151
|
Hinow P, Radunskaya A, Mackay SM, Reynolds JNJ, Schroeder M, Tan EW, Tucker IG. Signaled drug delivery and transport across the blood-brain barrier. J Liposome Res 2015; 26:233-45. [PMID: 26572864 DOI: 10.3109/08982104.2015.1102277] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
We use a mathematical model to describe the delivery of a drug to a specific region of the brain. The drug is carried by liposomes that can release their cargo by application of focused ultrasound (US). Thereupon, the drug is absorbed through the endothelial cells that line the brain capillaries and form the physiologically important blood-brain barrier (BBB). We present a compartmental model of a capillary that is able to capture the complex binding and transport processes the drug undergoes in the blood plasma and at the BBB. We apply this model to the delivery of levodopa (L-dopa, used to treat Parkinson's disease) and doxorubicin (an anticancer agent). The goal is to optimize the delivery of drug while at the same time minimizing possible side effects of the US.
Collapse
Affiliation(s)
- Peter Hinow
- a Department of Mathematical Sciences , University of Wisconsin , Milwaukee , WI , USA
| | - Ami Radunskaya
- b Department of Mathematics , Pomona College , Claremont , CA , USA
| | - Sean M Mackay
- c Department of Chemistry , University of Otago , Dunedin , New Zealand
| | - John N J Reynolds
- d Department of Anatomy and the Brain Health Research Centre , University of Otago , Dunedin , New Zealand
| | - Morgan Schroeder
- e Department of Biology , University of Oregon , Eugene , OR , USA , and
| | - Eng Wui Tan
- c Department of Chemistry , University of Otago , Dunedin , New Zealand
| | - Ian G Tucker
- f New Zealand's National School of Pharmacy, University of Otago , Dunedin , New Zealand
| |
Collapse
|
152
|
Optical coherence tomography-guided laser microsurgery for blood coagulation with continuous-wave laser diode. Sci Rep 2015; 5:16739. [PMID: 26568136 PMCID: PMC4645164 DOI: 10.1038/srep16739] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2015] [Accepted: 10/19/2015] [Indexed: 12/18/2022] Open
Abstract
Blood coagulation is the clotting and subsequent dissolution of the clot following repair to the damaged tissue. However, inducing blood coagulation is difficult for some patients with homeostasis dysfunction or during surgery. In this study, we proposed a method to develop an integrated system that combines optical coherence tomography (OCT) and laser microsurgery for blood coagulation. Also, an algorithm for positioning of the treatment location from OCT images was developed. With OCT scanning, 2D/3D OCT images and angiography of tissue can be obtained simultaneously, enabling to noninvasively reconstruct the morphological and microvascular structures for real-time monitoring of changes in biological tissues during laser microsurgery. Instead of high-cost pulsed lasers, continuous-wave laser diodes (CW-LDs) with the central wavelengths of 450 nm and 532 nm are used for blood coagulation, corresponding to higher absorption coefficients of oxyhemoglobin and deoxyhemoglobin. Experimental results showed that the location of laser exposure can be accurately controlled with the proposed approach of imaging-based feedback positioning. Moreover, blood coagulation can be efficiently induced by CW-LDs and the coagulation process can be monitored in real-time with OCT. This technology enables to potentially provide accurate positioning for laser microsurgery and control the laser exposure to avoid extra damage by real-time OCT imaging.
Collapse
|
153
|
Chu PC, Liu HL, Lai HY, Lin CY, Tsai HC, Pei YC. Neuromodulation accompanying focused ultrasound-induced blood-brain barrier opening. Sci Rep 2015; 5:15477. [PMID: 26490653 PMCID: PMC4614673 DOI: 10.1038/srep15477] [Citation(s) in RCA: 77] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2015] [Accepted: 09/25/2015] [Indexed: 01/07/2023] Open
Abstract
Burst-mode focused ultrasound (FUS) induces microbubble cavitation in the vasculature and temporarily disrupts the blood-brain barrier (BBB) to enable therapeutic agent delivery. However, it remains unclear whether FUS-induced BBB opening is accompanied by neuromodulation. Here we characterized the functional effects of FUS-induced BBB opening by measuring changes in somatosensory evoked potentials (SSEPs) and blood-oxygen-level dependent (BOLD) responses. Rats underwent burst-mode FUS (mechanical index (MI) of 0.3, 0.55 or 0.8) to the forelimb region in the left primary somatosensory cortex to induce BBB opening. Longitudinal measurements were followed for up to 1 week to characterize the temporal dynamics of neuromodulation. We observed that 0.8-MI FUS profoundly suppressed SSEP amplitude and prolonged latency, and this effect lasted 7 days. 0.55-MI FUS resulted in minimal and short-term suppression of SSEP for less than 60 minutes and didn’t affect latency. BOLD responses were also suppressed in an MI-dependent manner, mirroring the effect on SSEPs. Furthermore, repetitive delivery of 0.55-MI FUS every 3 days elicited no accumulative effects on SSEPs or tissue integrity. This is the first evidence that FUS-induced BBB opening is accompanied by reversible changes in neuron responses, and may provide valuable insight toward the development of FUS-induced BBB opening for clinical applications.
Collapse
Affiliation(s)
- Po-Chun Chu
- Department of Electrical Engineering, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan Tao-Yuan, Taiwan, 333
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan Tao-Yuan, Taiwan, 333.,Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, Taiwan 333
| | - Hsin-Yi Lai
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, 5 Fu-shin Street, Kwei-Shan, Tao-Yuan, Taiwan, 333, R.O.C
| | - Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, Taiwan 333
| | - Hong-Chieh Tsai
- School of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan Tao-Yuan, Taiwan, 333, R.O.C.,Department of Neurosurgery, Chang Gung Memorial Hospital, 5 Fu-shin Street, Kwei-Shan, Tao-Yuan, Taiwan, 333, R.O.C
| | - Yu-Cheng Pei
- Department of Physical Medicine and Rehabilitation, Chang Gung Memorial Hospital, 5 Fu-shin Street, Kwei-Shan, Tao-Yuan, Taiwan, 333, R.O.C.,School of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kwei-Shan Tao-Yuan, Taiwan, 333, R.O.C
| |
Collapse
|
154
|
Dilnawaz F, Sahoo SK. Therapeutic approaches of magnetic nanoparticles for the central nervous system. Drug Discov Today 2015; 20:1256-64. [DOI: 10.1016/j.drudis.2015.06.008] [Citation(s) in RCA: 49] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2015] [Revised: 05/20/2015] [Accepted: 06/11/2015] [Indexed: 11/26/2022]
|
155
|
Promising approaches to circumvent the blood-brain barrier: progress, pitfalls and clinical prospects in brain cancer. Ther Deliv 2015; 6:989-1016. [PMID: 26488496 DOI: 10.4155/tde.15.48] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Brain drug delivery is a major challenge for therapy of central nervous system (CNS) diseases. Biochemical modifications of drugs or drug nanocarriers, methods of local delivery, and blood-brain barrier (BBB) disruption with focused ultrasound and microbubbles are promising approaches which enhance transport or bypass the BBB. These approaches are discussed in the context of brain cancer as an example in CNS drug development. Targeting to receptors enabling transport across the BBB offers noninvasive delivery of small molecule and biological cancer therapeutics. Local delivery methods enable high dose delivery while avoiding systemic exposure. BBB disruption with focused ultrasound and microbubbles offers local and noninvasive treatment. Clinical trials show the prospects of these technologies and point to challenges for the future.
Collapse
|
156
|
Lin CY, Hsieh HY, Pitt WG, Huang CY, Tseng IC, Yeh CK, Wei KC, Liu HL. Focused ultrasound-induced blood-brain barrier opening for non-viral, non-invasive, and targeted gene delivery. J Control Release 2015; 212:1-9. [PMID: 26071631 DOI: 10.1016/j.jconrel.2015.06.010] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2015] [Revised: 05/28/2015] [Accepted: 06/09/2015] [Indexed: 12/22/2022]
Abstract
Focused ultrasound (FUS) exposure in the presence of microbubbles can temporally open the blood-brain barrier (BBB) and is an emerging technique for non-invasive brain therapeutic agent delivery. Given the potential to deliver large molecules into the CNS via this technique, we propose a reliable strategy to synergistically apply FUS-BBB opening for the non-invasive and targeted delivery of non-viral genes into the CNS for therapeutic purpose. In this study, we developed a gene-liposome system, in which the liposomes are designed to carry plasmid DNA (pDNA, containing luciferase reporter gene) to form a liposomal-plasmid DNA (LpDNA) complex. Pulsed FUS exposure was delivered to induce BBB opening (500-kHz, burst length=10ms, 1% duty cycle, PRF=1Hz). The longitudinal expression of luciferase was quantitated via an in vivo imaging system (IVIS). The reporter gene expression level was confirmed via immunoblotting, and histological staining was used to identify transfected cells via fluorescent microscopy. In a comparison of gene transduction efficiency, the LpDNA system showed better cell transduction than the pDNA system. With longitudinal observation of IVIS monitoring, animals with FUS treatment showed significant promotion of LpDNA release into the CNS and demonstrated enhanced expression of genes upon sonication with FUS-BBB opening, while both the luciferase and GDNF protein expression were successfully measured via Western blotting. The gene expression peak was observed at day 2, and the gene expression level was up to 5-fold higher than that in the untreated hemisphere (compared to a 1-fold increase in the direct-inject positive-control group). The transfection efficiency was also found to be LpDNA dose-dependent, where higher payloads of pDNA resulted in a higher transfection rate. Immunoblotting and histological staining confirmed the expression of reporter genes in glial cells as well as astrocytes. This study suggests that IV administration of LpDNA in combination with FUS-BBB opening can provide effective gene delivery and expression in the CNS, demonstrating the potential to achieve non-invasive and targeted gene delivery for treatment of CNS diseases.
Collapse
Affiliation(s)
- Chung-Yin Lin
- Medical Imaging Research Center, Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan 333, Taiwan
| | - Han-Yi Hsieh
- Department of Electrical Engineering, Chang Gung University, Taoyuan 333, Taiwan
| | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT 84602, United States
| | - Chiung-Yin Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - I-Chou Tseng
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu 300, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang Gung University, Taoyuan 333, Taiwan.
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang Gung University, Taoyuan 333, Taiwan; Health Aging Research Center, Chang Gung University, Taoyuan 333, Taiwan.
| |
Collapse
|
157
|
Aryal M, Park J, Vykhodtseva N, Zhang YZ, McDannold N. Enhancement in blood-tumor barrier permeability and delivery of liposomal doxorubicin using focused ultrasound and microbubbles: evaluation during tumor progression in a rat glioma model. Phys Med Biol 2015; 60:2511-27. [PMID: 25746014 DOI: 10.1088/0031-9155/60/6/2511] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Effective drug delivery to brain tumors is often challenging because of the heterogeneous permeability of the 'blood tumor barrier' (BTB) along with other factors such as increased interstitial pressure and drug efflux pumps. Focused ultrasound (FUS) combined with microbubbles can enhance the permeability of the BTB in brain tumors, as well as the blood-brain barrier in the surrounding tissue. In this study, dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was used to characterize the FUS-induced permeability changes of the BTB in a rat glioma model at different times after implantation. 9L gliosarcoma cells were implanted in both hemispheres in male rats. At day 9, 14, or 17 days after implantation, FUS-induced BTB disruption using 690 kHz ultrasound and definity microbubbles was performed in one tumor in each animal. Before FUS, liposomal doxorubicin was administered at a dose of 5.67 mg kg(-1). This chemotherapy agent was previously shown to improve survival in animal glioma models. The transfer coefficient Ktrans describing extravasation of the MRI contrast agent Gd-DTPA was measured via DCE-MRI before and after sonication. We found that tumor doxorubicin concentrations increased monotonically (823 ± 600, 1817 ± 732 and 2432 ± 448 ng g(-1)) in the control tumors at 9, 14 and 17 d. With FUS-induced BTB disruption, the doxorubicin concentrations were enhanced significantly (P < 0.05, P < 0.01, and P < 0.0001 at days 9, 14, and 17, respectively) and were greater than the control tumors by a factor of two or more (2222 ± 784, 3687 ± 796 and 5658 ± 821 ng g(-1)) regardless of the stage of tumor growth. The transfer coefficient Ktrans was significantly (P < 0.05) enhanced compared to control tumors only at day 9 but not at day 14 or 17. These results suggest that FUS-induced enhancements in tumor drug delivery are relatively consistent over time, at least in this tumor model. These results are encouraging for the use of large drug carriers, as they suggest that even large/late-stage tumors can benefit from FUS-induced drug enhancement. Corresponding enhancements in Ktrans were found to be variable in large/late-stage tumors and not significantly different than controls, perhaps reflecting the size mismatch between the liposomal drug (~100 nm) and Gd-DTPA (molecular weight: 938 Da; hydrodynamic diameter: ≃2 nm). It may be necessary to use a larger MRI contrast agent to effectively evaluate the sonication-induced enhanced permeabilization in large/late-stage tumors when a large drug carrier such as a liposome is used.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Physics, Boston College, 221 Longwood Avenue, Boston, MA 02115, USA. Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | |
Collapse
|
158
|
Aryal M, Vykhodtseva N, Zhang YZ, McDannold N. Multiple sessions of liposomal doxorubicin delivery via focused ultrasound mediated blood-brain barrier disruption: a safety study. J Control Release 2015; 204:60-9. [PMID: 25724272 DOI: 10.1016/j.jconrel.2015.02.033] [Citation(s) in RCA: 92] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 02/20/2015] [Accepted: 02/21/2015] [Indexed: 12/11/2022]
Abstract
Transcranial MRI-guided focused ultrasound is a rapidly advancing method for delivering therapeutic and imaging agents to the brain. It has the ability to facilitate the passage of therapeutics from the vasculature to the brain parenchyma, which is normally protected by the blood-brain barrier (BBB). The method's main advantages are that it is both targeted and noninvasive, and that it can be easily repeated. Studies have shown that liposomal doxorubicin (Lipo-DOX), a chemotherapy agent with promise for tumors in the central nervous system, can be delivered into the brain across BBB. However, prior studies have suggested that doxorubicin can be significantly neurotoxic, even at small concentrations. Here, we studied whether multiple sessions of Lipo-DOX administered after FUS-induced BBB disruption (FUS-BBBD) induces severe adverse events in the normal brain tissues. First, we used fluorometry to measure the doxorubicin concentrations in the brain after FUS-BBBD to ensure that a clinically relevant doxorubicin concentration was achieved in the brain. Next, we performed three weekly sessions with FUS-BBBD±Lipo-DOX administration. Five to twelve targets were sonicated each week, following a schedule described previously in a survival study in glioma-bearing rats (Aryal et al., 2013). Five rats received three weekly sessions where i.v. injected Lipo-DOX was combined with FUS-BBBD; an additional four rats received FUS-BBBD only. Animals were euthanized 70days from the first session and brains were examined in histology. We found that clinically-relevant concentrations of doxorubicin (4.8±0.5μg/g) were delivered to the brain with the sonication parameters (0.69MHz; 0.55-0.81MPa; 10ms bursts; 1Hz PRF; 60s duration), microbubble concentration (Definity, 10μl/kg), and the administered Lipo-DOX dose (5.67mg/kg) used. The resulting concentration of Lipo-DOX was reduced by 32% when it was injected 10min after the last sonication compared to cases where the agent was delivered before sonication. In histology, the severe neurotoxicity observed in some previous studies with doxorubicin by other investigators was not observed here. However, four of the five rats who received FUS-BBBD and Lipo-DOX had regions (dimensions: 0.5-2mm) at the focal targets with evidence of minor prior damage, either a small scar (n=4) or a small cyst (n=1). The focal targets were unaffected in rats who received FUS-BBBD alone. The result indicates that while delivery of Lipo-DOX to the rat brain might result in minor damage, the severe neurotoxicity seen in earlier works does not appear to occur with delivery via FUS-BBB disruption. The damage may be related to capillary damage produced by inertial cavitation, which might have resulted in excessive doxorubicin concentrations in some areas.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, USA.
| | - Natalia Vykhodtseva
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, USA
| | - Yong-Zhi Zhang
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, USA
| | - Nathan McDannold
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, USA
| |
Collapse
|
159
|
Wang XY, Lei R, Huang HD, Wang N, Yuan L, Xiao RY, Bai LD, Li X, Li LM, Yang XD. The permeability and transport mechanism of graphene quantum dots (GQDs) across the biological barrier. NANOSCALE 2015; 7:2034-41. [PMID: 25553649 DOI: 10.1039/c4nr04136d] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
As an emerging nanomaterial, graphene quantum dots (GQDs) have shown enormous potential in theranostic applications. However, many aspects of the biological properties of GQDs require further clarification. In the present work, we prepared two sizes of GQDs and for the first time investigated their membrane permeabilities, one of the key factors of all biomedical applications, and transport mechanisms on a Madin Darby Canine Kidney (MDCK) cell monolayer. The experimental results revealed that under ∼300 mg L(-1), GQDs were innoxious to MDCK and did not affect the morphology and integrity of the cell monolayer. The Papp values were determined to be 1-3 × 10(-6) cm s(-1) for the 12 nm GQDs and 0.5-1.5 × 10(-5) cm s(-1) for the 3 nm GQDs, indicating that the 3 nm GQDs are well-transported species while the 12 nm GQDs have a moderate membrane permeability. The transport and uptake of GQDs by MDCK cells were both time and concentration-dependent. Moreover, the incubation of cells with GQDs enhanced the formation of lipid rafts, while inhibition of lipid rafts with methyl-β-cyclodextrin almost eliminated the membrane transport of GQDs. Overall, the experimental results suggested that GQDs cross the MDCK cell monolayer mainly through a lipid raft-mediated transcytosis. The present work has indicated that GQDs are a novel, low-toxic, highly-efficient general carrier for drugs and/or diagnostic agents in biomedical applications.
Collapse
Affiliation(s)
- Xin-Yi Wang
- State Key laboratories of Natural and Mimetic Drugs and Department of Chemical Biology, School of Pharmaceutical Sciences, Peking University, Beijing 100083, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
160
|
Zhang Y, Aubry JF, Zhang J, Wang Y, Roy J, Mata JF, Miller W, Dumont E, Xie M, Lee K, Zuo Z, Wintermark M. Defining the optimal age for focal lesioning in a rat model of transcranial HIFU. ULTRASOUND IN MEDICINE & BIOLOGY 2015; 41:449-55. [PMID: 25542495 DOI: 10.1016/j.ultrasmedbio.2014.09.029] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 09/22/2014] [Accepted: 09/22/2014] [Indexed: 05/17/2023]
Abstract
This study aimed at determining the optimal age group for high-intensity focused ultrasound (HIFU) experiments for producing lesions in rats. Younger rats have thinner skulls, allowing for the acoustic waves to propagate easily through the skull without causing burns of the skin and brain surface. Younger rats however, have a smaller brain that can make HIFU focusing in the brain parenchyma challenging because of the focus size. In this study, we conducted transcranial HIFU sonications in rat pups of different ages (from 9 to 43 d) with a 1.5MHz MR compatible transducer. The electric power was selected to always reach a target temperature of at least 50°C in the parenchyma. The thickness of the skull and of the brain parenchyma was measured using T2-weighted MR imaging. Results showed that the thickness of the brain parenchyma increased quickly from P9 to P12, reaching 8.5 mm at P16, and then increasing gradually along with age. The skull thickness increased gradually from P9 to P26, and then more quickly after P30. The ratio between brain parenchyma thickness and skull thickness decreased gradually with age. For the pups at 30 d, the temperature in the brain tissue adjacent to the skull increased to 48.9°C, and those from the rodents older than 33 d reached 60°C or higher, which can produce undesired irreversible damage in this location. We conclude that young rats aged 16-26 d are optimal for experiments producing transcranial HIFU lesions in rats with an intact skull.
Collapse
Affiliation(s)
- Yanrong Zhang
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China; Neuroradiology Division, Department of Radiology, University of Virginia, Charlottesville, VA, USA
| | - Jean-François Aubry
- Department of Radiation Oncology, University of Virginia, Charlottesville, VA, USA; ESPCI ParisTech, PSL Research University, Institut Langevin, Paris, France; CNRS, Institut Langevin, Paris, France; INSERM, Institut Langevin, Paris, France
| | - Junfeng Zhang
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA; Department of Anesthesiology, Shanghai Sixth Hospital, Shanghai Jiaotong University, Shanghai, China
| | - Yi Wang
- Departments of Neuroscience and Neurologic Surgery, University of Virginia, Charlottesville, VA, USA
| | - Jack Roy
- Department of Radiology, University of Virginia, Charlottesville, VA, USA
| | - Jaime F Mata
- Department of Radiology, University of Virginia, Charlottesville, VA, USA
| | - Wilson Miller
- Department of Radiology, University of Virginia, Charlottesville, VA, USA
| | | | - Mingxing Xie
- Department of Ultrasound, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Kevin Lee
- Departments of Neuroscience and Neurologic Surgery, University of Virginia, Charlottesville, VA, USA
| | - Zhiyi Zuo
- Department of Anesthesiology, University of Virginia, Charlottesville, VA, USA
| | - Max Wintermark
- Neuroradiology Division, Department of Radiology, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
161
|
Shakeri-Zadeh A, Khoee S, Shiran MB, Sharifi AM, Khoei S. Synergistic effects of magnetic drug targeting using a newly developed nanocapsule and tumor irradiation by ultrasound on CT26 tumors in BALB/c mice. J Mater Chem B 2015; 3:1879-1887. [PMID: 32262260 DOI: 10.1039/c4tb01708k] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The aim of the current study was to magnetically target the 5-fluorouracil (5-Fu) loaded magnetic poly lactic-co-glycolic acid (PLGA) nanocapsules towards CT26 colon tumor model in BALB/c mice. In addition, we ultrasonicated the tumors impregnated by nanocapsules with the goal of aiding them in magnetic drug targeting (MDT) procedure. Newly synthesized 5-Fu-loaded PLGA magnetic nanocapsules were characterized. Various treatment modalities with the use of nanocapsules, magnetic fields, and ultrasound were applied to the tumors and appropriate controls were considered. Magnetic resonance imaging (MRI) and Prussian blue (PB) staining were performed to analyze the distribution of nanocapsules within the CT26 tumor. Finally, anti-tumor and pro-apoptotic effects of each treatment modality on CT26 tumors were investigated. The effective diameter of nanocapsules was approximately 70 nm. The histological staining of the tumor tissue with PB as well as MRI revealed a broad distribution of magnetic nanocapsules within the tumor and confirmed the targeting of nanocapsules to the tumors. Anti-tumor studies demonstrated that the combination of nanocapsules-MDT-ultrasound effectively inhibits the growth of CT26 tumors compared with injection of 5-Fu alone (P < 0.01). The present study exhibits potentials of the newly synthesized magnetic nanocapsule and suggests that the combination of MDT and ultrasound might help this new nanotechnology-based cancer chemotherapy agent in vivo.
Collapse
Affiliation(s)
- Ali Shakeri-Zadeh
- Medical Physics Department, School of Medicine, Iran University of Medical Sciences (IUMS), Associate Professor of Biophysics, Razi Drug Research Centre, P.O. Box: 14155-5983, Tehran, Iran.
| | | | | | | | | |
Collapse
|
162
|
Nacev A, Weinberg IN, Stepanov PY, Kupfer S, Mair LO, Urdaneta MG, Shimoji M, Fricke ST, Shapiro B. Dynamic inversion enables external magnets to concentrate ferromagnetic rods to a central target. NANO LETTERS 2015; 15:359-64. [PMID: 25457292 PMCID: PMC4296920 DOI: 10.1021/nl503654t] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2014] [Revised: 11/30/2014] [Indexed: 05/26/2023]
Abstract
The ability to use magnets external to the body to focus therapy to deep tissue targets has remained an elusive goal in magnetic drug targeting. Researchers have hitherto been able to manipulate magnetic nanotherapeutics in vivo with nearby magnets but have remained unable to focus these therapies to targets deep within the body using magnets external to the body. One of the factors that has made focusing of therapy to central targets between magnets challenging is Samuel Earnshaw's theorem as applied to Maxwell's equations. These mathematical formulations imply that external static magnets cannot create a stable potential energy well between them. We posited that fast magnetic pulses could act on ferromagnetic rods before they could realign with the magnetic field. Mathematically, this is equivalent to reversing the sign of the potential energy term in Earnshaw's theorem, thus enabling a quasi-static stable trap between magnets. With in vitro experiments, we demonstrated that quick, shaped magnetic pulses can be successfully used to create inward pointing magnetic forces that, on average, enable external magnets to concentrate ferromagnetic rods to a central location.
Collapse
Affiliation(s)
- A. Nacev
- Weinberg Medical Physics LLC, 5611
Roosevelt St, Bethesda, Maryland 20817, United States
| | - I. N. Weinberg
- Weinberg Medical Physics LLC, 5611
Roosevelt St, Bethesda, Maryland 20817, United States
| | - P. Y. Stepanov
- Weinberg Medical Physics LLC, 5611
Roosevelt St, Bethesda, Maryland 20817, United States
| | - S. Kupfer
- Weinberg Medical Physics LLC, 5611
Roosevelt St, Bethesda, Maryland 20817, United States
| | - L. O. Mair
- Weinberg Medical Physics LLC, 5611
Roosevelt St, Bethesda, Maryland 20817, United States
| | - M. G. Urdaneta
- Weinberg Medical Physics LLC, 5611
Roosevelt St, Bethesda, Maryland 20817, United States
| | - M. Shimoji
- Weinberg Medical Physics LLC, 5611
Roosevelt St, Bethesda, Maryland 20817, United States
| | - S. T. Fricke
- Children’s
National Medical Center, 11 Michigan
Ave NW, Washington, DC, 20010, United States
| | - B. Shapiro
- Fischell Department of Bioengineering and the Institute for Systems
Research, University of Maryland, College Park, Maryland 20742, United States
| |
Collapse
|
163
|
Targeted Drug Delivery Systems: Strategies and Challenges. ADVANCES IN DELIVERY SCIENCE AND TECHNOLOGY 2015. [DOI: 10.1007/978-3-319-11355-5_1] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/14/2023]
|
164
|
Caltagirone C, Bettoschi A, Garau A, Montis R. Silica-based nanoparticles: a versatile tool for the development of efficient imaging agents. Chem Soc Rev 2015; 44:4645-71. [DOI: 10.1039/c4cs00270a] [Citation(s) in RCA: 107] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
In this review a selection of the most recent examples of imaging techniques applied to silica-based NPs for imaging is reported.
Collapse
Affiliation(s)
- Claudia Caltagirone
- Università degli Studi di Cagliari
- Dipartimento di Scienze Chimiche e Geologiche
- 09042 Monserrato
- Italy
| | - Alexandre Bettoschi
- Università degli Studi di Cagliari
- Dipartimento di Scienze Chimiche e Geologiche
- 09042 Monserrato
- Italy
| | - Alessandra Garau
- Università degli Studi di Cagliari
- Dipartimento di Scienze Chimiche e Geologiche
- 09042 Monserrato
- Italy
| | - Riccardo Montis
- Università degli Studi di Cagliari
- Dipartimento di Scienze Chimiche e Geologiche
- 09042 Monserrato
- Italy
| |
Collapse
|
165
|
Zhang Q, Sauter T, Wang L, Fang L, Kratz K, Lendlein A. Preparation of Magneto-Sensitive Polymer Nanocomposite Microparticles from Copolyesterurethanes via Electrospraying. ACTA ACUST UNITED AC 2014. [DOI: 10.1002/masy.201400140] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Quanchao Zhang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; 14513 Teltow Germany
- Institute of Chemistry; University Potsdam; 14476 Potsdam Germany
| | - Tilman Sauter
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; 14513 Teltow Germany
- Institute of Biochemistry and Biology; University Potsdam; 14476 Potsdam Germany
| | - Li Wang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; 14513 Teltow Germany
- Institute of Chemistry; University Potsdam; 14476 Potsdam Germany
| | - Liang Fang
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; 14513 Teltow Germany
| | - Karl Kratz
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; 14513 Teltow Germany
| | - Andreas Lendlein
- Institute of Biomaterial Science and Berlin-Brandenburg Center for Regenerative Therapies; Helmholtz-Zentrum Geesthacht; 14513 Teltow Germany
- Institute of Chemistry; University Potsdam; 14476 Potsdam Germany
- Institute of Biochemistry and Biology; University Potsdam; 14476 Potsdam Germany
| |
Collapse
|
166
|
Mahmoudi K, Hadjipanayis CG. The application of magnetic nanoparticles for the treatment of brain tumors. Front Chem 2014; 2:109. [PMID: 25520952 PMCID: PMC4253533 DOI: 10.3389/fchem.2014.00109] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 11/17/2014] [Indexed: 12/18/2022] Open
Affiliation(s)
- Keon Mahmoudi
- Georgia Institute of Technology, School of Biology Atlanta, GA, USA
| | - Costas G Hadjipanayis
- Brain Tumor Nanotechnology Laboratory, Department of Neurosurgery, Winship Cancer Institute of Emory University, Emory University School of Medicine Atlanta, GA, USA
| |
Collapse
|
167
|
Fan CH, Yeh CK. Microbubble-enhanced Focused Ultrasound-induced Blood–brain Barrier Opening for Local and Transient Drug Delivery in Central Nervous System Disease. J Med Ultrasound 2014. [DOI: 10.1016/j.jmu.2014.11.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
168
|
Abstract
High intensity focused ultrasound (HIFU), is a promising, non-invasive modality for treatment of tumours in conjunction with magnetic resonance imaging or diagnostic ultrasound guidance. HIFU is being used increasingly for treatment of prostate cancer and uterine fibroids. Over the last 10 years a growing number of clinical trials have examined HIFU treatment of both benign and malignant tumours of the liver, breast, pancreas, bone, connective tissue, thyroid, parathyroid, kidney and brain. For some of these emerging indications, HIFU is poised to become a serious alternative or adjunct to current standard treatments--including surgery, radiation, gene therapy, immunotherapy, and chemotherapy. Current commercially available HIFU devices are marketed for their thermal ablation applications. In the future, lower energy treatments may play a significant role in mediating targeted drug and gene delivery for cancer treatment. In this article we introduce currently available HIFU systems, provide an overview of clinical trials in emerging oncological targets, and briefly discuss selected pre-clinical research that is relevant to future oncological HIFU applications.
Collapse
Affiliation(s)
- Ezekiel Maloney
- Department of Radiology, University of Washington , Seattle and
| | | |
Collapse
|
169
|
Yang CH, Tsai MT, Shen SC, Ng CY, Jung SM. Feasibility of ablative fractional laser-assisted drug delivery with optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2014; 5:3949-59. [PMID: 25426321 PMCID: PMC4242029 DOI: 10.1364/boe.5.003949] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2014] [Revised: 10/07/2014] [Accepted: 10/11/2014] [Indexed: 05/20/2023]
Abstract
Fractional resurfacing creates hundreds of microscopic wounds in the skin without injuring surrounding tissue. This technique allows rapid wound healing owing to small injury regions, and has been proven as an effective method for repairing photodamaged skin. Recently, ablative fractional laser (AFL) treatment has been demonstrated to facilitate topical drug delivery into skin. However, induced fractional photothermolysis depends on several parameters, such as incident angle, exposure energy, and spot size of the fractional laser. In this study, we used fractional CO2 laser to induce microscopic ablation array on the nail for facilitating drug delivery through the nail. To ensure proper energy delivery without damaging tissue structures beneath the nail plate, optical coherence tomography (OCT) was implemented for quantitative evaluation of induced microscopic ablation zone (MAZ). Moreover, to further study the feasibility of drug delivery, normal saline was dripped on the exposure area of fingernail and the speckle variance in OCT signal was used to observe water diffusion through the ablative channels into the nail plate. In conclusion, this study establishes OCT as an effective tool for the investigation of fractional photothermolysis and water/drug delivery through microscopic ablation channels after nail fractional laser treatment.
Collapse
Affiliation(s)
- Chih-Hsun Yang
- Department of Dermatology, Chang Gung Memorial Hospital, 5 Fusing St., Kwei-Shan, Tao- Yuan, 33302,
Taiwan
- College of Medicine, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302
Taiwan
| | - Meng-Tsan Tsai
- Department of Electrical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302
Taiwan
- Graduate Institute of Electro-Optical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302
Taiwan
| | - Su-Chin Shen
- College of Medicine, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302
Taiwan
- Department of Ophthalmology, Chang Gung Memorial Hospital, 5 Fusing St. Kwei-Shan, Tao- Yuan, 33302
Taiwan
| | - Chau Yee Ng
- Department of Dermatology, Chang Gung Memorial Hospital, 5 Fusing St., Kwei-Shan, Tao- Yuan, 33302,
Taiwan
- College of Medicine, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302
Taiwan
| | - Shih-Ming Jung
- College of Medicine, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302
Taiwan
- Department of Pathology, Chang Gung Memorial Hospital, 5 Fusing St., Kwei-Shan, Tao- Yuan, 33302
Taiwan
| |
Collapse
|
170
|
Mechanisms of microbubble-facilitated sonoporation for drug and gene delivery. Ther Deliv 2014; 5:467-86. [PMID: 24856171 DOI: 10.4155/tde.14.10] [Citation(s) in RCA: 109] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
171
|
Maguire CA, Ramirez SH, Merkel SF, Sena-Esteves M, Breakefield XO. Gene therapy for the nervous system: challenges and new strategies. Neurotherapeutics 2014; 11:817-39. [PMID: 25159276 PMCID: PMC4391389 DOI: 10.1007/s13311-014-0299-5] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Current clinical treatments for central nervous system (CNS) diseases, such as Parkinson's disease and glioblastoma do not halt disease progression and have significant treatment morbidities. Gene therapy has the potential to "permanently" correct disease by bringing in a normal gene to correct a mutant gene deficiency, knocking down mRNA of mutant alleles, and inducing cell-death in cancer cells using transgenes encoding apoptosis-inducing proteins. Promising results in clinical trials of eye disease (Leber's congenital aumorosis) and Parkinson's disease have shown that gene-based neurotherapeutics have great potential. The recent development of genome editing technology, such as zinc finger nucleases, TALENS, and CRISPR, has made the ultimate goal of gene correction a step closer. This review summarizes the challenges faced by gene-based neurotherapeutics and the current and recent strategies designed to overcome these barriers. We have chosen the following challenges to focus on in this review: (1) delivery vehicles (both virus and nonviral), (2) use of promoters for vector-mediated gene expression in CNS, and (3) delivery across the blood-brain barrier. The final section (4) focuses on promising pre-clinical/clinical studies of neurotherapeutics.
Collapse
Affiliation(s)
- Casey A Maguire
- Department of Neurology, Massachusetts General Hospital, and Neuroscience Program, Harvard Medical School, Molecular Neurogenetics Unit, 13th Street, Building 149, Charlestown, MA, 02129, USA,
| | | | | | | | | |
Collapse
|
172
|
Weiser JR, Saltzman WM. Controlled release for local delivery of drugs: barriers and models. J Control Release 2014; 190:664-73. [PMID: 24801251 PMCID: PMC4142083 DOI: 10.1016/j.jconrel.2014.04.048] [Citation(s) in RCA: 117] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Revised: 04/15/2014] [Accepted: 04/25/2014] [Indexed: 01/14/2023]
Abstract
Controlled release systems are an effective means for local drug delivery. In local drug delivery, the major goal is to supply therapeutic levels of a drug agent at a physical site in the body for a prolonged period. A second goal is to reduce systemic toxicities, by avoiding the delivery of agents to non-target tissues remote from the site. Understanding the dynamics of drug transport in the vicinity of a local drug delivery device is helpful in achieving both of these goals. Here, we provide an overview of controlled release systems for local delivery and we review mathematical models of drug transport in tissue, which describe the local penetration of drugs into tissue and illustrate the factors - such as diffusion, convection, and elimination - that control drug dispersion and its ultimate fate. This review highlights the important role of controlled release science in development of reliable methods for local delivery, as well as the barriers to accomplishing effective delivery in the brain, blood vessels, mucosal epithelia, and the skin.
Collapse
Affiliation(s)
- Jennifer R Weiser
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA.
| | - W Mark Saltzman
- Department of Biomedical Engineering, Yale University, New Haven, CT, 06511, USA.
| |
Collapse
|
173
|
Wu SK, Chiang CF, Hsu YH, Lin TH, Liou HC, Fu WM, Lin WL. Short-time focused ultrasound hyperthermia enhances liposomal doxorubicin delivery and antitumor efficacy for brain metastasis of breast cancer. Int J Nanomedicine 2014; 9:4485-94. [PMID: 25278753 PMCID: PMC4178504 DOI: 10.2147/ijn.s68347] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
The blood–brain/tumor barrier inhibits the uptake and accumulation of chemotherapeutic drugs. Hyperthermia can enhance the delivery of chemotherapeutic agent into tumors. In this study, we investigated the effects of short-time focused ultrasound (FUS) hyperthermia on the delivery and therapeutic efficacy of pegylated liposomal doxorubicin (PLD) for brain metastasis of breast cancer. Murine breast cancer 4T1-luc2 cells expressing firefly luciferase were injected into female BALB/c mice striatum tissues and used as a brain metastasis model. The mice were intravenously injected with PLD (5 mg/kg) with/without 10-minute transcranial FUS hyperthermia on day 6 after tumor implantation. The amounts of doxorubicin accumulated in the normal brain tissues and tumor tissues with/without FUS hyperthermia were measured using fluorometry. The tumor growth for the control, hyperthermia, PLD, and PLD + hyperthermia groups was measured using an IVIS spectrum system every other day from day 3 to day 11. Cell apoptosis and tumor characteristics were assessed using immunohistochemistry. Short-time FUS hyperthermia was able to significantly enhance the PLD delivery into brain tumors. The tumor growth was effectively inhibited by a single treatment of PLD + hyperthermia compared with both PLD alone and short-time FUS hyperthermia alone. Immunohistochemical examination further demonstrated the therapeutic efficacy of PLD plus short-time FUS hyperthermia for brain metastasis of breast cancer. The application of short-time FUS hyperthermia after nanodrug injection may be an effective approach to enhance nanodrug delivery and improve the treatment of metastatic cancers.
Collapse
Affiliation(s)
- Sheng-Kai Wu
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, Taipei, Taiwan
| | - Chi-Feng Chiang
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, Taipei, Taiwan
| | - Yu-Hone Hsu
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, Taipei, Taiwan ; Department of Neurosurgery, Cheng-Hsin General Hospital, Taipei, Taiwan
| | - Tzu-Hung Lin
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Houng-Chi Liou
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Wen-Mei Fu
- Institute of Pharmacology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Win-Li Lin
- Institute of Biomedical Engineering, College of Medicine and College of Engineering, Taipei, Taiwan ; Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
174
|
Burgess A, Dubey S, Yeung S, Hough O, Eterman N, Aubert I, Hynynen K. Alzheimer disease in a mouse model: MR imaging-guided focused ultrasound targeted to the hippocampus opens the blood-brain barrier and improves pathologic abnormalities and behavior. Radiology 2014; 273:736-45. [PMID: 25222068 DOI: 10.1148/radiol.14140245] [Citation(s) in RCA: 200] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
PURPOSE To validate whether repeated magnetic resonance (MR) imaging-guided focused ultrasound treatments targeted to the hippocampus, a brain structure relevant for Alzheimer disease ( AD Alzheimer disease ), could modulate pathologic abnormalities, plasticity, and behavior in a mouse model. MATERIALS AND METHODS All animal procedures were approved by the Animal Care Committee and are in accordance with the Canadian Council on Animal Care. Seven-month-old transgenic (TgCRND8) (Tg) mice and their nontransgenic (non-Tg) littermates were entered in the study. Mice were treated weekly with MR imaging-guided focused ultrasound in the bilateral hippocampus (1.68 MHz, 10-msec bursts, 1-Hz burst repetition frequency, 120-second total duration). After 1 month, spatial memory was tested in the Y maze with the novel arm prior to sacrifice and immunohistochemical analysis. The data were compared by using unpaired t tests and analysis of variance with Tukey post hoc analysis. RESULTS Untreated Tg mice spent 61% less time than untreated non-Tg mice exploring the novel arm of the Y maze because of spatial memory impairments (P < .05). Following MR imaging-guided focused ultrasound, Tg mice spent 99% more time exploring the novel arm, performing as well as their non-Tg littermates. Changes in behavior were correlated with a reduction of the number and size of amyloid plaques in the MR imaging-guided focused ultrasound-treated animals (P < .01). Further, after MR imaging-guided focused ultrasound treatment, there was a 250% increase in the number of newborn neurons in the hippocampus (P < .01). The newborn neurons had longer dendrites and more arborization after MR imaging-guided focused ultrasound, as well (P < .01). CONCLUSION Repeated MR imaging-guided focused ultrasound treatments led to spatial memory improvement in a Tg mouse model of AD Alzheimer disease . The behavior changes may be mediated by decreased amyloid pathologic abnormalities and increased neuronal plasticity.
Collapse
Affiliation(s)
- Alison Burgess
- From the Physical Sciences Platform (A.B., S.Y., O.H., N.E., K.H.) and Biological Sciences Platform (S.D., I.A.), Sunnybrook Research Institute, 2075 Bayview Ave, C713, Toronto, ON, Canada M4N 3M5; Department of Laboratory Medicine and Pathobiology (S.D., I.A.) and Department of Medical Biophysics (K.H.), University of Toronto, Toronto, Ontario, Canada
| | | | | | | | | | | | | |
Collapse
|
175
|
Lu YC, Luo PC, Huang CW, Leu YL, Wang TH, Wei KC, Wang HE, Ma YH. Augmented cellular uptake of nanoparticles using tea catechins: effect of surface modification on nanoparticle-cell interaction. NANOSCALE 2014; 6:10297-10306. [PMID: 25069428 DOI: 10.1039/c4nr00617h] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Nanoparticles may serve as carriers in targeted therapeutics; interaction of the nanoparticles with a biological system may determine their targeting effects and therapeutic efficacy. Epigallocatechin-3-gallate (EGCG), a major component of tea catechins, has been conjugated with nanoparticles and tested as an anticancer agent. We investigated whether EGCG may enhance nanoparticle uptake by tumor cells. Cellular uptake of a dextran-coated magnetic nanoparticle (MNP) was determined by confocal microscopy, flow cytometry or a potassium thiocyanate colorimetric method. We demonstrated that EGCG greatly enhanced interaction and/or internalization of MNPs (with or without polyethylene glycol) by glioma cells, but not vascular endothelial cells. The enhancing effects are both time- and concentration-dependent. Such effects may be induced by a simple mix of MNPs with EGCG at a concentration as low as 1-3 μM, which increased MNP uptake 2- to 7-fold. In addition, application of magnetic force further potentiated MNP uptake, suggesting a synergetic effect of EGCG and magnetic force. Because the effects of EGCG were preserved at 4 °C, but not when EGCG was removed from the culture medium prior to addition of MNPs, a direct interaction of EGCG and MNPs was implicated. Use of an MNP-EGCG composite produced by adsorption of EGCG and magnetic separation also led to an enhanced uptake. The results reveal a novel interaction of a food component and nanocarrier system, which may be potentially amenable to magnetofection, cell labeling/tracing, and targeted therapeutics.
Collapse
Affiliation(s)
- Yi-Ching Lu
- Department of Physiology and Pharmacology, Institute of Biomedical Sciences, College of Medicine, Chang Gung University, 259 Wen-Hwa 1st Road, Kuei-Shan, Tao-Yuan 33302, Taiwan, Republic of China.
| | | | | | | | | | | | | | | |
Collapse
|
176
|
Pajek D, Hynynen K. The application of sparse arrays in high frequency transcranial focused ultrasound therapy: a simulation study. Med Phys 2014; 40:122901. [PMID: 24320540 DOI: 10.1118/1.4829510] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
PURPOSE Transcranial focused ultrasound is an emerging therapeutic modality that can be used to perform noninvasive neurosurgical procedures. The current clinical transcranial phased array operates at 650 kHz, however the development of a higher frequency array would enable more precision, while reducing the risk of standing waves. However, the smaller wavelength and the skull's increased distortion at this frequency are problematic. It would require an order of magnitude more elements to create such an array. Random sparse arrays enable steering of a therapeutic array with fewer elements. However, the tradeoffs inherent in the use of sparsity in a transcranial phased array have not been systematically investigated and so the objective of this simulation study is to investigate the effect of sparsity on transcranial arrays at a frequency of 1.5 MHz that provides small focal spots for precise exposure control. METHODS Transcranial sonication simulations were conducted using a multilayer Rayleigh-Sommerfeld propagation model. Element size and element population were varied and the phased array's ability to steer was assessed. RESULTS The focal pressures decreased proportionally as elements were removed. However, off-focus hotspots were generated if a high degree of steering was attempted with very sparse arrays. A phased array consisting of 1588 elements 3 mm in size, a 10% population, was appropriate for steering up to 4 cm in all directions. However, a higher element population would be required if near-skull sonication is desired. CONCLUSIONS This study demonstrated that the development of a sparse, hemispherical array at 1.5 MHz could enable more precision in therapies that utilize lower intensity sonications.
Collapse
Affiliation(s)
- Daniel Pajek
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario M4N 3M5, Canada and Department of Medical Biophysics, University of Toronto, Toronto, Ontario M4N3M5, Canada
| | | |
Collapse
|
177
|
Fan CH, Lin WH, Ting CY, Chai WY, Yen TC, Liu HL, Yeh CK. Contrast-enhanced ultrasound imaging for the detection of focused ultrasound-induced blood-brain barrier opening. Theranostics 2014; 4:1014-25. [PMID: 25161701 PMCID: PMC4143942 DOI: 10.7150/thno.9575] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 07/10/2014] [Indexed: 01/08/2023] Open
Abstract
The blood-brain barrier (BBB) can be transiently and locally opened by focused ultrasound (FUS) in the presence of microbubbles (MBs). Various imaging modalities and contrast agents have been used to monitor this process. Unfortunately, direct ultrasound imaging of BBB opening with MBs as contrast agent is not feasible, due to the inability of MBs to penetrate brain parenchyma. However, FUS-induced BBB opening is accompanied by changes in blood flow and perfusion, suggesting the possibility of perfusion-based ultrasound imaging. Here we evaluated the use of MB destruction-replenishment, which was originally developed for analysis of ultrasound perfusion kinetics, for verifying and quantifying FUS-induced BBB opening. MBs were intravenously injected and the BBB was disrupted by 2 MHz FUS with burst-tone exposure at 0.5-0.7 MPa. A perfusion kinetic map was estimated by MB destruction-replenishment time-intensity curve analysis. Our results showed that the scale and distribution of FUS-induced BBB opening could be determined at high resolution by ultrasound perfusion kinetic analysis. The accuracy and sensitivity of this approach was validated by dynamic contrast-enhanced MRI. Our successful demonstration of ultrasound imaging to monitor FUS-induced BBB opening provides a new approach to assess FUS-dependent brain drug delivery, with the benefit of high temporal resolution and convenient integration with the FUS device.
Collapse
|
178
|
Servant A, Leon V, Jasim D, Methven L, Limousin P, Fernandez-Pacheco EV, Prato M, Kostarelos K. Graphene-based electroresponsive scaffolds as polymeric implants for on-demand drug delivery. Adv Healthc Mater 2014; 3:1334-43. [PMID: 24799416 DOI: 10.1002/adhm.201400016] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Revised: 03/27/2014] [Indexed: 01/25/2023]
Abstract
Stimuli-responsive biomaterials have attracted significant attention in the field of polymeric implants designed as active scaffolds for on-demand drug delivery. Conventional porous scaffolds suffer from drawbacks such as molecular diffusion and material degradation, allowing in most cases only a zero-order drug release profile. The possibility of using external stimulation to trigger drug release is particularly enticing. In this paper, the fabrication of previously unreported graphene hydrogel hybrid electro-active scaffolds capable of controlled small molecule release is presented. Pristine ball-milled graphene sheets are incorporated into a three dimensional macroporous hydrogel matrix to obtain hybrid gels with enhanced mechanical, electrical, and thermal properties. These electroactive scaffolds demonstrate controlled drug release in a pulsatile fashion upon the ON/OFF application of low electrical voltages, at low graphene concentrations (0.2 mg mL(-1) ) and by maintaining their structural integrity. Moreover, the in vivo performance of these electroactive scaffolds to release drug molecules without any "resistive heating" is demonstrated. In this study, an illustration of how the heat dissipating properties of graphene can provide significant and previously unreported advantages in the design of electroresponsive hydrogels, able to maintain optimal functionality by overcoming adverse effects due to unwanted heating, is offered.
Collapse
Affiliation(s)
- Ania Servant
- Nanomedicine Lab, Faculty of Life Sciences; University College London; London WC1N 1AX UK
- Faculty of Medical & Human Sciences and National Graphene Institute; University of Manchester; M19 9PT UK
| | - Veronica Leon
- Facultad de Ciencias Químicas; Universidad Castilla La-Mancha; Ciudad Real 13071 Spain
- Dipartimento Scienze Chimiche e Farmaceutiche; University of Trieste; Piazzale Europa 1 Trieste 34127 Italy
| | - Dhifaf Jasim
- Nanomedicine Lab, Faculty of Life Sciences; University College London; London WC1N 1AX UK
- Faculty of Medical & Human Sciences and National Graphene Institute; University of Manchester; M19 9PT UK
| | - Laura Methven
- Nanomedicine Lab, Faculty of Life Sciences; University College London; London WC1N 1AX UK
- Faculty of Medical & Human Sciences and National Graphene Institute; University of Manchester; M19 9PT UK
| | - Patricia Limousin
- UCL Institute of Neurology; University College London; Queen Square London WC1N 1AX UK
| | | | - Maurizio Prato
- Dipartimento Scienze Chimiche e Farmaceutiche; University of Trieste; Piazzale Europa 1 Trieste 34127 Italy
| | - Kostas Kostarelos
- Nanomedicine Lab, Faculty of Life Sciences; University College London; London WC1N 1AX UK
- Faculty of Medical & Human Sciences and National Graphene Institute; University of Manchester; M19 9PT UK
| |
Collapse
|
179
|
Combination of ultrasound and newly synthesized magnetic nanocapsules affects the temperature profile of CT26 tumors in BALB/c mice. J Med Ultrason (2001) 2014; 42:9-16. [DOI: 10.1007/s10396-014-0558-4] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2014] [Accepted: 06/10/2014] [Indexed: 11/26/2022]
|
180
|
Shapiro B, Kulkarni S, Nacev A, Sarwar A, Preciado D, Depireux D. Shaping Magnetic Fields to Direct Therapy to Ears and Eyes. Annu Rev Biomed Eng 2014; 16:455-81. [DOI: 10.1146/annurev-bioeng-071813-105206] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Affiliation(s)
- B. Shapiro
- Fischell Department of Bioengineering,
- The Institute for Systems Research (ISR), University of Maryland, College Park, Maryland 20742;
| | | | - A. Nacev
- Fischell Department of Bioengineering,
| | - A. Sarwar
- Fischell Department of Bioengineering,
| | - D. Preciado
- Otolaryngology, Sheikh Zayed Institute for Pediatric Surgical Innovation, Children's National Medical Center, Washington, DC 20010
| | - D.A. Depireux
- The Institute for Systems Research (ISR), University of Maryland, College Park, Maryland 20742;
| |
Collapse
|
181
|
Oh KS, Han H, Yoon BD, Lee M, Kim H, Seo DW, Seo JH, Kim K, Kwon IC, Yuk SH. Effect of HIFU treatment on tumor targeting efficacy of docetaxel-loaded Pluronic nanoparticles. Colloids Surf B Biointerfaces 2014; 119:137-44. [DOI: 10.1016/j.colsurfb.2014.05.007] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Revised: 05/02/2014] [Accepted: 05/03/2014] [Indexed: 10/25/2022]
|
182
|
Anthony KJP, Murugan M, Jeyaraj M, Rathinam NK, Sangiliyandi G. Synthesis of silver nanoparticles using pine mushroom extract: A potential antimicrobial agent against E. coli and B. subtilis. J IND ENG CHEM 2014. [DOI: 10.1016/j.jiec.2013.10.008] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
183
|
Tsai MT, Chang FY, Lee CK, Gong CSA, Lin YX, Lee JD, Yang CH, Liu HL. Investigation of temporal vascular effects induced by focused ultrasound treatment with speckle-variance optical coherence tomography. BIOMEDICAL OPTICS EXPRESS 2014; 5:2009-2022. [PMID: 25071945 PMCID: PMC4102345 DOI: 10.1364/boe.5.002009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2014] [Revised: 05/28/2014] [Accepted: 05/28/2014] [Indexed: 05/30/2023]
Abstract
Focused ultrasound (FUS) can be used to locally and temporally enhance vascular permeability, improving the efficiency of drug delivery from the blood vessels into the surrounding tissue. However, it is difficult to evaluate in real time the effect induced by FUS and to noninvasively observe the permeability enhancement. In this study, speckle-variance optical coherence tomography (SVOCT) was implemented for the investigation of temporal effects on vessels induced by FUS treatment. With OCT scanning, the dynamic change in vessels during FUS exposure can be observed and studied. Moreover, the vascular effects induced by FUS treatment with and without the presence of microbubbles were investigated and quantitatively compared. Additionally, 2D and 3D speckle-variance images were used for quantitative observation of blood leakage from vessels due to the permeability enhancement caused by FUS, which could be an indicator that can be used to determine the influence of FUS power exposure. In conclusion, SVOCT can be a useful tool for monitoring FUS treatment in real time, facilitating the dynamic observation of temporal effects and helping to determine the optimal FUS power.
Collapse
Affiliation(s)
- Meng-Tsan Tsai
- Department of Electrical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302 Taiwan
- Graduate Institute of Electro-Optical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302 Taiwan
| | - Feng-Yu Chang
- Department of Electrical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302 Taiwan
| | - Cheng-Kuang Lee
- Department of Electrical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302 Taiwan
| | - Cihun-Siyong Alex Gong
- Department of Electrical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302 Taiwan
| | - Yu-Xiang Lin
- Department of Electrical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302 Taiwan
| | - Jiann-Der Lee
- Department of Electrical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302 Taiwan
| | - Chih-Hsun Yang
- Department of Dermatology, Chang Gung Memorial Hospital, 5 Fusing Street, Kwei-Shan, Tao-Yaun 33302, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, School of Electrical and Computer Engineering, College of Engineering, Chang Gung University, 259, Wen-Hwa 1st Road, Kwei-Shan, Tao-Yuan, 33302 Taiwan
| |
Collapse
|
184
|
Sun Z, Worden M, Wroczynskyj Y, Yathindranath V, van Lierop J, Hegmann T, Miller DW. Magnetic field enhanced convective diffusion of iron oxide nanoparticles in an osmotically disrupted cell culture model of the blood-brain barrier. Int J Nanomedicine 2014; 9:3013-26. [PMID: 25018630 PMCID: PMC4073976 DOI: 10.2147/ijn.s62260] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
PURPOSE The present study examines the use of an external magnetic field in combination with the disruption of tight junctions to enhance the permeability of iron oxide nanoparticles (IONPs) across an in vitro model of the blood-brain barrier (BBB). The feasibility of such an approach, termed magnetic field enhanced convective diffusion (MFECD), along with the effect of IONP surface charge on permeability, was examined. METHODS The effect of magnetic field on the permeability of positively (aminosilane-coated [AmS]-IONPs) and negatively (N-(trimethoxysilylpropyl)ethylenediaminetriacetate [EDT]-IONPs) charged IONPs was evaluated in confluent monolayers of mouse brain endothelial cells under normal and osmotically disrupted conditions. RESULTS Neither IONP formulation was permeable across an intact cell monolayer. However, when tight junctions were disrupted using D-mannitol, flux of EDT-IONPs across the bEnd.3 monolayers was 28%, increasing to 44% when a magnetic field was present. In contrast, the permeability of AmS-IONPs after osmotic disruption was less than 5%. The cellular uptake profile of both IONPs was not altered by the presence of mannitol. CONCLUSIONS MFECD improved the permeability of EDT-IONPs through the paracellular route. The MFECD approach favors negatively charged IONPs that have low affinity for the brain endothelial cells and high colloidal stability. This suggests that MFECD may improve IONP-based drug delivery to the brain.
Collapse
Affiliation(s)
- Zhizhi Sun
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Matthew Worden
- Department of Chemistry and Biochemistry, Kent State University, Kent, OH, USA
| | - Yaroslav Wroczynskyj
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada
| | | | - Johan van Lierop
- Department of Physics and Astronomy, University of Manitoba, Winnipeg, Manitoba, Canada
| | - Torsten Hegmann
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada ; Department of Chemistry and Biochemistry, Kent State University, Kent, OH, USA ; Department of Chemistry, University of Manitoba, Winnipeg, Manitoba, Canada ; Chemical Physics Interdisciplinary Program, Liquid Crystal Institute, Kent State University, Kent, OH, USA
| | - Donald W Miller
- Department of Pharmacology and Therapeutics, University of Manitoba, Winnipeg, Manitoba, Canada
| |
Collapse
|
185
|
Focused ultrasound simultaneous irradiation/MRI imaging, and two-stage general kinetic model. PLoS One 2014; 9:e100280. [PMID: 24949997 PMCID: PMC4064987 DOI: 10.1371/journal.pone.0100280] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2013] [Accepted: 05/26/2014] [Indexed: 11/19/2022] Open
Abstract
Many studies have investigated how to use focused ultrasound (FUS) to temporarily disrupt the blood-brain barrier (BBB) in order to facilitate the delivery of medication into lesion sites in the brain. In this study, through the setup of a real-time system, FUS irradiation and injections of ultrasound contrast agent (UCA) and Gadodiamide (Gd, an MRI contrast agent) can be conducted simultaneously during MRI scanning. By using this real-time system, we were able to investigate in detail how the general kinetic model (GKM) is used to estimate Gd penetration in the FUS irradiated area in a rat's brain resulting from UCA concentration changes after single FUS irradiation. Two-stage GKM was proposed to estimate the Gd penetration in the FUS irradiated area in a rat's brain under experimental conditions with repeated FUS irradiation combined with different UCA concentrations. The results showed that the focal increase in the transfer rate constant of Ktrans caused by BBB disruption was dependent on the doses of UCA. Moreover, the amount of in vivo penetration of Evans blue in the FUS irradiated area in a rat's brain under various FUS irradiation experimental conditions was assessed to show the positive correlation with the transfer rate constants. Compared to the GKM method, the Two-stage GKM is more suitable for estimating the transfer rate constants of the brain treated with repeated FUS irradiations. This study demonstrated that the entire process of BBB disrupted by FUS could be quantitatively monitored by real-time dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI).
Collapse
|
186
|
Mikitsh JL, Chacko AM. Pathways for small molecule delivery to the central nervous system across the blood-brain barrier. PERSPECTIVES IN MEDICINAL CHEMISTRY 2014; 6:11-24. [PMID: 24963272 PMCID: PMC4064947 DOI: 10.4137/pmc.s13384] [Citation(s) in RCA: 123] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/26/2014] [Revised: 04/29/2014] [Accepted: 04/29/2014] [Indexed: 01/04/2023]
Abstract
The treatment of central nervous system (CNS) disease has long been difficult due to the ineffectiveness of drug delivery across the blood-brain barrier (BBB). This review summarizes important concepts of the BBB in normal versus pathophysiology and how this physical, enzymatic, and efflux barrier provides necessary protection to the CNS during drug delivery, and consequently treatment challenging. Small molecules account for the vast majority of available CNS drugs primarily due to their ability to penetrate the phospholipid membrane of the BBB by passive or carrier-mediated mechanisms. Physiochemical and biological factors relevant for designing small molecules with optimal capabilities for BBB permeability are discussed, as well as the most promising classes of transporters suitable for small-molecule drug delivery. Clinically translatable imaging methodologies for detecting and quantifying drug uptake and targeting in the brain are discussed as a means of further understanding and refining delivery parameters for both drugs and imaging probes in preclinical and clinical domains. This information can be used as a guide to design drugs with preserved drug action and better delivery profiles for improved treatment outcomes over existing therapeutic approaches.
Collapse
Affiliation(s)
- John L Mikitsh
- Department of Radiology, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Ann-Marie Chacko
- Department of Radiology, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- Center for Targeted Therapeutics and Translational Nanomedicine, Institute for Translational Medicine and Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| |
Collapse
|
187
|
Aryal M, Arvanitis CD, Alexander PM, McDannold N. Ultrasound-mediated blood-brain barrier disruption for targeted drug delivery in the central nervous system. Adv Drug Deliv Rev 2014; 72:94-109. [PMID: 24462453 DOI: 10.1016/j.addr.2014.01.008] [Citation(s) in RCA: 271] [Impact Index Per Article: 27.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2013] [Revised: 12/30/2013] [Accepted: 01/14/2014] [Indexed: 12/24/2022]
Abstract
The physiology of the vasculature in the central nervous system (CNS), which includes the blood-brain barrier (BBB) and other factors, complicates the delivery of most drugs to the brain. Different methods have been used to bypass the BBB, but they have limitations such as being invasive, non-targeted or requiring the formulation of new drugs. Focused ultrasound (FUS), when combined with circulating microbubbles, is a noninvasive method to locally and transiently disrupt the BBB at discrete targets. This review provides insight on the current status of this unique drug delivery technique, experience in preclinical models, and potential for clinical translation. If translated to humans, this method would offer a flexible means to target therapeutics to desired points or volumes in the brain, and enable the whole arsenal of drugs in the CNS that are currently prevented by the BBB.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Physics, Boston College, Chestnut Hill, USA; Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| | - Costas D Arvanitis
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA
| | - Phillip M Alexander
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA; Institute of Biomedical Engineering, Department of Engineering Science, and Brasenose College, University of Oxford, Oxford, UK
| | - Nathan McDannold
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, USA.
| |
Collapse
|
188
|
He K, Retterer ST, Srijanto BR, Conrad JC, Krishnamoorti R. Transport and dispersion of nanoparticles in periodic nanopost arrays. ACS NANO 2014; 8:4221-4227. [PMID: 24738873 DOI: 10.1021/nn404497z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Nanoparticles transported through highly confined porous media exhibit faster breakthrough than small molecule tracers. Despite important technological applications in advanced materials, human health, energy, and environment, the microscale mechanisms leading to early breakthrough have not been identified. Here, we measure dispersion of nanoparticles at the single-particle scale in regular arrays of nanoposts and show that for highly confined flows of dilute suspensions of nanoparticles the longitudinal and transverse velocities exhibit distinct scaling behaviors. The distributions of transverse particle velocities become narrower and more non-Gaussian when the particles are strongly confined. As a result, the transverse dispersion of highly confined nanoparticles at low Péclet numbers is significantly less important than longitudinal dispersion, leading to early breakthrough. This finding suggests a fundamental mechanism by which to control dispersion and thereby improve efficacy of nanoparticles applied for advanced polymer nanocomposites, drug delivery, hydrocarbon production, and environmental remediation.
Collapse
Affiliation(s)
- Kai He
- Department of Chemical and Biomolecular Engineering, University of Houston , Houston, Texas 77204-4004, United States
| | | | | | | | | |
Collapse
|
189
|
Bai JF, Liu P, Xu LX. Recent Advances in Thermal Treatment Techniques and Thermally Induced Immune Responses Against Cancer. IEEE Trans Biomed Eng 2014; 61:1497-505. [DOI: 10.1109/tbme.2014.2314357] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
190
|
Abstract
INTRODUCTION The presence of the blood-brain barrier (BBB) is a significant impediment to the delivery of therapeutic agents to the brain for treatment of brain diseases. Focused ultrasound (FUS) has been developed as a noninvasive method for transiently increasing the permeability of the BBB to promote drug delivery to targeted regions of the brain. AREAS COVERED The present review briefly compares the methods used to promote drug delivery to the brain and describes the benefits and limitations of FUS technology. We summarize the experimental data which shows that FUS, combined with intravascular microbubbles, increases therapeutic agent delivery into the brain leading to significant reductions in pathology in preclinical models of disease. The potential for translation of this technology to the clinic is also discussed. EXPERT OPINION The introduction of magnetic resonance imaging guidance and intravascular administration of microbubbles to FUS treatments permits the consistent, transient and targeted opening of the BBB. The development of feedback systems and real-time monitoring techniques improve the safety of BBB opening. Successful clinical translation of FUS has the potential to revolutionize the treatment of brain disease resulting in effective, less-invasive treatments without the need for expensive drug development.
Collapse
Affiliation(s)
- Alison Burgess
- Sunnybrook Research Institute, Physical Sciences , 2075 Bayview Avenue, S665, Toronto, ON M4N 3M5 , Canada +1 416 480 5765 ;
| | | |
Collapse
|
191
|
Bogart LK, Pourroy G, Murphy CJ, Puntes V, Pellegrino T, Rosenblum D, Peer D, Lévy R. Nanoparticles for imaging, sensing, and therapeutic intervention. ACS NANO 2014; 8:3107-22. [PMID: 24641589 PMCID: PMC4123720 DOI: 10.1021/nn500962q] [Citation(s) in RCA: 195] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/17/2014] [Indexed: 05/18/2023]
Abstract
Nanoparticles have the potential to contribute to new modalities in molecular imaging and sensing as well as in therapeutic interventions. In this Nano Focus article, we identify some of the current challenges and knowledge gaps that need to be confronted to accelerate the developments of various applications. Using specific examples, we journey from the characterization of these complex hybrid nanomaterials; continue with surface design and (bio)physicochemical properties, their fate in biological media and cells, and their potential for cancer treatment; and finally reflect on the role of animal models to predict their behavior in humans.
Collapse
Affiliation(s)
- Lara K. Bogart
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, Merseyside L69 3BX, United Kingdom
| | - Genevieve Pourroy
- Institut de Physique et Chimie des Matériaux de Strasbourg IPCMS, UMR 7504 CNRS-Université de Strasbourg, 23 rue du Loess BP 43, 67034 Strasbourg cedex 2, France
| | - Catherine J. Murphy
- Department of Chemistry, University of Illinois at Urbana—Champaign, 600 South Mathews Avenue, Urbana, Illinois 61801, United States
| | - Victor Puntes
- Insitut Català de Nanociencia I Nanotecnologia, campus UAB (CERCA-CSIC-ICREA), 08193 Barcelona, Spain
| | - Teresa Pellegrino
- Nanochemistry, Instituto Italiano di Tecnologia, via Morego 30, 16163 Genova, Italy
| | - Daniel Rosenblum
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Department of Materials Science and Engineering, Faculty of Engineering, and Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Dan Peer
- Laboratory of NanoMedicine, Department of Cell Research and Immunology, George S. Wise Faculty of Life Sciences, Department of Materials Science and Engineering, Faculty of Engineering, and Center for Nanoscience and Nanotechnology, Tel Aviv University, Tel Aviv 69978, Israel
| | - Raphaël Lévy
- Department of Biochemistry, Institute of Integrative Biology, University of Liverpool, Liverpool, Merseyside L69 3BX, United Kingdom
| |
Collapse
|
192
|
An L, Hu H, Du J, Wei J, Wang L, Yang H, Wu D, Shi H, Li F, Yang S. Paramagnetic hollow silica nanospheres for in vivo targeted ultrasound and magnetic resonance imaging. Biomaterials 2014; 35:5381-5392. [PMID: 24703718 DOI: 10.1016/j.biomaterials.2014.03.030] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Accepted: 03/13/2014] [Indexed: 11/29/2022]
Abstract
A series of hollow silica nanospheres (HSNSs) with sizes ranging from 100 to 400 nm were synthesized and used for primary ultrasound imaging (US) efficiency assessment. The 400 nm HSNSs were chosen as platform for conjugation with Gd-DTPA and cyclo-arginine-glycine-aspartic acid c(RGD) peptide to construct US and magnetic resonance imaging (MRI) dual-modal contrast agents (CAs): [HSNSs@(DTPA-Gd)-RGD]. The obtained CAs displayed good physiological stability, low cytotoxicity and negligible hemolytic activity in vitro. Furthermore, the passive accumulation and active-targeting of the HSNSs in the tumor site of mice was demonstrated by US and MR imaging, respectively. The qualitative and quantitative biodistribution of the HSNSs showed that they mainly accumulated in the tissues of liver, lung, tumor after intravenous administration and then be excreted from feces. In addition, histological, hematological, blood and biochemical analysis were used to further study toxicity of the HSNSs, and all results indicated that there were no covert toxicity of HSNSs in mice after long exposure times. Findings from this study indicated that the silica-based paramagnetic HSNSs can be used as a platform for long-term targeted imaging and therapy studies safely in vivo.
Collapse
Affiliation(s)
- Lu An
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, PR China
| | - He Hu
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, PR China.
| | - Jing Du
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, PR China
| | - Jie Wei
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, PR China
| | - Li Wang
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, PR China
| | - Hong Yang
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, PR China
| | - Dongmei Wu
- Shanghai Key Laboratory of Magnetic Resonance, Department of Physics, East China Normal University, Shanghai 200062, PR China
| | - Haili Shi
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, PR China
| | - Fenghua Li
- Department of Ultrasound, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, PR China
| | - Shiping Yang
- The Education Ministry Key Lab of Resource Chemistry and Shanghai Key Laboratory of Rare Earth Functional Materials, Shanghai Normal University, Shanghai 200234, PR China.
| |
Collapse
|
193
|
Barua S, Mitragotri S. Challenges associated with Penetration of Nanoparticles across Cell and Tissue Barriers: A Review of Current Status and Future Prospects. NANO TODAY 2014; 9:223-243. [PMID: 25132862 PMCID: PMC4129396 DOI: 10.1016/j.nantod.2014.04.008] [Citation(s) in RCA: 716] [Impact Index Per Article: 71.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
Nanoparticles (NPs) have emerged as an effective modality for the treatment of various diseases including cancer, cardiovascular and inflammatory diseases. Various forms of NPs including liposomes, polymer particles, micelles, dendrimers, quantum dots, gold NPs and carbon nanotubes have been synthesized and tested for therapeutic applications. One of the greatest challenges that limit the success of NPs is their ability to reach the therapeutic site at necessary doses while minimizing accumulation at undesired sites. The biodistribution of NPs is determined by body's biological barriers that manifest in several distinct ways. For intravascular delivery of NPs, the barrier manifests in the form of: (i) immune clearance in the liver and spleen, (ii) permeation across the endothelium into target tissues, (iii) penetration through the tissue interstitium, (iv) endocytosis in target cells, (v) diffusion through cytoplasm and (vi) eventually entry into the nucleus, if required. Certain applications of NPs also rely on delivery through alternate routes including skin and mucosal membranes of the nose, lungs, intestine and vagina. In these cases, the diffusive resistance of these tissues poses a significant barrier to delivery. This review focuses on the current understanding of penetration of NPs through biological barriers. Emphasis is placed on transport barriers and not immunological barriers. The review also discusses design strategies for overcoming the barrier properties.
Collapse
Affiliation(s)
- Sutapa Barua
- Center for Bioengineering, Department of Chemical Engineering University of California, Santa Barbara, CA 93106
| | - Samir Mitragotri
- Center for Bioengineering, Department of Chemical Engineering University of California, Santa Barbara, CA 93106
| |
Collapse
|
194
|
Tu SJ, Wu SY, Wang FS, Ma YH. Retention assessment of magnetic nanoparticles in rat arteries with micro-computed tomography. Phys Med Biol 2014; 59:1271-81. [DOI: 10.1088/0031-9155/59/5/1271] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
|
195
|
Liu HL, Fan CH, Ting CY, Yeh CK. Combining microbubbles and ultrasound for drug delivery to brain tumors: current progress and overview. Theranostics 2014; 4:432-44. [PMID: 24578726 PMCID: PMC3936295 DOI: 10.7150/thno.8074] [Citation(s) in RCA: 189] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2013] [Accepted: 01/20/2014] [Indexed: 02/06/2023] Open
Abstract
Malignant glioma is one of the most challenging central nervous system (CNS) diseases, which is typically associated with high rates of recurrence and mortality. Current surgical debulking combined with radiation or chemotherapy has failed to control tumor progression or improve glioma patient survival. Microbubbles (MBs) originally serve as contrast agents in diagnostic ultrasound but have recently attracted considerable attention for therapeutic application in enhancing blood-tissue permeability for drug delivery. MB-facilitated focused ultrasound (FUS) has already been confirmed to enhance CNS-blood permeability by temporally opening the blood-brain barrier (BBB), thus has potential to enhance delivery of various kinds of therapeutic agents into brain tumors. Here we review the current preclinical studies which demonstrate the reports by using FUS with MB-facilitated drug delivery technology in brain tumor treatment. In addition, we review newly developed multifunctional theranostic MBs for FUS-induced BBB opening for brain tumor therapy.
Collapse
Affiliation(s)
- Hao-Li Liu
- 1. Department of Electrical Engineering, Chang-Gung University, 259 Wen-Hwa 1st Road, Kuei-Shan, Tao-Yuan, Taiwan 33302
| | - Ching-Hsiang Fan
- 2. Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu, Taiwan 30013
| | - Chien-Yu Ting
- 2. Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu, Taiwan 30013
| | - Chih-Kuang Yeh
- 2. Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, No. 101, Section 2, Kuang-Fu Road, Hsinchu, Taiwan 30013
| |
Collapse
|
196
|
Cheng Y, Morshed RA, Auffinger B, Tobias AL, Lesniak MS. Multifunctional nanoparticles for brain tumor imaging and therapy. Adv Drug Deliv Rev 2014; 66:42-57. [PMID: 24060923 PMCID: PMC3948347 DOI: 10.1016/j.addr.2013.09.006] [Citation(s) in RCA: 230] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2013] [Revised: 08/28/2013] [Accepted: 09/13/2013] [Indexed: 12/16/2022]
Abstract
Brain tumors are a diverse group of neoplasms that often carry a poor prognosis for patients. Despite tremendous efforts to develop diagnostic tools and therapeutic avenues, the treatment of brain tumors remains a formidable challenge in the field of neuro-oncology. Physiological barriers including the blood-brain barrier result in insufficient accumulation of therapeutic agents at the site of a tumor, preventing adequate destruction of malignant cells. Furthermore, there is a need for improvements in brain tumor imaging to allow for better characterization and delineation of tumors, visualization of malignant tissue during surgery, and tracking of response to chemotherapy and radiotherapy. Multifunctional nanoparticles offer the potential to improve upon many of these issues and may lead to breakthroughs in brain tumor management. In this review, we discuss the diagnostic and therapeutic applications of nanoparticles for brain tumors with an emphasis on innovative approaches in tumor targeting, tumor imaging, and therapeutic agent delivery. Clinically feasible nanoparticle administration strategies for brain tumor patients are also examined. Furthermore, we address the barriers towards clinical implementation of multifunctional nanoparticles in the context of brain tumor management.
Collapse
Affiliation(s)
- Yu Cheng
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | - Ramin A Morshed
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | - Brenda Auffinger
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | - Alex L Tobias
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA
| | - Maciej S Lesniak
- The Brain Tumor Center, The University of Chicago, Chicago, IL, USA.
| |
Collapse
|
197
|
Aguiló-Aguayo N, Maurizi L, Galmarini S, Ollivier-Beuzelin MG, Coullerez G, Bertran E, Hofmann H. Aqueous stabilisation of carbon-encapsulated superparamagnetic α-iron nanoparticles for biomedical applications. Dalton Trans 2014; 43:13764-75. [DOI: 10.1039/c4dt00085d] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Carbon-encapsulated superparamagnetic α-Fe nanoparticles were stabilised in aqueous media allowing their cell internalisation.
Collapse
Affiliation(s)
- Noemí Aguiló-Aguayo
- FEMAN Group
- IN2 UB
- Department of Applied Physics and Optics
- Universitat de Barcelona
- 08028 Barcelona, Spain
| | - Lionel Maurizi
- Laboratoire de Technologie de Poudres (LTP)
- EPFL-STI-IMX-LTP
- Lausanne, Switzerland
| | - Sandra Galmarini
- Laboratoire de Technologie de Poudres (LTP)
- EPFL-STI-IMX-LTP
- Lausanne, Switzerland
| | | | - Géraldine Coullerez
- Laboratoire de Technologie de Poudres (LTP)
- EPFL-STI-IMX-LTP
- Lausanne, Switzerland
| | - Enric Bertran
- FEMAN Group
- IN2 UB
- Department of Applied Physics and Optics
- Universitat de Barcelona
- 08028 Barcelona, Spain
| | - Heinrich Hofmann
- Laboratoire de Technologie de Poudres (LTP)
- EPFL-STI-IMX-LTP
- Lausanne, Switzerland
| |
Collapse
|
198
|
|
199
|
Carenza E, Barceló V, Morancho A, Levander L, Boada C, Laromaine A, Roig A, Montaner J, Rosell A. In vitro angiogenic performance and in vivo brain targeting of magnetized endothelial progenitor cells for neurorepair therapies. NANOMEDICINE : NANOTECHNOLOGY, BIOLOGY, AND MEDICINE 2014; 10:225-34. [PMID: 23792330 DOI: 10.1016/j.nano.2013.06.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2013] [Revised: 05/27/2013] [Accepted: 06/11/2013] [Indexed: 01/01/2023]
Abstract
Endothelial progenitor cells (EPCs) represent a promising approach for cell-based therapies to induce tissue repair; however, their effective delivery into the brain has remained a challenge. We loaded EPCs with superparamagnetic iron oxide nanoparticles (SPIONs), assessed their angiogenic potential and evaluated their guidance to the brain using an external magnet. SPIONs were stored in the cytoplasm within endosomes/lysosomes as observed by transmission electron microscopy (TEM) and could be visualized as hypointense signals by magnetic resonance imaging (MRI) T2-weighted images. In vitro SPION-loaded EPCs were fully functional, forming vessel-like structures in Matrigel®, and displayed enhanced migration and secretion of growth factors (VEGF and FGF), which was associated with a moderate increase in reactive oxygen species production. Furthermore, in vivo MRI of treated mice showed accumulated hypointense signals consistent with SPION-loaded EPCs engraftment. Thus, we demonstrate that loading EPCs with SPIONs represents a safe and effective strategy for precise cell guidance into specific brain areas. FROM THE CLINICAL EDITOR This study investigates the potential role of endothelial progenitor cells in neuro-repair strategies of the central nervous system using SPION-loaded EPCs and magnetic guidance to the target organ. The authors demonstrate ex vivo cellular viability and maintained function following SPION load as well as successful guidance of the EPCs to the target site via MR imaging in a murine model.
Collapse
Affiliation(s)
- Elisa Carenza
- Institut de Ciència de Materials de Barcelona, Consejo Superior de Investigaciones Científicas (ICMAB-CSIC), Campus de la UAB, Bellaterra, Catalunya, Spain
| | - Verónica Barceló
- Neurovascular Research Laboratory and Neurovascular Unit, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, Barcelona, Catalunya, Spain
| | - Anna Morancho
- Neurovascular Research Laboratory and Neurovascular Unit, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, Barcelona, Catalunya, Spain
| | - Lisa Levander
- Institut de Ciència de Materials de Barcelona, Consejo Superior de Investigaciones Científicas (ICMAB-CSIC), Campus de la UAB, Bellaterra, Catalunya, Spain
| | - Cristina Boada
- Neurovascular Research Laboratory and Neurovascular Unit, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, Barcelona, Catalunya, Spain
| | - Anna Laromaine
- Institut de Ciència de Materials de Barcelona, Consejo Superior de Investigaciones Científicas (ICMAB-CSIC), Campus de la UAB, Bellaterra, Catalunya, Spain
| | - Anna Roig
- Institut de Ciència de Materials de Barcelona, Consejo Superior de Investigaciones Científicas (ICMAB-CSIC), Campus de la UAB, Bellaterra, Catalunya, Spain.
| | - Joan Montaner
- Neurovascular Research Laboratory and Neurovascular Unit, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, Barcelona, Catalunya, Spain
| | - Anna Rosell
- Neurovascular Research Laboratory and Neurovascular Unit, Vall d'Hebron Institut de Recerca, Hospital Universitari Vall d'Hebron, Universitat Autònoma de Barcelona, Passeig Vall d'Hebron 119-129, Barcelona, Catalunya, Spain.
| |
Collapse
|
200
|
Gdowski A, Ranjan AP, Mukerjee A, Vishwanatha JK. Nanobiosensors: role in cancer detection and diagnosis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2014; 807:33-58. [PMID: 24619617 DOI: 10.1007/978-81-322-1777-0_4] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The ability to detect many cancers at an early stage in its clinical course has the potential to improve patient outcomes in terms of morbidity and mortality. Nanosized components incorporated into existing clinical diagnostic and detection systems as well as novel nanobiosensors have demonstrated improved sensitivity and specificity compared with traditional cancer testing approaches. Nanoparticles, nanowires, nanotubes, and nanocantilevers are examples of four nanobiosensor systems that have been used experimentally in the context of detection and diagnosis of prostate, breast, pancreatic, lung, and brain cancers over the past few years. Nanobiosensors will begin to transition into clinically validated tests as experimental and engineering techniques advance. This paper presents examples of some such nanobiosensors for cancer diagnosis and detection.
Collapse
Affiliation(s)
- Andrew Gdowski
- Department of Molecular Biology and Immunology, Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, 76107, USA
| | | | | | | |
Collapse
|