaques (VASPs). However, the precise detection and treatment of VASPs remains a difficult challenge. Here, we present the development of noninvasive human serum albumin (HSA)-based theranostic nanomedicines (NMs) for the specific diagnosis and effective therapy of VASPs. Methods: The ICG/SRT@HSA-pept NMs were formulated to contain payloads of the near-infrared (NIR) fluorescent dye indocyanine green (ICG) and the sirtuin 1 (Sirt1) activator SRT1720, and modified with a peptide moiety targeting osteopontin (OPN). The in vivo atherosclerotic mouse model was established with the high-fat diet (HFD). The in vitro vascular smooth muscle cells (VSMCs) phenotypic switching was induced using the ox-LDL stimulation. Results: Due to the overexpression of OPN in activated VSMCs and VASPs, the targeted NMs specifically accumulated within the VASPs region after intravenous injection into the atherosclerotic mice, achieving the precise detection of VASPs. In addition, in the presence of SRT1720, the NMs could activate intracellular Sirt1 and activate an antiatherogenesis effect by inhibiting the phenotypic switching of VSMCs, which is an essential contributor to the vulnerability and progression of atherosclerotic plaques. After therapeutic administration of the ICG/SRT@HSA-pept NMs for two weeks, the physiological sizes and plaque compositions of VASPs were markedly improved. Furthermore, ICG/SRT@HSA-pept NMs-treated mice presented a more favorable plaque phenotype than that was observed in free SRT1720-treated mice, suggesting the enhanced delivery of pharmaceutical agents to the atherosclerotic lesions and improved therapeutic efficacy of NMs compared with free SRT1720. Conclusions: The theranostic ICG/SRT@HSA-pept NMs showed great potential for the precise identification and targeted treatment of atherosclerotic diseases.
Collapse
|
31
|
Li X, Sui Z, Li X, Xu W, Guo Q, Sun J, Jing F. Perfluorooctylbromide nanoparticles for ultrasound imaging and drug delivery. Int J Nanomedicine 2018; 13:3053-3067. [PMID: 29872293 PMCID: PMC5975599 DOI: 10.2147/ijn.s164905] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Perfluorooctylbromide nanoparticles (PFOB NPs) are a type of multifunctional nanotechnology that has been studied for various medical applications. Commercial ultrasound contrast agents (UCAs) suffer from the following limitations: short half-lives in vivo, high background signal and restricted distribution in the vascular circulation due to their micrometer dimensions. PFOB NPs are new potential UCAs that persist for long periods in the circulatory system, possess a relatively stable echogenic response without increasing the background signal and exhibit lower acoustic attenuation than commercial UCAs. Furthermore, PFOB NPs may also serve as drug delivery vehicles in which drugs are dissolved in the outer lipid or polymer layer for subsequent delivery to target sites in site-targeted therapy. The use of PFOB NPs as carriers has the potential advantage of selectively delivering payloads to the target site while improving visualization of the site using ultrasound (US) imaging. Unfortunately, the application of PFOB NPs to the field of ultrasonography has been limited because of the low intensity of US reflection. Numerous researchers have realized the potential use of PFOB NPs as UCAs and thus have developed alternative approaches to apply PFOB NPs in ultrasonography. In this article, we review the latest approaches for using PFOB NPs to enhance US imaging in vivo. In addition, this article emphasizes the application of PFOB NPs as promising drug delivery carriers for cancer and atherosclerosis treatments, as PFOB NPs can transport different drug payloads for various applications with good efficacy. We also note the challenges and future study directions for the application of PFOB NPs as both a delivery system for therapeutic agents and a diagnostic agent for ultrasonography.
Collapse
Affiliation(s)
- Xiao Li
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Zhongguo Sui
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Xin Li
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Wen Xu
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Qie Guo
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Jialin Sun
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| | - Fanbo Jing
- Department of Clinical Pharmacy, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, People's Republic of China
| |
Collapse
|
32
|
Anti-angiogenic drug loaded liposomes: Nanotherapy for early atherosclerotic lesions in mice. PLoS One 2018; 13:e0190540. [PMID: 29338009 PMCID: PMC5770017 DOI: 10.1371/journal.pone.0190540] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 12/15/2017] [Indexed: 02/08/2023] Open
Abstract
Fumagillin-loaded liposomes were injected into ApoE-KO mice. The animals were divided into several groups to test the efficacy of this anti-angiogenic drug for early treatment of atherosclerotic lesions. Statistical analysis of the lesions revealed a decrease in the lesion size after 5 weeks of treatment.
Collapse
|
33
|
Su T, Wang YB, Han D, Wang J, Qi S, Gao L, Shao YH, Qiao HY, Chen JW, Liang SH, Nie YZ, Li JY, Cao F. Multimodality Imaging of Angiogenesis in a Rabbit Atherosclerotic Model by GEBP11 Peptide Targeted Nanoparticles. Am J Cancer Res 2017; 7:4791-4804. [PMID: 29187904 PMCID: PMC5706100 DOI: 10.7150/thno.20767] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2017] [Accepted: 09/16/2017] [Indexed: 12/12/2022] Open
Abstract
Background and Aims: Angiogenesis is an important pathological process during progression of plaque formation, which can result in plaque hemorrhage and vulnerability. This study aims to explore non-invasive imaging of angiogenesis in atherosclerotic plaque through magnetic resonance imaging (MRI) and positron emission tomography (PET) by using GEBP11 peptide targeted magnetic iron oxide nanoparticles in a rabbit model of atherosclerosis. Methods: The dual-modality imaging probe was constructed by coupling 2, 3-dimercaptosuccinnic acid-coated paramagnetic nanoparticles (DMSA-MNPs) and the PET 68Ga chelator 1,4,7-triazacyclononane-N, N', N''-triacetic acid (NOTA) to GEBP11 peptide. The atherosclerosis model was induced in New Zealand white rabbits by abdominal aorta balloon de-endothelialization and atherogenic diet for 12 weeks. The plaque areas in abdominal artery were detected by ultrasound imaging and Oil Red O staining. Immunofluorescence staining and Prussian blue staining were applied respectively to investigate the affinity of GEBP11 peptide. MTT and flow cytometric analysis were performed to detect the effects of NGD-MNPs on cell proliferation, cell cycle and apoptosis in Human umbilical vein endothelial cells (HUVECs). In vivo MRI and PET imaging of atherosclerotic plaque were carried out at different time points after intravenous injection of nanoparticles. Results: The NGD-MNPs with hydrodynamic diameter of 130.8 nm ± 7.1 nm exhibited good imaging properties, high stability, low immunogenicity and little cytotoxicity. In vivo PET/MR imaging revealed that 68Ga-NGD-MNPs were successfully applied to visualize atherosclerotic plaque angiogenesis in the rabbit abdominal aorta. Prussian blue and CD31 immunohistochemical staining confirmed that NGD-MNPs were well co-localized within the blood vessels' plaques. Conclusion:68Ga-NGD-MNPs might be a promising MR and PET dual imaging probe for visualizing the vulnerable plaques.
Collapse
|
34
|
Alaarg A, Pérez-Medina C, Metselaar JM, Nahrendorf M, Fayad ZA, Storm G, Mulder WJM. Applying nanomedicine in maladaptive inflammation and angiogenesis. Adv Drug Deliv Rev 2017; 119:143-158. [PMID: 28506745 PMCID: PMC5682240 DOI: 10.1016/j.addr.2017.05.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2017] [Revised: 04/12/2017] [Accepted: 05/09/2017] [Indexed: 12/11/2022]
Abstract
Inflammation and angiogenesis drive the development and progression of multiple devastating diseases such as atherosclerosis, cancer, rheumatoid arthritis, and inflammatory bowel disease. Though these diseases have very different phenotypic consequences, they possess several common pathophysiological features in which monocyte recruitment, macrophage polarization, and enhanced vascular permeability play critical roles. Thus, developing rational targeting strategies tailored to the different stages of the journey of monocytes, from bone marrow to local lesions, and their extravasation from the vasculature in diseased tissues will advance nanomedicine. The integration of in vivo imaging uniquely allows studying nanoparticle kinetics, accumulation, clearance, and biological activity, at levels ranging from subcellular to an entire organism, and will shed light on the fate of intravenously administered nanomedicines. We anticipate that convergence of nanomedicines, biomedical engineering, and life sciences will help to advance clinically relevant therapeutics and diagnostic agents for patients with chronic inflammatory diseases.
Collapse
Affiliation(s)
- Amr Alaarg
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Carlos Pérez-Medina
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Josbert M Metselaar
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Institute for Experimental Molecular Imaging, University Clinic, Helmholtz Institute for Biomedical Engineering, Aachen, Germany
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Gert Storm
- Department of Biomaterials Science and Technology, MIRA Institute for Biomedical Technology and Technical Medicine, University of Twente, Enschede, The Netherlands; Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Faculty of Science, Utrecht University, Utrecht, The Netherlands
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands.
| |
Collapse
|
35
|
Li Z, Huang H, Huang L, Du L, Sun Y, Duan Y. Prevention of Oxidized Low Density Lipoprotein-Induced Endothelial Cell Injury by DA-PLGA-PEG-cRGD Nanoparticles Combined with Ultrasound. Int J Mol Sci 2017; 18:ijms18040815. [PMID: 28406431 PMCID: PMC5412399 DOI: 10.3390/ijms18040815] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 04/06/2017] [Accepted: 04/07/2017] [Indexed: 02/07/2023] Open
Abstract
In general, atherosclerosis is considered to be a form of chronic inflammation. Dexamethasone has anti-inflammatory effects in atherosclerosis, but it was not considered for long-term administration on account of a poor pharmacokinetic profile and adverse side effects. Nanoparticles in which drugs can be dissolved, encapsulated, entrapped or chemically attached to the particle surface have abilities to incorporate dexamethasone and to be used as controlled or targeted drug delivery system. Long circulatory polymeric nanoparticles present as an assisting approach for controlled and targeted release of the encapsulated drug at the atherosclerotic site. Polymeric nanoparticles combined with ultrasound (US) are widely applied in cancer treatment due to their time applications, low cost, simplicity, and safety. However, there are few studies on atherosclerosis treatment using polymeric nanoparticles combined with US. In this study, targeted dexamethasone acetate (DA)-loaded poly (lactide-glycolide)-polyethylene glycol-cRGD (PLGA-PEG-cRGD) nanoparticles (DA-PLGA-PEG-cRGD NPs) were prepared by the emulsion-evaporation method using cRGD modified PLGA-PEG polymeric materials (PLGA-PEG-cRGD) prepared as the carrier. The average particle size of DA-PLGA-PEG-cRGD NPs was 221.6 ± 0.9 nm. Morphology of the nanoparticles was spherical and uniformly dispersed. In addition, the DA released profiles suggested that ultrasound could promote drug release from the nanocarriers and accelerate the rate of release. In vitro, the cellular uptake process of fluorescein isothiocyanate (FITC)@DA-PLGA-PEG-cRGD NPs combined with US into the damaged human umbilical vein endothelial cells (HUVECs) indicated that US promoted rapid intracellular uptake of FITC@DA- PLGA-PEG-cRGD NPs. The cell viability of DA-PLGA-PEG-cRGD NPs combined with US reached 91.9% ± 0.2%, which demonstrated that DA-PLGA-PEG-cRGD NPs combined with US had a positive therapeutic effect on damaged HUVECs. Overall, DA-PLGA-PEG-cRGD NPs in combination with US may provide a promising drug delivery system to enhance the therapeutic effects of these chemotherapeutics at the cellular level.
Collapse
Affiliation(s)
- Zhaojun Li
- Department of Ultrasound, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China.
| | - Hui Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China.
| | - Lili Huang
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China.
| | - Lianfang Du
- Department of Ultrasound, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200080, China.
| | - Ying Sun
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China.
| | - Yourong Duan
- State Key Laboratory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200032, China.
| |
Collapse
|
36
|
Atukorale PU, Covarrubias G, Bauer L, Karathanasis E. Vascular targeting of nanoparticles for molecular imaging of diseased endothelium. Adv Drug Deliv Rev 2017; 113:141-156. [PMID: 27639317 DOI: 10.1016/j.addr.2016.09.006] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2016] [Revised: 09/02/2016] [Accepted: 09/08/2016] [Indexed: 01/08/2023]
Abstract
This review seeks to highlight the enormous potential of targeted nanoparticles for molecular imaging applications. Being the closest point-of-contact, circulating nanoparticles can gain direct access to targetable molecular markers of disease that appear on the endothelium. Further, nanoparticles are ideally suitable to vascular targeting due to geometrically enhanced multivalent attachment on the vascular target. This natural synergy between nanoparticles, vascular targeting and molecular imaging can provide new avenues for diagnosis and prognosis of disease with quantitative precision. In addition to the obvious applications of targeting molecular signatures of vascular diseases (e.g., atherosclerosis), deep-tissue diseases often manifest themselves by continuously altering and remodeling their neighboring blood vessels (e.g., cancer). Thus, the remodeled endothelium provides a wide range of targets for nanoparticles and molecular imaging. To demonstrate the potential of molecular imaging, we present a variety of nanoparticles designed for molecular imaging of cancer or atherosclerosis using different imaging modalities.
Collapse
|
37
|
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: 2.0] [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
|
38
|
Ramos-Cabrer P, Fay F, Sanchez-Gaytan BL, Tang J, Castillo J, Fayad ZA, Mulder WM. Conformational Changes in High-Density Lipoprotein Nanoparticles Induced by High Payloads of Paramagnetic Lipids. ACS OMEGA 2016; 1:470-475. [PMID: 27713933 PMCID: PMC5046173 DOI: 10.1021/acsomega.6b00108] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2016] [Accepted: 09/09/2016] [Indexed: 06/06/2023]
Abstract
High-density lipoprotein (HDL) nanoparticles doped with gadolinium lipids can be used as magnetic resonance imaging diagnostic agents for atherosclerosis. In this study, HDL nanoparticles with different molar fractions of gadolinium lipids (0 < xGd-lipids < 0.33) were prepared, and the MR relaxivity values (r1 and r2) for all compositions were measured. Both r1 and r2 parameters reached a maximal value at a molar fraction of approximately xGd-lipids = 0.2. Higher payloads of gadolinium did not significantly increase relaxivity values but induced changes in the structure of HDL, increasing the size of the particles from dH = 8.2 ± 1.6 to 51.7 ± 7.3 nm. High payloads of gadolinium lipids trigger conformational changes in HDL, with potential effects on the in vivo behavior of the nanoparticles.
Collapse
Affiliation(s)
- Pedro Ramos-Cabrer
- Molecular
Imaging Unit, CIC biomaGUNE, Paseo Miramón 182, 20009 Donostia-San Sebastián, Spain
- Ikerbasque,
Basque Foundation for Science, Maria Diaz de Haro 3, 48011 Bilbao, Spain
- Clinical
Neurosciences Research Laboratory, Department of Neurology, University Clinical Hospital Santiago, Health Sciences
Institute (IDIS), Travesa
da choupana s/n, 15706 Santiago de Compostela, Spain
| | - Francois Fay
- Translational
and Molecular Imaging Institute, Icahn School
of Medicine at Mount Sinai, One Gustave Levy Place, New York, New York 10029, United
States
| | - Brenda L. Sanchez-Gaytan
- Translational
and Molecular Imaging Institute, Icahn School
of Medicine at Mount Sinai, One Gustave Levy Place, New York, New York 10029, United
States
| | - Jun Tang
- Translational
and Molecular Imaging Institute, Icahn School
of Medicine at Mount Sinai, One Gustave Levy Place, New York, New York 10029, United
States
- Radiology
Department, Memorial Sloan Kettering Cancer
Center, 1275 York Avenue, New York, New York 10065, United States
| | - José Castillo
- Clinical
Neurosciences Research Laboratory, Department of Neurology, University Clinical Hospital Santiago, Health Sciences
Institute (IDIS), Travesa
da choupana s/n, 15706 Santiago de Compostela, Spain
| | - Zahi A. Fayad
- Translational
and Molecular Imaging Institute, Icahn School
of Medicine at Mount Sinai, One Gustave Levy Place, New York, New York 10029, United
States
| | - Willem
J. M. Mulder
- Translational
and Molecular Imaging Institute, Icahn School
of Medicine at Mount Sinai, One Gustave Levy Place, New York, New York 10029, United
States
| |
Collapse
|
39
|
Molecular Imaging of Angiogenesis and Vascular Remodeling in Cardiovascular Pathology. J Clin Med 2016; 5:jcm5060057. [PMID: 27275836 PMCID: PMC4929412 DOI: 10.3390/jcm5060057] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 05/19/2016] [Accepted: 05/31/2016] [Indexed: 12/20/2022] Open
Abstract
Angiogenesis and vascular remodeling are involved in a wide array of cardiovascular diseases, from myocardial ischemia and peripheral arterial disease, to atherosclerosis and aortic aneurysm. Molecular imaging techniques to detect and quantify key molecular and cellular players in angiogenesis and vascular remodeling (e.g., vascular endothelial growth factor and its receptors, αvβ3 integrin, and matrix metalloproteinases) can advance vascular biology research and serve as clinical tools for early diagnosis, risk stratification, and selection of patients who would benefit most from therapeutic interventions. To target these key mediators, a number of molecular imaging techniques have been developed and evaluated in animal models of angiogenesis and vascular remodeling. This review of the state of the art molecular imaging of angiogenesis and vascular (and valvular) remodeling, will focus mostly on nuclear imaging techniques (positron emission tomography and single photon emission tomography) that offer high potential for clinical translation.
Collapse
|
40
|
Zhang J, Zu Y, Dhanasekara CS, Li J, Wu D, Fan Z, Wang S. Detection and treatment of atherosclerosis using nanoparticles. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2016; 9. [PMID: 27241794 DOI: 10.1002/wnan.1412] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2015] [Revised: 03/25/2016] [Accepted: 04/12/2016] [Indexed: 01/10/2023]
Abstract
Atherosclerosis is the key pathogenesis of cardiovascular disease, which is a silent killer and a leading cause of death in the United States. Atherosclerosis starts with the adhesion of inflammatory monocytes on the activated endothelial cells in response to inflammatory stimuli. These monocytes can further migrate into the intimal layer of the blood vessel where they differentiate into macrophages, which take up oxidized low-density lipoproteins and release inflammatory factors to amplify the local inflammatory response. After accumulation of cholesterol, the lipid-laden macrophages are transformed into foam cells, the hallmark of the early stage of atherosclerosis. Foam cells can die from apoptosis or necrosis, and the intracellular lipid is deposed in the artery wall forming lesions. The angiogenesis for nurturing cells is enhanced during lesion development. Proteases released from macrophages, foam cells, and other cells degrade the fibrous cap of the lesion, resulting in rupture of the lesion and subsequent thrombus formation. Thrombi can block blood circulation, which represents a major cause of acute heart events and stroke. There are generally no symptoms in the early stages of atherosclerosis. Current detection techniques cannot easily, safely, and effectively detect the lesions in the early stages, nor can they characterize the lesion features such as the vulnerability. While the available therapeutic modalities cannot target specific molecules, cells, and processes in the lesions, nanoparticles appear to have a promising potential in improving atherosclerosis detection and treatment via targeting the intimal macrophages, foam cells, endothelial cells, angiogenesis, proteolysis, apoptosis, and thrombosis. Indeed, many nanoparticles have been developed in improving blood lipid profile and decreasing inflammatory response for enhancing therapeutic efficacy of drugs and decreasing their side effects. WIREs Nanomed Nanobiotechnol 2017, 9:e1412. doi: 10.1002/wnan.1412 For further resources related to this article, please visit the WIREs website.
Collapse
Affiliation(s)
- Jia Zhang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | - Yujiao Zu
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| | | | - Jun Li
- Laboratory Animal Center, Peking University, Beijing, PR China
| | - Dayong Wu
- Nutritional Immunology Laboratory, Jean Mayer Human Nutrition Research Center on Aging, Tufts University, Boston, MA, USA
| | - Zhaoyang Fan
- Department of Electrical and Computer Engineering and Nano Tech Center, Texas Tech University, Lubbock, TX, USA
| | - Shu Wang
- Department of Nutritional Sciences, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
41
|
Palekar RU, Jallouk AP, Lanza GM, Pan H, Wickline SA. Molecular imaging of atherosclerosis with nanoparticle-based fluorinated MRI contrast agents. Nanomedicine (Lond) 2016; 10:1817-32. [PMID: 26080701 DOI: 10.2217/nnm.15.26] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
As atherosclerosis remains one of the most prevalent causes of patient mortality, the ability to diagnose early signs of plaque rupture and thrombosis represents a significant clinical need. With recent advances in nanotechnology, it is now possible to image specific molecular processes noninvasively with MRI, using various types of nanoparticles as contrast agents. In the context of cardiovascular disease, it is possible to specifically deliver contrast agents to an epitope of interest for detecting vascular inflammatory processes, which serve as predecessors to atherosclerotic plaque development. Herein, we review various applications of nanotechnology in detecting atherosclerosis using MRI, with an emphasis on perfluorocarbon nanoparticles and fluorine imaging, along with theranostic prospects of nanotechnology in cardiovascular disease.
Collapse
Affiliation(s)
- Rohun U Palekar
- Department of Biomedical Engineering, Washington University, Whitaker Hall, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA
| | - Andrew P Jallouk
- Department of Medicine, Washington University, Campus Box 8215, 4320 Forest Park Avenue, St Louis, MO 63108, USA
| | - Gregory M Lanza
- Department of Biomedical Engineering, Washington University, Whitaker Hall, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA.,Department of Medicine, Washington University, Campus Box 8215, 4320 Forest Park Avenue, St Louis, MO 63108, USA
| | - Hua Pan
- Department of Medicine, Washington University, Campus Box 8215, 4320 Forest Park Avenue, St Louis, MO 63108, USA
| | - Samuel A Wickline
- Department of Biomedical Engineering, Washington University, Whitaker Hall, Campus Box 1097, One Brookings Drive, St. Louis, MO 63130, USA.,Department of Medicine, Washington University, Campus Box 8215, 4320 Forest Park Avenue, St Louis, MO 63108, USA
| |
Collapse
|
42
|
Arosio D, Casagrande C. Advancement in integrin facilitated drug delivery. Adv Drug Deliv Rev 2016; 97:111-43. [PMID: 26686830 DOI: 10.1016/j.addr.2015.12.001] [Citation(s) in RCA: 101] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2015] [Revised: 11/27/2015] [Accepted: 12/03/2015] [Indexed: 02/06/2023]
Abstract
The research of integrin-targeted anticancer agents has recorded important advancements in ingenious design of delivery systems, based either on the prodrug approach, or on nanoparticle carriers, but for now, none of these has reached a clinical stage of development. Past work in this area has been extensively reviewed by us and others. Thus, the purpose and scope of the present review is to survey the advancement reported in the last 3years, with focus on innovative delivery systems that appear to afford openings for future developments. These systems exploit the labelling with conventional and novel integrin ligands for targeting the interface of cancer cells and of endothelial cells involved in cancer angiogenesis, with the proteins of the extracellular matrix, in the circulation, in tissues, and in tumour stroma, as the site of progression and metastatic evolution of the disease. Furthermore, these systems implement the expertise in the development of nanomedicines to the purpose of achieving preferential biodistribution and uptake in cancer tissues, internalisation in cancer cells, and release of the transported drugs at intracellular sites. The assessment of the value of controlling these factors, and their combination, for future developments requires support of biological testing in appropriate mechanistic models, but also imperatively demand confirmation in therapeutically relevant in vivo models for biodistribution, efficacy, and lack of off-target effects. Thus, among many studies, we have tried to point out the results supported by relevant in vivo studies, and we have emphasised in specific sections those addressing the medical needs of drug delivery to brain tumours, as well as the delivery of oligonucleotides modulating gene-dependent pathological mechanism. The latter could constitute the basis of a promising third branch in the therapeutic armamentarium against cancer, in addition to antibody-based agents and to cytotoxic agents.
Collapse
Affiliation(s)
- Daniela Arosio
- Istituto di Scienze e Tecnologie Molecolari (ISTM), CNR, Via C. Golgi 19, I-20133 Milan, Italy.
| | - Cesare Casagrande
- Università degli Studi di Milano, Dipartimento di Chimica, Via C. Golgi 19, I-20133 Milan, Italy.
| |
Collapse
|
43
|
Mulder WJM. Combination nanotherapy penetrates atherosclerosis. Cardiovasc Res 2016; 109:191-2. [PMID: 26678357 DOI: 10.1093/cvr/cvv271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Affiliation(s)
- Willem J M Mulder
- Translational and Molecular Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA Department of Medical Biochemistry, Academic Medical Center, Amsterdam, The Netherlands
| |
Collapse
|
44
|
Drug-Loaded Perfluorocarbon Nanodroplets for Ultrasound-Mediated Drug Delivery. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2016; 880:221-41. [DOI: 10.1007/978-3-319-22536-4_13] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
45
|
Schmieder AH, Caruthers SD, Keupp J, Wickline SA, Lanza GM. Recent Advances in 19Fluorine Magnetic Resonance Imaging with Perfluorocarbon Emulsions. ENGINEERING (BEIJING, CHINA) 2015; 1:475-489. [PMID: 27110430 PMCID: PMC4841681 DOI: 10.15302/j-eng-2015103] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
The research roots of 19fluorine (19F) magnetic resonance imaging (MRI) date back over 35 years. Over that time span, 1H imaging flourished and was adopted worldwide with an endless array of applications and imaging approaches, making magnetic resonance an indispensable pillar of biomedical diagnostic imaging. For many years during this timeframe, 19F imaging research continued at a slow pace as the various attributes of the technique were explored. However, over the last decade and particularly the last several years, the pace and clinical relevance of 19F imaging has exploded. In part, this is due to advances in MRI instrumentation, 19F/1H coil designs, and ultrafast pulse sequence development for both preclinical and clinical scanners. These achievements, coupled with interest in the molecular imaging of anatomy and physiology, and combined with a cadre of innovative agents, have brought the concept of 19F into early clinical evaluation. In this review, we attempt to provide a slice of this rich history of research and development, with a particular focus on liquid perfluorocarbon compound-based agents.
Collapse
Affiliation(s)
- Anne H. Schmieder
- Division of Cardiology, Washington University School of Medical, St. Louis, MO 63110, USA
| | - Shelton D. Caruthers
- Toshiba Medical Research Institute USA, Inc., Cleveland, OH 44143, USA
- Department of Biomedical Engineering, Washington University, St. Louis, MO 63130, USA
| | - Jochen Keupp
- Philips Research Hamburg, Hamburg 22335, Germany
| | - Samuel A. Wickline
- Division of Cardiology, Washington University School of Medical, St. Louis, MO 63110, USA
| | - Gregory M. Lanza
- Division of Cardiology, Washington University School of Medical, St. Louis, MO 63110, USA
- Correspondence author.
| |
Collapse
|
46
|
Pan D, Pham CTN, Weilbaecher KN, Tomasson MH, Wickline SA, Lanza GM. Contact-facilitated drug delivery with Sn2 lipase labile prodrugs optimize targeted lipid nanoparticle drug delivery. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2015; 8:85-106. [PMID: 26296541 PMCID: PMC4709477 DOI: 10.1002/wnan.1355] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 05/18/2015] [Indexed: 01/10/2023]
Abstract
Sn2 lipase labile phospholipid prodrugs in conjunction with contact-facilitated drug delivery offer an important advancement in Nanomedicine. Many drugs incorporated into nanosystems, targeted or not, are substantially lost during circulation to the target. However, favorably altering the pharmacokinetics and volume of distribution of systemic drug delivery can offer greater efficacy with lower toxicity, leading to new prolonged-release nanoexcipients. However, the concept of achieving Paul Erhlich's inspired vision of a 'magic bullet' to treat disease has been largely unrealized due to unstable nanomedicines, nanosystems achieving low drug delivery to target cells, poor intracellular bioavailability of endocytosed nanoparticle payloads, and the substantial biological barriers of extravascular particle penetration into pathological sites. As shown here, Sn2 phospholipid prodrugs in conjunction with contact-facilitated drug delivery prevent premature drug diffusional loss during circulation and increase target cell bioavailability. The Sn2 phospholipid prodrug approach applies equally well for vascular constrained lipid-encapsulated particles and micelles the size of proteins that penetrate through naturally fenestrated endothelium in the bone marrow or thin-walled venules of an inflamed microcirculation. At one time Nanomedicine was considered a 'Grail Quest' by its loyal opposition and even many in the field adsorbing the pains of a long-learning curve about human biology and particles. However, Nanomedicine with innovations like Sn2 phospholipid prodrugs has finally made 'made the turn' toward meaningful translational success.
Collapse
Affiliation(s)
- Dipanjan Pan
- Departments of Bioengineering, Materials Science and Engineering, Beckman Institute, University of Illinois, Urbana-Champaign, IL, USA
| | - Christine T N Pham
- Division of Rheumatology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.,Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Katherine N Weilbaecher
- Division of Oncology, Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | - Michael H Tomasson
- Division of Oncology, Department of Medicine, Washington University Medical School, St. Louis, MO, USA
| | - Samuel A Wickline
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Gregory M Lanza
- Division of Cardiology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|
47
|
Yoshimura K, Nagasawa A, Kudo J, Onoda M, Morikage N, Furutani A, Aoki H, Hamano K. Inhibitory effect of statins on inflammation-related pathways in human abdominal aortic aneurysm tissue. Int J Mol Sci 2015; 16:11213-28. [PMID: 25993292 PMCID: PMC4463697 DOI: 10.3390/ijms160511213] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2015] [Revised: 03/27/2015] [Accepted: 03/30/2015] [Indexed: 12/03/2022] Open
Abstract
HMG-CoA (3-hydroxy-3-methylglutaryl-coenzyme A) reductase inhibitors (statins) have been suggested to attenuate abdominal aortic aneurysm (AAA) growth. However, the effects of statins in human AAA tissues are not fully elucidated. The aim of this study was to investigate the direct effects of statins on proinflammatory molecules in human AAA walls in ex vivo culture. Simvastatin strongly inhibited the activation of nuclear factor (NF)-κB induced by tumor necrosis factor (TNF)-α in human AAA walls, but showed little effect on c-jun N-terminal kinase (JNK) activation. Simvastatin, as well as pitavastatin significantly reduced the secretion of matrix metalloproteinase (MMP)-9, monocyte chemoattractant protein (MCP)-2 and epithelial neutrophil-activating peptide (CXCL5) under both basal and TNF-α-stimulated conditions. Similar to statins, the Rac1 inhibitor NSC23766 significantly inhibited the activation of NF-κB, accompanied by a decreased secretion of MMP-9, MCP-2 and CXCL5. Moreover, the effect of simvastatin and the JNK inhibitor SP600125 was additive in inhibiting the secretion of MMP-9, MCP-2 and CXCL5. These findings indicate that statins preferentially inhibit the Rac1/NF-κB pathway to suppress MMP-9 and chemokine secretion in human AAA, suggesting a mechanism for the potential effect of statins in attenuating AAA progression.
Collapse
Affiliation(s)
- Koichi Yoshimura
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
- Graduate School of Health and Welfare, Yamaguchi Prefectural University, Yamaguchi 753-8502, Japan.
| | - Ayako Nagasawa
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
- Division of Thoracic and Cardiovascular Surgery, Niigata University Graduate School of Medical and Dental Science, Niigata 951-8510, Japan.
| | - Junichi Kudo
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| | - Masahiko Onoda
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| | - Noriyasu Morikage
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| | - Akira Furutani
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| | - Hiroki Aoki
- Cardiovascular Research Institute, Kurume University, Kurume 830-0011, Japan.
| | - Kimikazu Hamano
- Department of Surgery and Clinical Science, Yamaguchi University Graduate School of Medicine, Ube 755-8505, Japan.
| |
Collapse
|
48
|
Wang K, Pan D, Schmieder AH, Senpan A, Caruthers SD, Cui G, Allen JS, Zhang H, Shen B, Lanza GM. Atherosclerotic neovasculature MR imaging with mixed manganese-gadolinium nanocolloids in hyperlipidemic rabbits. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2015; 11:569-78. [PMID: 25652897 DOI: 10.1016/j.nano.2014.12.008] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2014] [Revised: 12/09/2014] [Accepted: 12/13/2014] [Indexed: 10/24/2022]
Abstract
A high r1 relaxivity manganese-gadolinium nanocolloid (αvβ3-MnOL-Gd NC) was developed and effectively detected atherosclerotic angiogenesis in rabbits fed cholesterol-rich diets for 12 months using a clinical MRI scanner (3T). 3D mapping of neovasculature signal intensity revealed the spatial coherence and intensity of plaque angiogenic expansion, which may, with other high risk MR bioindicators, help identify high-risk patients with moderate (40% to 60%) vascular stenosis. Microscopy confirmed the predominant media and plaque distribution of fluorescent αvβ3-MnOL-Gd NC, mirroring the MR data. An expected close spatial association of αvβ3-integrin neovasculature and macrophages was noted, particularly within plaque shoulder regions. Manganese oleate bioelimination occurred via the biliary system into feces. Gd-DOTA was eliminated through the bile-fecal and renal excretion routes. αvβ3-MnOL-Gd NC offers an effective vehicle for T1w neovascular imaging in atherosclerosis. From the clinical editor: Cerebrovascular accidents are a leading cause of mortality and morbidity worldwide. The acute formation of thrombus following atherosclerotic plaque rupture has been well recognized as the etiology of stroke. The authors studied microanatomical features of vulnerable atherosclerotic plaque in this article, in an attempt to identify those with high risk of rupture. Gadolinium-manganese hybrid nanocolloid (MnOL-Gd NC) was developed as a novel contrast agent for MRI. They show that this agent is effective in providing neovascular imaging.
Collapse
Affiliation(s)
- Kezheng Wang
- Department of Radiology, the Fourth Hospital of Harbin Medical University and Molecular Imaging Center of Harbin Medical University, Harbin, China; Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Dipanjan Pan
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Anne H Schmieder
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Angana Senpan
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Shelton D Caruthers
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Grace Cui
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - John S Allen
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Huiying Zhang
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Baozhong Shen
- Department of Radiology, the Fourth Hospital of Harbin Medical University and Molecular Imaging Center of Harbin Medical University, Harbin, China.
| | - Gregory M Lanza
- Division of Cardiology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
49
|
Mulder WJM, Jaffer FA, Fayad ZA, Nahrendorf M. Imaging and nanomedicine in inflammatory atherosclerosis. Sci Transl Med 2015; 6:239sr1. [PMID: 24898749 DOI: 10.1126/scitranslmed.3005101] [Citation(s) in RCA: 147] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Bioengineering provides unique opportunities to better understand and manage atherosclerotic disease. The field is entering a new era that merges the latest biological insights into inflammatory disease processes with targeted imaging and nanomedicine. Preclinical cardiovascular molecular imaging allows the in vivo study of targeted nanotherapeutics specifically directed toward immune system components that drive atherosclerotic plaque development and complication. The first multicenter trials highlight the potential contribution of multimodality imaging to more efficient drug development. This review describes how the integration of engineering, nanotechnology, and cardiovascular immunology may yield precision diagnostics and efficient therapeutics for atherosclerosis and its ischemic complications.
Collapse
Affiliation(s)
- Willem J M Mulder
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA. Department of Vascular Medicine, Academic Medical Center, Meibergdreef 9, 1105 AZ Amsterdam, the Netherlands.
| | - Farouc A Jaffer
- Cardiovascular Research Center and Cardiology Division, Massachusetts General Hospital and Harvard Medical School, Simches Research Building, 185 Cambridge Street, Boston, MA 02114, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Department of Radiology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Matthias Nahrendorf
- Center for Systems Biology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA
| |
Collapse
|
50
|
Maranhão RC, Tavares ER. Advances in non-invasive drug delivery for atherosclerotic heart disease. Expert Opin Drug Deliv 2015; 12:1135-47. [DOI: 10.1517/17425247.2015.999663] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|