101
|
Cellular imaging of endosome entrapped small gold nanoparticles. MethodsX 2015; 2:306-15. [PMID: 26151001 PMCID: PMC4487928 DOI: 10.1016/j.mex.2015.06.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 05/26/2015] [Accepted: 06/05/2015] [Indexed: 12/05/2022] Open
Abstract
Small gold nanoparticles (sAuNPs, <10 nm in a core diameter) have been used for drug delivery and cancer therapy due to their high payload to carrier ratio. Information about the amount and location of sAuNPs in cells and tissues is critical to many applications. However, the current detection method (i.e., transmission electron microscopy) for such sAuNPs is limited due to the extensive sample preparation and the limited field of view. Here we use confocal laser scanning microscopy to provide endosome-entrapped sAuNP distributions and to quantify particle uptake into cells. The quantitative capabilities of the system were confirmed by inductively coupled plasma-mass spectrometry, with an observed linear relation between scattering intensity and the initial cellular uptake of sAuNPs using 4 nm and 6 nm core particles. The summary of the method is: This non-invasive imaging strategy provides a tool for label-free real-time tracking and quantification of sAuNPs using a commercially available confocal laser scanning microscope. Scattering intensity depends on particle size. The linear relation established between scattering intensity and uptaken gold amount enables simultaneous quantitative assessment through simple image analysis.
Collapse
|
102
|
Meyers JD, Cheng Y, Broome AM, Agnes RS, Schluchter MD, Margevicius S, Wang X, Kenney ME, Burda C, Basilion JP. Peptide-Targeted Gold Nanoparticles for Photodynamic Therapy of Brain Cancer. PARTICLE & PARTICLE SYSTEMS CHARACTERIZATION : MEASUREMENT AND DESCRIPTION OF PARTICLE PROPERTIES AND BEHAVIOR IN POWDERS AND OTHER DISPERSE SYSTEMS 2015; 32:448-457. [PMID: 25999665 PMCID: PMC4437573 DOI: 10.1002/ppsc.201400119] [Citation(s) in RCA: 93] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
Targeted drug delivery using epidermal growth factor peptide-targeted gold nanoparticles (EGFpep-Au NPs) is investigated as a novel approach for delivery of photodynamic therapy (PDT) agents, specifically Pc 4, to cancer. In vitro studies of PDT show that EGFpep-Au NP-Pc 4 is twofold better at killing tumor cells than free Pc 4 after increasing localization in early endosomes. In vivo studies show that targeting with EGFpep-Au NP-Pc 4 improves accumulation of fluorescence of Pc 4 in subcutaneous tumors by greater than threefold compared with untargeted Au NPs. Targeted drug delivery and treatment success can be imaged via the intrinsic fluorescence of the PDT drug Pc 4. Using Pc 4 fluorescence, it is demonstrated in vivo that EGFpep-Au NP-Pc 4 impacts biodistribution of the NPs by decreasing the initial uptake by the reticuloendothelial system (RES) and by increasing the amount of Au NPs circulating in the blood 4 h after IV injection. Interestingly, in vivo PDT with EGFpep-Au NP-Pc 4 results in interrupted tumor growth when compared with EGFpep-Au NP control mice when selectively activated with light. These data demonstrate that EGFpep-Au NP-Pc 4 utilizes cancer-specific biomarkers to improve drug delivery and therapeutic efficacy over untargeted drug delivery.
Collapse
Affiliation(s)
- Joseph D. Meyers
- Departments of Biomedical Engineering and Radiology, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Yu Cheng
- Department of Chemistry, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Ann-Marie Broome
- Departments of Biomedical Engineering and Radiology, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Richard S. Agnes
- Departments of Biomedical Engineering and Radiology, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Mark D. Schluchter
- Departments of Epidemiology and Biostatistics, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Seunghee Margevicius
- Departments of Epidemiology and Biostatistics, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Xinning Wang
- Departments of Biomedical Engineering and Radiology, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Malcolm E. Kenney
- Department of Chemistry, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - Clemens Burda
- Department of Chemistry, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| | - James P. Basilion
- Departments of Biomedical Engineering and Radiology, NFCR Center for Molecular Imaging, Case Western Reserve University, 11100 Euclid Ave., Cleveland, OH 44106, USA
| |
Collapse
|
103
|
Zhang F, Mastorakos P, Mishra MK, Mangraviti A, Hwang L, Zhou J, Hanes J, Brem H, Olivi A, Tyler B, Kannan RM. Uniform brain tumor distribution and tumor associated macrophage targeting of systemically administered dendrimers. Biomaterials 2015; 52:507-16. [PMID: 25818456 DOI: 10.1016/j.biomaterials.2015.02.053] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Revised: 02/11/2015] [Accepted: 02/13/2015] [Indexed: 02/07/2023]
Abstract
Effective blood-brain tumor barrier penetration and uniform solid tumor distribution can significantly enhance therapeutic delivery to brain tumors. Hydroxyl-functionalized, generation-4 poly(amidoamine) (PAMAM) dendrimers, with their small size, near-neutral surface charge, and the ability to selectively localize in cells associated with neuroinflammation may offer new opportunities to address these challenges. In this study we characterized the intracranial tumor biodistribution of systemically delivered PAMAM dendrimers in an intracranial rodent gliosarcoma model using fluorescence-based quantification methods and high resolution confocal microscopy. We observed selective and homogeneous distribution of dendrimer throughout the solid tumor (∼6 mm) and peritumoral area within fifteen minutes after systemic administration, with subsequent accumulation and retention in tumor associated microglia/macrophages (TAMs). Neuroinflammation and TAMs have important growth promoting and pro-invasive effects in brain tumors. The rapid clearance of systemically administered dendrimers from major organs promises minimal off-target adverse effects of conjugated drugs. Therefore, selective delivery of immunomodulatory molecules to TAM, using hydroxyl PAMAM dendrimers, may hold promise for therapy of glioblastoma.
Collapse
Affiliation(s)
- Fan Zhang
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Panagiotis Mastorakos
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Manoj K Mishra
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States
| | - Antonella Mangraviti
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Lee Hwang
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Jinyuan Zhou
- F.M. Kirby Research Center for Functional Brain Imaging, Kennedy Krieger Institute, Baltimore, MD 21205, United States
| | - Justin Hanes
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States
| | - Henry Brem
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States; Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States; Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Alessandro Olivi
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Betty Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, United States
| | - Rangaramanujam M Kannan
- Center for Nanomedicine at the Wilmer Eye Institute, Department of Ophthalmology, Johns Hopkins University School of Medicine, Baltimore, MD 21231, United States; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD 21218, United States; Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, United States.
| |
Collapse
|
104
|
Gooding M, Malhotra M, McCarthy DJ, Godinho BMDC, Cryan JF, Darcy R, O'Driscoll CM. Synthesis and characterization of rabies virus glycoprotein-tagged amphiphilic cyclodextrins for siRNA delivery in human glioblastoma cells: in vitro analysis. Eur J Pharm Sci 2015; 71:80-92. [PMID: 25703259 DOI: 10.1016/j.ejps.2015.02.007] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2014] [Revised: 12/23/2014] [Accepted: 02/11/2015] [Indexed: 11/28/2022]
Abstract
In man brain cancer is an aggressive, malignant form of tumour, it is highly infiltrative in nature, is associated with cellular heterogeneity and affects cerebral hemispheres of the brain. Current drug therapies are inadequate and an unmet clinical need exists to develop new improved therapeutics. The ability to silence genes associated with disease progression by using short interfering RNA (siRNA) presents the potential to develop safe and effective therapies. In this work, in order to protect the siRNA from degradation, promote cell specific uptake and enhance gene silencing efficiency, a PEGylated cyclodextrin (CD)-based nanoparticle, tagged with a CNS-targeting peptide derived from the rabies virus glycoprotein (RVG) was formulated and characterized. The modified cyclodextrin derivatives were synthesized and co-formulated to form nanoparticles containing siRNA which were analysed for size, surface charge, stability, cellular uptake and gene-knockdown in brain cancer cells. The results identified an optimised co-formulation prototype at a molar ratio of 1:1.5:0.5 (cationic cyclodextrin:PEGylated cyclodextrin:RVG-tagged PEGylated cyclodextrin) with a size of 281 ± 39.72 nm, a surface charge of 26.73 ± 3 mV, with efficient cellular uptake and a 27% gene-knockdown ability. This CD-based formulation represents a potential nanocomplex for systemic delivery of siRNA targeting brain cancer.
Collapse
Affiliation(s)
- Matt Gooding
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland; Centre for Synthesis and Chemical Biology, University College Dublin, Dublin, Ireland
| | - Meenakshi Malhotra
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - David J McCarthy
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland
| | - Bruno M D C Godinho
- Pharmacodelivery Group, School of Pharmacy, University College Cork, Cork, Ireland; Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland; Lisbon School of Health Technology, Polytechnic Institute of Lisbon, Lisbon, Portugal
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Cork, Ireland
| | - Raphael Darcy
- Centre for Synthesis and Chemical Biology, University College Dublin, Dublin, Ireland
| | | |
Collapse
|
105
|
Gnach A, Lipinski T, Bednarkiewicz A, Rybka J, Capobianco JA. Upconverting nanoparticles: assessing the toxicity. Chem Soc Rev 2015; 44:1561-84. [DOI: 10.1039/c4cs00177j] [Citation(s) in RCA: 438] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Based on a survey of existing studies, low nanotoxicity of lanthanide doped upconverting nanoparticles holds promise for their safety and suitability for biomedical detection and imaging.
Collapse
Affiliation(s)
- Anna Gnach
- Wrocław Research Center EIT+
- 54-066 Wrocław
- Poland
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy
- PAS
| | - Tomasz Lipinski
- Wrocław Research Center EIT+
- 54-066 Wrocław
- Poland
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy
- PAS
| | - Artur Bednarkiewicz
- Wrocław Research Center EIT+
- 54-066 Wrocław
- Poland
- Institute of Low Temp&Structure Research
- PAS
| | - Jacek Rybka
- Wrocław Research Center EIT+
- 54-066 Wrocław
- Poland
- Ludwik Hirszfeld Institute of Immunology and Experimental Therapy
- PAS
| | - John A. Capobianco
- Department of Chemistry and Biochemistry and Centre for NanoScience Research
- Concordia University
- Montreal
- H4B 1R6 Canada
| |
Collapse
|
106
|
Cheng Y, Dai Q, Morshed R, Fan X, Wegscheid ML, Wainwright DA, Han Y, Zhang L, Auffinger B, Tobias AL, Rincón E, Thaci B, Ahmed AU, Warnke P, He C, Lesniak MS. Blood-brain barrier permeable gold nanoparticles: an efficient delivery platform for enhanced malignant glioma therapy and imaging. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:5137-50. [PMID: 25104165 PMCID: PMC4268041 DOI: 10.1002/smll.201400654] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 06/27/2014] [Indexed: 05/19/2023]
Abstract
The blood-brain barrier (BBB) remains a formidable obstacle in medicine, preventing efficient penetration of chemotherapeutic and diagnostic agents to malignant gliomas. Here, a transactivator of transcription (TAT) peptide-modified gold nanoparticle platform (TAT-Au NP) with a 5 nm core size is demonstrated to be capable of crossing the BBB efficiently and delivering cargoes such as the anticancer drug doxorubicin (Dox) and Gd(3+) contrast agents to brain tumor tissues. Treatment of mice bearing intracranial glioma xenografts with pH-sensitive Dox-conjugated TAT-Au NPs via a single intravenous administration leads to significant survival benefit when compared to the free Dox. Furthermore, it is demonstrated that TAT-Au NPs are capable of delivering Gd(3+) chelates for enhanced brain tumor imaging with a prolonged retention time of Gd(3+) when compared to the free Gd(3+) chelates. Collectively, these results show promising applications of the TAT-Au NPs for enhanced malignant brain tumor therapy and non-invasive imaging.
Collapse
Affiliation(s)
| | | | - Ramin Morshed
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Xiaobing Fan
- Department of Radiology, The University of Chicago, Chicago, Illinois, USA
| | - Michelle L. Wegscheid
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Derek A. Wainwright
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Yu Han
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Lingjiao Zhang
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Brenda Auffinger
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Alex L. Tobias
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Esther Rincón
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Bart Thaci
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Atique U. Ahmed
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| | - Peter Warnke
- Department of Surgery, The University of Chicago, Chicago, Illinois, USA
| | - Chuan He
- Department of Chemistry, Institute of Biophysics Dynamics and Howard Hughes Medical Institute, The University of Chicago, Chicago, Illinois, USA
| | - Maciej S. Lesniak
- The Brain Tumor Center, The University of Chicago, Chicago, Illinois, USA, Fax: +1 773 834 2608
| |
Collapse
|
107
|
Nanoparticles and the blood-brain barrier: advancing from in-vitro models towards therapeutic significance. Pharm Res 2014; 32:1161-85. [PMID: 25446769 DOI: 10.1007/s11095-014-1545-6] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2014] [Accepted: 10/06/2014] [Indexed: 01/12/2023]
Abstract
The blood-brain barrier is a unique cell-based restrictive barrier that prevents the entry of many substances, including most therapeutics, into the central nervous system. A wide range of nanoparticulate delivery systems have been investigated with the aim of targeting therapeutics (drugs, nucleic acids, proteins) to the brain following administration by various routes. This review provides a comprehensive description of the design and formulation of these nanoparticles including the rationale behind individual approaches. In addition, the ability of currently available in-vitro BBB models to accurately predict the in-vivo performance of targeted nanoparticles is critically assessed.
Collapse
|
108
|
Mooney MA, Zehri AH, Georges JF, Nakaji P. Laser scanning confocal endomicroscopy in the neurosurgical operating room: a review and discussion of future applications. Neurosurg Focus 2014; 36:E9. [PMID: 24484262 DOI: 10.3171/2013.11.focus13484] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Laser scanning confocal endomicroscopy (LSCE) is an emerging technology for examining brain neoplasms in vivo. While great advances have been made in macroscopic fluorescence in recent years, the ability to perform confocal microscopy in vivo expands the potential of fluorescent tumor labeling, can improve intraoperative tissue diagnosis, and provides real-time guidance for tumor resection intraoperatively. In this review, the authors highlight the technical aspects of confocal endomicroscopy and fluorophores relevant to the neurosurgeon, provide a comprehensive summary of LSCE in animal and human neurosurgical studies to date, and discuss the future directions and potential for LSCE in neurosurgery.
Collapse
|
109
|
Zheng S, Bai YY, Changyi Y, Gao X, Zhang W, Wang Y, Zhou L, Ju S, Li C. Multimodal nanoprobes evaluating physiological pore size of brain vasculatures in ischemic stroke models. Adv Healthc Mater 2014; 3:1909-18. [PMID: 24898608 DOI: 10.1002/adhm.201400159] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2014] [Revised: 04/29/2014] [Indexed: 12/25/2022]
Abstract
Ischemic stroke accounts for 80% strokes and originates from a reduction of cerebral blood flow (CBF) after vascular occlusion. For treatment, the first action is to restore CBF by thrombolytic agent recombinant tissue-type plasminogen activator (rt-PA). Although rt-PA benefits clinical outcome, its application is limited by short therapeutic time window and risk of brain hemorrhage. Different to thrombolytic agents, neuroprotectants reduce neurological injuries by blocking ischemic cascade events such as excitotoxicity and oxidative stress. Nano-neuroprotectants demonstrate higher therapeutic effect than small molecular analogues due to their prolonged circulation lifetime and disrupted blood-brain barrier (BBB) in ischemic region. Even enhanced BBB permeability in ischemic territories is verified, the pore size of ischemic vasculatures determining how large and how efficient the therapeutics can pass is barely studied. In this work, nanoprobes (NPs) with different diameters are developed. In vivo multimodal imaging indicates that NP uptakes in ischemic region depended on their diameters and the pore size upper limit of ischemic vasculatures is determined as 10-11 nm. Additionally, penumbra defined as salvageable ischemic tissues performed a higher BBB permeability than infarct core. This work provides a guideline for developing nano-neuroprotectants by taking advantage of the locally enhanced BBB permeability in ischemic brain tissues.
Collapse
Affiliation(s)
- Shuyan Zheng
- Key Laboratory of Smart Drug Delivery Ministry of Education; School of Pharmacy Fudan University; Shanghai 201203 China
| | - Ying-Ying Bai
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology; Zhongda Hospital Medical School of Southeast University; Nanjing 210009 China
| | - Yinzhi Changyi
- Key Laboratory of Smart Drug Delivery Ministry of Education; School of Pharmacy Fudan University; Shanghai 201203 China
| | - Xihui Gao
- Key Laboratory of Smart Drug Delivery Ministry of Education; School of Pharmacy Fudan University; Shanghai 201203 China
| | - Wenqing Zhang
- Key Laboratory of Smart Drug Delivery Ministry of Education; School of Pharmacy Fudan University; Shanghai 201203 China
| | - Yuancheng Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology; Zhongda Hospital Medical School of Southeast University; Nanjing 210009 China
| | - Lu Zhou
- Key Laboratory of Smart Drug Delivery Ministry of Education; School of Pharmacy Fudan University; Shanghai 201203 China
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology; Zhongda Hospital Medical School of Southeast University; Nanjing 210009 China
| | - Cong Li
- Key Laboratory of Smart Drug Delivery Ministry of Education; School of Pharmacy Fudan University; Shanghai 201203 China
| |
Collapse
|
110
|
Sivasubramanian M, Hsia Y, Lo LW. Nanoparticle-facilitated functional and molecular imaging for the early detection of cancer. Front Mol Biosci 2014; 1:15. [PMID: 25988156 PMCID: PMC4428449 DOI: 10.3389/fmolb.2014.00015] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 09/19/2014] [Indexed: 01/08/2023] Open
Abstract
Cancer detection in its early stages is imperative for effective cancer treatment and patient survival. In recent years, biomedical imaging techniques, such as magnetic resonance imaging, computed tomography and ultrasound have been greatly developed and have served pivotal roles in clinical cancer management. Molecular imaging (MI) is a non-invasive imaging technique that monitors biological processes at the cellular and sub-cellular levels. To achieve these goals, MI uses targeted imaging agents that can bind targets of interest with high specificity and report on associated abnormalities, a task that cannot be performed by conventional imaging techniques. In this respect, MI holds great promise as a potential therapeutic tool for the early diagnosis of cancer. Nevertheless, the clinical applications of targeted imaging agents are limited due to their inability to overcome biological barriers inside the body. The use of nanoparticles has made it possible to overcome these limitations. Hence, nanoparticles have been the subject of a great deal of recent studies. Therefore, developing nanoparticle-based imaging agents that can target tumors via active or passive targeting mechanisms is desirable. This review focuses on the applications of various functionalized nanoparticle-based imaging agents used in MI for the early detection of cancer.
Collapse
Affiliation(s)
- Maharajan Sivasubramanian
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes Zhunan, Taiwan
| | - Yu Hsia
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes Zhunan, Taiwan ; Institute of Biotechnology, National Tsing Hua University Hsinchu, Taiwan
| | - Leu-Wei Lo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes Zhunan, Taiwan
| |
Collapse
|
111
|
Zhou Z, Lu ZR. Dendritic nanoglobules with polyhedral oligomeric silsesquioxane core and their biomedical applications. Nanomedicine (Lond) 2014; 9:2387-401. [DOI: 10.2217/nnm.14.133] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Dendrimers have been recognized as promising materials for biomedical applications due to their unique properties, such as well-defined unimolecular structures, precise molecular weights and high degrees of branching and surface functionality. The dendrimers with a polyhedral oligomeric silsesquioxane core – nanoglobules – have many advantageous features over traditional dendrimers, including more functional groups at the same generation of dendrimers, well-defined 3D structures and relatively compact morphologies. Various nanoglobules of polyhedral oligomeric silsesquioxane-core dendrimers have been synthesized and investigated for biomedical applications. Nanoglobules have been used as carriers for developing drug-delivery systems, gene-delivery systems and imaging contrast agents with precisely defined structures and sizes. This article summarizes the recent developments in nanoglobules for biomedical applications.
Collapse
Affiliation(s)
- Zhuxian Zhou
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| | - Zheng-Rong Lu
- Department of Biomedical Engineering, Case Western Reserve University, 10900 Euclid Avenue, Cleveland, OH 44106, USA
| |
Collapse
|
112
|
Zeng Y, Zhang D, Wu M, Liu Y, Zhang X, Li L, Li Z, Han X, Wei X, Liu X. Lipid-AuNPs@PDA nanohybrid for MRI/CT imaging and photothermal therapy of hepatocellular carcinoma. ACS APPLIED MATERIALS & INTERFACES 2014; 6:14266-14277. [PMID: 25090604 DOI: 10.1021/am503583s] [Citation(s) in RCA: 110] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
Multifunctional theranostic nanoparticles represent an emerging agent with the potential to offer extremely sensitive diagnosis and targeted cancer therapy. Herein, we report the synthesis and characterization of a multifunctional theranostic agent (referred to as LA-LAPNHs) for targeted magnetic resonance imaging/computed X-ray tomography (MRI/CT) dual-mode imaging and photothermal therapy of hepatocellular carcinoma. The LA-LAPNHs were characterized as having a core-shell structure with the gold nanoparticles (AuNPs)@polydopamine (PDA) as the inner core, the indocyanine green (ICG), which is electrostatically absorbed onto the surface of PDA, as the photothermal therapeutic agent, and the lipids modified with gadolinium-1,4,7,10-tetraacetic acid and lactobionic acid (LA), which is self-assembled on the outer surface as the shell. The LA-LAPNHs could be selectively internalized into the hepatocellular cell line (HepG2 cells) but not into HeLa cells due to the specific recognition ability of LA to asialoglycoprotein receptor. Additionally, the dual-mode imaging ability of the LA-LAPNH aqueous solution was confirmed by enhanced MR and CT imaging showing a shorter T1 relaxation time and a higher Hounsfield unit value, respectively. In addition, the LA-LAPNHs showed significant photothermal cytotoxicity against liver cancer cells with near-infrared irradiation due to their strong absorbance in the region between 700 and 850 nm. In summary, this study demonstrates that LA-LAPNHs may be a promising candidate for targeted MR/CT dual-mode imaging and photothermal therapy of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Yongyi Zeng
- Liver Disease Center, The First Affiliated Hospital of Fujian Medical University , Fuzhou 350005, P. R. China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
113
|
Chong K, Ku T, Saw PE, Jon S, Park JH, Choi C. Enhancement of the photocytotoxic efficiency of sub-12 nm therapeutic polymeric micelles with increased co-localisation in mitochondria. Chem Commun (Camb) 2014; 49:11476-8. [PMID: 24064984 DOI: 10.1039/c3cc46166a] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
We engineered phototherapeutic sub-12 nm-sized polymeric micelles to treat malignant brain tumours (MBTs). The engineered nanoparticles in MBT cells enhanced the photocytotoxic efficiency more than 2.5-fold compared with parental and PEGylated photosensitisers (PSs). Increased subcellular co-localisation of PSs in mitochondria was observed.
Collapse
Affiliation(s)
- Kyuha Chong
- Graduate School of Medical Science and Engineering, KAIST, 291 Daehak-ro, Yuseong-gu, Daejeon 305-701, Republic of Korea
| | | | | | | | | | | |
Collapse
|
114
|
Lu CT, Zhao YZ, Wong HL, Cai J, Peng L, Tian XQ. Current approaches to enhance CNS delivery of drugs across the brain barriers. Int J Nanomedicine 2014; 9:2241-57. [PMID: 24872687 PMCID: PMC4026551 DOI: 10.2147/ijn.s61288] [Citation(s) in RCA: 188] [Impact Index Per Article: 18.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Although many agents have therapeutic potentials for central nervous system (CNS) diseases, few of these agents have been clinically used because of the brain barriers. As the protective barrier of the CNS, the blood–brain barrier and the blood–cerebrospinal fluid barrier maintain the brain microenvironment, neuronal activity, and proper functioning of the CNS. Different strategies for efficient CNS delivery have been studied. This article reviews the current approaches to open or facilitate penetration across these barriers for enhanced drug delivery to the CNS. These approaches are summarized into three broad categories: noninvasive, invasive, and miscellaneous techniques. The progresses made using these approaches are reviewed, and the associated mechanisms and problems are discussed.
Collapse
Affiliation(s)
- Cui-Tao Lu
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, People's Republic of China
| | - Ying-Zheng Zhao
- Hainan Medical College, Haikou City, Hainan Province, People's Republic of China ; College of Pharmaceutical Sciences, Wenzhou Medical University, Zhejiang Province, People's Republic of China
| | - Ho Lun Wong
- School of Pharmacy, Temple University, Philadelphia, PA, USA
| | - Jun Cai
- Departments of Pediatrics and Anatomical Sciences and Neurobiology, University of Louisville School of Medicine Louisville, KY, USA
| | - Lei Peng
- Hainan Medical College, Haikou City, Hainan Province, People's Republic of China
| | - Xin-Qiao Tian
- The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou City, Zhejiang Province, People's Republic of China
| |
Collapse
|
115
|
Voigt N, Henrich-Noack P, Kockentiedt S, Hintz W, Tomas J, Sabel BA. Surfactants, not size or zeta-potential influence blood–brain barrier passage of polymeric nanoparticles. Eur J Pharm Biopharm 2014; 87:19-29. [DOI: 10.1016/j.ejpb.2014.02.013] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2013] [Revised: 01/16/2014] [Accepted: 02/24/2014] [Indexed: 10/25/2022]
|
116
|
How to study dendrimers and dendriplexes III. Biodistribution, pharmacokinetics and toxicity in vivo. J Control Release 2014; 181:40-52. [DOI: 10.1016/j.jconrel.2014.02.021] [Citation(s) in RCA: 83] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2014] [Revised: 02/21/2014] [Accepted: 02/22/2014] [Indexed: 12/15/2022]
|
117
|
Lamberti M, Zappavigna S, Sannolo N, Porto S, Caraglia M. Advantages and risks of nanotechnologies in cancer patients and occupationally exposed workers. Expert Opin Drug Deliv 2014; 11:1087-101. [DOI: 10.1517/17425247.2014.913568] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
|
118
|
LIANG RUICHAO, FANG FANG. THE APPLICATION OF NANOMATERIALS IN DIAGNOSIS AND TREATMENT FOR MALIGNANT PRIMARY BRAIN TUMORS. NANO 2014. [DOI: 10.1142/s1793292014300011] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Malignant primary brain tumors have a very high morbidity and mortality. Even though enormous advances have been made in primary brain tumor management, in the case of malignant primary brain tumors, current diagnostic strategies cannot identify exact infiltrating margins, surgery alone cannot achieve total mass resection, and adjuvant therapies cannot improve survivals. Therefore, there is an urgent need to explore novel strategies to diagnose and treat such infiltrating brain tumors. Nanomaterials, particularly zero-dimensional and one-dimensional platforms, can carry various compounds such as contrast agents, anticancer drugs and genes into brain tumor cells specifically. Thus, contrast agent-based nanomaterials can selectively present infiltrating tumor outlines, while anticancer agent-based nanomaterials can specifically kill malignant tumor cells. In addition, dual-targeting nanomaterials, multifunctional nanocarriers, theranostic nanovehicles as well as convection-enhanced delivery technology hold promise to increase drug accumulation in tumor tissues, which could largely improve anticancer efficacy. In this review, we will mainly focus on the application of nanomaterials in preoperative diagnosis, intraoperative diagnosis and adjuvant treatment for malignant primary brain tumors.
Collapse
Affiliation(s)
- RUICHAO LIANG
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| | - FANG FANG
- Department of Neurosurgery, West China Hospital, Sichuan University, Chengdu, Sichuan, P. R. China
| |
Collapse
|
119
|
Gao X, Li C. Nanoprobes visualizing gliomas by crossing the blood brain tumor barrier. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2014; 10:426-440. [PMID: 24106064 DOI: 10.1002/smll.201301673] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/04/2013] [Indexed: 06/02/2023]
Abstract
The difficulty in delineating the glioma margins in brain is a major obstacle for its completed resection, which leads to the disproportionately high recurrence and mortality. Besides the fast exertion rate, inadequate sensitivity and non-targeting specificity, the main reason leading to failure of small molecular probes to define gliomas is their incapability to efficiently cross the blood brain tumor barrier (BBTB). Nanoprobes (NPs) show promise to precisely delineate the geographically irregular tumor margins due to their tunable size/circulation lifetime that maximize their passive intratumoral accumulation and their convenience for surface modification that increases the BBTB transcytosis efficacy, imaging sensitivity and receptor targeting specificity. In this work, the characteristics of the BBTB are addressed from biological and physiological perspectives, strategies are presented to deliver NPs across the BBTB, recent developments of NPs are reviewed for glioma visualization and finally the difficulty and promise for clinical translation of NPs are described. Overall, NPs hold great potential for glioma imaging and treatment by pre-surgically delineating tumor margins and intra-operatively guiding tumor excision.
Collapse
Affiliation(s)
- Xihui Gao
- Key Laboratory of Smart Drug Delivery, Ministry of Education, School of Pharmacy, Fudan University 826 Zhangheng Rd., Shanghai, 201203, China
| | | |
Collapse
|
120
|
Abstract
Phenomenal advances in nanotechnology and nanoscience have been accompanied by exciting progress in de novo design of nanomedicines. Nanoparticles with their large space of structural amenability and excellent mechanical and electrical properties have become ideal candidates for high efficacy nanomedicines in both diagnostics and therapeutics. The therapeutic nanomedicines can be further categorized into nanocarriers for conventional drugs and nanodrugs with direct curing of target diseases. Here we review some of the recent advances in de novo design of nanodrugs, with an emphasis on the molecular level understanding of their interactions with biological systems including key proteins and cell membranes. We also include some of the latest advances in the development of nanocarriers with both passive and active targeting for completeness. These studies may shed light on a better understanding of the molecular mechanisms behind these nanodrugs, and also provide new insights and direction for the future design of nanomedicines.
Collapse
Affiliation(s)
- Zaixing Yang
- School of Radiation Medicine and Protection, Medical College of Soochow University & Collaborative Innovation Center of Suzhou Nano Science and Technology, Jiangsu Provincial Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou 215123, China.
| | | | | |
Collapse
|
121
|
Abstract
Patients affected by malignant brain tumors present an extremely poor prognosis, notwithstanding improvements in surgery techniques and therapeutic protocols. Brain tumor treatment has been principally hampered by limited drug delivery across the blood–brain barrier (BBB). An efficacious chemotherapeutic treatment requires a pharmacological agent that can penetrate the BBB and target neoplastic cells. Nanotechnology involves the design, synthesis and characterization of materials that have a functional organization in at least one dimension on the nanometer scale. Nanoparticle systems can represent optimal devices for delivery of various drugs into the brain across the BBB. Nanoparticle drug-delivery systems can also be used to provide targeted delivery of drugs, improve bioavailability and sustain release of drugs for systemic delivery. In this patent review, the recent studies of certain nanoparticle systems in treatment of brain tumors are summarized. Common nanoparticles systems include polymeric nanoparticles, lipid nanoparticles and inorganic nanoparticles. Various patents of nanoparticle systems able to across the BBB to target brain tumors are also reported and discussed.
Collapse
|
122
|
A choline derivate-modified nanoprobe for glioma diagnosis using MRI. Sci Rep 2013; 3:1623. [PMID: 23563908 PMCID: PMC3619251 DOI: 10.1038/srep01623] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Accepted: 03/15/2013] [Indexed: 12/26/2022] Open
Abstract
Gadolinium (Gd) chelate contrast-enhanced magnetic resonance imaging (MRI) is a preferred method of glioma detection and preoperative localisation because it offers high spatial resolution and non-invasive deep tissue penetration. Gd-based contrast agents, such as Gd-diethyltriaminepentaacetic acid (DTPA-Gd, Magnevist), are widely used clinically for tumor diagnosis. However, the Gd-based MRI approach is limited for patients with glioma who have an uncompromised blood-brain barrier (BBB). Moreover, the rapid renal clearance and non-specificity of such contrast agents further hinders their prevalence. We present a choline derivate (CD)-modified nanoprobe with BBB permeability, glioma specificity and a long blood half-life. Specific accumulation of the nanoprobe in gliomas and subsequent MRI contrast enhancement are demonstrated in vitro in U87 MG cells and in vivo in a xenograft nude model. BBB and glioma dual targeting by this nanoprobe may facilitate precise detection of gliomas with an uncompromised BBB and may offer better preoperative and intraoperative tumor localization.
Collapse
|
123
|
Li Y, Rey-Dios R, Roberts DW, Valdés PA, Cohen-Gadol AA. Intraoperative fluorescence-guided resection of high-grade gliomas: a comparison of the present techniques and evolution of future strategies. World Neurosurg 2013; 82:175-85. [PMID: 23851210 DOI: 10.1016/j.wneu.2013.06.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 04/15/2013] [Accepted: 06/29/2013] [Indexed: 12/30/2022]
Abstract
OBJECTIVE Fluorescence guidance has a demonstrated potential in maximizing the extent of high-grade glioma resection. Different fluorophores (fluorescent biomarkers), including 5-aminolevulinic acid (5-ALA) and fluorescein, have been examined with the use of several imaging techniques. Our goal was to review the state of this technology and discuss strategies for more widespread adoption. METHODS We performed a Medline search using the key words "fluorescence," "intraoperative fluorescence-guided resection," "intraoperative image-guided resection," and "brain glioma" for articles from 1960 until the present. This initial search revealed 267 articles. Each abstract and article was reviewed and the reference lists from select articles were further evaluated for relevance. A total of 64 articles included information about the role of fluorescence in resection of high-grade gliomas and therefore were selectively included for our analysis. RESULTS 5-ALA and fluorescein sodium have shown promise as fluorescent markers in detecting residual tumor intraoperatively. These techniques have demonstrated a significant increase in the extent of tumor resection. Regulatory barriers have limited the use of 5-ALA and technological challenges have restricted the use of fluorescein and its derivatives in the United States. Limitations to this technology currently exist, such as the fact that fluorescence at tumor margins is not always reliable for identification of tumor-brain interface. CONCLUSIONS These techniques are safe and effective for increasing gross total resection. The development of more tumor-specific fluorophores is needed to resolve problems with subjective interpretation of fluorescent signal at tumor margins. Techniques such as quantum dots and polymer or iron oxide-based nanoparticles have shown promise as potential future tools.
Collapse
Affiliation(s)
- Yiping Li
- Goodman Campbell Brain and Spine, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Roberto Rey-Dios
- Department of Neurosurgery, University of Mississippi Medical Center, Jackson, Mississippi, USA
| | - David W Roberts
- Section of Neurosurgery, Department of Surgery, Dartmouth Medical School, Lebanon, New Hampshire, USA; Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Pablo A Valdés
- Section of Neurosurgery, Department of Surgery, Dartmouth Medical School, Lebanon, New Hampshire, USA; Dartmouth Medical School, Hanover, New Hampshire, USA
| | - Aaron A Cohen-Gadol
- Goodman Campbell Brain and Spine, Department of Neurological Surgery, Indiana University School of Medicine, Indianapolis, Indiana, USA.
| |
Collapse
|
124
|
Chacko AM, Li C, Pryma DA, Brem S, Coukos G, Muzykantov V. Targeted delivery of antibody-based therapeutic and imaging agents to CNS tumors: crossing the blood-brain barrier divide. Expert Opin Drug Deliv 2013; 10:907-26. [PMID: 23751126 PMCID: PMC4089357 DOI: 10.1517/17425247.2013.808184] [Citation(s) in RCA: 75] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
INTRODUCTION Brain tumors are inherently difficult to treat in large part due to the cellular blood-brain barriers (BBBs) that limit the delivery of therapeutics to the tumor tissue from the systemic circulation. Virtually no large molecules, including antibody-based proteins, can penetrate the BBB. With antibodies fast becoming attractive ligands for highly specific molecular targeting to tumor antigens, a variety of methods are being investigated to enhance the access of these agents to intracranial tumors for imaging or therapeutic applications. AREAS COVERED This review describes the characteristics of the BBB and the vasculature in brain tumors, described as the blood-brain tumor barrier (BBTB). Antibodies targeted to molecular markers of central nervous system (CNS) tumors will be highlighted, and current strategies for enhancing the delivery of antibodies across these cellular barriers into the brain parenchyma to the tumor will be discussed. Noninvasive imaging approaches to assess BBB/BBTB permeability and/or antibody targeting will be presented as a means of guiding the optimal delivery of targeted agents to brain tumors. EXPERT OPINION Preclinical and clinical studies highlight the potential of several approaches in increasing brain tumor delivery across the BBB divide. However, each carries its own risks and challenges. There is tremendous potential in using neuroimaging strategies to assist in understanding and defining the challenges to translating and optimizing molecularly targeted antibody delivery to CNS tumors to improve clinical outcomes.
Collapse
Affiliation(s)
- Ann-Marie Chacko
- University of Pennsylvania, Perelman School of Medicine, Nuclear Medicine & Clinical Molecular Imaging, Department of Radiology, 231 S. 34 Street, Room 288, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
125
|
Ramachandran PS, Keiser MS, Davidson BL. Recent advances in RNA interference therapeutics for CNS diseases. Neurotherapeutics 2013; 10:473-85. [PMID: 23589092 PMCID: PMC3701762 DOI: 10.1007/s13311-013-0183-8] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
Over the last decade, RNA interference technology has shown therapeutic promise in rodent models of dominantly inherited brain diseases, including those caused by polyglutamine repeat expansions in the coding region of the affected gene. For some of these diseases, proof-of concept studies in model organisms have transitioned to safety testing in larger animal models, such as the nonhuman primate. Here, we review recent progress on RNA interference-based therapies in various model systems. We also highlight outstanding questions or concerns that have emerged as a result of an improved (and ever advancing) understanding of the technologies employed.
Collapse
Affiliation(s)
| | - Megan S. Keiser
- />Interdisciplinary program in Neuroscience, University of Iowa, Iowa City, IA USA
| | - Beverly L. Davidson
- />Interdisciplinary program in Genetics, University of Iowa, Iowa City, IA 52242 USA
- />Interdisciplinary program in Neuroscience, University of Iowa, Iowa City, IA USA
- />Department of Internal Medicine, University of Iowa, Iowa City, USA
| |
Collapse
|
126
|
Gao H, Pang Z, Jiang X. Targeted Delivery of Nano-Therapeutics for Major Disorders of the Central Nervous System. Pharm Res 2013; 30:2485-98. [DOI: 10.1007/s11095-013-1122-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2013] [Accepted: 06/11/2013] [Indexed: 12/22/2022]
|
127
|
Suh KS, Lee YS, Seo SH, Kim YS, Choi EM. Gold nanoparticles attenuates antimycin A-induced mitochondrial dysfunction in MC3T3-E1 osteoblastic cells. Biol Trace Elem Res 2013; 153:428-36. [PMID: 23645457 DOI: 10.1007/s12011-013-9679-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/12/2013] [Accepted: 04/18/2013] [Indexed: 01/09/2023]
Abstract
Gold nanoparticles have shown promising biological applications due to their unique properties. Understanding the interaction mechanisms between nanomaterials and biological cells is important for the control and manipulation of these interactions for biomedical applications. In the present study, we investigated the effects of gold nanoparticles on the differentiation of osteoblastic MC3T3-E1 cells and antimycin A-induced mitochondrial dysfunction. The results showed that gold nanoparticles (5, 10, and 20 nm) caused a significant elevation of cell growth, alkaline phosphatase activity, collagen synthesis, and osteocalcin content in the cells (P < 0.05). Moreover, pretreatment with gold nanoparticles prior to antimycin A exposure significantly reduced antimycin A-induced cell damage by preventing mitochondrial membrane potential dissipation, complex IV inactivation, ATP loss, cytochrome c release, cardiolipin peroxidation, and reactive oxygen species generation. Taken together, our study indicated that gold nanoparticles may improve the differentiation and have protective effects on mitochondrial dysfunction of osteoblastic cells.
Collapse
Affiliation(s)
- Kwang Sik Suh
- Research Institute of Endocrinology, Kyung Hee University Hospital, 1, Hoegi-dong, Dongdaemun-gu, Seoul 130-702, South Korea
| | | | | | | | | |
Collapse
|
128
|
Masserini M. Nanoparticles for brain drug delivery. ISRN BIOCHEMISTRY 2013; 2013:238428. [PMID: 25937958 PMCID: PMC4392984 DOI: 10.1155/2013/238428] [Citation(s) in RCA: 262] [Impact Index Per Article: 23.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2013] [Accepted: 04/11/2013] [Indexed: 12/24/2022]
Abstract
The central nervous system, one of the most delicate microenvironments of the body, is protected by the blood-brain barrier (BBB) regulating its homeostasis. BBB is a highly complex structure that tightly regulates the movement of ions of a limited number of small molecules and of an even more restricted number of macromolecules from the blood to the brain, protecting it from injuries and diseases. However, the BBB also significantly precludes the delivery of drugs to the brain, thus, preventing the therapy of a number of neurological disorders. As a consequence, several strategies are currently being sought after to enhance the delivery of drugs across the BBB. Within this review, the recently born strategy of brain drug delivery based on the use of nanoparticles, multifunctional drug delivery systems with size in the order of one-billionth of meters, is described. The review also includes a brief description of the structural and physiological features of the barrier and of the most utilized nanoparticles for medical use. Finally, the potential neurotoxicity of nanoparticles is discussed, and future technological approaches are described. The strong efforts to allow the translation from preclinical to concrete clinical applications are worth the economic investments.
Collapse
Affiliation(s)
- Massimo Masserini
- Department of Health Sciences, University of Milano-Bicocca, Via Cadore 48, 20900 Monza, Italy
| |
Collapse
|
129
|
Halamoda Kenzaoui B, Angeloni S, Overstolz T, Niedermann P, Chapuis Bernasconi C, Liley M, Juillerat-Jeanneret L. Transfer of ultrasmall iron oxide nanoparticles from human brain-derived endothelial cells to human glioblastoma cells. ACS APPLIED MATERIALS & INTERFACES 2013; 5:3581-3586. [PMID: 23578059 DOI: 10.1021/am401310s] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Nanoparticles (NPs) are being used or explored for the development of biomedical applications in diagnosis and therapy, including imaging and drug delivery. Therefore, reliable tools are needed to study the behavior of NPs in biological environment, in particular the transport of NPs across biological barriers, including the blood-brain tumor barrier (BBTB), a challenging question. Previous studies have addressed the translocation of NPs of various compositions across cell layers, mostly using only one type of cells. Using a coculture model of the human BBTB, consisting in human cerebral endothelial cells preloaded with ultrasmall superparamagnetic iron oxide nanoparticles (USPIO NPs) and unloaded human glioblastoma cells grown on each side of newly developed ultrathin permeable silicon nitride supports as a model of the human BBTB, we demonstrate for the first time the transfer of USPIO NPs from human brain-derived endothelial cells to glioblastoma cells. The reduced thickness of the permeable mechanical support compares better than commercially available polymeric supports to the thickness of the basement membrane of the cerebral vascular system. These results are the first report supporting the possibility that USPIO NPs could be directly transferred from endothelial cells to glioblastoma cells across a BBTB. Thus, the use of such ultrathin porous supports provides a new in vitro approach to study the delivery of nanotherapeutics to brain cancers. Our results also suggest a novel possibility for nanoparticles to deliver therapeutics to the brain using endothelial to neural cells transfer.
Collapse
Affiliation(s)
- Blanka Halamoda Kenzaoui
- Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | | | | | | | | | | | | |
Collapse
|
130
|
Lee BS, Amano T, Wang HQ, Pantoja JL, Yoon CW, Hanson CJ, Amatya R, Yen A, Black KL, Yu JS. Reactive oxygen species responsive nanoprodrug to treat intracranial glioblastoma. ACS NANO 2013; 7:3061-3077. [PMID: 23557138 DOI: 10.1021/nn400347j] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Chemotherapy for intracranial gliomas is hampered by limited delivery of therapeutic agents through the blood brain barrier (BBB). An optimal therapeutic agent for brain tumors would selectively cross the BBB, accumulates in the tumor tissue and be activated from an innocuous prodrug within the tumor. Here we show brain tumor-targeted delivery and therapeutic efficacy of a nanometer-sized prodrug (nanoprodrug) of camptothecin (CPT) to treat experimental glioblastoma multiforme (GBM). The CPT nanoprodrug was prepared using spontaneous nanoemulsification of a biodegradable, antioxidant CPT prodrug and α-tocopherol. The oxidized nanoprodrug was activated more efficiently than nonoxidized nanoprodrug, suggesting enhanced therapeutic efficacy in the oxidative tumor microenvironment. The in vitro imaging of U-87 MG glioma cells revealed an efficient intracellular uptake of the nanoprodrug via direct cell membrane penetration rather than via endocytosis. The in vivo study in mice demonstrated that the CPT nanoprodrug passed through the BBB and specifically accumulated in brain tumor tissue, but not in healthy brain tissue and other organs. The accumulation preferably occurred at the periphery of the tumor where cancer cells are most actively proliferating, suggesting optimal therapeutic efficacy of the nanoprodrug. The nanoprodrug was effective in treating subcutaneous and intracranial tumors. The nanoprodrug inhibited subcutaneous tumor growth more than 80% compared with control. The median survival time of mice implanted with an intracranial tumor increased from 40.5 days for control to 72.5 days for CPT nanoprodrug. This nanoprodrug approach is a versatile method for developing therapeutic nanoparticles enabling tumor-specific targeting and treatment. The nontoxic, tumor-specific targeting properties of the nanoprodrug system make it a safe, low cost, and versatile nanocarrier for pharmaceuticals, imaging agents, and diagnostic agents.
Collapse
Affiliation(s)
- Bong-Seop Lee
- Department of Neurosurgery, Cedars-Sinai Medical Center, 8631 West Third Street, Suite 800 East, Los Angeles, California 90048, United States
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Pharmacodynamic analysis of magnetic resonance imaging-monitored focused ultrasound-induced blood-brain barrier opening for drug delivery to brain tumors. BIOMED RESEARCH INTERNATIONAL 2013; 2013:627496. [PMID: 23607093 PMCID: PMC3626247 DOI: 10.1155/2013/627496] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/21/2013] [Accepted: 02/25/2013] [Indexed: 12/22/2022]
Abstract
Microbubble-enhanced focused ultrasound (FUS) can enhance the delivery of therapeutic agents into the brain for brain tumor treatment. The purpose of this study was to investigate the influence of brain tumor conditions on the distribution and dynamics of small molecule leakage into targeted regions of the brain after FUS-BBB opening. A total of 34 animals were used, and the process was monitored by 7T-MRI. Evans blue (EB) dye as well as Gd-DTPA served as small molecule substitutes for evaluation of drug behavior. EB was quantified spectrophotometrically. Spin-spin (R1) relaxometry and area under curve (AUC) were measured by MRI to quantify Gd-DTPA. We found that FUS-BBB opening provided a more significant increase in permeability with small tumors. In contrast, accumulation was much higher in large tumors, independent of FUS. The AUC values of Gd-DTPA were well correlated with EB delivery, suggesting that Gd-DTPA was a good indicator of total small-molecule accumulation in the target region. The peripheral regions of large tumors exhibited similar dynamics of small-molecule leakage after FUS-BBB opening as small tumors, suggesting that FUS-BBB opening may have the most significant permeability-enhancing effect on tumor peripheral. This study provides useful information toward designing an optimized FUS-BBB opening strategy to deliver small-molecule therapeutic agents into brain tumors.
Collapse
|
132
|
Daneshvar N, Abdullah R, Shamsabadi FT, How CW, Mh MA, Mehrbod P. PAMAM dendrimer roles in gene delivery methods and stem cell research. Cell Biol Int 2013; 37:415-9. [PMID: 23504853 DOI: 10.1002/cbin.10051] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Accepted: 01/10/2013] [Indexed: 12/21/2022]
Abstract
Nanotechnology has provided new technological opportunities, which could help in challenges confronting stem cell research. Polyamidoamine (PAMAM) dendrimers, a new class of macromolecular polymers with high molecular uniformity, narrow molecular distribution specific size and shape and highly functionalised terminal surface have been extensively explored for biomedical application. PAMAM dendrimers are also nanospherical, hyperbranched and monodispersive molecules exhibiting exclusive properties which make them potential carriers for drug and gene delivery.
Collapse
Affiliation(s)
- Nasibeh Daneshvar
- Institute of Bioscience, Universiti Putra Malaysia, 43400 UPM Serdang, Selangor, Malaysia
| | | | | | | | | | | |
Collapse
|
133
|
Siegal T. Which drug or drug delivery system can change clinical practice for brain tumor therapy? Neuro Oncol 2013; 15:656-69. [PMID: 23502426 DOI: 10.1093/neuonc/not016] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The prognosis and treatment outcome for primary brain tumors have remained unchanged despite advances in anticancer drug discovery and development. In clinical trials, the majority of promising experimental agents for brain tumors have had limited impact on survival or time to recurrence. These disappointing results are partially explained by the inadequacy of effective drug delivery to the CNS. The impediments posed by the various specialized physiological barriers and active efflux mechanisms lead to drug failure because of inability to reach the desired target at a sufficient concentration. This perspective reviews the leading strategies that aim to improve drug delivery to brain tumors and their likelihood to change clinical practice. The English literature was searched for defined search items. Strategies that use systemic delivery and those that use local delivery are critically reviewed. In addition, challenges posed for drug delivery by combined treatment with anti-angiogenic therapy are outlined. To impact clinical practice and to achieve more than just a limited local control, new drugs and delivery systems must adhere to basic clinical expectations. These include, in addition to an antitumor effect, a verified favorable adverse effects profile, easy introduction into clinical practice, feasibility of repeated or continuous administration, and compatibility of the drug or delivery system with any tumor size and brain location.
Collapse
Affiliation(s)
- Tali Siegal
- Gaffin Center for Neuro-Oncology, Hadassah Hebrew-University Medical Center, Ein Kerem, P.O. Box 12000, Jerusalem 91120, Israel.
| |
Collapse
|
134
|
"Nanogold detoxifying machine" to remove idle nanogold particles from blood stream of cancer patients treated with antibody-nanogold therapeutics. Med Hypotheses 2013; 80:601-5. [PMID: 23462370 DOI: 10.1016/j.mehy.2013.01.035] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 01/21/2013] [Accepted: 01/29/2013] [Indexed: 11/21/2022]
Abstract
This hypothesis would help to elevate safety in nanogold-therapy of some cancers. It has no previous record in published media and there is no existing protocol for what is expressed in this paper. The hypothesis makes some experimental recommendations for future research at the interface of nanotechnology and biological systems. It is about reducing toxicity and side effects of nanogold particles (NGPs) in cancer patients treated with antibody-nanogold therapeutics. The hypothesis expresses that how to eliminate idle NGPs from blood stream of treated cancer patients in order to reduce undesirable deposition and accumulation of NGPs in non-target tissues and organs in post therapy. In this regard, at the end of therapy period, blood of treated cancer patient which contains idle nanogold-antibodies is passed through a machine named "Nanogold detoxifying machine" (NDM). This machine behaves like "hemodialysis machine" used for patients with lost kidney function but, instead of dialysis membrane, NDM is equipped with impermeable membrane containing nanotubes to which cancer specific-antigens (CSAs) are attached. While blood circulates in NDM, CSAs attract and immobilize the idle nanogold-antibodies. Implementation of NDM clears patient's blood from idle nanogold-antibodies which were not attached to their targets, the cancer cells. Elimination of idle NGPs from blood would minimize side effects resulting from their deposition or accumulation on non-target cells or tissues. Since NDM reduces anxieties about side effects, it would provide patients with better feelings too. NDM would enhance efficacy of NGPs targeted toward specific cancers. Before using it for clinical trials in human cases, NDM should pass animal evaluations; however, research works should optimize its implementation. I think NDM method holds great promise to reduce the time, effort, and expense in cancer research, screening, detection, and therapy.
Collapse
|
135
|
Iqbal A, Ahmad I, Khalid MH, Nawaz MS, Gan SH, Kamal MA. Nanoneurotoxicity to nanoneuroprotection using biological and computational approaches. JOURNAL OF ENVIRONMENTAL SCIENCE AND HEALTH. PART C, ENVIRONMENTAL CARCINOGENESIS & ECOTOXICOLOGY REVIEWS 2013; 31:256-284. [PMID: 24024521 DOI: 10.1080/10590501.2013.829706] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/02/2023]
Abstract
Nanoparticles (NPs) that are ∼100 nm in diameter can potentially cause toxicity in the central nervous system (CNS). Although NPs exhibit positive aspects, these molecules primarily exert negative or harmful effects. Thus, the beneficial and harmful effects should be compared. The prevalence of neurodegenerative diseases, such as Alzheimer disease, Parkinson disease, and some brain tumors, has increased. However, the major cause of these diseases remains unknown. NPs have been considered as one of the major potential causes of these diseases, penetrating the human body via different pathways. This review summarizes various pathways for NP-induced neurotoxicity, suggesting the development of strategies for nanoneuroprotection using in silico and biological methods. Studies of oxidative stress associated with gene expression analyses provide efficient information for understanding neuroinflammation and neurodegeneration associated with NPs. The brain is a sensitive and fragile organ, and evolution has developed mechanisms to protect it from injury; however, this protection also hinders the methods used for therapeutic purposes. Thus, brain and CNS-related diseases that are the cause of disability and disorder are the most difficult to treat. There are many obstacles to drug delivery in the CNS, such as the blood brain barrier and blood tumor barrier. Considering these barriers, we have reviewed the strategies available to map NPs using biological techniques. The surface adsorption energy of NPs is the basic force driving NP gathering, protein corona formation, and many other interactions of NPs within biological systems. These interactions can be described using an approach named the biological surface adsorption index. A quantitative structural activity relationship study helps to understand different protein-protein or protein-ligand interactions. Moreover, equilibrium between cerebrovascular permeability is required when a drug is transferred via the circulatory system for the therapy of neurodegenerative diseases. Various drug delivery approaches, such as chemical drug delivery and carrier-mediated drug delivery, have been established to avoid different barriers inhibiting CNS penetration by therapeutic substances. Developing an improved understanding of drug receptors and the sites of drug action, together with advances in medicinal chemistry, will make it possible to design drugs with greatly enhanced activity and selectivity; this may result in a significant increase in the therapeutic index.
Collapse
Affiliation(s)
- Almas Iqbal
- a Department of Biosciences , COMSATS Institute of Information Technology , Chak Shahzad , Islamabad , Pakistan
| | | | | | | | | | | |
Collapse
|
136
|
Meyers JD, Doane T, Burda C, Basilion JP. Nanoparticles for imaging and treating brain cancer. Nanomedicine (Lond) 2013; 8:123-43. [PMID: 23256496 PMCID: PMC3564670 DOI: 10.2217/nnm.12.185] [Citation(s) in RCA: 80] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Brain cancer tumors cause disruption of the selective properties of vascular endothelia, even causing disruptions in the very selective blood-brain barrier, which are collectively referred to as the blood-brain-tumor barrier. Nanoparticles (NPs) have previously shown great promise in taking advantage of this increased vascular permeability in other cancers, which results in increased accumulation in these cancers over time due to the accompanying loss of an effective lymph system. NPs have therefore attracted increased attention for treating brain cancer. While this research is just beginning, there have been many successes demonstrated thus far in both the laboratory and clinical setting. This review serves to present the reader with an overview of NPs for treating brain cancer and to provide an outlook on what may come in the future. For NPs, just like the blood-brain-tumor barrier, the future is wide open.
Collapse
Affiliation(s)
- Joseph D Meyers
- Departments of Biomedical Engineering & Radiology, Case Western Reserve University, Cleveland, OH 44106, USA
- National Foundation for Cancer Research (NFCR), Bethesda, MD, USA
| | - Tennyson Doane
- Department of Chemistry, NFCR Center for Molecular Imaging, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Clemens Burda
- Department of Chemistry, NFCR Center for Molecular Imaging, Case Western Reserve University, Cleveland, OH 44106, USA
| | - James P Basilion
- Departments of Biomedical Engineering & Radiology, Case Western Reserve University, Cleveland, OH 44106, USA
- National Foundation for Cancer Research (NFCR), Bethesda, MD, USA
| |
Collapse
|
137
|
Ali MM, Bhuiyan MP, Janic B, Varma NR, Mikkelsen T, Ewing JR, Knight RA, Pagel MD, Arbab AS. A nano-sized PARACEST-fluorescence imaging contrast agent facilitates and validates in vivo CEST MRI detection of glioma. Nanomedicine (Lond) 2012; 7:1827-37. [PMID: 22891866 DOI: 10.2217/nnm.12.92] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
AIM The authors have investigated the usefulness of in vivo chemical exchange saturation transfer MRI for detecting gliomas using a dual-modality imaging contrast agent. MATERIALS & METHODS A paramagnetic chemical exchange saturation transfer MRI contrast agent, Eu-1,4,7,10-tetraazacclododecane-1,4,7,10-tetraacetic acid-Gly(4) and a fluorescent agent, DyLight 680, were conjugated to a generation 5 polyamidoamine dendrimer to create the dual-modality, nano-sized imaging contrast agent. RESULTS The agent was detected with in vivo chemical exchange saturation transfer MRI in an U87 glioma model. These results were validated using in vivo and ex vivo fluorescence imaging. CONCLUSION This study demonstrated the merits of using a nano-sized imaging contrast agent for detecting gliomas and using a dual-modality agent for detecting gliomas at different spatial scales.
Collapse
Affiliation(s)
- Meser M Ali
- Henry Ford Hospital, Detroit, MI 48202, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Kannan S, Dai H, Navath RS, Balakrishnan B, Jyoti A, Janisse J, Romero R, Kannan RM. Dendrimer-based postnatal therapy for neuroinflammation and cerebral palsy in a rabbit model. Sci Transl Med 2012; 4:130ra46. [PMID: 22517883 DOI: 10.1126/scitranslmed.3003162] [Citation(s) in RCA: 273] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cerebral palsy (CP) is a chronic childhood disorder with no effective cure. Neuroinflammation, caused by activated microglia and astrocytes, plays a key role in the pathogenesis of CP and disorders such as Alzheimer's disease and multiple sclerosis. Targeting neuroinflammation can be a potent therapeutic strategy. However, delivering drugs across the blood-brain barrier to the target cells for treating diffuse brain injury is a major challenge. We show that systemically administered polyamidoamine dendrimers localize in activated microglia and astrocytes in the brain of newborn rabbits with CP, but not healthy controls. We further demonstrate that dendrimer-based N-acetyl-l-cysteine (NAC) therapy for brain injury suppresses neuroinflammation and leads to a marked improvement in motor function in the CP kits. The well-known and safe clinical profile for NAC, when combined with dendrimer-based targeting, provides opportunities for clinical translation in the treatment of neuroinflammatory disorders in humans. The effectiveness of the dendrimer-NAC treatment, administered in the postnatal period for a prenatal insult, suggests a window of opportunity for treatment of CP in humans after birth.
Collapse
Affiliation(s)
- Sujatha Kannan
- Perinatology Research Branch, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Detroit, MI 48201, USA.
| | | | | | | | | | | | | | | |
Collapse
|
139
|
Sarantseva SV, Bolshakova OI, Timoshenko SI, Kolobov AA, Schwarzman AL. Dendrimer D5 is a vector for peptide transport to brain cells. Bull Exp Biol Med 2012; 150:429-31. [PMID: 22268035 DOI: 10.1007/s10517-011-1160-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Dendrimers are a new class of nonviral vectors for gene or drug transport. Dendrimer capacity to penetrate through the blood-brain barrier remaines little studied. Biotinylated polylysine dendrimer D5, similarly to human growth hormone biotinylated fragment covalently bound to D5 dendrimer, penetrates through the blood-brain barrier and accumulates in Drosophila brain after injection into the abdomen. Hence, D5 dendrimer can serve as a vector for peptide transport to brain cells.
Collapse
Affiliation(s)
- S V Sarantseva
- B. P. Konstantinov Petersburg Nuclear Physics Institute, Russian Academy of Sciences, Leningrad region, Gatchina, Russia.
| | | | | | | | | |
Collapse
|
140
|
Liu Y, Lu W. Recent advances in brain tumor-targeted nano-drug delivery systems. Expert Opin Drug Deliv 2012; 9:671-86. [DOI: 10.1517/17425247.2012.682726] [Citation(s) in RCA: 124] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
|
141
|
Zhang F, Huang X, Zhu L, Guo N, Niu G, Swierczewska M, Lee S, Xu H, Wang AY, Mohamedali KA, Rosenblum MG, Lu G, Chen X. Noninvasive monitoring of orthotopic glioblastoma therapy response using RGD-conjugated iron oxide nanoparticles. Biomaterials 2012; 33:5414-22. [PMID: 22560667 DOI: 10.1016/j.biomaterials.2012.04.032] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2012] [Accepted: 04/11/2012] [Indexed: 02/07/2023]
Abstract
Noninvasive imaging techniques have been considered important strategies in the clinic to monitor tumor early response to therapy. In the present study, we applied RGD peptides conjugated to iron oxide nanoparticles (IONP-RGD) as contrast agents in magnetic resonance imaging (MRI) to noninvasively monitor the response of a vascular disrupting agent VEGF(121)/rGel in an orthotopic glioblastoma model. RGD peptides were firstly coupled to IONPs coated with a crosslinked PEGylated amphiphilic triblock copolymer. In vitro binding assays confirmed that cellular uptake of particles was mainly dependent on the interaction between RGD and integrin α(v)β(3) of human umbilical vein endothelial cells (HUVEC). The tumor targeting of IONP-RGD was observed in an orthotopic U87 glioblastoma model. Finally, noninvasive monitoring of the tumor response to VEGF(121)/rGel therapy at early stages of treatment was successfully accomplished using IONP-RGD as a contrast agent for MRI, a superior method over common anatomical approaches which are based on tumor size measurements. This preclinical study can accelerate anticancer drug development and promote clinical translation of nanoprobes.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Radiology, Nanjing Jinling Hospital, Clinical School of Medical College of Nanjing University, Nanjing 210002, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
142
|
Vlieghe P, Khrestchatisky M. Medicinal chemistry based approaches and nanotechnology-based systems to improve CNS drug targeting and delivery. Med Res Rev 2012; 33:457-516. [PMID: 22434495 DOI: 10.1002/med.21252] [Citation(s) in RCA: 53] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
The central nervous system (CNS) is protected by various barriers, which regulate nervous tissue homeostasis and control the selective and specific uptake, efflux, and metabolism of endogenous and exogenous molecules. Among these barriers is the blood-brain barrier (BBB), a physical and physiological barrier that filters very efficiently and selectively the entry of compounds from the blood to the brain and protects nervous tissue from harmful substances and infectious agents present in the bloodstream. The BBB also prevents the entry of potential drugs. As a result, various drug targeting and delivery strategies are currently being developed to enhance the transport of drugs from the blood to the brain. Following a general introduction, we briefly overview in this review article the fundamental physiological properties of the BBB. Then, we describe current strategies to bypass the BBB (i.e., invasive methods, alternative approaches, and temporary opening) and to cross it (i.e., noninvasive approaches). This section is followed by a chapter addressing the chemical and technological solutions developed to cross the BBB. A special emphasis is given to prodrug-targeting approaches and targeted nanotechnology-based systems, two promising strategies for BBB targeting and delivery of drugs to the brain.
Collapse
Affiliation(s)
- Patrick Vlieghe
- VECT-HORUS S.A.S., Faculté de Médecine Secteur Nord, CS80011, Boulevard Pierre Dramard, 13344 Marseille Cedex 15, France.
| | | |
Collapse
|
143
|
In vivo characterization of changing blood-tumor barrier permeability in a mouse model of breast cancer metastasis: a complementary magnetic resonance imaging approach. Invest Radiol 2012; 46:718-25. [PMID: 21788908 DOI: 10.1097/rli.0b013e318226c427] [Citation(s) in RCA: 62] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
OBJECTIVES The current lack of efficacy for any chemo- or molecular therapeutic in the treatment of brain metastases is thought to be due, in part, to the heterogeneous permeability of the blood-brain-barrier (BBB). Little is known about how heterogeneous permeability develops, or how it varies among individual metastases. Understanding the BBB's role in metastasis will be crucial to the development of new, more effective therapies. In this article, we developed the first magnetic resonance imaging-based strategy to detect and measure the volumes of BBB permeable and nonpermeable metastases and studied the development of altered BBB permeability in metastases in vivo, over time in a mouse model of breast cancer metastasis to the brain. MATERIALS AND METHODS Animals bearing human experimental brain metastases of breast cancer (231-BR cells) were imaged, using 3-dimensional balanced steady-state free precession to visualize total metastases, and contrast-enhanced T1-weighted spin echo with gadopentetic acid (Gd-DTPA) to visualize which of these displayed contrast enhancement, as Gd-DTPA leakage is indicative of altered BBB permeability. RESULTS Metastases detected 20 days after injection showed no Gd-DTPA enhancement. At day 25, 6.1% ± 6.3% (mean ± standard deviation) of metastases enhanced, and by day 30, 28.1% ± 14.2% enhanced (P < 0.05). Enhancing metastases (mid: 0.14 ± 0.18 mm, late: 0.24 ± 0.32 mm) had larger volumes than nonenhancing (mid: 0.04 ± 0.04 mm, late: 0.09 ± 0.09 mm, P < 0.05); however, there was no significant difference between the growth rates of the 2. CONCLUSIONS A significant number of brain metastases were uniformly nonpermeable, which highlights the need for developing treatment strategies that can overcome the permeability of the BBB. The model developed herein can provide the basis for in vivo evaluation of both BBB permeable and nonpermeable metastases response to therapy.
Collapse
|
144
|
Irinophore C™, a lipid-based nanoparticulate formulation of irinotecan, is more effective than free irinotecan when used to treat an orthotopic glioblastoma model. J Control Release 2012; 158:34-43. [DOI: 10.1016/j.jconrel.2011.09.095] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2011] [Accepted: 09/27/2011] [Indexed: 11/24/2022]
|
145
|
Yan H, Wang L, Wang J, Weng X, Lei H, Wang X, Jiang L, Zhu J, Lu W, Wei X, Li C. Two-order targeted brain tumor imaging by using an optical/paramagnetic nanoprobe across the blood brain barrier. ACS NANO 2012; 6:410-420. [PMID: 22148835 DOI: 10.1021/nn203749v] [Citation(s) in RCA: 142] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Surgical resection is a mainstay of brain tumor treatments. However, the completed excision of malignant brain tumor is challenged by its infiltrative nature. Contrast enhanced magnetic resonance imaging is widely used for defining brain tumor in clinic. However its ability in tumor visualization is hindered by the transient circulation lifetime, nontargeting specificity, and poor blood brain barrier (BBB) permeability of the commercially available MR contrast agents. In this work, we developed a two-order targeted nanoprobe in which MR/optical imaging reporters, tumor vasculature targeted cyclic [RGDyK] peptides, and BBB-permeable Angiopep-2 peptides are labeled on the PAMAM-G5 dendrimer. This nanoprobe is supposed to first target the α(V)β(3) integrin on tumor vasculatures. Increased local concentration of nanoprobe facilitates the association between BBB-permeable peptides and the low-density lipoprotein receptor-related protein (LRP) receptors on the vascular endothelial cells, which further accelerates BBB transverse of the nanoprobe via LRP receptor-mediated endocytosis. The nanoprobes that have penetrated the BBB secondly target the brain tumor because both α(V)β(3) integrin and LRP receptor are highly expressed on the tumor cells. In vivo imaging studies demonstrated that this nanoprobe not only efficiently crossed intact BBB in normal mice, but also precisely delineated the boundary of the orthotropic U87MG human glioblastoma xenograft with high target to background signal ratio. Overall, this two-order targeted nanoprobe holds the promise to noninvasively visualize brain tumors with uncompromised BBB and provides the possibility for real-time optical-image-guided brain tumor resection during surgery.
Collapse
Affiliation(s)
- Huihui Yan
- Department of Gastroenterology, Zhongshan Hospital affiliated with Fudan University, 180 Fenglin Road, Shanghai 200032, China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
146
|
Kenzaoui BH, Bernasconi CC, Hofmann H, Juillerat-Jeanneret L. Evaluation of uptake and transport of ultrasmall superparamagnetic iron oxide nanoparticles by human brain-derived endothelial cells. Nanomedicine (Lond) 2012; 7:39-53. [DOI: 10.2217/nnm.11.85] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Aim: Ultrasmall superparamagnetic iron oxide nanoparticles (USPIO-NPs) are under development for imaging and drug delivery; however, their interaction with human blood–brain barrier models is not known. Materials & Methods: The uptake, reactive oxygen species production and transport of USPIO-NPs across human brain-derived endothelial cells as models of the blood–brain tumor barrier were evaluated for either uncoated, oleic acid-coated or polyvinylamine-coated USPIO-NPs. Results: Reactive oxygen species production was observed for oleic acid-coated and polyvinylamine-coated USPIO-NPs. The uptake and intracellular localization of the iron oxide core of the USPIO-NPs was confirmed by transmission electron microscopy. However, while the uptake of these USPIO-NPs by cells was observed, they were neither released by nor transported across these cells even in the presence of an external dynamic magnetic field. Conclusion: USPIO-NP-loaded filopodia were observed to invade the polyester membrane, suggesting that they can be transported by migrating angiogenic brain-derived endothelial cells.
Collapse
Affiliation(s)
- Blanka Halamoda Kenzaoui
- Centre Hospitalier Universitaire Vaudois (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | | | - Heinrich Hofmann
- Laboratory of Powder Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | | |
Collapse
|
147
|
Iezzi R, Guru BR, Glybina IV, Mishra MK, Kennedy A, Kannan RM. Dendrimer-based targeted intravitreal therapy for sustained attenuation of neuroinflammation in retinal degeneration. Biomaterials 2012; 33:979-88. [DOI: 10.1016/j.biomaterials.2011.10.010] [Citation(s) in RCA: 119] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2011] [Accepted: 10/06/2011] [Indexed: 10/15/2022]
|
148
|
Kudgus RA, Bhattacharya R, Mukherjee P. Cancer nanotechnology: emerging role of gold nanoconjugates. Anticancer Agents Med Chem 2011; 11:965-73. [PMID: 21864234 PMCID: PMC4684088 DOI: 10.2174/187152011797927652] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2011] [Revised: 07/04/2011] [Accepted: 07/04/2011] [Indexed: 01/20/2023]
Abstract
Over the last few decades, the study of nanotechnology has grown exponentially. Nanotechnology bridges science, engineering and technology; it continues to expand in definition as well as practice. One sub-set of nanotechnology is bionanotechnology, this will be the focus of this review. Currently, bionanotechnology is being studied and exploited for utility within medicinal imaging, diagnosis and therapy in regard to cancer. Cancer is a world-wide health problem and the implication rate as well as the death rate increase year to year. However promising work is being done with gold nanoparticles for detection, diagnosis and targeted drug delivery therapy. Gold nanoparticles can be synthesized in various shapes and sizes, which directly correlates to the color; they can also be manipulated to carry various antibody, protein, plasmid, DNA or small molecule drug. Herein we summarize some of the very influential research being done in the field of Cancer Nanotechnology with an emphasis on gold nanoparticles.
Collapse
Affiliation(s)
- Rachel A. Kudgus
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Resham Bhattacharya
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| | - Priyabrata Mukherjee
- Department of Biochemistry and Molecular Biology, College of Medicine, Mayo Clinic, Rochester, Minnesota 55905, USA
| |
Collapse
|
149
|
Fu Y, An N, Li K, Zheng Y, Liang A. Chlorotoxin-conjugated nanoparticles as potential glioma-targeted drugs. J Neurooncol 2011; 107:457-62. [DOI: 10.1007/s11060-011-0763-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2011] [Accepted: 11/08/2011] [Indexed: 10/15/2022]
|
150
|
De M, Chou SS, Joshi HM, Dravid VP. Hybrid magnetic nanostructures (MNS) for magnetic resonance imaging applications. Adv Drug Deliv Rev 2011; 63:1282-99. [PMID: 21851844 DOI: 10.1016/j.addr.2011.07.001] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2011] [Revised: 06/29/2011] [Accepted: 07/02/2011] [Indexed: 12/13/2022]
Abstract
The development of MRI contrast agents has experienced its version of the gilded age over the past decade, thanks largely to the rapid advances in nanotechnology. In addition to progress in single mode contrast agents, which ushered in unprecedented R(1) or R(2) sensitivities, there has also been a boon in the development of agents covering more than one mode of detection. These include T(1)-PET, T(2)-PET T(1)-optical, T(2)-optical, T(1)-T(2) agents and many others. In this review, we describe four areas which we feel have experienced particular growth due to nanotechnology, specifically T(2) magnetic nanostructure development, T(1)/T(2)-optical dual mode agents, and most recently the T(1)-T(2) hybrid imaging systems. In each of these systems, we describe applications including in vitro, in vivo usage and assay development. In all, while the benefits and drawbacks of most MRI contrast agents depend on the application at hand, the recent development in multimodal nanohybrids may curtail the shortcomings of single mode agents in diagnostic and clinical settings by synergistically incorporating functionality. It is hoped that as nanotechnology advances over the next decade, it will produce agents with increased diagnostics and assay relevant capabilities in streamlined packages that can meaningfully improve patient care and prognostics. In this review article, we focus on T(2) materials, its surface functionalization and coupling with optical and/or T(1) agents.
Collapse
|