101
|
|
102
|
O'Reilly MA, Hynynen K. Ultrasound and Microbubble-Mediated Blood-Brain Barrier Disruption for Targeted Delivery of Therapeutics to the Brain. Methods Mol Biol 2018; 1831:111-119. [PMID: 30051428 DOI: 10.1007/978-1-4939-8661-3_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
Ultrasound and microbubble-mediated disruption of the Blood-Brain barrier is a noninvasive and targetable technique that permits the investigation of pharmacological interventions in the brain and CNS. This technique provides an alternative to direct injection of agents into the brain parenchyma or chemical disruption of the Blood-Brain barrier. Here, we detail one protocol for inducing transient Blood-Brain barrier disruption in a rodent model using a commercially available microbubble contrast agent (Definity).
Collapse
Affiliation(s)
- Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada.
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, ON, Canada
- Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
103
|
Magnetic Nanoparticles in the Central Nervous System: Targeting Principles, Applications and Safety Issues. Molecules 2017; 23:molecules23010009. [PMID: 29267188 PMCID: PMC5943969 DOI: 10.3390/molecules23010009] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Revised: 12/12/2017] [Accepted: 12/19/2017] [Indexed: 02/07/2023] Open
Abstract
One of the most challenging goals in pharmacological research is overcoming the Blood Brain Barrier (BBB) to deliver drugs to the Central Nervous System (CNS). The use of physical means, such as steady and alternating magnetic fields to drive nanocarriers with proper magnetic characteristics may prove to be a useful strategy. The present review aims at providing an up-to-date picture of the applications of magnetic-driven nanotheranostics agents to the CNS. Although well consolidated on physical ground, some of the techniques described herein are still under investigation on in vitro or in silico models, while others have already entered in—or are close to—clinical validation. The review provides a concise overview of the physical principles underlying the behavior of magnetic nanoparticles (MNPs) interacting with an external magnetic field. Thereafter we describe the physiological pathways by which a substance can reach the brain from the bloodstream and then we focus on those MNP applications that aim at a nondestructive crossing of the BBB such as static magnetic fields to facilitate the passage of drugs and alternating magnetic fields to increment BBB permeability by magnetic heating. In conclusion, we briefly cite the most notable biomedical applications of MNPs and some relevant remarks about their safety and potential toxicity.
Collapse
|
104
|
Hersh DS, Kim AJ, Winkles JA, Eisenberg HM, Woodworth GF, Frenkel V. Emerging Applications of Therapeutic Ultrasound in Neuro-oncology: Moving Beyond Tumor Ablation. Neurosurgery 2017; 79:643-654. [PMID: 27552589 DOI: 10.1227/neu.0000000000001399] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
: Transcranial focused ultrasound (FUS) can noninvasively transmit acoustic energy with a high degree of accuracy and safety to targets and regions within the brain. Technological advances, including phased-array transducers and real-time temperature monitoring with magnetic resonance thermometry, have created new opportunities for FUS research and clinical translation. Neuro-oncology, in particular, has become a major area of interest because FUS offers a multifaceted approach to the treatment of brain tumors. FUS has the potential to generate cytotoxicity within tumor tissue, both directly via thermal ablation and indirectly through radiosensitization and sonodynamic therapy; to enhance the delivery of therapeutic agents to brain tumors by transiently opening the blood-brain barrier or improving distribution through the brain extracellular space; and to modulate the tumor microenvironment to generate an immune response. In this review, we describe each of these applications for FUS, the proposed mechanisms of action, and the preclinical and clinical studies that have set the foundation for using FUS in neuro-oncology. ABBREVIATIONS BBB, blood-brain barrierCED, convection-enhanced delivery5-Ala, 5-aminolevulinic acidFUS, focused ultrasoundGBM, glioblastoma multiformeHSP, heat shock proteinMRgFUS, magnetic resonance-guided focused ultrasoundpFUS, pulsed focused ultrasound.
Collapse
Affiliation(s)
- David S Hersh
- *Department of Neurosurgery,‡Marlene and Stewart Greenebaum Cancer Center,¶Center for Biomedical Engineering and Technology,‖Department of Surgery,#Center for Vascular and Inflammatory Diseases, and**Department of Diagnostic Radiology and Nuclear Medicine, University of Maryland School of Medicine, Baltimore, Maryland;§Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland
| | | | | | | | | | | |
Collapse
|
105
|
Dai C, Zhang S, Liu Z, Wu R, Chen Y. Two-Dimensional Graphene Augments Nanosonosensitized Sonocatalytic Tumor Eradication. ACS NANO 2017; 11:9467-9480. [PMID: 28829584 DOI: 10.1021/acsnano.7b05215] [Citation(s) in RCA: 183] [Impact Index Per Article: 26.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/22/2023]
Abstract
Ultrasound (US) can activate sonosensitizers for sonodynamic therapy (SDT), but the low activation efficiency and therapeutic outcome significantly hinder its further clinical translation. Inspired by the principles of semiconductor physics and photocatalysis chemistry, we herein report on augmenting the sonocatalytic efficiency of semiconductor TiO2-based nanosonosensitizers for highly efficient SDT by the integration of two-dimensional (2D) ultrathin graphene with TiO2 nanosonosensitizers. The high electroconductivity of graphene facilitates the separation of the electron (e-) and hole (h+) pairs from the energy band of TiO2 and avoids their recombination upon external US irradiation; thus it significantly augments the therapeutic efficiency of TiO2 nanosonosensitizers for SDT against tumors. By further MnOx functionalization, these 2D composite nanosonosensitizers achieved tumor microenvironment-sensitive (mild acidity) T1-weighted magnetic resonance imaging of tumors for therapeutic guidance and monitoring. The high photothermal-conversion capability of graphene also synergistically enhanced the SDT efficiency, achieving the complete eradication of a tumor without reoccurrence. This work provides a paradigm for augmenting semiconductor TiO2-based sonocatalytic therapeutic nanomedicine by learning the physiochemical principles from traditional photocatalysis, which also demonstrates a highly efficient noninvasive and safe therapeutic modality for tumor eradication by the nanosonosensitized sonocatalytic process.
Collapse
Affiliation(s)
- Chen Dai
- Department of Ultrasound in Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai 200072, People's Republic of China
- Department of Ultrasound, Shanghai East Hospital, Tongji University School of Medicine , Shanghai 200120, People's Republic of China
| | - Shengjian Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center , Shanghai 200032, People's Republic of China
| | - Zhuang Liu
- Department of Radiology, Fudan University Shanghai Cancer Center , Shanghai 200032, People's Republic of China
| | - Rong Wu
- Department of Ultrasound in Medicine, Shanghai Tenth People's Hospital, Tongji University School of Medicine , Shanghai 200072, People's Republic of China
| | - Yu Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences , Shanghai 200050, People's Republic of China
| |
Collapse
|
106
|
Browning RJ, Reardon PJT, Parhizkar M, Pedley RB, Edirisinghe M, Knowles JC, Stride E. Drug Delivery Strategies for Platinum-Based Chemotherapy. ACS NANO 2017; 11:8560-8578. [PMID: 28829568 DOI: 10.1021/acsnano.7b04092] [Citation(s) in RCA: 153] [Impact Index Per Article: 21.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Few chemotherapeutics have had such an impact on cancer management as cis-diamminedichloridoplatinum(II) (CDDP), also known as cisplatin. The first member of the platinum-based drug family, CDDP's potent toxicity in disrupting DNA replication has led to its widespread use in multidrug therapies, with particular benefit in patients with testicular cancers. However, CDDP also produces significant side effects that limit the maximum systemic dose. Various strategies have been developed to address this challenge including encapsulation within micro- or nanocarriers and the use of external stimuli such as ultrasound to promote uptake and release. The aim of this review is to look at these strategies and recent scientific and clinical developments.
Collapse
Affiliation(s)
- Richard J Browning
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford , Oxford OX1 2JD, United Kingdom
| | | | | | | | | | - Jonathan C Knowles
- Department of Nanobiomedical Science and BK21 Plus NBM, Global Research Center for Regenerative Medicine, Dankook University , 518-10 Anseo-dong, Dongnam-gu, Cheonan, Chungcheongnam-do, Republic of Korea
- The Discoveries Centre for Regenerative and Precision Medicine, UCL Campus , Gower Street, London WC1E 6BT, United Kingdom
| | - Eleanor Stride
- Institute of Biomedical Engineering, Department of Engineering Science, University of Oxford , Oxford OX1 2JD, United Kingdom
| |
Collapse
|
107
|
Askoxylakis V, Arvanitis CD, Wong CSF, Ferraro GB, Jain RK. Emerging strategies for delivering antiangiogenic therapies to primary and metastatic brain tumors. Adv Drug Deliv Rev 2017. [PMID: 28648712 DOI: 10.1016/j.addr.2017.06.011] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Five-year survival rates have not increased appreciably for patients with primary and metastatic brain tumors. Nearly 17,000 patients die from primary brain tumors, whereas approximately 200,000 cases are diagnosed with brain metastasis every year in the US alone. At the same time, with improved control of systemic disease, the incidence of brain metastasis is increasing. Thus, novel approaches for improving the treatment outcome for these uniformly fatal diseases are needed urgently. In the review, we summarize the challenges in the treatment of these diseases using antiangiogenic therapies alone or in combination with radio-, chemo- and immuno-therapies. We also discuss the emerging strategies to improve the treatment outcome using both pharmacological approaches to normalize the tumor microenvironment and physical approaches (e.g., focused ultrasound) to modulate the blood-tumor-barrier, along with limitations of each approach. Finally, we offer some new avenues of future research.
Collapse
Affiliation(s)
- Vasileios Askoxylakis
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH), Harvard Medical School (HMS), Boston, MA, 02114, USA
| | - Costas D Arvanitis
- School of Mechanical Engineering, Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Christina S F Wong
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH), Harvard Medical School (HMS), Boston, MA, 02114, USA
| | - Gino B Ferraro
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH), Harvard Medical School (HMS), Boston, MA, 02114, USA
| | - Rakesh K Jain
- Edwin L. Steele Laboratories, Department of Radiation Oncology, Massachusetts General Hospital (MGH), Harvard Medical School (HMS), Boston, MA, 02114, USA.
| |
Collapse
|
108
|
The role of non-endothelial cells on the penetration of nanoparticles through the blood brain barrier. Prog Neurobiol 2017; 159:39-49. [PMID: 28899762 DOI: 10.1016/j.pneurobio.2017.09.001] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 08/30/2017] [Accepted: 09/08/2017] [Indexed: 12/26/2022]
Abstract
The blood brain barrier (BBB) is a well-established cell-based membrane that circumvents the central nervous system (CNS), protecting it from harmful substances. Due to its robustness and cell integrity, it is also an outstanding opponent when it comes to the delivery of several therapeutic agents to the brain, which requires the crossing through its highly-organized structure. This regulation and cell-cell communications occur mostly between astrocytes, pericytes and endothelial cells. Therefore, alternative ways to deliver drugs to the CNS, overcoming the BBB are required, to improve the efficacy of brain target drugs. Nanoparticles emerge here as a promising drug delivery strategy, due to their ability of high drug loading and the capability to exploit specific delivery pathways that most drugs are unable to when administered freely, increasing their bioavailability in the CNS. Thus, further attempts to assess the possible influence of non-endothelial may have on the BBB translocation of nanoparticles are here revised. Furthermore, the use of macrophages and/or monocytes as nanoparticle delivery cells are also approached. Lastly, the temporarily disruption of the overall organization and normal structure of the BBB to promote the penetration of nanoparticles aimed at the CNS is described, as a synergistic path.
Collapse
|
109
|
Zhou Y, Han X, Jing X, Chen Y. Construction of Silica-Based Micro/Nanoplatforms for Ultrasound Theranostic Biomedicine. Adv Healthc Mater 2017; 6. [PMID: 28795530 DOI: 10.1002/adhm.201700646] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2017] [Revised: 06/24/2017] [Indexed: 12/20/2022]
Abstract
Ultrasound (US)-based biomedicine has been extensively explored for its applications in both diagnostic imaging and disease therapy. The fast development of theranostic nanomedicine significantly promotes the development of US-based biomedicine. This progress report summarizes and discusses the recent developments of rational design and fabrication of silica-based micro/nanoparticles for versatile US-based biomedical applications. The synthetic strategies and surface-engineering approaches of silica-based micro/nanoparticles are initially discussed, followed by detailed introduction on their US-based theranostic applications. They have been extensively explored in contrast-enhanced US imaging, US-based multi-modality imaging, synergistic high-intensity focused US (HIFU) ablation, sonosensitizer-enhanced sonodynamic therapy (SDT), as well as US-triggered chemotherapy. Their biological effects and biosafety have been briefly discussed to guarantee further clinical translation. Based on the high biocompatibility, versatile composition/structure and high performance in US-based theranostic biomedicine, these silica-based theranostic agents are expected to pave a new way for achieving efficient US-based theranostics of disease by taking the specific advantages of material science, nanotechnology and US-based biomedicine.
Collapse
Affiliation(s)
- Yang Zhou
- Department of Ultrasound the Third People's Hospital of Chengdu City the Affiliated Hospital of Southwest Jiaotong University Chengdu 600031 P. R. China
| | - Xiaoxia Han
- Institute of Ultrasound Imaging and Department of Ultrasound Second Affiliated Hospital of Chongqing Medical University Chongqing 400010 P. R. China
| | - Xiangxiang Jing
- Department of Ultrasound Hainan General Hospital Haikou 570311 P. R. China
| | - Yu Chen
- State Key Lab of High Performance Ceramics and Superfine Microstructure Shanghai Institute of Ceramics Chinese Academy of Sciences Shanghai 200050 P. R. China
| |
Collapse
|
110
|
Lin FW, Chen PY, Wei KC, Huang CY, Wang CK, Yang HW. Rapid In Situ MRI Traceable Gel-forming Dual-drug Delivery for Synergistic Therapy of Brain Tumor. Am J Cancer Res 2017; 7:2524-2536. [PMID: 28744332 PMCID: PMC5525754 DOI: 10.7150/thno.19856] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Accepted: 04/21/2017] [Indexed: 01/03/2023] Open
Abstract
Preventing tumor recurrence after surgical resection of a brain tumor is a significant clinical challenge because current methods deliver chemotherapeutic agents in a rapid manner and are not effective against the residual tumor cells. To overcome this drawback, we report a simple method to prepare magnetic resonance imaging (MRI) traceable ultra-thermosensitive hydrogels with rapid gelation ability from aqueous solution within 4 s at 28 °C for hydrophilic (epirubicin, EPI) and hydrophobic (paclitaxel, PTX) drugs co-delivery with bovine serum albumin nanoparticles (BSA NPs) incorporation. The results showed the average survival of gliosarcoma-bearing (MBR 614 or U87) mice receiving BSA/PTX NPs incorporated hydrogelGd/EPI increased to 63 days or 69 days with no tumor recurrence observed. Our synergistic strategy presents a new approach to the development of a local drug delivery system for the prevention of brain tumor recurrence.
Collapse
|
111
|
Lee TH, Yeh JC, Tsai CH, Yang JT, Lou SL, Seak CJ, Wang CY, Wei KC, Liu HL. Improved thrombolytic effect with focused ultrasound and neuroprotective agent against acute carotid artery thrombosis in rat. Sci Rep 2017; 7:1638. [PMID: 28487554 PMCID: PMC5431649 DOI: 10.1038/s41598-017-01769-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2016] [Accepted: 03/30/2017] [Indexed: 01/19/2023] Open
Abstract
Combination therapy with focused ultrasound (FUS) and a neuroprotective agent, BNG-1, was examined in an acute carotid thrombotic occlusion model using LED irradiation in rat to improve the thrombolytic effect of rt-PA. Seven treatment groups included (A) intravenous bolus injection of 0.45 mg/kg rt-PA, (B) intravenous bolus injection of 0.9 mg/kg, (C) sonothrombolysis with FUS alone, (D) oral administration of 2 g/kg BNG-1 for 7 days alone, (E) A + D, (F) A + C, and (G) A + C + D. Four comparison groups were made including (H) 0.45 mg/kg rt-PA 20% bolus +80% IV fusion + FUS, (I) 0.9 mg/kg rt-PA with 10% bolus + 90% intravenous fusion, (J) B + C, (K) B + D. At 7 days after carotid occlusion, small-animal carotid ultrasound and 7 T MR angiography showed the recanalization rate of ≤50% stenosis was 50% in group B and 83% in group I, but 0% in groups A and C and 17% in group D. Combination therapy improved recanalization rate to 50–63% in groups E and F, to 67–83% in groups J and K, and to 100% in groups G and H. Our study demonstrated combination therapy with different remedies can be a feasible strategy to improve the thrombolytic effect of rt-PA.
Collapse
Affiliation(s)
- Tsong-Hai Lee
- Stroke Center and Department of Neurology, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Jih-Chao Yeh
- Stroke Center and Department of Neurology, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chih-Hung Tsai
- Departments of Electrical Engineering, Graduate Institute of Clinical Medical Sciences, Chang-Gung University, Taoyuan, 333, Taiwan
| | - Jen-Tsung Yang
- Department of Neurosurgery, Chiayi Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Chiayi, Taiwan
| | - Shyh-Liang Lou
- Department of Biomedical Engineering, Chung Yuan Christian University, Chung Li, Taiwan
| | - Chen-June Seak
- Department of Emergency Medicine, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Chao-Yung Wang
- Department of Cardiology, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan
| | - Kuo-Chen Wei
- Department of Neurosurgery, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan.
| | - Hao-Li Liu
- Departments of Electrical Engineering, Graduate Institute of Clinical Medical Sciences, Chang-Gung University, Taoyuan, 333, Taiwan. .,Department of Neurosurgery, Linkou Chang Gung Memorial Hospital and College of Medicine, Chang Gung University, Taoyuan, 333, Taiwan. .,Institute for Radiological Research, Chang Gung University/Chang Gung Memorial Hospital, Taoyuan, 333, Taiwan.
| |
Collapse
|
112
|
Biological effects of blood-brain barrier disruption using a focused ultrasound. Biomed Eng Lett 2017; 7:115-120. [PMID: 30603158 DOI: 10.1007/s13534-017-0025-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2016] [Revised: 02/19/2017] [Accepted: 03/24/2017] [Indexed: 01/06/2023] Open
Abstract
With focused ultrasound (FUS) and microbubbles, BBB can be transiently disrupted with a localized and non-invasive approach. BBB disruption induced by FUS has made progressions to move forward on delivery of therapeutic agents into a brain in a specific area of brain for better treatment of neurological diseases. In addition to be used as an improvement of drug delivery, BBB disruption has been found to induce biological effects such as a clearance of protein aggregation which cause Alzheimer's disease, regulation of proteins which facilitate drug uptake, and modulation of neuronal function and neurogenesis. In this review, we discuss overview about the principles of BBB opening with FUS and milestones in these biological effects of FUS-induced BBB disruption.
Collapse
|
113
|
Jin Z, Wen Y, Hu Y, Chen W, Zheng X, Guo W, Wang T, Qian Z, Su BL, He Q. MRI-guided and ultrasound-triggered release of NO by advanced nanomedicine. NANOSCALE 2017; 9:3637-3645. [PMID: 28247895 DOI: 10.1039/c7nr00231a] [Citation(s) in RCA: 98] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/19/2023]
Abstract
Nitric oxide (NO) has been well identified as a specific free radical molecule possessing wide-ranging therapeutic effects. Targeted delivery and controlled release of NO are highly desired to realize precision gas therapy, but are still challenging owing to the non-targetability and uncontrollability of NO itself. Herein, we propose a new concept of MRI-guided and ultrasound-triggered gas release for precision gas therapy. Based on this concept, we develop a novel ultrasound-responsive BNN-type NO-releasing molecule (NORM) and an advanced rattle-type nano-carrier of superparamagnetic iron oxide-encapsulated mesoporous silica nanoparticles (SPION@hMSN), and use them to construct a new intelligent nanomedicine (BNN6-SPION@hMSN) for the first time. The BNN6-SPION@hMSN nanomedicine exhibits excellent passive tumor-targeting capability, high MRI-guided tumor localization performance and a unique ultrasound-triggered NO release profile. The tumor-targeted, MRI-guided and ultrasound-triggered release profiles of the developed nanomedicine enable the tumor site-specific controlled release of NO in favor of high-efficacy and safe NO gas therapy of tumor.
Collapse
Affiliation(s)
- Zhaokui Jin
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, Guangdong, P. R. China.
| | - Yanyuan Wen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, Guangdong, P. R. China.
| | - Yaxin Hu
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, Guangdong, P. R. China.
| | - Wenwen Chen
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, Guangdong, P. R. China.
| | - Xianfeng Zheng
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Weisheng Guo
- Chinese Academy of Sciences (CAS) Center for Excellence in Nanoscience and CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, National Center for Nanoscience and Technology of China, Beijing 100190, P. R. China
| | - Tianfu Wang
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, Guangdong, P. R. China.
| | - Zhiyong Qian
- State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center for Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Bao-Lian Su
- State Key Laboratory of Advanced Technology for Materials Synthesis and Processing, Wuhan University of Technology, Wuhan 430070, P. R. China
| | - Qianjun He
- National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University, Shenzhen 518060, Guangdong, P. R. China.
| |
Collapse
|
114
|
O'Reilly MA, Hough O, Hynynen K. Blood-Brain Barrier Closure Time After Controlled Ultrasound-Induced Opening Is Independent of Opening Volume. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2017; 36:475-483. [PMID: 28108988 PMCID: PMC5319892 DOI: 10.7863/ultra.16.02005] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2016] [Accepted: 06/03/2016] [Indexed: 05/15/2023]
Abstract
OBJECTIVES Microbubble-mediated focused ultrasound (US) opening of the blood-brain barrier (BBB) has shown promising results for the treatment of brain tumors and conditions such as Alzheimer disease. Practical clinical implementation of focused US treatments would aim to treat a substantial portion of the brain; thus, the safety of opening large volumes must be investigated. This study investigated whether the opened volume affects the time for the BBB to be restored after treatment. METHODS Sprague Dawley rats (n = 5) received bilateral focused US treatments. One hemisphere received a single sonication, and the contralateral hemisphere was targeted with 4 overlapping foci. Contrast-enhanced T1-weighted magnetic resonance imaging was used to assess the integrity of the BBB at 0, 6, and 24 hours after focused US. RESULTS At time 0, there was no significant difference in the mean enhancement between the single- and multi-point sonications (mean ± SD, 29.7% ± 18.4% versus 29.7% ± 24.1%; P = .9975). The mean cross-sectional area of the BBB opening resulting from the multi-point sonication was approximately 3.5-fold larger than that of the single-point case (14.2 ± 4.7 versus 4.1 ± 3.3 mm2 ; P < .0001). The opened volumes in 9 of 10 hemispheres were closed by 6 hours after focused US. The remaining treatment location had substantially reduced enhancement at 6 hours and was closed by 24 hours. Histologic analysis revealed small morphologic changes associated with this location. T2-weighted images at 6 and 24 hours showed no signs of edema. T2*-weighted images obtained at 6 hours also showed no signs hemorrhage in any animal. CONCLUSIONS The time for the BBB to close after focused US was independent of the opening volume on the time scale investigated. No differences in treatment effects were observable by magnetic resonance imaging follow-up between larger- and smaller-volume sonications, suggesting that larger-volume BBB opening can be performed safely.
Collapse
Affiliation(s)
- Meaghan A O'Reilly
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
| | - Olivia Hough
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Kullervo Hynynen
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, Ontario, Canada
- Department of Medical Biophysics, University of Toronto, Toronto, Ontario, Canada
- Department of Institute of Biomaterials and Biomedical Engineering, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
115
|
Aryal M, Fischer K, Gentile C, Gitto S, Zhang YZ, McDannold N. Effects on P-Glycoprotein Expression after Blood-Brain Barrier Disruption Using Focused Ultrasound and Microbubbles. PLoS One 2017; 12:e0166061. [PMID: 28045902 PMCID: PMC5207445 DOI: 10.1371/journal.pone.0166061] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2016] [Accepted: 10/21/2016] [Indexed: 12/31/2022] Open
Abstract
Many blood-borne substances attempting to pass through the luminal membrane of brain endothelial cells are acted upon by a variety of metabolizing enzymes or are actively expelled back into the capillary lumen by embedded efflux transporters, such as Permeability-glycoprotein (Pgp). Overexpression of this protein has also been linked to multidrug resistance in cancer cells. Previous studies have shown that focused ultrasound (FUS), when combined with a microbubble agent, has ability to temporarily disrupt blood-brain barrier (BBBD). In this work, we investigated whether modulation of Pgp expression is part of the FUS-induced effects. We found that ultrasound can temporarily suppress Pgp expression. When BBBD was produced at 0.55 MPa, Pgp was suppressed up to 48 hours and restored by 72 hours. At 0.81 MPa, suppression can last 72 hours or longer. These findings support the idea that microbubble-enhanced FUS disrupts the functional components of the BBB through suppression of drug efflux.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Radiology, Brigham & Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Krisztina Fischer
- Department of Radiology, Brigham & Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Caroline Gentile
- Department of Radiology, Brigham & Women’s Hospital, Harvard Medical School, Boston, United States of America
- Department of Neurobiology, Harvard College, Cambridge, United States of America
| | | | - Yong-Zhi Zhang
- Department of Radiology, Brigham & Women’s Hospital, Harvard Medical School, Boston, United States of America
| | - Nathan McDannold
- Department of Radiology, Brigham & Women’s Hospital, Harvard Medical School, Boston, United States of America
| |
Collapse
|
116
|
Ai P, Wang H, Liu K, Wang T, Gu W, Ye L, Yan C. The relative length of dual-target conjugated on iron oxide nanoparticles plays a role in brain glioma targeting. RSC Adv 2017. [DOI: 10.1039/c7ra02102j] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Cy5.5-labeled, CTX/PEG-FA dual-target conjugated Fe3O4 NPs were prepared and the effect of dual-target relative length on glioma targeting was investigated.
Collapse
Affiliation(s)
- Penghui Ai
- Department of Neurosurgery
- Beijing Sanbo Brain Hospital
- Capital Medical University
- Beijing 100093
- P. R. China
| | - Hao Wang
- Department of Anatomy
- School of Basic Medical Sciences
- Capital Medical University
- Beijing 100069
- P. R. China
| | - Kang Liu
- School of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069
- P. R. China
| | - Tingjian Wang
- Department of Neurosurgery
- Beijing Sanbo Brain Hospital
- Capital Medical University
- Beijing 100093
- P. R. China
| | - Wei Gu
- School of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069
- P. R. China
| | - Ling Ye
- School of Pharmaceutical Sciences
- Capital Medical University
- Beijing 100069
- P. R. China
| | - Changxiang Yan
- Department of Neurosurgery
- Beijing Sanbo Brain Hospital
- Capital Medical University
- Beijing 100093
- P. R. China
| |
Collapse
|
117
|
Yoon HS, Chang C, Jang JH, Bhuyan A, Choe JW, Nikoozadeh A, Watkins RD, Stephens DN, Butts Pauly K, Khuri-Yakub BT. Ex Vivo HIFU Experiments Using a $32 \times 32$ -Element CMUT Array. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2016; 63:2150-2158. [PMID: 27913330 PMCID: PMC5241055 DOI: 10.1109/tuffc.2016.2606126] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/17/2023]
Abstract
High-intensity focused ultrasound (HIFU) has been used as noninvasive treatment for various diseases. For these therapeutic applications, capacitive micromachined ultrasonic transducers (CMUTs) have advantages that make them potentially preferred transducers over traditional piezoelectric transducers. In this paper, we present the design and the fabrication process of an 8 ×8 -mm 2 32 ×32 -element 2-D CMUT array for HIFU applications. To reduce the system complexity for addressing the 1024 transducer elements, we propose to group the CMUT array elements into eight HIFU channels based on the phase delay from the CMUT element to the targeted focal point. Designed to focus at an 8-mm depth with a 5-MHz exciting frequency, this grouping scheme was realized using a custom application-specific integrated circuit. With a 40-V dc bias and a 60-V peak-to-peak ac excitation, the surface pressure was measured 1.2 MPa peak-to-peak and stayed stable for a long enough time to create a lesion. With this dc and ac voltage combination, the measured peak-to-peak output pressure at the focus was 8.5 MPa, which is expected to generate a lesion in a minute according to the temperature simulation. The following ex vivo tissue experiments successfully demonstrated its capability to make lesions in both bovine muscle and liver tissue.
Collapse
|
118
|
Wang F, Li C, Cheng J, Yuan Z. Recent Advances on Inorganic Nanoparticle-Based Cancer Therapeutic Agents. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2016; 13:E1182. [PMID: 27898016 PMCID: PMC5201323 DOI: 10.3390/ijerph13121182] [Citation(s) in RCA: 72] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/25/2016] [Revised: 11/21/2016] [Accepted: 11/22/2016] [Indexed: 12/19/2022]
Abstract
Inorganic nanoparticles have been widely investigated as therapeutic agents for cancer treatments in biomedical fields due to their unique physical/chemical properties, versatile synthetic strategies, easy surface functionalization and excellent biocompatibility. This review focuses on the discussion of several types of inorganic nanoparticle-based cancer therapeutic agents, including gold nanoparticles, magnetic nanoparticles, upconversion nanoparticles and mesoporous silica nanoparticles. Several cancer therapy techniques are briefly introduced at the beginning. Emphasis is placed on how these inorganic nanoparticles can provide enhanced therapeutic efficacy in cancer treatment through site-specific accumulation, targeted drug delivery and stimulated drug release, with elaborations on several examples to highlight the respective strategies adopted. Finally, a brief summary and future challenges are included.
Collapse
Affiliation(s)
- Fenglin Wang
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China.
| | - Chengyao Li
- School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou 510515, China.
| | - Jing Cheng
- Hunan Key Laboratory of Food Safety Science & Technology, Technology Center of Hunan Entry-Exit Inspection and Quarantine Bureau, Hunan Academy of Inspection and Quarantine, Changsha 410004, China.
| | - Zhiqin Yuan
- State Key Laboratory of Chemical Resource Engineering, Beijing University of Chemical Technology, Beijing 100029, China.
| |
Collapse
|
119
|
Nanomaterial-based in vitro analytical system for diagnosis and therapy in microfluidic device. BIOCHIP JOURNAL 2016. [DOI: 10.1007/s13206-016-0409-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
120
|
Vegerhof A, Barnoy EA, Motiei M, Malka D, Danan Y, Zalevsky Z, Popovtzer R. Targeted Magnetic Nanoparticles for Mechanical Lysis of Tumor Cells by Low-Amplitude Alternating Magnetic Field. MATERIALS 2016; 9:ma9110943. [PMID: 28774062 PMCID: PMC5457194 DOI: 10.3390/ma9110943] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 10/30/2016] [Accepted: 11/17/2016] [Indexed: 11/16/2022]
Abstract
Currently available cancer therapies can cause damage to healthy tissue. We developed a unique method for specific mechanical lysis of cancer cells using superparamagnetic iron oxide nanoparticle rotation under a weak alternating magnetic field. Iron oxide core nanoparticles were coated with cetuximab, an anti-epidermal growth factor receptor antibody, for specific tumor targeting. Nude mice bearing a head and neck tumor were treated with cetuximab-coated magnetic nanoparticles (MNPs) and then received a 30 min treatment with a weak external alternating magnetic field (4 Hz) applied on alternating days (total of seven treatments, over 14 days). This treatment, compared to a pure antibody, exhibited a superior cell death effect over time. Furthermore, necrosis in the tumor site was detected by magnetic resonance (MR) images. Thermal camera images of head and neck squamous cell carcinoma cultures demonstrated that cell death occurred purely by a mechanical mechanism.
Collapse
Affiliation(s)
- Adi Vegerhof
- Faculty of Engineering & The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Eran A Barnoy
- Faculty of Engineering & The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Menachem Motiei
- Faculty of Engineering & The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Dror Malka
- Faculty of Engineering Holon Institute of Technology, Holon 5810201, Israel.
| | - Yossef Danan
- Faculty of Engineering & The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Zeev Zalevsky
- Faculty of Engineering & The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| | - Rachela Popovtzer
- Faculty of Engineering & The Institute of Nanotechnology and Advanced Materials, Bar-Ilan University, Ramat-Gan 5290002, Israel.
| |
Collapse
|
121
|
Tsai MT, Li DR, Chan MC. Non-invasive image-guided laser microsurgery by a dual-wavelength fiber laser and an integrated fiber-optic multi-modal system. OPTICS LETTERS 2016; 41:4847-4850. [PMID: 28005846 DOI: 10.1364/ol.41.004847] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/28/2023]
Abstract
A new approach to non-invasive image-guided laser micro-treatment is demonstrated by a dual-wavelength fiber laser source and an integrated fiber-based multi-modal system. The fiber-based source, operated in 1.55 and 1.2 μm simultaneously, was directly connected to an integrated fiber-based multi-modal system for imaging and laser micro-treatment at the same time. The 1.2 μm radiations, within the 1.2-1.35 μm bio-penetration window of skin, were utilized for spectral domain optical coherence tomography imaging. The 1.55 μm radiations, highly absorptive to waters, were utilized for laser microsurgery. The new approach, which is simple in configuration and accurately controls the positions and exposure time of the laser microsurgery, shows great promises for future clinical applications.
Collapse
|
122
|
Sherwood J, Lovas K, Rich M, Yin Q, Lackey K, Bolding MS, Bao Y. Shape-dependent cellular behaviors and relaxivity of iron oxide-based T 1 MRI contrast agents. NANOSCALE 2016; 8:17506-17515. [PMID: 27714177 DOI: 10.1039/c6nr06158c] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Recent research efforts about iron oxide nanoparticles has focused on the development of iron oxide-based T1 contrast agents for magnetic resonance imaging (MRI), such as ultrasmall iron oxide nanospheres (USNPs <4 nm) and ultrathin nanowires (NW, diameter <4 nm). In this paper, we report the cellular uptake behaviors of these two types of ultrasmall scale nanostructures on HepG2 cells. Both these two nanostructures were functionalized with tannic acid and their physical and chemical properties were carefully analyzed before cellular tests. Both USNPs and NWs exhibited strong paramagnetic signals, a property suitable for T1 MRI contrast agents. The distinct shapes also caused much difference in their cellular uptake behaviors. Specifically, the uptake of USNPs was five times higher than that of NWs after 72 hours incubation. The shape-dependent cellular uptake can potentially lead to different blood circulation times, and subsequently different applications of these two types of ultrasmall nanostructures.
Collapse
Affiliation(s)
- J Sherwood
- Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA.
| | - K Lovas
- Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA.
| | - M Rich
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Q Yin
- Alabama Innovation and Mentoring of Entrepreneurs, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - K Lackey
- Department of Biological Science, The University of Alabama, Tuscaloosa, AL 35487, USA
| | - M S Bolding
- Department of Radiology, The University of Alabama at Birmingham, Birmingham, AL 35233, USA.
| | - Y Bao
- Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL 35487, USA.
| |
Collapse
|
123
|
Park J, Aryal M, Vykhodtseva N, Zhang YZ, McDannold N. Evaluation of permeability, doxorubicin delivery, and drug retention in a rat brain tumor model after ultrasound-induced blood-tumor barrier disruption. J Control Release 2016; 250:77-85. [PMID: 27742444 DOI: 10.1016/j.jconrel.2016.10.011] [Citation(s) in RCA: 103] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2016] [Revised: 08/09/2016] [Accepted: 10/11/2016] [Indexed: 02/07/2023]
Abstract
Drug delivery in brain tumors is challenging because of the presence of blood-brain barrier (BBB) and the blood-tumor barrier (BTB). Focused ultrasound (FUS) combined with microbubbles can enhance the permeability of the BTB in brain tumors, as well as disrupting the BBB in the surrounding tissue. In this study, dynamic contrast-enhanced Magnetic Resonance Imaging (DCE-MRI) was used to characterize FUS-induced permeability changes in a rat glioma model and in the normal brain and to investigate the relationship between these changes and the resulting concentration of the chemotherapy agent doxorubicin (DOX). 9L gliosarcoma cells were implanted in both hemispheres in male rats. At day 10-12 after implantation, FUS-induced BTB disruption using 690kHz ultrasound and Definity microbubbles was performed in one of the tumors and in a normal brain region in each animal. After FUS, DOX was administered at a dose of 5.67mg/kg. The resulting DOX concentration was measured via fluorometry at 1 or 24h after FUS. The transfer coefficient Ktrans describing extravasation of the MRI contrast agent Gd-DTPA was significantly increased in both the sonicated tumors and in the normal brain tissue (P<0.001) between the two DCE-MRI acquisitions obtained before and after FUS, while no significant difference was found in the controls (non-sonicated tumor/normal brain tissue). DOX concentrations were also significantly larger than controls in both the sonicated tumors and in the normal tissue volumes at 1 and 24h after sonication. The DOX concentrations were significantly larger (P<0.01) in the control tumors harvested 1h after FUS than in those harvested at 24h, when the tumor concentrations were not significantly different than in the non-sonicated normal brain. In contrast, there was no significant difference in the DOX concentrations between the tumors harvested at 1 and 24h after FUS or in the concentrations measured in the brain at these time points. The transfer coefficient Ktrans for Gd-DTPA and the drug concentrations showed a good linear correlation (R2=0.56). Overall, these data suggest that FUS and microbubbles can not only increase DOX delivery across the BBB and BTB, but that it is retained in the tissue at significantly enhanced levels for at least 24h. Such enhanced retention may increase the potency of this chemotherapy agent and allow for reduced systemic doses. Furthermore, MRI-based estimates of Gd-DTPA transport across these barriers might be useful to estimate local DOX concentrations in the tumor and in the surrounding normal tissue.
Collapse
Affiliation(s)
- Juyoung Park
- Medical Device Development Center, Daegu-Gyeongbuk, Medical Innovation Foundation, Daegu, Republic of Korea
| | - 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
|
124
|
Concepts, technologies, and practices for drug delivery past the blood–brain barrier to the central nervous system. J Control Release 2016; 240:251-266. [DOI: 10.1016/j.jconrel.2015.12.041] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2015] [Revised: 12/21/2015] [Accepted: 12/23/2015] [Indexed: 12/29/2022]
|
125
|
Qian X, Zheng Y, Chen Y. Micro/Nanoparticle-Augmented Sonodynamic Therapy (SDT): Breaking the Depth Shallow of Photoactivation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2016; 28:8097-8129. [PMID: 27384408 DOI: 10.1002/adma.201602012] [Citation(s) in RCA: 504] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 05/28/2016] [Indexed: 05/08/2023]
Abstract
The fast development of photoactivation for cancer treatment provides an efficient photo-therapeutic strategy for cancer treatment, but traditional photodynamic or photothermal therapy suffers from the critical issue of low in vivo penetration depth of tissues. As a non-invasive therapeutic modality, sonodynamic therapy (SDT) can break the depth barrier of photoactivation because ultrasound has an intrinsically high tissue-penetration performance. Micro/nanoparticles can efficiently augment the SDT efficiency based on nanobiotechnology. The state-of-art of the representative achievements on micro/nanoparticle-enhanced SDT is summarized, and specific functions of micro/nanoparticles for SDT are discussed, from the different viewpoints of ultrasound medicine, material science and nanobiotechnology. Emphasis is put on the relationship of structure/composition-SDT performance of micro/nanoparticle-based sonosensitizers. Three types of micro/nanoparticle-augmented SDT are discussed, including organic and inorganic sonosensitizers and micro/nanoparticle-based but sonosensitizer-free strategies to enhance the SDT outcome. SDT-based synergistic cancer therapy augmented by micro/nanoparticles and their biosafety are also included. Some urgent critical issues and potential developments of micro/nanoparticle-augmented SDT for efficient cancer treatment are addressed. It is highly expected that micro/nanoparticle-augmented SDT will be quickly developed as a new and efficient therapeutic modality which will find practical applications in cancer treatment. At the same time, fundamental disciplines regarding materials science, chemistry, medicine and nanotechnology will be advanced.
Collapse
Affiliation(s)
- Xiaoqin Qian
- Department of Ultrasound, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, 212002, P. R. China
| | - Yuanyi Zheng
- Sixth Affiliated Hospital of Shanghai Jiaotong University & Shanghai Institute of Ultrasound in Medicine, Shanghai, 200233, P. R. China.
| | - Yu Chen
- State Key Laboratory of High Performance Ceramic and Superfine Microstructures, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, P. R. China.
| |
Collapse
|
126
|
Focused Ultrasound-Induced Blood-Brain Barrier Opening: Association with Mechanical Index and Cavitation Index Analyzed by Dynamic Contrast-Enhanced Magnetic-Resonance Imaging. Sci Rep 2016; 6:33264. [PMID: 27630037 PMCID: PMC5024096 DOI: 10.1038/srep33264] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2016] [Accepted: 08/22/2016] [Indexed: 01/07/2023] Open
Abstract
Focused ultrasound (FUS) with microbubbles can temporally open the blood-brain barrier (BBB), and the cavitation activities of microbubbles play a key role in the BBB-opening process. Previous attempts used contrast-enhanced magnetic resonance imaging (CE-MRI) to correlate the mechanical index (MI) with the scale of BBB-opening, but MI only partially gauged acoustic activities, and CE-MRI did not fully explore correlations of pharmacodynamic/pharmacokinetic behaviors. Recently, the cavitation index (CI) has been derived to serve as an indicator of microbubble-ultrasound stable cavitation, and may also serve as a valid indicator to gauge the level of FUS-induced BBB opening. This study investigates the feasibility of gauging FUS-induced BBB opened level via the two indexes, MI and CI, through dynamic contrast-enhanced (DCE)-MRI analysis as well as passive cavitation detection (PCD) analysis. Pharmacodynamic/pharmacokinetic parameters derived from DCE-MRI were characterized to identify the scale of FUS-induced BBB opening. Our results demonstrated that DCE-MRI can successfully access pharmacodynamic/pharmacokinetic BBB-opened behavior, and was highly correlated both with MI and CI, implying the feasibility in using these two indices to gauge the scale of FUS-induced BBB opening. The proposed finding may facilitate the design toward using focused ultrasound as a safe and reliable noninvasive CNS drug delivery.
Collapse
|
127
|
Non-invasive focused ultrasound-based synergistic treatment of brain tumors. JOURNAL OF CANCER RESEARCH AND PRACTICE 2016. [DOI: 10.1016/j.jcrpr.2016.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
|
128
|
Xu HL, Mao KL, Lu CT, Fan ZL, Yang JJ, Xu J, Chen PP, ZhuGe DL, Shen BX, Jin BH, Xiao J, Zhao YZ. An injectable acellular matrix scaffold with absorbable permeable nanoparticles improves the therapeutic effects of docetaxel on glioblastoma. Biomaterials 2016; 107:44-60. [PMID: 27614158 DOI: 10.1016/j.biomaterials.2016.08.026] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 08/13/2016] [Accepted: 08/16/2016] [Indexed: 10/21/2022]
Abstract
Intratumoral drug delivery (IT) is an inherently appealing approach for concentrating toxic chemotherapies at the site of action. However, for most chemotherapies, poor tumor penetration and short retention at the administration site limit their anti-tumor effects. In this work, we describe permeable nanoparticles (NPs) prepared with a novel amphiphilic polymer, RRR-α-tocopheryl succinate-grafted-ε-polylysine conjugate (VES-g-ε-PLL). The nanoparticles (NPs) of VES-g-ε-PLL exhibited an ultra-small hydrodynamic diameter (20.8 nm) and positive zeta potential (20.6 mV), which facilitate strong glioma spheroid penetration ability in vitro. Additionally, the hydrophobic model drug docetaxel (DTX) could be effectively encapsulated in the nanoparticles with 3.99% drug loading and 73.37% encapsulation efficiency. To prolong the retention time of DTX-loaded nanoparticles (DTX-NPs) in the tumor, intact decellularized brain extracellular matrix (dBECM) derived from healthy rats was used as a drug depot to adsorb the ultra-small DTX-NPs. The intact DTX-NPs-adsorbing dBECM scaffold was further homogenized into an injectable DTX-NPs-dBECM suspension for intratumoral administration. The DTX-NPs-dBECM suspension exhibited slower DTX release than naked DTX-NPs without compromising the tumor penetration ability of DTX-NPs. An antitumor study showed that the DTX-NPs-dBECM suspension exhibited more powerful in vitro inhibition of tumor spheroid growth than free DTX solution or DTX-NPs. Due to strong tumor penetration ability and prolonged retention, DTX-NPs-dBECM led to complete suppression of glioma growth in vivo at 28 days after treatment. The therapeutic mechanism was due to enhanced proliferation inhibition and apoptosis of tumor cells and angiogenesis inhibition of glioma after treatment with DTX-NPs-dBECM. Finally, the safety of DTX-NPs-dBECM at the therapeutic dose was demonstrated via pathological HE assay from heart, liver, spleen, lung and kidney tissues. In conclusion, permeable nanoparticle-absorbing dBECM is a potential carrier for intratumoral delivery of common chemotherapeutics.
Collapse
Affiliation(s)
- He-Lin Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Kai-Li Mao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Cui-Tao Lu
- The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China.
| | - Zi-Liang Fan
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Jing-Jing Yang
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Jie Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Pian-Pian Chen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - De-Li ZhuGe
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Bi-Xin Shen
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Bing-Hui Jin
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China
| | - Jian Xiao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China.
| | - Ying-Zheng Zhao
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China; The Second Affiliated Hospital, Wenzhou Medical University, Wenzhou, Zhejiang Province 325035, China.
| |
Collapse
|
129
|
Wei KC, Lin FW, Huang CY, Ma CCM, Chen JY, Feng LY, Yang HW. 1,3-Bis(2-chloroethyl)-1-nitrosourea-loaded bovine serum albumin nanoparticles with dual magnetic resonance-fluorescence imaging for tracking of chemotherapeutic agents. Int J Nanomedicine 2016; 11:4065-75. [PMID: 27601895 PMCID: PMC5003552 DOI: 10.2147/ijn.s113589] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
To date, knowing how to identify the location of chemotherapeutic agents in the human body after injection is still a challenge. Therefore, it is urgent to develop a drug delivery system with molecular imaging tracking ability to accurately understand the distribution, location, and concentration of a drug in living organisms. In this study, we developed bovine serum albumin (BSA)-based nanoparticles (NPs) with dual magnetic resonance (MR) and fluorescence imaging modalities (fluorescein isothiocyanate [FITC]-BSA-Gd/1,3-bis(2-chloroethyl)-1-nitrosourea [BCNU] NPs) to deliver BCNU for inhibition of brain tumor cells (MBR 261-2). These BSA-based NPs are water dispersible, stable, and biocompatible as confirmed by XTT cell viability assay. In vitro phantoms and in vivo MR and fluorescence imaging experiments show that the developed FITC-BSA-Gd/BCNU NPs enable dual MR and fluorescence imaging for monitoring cellular uptake and distribution in tumors. The T1 relaxivity (R1) of FITC-BSA-Gd/BCNU NPs was 3.25 mM(-1) s(-1), which was similar to that of the commercial T1 contrast agent (R1 =3.36 mM(-1) s(-1)). The results indicate that this multifunctional drug delivery system has potential bioimaging tracking of chemotherapeutic agents ability in vitro and in vivo for cancer therapy.
Collapse
Affiliation(s)
- Kuo-Chen Wei
- Department of Neurosurgery, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan
| | - Feng-Wei Lin
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung
| | - Chiung-Yin Huang
- Department of Neurosurgery, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan
| | - Chen-Chi M Ma
- Department of Chemical Engineering, National Tsing Hua University, Hsinchu, Taiwan, Republic of China
| | - Ju-Yu Chen
- Department of Neurosurgery, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan
| | - Li-Ying Feng
- Department of Neurosurgery, Chang Gung Memorial Hospital, School of Medicine, Chang Gung University, Taoyuan
| | - Hung-Wei Yang
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung
| |
Collapse
|
130
|
Xu HL, Mao KL, Huang YP, Yang JJ, Xu J, Chen PP, Fan ZL, Zou S, Gao ZZ, Yin JY, Xiao J, Lu CT, Zhang BL, Zhao YZ. Glioma-targeted superparamagnetic iron oxide nanoparticles as drug-carrying vehicles for theranostic effects. NANOSCALE 2016; 8:14222-14236. [PMID: 27396404 DOI: 10.1039/c6nr02448c] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
Multifunctional nanoparticles capable of the specific delivery of therapeutics to diseased cells and the real-time imaging of these sites have the potential to improve cancer treatment through personalized therapy. In this study, we have proposed a multifunctional nanoparticle that integrate magnetic targeting, drug-carrier functionality and real-time MRI imaging capabilities in one platform for the theranostic treatment of tumors. The multifunctional nanoparticle was designed with a superparamagnetic iron oxide core and a multifunctional shell composed of PEG/PEI/polysorbate 80 (Ps 80) and was used to encapsulate DOX. DOX-loaded multifunctional nanoparticles (DOX@Ps 80-SPIONs) with a Dh of 58.0 nm, a zeta potential of 28.0 mV, and a drug loading content of 29.3% presented superior superparamagnetic properties with a saturation magnetization (Ms) of 24.1 emu g(-1). The cellular uptake of DOX@Ps 80-SPIONs by C6 cells under a magnetic field was significantly enhanced over that of free DOX in solution, resulting in stronger in vitro cytotoxicity. The real-time therapeutic outcome of DOX@Ps 80-SPIONs was easily monitored by MRI. Furthermore, the negative contrast enhancement effect of the nanoparticles was confirmed in glioma-bearing rats. Prussian blue staining and ex vivo DOX fluorescence assays showed that the magnetic Ps 80-SPIONs and encapsulated DOX were delivered to gliomas by imposing external magnetic fields, indicating effective magnetic targeting. Due to magnetic targeting and Ps 80-mediated endocytosis, DOX@Ps 80-SPIONs in the presence of a magnetic field led to the complete suppression of glioma growth in vivo at 28 days after treatment. The therapeutic mechanism of DOX@Ps 80-SPIONs acted by inducing apoptosis through the caspase-3 pathway. Finally, DOX@Ps 80-SPIONs' safety at therapeutic dosage was verified using pathological HE assays of the heart, liver, spleen, lung and kidney. Multifunctional SPIONs could be used as potential carriers for the theranostic treatment of CNS diseases.
Collapse
Affiliation(s)
- He-Lin Xu
- School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou City, Zhejiang Province 325035, China.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Nair M, Jayant RD, Kaushik A, Sagar V. Getting into the brain: Potential of nanotechnology in the management of NeuroAIDS. Adv Drug Deliv Rev 2016; 103:202-217. [PMID: 26944096 PMCID: PMC4935582 DOI: 10.1016/j.addr.2016.02.008] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 02/17/2016] [Accepted: 02/18/2016] [Indexed: 12/18/2022]
Abstract
In spite of significant advances in antiretroviral (ARV) therapy, the elimination of human immunodeficiency virus (HIV) reservoirs from the periphery and the central nervous system (CNS) remains a formidable task. The incapability of ARV to go across the blood-brain barrier (BBB) after systemic administration makes the brain one of the dominant HIV reservoirs. Thus, screening, monitoring, and elimination of HIV reservoirs from the brain remain a clinically daunting and key task. The practice and investigation of nanomedicine possesses potentials for therapeutics against neuroAIDS. This review highlights the advancements in nanoscience and nanotechnology to design and develop specific size therapeutic cargo for efficient navigation across BBB so as to recognize and eradicate HIV brain reservoirs. Different navigation and drug release strategies, their biocompatibility and efficacy with related challenges and future prospects are also discussed. This review would be an excellent platform to understand nano-enable multidisciplinary research to formulate efficient nanomedicine for the management of neuroAIDS.
Collapse
Key Words
- Anti-retroviral (ARV) therapy
- Blood–brain barrier (BBB)
- Bradykinin (PubChem CID: 439,201)
- CNS drug delivery
- Enfuvirtide (PubChem CID: 16,130,199), Lamivudine & Zidovudine (PubChem CID: 160,352)
- Ferrous oxide or iron (II) oxide (PubChem CID: 14,945)
- Foscarnet sodium (PubChem CID: 44,561)
- HIV monitoring
- HIV-1
- Magnetic nanoparticle
- Mannitol (PubChem CID: 6251)
- Nanotechnology
- Neopterin (PubChem CID: 4455)
- NeuroAIDS
- Pluronic-P85 (PubChem CID: 24,751)
- Saquinavir mesylate (PubChem CID: 60,934)
- Tenofovir disoproxil fumarate (PubChem CID: 6,398,764)
Collapse
Affiliation(s)
- Madhavan Nair
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Rahul Dev Jayant
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA.
| | - Ajeet Kaushik
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| | - Vidya Sagar
- Center for Personalized Nanomedicine, Institute of NeuroImmune Pharmacology, Department of Immunology, Herbert Wertheim College of Medicine, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
132
|
Lin CY, Hsieh HY, Chen CM, Wu SR, Tsai CH, Huang CY, Hua MY, Wei KC, Yeh CK, Liu HL. Non-invasive, neuron-specific gene therapy by focused ultrasound-induced blood-brain barrier opening in Parkinson's disease mouse model. J Control Release 2016; 235:72-81. [DOI: 10.1016/j.jconrel.2016.05.052] [Citation(s) in RCA: 70] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 04/28/2016] [Accepted: 05/24/2016] [Indexed: 02/01/2023]
|
133
|
Appelboom G, Detappe A, LoPresti M, Kunjachan S, Mitrasinovic S, Goldman S, Chang SD, Tillement O. Stereotactic modulation of blood-brain barrier permeability to enhance drug delivery. Neuro Oncol 2016; 18:1601-1609. [PMID: 27407134 DOI: 10.1093/neuonc/now137] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2016] [Accepted: 05/23/2016] [Indexed: 12/14/2022] Open
Abstract
Drug delivery in the CNS is limited by endothelial tight junctions forming the impermeable blood-brain barrier. The development of new treatment paradigms has previously been hampered by the restrictiveness of the blood-brain barrier to systemically administered therapeutics. With recent advances in stereotactic localization and noninvasive imaging, we have honed the ability to modulate, ablate, and rewire millimetric brain structures to precisely permeate the impregnable barrier. The wide range of focused radiations offers endless possibilities to disrupt endothelial permeability with different patterns and intensity following 3-dimensional coordinates offering a new world of possibilities to access the CNS, as well as to target therapies. We propose a review of the current state of knowledge in targeted drug delivery using noninvasive image-guided approaches. To this end, we focus on strategies currently used in clinics or in clinical trials such as targeted radiotherapy and magnetic resonance guided focused ultrasound, but also on more experimental approaches such as magnetically heated nanoparticles, electric fields, and lasers, techniques which demonstrated remarkable results both in vitro and in vivo. We envision that biodistribution and efficacy of systemically administered drugs will be enhanced with further developments of these promising strategies. Besides therapeutic applications, stereotactic platforms can be highly valuable in clinical applications for interventional strategies that can improve the targetability and efficacy of drugs and macromolecules. It is our hope that by showcasing and reviewing the current state of this field, we can lay the groundwork to guide future research in this realm.
Collapse
Affiliation(s)
- Geoff Appelboom
- Department of Neurosurgery, Stanford Medical Center, Stanford, California (G.A., S.D.C.); Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.D., S.K.); Institut Lumière Matière, Université Claude Bernard Lyon 1, Villeurbanne, France (A.D., O.T.); Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (M.L.); Department of Neurological Surgery, Columbia University Medical Center, New York, New York (S.M.); Department of Nuclear Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium (S.G.)
| | - Alexandre Detappe
- Department of Neurosurgery, Stanford Medical Center, Stanford, California (G.A., S.D.C.); Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.D., S.K.); Institut Lumière Matière, Université Claude Bernard Lyon 1, Villeurbanne, France (A.D., O.T.); Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (M.L.); Department of Neurological Surgery, Columbia University Medical Center, New York, New York (S.M.); Department of Nuclear Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium (S.G.)
| | - Melissa LoPresti
- Department of Neurosurgery, Stanford Medical Center, Stanford, California (G.A., S.D.C.); Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.D., S.K.); Institut Lumière Matière, Université Claude Bernard Lyon 1, Villeurbanne, France (A.D., O.T.); Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (M.L.); Department of Neurological Surgery, Columbia University Medical Center, New York, New York (S.M.); Department of Nuclear Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium (S.G.)
| | - Sijumon Kunjachan
- Department of Neurosurgery, Stanford Medical Center, Stanford, California (G.A., S.D.C.); Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.D., S.K.); Institut Lumière Matière, Université Claude Bernard Lyon 1, Villeurbanne, France (A.D., O.T.); Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (M.L.); Department of Neurological Surgery, Columbia University Medical Center, New York, New York (S.M.); Department of Nuclear Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium (S.G.)
| | - Stefan Mitrasinovic
- Department of Neurosurgery, Stanford Medical Center, Stanford, California (G.A., S.D.C.); Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.D., S.K.); Institut Lumière Matière, Université Claude Bernard Lyon 1, Villeurbanne, France (A.D., O.T.); Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (M.L.); Department of Neurological Surgery, Columbia University Medical Center, New York, New York (S.M.); Department of Nuclear Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium (S.G.)
| | - Serge Goldman
- Department of Neurosurgery, Stanford Medical Center, Stanford, California (G.A., S.D.C.); Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.D., S.K.); Institut Lumière Matière, Université Claude Bernard Lyon 1, Villeurbanne, France (A.D., O.T.); Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (M.L.); Department of Neurological Surgery, Columbia University Medical Center, New York, New York (S.M.); Department of Nuclear Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium (S.G.)
| | - Steve D Chang
- Department of Neurosurgery, Stanford Medical Center, Stanford, California (G.A., S.D.C.); Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.D., S.K.); Institut Lumière Matière, Université Claude Bernard Lyon 1, Villeurbanne, France (A.D., O.T.); Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (M.L.); Department of Neurological Surgery, Columbia University Medical Center, New York, New York (S.M.); Department of Nuclear Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium (S.G.)
| | - Olivier Tillement
- Department of Neurosurgery, Stanford Medical Center, Stanford, California (G.A., S.D.C.); Department of Radiation Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts (A.D., S.K.); Institut Lumière Matière, Université Claude Bernard Lyon 1, Villeurbanne, France (A.D., O.T.); Department of Neurosurgery, Baylor College of Medicine, Houston, Texas (M.L.); Department of Neurological Surgery, Columbia University Medical Center, New York, New York (S.M.); Department of Nuclear Medicine, Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium (S.G.)
| |
Collapse
|
134
|
Gao H. Progress and perspectives on targeting nanoparticles for brain drug delivery. Acta Pharm Sin B 2016; 6:268-86. [PMID: 27471668 PMCID: PMC4951594 DOI: 10.1016/j.apsb.2016.05.013] [Citation(s) in RCA: 294] [Impact Index Per Article: 36.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 05/06/2016] [Accepted: 05/09/2016] [Indexed: 02/06/2023] Open
Abstract
Due to the ability of the blood-brain barrier (BBB) to prevent the entry of drugs into the brain, it is a challenge to treat central nervous system disorders pharmacologically. The development of nanotechnology provides potential to overcome this problem. In this review, the barriers to brain-targeted drug delivery are reviewed, including the BBB, blood-brain tumor barrier (BBTB), and nose-to-brain barrier. Delivery strategies are focused on overcoming the BBB, directly targeting diseased cells in the brain, and dual-targeted delivery. The major concerns and perspectives on constructing brain-targeted delivery systems are discussed.
Collapse
|
135
|
Fan CH, Cheng YH, Ting CY, Ho YJ, Hsu PH, Liu HL, Yeh CK. Ultrasound/Magnetic Targeting with SPIO-DOX-Microbubble Complex for Image-Guided Drug Delivery in Brain Tumors. Theranostics 2016; 6:1542-56. [PMID: 27446489 PMCID: PMC4955054 DOI: 10.7150/thno.15297] [Citation(s) in RCA: 86] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 04/27/2016] [Indexed: 11/18/2022] Open
Abstract
One of the greatest challenges in the deployment of chemotherapeutic drugs against brain tumors is ensuring that sufficient drug concentrations reach the tumor, while minimizing drug accumulation at undesired sites. Recently, injection of therapeutic agents following blood-brain barrier (BBB) opening by focused ultrasound (FUS) with microbubbles (MBs) has been shown to enhance drug delivery in targeted brain regions. Nevertheless, the distribution and quantitative deposition of agents delivered to the brain are still hard to estimate. Based on our previous work on superparamagnetic iron oxide (SPIO)-loaded MBs, we present a novel theranostic complex of SPIO-Doxorubicin (DOX)-conjugated MB (SD-MB) for drug delivery to the brain. Magnetic labeling of the drug enables direct visualization via magnetic resonance imaging, and also facilitates magnetic targeting (MT) to actively enhance targeted deposition of the drug. In a rat glioma model, we demonstrated that FUS sonication can be used with SD-MBs to simultaneously facilitate BBB opening and allow dual ultrasound/magnetic targeting of chemotherapeutic agent (DOX) delivery. The accumulation of SD complex within brain tumors can be significantly enhanced by MT (25.7 fold of DOX, 7.6 fold of SPIO). The change in relaxation rate R2 (1/T2) within tumors was highly correlated with SD deposition as quantified by high performance liquid chromatography (R2 = 0.93) and inductively coupled plasma-atomic emission spectroscopy (R2 = 0.94), demonstrating real-time monitoring of DOX distribution. Our results suggest that SD-MBs can serve as multifunction agents to achieve advanced molecular theranostics.
Collapse
|
136
|
Liu H, Tan Y, Xie L, Yang L, Zhao J, Bai J, Huang P, Zhan W, Wan Q, Zou C, Han Y, Wang Z. Self-assembled dual-modality contrast agents for non-invasive stem cell tracking via near-infrared fluorescence and magnetic resonance imaging. J Colloid Interface Sci 2016; 478:217-26. [PMID: 27299677 DOI: 10.1016/j.jcis.2016.05.063] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2016] [Revised: 05/26/2016] [Accepted: 05/27/2016] [Indexed: 01/15/2023]
Abstract
Stem cells hold great promise for treating various diseases. However, one of the main drawbacks of stem cell therapy is the lack of non-invasive image-tracking technologies. Although magnetic resonance imaging (MRI) and near-infrared fluorescence (NIRF) imaging have been employed to analyse cellular and subcellular events via the assistance of contrast agents, the sensitivity and temporal resolution of MRI and the spatial resolution of NIRF are still shortcomings. In this study, superparamagnetic iron oxide nanocrystals and IR-780 dyes were co-encapsulated in stearic acid-modified polyethylenimine to form a dual-modality contrast agent with nano-size and positive charge. These resulting agents efficiently labelled stem cells and did not influence the cellular viability and differentiation. Moreover, the labelled cells showed the advantages of dual-modality imaging in vivo.
Collapse
Affiliation(s)
- Hong Liu
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Key Laboratory for MRI, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China; School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong 510006, PR China
| | - Yan Tan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Key Laboratory for MRI, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Lisi Xie
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, Fujian 361005, PR China
| | - Lei Yang
- Laboratory for Gene and Cell Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Jing Zhao
- Laboratory for Gene and Cell Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Jingxuan Bai
- Laboratory for Gene and Cell Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Ping Huang
- Laboratory for Gene and Cell Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Wugen Zhan
- Laboratory for Gene and Cell Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Qian Wan
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Key Laboratory for MRI, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Chao Zou
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Key Laboratory for MRI, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Yali Han
- School of Chemical Engineering and Light Industry, Guangdong University of Technology, Guangzhou, Guangdong 510006, PR China.
| | - Zhiyong Wang
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Key Laboratory for MRI, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China.
| |
Collapse
|
137
|
Liang PC, Chen YC, Chiang CF, Mo LR, Wei SY, Hsieh WY, Lin WL. Doxorubicin-modified magnetic nanoparticles as a drug delivery system for magnetic resonance imaging-monitoring magnet-enhancing tumor chemotherapy. Int J Nanomedicine 2016; 11:2021-37. [PMID: 27274233 PMCID: PMC4869666 DOI: 10.2147/ijn.s94139] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
In this study, we developed functionalized superparamagnetic iron oxide (SPIO) nanoparticles consisting of a magnetic Fe3O4 core and a shell of aqueous stable polyethylene glycol (PEG) conjugated with doxorubicin (Dox) (SPIO-PEG-D) for tumor magnetic resonance imaging (MRI) enhancement and chemotherapy. The size of SPIO nanoparticles was ~10 nm, which was visualized by transmission electron microscope. The hysteresis curve, generated with vibrating-sample magnetometer, showed that SPIO-PEG-D was superparamagnetic with an insignificant hysteresis. The transverse relaxivity (r 2) for SPIO-PEG-D was significantly higher than the longitudinal relaxivity (r 1) (r 2/r 1 >10). The half-life of Dox in blood circulation was prolonged by conjugating Dox on the surface of SPIO with PEG to reduce its degradation. The in vitro experiment showed that SPIO-PEG-D could cause DNA crosslink more serious, resulting in a lower DNA expression and a higher cell apoptosis for HT-29 cancer cells. The Prussian blue staining study showed that the tumors treated with SPIO-PEG-D under a magnetic field had a much higher intratumoral iron density than the tumors treated with SPIO-PEG-D alone. The in vivo MRI study showed that the T2-weighted signal enhancement was stronger for the group under a magnetic field, indicating that it had a better accumulation of SPIO-PEG-D in tumor tissues. In the anticancer efficiency study for SPIO-PEG-D, the results showed that there was a significantly smaller tumor size for the group with a magnetic field than the group without. The in vivo experiments also showed that this drug delivery system combined with a local magnetic field could reduce the side effects of cardiotoxicity and hepatotoxicity. The results showed that the developed SPIO-PEG-D nanoparticles own a great potential for MRI-monitoring magnet-enhancing tumor chemotherapy.
Collapse
Affiliation(s)
- Po-Chin Liang
- Institute of Biomedical Engineering, College of Medicine, College of Engineering, National Taiwan University Hospital, Taipei, Taiwan
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Yung-Chu Chen
- Institute of Biomedical Engineering, College of Medicine, College of Engineering, National Taiwan University Hospital, Taipei, Taiwan
- Biomedical Technology and Device Research Labs, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Chi-Feng Chiang
- Institute of Biomedical Engineering, College of Medicine, College of Engineering, National Taiwan University Hospital, Taipei, Taiwan
| | - Lein-Ray Mo
- Division of Gastroenterology, Department of Internal Medicine, E-Da Hospital, Kaohsiung, Taiwan
| | - Shwu-Yuan Wei
- Department of Medical Imaging, National Taiwan University Hospital, Taipei, Taiwan
| | - Wen-Yuan Hsieh
- Biomedical Technology and Device Research Labs, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Win-Li Lin
- Institute of Biomedical Engineering, College of Medicine, College of Engineering, National Taiwan University Hospital, Taipei, Taiwan
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Miaoli, Taiwan
| |
Collapse
|
138
|
Liu H, Zhang J, Chen X, Du XS, Zhang JL, Liu G, Zhang WG. Application of iron oxide nanoparticles in glioma imaging and therapy: from bench to bedside. NANOSCALE 2016; 8:7808-7826. [PMID: 27029509 DOI: 10.1039/c6nr00147e] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Gliomas are the most common primary brain tumors and have a very dismal prognosis. However, recent advancements in nanomedicine and nanotechnology provide opportunities for personalized treatment regimens to improve the poor prognosis of patients suffering from glioma. This comprehensive review starts with an outline of the current status facing glioma. It then provides an overview of the state-of-the-art applications of iron oxide nanoparticles (IONPs) to glioma diagnostics and therapeutics, including MR contrast enhancement, drug delivery, cell labeling and tracking, magnetic hyperthermia treatment and magnetic particle imaging. It also addresses current challenges associated with the biological barriers and IONP design with an emphasis on recent advances and innovative approaches for glioma targeting strategies. Opportunities for future development are highlighted.
Collapse
Affiliation(s)
- Heng Liu
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China and State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Jun Zhang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China. and Sichuan Key Laboratory of Medical Imaging, Affiliated Hospital of North Sichuan Medical College, North Sichuan Medical College, Nanchong 637007, China
| | - Xiao Chen
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Xue-Song Du
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Jin-Long Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China.
| | - Wei-Guo Zhang
- Department of Radiology, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China and The State Key Laboratory of Trauma, Burns and Combined Injury, Institute of Surgery Research, Daping Hospital, Third Military Medical University, Chongqing, 400042, China.
| |
Collapse
|
139
|
Das S, Carnicer-Lombarte A, Fawcett JW, Bora U. Bio-inspired nano tools for neuroscience. Prog Neurobiol 2016; 142:1-22. [PMID: 27107796 DOI: 10.1016/j.pneurobio.2016.04.008] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2015] [Revised: 04/14/2016] [Accepted: 04/15/2016] [Indexed: 01/19/2023]
Abstract
Research and treatment in the nervous system is challenged by many physiological barriers posing a major hurdle for neurologists. The CNS is protected by a formidable blood brain barrier (BBB) which limits surgical, therapeutic and diagnostic interventions. The hostile environment created by reactive astrocytes in the CNS along with the limited regeneration capacity of the PNS makes functional recovery after tissue damage difficult and inefficient. Nanomaterials have the unique ability to interface with neural tissue in the nano-scale and are capable of influencing the function of a single neuron. The ability of nanoparticles to transcend the BBB through surface modifications has been exploited in various neuro-imaging techniques and for targeted drug delivery. The tunable topography of nanofibers provides accurate spatio-temporal guidance to regenerating axons. This review is an attempt to comprehend the progress in understanding the obstacles posed by the complex physiology of the nervous system and the innovations in design and fabrication of advanced nanomaterials drawing inspiration from natural phenomenon. We also discuss the development of nanomaterials for use in Neuro-diagnostics, Neuro-therapy and the fabrication of advanced nano-devices for use in opto-electronic and ultrasensitive electrophysiological applications. The energy efficient and parallel computing ability of the human brain has inspired the design of advanced nanotechnology based computational systems. However, extensive use of nanomaterials in neuroscience also raises serious toxicity issues as well as ethical concerns regarding nano implants in the brain. In conclusion we summarize these challenges and provide an insight into the huge potential of nanotechnology platforms in neuroscience.
Collapse
Affiliation(s)
- Suradip Das
- Bioengineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India
| | - Alejandro Carnicer-Lombarte
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Robinson Way, Cambridge CB2 0PY, United Kingdom
| | - James W Fawcett
- John Van Geest Centre for Brain Repair, Department of Clinical Neurosciences, University of Cambridge, Robinson Way, Cambridge CB2 0PY, United Kingdom
| | - Utpal Bora
- Bioengineering Laboratory, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India; Mugagen Laboratories Private Limited, Technology Incubation Complex, Indian Institute of Technology Guwahati, Guwahati 781039, Assam, India.
| |
Collapse
|
140
|
Tsai CH, Zhang JW, Liao YY, Liu HL. Real-time monitoring of focused ultrasound blood-brain barrier opening via subharmonic acoustic emission detection: implementation of confocal dual-frequency piezoelectric transducers. Phys Med Biol 2016; 61:2926-46. [PMID: 26988240 DOI: 10.1088/0031-9155/61/7/2926] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Burst-tone focused ultrasound exposure in the presence of microbubbles has been demonstrated to be effective at inducing temporal and local opening of the blood-brain barrier (BBB), which promises significant clinical potential to deliver therapeutic molecules into the central nervous system (CNS). Traditional contrast-enhanced imaging confirmation after focused ultrasound (FUS) exposure serves as a post-operative indicator of the effectiveness of FUS-BBB opening, however, an indicator that can concurrently report the BBB status and BBB-opening effectiveness is required to provide effective feedback to implement this treatment clinically. In this study, we demonstrate the use of subharmonic acoustic emission detection with implementation on a confocal dual-frequency piezoelectric ceramic structure to perform real-time monitoring of FUS-BBB opening. A confocal dual-frequency (0.55 MHz/1.1 MHz) focused ultrasound transducer was designed. The 1.1 MHz spherically-curved ceramic was employed to deliver FUS exposure to induce BBB-opening, whereas the outer-ring 0.55 MHz ceramic was employed to detect the subharmonic acoustic emissions originating from the target position. In stage-1 experiments, we employed spectral analysis and performed an energy spectrum density (ESD) calculation. An optimized 0.55 MHz ESD level change was shown to effectively discriminate the occurrence of BBB-opening. Wideband acoustic emissions received from 0.55 MHz ceramics were also analyzed to evaluate its correlations with erythrocyte extravasations. In stage-2 real-time monitoring experiments, we applied the predetermined ESD change as a detection threshold in PC-controlled algorithm to predict the FUS exposure intra-operatively. In stage-1 experiment, we showed that subharmonic ESD presents distinguishable dynamics between intact BBB and opened BBB, and therefore a threshold ESD change level (5.5 dB) can be identified for BBB-opening prediction. Using this ESD change threshold detection as a surrogate to on/off control the FUS exposure in stage-2 experiments, we demonstrated both excellent sensitivity (92%) and specificity (92.3%) in discriminating BBB-opening occurrence can be obtained in animal treatments, while concurrently achieving a high positive predicted value (95.8%). Wideband ESD was also highly correlated with the occurrence and level of erythrocyte extravasations (r (2) = 0.81). The proposed system configuration and corresponding analysis based on subharmonic acoustic emissions has the potential to be implemented as a real-time feedback control structure for reliable indication of intact FUS-BBB opening for CNS brain drug delivery.
Collapse
Affiliation(s)
- Chih-Hung Tsai
- Department of Electrical Engineering, Chang Gung University, Taoyuan, Taiwan
| | | | | | | |
Collapse
|
141
|
da Ros M, Iorio AL, Lucchesi M, Stival A, de Martino M, Sardi I. The Use of Anthracyclines for Therapy of CNS Tumors. Anticancer Agents Med Chem 2016; 15:721-7. [PMID: 25846760 PMCID: PMC4997942 DOI: 10.2174/1871520615666150407155319] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2014] [Revised: 03/26/2015] [Accepted: 04/05/2015] [Indexed: 02/07/2023]
Abstract
Despite being long lived, anthracyclines remain the “evergreen” drugs in clinical practice of oncology, showing a potent effect in inhibiting cell growth in many types of tumors, including brain neoplasms. Unfortunately, they suffer from a poor penetration into the brain when intravenously administered due to multidrug resistance mechanism, which hampers their delivery across the blood brain barrier. In this paper, we summarize the current literature on the role of anthracyclines in cancer therapy and highlight recent efforts on 1) development of tumor cell resistance to anthracyclines and 2) the new approaches to brain drug delivery across the blood brain barrier.
Collapse
Affiliation(s)
| | | | | | | | | | - Iacopo Sardi
- Neuro-Oncology Unit, Department of Paediatric Medicine, Meyer Children's Hospital. Viale G. Pieraccini 24, 50139 Florence, Italy.
| |
Collapse
|
142
|
Focused ultrasound to transiently disrupt the blood brain barrier. J Clin Neurosci 2016; 28:187-9. [PMID: 26883350 DOI: 10.1016/j.jocn.2015.12.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2015] [Accepted: 12/17/2015] [Indexed: 10/22/2022]
|
143
|
Burgess A, Shah K, Hough O, Hynynen K. Focused ultrasound-mediated drug delivery through the blood-brain barrier. Expert Rev Neurother 2016; 15:477-91. [PMID: 25936845 DOI: 10.1586/14737175.2015.1028369] [Citation(s) in RCA: 149] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Despite recent advances in blood-brain barrier (BBB) research, it remains a significant hurdle for the pharmaceutical treatment of brain diseases. Focused ultrasound (FUS) is one method to transiently increase permeability of the BBB to promote drug delivery to specific brain regions. An introduction to the BBB and a brief overview of the methods, which can be used to circumvent the BBB to promote drug delivery, is provided. In particular, we discuss the advantages and limitations of FUS technology and the efficacy of FUS-mediated drug delivery in models of disease. MRI for targeting and evaluating FUS treatments, combined with administration of microbubbles, allows for transient, reproducible BBB opening. The integration of a real-time acoustic feedback controller has improved treatment safety. Successful clinical translation of FUS has the potential to transform the treatment of brain disease worldwide without requiring the development of new pharmaceutical agents.
Collapse
Affiliation(s)
- Alison Burgess
- Physical Sciences, Sunnybrook Research Institute, 2075 Bayview Ave, S665, Toronto, ON M4N 3M5, Canada
| | | | | | | |
Collapse
|
144
|
Lang C, Zhang X, Dong Z, Luo Q, Qiao S, Huang Z, Fan X, Xu J, Liu J. Selenium-containing organic nanoparticles as silent precursors for ultra-sensitive thiol-responsive transmembrane anion transport. NANOSCALE 2016; 8:2960-2966. [PMID: 26783054 DOI: 10.1039/c5nr07808c] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
An anion transporter with a selenoxide group was able to form nanoparticles in water, whose activity was fully turned off due to the aggregation effect. The formed nanoparticles have a uniform size and can be readily dispersed in water at high concentrations. Turn-on of the nanoparticles by reducing molecules is proposed to be a combined process, including the reduction of selenoxide to selenide, disassembly of the nanoparticles and location of the transporter to the lipid membrane. Accordingly, a special acceleration phase can be observed in the turn-on kinetic curves. Since turn-on of the nanoparticles is quantitatively related to the amount of reductant, the nanoparticles can be activated in a step-by-step manner. Due to the sensibility of this system to thiols, cysteine can be detected at low nanomolar concentrations. This ultra-sensitive thiol-responsive transmembrane anion transport system is quite promising in biological applications.
Collapse
Affiliation(s)
- Chao Lang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Xin Zhang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Zeyuan Dong
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Quan Luo
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Shanpeng Qiao
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Zupeng Huang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Xiaotong Fan
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Jiayun Xu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| | - Junqiu Liu
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, 2699 Qianjin Street, Changchun 130012, P. R. China.
| |
Collapse
|
145
|
Hersh DS, Wadajkar AS, Roberts NB, Perez JG, Connolly NP, Frenkel V, Winkles JA, Woodworth GF, Kim AJ. Evolving Drug Delivery Strategies to Overcome the Blood Brain Barrier. Curr Pharm Des 2016; 22:1177-1193. [PMID: 26685681 PMCID: PMC4900538 DOI: 10.2174/1381612822666151221150733] [Citation(s) in RCA: 192] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 12/18/2015] [Indexed: 01/10/2023]
Abstract
The blood-brain barrier (BBB) poses a unique challenge for drug delivery to the central nervous system (CNS). The BBB consists of a continuous layer of specialized endothelial cells linked together by tight junctions, pericytes, nonfenestrated basal lamina, and astrocytic foot processes. This complex barrier controls and limits the systemic delivery of therapeutics to the CNS. Several innovative strategies have been explored to enhance the transport of therapeutics across the BBB, each with individual advantages and disadvantages. Ongoing advances in delivery approaches that overcome the BBB are enabling more effective therapies for CNS diseases. In this review, we discuss: (1) the physiological properties of the BBB, (2) conventional strategies to enhance paracellular and transcellular transport through the BBB, (3) emerging concepts to overcome the BBB, and (4) alternative CNS drug delivery strategies that bypass the BBB entirely. Based on these exciting advances, we anticipate that in the near future, drug delivery research efforts will lead to more effective therapeutic interventions for diseases of the CNS.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Graeme F. Woodworth
- Address correspondence to these authors at the Department of Neurosurgery, University of Maryland School of Medicine, 22 South Greene Street, Baltimore, MD 21201; E-mail: , Departments of Neurosurgery and Pharmaceutical Sciences, University of Maryland, Baltimore, 655 W. Baltimore Street, Baltimore, MD 21201;, E-mail:
| | - Anthony J. Kim
- Address correspondence to these authors at the Department of Neurosurgery, University of Maryland School of Medicine, 22 South Greene Street, Baltimore, MD 21201; E-mail: , Departments of Neurosurgery and Pharmaceutical Sciences, University of Maryland, Baltimore, 655 W. Baltimore Street, Baltimore, MD 21201;, E-mail:
| |
Collapse
|
146
|
Tagami T, Taki M, Ozeki T. Nanoparticulate Drug Delivery Systems to Overcome the Blood–Brain Barrier. METHODS IN PHARMACOLOGY AND TOXICOLOGY 2016. [DOI: 10.1007/978-1-4939-3121-7_17] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
|
147
|
Hudson R. Coupling the magnetic and heat dissipative properties of Fe3O4 particles to enable applications in catalysis, drug delivery, tissue destruction and remote biological interfacing. RSC Adv 2016. [DOI: 10.1039/c5ra22260e] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
As interest in nanomaterials continues to grow, and the scope of their applications widens, one subset of materials has set itself apart: magnetic nanoparticles (MNPs).
Collapse
Affiliation(s)
- R. Hudson
- Department of Chemistry
- Colby College
- Waterville
- USA
| |
Collapse
|
148
|
Hybrid Nanomaterials Based on Iron Oxide Nanoparticles and Mesoporous Silica Nanoparticles: Overcoming Challenges in Current Cancer Treatments. J CHEM-NY 2016. [DOI: 10.1155/2016/2672740] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The current approaches used for the treatment of cancer face some clinical limitations such as induction of severe side effects, multidrug resistance (MDR), and low specificity toward metastatic cancer cells. Hybrid nanomaterials hold a great potential to overcome all these challenges. Among hybrid nanoparticles, those based on mesoporous silica and iron oxide nanoparticles (MSNs and IONPs) have gained a privileged place in the biomedical field because of their outstanding properties. There are many studies demonstrating their effectiveness as drug delivery systems, nanoheaters, and imaging contrast agents. This review summarizes the advances related to the utilization of IONPs and MSNs for reducing side effects, overcoming MDR, and inhibiting metastasis. Furthermore, we give a future perspective of the clinical application of these technologies.
Collapse
|
149
|
Peng HH, Wu CH, Kang ST, Zhang JW, Liu HL, Chen WS, Wang CH, Yeh CK. Real-time monitoring of inertial cavitation effects of microbubbles by using MRI: In vitro experiments. Magn Reson Med 2015; 77:102-111. [PMID: 26714923 DOI: 10.1002/mrm.26082] [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] [Received: 08/26/2015] [Revised: 11/16/2015] [Accepted: 11/19/2015] [Indexed: 12/23/2022]
Abstract
PURPOSE To investigate the feasibility of half-Fourier acquisition single-shot turbo spin-echo (HASTE) for real-time monitoring of signal changes because of water flow induced by inertial cavitation (IC) during microbubbles (MBs)-present focused ultrasound (FUS) exposure. THEORY AND METHODS Strong turbulence produced in MB solution at the onset of IC results in the difficulty to refocus signal echoes and thus the decrease in signal intensity (SI). Fundamental investigations were conducted using an agar phantom containing MB dilutions exposed to 1.85-MHz FUS. The effects of various experimental conditions including MB concentrations, imaging slice thicknesses, chamber diameters, acoustic pressures, duty cycles, and pulse repetition frequencies (PRFs) were discussed. RESULTS Continuous 2.8 MPa FUS exposure resulted in SI changed from 11% to 55% when MBs concentrations increased from 0.025% to 0.1%. When slice thickness increased from 3 mm to 6 or 8 mm, smaller SI changes were observed (84%, 59%, and 46%). Images acquired with chamber diameter of 6 and 3 mm showed SI changes of 84% and 35%, respectively. In burst modes, higher duty cycles exhibited higher SI changes, and lower PRFs exhibited smaller and longer SI decrease. CONCLUSION Under various conditions, substantial signal changes were observable, suggesting the feasibility of applying HASTE to real-time monitor IC effect under FUS exposure. Magn Reson Med 77:102-111, 2017. © 2015 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hsu-Hsia Peng
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chen-Hua Wu
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Shih-Tsung Kang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Jia-Wei Zhang
- Department of Electrical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Hao-Li Liu
- Department of Electrical Engineering, Chang Gung University, Taoyuan, Taiwan
| | - Wen-Shiang Chen
- Department of Physical Medicine and Rehabilitation, National Taiwan University Hospital and National Taiwan University College of Medicine, Taipei, Taiwan.,Division of Medical Engineering Research, National Health Research Institutes, Miaoli, Taiwan
| | - Chung-Hsin Wang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| | - Chih-Kuang Yeh
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan
| |
Collapse
|
150
|
Haritonova A, Liu D, Ebbini ES. In Vivo application and localization of transcranial focused ultrasound using dual-mode ultrasound arrays. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2015; 62:2031-42. [PMID: 26670845 PMCID: PMC4683405 DOI: 10.1109/tuffc.2014.006882] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
Focused ultrasound (FUS) has been proposed for a variety of transcranial applications, including neuromodulation, tumor ablation, and blood-brain barrier opening. A flurry of activity in recent years has generated encouraging results demonstrating its feasibility in these and other applications. To date, monitoring of FUS beams has been primarily accomplished using MR guidance, where both MR thermography and elastography have been used. The recent introduction of real-time dual-mode ultrasound array (DMUA) systems offers a new paradigm in transcranial focusing. In this paper, we present first experimental results of ultrasound-guided transcranial FUS (tFUS) application in a rodent brain, both ex vivo and in vivo. DMUA imaging is used for visualization of the treatment region for placement of the focal spot within the brain. This includes the detection and localization of pulsating blood vessels at or near the target point(s). In addition, DMUA imaging is used to monitor and localize the FUS-tissue interactions in real time. In particular, a concave (40 mm radius of curvature), 32-element, 3.5-MHz DMUA prototype was used for imaging and tFUS application in ex vivo and in vivo rat models. The ex vivo experiments were used to evaluate the point spread function of the transcranial DMUA imaging at various points within the brain. In addition, DMUA-based transcranial ultrasound thermography measurements were compared with thermocouple measurements of subtherapeutic tFUS heating in rat brain ex vivo. The ex vivo setting was also used to demonstrate the capability of DMUA to produce localized thermal lesions. The in vivo experiments were designed to demonstrate the ability of the DMUA to apply, monitor, and localize subtherapeutic tFUS patterns that could be beneficial in transient blood-brain barrier opening. The results show that although the DMUA focus is degraded due to the propagation through the skull, it still produces localized heating effects within a sub-millimeter volume. In addition, DMUA transcranial echo data from brain tissue allow for reliable estimation of temperature change.
Collapse
Affiliation(s)
- Alyona Haritonova
- Department of Biomedical Engineering, University of Minnesota Twin Cities
| | - Dalong Liu
- Department of Electrical and Computer Engineering, University of Minnesota Twin Cities
| | - Emad S. Ebbini
- Department of Electrical and Computer Engineering, University of Minnesota Twin Cities
| |
Collapse
|