1
|
Ullah A, Ullah M, Lim SI. Recent advancements in nanotechnology based drug delivery for the management of cardiovascular disease. Curr Probl Cardiol 2024; 49:102396. [PMID: 38266693 DOI: 10.1016/j.cpcardiol.2024.102396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2024] [Accepted: 01/14/2024] [Indexed: 01/26/2024]
Abstract
Cardiovascular diseases (CVDs) constitute a predominant cause of both global mortality and morbidity. To address the challenges in the early diagnosis and management of CVDs, there is growing interest in the field of nanotechnology and nanomaterials to develop innovative diagnostic and therapeutic approaches. This review focuses on the recent advancements in nanotechnology-based diagnostic techniques, including cardiac immunoassays (CIA), cardiac circulating biomarkers, cardiac exosomal biomarkers, and molecular Imaging (MOI). Moreover, the article delves into the exciting developments in nanoparticles (NPs), biomimetic NPs, nanofibers, nanogels, and nanopatchs for cardiovascular applications. And discuss how these nanoscale technologies can improve the precision, sensitivity, and speed of CVD diagnosis and management. While highlighting their vast potential, we also address the limitations and challenges that must be overcome to harness these innovations successfully. Furthermore, this review focuses on the emerging opportunities for personalized and effective cardiovascular care through the integration of nanotechnology, ultimately aiming to reduce the global burden of CVDs.
Collapse
Affiliation(s)
- Aziz Ullah
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea
| | - Muneeb Ullah
- College of Pharmacy, Pusan National University, Busandaehak-ro 63 beon-gil 2, Geumjeong-gu, Busan 46241, Republic of Korea
| | - Sung In Lim
- Department of Chemical Engineering, Pukyong National University, Yongso-ro 45, Nam-gu, Engineering Bldg#1, Rm1108, Busan 48513, Republic of Korea.
| |
Collapse
|
2
|
Chen Q, Duan X, Yu Y, Ni R, Song G, Yang X, Zhu L, Zhong Y, Zhang K, Qu K, Qin X, Wu W. Target Functionalized Carbon Dot Nanozymes with Dual-Model Photoacoustic and Fluorescence Imaging for Visual Therapy in Atherosclerosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2307441. [PMID: 38145362 PMCID: PMC10853701 DOI: 10.1002/advs.202307441] [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: 10/06/2023] [Revised: 11/21/2023] [Indexed: 12/26/2023]
Abstract
Multifunctional nanomedicines have been used in atherosclerosis theranostics. Herein, phosphatidylserine-specific peptide CLIKKPF-functionalized carbon-dots nanozymes (pep-CDs) are reported for specific and efficient noninvasive theranostic of atherosclerosis. Surprisingly, pep-CDs are discovered to not only inherit the inherent properties of carbon dots (CDs), including deep-red fluorescence emission, photoacoustic response, and superoxide dismutase-like antioxidant, and anti-inflammatory activities but also possess the ability to target recognition on foam cells and target localization on plaques due to the specific interaction of CLIKKPF with phosphatidylserine on the membrane outer surface of foam cells. Furthermore, the target localization effect of pep-CDs vastly promotes the efficient accumulation of CDs in plaque, thus maximizing AS theranostic of CDs. Interestingly, pep-CDs could be developed to image plaque for monitoring atherosclerosis pathological progression in real-time resulting from the different content of foam cells. This work on the one hand proposes a simple and feasible strategy to construct theranostic nanoplatform employing only a single functional unit (i.e., multifunctional CDs) to simplify the fabrication procedure, on the other hand, highlights the advantages of the active target auxiliary mode for atherosclerosis theranostic applications.
Collapse
Affiliation(s)
- Qiao Chen
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
| | - Xinmei Duan
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
| | - Yao Yu
- Thyroid Breast Surgery DepartmentDazhou Central HospitalDazhou635000China
| | - Rongrong Ni
- Medical DepartmentSouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Guojing Song
- UrologySouthwest HospitalThird Military Medical UniversityChongqing400038China
| | - Xu Yang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
| | - Li Zhu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
| | - Yuan Zhong
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
| | - Kun Zhang
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
- Chongqing University Three Gorges HospitalChongqing404000China
| | - Kai Qu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
- Chongqing University Three Gorges HospitalChongqing404000China
| | - Xian Qin
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
- Chongqing University Three Gorges HospitalChongqing404000China
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of EducationState and Local Joint Engineering Laboratory for Vascular ImplantsBioengineering College of Chongqing UniversityChongqing400044China
- Jin Feng LaboratoryChongqing401329China
| |
Collapse
|
3
|
Smith BR, Edelman ER. Nanomedicines for cardiovascular disease. NATURE CARDIOVASCULAR RESEARCH 2023; 2:351-367. [PMID: 39195953 DOI: 10.1038/s44161-023-00232-y] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Accepted: 01/25/2023] [Indexed: 08/29/2024]
Abstract
The leading cause of death in the world, cardiovascular disease (CVD), remains a formidable condition for researchers, clinicians and patients alike. CVD comprises a broad collection of diseases spanning the heart, the vasculature and the blood that runs through and interconnects them. Limitations in CVD therapeutic and diagnostic landscapes have generated excitement for advances in nanomedicine, a field focused on improving patient outcomes through transformative therapies, imaging agents and ex vivo diagnostics. CVD nanomedicines are fundamentally shaped by their intended clinical application, including (1) cardiac or heart-related biomaterials, which can be functionally (for example, mechanically, immunologically, electrically) improved by incorporating nanomaterials; (2) the vasculature, involving systemically injected nanotherapeutics and imaging nanodiagnostics, nano-enabled biomaterials or tissue-nanoengineered solutions; and (3) improving the sensitivity and/or specificity of ex vivo diagnostic devices for patient samples. While immunotherapy has developed into a key pillar of oncology in the past dozen years, CVD immunotherapy and immunoimaging are recently emergent and likely to factor substantially in CVD management in the coming decade. The nanomaterials in CVD-related clinical trials and many promising preclinical strategies indicate that nanomedicine is on the cusp of greatly impacting patients with CVD. Here we review these recent advances, highlighting key clinical opportunities in the rapidly emerging field of CVD nanomedicine.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Department of Biomedical Engineering and Institute for Quantitative Health Science and Engineering (IQ), Michigan State University, East Lansing, MI, USA.
| | - Elazer R Edelman
- Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, MA, USA
- Cardiovascular Division, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
4
|
Tuguntaev RG, Hussain A, Fu C, Chen H, Tao Y, Huang Y, Liu L, Liang XJ, Guo W. Bioimaging guided pharmaceutical evaluations of nanomedicines for clinical translations. J Nanobiotechnology 2022; 20:236. [PMID: 35590412 PMCID: PMC9118863 DOI: 10.1186/s12951-022-01451-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Accepted: 05/05/2022] [Indexed: 11/25/2022] Open
Abstract
Nanomedicines (NMs) have emerged as an efficient approach for developing novel treatment strategies against a variety of diseases. Over the past few decades, NM formulations have received great attention, and a large number of studies have been performed in this field. Despite this, only about 60 nano-formulations have received industrial acceptance and are currently available for clinical use. Their in vivo pharmaceutical behavior is considered one of the main challenges and hurdles for the effective clinical translation of NMs, because it is difficult to monitor the pharmaceutic fate of NMs in the biological environment using conventional pharmaceutical evaluations. In this context, non-invasive imaging modalities offer attractive solutions, providing the direct monitoring and quantification of the pharmacokinetic and pharmacodynamic behavior of labeled NMs in a real-time manner. Imaging evaluations have great potential for revealing the relationship between the physicochemical properties of NMs and their pharmaceutical profiles in living subjects. In this review, we introduced imaging techniques that can be used for in vivo NM evaluations. We also provided an overview of various studies on the influence of key parameters on the in vivo pharmaceutical behavior of NMs that had been visualized in a non-invasive and real-time manner.
Collapse
Affiliation(s)
- Ruslan G Tuguntaev
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Abid Hussain
- Advanced Research Institute of Multidisciplinary Science, School of Life Science, School of Medical Technology (Institute of Engineering Medicine), Key Laboratory of Molecular Medicine and Biotherapy, Key Laboratory of Medical Molecular Science and Pharmaceutics Engineering, Beijing Institute of Technology, Beijing, 100081, China
| | - Chenxing Fu
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Haoting Chen
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Ying Tao
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China
| | - Yan Huang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, 226001, China
| | - Lu Liu
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China.
| | - Xing-Jie Liang
- Chinese Academy of Sciences (CAS) Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing, 100190, People's Republic of China.
| | - Weisheng Guo
- Department of Minimally Invasive Interventional Radiology, Key Laboratory of Molecular Target & Clinical Pharmacology, School of Pharmaceutical Sciences & the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510260, People's Republic of China.
| |
Collapse
|
5
|
Gifani M, Eddins DJ, Kosuge H, Zhang Y, Paluri SLA, Larson T, Leeper N, Herzenberg LA, Gambhir SS, McConnell MV, Ghosn EEB, Smith BR. Ultra-selective carbon nanotubes for photoacoustic imaging of inflamed atherosclerotic plaques. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2101005. [PMID: 34733130 PMCID: PMC8559995 DOI: 10.1002/adfm.202101005] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Indexed: 05/29/2023]
Abstract
Disruption of vulnerable atherosclerotic plaques often leads to myocardial infarction and stroke, the leading causes of morbidity and mortality in the United States. A diagnostic method that detects high-risk atherosclerotic plaques at early stages could prevent these sequelae. The abundance of immune cells in the arterial wall, especially inflammatory Ly-6Chi monocytes and foamy macrophages, is indicative of plaque inflammation, and may be associated with plaque vulnerability. Hence, we sought to develop a new method that specifically targets these immune cells to offer clinically-relevant diagnostic information about cardiovascular disease. We combine ultra-selective nanoparticle targeting of Ly-6Chi monocytes and foamy macrophages with clinically-viable photoacoustic imaging (PAI) in order to precisely and specifically image inflamed plaques ex vivo in a mouse model that mimics human vulnerable plaques histopathologically. Within the plaques, high-dimensional single-cell flow cytometry (13-parameter) showed that our nanoparticles were almost-exclusively taken up by the Ly-6Chi monocytes and foamy macrophages that heavily infiltrate plaques. PAI identified inflamed atherosclerotic plaques that display ~6-fold greater signal compared to controls (P<0.001) six hours after intravenous injection of ultra-selective carbon nanotubes, with in vivo corroboration via optical imaging. Our highly selective strategy may provide a targeted, non-invasive imaging strategy to accurately identify and diagnose inflamed atherosclerotic lesions.
Collapse
Affiliation(s)
- Mahsa Gifani
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Devon J. Eddins
- Departments of Medicine and Pediatrics, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - Hisanori Kosuge
- Division of Cardiovascular Medicine and Vascular Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Yapei Zhang
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Sesha L. A. Paluri
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
| | - Timothy Larson
- Departments of Radiology, Bioengineering, and Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Nicholas Leeper
- Division of Cardiovascular Medicine and Vascular Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Sanjiv Sam Gambhir
- Departments of Radiology, Bioengineering, and Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Michael V. McConnell
- Division of Cardiovascular Medicine and Vascular Surgery, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Eliver E. B. Ghosn
- Departments of Medicine and Pediatrics, Lowance Center for Human Immunology, Emory University, Atlanta, GA 30322, USA
| | - Bryan Ronain Smith
- Department of Biomedical Engineering, Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, MI 48824, USA
- Departments of Radiology, Bioengineering, and Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
6
|
Friedrich RP, Cicha I, Alexiou C. Iron Oxide Nanoparticles in Regenerative Medicine and Tissue Engineering. NANOMATERIALS 2021; 11:nano11092337. [PMID: 34578651 PMCID: PMC8466586 DOI: 10.3390/nano11092337] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Revised: 09/03/2021] [Accepted: 09/06/2021] [Indexed: 12/13/2022]
Abstract
In recent years, many promising nanotechnological approaches to biomedical research have been developed in order to increase implementation of regenerative medicine and tissue engineering in clinical practice. In the meantime, the use of nanomaterials for the regeneration of diseased or injured tissues is considered advantageous in most areas of medicine. In particular, for the treatment of cardiovascular, osteochondral and neurological defects, but also for the recovery of functions of other organs such as kidney, liver, pancreas, bladder, urethra and for wound healing, nanomaterials are increasingly being developed that serve as scaffolds, mimic the extracellular matrix and promote adhesion or differentiation of cells. This review focuses on the latest developments in regenerative medicine, in which iron oxide nanoparticles (IONPs) play a crucial role for tissue engineering and cell therapy. IONPs are not only enabling the use of non-invasive observation methods to monitor the therapy, but can also accelerate and enhance regeneration, either thanks to their inherent magnetic properties or by functionalization with bioactive or therapeutic compounds, such as drugs, enzymes and growth factors. In addition, the presence of magnetic fields can direct IONP-labeled cells specifically to the site of action or induce cell differentiation into a specific cell type through mechanotransduction.
Collapse
|
7
|
Zia A, Wu Y, Nguyen T, Wang X, Peter K, Ta HT. The choice of targets and ligands for site-specific delivery of nanomedicine to atherosclerosis. Cardiovasc Res 2021; 116:2055-2068. [PMID: 32077918 DOI: 10.1093/cvr/cvaa047] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2019] [Revised: 12/23/2019] [Accepted: 02/17/2020] [Indexed: 12/22/2022] Open
Abstract
As nanotechnologies advance into clinical medicine, novel methods for applying nanomedicine to cardiovascular diseases are emerging. Extensive research has been undertaken to unlock the complex pathogenesis of atherosclerosis. However, this complexity presents challenges to develop effective imaging and therapeutic modalities for early diagnosis and acute intervention. The choice of ligand-receptor system vastly influences the effectiveness of nanomedicine. This review collates current ligand-receptor systems used in targeting functionalized nanoparticles for diagnosis and treatment of atherosclerosis. Our focus is on the binding affinity and selectivity of ligand-receptor systems, as well as the relative abundance of targets throughout the development and progression of atherosclerosis. Antibody-based targeting systems are currently the most commonly researched due to their high binding affinities when compared with other ligands, such as antibody fragments, peptides, and other small molecules. However, antibodies tend to be immunogenic due to their size. Engineering antibody fragments can address this issue but will compromise their binding affinity. Peptides are promising ligands due to their synthetic flexibility and low production costs. Alongside the aforementioned binding affinity of ligands, the choice of target and its abundance throughout distinct stages of atherosclerosis and thrombosis is relevant to the intended purpose of the nanomedicine. Further studies to investigate the components of atherosclerotic plaques are required as their cellular and molecular profile shifts over time.
Collapse
Affiliation(s)
- Adil Zia
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Yuao Wu
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| | - Tuan Nguyen
- School of Chemical Engineering, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Xiaowei Wang
- Baker Heart and Diabetes Institute, Melbourne, VIC 3000, Australia
| | - Karlheinz Peter
- Baker Heart and Diabetes Institute, Melbourne, VIC 3000, Australia
| | - Hang T Ta
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, Brisbane, QLD 4072, Australia.,School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, Woolloongabba, QLD 4102, Australia
| |
Collapse
|
8
|
Chauhan DS, Dhasmana A, Laskar P, Prasad R, Jain NK, Srivastava R, Jaggi M, Chauhan SC, Yallapu MM. Nanotechnology synergized immunoengineering for cancer. Eur J Pharm Biopharm 2021; 163:72-101. [PMID: 33774162 PMCID: PMC8170847 DOI: 10.1016/j.ejpb.2021.03.010] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 03/06/2021] [Accepted: 03/15/2021] [Indexed: 12/26/2022]
Abstract
Novel strategies modulating the immune system yielded enhanced anticancer responses and improved cancer survival. Nevertheless, the success rate of immunotherapy in cancer treatment has been below expectation(s) due to unpredictable efficacy and off-target effects from systemic dosing of immunotherapeutic(s). As a result, there is an unmet clinical need for improving conventional immunotherapy. Nanotechnology offers several new strategies, multimodality, and multiplex biological targeting advantage to overcome many of these challenges. These efforts enable programming the pharmacodynamics, pharmacokinetics, and delivery of immunomodulatory agents/co-delivery of compounds to prime at the tumor sites for improved therapeutic benefits. This review provides an overview of the design and clinical principles of biomaterials driven nanotechnology and their potential use in personalized nanomedicines, vaccines, localized tumor modulation, and delivery strategies for cancer immunotherapy. In this review, we also summarize the latest highlights and recent advances in combinatorial therapies availed in the treatment of cold and complicated tumors. It also presents key steps and parameters implemented for clinical success. Finally, we analyse, discuss, and provide clinical perspectives on the integrated opportunities of nanotechnology and immunology to achieve synergistic and durable responses in cancer treatment.
Collapse
Affiliation(s)
- Deepak S Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Anupam Dhasmana
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Partha Laskar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Rajendra Prasad
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Nishant K Jain
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Rohit Srivastava
- Department of Biosciences and Bioengineering, Indian Institute of Technology Bombay, Powai, Mumbai 400076, India
| | - Meena Jaggi
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Subhash C Chauhan
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA
| | - Murali M Yallapu
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA; South Texas Center of Excellence in Cancer Research, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX 78504, USA.
| |
Collapse
|
9
|
Mauro N, Utzeri MA, Varvarà P, Cavallaro G. Functionalization of Metal and Carbon Nanoparticles with Potential in Cancer Theranostics. Molecules 2021; 26:3085. [PMID: 34064173 PMCID: PMC8196792 DOI: 10.3390/molecules26113085] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 01/19/2023] Open
Abstract
Cancer theranostics is a new concept of medical approach that attempts to combine in a unique nanoplatform diagnosis, monitoring and therapy so as to provide eradication of a solid tumor in a non-invasive fashion. There are many available solutions to tackle cancer using theranostic agents such as photothermal therapy (PTT) and photodynamic therapy (PDT) under the guidance of imaging techniques (e.g., magnetic resonance-MRI, photoacoustic-PA or computed tomography-CT imaging). Additionally, there are several potential theranostic nanoplatforms able to combine diagnosis and therapy at once, such as gold nanoparticles (GNPs), graphene oxide (GO), superparamagnetic iron oxide nanoparticles (SPIONs) and carbon nanodots (CDs). Currently, surface functionalization of these nanoplatforms is an extremely useful protocol for effectively tuning their structures, interface features and physicochemical properties. This approach is much more reliable and amenable to fine adjustment, reaching both physicochemical and regulatory requirements as a function of the specific field of application. Here, we summarize and compare the most promising metal- and carbon-based theranostic tools reported as potential candidates in precision cancer theranostics. We focused our review on the latest developments in surface functionalization strategies for these nanosystems, or hybrid nanocomposites consisting of their combination, and discuss their main characteristics and potential applications in precision cancer medicine.
Collapse
Affiliation(s)
- Nicolò Mauro
- Lab of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy; (M.A.U.); (P.V.); (G.C.)
| | - Mara Andrea Utzeri
- Lab of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy; (M.A.U.); (P.V.); (G.C.)
| | - Paola Varvarà
- Lab of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy; (M.A.U.); (P.V.); (G.C.)
| | - Gennara Cavallaro
- Lab of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, via Archirafi 32, 90123 Palermo, Italy; (M.A.U.); (P.V.); (G.C.)
- Advanced Technologies Network Center, University of Palermo, Viale delle Scienze, Ed. 18, 90128 Palermo, Italy
| |
Collapse
|
10
|
He Y, Yi C, Zhang X, Zhao W, Yu D. Magnetic graphene oxide: Synthesis approaches, physicochemical characteristics, and biomedical applications. Trends Analyt Chem 2021. [DOI: 10.1016/j.trac.2021.116191] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
11
|
Wang P, Kim T, Harada M, Contag C, Huang X, Smith BR. Nano-immunoimaging. NANOSCALE HORIZONS 2020; 5:628-653. [PMID: 32226975 DOI: 10.1039/c9nh00514e] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Immunoimaging is a rapidly growing field stoked in large part by the intriguing triumphs of immunotherapy. On the heels of immunotherapy's successes, there exists a growing need to evaluate tumor response to therapy particularly immunotherapy, stratify patients into responders vs. non-responders, identify inflammation, and better understand the fundamental roles of immune system components to improve both immunoimaging and immunotherapy. Innovative nanomaterials have begun to provide novel opportunities for immunoimaging, in part due to their sensitivity, modularity, capacity for many potentially varied ligands (high avidity), and potential for multifunctionality/multimodality imaging. This review strives to comprehensively summarize the integration of nanotechnology and immunoimaging, and the field's potential for clinical applications.
Collapse
Affiliation(s)
- Ping Wang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA
| | - Taeho Kim
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Masako Harada
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA
| | - Christopher Contag
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Precision Health Program, Michigan State University, East Lansing, MI 488824, USA and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Microbiology & Molecular Genetics, Michigan State University, East Lansing, MI 488824, USA
| | - Xuefei Huang
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Chemistry, Michigan State University, East Lansing, MI 488824, USA
| | - Bryan Ronain Smith
- Institute for Quantitative Health Science and Engineering, Michigan State University, 775 Woodlot Drive, Room #1118, East Lansing, MI 488824, USA. and Department of Biomedical Engineering, Michigan State University, East Lansing, MI 488824, USA and Department of Radiology, Stanford University, Stanford, CA 94306, USA
| |
Collapse
|
12
|
Abstract
The past several decades have brought significant advances in the application of clinical and preclinical nanoparticulate drugs in the field of cancer, but nanodrug development in cardiovascular disease has lagged in comparison. Improved understanding of the spatiotemporal kinetics of nanoparticle delivery to atherosclerotic plaques is required to optimize preclinical nanodrug delivery and to drive their clinical translation. Mechanistic studies using super-resolution and correlative light microscopy/electron microscopy permit a broad, ultra-high-resolution picture of how endothelial barrier integrity impacts the enhanced permeation and retention (EPR) effect for nanoparticles as a function of both atherosclerosis progression and metabolic therapy. Studies by Beldman et al. in the December issue of ACS Nano suggest atherosclerotic plaque progression supports endothelial junction stabilization, which can reduce nanoparticle entry into plaques, and metabolic therapy may induce similar effects. Herein, we examine the potential for advanced dynamic intravital microscopy-based mechanistic studies of nanoparticle entry into atherosclerotic plaques to shed light on the advantages of free extravasation versus immune-mediated nanoparticle uptake for effective clinical translation. We further explore the potential combination of metabolic therapy with another emerging cardiovascular disease treatment paradigm-efferocytosis stimulation-to enhance atherosclerotic plaque regression.
Collapse
Affiliation(s)
- Yogendra Kanthi
- Division of Cardiovascular Medicine, Frankel Cardiovascular Center , University of Michigan , Ann Arbor , Michigan 48109 , United States
- Section of Cardiology , Ann Arbor Veterans Health System , Ann Arbor , Michigan 48109 , United States
| | - Adam de la Zerda
- Department of Structural Biology , Stanford University , Stanford , California 94305 , United States
- Department of Electrical Engineering , Stanford University , Stanford , California 94305 , United States
- Molecular Imaging Program at Stanford and the Bio-X Program , Stanford , California 94305 , United States
- Biophysics Program at Stanford , Stanford , California 94305 , United States
- The Chan Zuckerberg Biohub , San Francisco , California 94158 , United States
| | - Bryan Ronain Smith
- Department of Biomedical Engineering , Michigan State University , East Lansing , Michigan 48824 , United States
- Institute for Quantitative Health Science and Engineering , East Lansing , Michigan 48824 , United States
| |
Collapse
|
13
|
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
|
14
|
Lavin Plaza B, Theodoulou I, Rashid I, Hajhosseiny R, Phinikaridou A, Botnar RM. Molecular Imaging in Ischemic Heart Disease. CURRENT CARDIOVASCULAR IMAGING REPORTS 2019; 12:31. [PMID: 31281564 PMCID: PMC6557873 DOI: 10.1007/s12410-019-9500-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Purpose of Review The purpose of this paper is to review current and new modalities to image key biological processes in ischemic heart disease and after myocardial infarction non-invasively. Recent Findings New imaging targets have been developed to detect and quantify myocardial damage after ischemia. Although positron emission tomography (PET) has been leading the development of new probes in the past, continuous improvements of magnetic resonance imaging (MRI) together with the development of new novel MRI contrast agents opens new research avenues including the combination of both PET and MRI to obtain anatomic, functional, and molecular information simultaneously, which is not possible from a single imaging session. Summary This review summarizes the state of art of non-invasive molecular imaging of the myocardium during ischemia and after myocardial infarction using PET and MRI. We also describe the different contrast agents that have been developed to image the different phases of cardiac healing and the biological processes associated with each of those phases. Importantly, here we focus on imaging of inflammation as it is the key biological process that orchestrates clearance of dead cells, tissue remodeling, cardiac repair, and future outcome. We also focus on clinical translation of some of the novel contrast agents that have been tested in patients and discuss the need for larger, multi-center patient studies to fully validate the applicability of new imaging probes.
Collapse
Affiliation(s)
- Begoña Lavin Plaza
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK
| | - Iakovos Theodoulou
- 2Faculty of Life Sciences and Medicine, King's College London, London, UK
| | - Imran Rashid
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK
| | - Reza Hajhosseiny
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK
| | - Alkystis Phinikaridou
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK
| | - Rene M Botnar
- 1School of Biomedical Engineering and Imaging Sciences, King's College London, 3rd Floor, Lambeth wing, St Thomas Hospital, London, SE1 7EH UK.,3Escuela de Ingeniería, Pontificia Universidad Católica de Chile, Santiago, Chile
| |
Collapse
|
15
|
Labeling of endothelial cells with magnetic microbeads by angiophagy. Biotechnol Lett 2018; 40:1189-1200. [PMID: 29876793 DOI: 10.1007/s10529-018-2581-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2018] [Accepted: 05/28/2018] [Indexed: 10/14/2022]
Abstract
OBJECTIVES Attachment of magnetic particles to cells is needed for a variety of applications but is not always possible or efficient. Simpler and more convenient methods are thus desirable. In this study, we tested the hypothesis that endothelial cells (EC) can be loaded with micron-size magnetic beads by the phagocytosis-like mechanism 'angiophagy'. To this end, human umbilical vein EC (HUVEC) were incubated with magnetic beads conjugated or not (control) with an anti-VEGF receptor 2 antibody, either in suspension, or in culture followed by re-suspension using trypsinization. RESULTS In all conditions tested, HUVEC incubation with beads induced their uptake by angiophagy, which was confirmed by (i) increased cell granularity assessed by flow cytometry, and (ii) the presence of an F-actin rich layer around many of the intracellular beads, visualized by confocal microscopy. For confluent cultures, the average number of beads per cell was 4.4 and 4.2, with and without the presence of the anti-VEGFR2 antibody, respectively. However, while the actively dividing cells took up 2.9 unconjugated beads on average, this number increased to 5.2 if binding was mediated by the antibody. Magnetic pulldown increased the cell density of beads-loaded cells in porous electrospun poly-capro-lactone scaffolds by a factor of 4.5 after 5 min, as compared to gravitational settling (p < 0.0001). CONCLUSION We demonstrated that EC can be readily loaded by angiophagy with micron-sized beads while attached in monolayer culture, then dispersed in single-cell suspensions for pulldown in porous scaffolds and for other applications.
Collapse
|
16
|
The Multifaceted Uses and Therapeutic Advantages of Nanoparticles for Atherosclerosis Research. MATERIALS 2018; 11:ma11050754. [PMID: 29738480 PMCID: PMC5978131 DOI: 10.3390/ma11050754] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 04/29/2018] [Accepted: 04/30/2018] [Indexed: 12/27/2022]
Abstract
Nanoparticles are uniquely suited for the study and development of potential therapies against atherosclerosis by virtue of their size, fine-tunable properties, and ability to incorporate therapies and/or imaging modalities. Furthermore, nanoparticles can be specifically targeted to the atherosclerotic plaque, evading off-target effects and/or associated cytotoxicity. There has been a wealth of knowledge available concerning the use of nanotechnologies in cardiovascular disease and atherosclerosis, in particular in animal models, but with a major focus on imaging agents. In fact, roughly 60% of articles from an initial search for this review included examples of imaging applications of nanoparticles. Thus, this review focuses on experimental therapy interventions applied to and observed in animal models. Particular emphasis is placed on how nanoparticle materials and properties allow researchers to learn a great deal about atherosclerosis. The objective of this review was to provide an update for nanoparticle use in imaging and drug delivery studies and to illustrate how nanoparticles can be used for sensing and modelling, for studying fundamental biological mechanisms, and for the delivery of biotherapeutics such as proteins, peptides, nucleic acids, and even cells all with the goal of attenuating atherosclerosis. Furthermore, the various atherosclerosis processes targeted mainly for imaging studies have been summarized in the hopes of inspiring new and exciting targeted therapeutic and/or imaging strategies.
Collapse
|
17
|
Nasr SH, Tonson A, El-Dakdouki MH, Zhu DC, Agnew D, Wiseman R, Qian C, Huang X. Effects of Nanoprobe Morphology on Cellular Binding and Inflammatory Responses: Hyaluronan-Conjugated Magnetic Nanoworms for Magnetic Resonance Imaging of Atherosclerotic Plaques. ACS APPLIED MATERIALS & INTERFACES 2018; 10:11495-11507. [PMID: 29558108 PMCID: PMC5995107 DOI: 10.1021/acsami.7b19708] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Atherosclerosis is an inflammatory disease of arterial walls and the rupturing of atherosclerotic plaques is a major cause of heart attack and stroke. Imaging techniques that can enable the detection of atherosclerotic plaques before clinical manifestation are urgently needed. Magnetic resonance imaging (MRI) is a powerful technique to image the morphology of atherosclerotic plaques. In order to better analyze molecular processes in plaques, contrast agents that can selectively bind to plaque receptors will prove invaluable. CD44 is a cell surface protein overexpressed in plaque tissues, the level of which can be correlated with the risks of plaque rupture. Thus, targeting CD44 is an attractive strategy for detection of atherosclerotic plaques. Herein, we report the synthesis of hyaluronan-conjugated iron oxide nanoworms (HA-NWs). A new purification and gel electrophoresis protocol was developed to ensure the complete removal of free HA from HA-NWs. Compared to the more traditional spherical HA-bearing nanoparticles, HA-NWs had an elongated shape, which interacted much stronger with CD44-expressing cells in CD44- and HA-dependent manners. Furthermore, the HA-NWs did not induce much inflammatory response compared to the spherical HA nanoparticles. When assessed in vivo, HA-NWs enabled successful imaging of atherosclerotic plaques in a clinically relevant model of ApoE knockout transgenic mice for noninvasive plaque detection by MRI. Thus, nanoprobe shape engineering can be a useful strategy to significantly enhance their desired biological properties.
Collapse
Affiliation(s)
| | - Anne Tonson
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Mohammad H. El-Dakdouki
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Chemistry, Beirut Arab University, P.O. Box 11-5020, Riad El Solh 11072809, Beirut, Lebanon
| | - David C. Zhu
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Dalen Agnew
- Department of Pathobiology and Diagnostic Investigation, Michigan State University, East Lansing, Michigan 48824, United States
| | - Robert Wiseman
- Department of Physiology, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Chunqi Qian
- Department of Radiology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Xuefei Huang
- Department of Chemistry, Michigan State University, East Lansing, Michigan 48824, United States
- Department of Biomedical Engineering, Michigan State University, East Lansing, Michigan 48824, United States
- Institute for Quantitative Health Science and Engineering, Michigan State University, East Lansing, Michigan 48824, United States
| |
Collapse
|
18
|
Abdollah MRA, Carter TJ, Jones C, Kalber TL, Rajkumar V, Tolner B, Gruettner C, Zaw-Thin M, Baguña Torres J, Ellis M, Robson M, Pedley RB, Mulholland P, T M de Rosales R, Chester KA. Fucoidan Prolongs the Circulation Time of Dextran-Coated Iron Oxide Nanoparticles. ACS NANO 2018; 12:1156-1169. [PMID: 29341587 DOI: 10.1021/acsnano.7b06734] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/11/2023]
Abstract
The magnetic properties and safety of dextran-coated superparamagnetic iron oxide nanoparticles (SPIONs) have facilitated their clinical use as MRI contrast agents and stimulated research on applications for SPIONs in particle imaging and magnetic hyperthermia. The wider clinical potential of SPIONs, however, has been limited by their rapid removal from circulation via the reticuloendothelial system (RES). We explored the possibility of extending SPION circulatory time using fucoidan, a seaweed-derived food supplement, to inhibit RES uptake. The effects of fucoidan on SPION biodistribution were evaluated using ferucarbotran, which in its pharmaceutical formulation (Resovist) targets the RES. Ferucarbotran was radiolabeled at the iron oxide core with technetium-99m (99mTc; t1/2 = 6 h) or zirconium-89 (89Zr; t1/2 = 3.3 days). Results obtained with 99mTc-ferucarbotran demonstrated that administration of fucoidan led to a 4-fold increase in the circulatory half-life (t1/2 slow) from 37.4 to 150 min (n = 4; P < 0.0001). To investigate whether a longer circulatory half-life could lead to concomitant increased tumor uptake, the effects of fucoidan were tested with 89Zr-ferucarbotran in mice bearing syngeneic subcutaneous (GL261) tumors. In this model, the longer circulatory half-life achieved with fucoidan was associated with a doubling in tumor SPION uptake (n = 5; P < 0.001). Fucoidan was also effective in significantly increasing the circulatory half-life of perimag-COOH, a commercially available SPION with a larger hydrodynamic size (130 nm) than ferucarbotran (65 nm). These findings indicate successful diversion of SPIONs away from the hepatic RES and show realistic potential for future clinical applications.
Collapse
Affiliation(s)
- Maha R A Abdollah
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
- Department of Pharmacology and Biochemistry, Faculty of Pharmacy, The British University in Egypt (BUE) , El Shorouk City, Misr- Ismalia Desert Road, Cairo 11837, Egypt
| | - Thomas J Carter
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Clare Jones
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Tammy L Kalber
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London , London WC1E 6DD, U.K
| | - Vineeth Rajkumar
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Berend Tolner
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Cordula Gruettner
- Micromod Partikeltechnologie GmbH , Friedrich-Barnewitz-Str. 4, D-18119 Rostock, Germany
| | - May Zaw-Thin
- Centre for Advanced Biomedical Imaging, Division of Medicine and Institute of Child Health, University College London , London WC1E 6DD, U.K
| | - Julia Baguña Torres
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Matthew Ellis
- Division of Neuropathology, Department of Neurodegenerative Disease, UCL Institute of Neurology (ION), University College London (UCL) , Queen Square, London WC1N 3BG, U.K
| | - Mathew Robson
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - R Barbara Pedley
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Paul Mulholland
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| | - Rafael T M de Rosales
- School of Biomedical Engineering & Imaging Sciences, King's College London (KCL) , St Thomas' Hospital, London SE1 7EH, U.K
| | - Kerry Ann Chester
- UCL Cancer Institute, University College London (UCL) , Paul O'Gorman Building, 72 Huntley Street, London WC1E 6JD, U.K
| |
Collapse
|
19
|
Ha Y, Ko S, Kim I, Huang Y, Mohanty K, Huh C, Maynard JA. Recent Advances Incorporating Superparamagnetic Nanoparticles into Immunoassays. ACS APPLIED NANO MATERIALS 2018; 1:512-521. [PMID: 29911680 PMCID: PMC5999228 DOI: 10.1021/acsanm.7b00025] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Accepted: 01/31/2018] [Indexed: 05/09/2023]
Abstract
Superparamagnetic nanoparticles (SPMNPs) have attracted interest for various biomedical applications due to their unique magnetic behavior, excellent biocompatibility, easy surface modification, and low cost. Their unique magnetic properties, superparamagnetism, and magnetophoretic mobility have led to their inclusion in immunoassays to enhance biosensor sensitivity and allow for rapid detection of various analytes. In this review, we describe SPMNP characteristics valuable for incorporation into biosensors, including the use of SPMNPs to increase detection capabilities of surface plasmon resonance and giant magneto-resistive biosensors. The current status of SPMNP-based immunoassays to improve the sensitivity of rapid diagnostic tests is reviewed, and suggested strategies for the successful adoption of SPMNPs for immunoassays are presented.
Collapse
Affiliation(s)
- Yeonjeong Ha
- Department
of Chemical Engineering and Petroleum and Geosystems Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- E-mail: . (J.A.M.)
| | - Saebom Ko
- Department
of Chemical Engineering and Petroleum and Geosystems Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Ijung Kim
- Department
of Civil and Environmental Engineering, Western New England University, Springfield, Massachusetts 01119, United States
| | - Yimin Huang
- Department
of Chemical Engineering and Petroleum and Geosystems Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Kishore Mohanty
- Department
of Chemical Engineering and Petroleum and Geosystems Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Chun Huh
- Department
of Chemical Engineering and Petroleum and Geosystems Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
| | - Jennifer A. Maynard
- Department
of Chemical Engineering and Petroleum and Geosystems Engineering, The University of Texas at Austin, Austin, Texas 78712, United States
- E-mail: . (Y.-J.H.)
| |
Collapse
|
20
|
Zhou C, Wu H, Wang M, Huang C, Yang D, Jia N. Functionalized graphene oxide/Fe 3O 4 hybrids for cellular magnetic resonance imaging and fluorescence labeling. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2017; 78:817-825. [PMID: 28576054 DOI: 10.1016/j.msec.2017.04.139] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2016] [Revised: 04/10/2017] [Accepted: 04/13/2017] [Indexed: 12/24/2022]
Abstract
In this work, we developed a T2-weighted contrast agent based on graphene oxide (GO)/Fe3O4 hybrids for efficient cellular magnetic resonance imaging (MRI). The GO/Fe3O4 hybrids were obtained by combining with co-precipitation method and pyrolysis method. The structural, surface and magnetic characteristics of the hybrids were systematically characterized by transmission electron microscopy (TEM), vibrating sample magnetometer (VSM), AFM, Raman, FT-IR and XRD. The GO/Fe3O4 hybrids were functionalized by modifying with anionic and cationic polyelectrolyte through layer-by-layer assembling. The fluorescence probe fluorescein isothiocyanate (FITC) was further loaded on the surface of functionalized GO/Fe3O4 hybrids to trace the location of GO/Fe3O4 hybrids in cells. Functionalized GO/Fe3O4 hybrids possess good hydrophilicity, less cytotoxicity, high MRI enhancement with the relaxivity (r2) of 493mM-1s-1 as well as cellular MRI contrast effect. These obtained results indicated that the functionalized GO/Fe3O4 hybrids could have great potential to be utilized as cellular MRI contrast agents for tumor early diagnosis and monitoring.
Collapse
Affiliation(s)
- Chaohui Zhou
- The Education Ministry Key Laboratory of Resource Chemistry, Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Department of Chemistry, Shanghai Normal University, Shanghai 200234, China
| | - Hui Wu
- The Education Ministry Key Laboratory of Resource Chemistry, Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Department of Chemistry, Shanghai Normal University, Shanghai 200234, China
| | - Mingliang Wang
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China.
| | - Chusen Huang
- The Education Ministry Key Laboratory of Resource Chemistry, Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Department of Chemistry, Shanghai Normal University, Shanghai 200234, China
| | - Dapeng Yang
- College of Chemical Engineering and Materials Science, Quanzhou Normal University, Quanzhou 362000, Fujian Province, China.
| | - Nengqin Jia
- The Education Ministry Key Laboratory of Resource Chemistry, Shanghai Key Laboratory of Rare Earth Functional Materials and Shanghai Municipal Education Committee Key Laboratory of Molecular Imaging Probes and Sensors, Department of Chemistry, Shanghai Normal University, Shanghai 200234, China; College of Chemical Engineering and Materials Science, Quanzhou Normal University, Quanzhou 362000, Fujian Province, China.
| |
Collapse
|
21
|
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
|
22
|
Abstract
In vivo imaging, which enables us to peer deeply within living subjects, is producing tremendous opportunities both for clinical diagnostics and as a research tool. Contrast material is often required to clearly visualize the functional architecture of physiological structures. Recent advances in nanomaterials are becoming pivotal to generate the high-resolution, high-contrast images needed for accurate, precision diagnostics. Nanomaterials are playing major roles in imaging by delivering large imaging payloads, yielding improved sensitivity, multiplexing capacity, and modularity of design. Indeed, for several imaging modalities, nanomaterials are now not simply ancillary contrast entities, but are instead the original and sole source of image signal that make possible the modality's existence. We address the physicochemical makeup/design of nanomaterials through the lens of the physical properties that produce contrast signal for the cognate imaging modality-we stratify nanomaterials on the basis of their (i) magnetic, (ii) optical, (iii) acoustic, and/or (iv) nuclear properties. We evaluate them for their ability to provide relevant information under preclinical and clinical circumstances, their in vivo safety profiles (which are being incorporated into their chemical design), their modularity in being fused to create multimodal nanomaterials (spanning multiple different physical imaging modalities and therapeutic/theranostic capabilities), their key properties, and critically their likelihood to be clinically translated.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- Stanford University , 3155 Porter Drive, #1214, Palo Alto, California 94304-5483, United States
| | - Sanjiv Sam Gambhir
- The James H. Clark Center , 318 Campus Drive, First Floor, E-150A, Stanford, California 94305-5427, United States
| |
Collapse
|
23
|
Dieckhoff J, Kaul MG, Mummert T, Jung C, Salamon J, Adam G, Knopp T, Ludwig F, Balceris C, Ittrich H. In vivo liver visualizations with magnetic particle imaging based on the calibration measurement approach. Phys Med Biol 2016; 62:3470-3482. [PMID: 28035904 DOI: 10.1088/1361-6560/aa562d] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Magnetic particle imaging (MPI) facilitates the rapid determination of 3D in vivo magnetic nanoparticle distributions. In this work, liver MPI following intravenous injections of ferucarbotran (Resovist®) was studied. The image reconstruction was based on a calibration measurement, the so called system function. The application of an enhanced system function sample reflecting the particle mobility and aggregation status of ferucarbotran resulted in significantly improved image reconstructions. The finding was supported by characterizations of different ferucarbotran compositions with the magnetorelaxometry and magnetic particle spectroscopy technique. For instance, similar results were obtained between ferucarbotran embedded in freeze-dried mannitol sugar and liver tissue harvested after a ferucarbotran injection. In addition, the combination of multiple shifted measurement patches for a joint reconstruction of the MPI data enlarged the field of view and increased the covering of liver MPI on magnetic resonance images noticeably.
Collapse
Affiliation(s)
- J Dieckhoff
- Department for Diagnostic and Interventional Radiology and Nuclear Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
24
|
Oil/water nano-emulsion loaded with cobalt ferrite oxide nanocubes for photo-acoustic and magnetic resonance dual imaging in cancer: in vitro and preclinical studies. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2016; 13:275-286. [PMID: 27565688 DOI: 10.1016/j.nano.2016.08.022] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Revised: 07/12/2016] [Accepted: 08/12/2016] [Indexed: 01/31/2023]
Abstract
Dual imaging dramatically improves detection and early diagnosis of cancer. In this work we present an oil in water (O/W) nano-emulsion stabilized with lecithin and loaded with cobalt ferrite oxide (Co0.5Fe2.5O4) nanocubes for photo-acoustic and magnetic resonance dual imaging. The nanocarrier is responsive in in vitro photo-acoustic and magnetic resonance imaging (MRI) tests. A clear and significant time-dependent accumulation in tumor tissue is shown in in vivo photo-acoustic studies on a murine melanoma xenograft model. The proposed O/W nano-emulsion exhibits also high values of r2/r1 (ranging from 45 to 85, depending on the magnetic field) suggesting a possible use as T2 weighted image contrast agents. In addition, viability and cellular uptake studies show no significant cytotoxicity on the fibroblast cell line. We also tested the O/W nano-emulsion loaded with curcumin against melanoma cancer cells demonstrating a significant cytotoxicity and thus showing possible therapeutic effects in addition to the in vivo imaging.
Collapse
|
25
|
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
|
26
|
Value of Functionalized Superparamagnetic Iron Oxide Nanoparticles in the Diagnosis and Treatment of Acute Temporal Lobe Epilepsy on MRI. Neural Plast 2016; 2016:2412958. [PMID: 26925269 PMCID: PMC4748095 DOI: 10.1155/2016/2412958] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2015] [Revised: 12/27/2015] [Accepted: 01/03/2016] [Indexed: 12/18/2022] Open
Abstract
Purpose. Although active targeting of drugs using a magnetic-targeted drug delivery system (MTDS) with superparamagnetic iron oxide nanoparticles (SPIONs) is a very effective treatment approach for tumors and other illnesses, successful results of drug-resistant temporal lobe epilepsy (TLE) are unprecedented. A hallmark in the neuropathology of TLE is brain inflammation, in particular the activation of interleukin-1β (IL-1β) induced by activated glial cells, which has been considered a new mechanistic target for treatment. The purpose of this study was to determine the feasibility of the functionalized SPIONs with anti-IL-1β monoclonal antibody (mAb) attached to render MRI diagnoses and simultaneously provide targeted therapy with the neutralization of IL-1β overexpressed in epileptogenic zone of an acute rat model of TLE. Experimental Design. The anti-IL-1β mAb-SPIONs were studied in vivo versus plain SPIONs and saline. Lithium-chloride pilocarpine-induced TLE models (n = 60) were followed by Western blot, Perl's iron staining, Nissl staining, and immunofluorescent double-label staining after MRI examination. Results. The magnetic anti-IL-1β mAb-SPION administered intravenously, which crossed the BBB and was concentrated in the astrocytes and neurons in epileptogenic tissues, rendered these tissues visible on MRI and simultaneously delivered anti-IL-1β mAb to the epileptogenic focus. Conclusions. Our study provides the first evidence that the novel approach enhanced accumulation and the therapeutic effect of anti-IL-1β mAb by MTDS using SPIONs.
Collapse
|
27
|
Nörenberg D, Ebersberger HU, Diederichs G, Hamm B, Botnar RM, Makowski MR. Molecular magnetic resonance imaging of atherosclerotic vessel wall disease. Eur Radiol 2015; 26:910-20. [DOI: 10.1007/s00330-015-3881-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/27/2015] [Accepted: 06/08/2015] [Indexed: 11/29/2022]
|
28
|
Lavin B, Phinikaridou A, Henningsson M, Botnar RM. Current Development of Molecular Coronary Plaque Imaging using Magnetic Resonance Imaging towards Clinical Application. CURRENT CARDIOVASCULAR IMAGING REPORTS 2014. [DOI: 10.1007/s12410-014-9309-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|
29
|
Fernández-Friera L, Ibáñez B, Fuster V. Imaging subclinical atherosclerosis: is it ready for prime time? A review. J Cardiovasc Transl Res 2014; 7:623-34. [PMID: 25119855 DOI: 10.1007/s12265-014-9582-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 07/28/2014] [Indexed: 01/02/2023]
Abstract
Imaging subclinical atherosclerosis holds the promise of individualized cardiovascular (CV) risk assessment. The large arsenal of noninvasive imaging techniques available today is playing an increasingly important role in the diagnosis and monitoring of subclinical atherosclerosis. However, there is a debate about the advisability of clinical screens for subclinical atherosclerosis and which modality is the most appropriate for monitoring risk and atherosclerosis progression. This article offers an overview of the traditional and emerging noninvasive imaging modalities used to detect early atherosclerosis, surveys population studies addressing the value of subclinical atherosclerosis detection, and also examines guideline recommendations for their clinical implementation. The clinical relevance of this manuscript lies in the potential of current imaging technology to improve CV risk prediction based on traditional risk factors and the present recommendations for subclinical atherosclerosis assessment. Noninvasive imaging will also help to identify individuals at high CV who would benefit from intensive prevention or therapeutic interventions.
Collapse
|
30
|
Herranz F, Salinas B, Groult H, Pellico J, Lechuga-Vieco AV, Bhavesh R, Ruiz-Cabello J. Superparamagnetic Nanoparticles for Atherosclerosis Imaging. NANOMATERIALS (BASEL, SWITZERLAND) 2014; 4:408-438. [PMID: 28344230 PMCID: PMC5304673 DOI: 10.3390/nano4020408] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/25/2014] [Revised: 05/15/2014] [Accepted: 05/16/2014] [Indexed: 12/12/2022]
Abstract
The production of magnetic nanoparticles of utmost quality for biomedical imaging requires several steps, from the synthesis of highly crystalline magnetic cores to the attachment of the different molecules on the surface. This last step probably plays the key role in the production of clinically useful nanomaterials. The attachment of the different biomolecules should be performed in a defined and controlled fashion, avoiding the random adsorption of the components that could lead to undesirable byproducts and ill-characterized surface composition. In this work, we review the process of creating new magnetic nanomaterials for imaging, particularly for the detection of atherosclerotic plaque, in vivo. Our focus will be in the different biofunctionalization techniques that we and several other groups have recently developed. Magnetic nanomaterial functionalization should be performed by chemoselective techniques. This approach will facilitate the application of these nanomaterials in the clinic, not as an exception, but as any other pharmacological compound.
Collapse
Affiliation(s)
- Fernando Herranz
- Advanced Imaging Unit, Department of Epidemiology, Atherothrombosis and Imaging, Spanish National Centre for Cardiovascular Research (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain.
- CIBER of Pulmonary Diseases, Biomedical Research Network, Carlos III Health Institute, 28029 Madrid, Spain.
| | - Beatriz Salinas
- Advanced Imaging Unit, Department of Epidemiology, Atherothrombosis and Imaging, Spanish National Centre for Cardiovascular Research (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain.
- CIBER of Pulmonary Diseases, Biomedical Research Network, Carlos III Health Institute, 28029 Madrid, Spain.
| | - Hugo Groult
- Advanced Imaging Unit, Department of Epidemiology, Atherothrombosis and Imaging, Spanish National Centre for Cardiovascular Research (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain.
- CIBER of Pulmonary Diseases, Biomedical Research Network, Carlos III Health Institute, 28029 Madrid, Spain.
| | - Juan Pellico
- Advanced Imaging Unit, Department of Epidemiology, Atherothrombosis and Imaging, Spanish National Centre for Cardiovascular Research (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain.
- CIBER of Pulmonary Diseases, Biomedical Research Network, Carlos III Health Institute, 28029 Madrid, Spain.
| | - Ana V Lechuga-Vieco
- Advanced Imaging Unit, Department of Epidemiology, Atherothrombosis and Imaging, Spanish National Centre for Cardiovascular Research (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain.
- CIBER of Pulmonary Diseases, Biomedical Research Network, Carlos III Health Institute, 28029 Madrid, Spain.
| | - Riju Bhavesh
- Advanced Imaging Unit, Department of Epidemiology, Atherothrombosis and Imaging, Spanish National Centre for Cardiovascular Research (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain.
| | - J Ruiz-Cabello
- Advanced Imaging Unit, Department of Epidemiology, Atherothrombosis and Imaging, Spanish National Centre for Cardiovascular Research (CNIC), Melchor Fernández Almagro, 3, 28029 Madrid, Spain.
- CIBER of Pulmonary Diseases, Biomedical Research Network, Carlos III Health Institute, 28029 Madrid, Spain.
- Department of Physicochemistry II, Faculty of Pharmacy, Complutense University Madrid (UCM), Plaza Ramón y Cajal s/n Ciudad Universitaria, 28040 Madrid, Spain.
| |
Collapse
|
31
|
Smith BR, Ghosn EEB, Rallapalli H, Prescher JA, Larson T, Herzenberg LA, Gambhir SS. Selective uptake of single-walled carbon nanotubes by circulating monocytes for enhanced tumour delivery. NATURE NANOTECHNOLOGY 2014; 9:481-7. [PMID: 24727688 PMCID: PMC4236538 DOI: 10.1038/nnano.2014.62] [Citation(s) in RCA: 179] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2013] [Accepted: 02/26/2014] [Indexed: 05/05/2023]
Abstract
In cancer imaging, nanoparticle biodistribution is typically visualized in living subjects using 'bulk' imaging modalities such as magnetic resonance imaging, computerized tomography and whole-body fluorescence. Accordingly, nanoparticle influx is observed only macroscopically, and the mechanisms by which they target cancer remain elusive. Nanoparticles are assumed to accumulate via several targeting mechanisms, particularly extravasation (leakage into tumour). Here, we show that, in addition to conventional nanoparticle-uptake mechanisms, single-walled carbon nanotubes are almost exclusively taken up by a single immune cell subset, Ly-6C(hi) monocytes (almost 100% uptake in Ly-6C(hi) monocytes, below 3% in all other circulating cells), and delivered to the tumour in mice. We also demonstrate that a targeting ligand (RGD) conjugated to nanotubes significantly enhances the number of single-walled carbon nanotube-loaded monocytes reaching the tumour (P < 0.001, day 7 post-injection). The remarkable selectivity of this tumour-targeting mechanism demonstrates an advanced immune-based delivery strategy for enhancing specific tumour delivery with substantial penetration.
Collapse
Affiliation(s)
- Bryan Ronain Smith
- 1] Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA [2] Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Eliver Eid Bou Ghosn
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Harikrishna Rallapalli
- Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA
| | - Jennifer A Prescher
- 1] Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA [2]
| | - Timothy Larson
- 1] Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA [2] Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Leonore A Herzenberg
- Department of Genetics, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Sanjiv Sam Gambhir
- 1] Molecular Imaging Program at Stanford, The James H Clark Center, Stanford University, Stanford, California 94305, USA [2] Department of Radiology, Stanford University School of Medicine, Stanford, California 94305, USA [3] Department of Bioengineering and Department of Materials Science & Engineering, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
32
|
Modeling and experiments of magneto-nanosensors for diagnostics of radiation exposure and cancer. Biomed Microdevices 2014; 15:665-671. [PMID: 22763391 DOI: 10.1007/s10544-012-9678-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
We present a resistive network model, protein assay data, and outlook of the giant magnetoresistive (GMR) spin-valve magneto-nanosensor platform ideal for multiplexed detection of protein biomarkers in solutions. The magneto-nanosensors are designed to have optimal performance considering several factors such as sensor dimension, shape anisotropy, and magnetic nanoparticle tags. The resistive network model indicates that thinner spin-valve sensors with narrower width lead to higher signals from magnetic nanoparticle tags. Standard curves and real-time measurements showed a sensitivity of ~10 pM for phosphorylated-structural maintenance of chromosome 1 (phosphor-SMC1), ~53 fM for granulocyte colony stimulation factor (GCSF), and ~460 fM for interleukin-6 (IL6), which are among the representative biomarkers for radiation exposure and cancer.
Collapse
|
33
|
Zhang Y, Bai Y, Jia J, Gao N, Li Y, Zhang R, Jiang G, Yan B. Perturbation of physiological systems by nanoparticles. Chem Soc Rev 2014; 43:3762-809. [PMID: 24647382 DOI: 10.1039/c3cs60338e] [Citation(s) in RCA: 91] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Nanotechnology is having a tremendous impact on our society. However, societal concerns about human safety under nanoparticle exposure may derail the broad application of this promising technology. Nanoparticles may enter the human body via various routes, including respiratory pathways, the digestive tract, skin contact, intravenous injection, and implantation. After absorption, nanoparticles are carried to distal organs by the bloodstream and the lymphatic system. During this process, they interact with biological molecules and perturb physiological systems. Although some ingested or absorbed nanoparticles are eliminated, others remain in the body for a long time. The human body is composed of multiple systems that work together to maintain physiological homeostasis. The unexpected invasion of these systems by nanoparticles disturbs normal cell signaling, impairs cell and organ functions, and may even cause pathological disorders. This review examines the comprehensive health risks of exposure to nanoparticles by discussing how nanoparticles perturb various physiological systems as revealed by animal studies. The potential toxicity of nanoparticles to each physiological system and the implications of disrupting the balance among systems are emphasized.
Collapse
Affiliation(s)
- Yi Zhang
- Key Laboratory for Colloid and Interface Chemistry, Ministry of Education, School of Chemistry and Chemical Engineering, Shandong University, Jinan 250100, China.
| | | | | | | | | | | | | | | |
Collapse
|
34
|
Cicha I, Garlichs CD, Alexiou C. Cardiovascular therapy through nanotechnology – how far are we still from bedside? EUROPEAN JOURNAL OF NANOMEDICINE 2014. [DOI: 10.1515/ejnm-2014-0001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
AbstractRecent years brought about a widespread interest in the potential applications of nanotechnology for the diagnostics and the therapy of human diseases. With its promise of disease-targeted, patient-tailored treatment and reduced side effects, nanomedicine brings hope for millions of patients suffering of non-communicable diseases such as cancer or cardiovascular disorders. However, the emergence of the complex, multicomponent products based on new technologies poses multiple challenges to successful approval in clinical practice. Regulatory and development considerations, including properties of the components, reproducible manufacturing and appropriate characterization methods, as well as nanodrugs’ safety and efficacy are critical for rapid marketing of the new products. This review discusses the recent advances in cardiovascular applications of nanotechnologies and highlights the challenges that must be overcome in order to fill the gap existing between the promising bench trials and the successful bedside applications.
Collapse
|
35
|
Superparamagnetic iron oxide based nanoprobes for imaging and theranostics. Adv Colloid Interface Sci 2013; 199-200:95-113. [PMID: 23891347 DOI: 10.1016/j.cis.2013.06.007] [Citation(s) in RCA: 61] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2013] [Revised: 06/21/2013] [Accepted: 06/27/2013] [Indexed: 12/11/2022]
Abstract
The need to target, deliver and subsequently evaluate the efficacy of therapeutics in the treatment of a disease has provided added impetus in developing novel and highly efficient contrast agents. Superparamagnetic iron oxide nanoparticles (SPIONs) have offered tremendous potential in designing advanced magnetic resonance imaging (MRI) diagnostic agents, due to their unique physicochemical properties. There has been tremendous effort devoted in the recent past in developing synthetic methodologies through which their size, hydrodynamic radii, chemical composition and morphologies could be tailored at the nanoscale. This enables one to fine tune their magnetic behavior, and thus their MRI response. While novel synthetic strategies are being assembled for directing SPIONs to the diseased site as well as imparting them stealth and biocompatibility, it is also essential to evaluate their biological toxicological profiles. This review highlights recent advances that have been made in the synthesis of SPIONs, subsequent functionalization with desired entities, and a discussion on their use as MRI contrast agents in cardiovascular research.
Collapse
|
36
|
CD44 targeting magnetic glyconanoparticles for atherosclerotic plaque imaging. Pharm Res 2013; 31:1426-37. [PMID: 23568520 DOI: 10.1007/s11095-013-1021-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2012] [Accepted: 03/04/2013] [Indexed: 12/21/2022]
Abstract
PURPOSE The cell surface adhesion molecule CD44 plays important roles in the initiation and development of atherosclerotic plaques. We aim to develop nanoparticles that can selectively target CD44 for the non-invasive detection of atherosclerotic plaques by magnetic resonance imaging. METHODS Magnetic glyconanoparticles with hyaluronan immobilized on the surface have been prepared. The binding of these nanoparticles with CD44 was evaluated in vitro by enzyme linked immunosorbent assay, flow cytometry and confocal microscopy. In vivo magnetic resonance imaging of plaques was performed on an atherosclerotic rabbit model. RESULTS The magnetic glyconanoparticles can selectively bind CD44. In T2* weighted magnetic resonance images acquired in vivo, significant contrast changes in aorta walls were observed with a very low dose of the magnetic nanoparticles, allowing the detection of atherosclerotic plaques. Furthermore, imaging could be performed without significant delay after probe administration. The selectivity of hyaluronan nanoparticles in plaque imaging was established by several control experiments. CONCLUSIONS Magnetic nanoparticles bearing surface hyaluronan enabled the imaging of atherosclerotic plaques in vivo by magnetic resonance imaging. The low dose of nanoparticles required, the possibility to image without much delay and the high biocompatibility are the advantages of these nanoparticles as contrast agents for plaque imaging.
Collapse
|
37
|
Phinikaridou A, Andia ME, Shah AM, Botnar RM. Advances in molecular imaging of atherosclerosis and myocardial infarction: shedding new light on in vivo cardiovascular biology. Am J Physiol Heart Circ Physiol 2012; 303:H1397-410. [PMID: 23064836 DOI: 10.1152/ajpheart.00583.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Molecular imaging of the cardiovascular system heavily relies on the development of new imaging probes and technologies to facilitate visualization of biological processes underlying or preceding disease. Molecular imaging is a highly active research discipline that has seen tremendous growth over the past decade. It has broadened our understanding of oncologic, neurologic, and cardiovascular diseases by providing new insights into the in vivo biology of disease progression and therapeutic interventions. As it allows for the longitudinal evaluation of biological processes, it is ideally suited for monitoring treatment response. In this review, we will concentrate on the major accomplishments and advances in the field of molecular imaging of atherosclerosis and myocardial infarction with a special focus on magnetic resonance imaging.
Collapse
Affiliation(s)
- Alkystis Phinikaridou
- Division of Imaging Science and Biomedical Engineering, King's College London, United Kingdom.
| | | | | | | |
Collapse
|
38
|
Kanwar RK, Chaudhary R, Tsuzuki T, Kanwar JR. Emerging engineered magnetic nanoparticulate probes for molecular MRI of atherosclerosis: how far have we come? Nanomedicine (Lond) 2012; 7:899-916. [DOI: 10.2217/nnm.12.57] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Atherosclerosis is a chronic, progressive, immunoinflammatory disease of the large and medium-sized arteries, and a major cause of cardiovascular diseases. Atherosclerosis often progresses silently for decades until the occurrence of a major catastrophic clinical event such as myocardial infarction, cardiac arrest and stroke. The main challenge in the diagnosis and management of atherosclerosis is to develop a safe, noninvasive technique that is accurate and reproducible, which can detect the biologically active high-risk vulnerable plaques (with ongoing active inflammation, angiogenesis and apoptosis) before the occurrence of an acute clinical event. This article reviews the events involved in the pathogenesis of atherosclerosis in light of recently advanced understanding of the molecular pathogenesis of the disease. Next, we elaborate on the interesting developments in molecular MRI, by describing the recently engineered magnetic nanoparticulate probes targeting clinically promising molecular and cellular players/processes, involved in early atherosclerotic lesion formation to plaque rupture and erosion.
Collapse
Affiliation(s)
- Rupinder K Kanwar
- Nanomedicine, Laboratory of Immunology & Molecular Biomedical Research (LIMBR), Center for Biotechnology & Interdisciplinary Biosciences, Institute for Frontier Materials (IFM), Deakin University, Waurn Ponds, Victoria 3217, Australia
| | - Rajneesh Chaudhary
- Nanomaterials, Institute for Frontier Materials (IFM), Deakin University, Waurn Ponds, Victoria 3217, Australia
| | - Takuya Tsuzuki
- Nanomaterials, Institute for Frontier Materials (IFM), Deakin University, Waurn Ponds, Victoria 3217, Australia
| | - Jagat R Kanwar
- Nanomedicine, Laboratory of Immunology & Molecular Biomedical Research (LIMBR), Center for Biotechnology & Interdisciplinary Biosciences, Institute for Frontier Materials (IFM), Deakin University, Waurn Ponds, Victoria 3217, Australia
| |
Collapse
|
39
|
Varna M, Ratajczak P, Ferreira I, Leboeuf C, Bousquet G, Janin A. <i>In vivo</i> Distribution of Inorganic Nanoparticles in Preclinical Models. ACTA ACUST UNITED AC 2012. [DOI: 10.4236/jbnb.2012.322033] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
40
|
Mudshinge SR, Deore AB, Patil S, Bhalgat CM. Nanoparticles: Emerging carriers for drug delivery. Saudi Pharm J 2011; 19:129-41. [PMID: 23960751 PMCID: PMC3744999 DOI: 10.1016/j.jsps.2011.04.001] [Citation(s) in RCA: 258] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2011] [Accepted: 04/12/2011] [Indexed: 01/16/2023] Open
Abstract
The core objective of nanoparticles is to control and manipulate biomacromolecular constructs and supramolecular assemblies that are critical to living cells in order to improve the quality of human health. By definition, these constructs and assemblies are nanoscale and include entities such as drugs, proteins, DNA/RNA, viruses, cellular lipid bilayers, cellular receptor sites and antibody variable regions critical for immunology and are involved in events of nanoscale proportions. The emergence of such nanotherapeutics/diagnostics will allow a deeper understanding of human longevity and human ills that include cancer, cardiovascular disease and genetic disorders. A technology platform that provides a wide range of synthetic nanostructures that may be controlled as a function of size, shape and surface chemistry and scale to these nanotechnical dimensions will be a critical first step in developing appropriate tools and a scientific basis for understanding nanoparticles.
Collapse
Affiliation(s)
| | - Amol B. Deore
- NDMVP’s Institute of Pharmaceutical Sciences, Adgaon, Nashik, Maharashtra, India
| | - Sachin Patil
- Mayani College of Pharmacy, Mayani, Satara Dist., Maharashtra, India
| | - Chetan M. Bhalgat
- S.A.C. College of Pharmacy, B.G. Nagara 571448, Nagamangala (Tq), Mandya Dist., Karnataka, India
| |
Collapse
|
41
|
Young VEL, Sadat U, Gillard JH. Noninvasive carotid artery imaging with a focus on the vulnerable plaque. Neuroimaging Clin N Am 2011; 21:391-405, xi-xii. [PMID: 21640306 DOI: 10.1016/j.nic.2011.01.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Currently carotid imaging has 2 main focuses: assessment of luminal stenosis and classification of atherosclerotic plaque characteristics. Measurement of the degree of stenosis is the main assessment used for current treatment decision making, but an evolving idea that is now driving imaging is the concept of vulnerable plaque, which is where plaque components are identified and used to define which plaques are at high risk of causing symptoms compared with those at low risk. This review article covers the methods used for noninvasive assessment of carotid luminal stenosis and the options available for plaque imaging.
Collapse
Affiliation(s)
- V E L Young
- University Department of Radiology, Addenbrookes Hospital, Box 218, Hills Road, Cambridge CB2 0QQ, UK.
| | | | | |
Collapse
|
42
|
Gupta AS. Nanomedicine approaches in vascular disease: a review. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2011; 7:763-79. [PMID: 21601009 DOI: 10.1016/j.nano.2011.04.001] [Citation(s) in RCA: 69] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2011] [Revised: 03/11/2011] [Accepted: 04/05/2011] [Indexed: 01/26/2023]
Abstract
UNLABELLED Nanomedicine approaches have revolutionized the treatment of cancer and vascular diseases, where the limitations of rapid nonspecific clearance, poor biodistribution and harmful side effects associated with direct systemic drug administration can be overcome by packaging the agents within sterically stabilized, long-circulating nanovehicles that can be further surface-modified with ligands to actively target cellular/molecular components of the disease. With significant advancements in genetics, proteomics, cellular and molecular biology and biomaterials engineering, the nanomedicine strategies have become progressively refined regarding the modulation of surface and bulk chemistry of the nanovehicles, control of drug release kinetics, manipulation of nanoconstruct geometry and integration of multiple functionalities on single nanoplatforms. The current review aims to capture the various nanomedicine approaches directed specifically toward vascular diseases during the past two decades. Analysis of the promises and limitations of these approaches will help identify and optimize vascular nanomedicine systems to enhance their efficacy and clinical translation in the future. FROM THE CLINICAL EDITOR Nanomedicine-based approaches have had a major impact on the treatment and diagnosis of malignancies and vascular diseases. This review discusses various nanomedicine approaches directed specifically toward vascular diseases during the past two decades, highlighting their advantages, limitations and offering new perspectives on future applications.
Collapse
Affiliation(s)
- Anirban Sen Gupta
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio 44106, USA.
| |
Collapse
|
43
|
Fitzgerald KT, Holladay CA, McCarthy C, Power KA, Pandit A, Gallagher WM. Standardization of models and methods used to assess nanoparticles in cardiovascular applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2011; 7:705-717. [PMID: 21319299 DOI: 10.1002/smll.201001347] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/05/2010] [Revised: 09/22/2010] [Indexed: 05/30/2023]
Abstract
Nanotechnology has the potential to revolutionize the management and treatment of cardiovascular disease. Controlled drug delivery and nanoparticle-based molecular imaging agents have advanced cardiovascular disease therapy and diagnosis. However, the delivery vehicles (dendrimers, nanocrystals, nanotubes, nanoparticles, nanoshells, etc.), as well as the model systems that are used to mimic human cardiac disease, should be questioned in relation to their suitability. This review focuses on the variations of the biological assays and preclinical models that are currently being used to study the biocompatibility and suitability of nanomaterials in cardiovascular applications. There is a need to standardize appropriate models and methods that will promote the development of novel nanomaterial-based cardiovascular therapies.
Collapse
Affiliation(s)
- Kathleen T Fitzgerald
- UCD School of Biomolecular and Biomedical Science, UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin 4, Ireland
| | | | | | | | | | | |
Collapse
|
44
|
Clinical applications in molecular imaging. Pediatr Radiol 2011; 41:199-207. [PMID: 21127854 DOI: 10.1007/s00247-010-1902-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/20/2010] [Revised: 09/21/2010] [Accepted: 10/10/2010] [Indexed: 10/18/2022]
Abstract
Molecular imaging is aimed at the noninvasive in vivo characterization and measurement of processes at a cellular and molecular level with clinical imaging methods. Contrast agents are constructed to target markers that are specific either for certain diseases or for functional states of specialized tissues. Efforts are currently focused mainly on processes involved in angiogenesis, inflammation, and apoptosis. Cell tracking is performed for diagnostic purposes as well as for monitoring of novel cell therapies. Visualization of these processes would provide more precise information about disease expansion as well as treatment response, and could lead to a more individualized therapy for patients. Many attempts have shown promising results in preclinical studies; however, translation into the clinic remains a challenge. This applies especially to paediatrics because of more stringent safety concerns and the low prevalence of individual diseases. The most promising modalities for clinical translation are nuclear medicine methods (positron emission tomography [PET] and single photon emission CT [SPECT]) due to their high sensitivity, which allows concentrations below biological activity. However, special dose consideration is required for any application of ionizing radiation especially in children. While very little has been published on molecular imaging in a paediatric patient population beyond fluorodeoxyglucose (FDG)-PET and metaiodobenzylguanidine (MIBG) tracers, this review will attempt to discuss approaches that we believe have promise for paediatric imaging. These will include agents that already reached clinical trials as well as preclinical developments with high potential for clinical application.
Collapse
|
45
|
van Tilborg GAF, Vucic E, Strijkers GJ, Cormode DP, Mani V, Skajaa T, Reutelingsperger CPM, Fayad ZA, Mulder WJM, Nicolay K. Annexin A5-functionalized bimodal nanoparticles for MRI and fluorescence imaging of atherosclerotic plaques. Bioconjug Chem 2011; 21:1794-803. [PMID: 20804153 DOI: 10.1021/bc100091q] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Apoptosis and macrophage burden are believed to correlate with atherosclerotic plaque vulnerability and are therefore considered important diagnostic and therapeutic targets for atherosclerosis. These cell types are characterized by the exposure of phosphatidylserine (PS) at their surface. In the present study, we developed and applied a small micellar fluorescent annexin A5-functionalized nanoparticle for noninvasive magnetic resonance imaging (MRI) of PS exposing cells in atherosclerotic lesions. Annexin A5-mediated target-specificity was confirmed with ellipsometry and in vitro binding to apoptotic Jurkat cells. In vivo T(1)-weighted MRI of the abdominal aorta in atherosclerotic ApoE(-/-) mice revealed enhanced uptake of the annexin A5-micelles as compared to control-micelles, which was corroborated with ex vivo near-infrared fluorescence images of excised whole aortas. Confocal laser scanning microscopy (CLSM) demonstrated that the targeted agent was associated with macrophages and apoptotic cells, whereas the nonspecific control agent showed no clear uptake by such cells. In conclusion, the annexin A5-conjugated bimodal micelles displayed potential for noninvasive assessment of cell types that are considered to significantly contribute to plaque instability and therefore may be of great value in the assessment of atherosclerotic lesion phenotype.
Collapse
Affiliation(s)
- Geralda A F van Tilborg
- Biomedical NMR, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
46
|
Zou J, Zhang W, Poe D, Qin J, Fornara A, Zhang Y, Ramadan UA, Muhammed M, Pyykkö I. MRI manifestation of novel superparamagnetic iron oxide nanoparticles in the rat inner ear. Nanomedicine (Lond) 2010; 5:739-54. [PMID: 20662645 DOI: 10.2217/nnm.10.45] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
AIM Superparamagnetic iron oxide nanoparticles hierarchically coated with oleic acid and Pluronic F127 copolymers (POA@SPION) have shown exceptional T2 contrast enhancement. The aim of the present work was to investigate the MRI manifestation of POA@SPION in the inner ear. MATERIALS & METHODS A total of 26 male Wister rats were selected for testing POA@SPION administered through intracochlear, intratympanic and intravenous routes. MRI was performed with a 4.7 T MR scanner. RESULTS & CONCLUSION POA@SPION can be introduced into the perilymph space, after which it becomes widely distributed and can demonstrate the integrity of the perilymph-endolymph barrier. Positive highlighting of the endolymph compartment against the darkened perilymph was visualized for the first time. POA@SPION passed through the middle-inner ear barriers in only small amounts, but stayed in the perilymph for 3 days. They did not traverse the blood-perilymph barrier or blood-endolymph barrier. The inner ear distribution of POA@SPION was confirmed by histology. POA@SPION is a promising T2 negative contrast agent.
Collapse
Affiliation(s)
- Jing Zou
- Department of Otolaryngology, University of Tampere, FM1, 3rd Floor, Biokatu 6, 33520 Tampere, Finland.
| | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Chen W, Cormode DP, Fayad ZA, Mulder WJM. Nanoparticles as magnetic resonance imaging contrast agents for vascular and cardiac diseases. WILEY INTERDISCIPLINARY REVIEWS-NANOMEDICINE AND NANOBIOTECHNOLOGY 2010; 3:146-161. [PMID: 20967875 DOI: 10.1002/wnan.114] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Advances in nanoparticle contrast agents for molecular imaging have made magnetic resonance imaging a promising modality for noninvasive visualization and assessment of vascular and cardiac disease processes. This review provides a description of the various nanoparticles exploited for imaging cardiovascular targets. Nanoparticle probes detecting inflammation, apoptosis, extracellular matrix, and angiogenesis may provide tools for assessing the risk of progressive vascular dysfunction and heart failure. The utility of nanoparticles as multimodal probes and/or theranostic agents has also been investigated. Although clinical application of these nanoparticles is largely unexplored, the potential for enhancing disease diagnosis and treatment is considerable.
Collapse
Affiliation(s)
- Wei Chen
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - David P Cormode
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, New York, NY, USA
| | - Zahi A Fayad
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, New York, NY, USA.,Department of Radiology, Mount Sinai School of Medicine, New York, NY, USA
| | - Willem J M Mulder
- Translational and Molecular Imaging Institute, Mount Sinai School of Medicine, New York, NY, USA.,Department of Gene and Cell Medicine, Mount Sinai School of Medicine, New York, NY, USA
| |
Collapse
|
48
|
Jayagopal A, Linton MF, Fazio S, Haselton FR. Insights into atherosclerosis using nanotechnology. Curr Atheroscler Rep 2010; 12:209-15. [PMID: 20425261 DOI: 10.1007/s11883-010-0106-7] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
A developing forefront in vascular disease research is the application of nanotechnology, the engineering of devices at the molecular scale, for diagnostic and therapeutic applications in atherosclerosis. Promising research in this field over the past decade has resulted in the preclinical validation of nanoscale devices that target cellular and molecular components of the atherosclerotic plaque, including one of its prominent cell types, the macrophage. Nanoscale contrast agents targeting constituents of plaque biology have been adapted for application in multiple imaging modalities, leading toward more detailed diagnostic readouts, whereas nanoscale drug delivery devices can be tailored for site-specific therapeutic activity. This review highlights recent progress in utilizing nanotechnology for the clinical management of atherosclerosis, drawing upon recent preclinical studies relevant to diagnosis and treatment of the plaque and promising future applications.
Collapse
Affiliation(s)
- Ashwath Jayagopal
- Department of Chemistry, Vanderbilt University, VU Station B Box 351822, Nashville, TN 37232, USA.
| | | | | | | |
Collapse
|
49
|
Abstract
Cardiovascular disease (CVD) is the leading cause of death worldwide, and its prevalence is likely to increase in the near future. The morbidity and mortality associated with CVD causes an enormous economic burden, which has become a major problem for many societies across the globe. The current prevention strategies are aimed at identifying and reducing established risk factors for atherosclerosis including hypertension, hypercholesterolemia, diabetes, obesity, smoking, and sedentary lifestyle. However, some of our prevention goals, such as reducing LDL cholesterol, change dramatically once a subject has been diagnosed with coronary atherosclerosis. At the present time, atherosclerosis is frequently diagnosed relatively late in the course of the disease, when a patient develops symptoms or presents with acute events such as an acute coronary syndrome or a stroke. Several studies have demonstrated that novel noninvasive imaging techniques have the potential to identify subclinical atherosclerosis and high-risk plaques. Early detection of subclinical atherosclerosis may enable clinicians to improve the control of cardiovascular risk factors in affected patients earlier, thereby helping to prevent some of the manifestations of CVD.
Collapse
Affiliation(s)
- Valentin Fuster
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain.
| | | |
Collapse
|
50
|
Huang RB, Mocherla S, Heslinga MJ, Charoenphol P, Eniola-Adefeso O. Dynamic and cellular interactions of nanoparticles in vascular-targeted drug delivery. Mol Membr Biol 2010; 27:312-27. [PMID: 21028938 DOI: 10.3109/09687688.2010.522117] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Vascular-targeted drug delivery systems could provide more efficient and effective pharmaceutical interventions for treating a variety of diseases including cardiovascular, pulmonary, inflammatory, and malignant disorders. However, several factors must be taken into account when designing these systems. The diverse blood hemodynamics and rheology, and the natural clearance process that tend to decrease the circulation time of foreign particles all lessen the probability of successful carrier interaction with the vascular wall. An effective vascular-targeted drug delivery system must be able to navigate through the bloodstream while avoiding immune clearance, attach to the vascular wall, and release its therapeutic cargo at the intended location. This review will summarize and analyze current literature reporting on (1) nanocarrier fabrication methods and materials that allow for optimum therapeutic encapsulation, protection, and release; (2) localization and binding dynamics of nanocarriers as influenced by hemodynamics and blood rheology in medium-to-large vessels; (3) blood cells' responses to various types of nanocarrier compositions and its effects on particle circulation time; and (4) properties that affect nanocarrier internalization at the target site.
Collapse
Affiliation(s)
- Ryan B Huang
- Department of Chemical Engineering, University of Michigan, Ann Arbor, Michigan 48109, USA
| | | | | | | | | |
Collapse
|