1
|
Aminyavari S, Afshari AR, Ahmadi SS, Kesharwani P, Sanati M, Sahebkar A. Unveiling the theranostic potential of SPIONs in Alzheimer's disease management. J Psychiatr Res 2024; 179:244-256. [PMID: 39321523 DOI: 10.1016/j.jpsychires.2024.09.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/22/2024] [Accepted: 09/19/2024] [Indexed: 09/27/2024]
Abstract
Alzheimer's disease (AD) is a devastating kind of dementia that is becoming more common worldwide. Toxic amyloid-beta (Aβ) aggregates are the primary cause of AD onset and development. Superparamagnetic iron oxide nanoparticles (SPIONs) have received a lot of interest in AD therapy over the last decade because of their ability to redirect the Aβ fibrillation process and improve associated brain dysfunction. The potential diagnostic application of SPIONs in AD has dramatically increased this interest. Furthermore, surface-modified engineered SPIONs function as drug carriers to improve the efficacy of current therapies. Various preclinical and clinical studies on the role of SPIONs in AD pathology have produced encouraging results. However, due to their physicochemical properties (e.g., size, surface charge, and particle concentration) in the biological milieu, SPIONs may play the role of a preventive or accelerative agent in AD. Even though SPIONs are potential therapeutic and diagnostic options in AD, significant efforts are still needed to overcome the inconsistencies and safety concerns. This review evaluated the current understanding of how various SPIONs interact with AD models and explored the discrepancies in their efficacy and safety.
Collapse
Affiliation(s)
- Samaneh Aminyavari
- Department of Neuroscience and Addiction Studies, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir R Afshari
- Department of Basic Sciences, Faculty of Medicine, Mashhad Medical Sciences, Islamic Azad University, Mashhad, Iran
| | - Seyed Sajad Ahmadi
- Imam Reza Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India.
| | - Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran; Experimental and Animal Study Center, Birjand University of Medical Sciences, Birjand, Iran.
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
2
|
Balaji PG, Bhimrao LS, Yadav AK. Revolutionizing Stroke Care: Nanotechnology-Based Brain Delivery as a Novel Paradigm for Treatment and Diagnosis. Mol Neurobiol 2024:10.1007/s12035-024-04215-3. [PMID: 38829514 DOI: 10.1007/s12035-024-04215-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/03/2024] [Indexed: 06/05/2024]
Abstract
Stroke, a severe medical condition arising from abnormalities in the coagulation-fibrinolysis cycle and metabolic processes, results in brain cell impairment and injury due to blood flow obstruction within the brain. Prompt and efficient therapeutic approaches are imperative to control and preserve brain functions. Conventional stroke medications, including fibrinolytic agents, play a crucial role in facilitating reperfusion to the ischemic brain. However, their clinical efficacy is hampered by short plasma half-lives, limited brain tissue distribution attributed to the blood-brain barrier (BBB), and lack of targeted drug delivery to the ischemic region. To address these challenges, diverse nanomedicine strategies, such as vesicular systems, polymeric nanoparticles, dendrimers, exosomes, inorganic nanoparticles, and biomimetic nanoparticles, have emerged. These platforms enhance drug pharmacokinetics by facilitating targeted drug accumulation at the ischemic site. By leveraging nanocarriers, engineered drug delivery systems hold the potential to overcome challenges associated with conventional stroke medications. This comprehensive review explores the pathophysiological mechanism underlying stroke and BBB disruption in stroke. Additionally, this review investigates the utilization of nanocarriers for current therapeutic and diagnostic interventions in stroke management. By addressing these aspects, the review aims to provide insight into potential strategies for improving stroke treatment and diagnosis through a nanomedicine approach.
Collapse
Affiliation(s)
- Paul Gajanan Balaji
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Londhe Sachin Bhimrao
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India
| | - Awesh K Yadav
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Raebareli (An Institute of National Importance under Department of Pharmaceuticals, Ministry of Chemicals and Fertilizers, GOI), A Transit Campus at Bijnor-Sisendi Road, Near CRPF Base Camp, Sarojini Nagar, Lucknow, 226002, Uttar Pradesh, India.
| |
Collapse
|
3
|
Chiang MC, Yang YP, Nicol CJB, Wang CJ. Gold Nanoparticles in Neurological Diseases: A Review of Neuroprotection. Int J Mol Sci 2024; 25:2360. [PMID: 38397037 PMCID: PMC10888679 DOI: 10.3390/ijms25042360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 02/10/2024] [Accepted: 02/14/2024] [Indexed: 02/25/2024] Open
Abstract
This review explores the diverse applications of gold nanoparticles (AuNPs) in neurological diseases, with a specific focus on Alzheimer's disease (AD), Parkinson's disease (PD), and stroke. The introduction highlights the pivotal role of neuroinflammation in these disorders and introduces the unique properties of AuNPs. The review's core examines the mechanisms by which AuNPs exert neuroprotection and anti-neuro-inflammatory effects, elucidating various pathways through which they manifest these properties. The potential therapeutic applications of AuNPs in AD are discussed, shedding light on promising avenues for therapy. This review also explores the prospects of utilizing AuNPs in PD interventions, presenting a hopeful outlook for future treatments. Additionally, the review delves into the potential of AuNPs in providing neuroprotection after strokes, emphasizing their significance in mitigating cerebrovascular accidents' aftermath. Experimental findings from cellular and animal models are consolidated to provide a comprehensive overview of AuNPs' effectiveness, offering insights into their impact at both the cellular and in vivo levels. This review enhances our understanding of AuNPs' applications in neurological diseases and lays the groundwork for innovative therapeutic strategies in neurology.
Collapse
Affiliation(s)
- Ming-Chang Chiang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| | - Yu-Ping Yang
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, FL 33136, USA;
- Department of Biochemistry and Molecular Biology, Miller School of Medicine, University of Miami, Miami, FL 33136, USA
| | - Christopher J. B. Nicol
- Departments of Pathology & Molecular Medicine and Biomedical & Molecular Sciences, Cancer Biology and Genetics Division, Cancer Research Institute, Queen’s University, Kingston, ON K7L 3N6, Canada;
| | - Chieh-Ju Wang
- Department of Life Science, College of Science and Engineering, Fu Jen Catholic University, New Taipei City 242, Taiwan
| |
Collapse
|
4
|
Martinez PJ, Green AL, Borden MA. Targeting diffuse midline gliomas: The promise of focused ultrasound-mediated blood-brain barrier opening. J Control Release 2024; 365:412-421. [PMID: 38000663 PMCID: PMC10842695 DOI: 10.1016/j.jconrel.2023.11.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/13/2023] [Accepted: 11/18/2023] [Indexed: 11/26/2023]
Abstract
Diffuse midline gliomas (DMGs), including diffuse intrinsic pontine glioma, have among the highest mortality rates of all childhood cancers, despite recent advancements in cancer therapeutics. This is partly because, unlike some CNS tumors, the blood-brain barrier (BBB) of DMG tumor vessels remains intact. The BBB prevents the permeation of many molecular therapies into the brain parenchyma, where the cancer cells reside. Focused ultrasound (FUS) with microbubbles has recently emerged as an innovative and exciting technology that non-invasively permeabilizes the BBB in a small focal region with millimeter precision. In this review, current treatment methods and biological barriers to treating DMGs are discussed. State-of-the-art FUS-mediated BBB opening is then examined, with a focus on the effects of various ultrasound parameters and the treatment of DMGs.
Collapse
Affiliation(s)
- Payton J Martinez
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80303, United States; Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO 80303, United States.
| | - Adam L Green
- Morgan Adams Foundation Pediatric Brain Tumor Research Program, Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO 80045, United States
| | - Mark A Borden
- Biomedical Engineering Program, University of Colorado Boulder, Boulder, CO 80303, United States; Department of Mechanical Engineering, University of Colorado Boulder, Boulder, CO 80303, United States
| |
Collapse
|
5
|
Kulkarni M, Patel K, Patel A, Patel S, Desai J, Patel M, Shah U, Patel A, Solanki N. Nanomaterials as drug delivery agents for overcoming the blood-brain barrier: A comprehensive review. ADMET AND DMPK 2023; 12:63-105. [PMID: 38560713 PMCID: PMC10974816 DOI: 10.5599/admet.2043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/23/2023] [Indexed: 04/04/2024] Open
Abstract
Background and Purpose The blood-brain barrier (BBB), a critical interface of specialized endothelial cells, plays a pivotal role in regulating molecular and ion transport between the central nervous system (CNS) and systemic circulation. Experimental Approach This review aims to delve into the intricate architecture and functions of the BBB while addressing challenges associated with delivering therapeutics to the brain. Historical milestones and contemporary insights underscore the BBB's significance in protecting the CNS. Key Results Innovative approaches for enhanced drug transport include intranasal delivery exploiting olfactory and trigeminal pathways, as well as techniques like temporary BBB opening through chemicals, receptors, or focused ultrasound. These avenues hold the potential to reshape conventional drug delivery paradigms and address the limitations posed by the BBB's selectivity. Conclusion This review underscores the vital role of the BBB in maintaining CNS health and emphasizes the importance of effective drug delivery through this barrier. Nanoparticles emerge as promising candidates to overcome BBB limitations and potentially revolutionize the treatment of CNS disorders. As research progresses, the application of nanomaterials shows immense potential for advancing neurological therapeutics, albeit with careful consideration of safety aspects.
Collapse
Affiliation(s)
- Mangesh Kulkarni
- Department of Pharmaceutical Technology; L J Institute of Pharmacy; L J University; Opp. Kataria Motors; Sarkhej-Gandhinagar Highway-382210, India
| | - Krishi Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, India
| | - Ayush Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, India
| | - Swayamprakash Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, India
| | - Jagruti Desai
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, India
| | - Mehul Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, India
| | - Umang Shah
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, India
| | - Ashish Patel
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, India
| | - Nilay Solanki
- Department of Pharmaceutics, Ramanbhai Patel College of Pharmacy, Charotar University of Science and Technology (CHARUSAT), CHARUSAT Campus, Changa 388421, India
| |
Collapse
|
6
|
Gareev K, Tagaeva R, Bobkov D, Yudintceva N, Goncharova D, Combs SE, Ten A, Samochernych K, Shevtsov M. Passing of Nanocarriers across the Histohematic Barriers: Current Approaches for Tumor Theranostics. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1140. [PMID: 37049234 PMCID: PMC10096980 DOI: 10.3390/nano13071140] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 03/19/2023] [Accepted: 03/20/2023] [Indexed: 06/19/2023]
Abstract
Over the past several decades, nanocarriers have demonstrated diagnostic and therapeutic (i.e., theranostic) potencies in translational oncology, and some agents have been further translated into clinical trials. However, the practical application of nanoparticle-based medicine in living organisms is limited by physiological barriers (blood-tissue barriers), which significantly hampers the transport of nanoparticles from the blood into the tumor tissue. This review focuses on several approaches that facilitate the translocation of nanoparticles across blood-tissue barriers (BTBs) to efficiently accumulate in the tumor. To overcome the challenge of BTBs, several methods have been proposed, including the functionalization of particle surfaces with cell-penetrating peptides (e.g., TAT, SynB1, penetratin, R8, RGD, angiopep-2), which increases the passing of particles across tissue barriers. Another promising strategy could be based either on the application of various chemical agents (e.g., efflux pump inhibitors, disruptors of tight junctions, etc.) or physical methods (e.g., magnetic field, electroporation, photoacoustic cavitation, etc.), which have been shown to further increase the permeability of barriers.
Collapse
Affiliation(s)
- Kamil Gareev
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Department of Micro and Nanoelectronics, Saint Petersburg Electrotechnical University “LETI”, 197022 Saint Petersburg, Russia
| | - Ruslana Tagaeva
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Danila Bobkov
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Natalia Yudintceva
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Daria Goncharova
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Stephanie E. Combs
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
| | - Artem Ten
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| | - Konstantin Samochernych
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
| | - Maxim Shevtsov
- Institute of Cytology of the Russian Academy of Sciences (RAS), 194064 Saint Petersburg, Russia
- Personalized Medicine Centre, Almazov National Medical Research Centre, 2 Akkuratova Str., 197341 Saint Petersburg, Russia
- Department of Radiation Oncology, Technishe Universität München (TUM), Klinikum rechts der Isar, Ismaningerstr. 22, 81675 Munich, Germany
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 690922 Vladivostok, Russia
| |
Collapse
|
7
|
Georgiou C, Cai Z, Alsaden N, Cho H, Behboudi M, Winnik MA, Rutka JT, Reilly RM. Treatment of Orthotopic U251 Human Glioblastoma Multiforme Tumors in NRG Mice by Convection-Enhanced Delivery of Gold Nanoparticles Labeled with the β-Particle-Emitting Radionuclide, 177Lu. Mol Pharm 2023; 20:582-592. [PMID: 36516432 PMCID: PMC9812026 DOI: 10.1021/acs.molpharmaceut.2c00815] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
In this study, we investigated convection-enhanced delivery (CED) of 23 ± 3 nm gold nanoparticles (AuNPs) labeled with the β-particle-emitting radionuclide 177Lu (177Lu-AuNPs) for treatment of orthotopic U251-Luc human glioblastoma multiforme (GBM) tumors in NRG mice. The cytotoxicity in vitro of 177Lu-AuNPs (0.0-2.0 MBq, 4 × 1011 AuNPs) on U251-Luc cells was also studied by a clonogenic survival assay, and DNA double-strand breaks (DSBs) caused by β-particle emissions of 177Lu were measured by confocal immunofluorescence microscopy for γH2AX. NRG mice with U251-Luc tumors in the right cerebral hemisphere of the brain were treated by CED of 1.1 ± 0.2 MBq of 177Lu-AuNPs (4 × 1011 AuNPs). Control mice received unlabeled AuNPs or normal saline. Tumor retention of 177Lu-AuNPs was assessed by single-photon emission computed tomography/computed tomography (SPECT/CT) imaging and biodistribution studies. Radiation doses were estimated for the tumor, brain, and other organs. The effectiveness for treating GBM tumors was determined by bioluminescence imaging (BLI) and T2-weighted magnetic resonance imaging (MRI) and by Kaplan-Meier median survival. Normal tissue toxicity was assessed by monitoring body weight and hematology and blood biochemistry analyses at 14 d post-treatment. 177Lu-AuNPs (2.0 MBq, 4 × 1011 AuNPs) decreased the clonogenic survival of U251-Luc cells to 0.005 ± 0.002 and increased DNA DSBs by 14.3-fold compared to cells treated with unlabeled AuNPs or normal saline. A high proportion of 177Lu-AuNPs was retained in the U251-Luc tumor in NRG mice up to 21 d with minimal re-distribution to the brain or other organs. The radiation dose in the tumor was high (599 Gy). The dose in the normal right cerebral hemisphere of the brain excluding the tumor was 93-fold lower (6.4 Gy), and 2000-3000-fold lower doses were calculated for the contralateral left cerebral hemisphere (0.3 Gy) or cerebellum (0.2 Gy). The doses in peripheral organs were <0.1 Gy. BLI revealed almost complete tumor growth arrest in mice treated with 177Lu-AuNPs, while tumors grew rapidly in control mice. MRI at 28 d post-treatment and histological staining showed no visible tumor in mice treated with 177Lu-AuNPs but large GBM tumors in control mice. All control mice reached a humane endpoint requiring sacrifice within 39 d (normal saline) or 45 d post-treatment (unlabeled AuNPs), while 5/8 mice treated with 177Lu-AuNPs survived up to 150 d. No normal tissue toxicity was observed in mice treated with 177Lu-AuNPs. We conclude that CED of 177Lu-AuNPs was highly effective for treating U251-Luc human GBM tumors in the brain in NRG mice at amounts that were non-toxic to normal tissues. These 177Lu-AuNPs administered by CED hold promise for treating patients with GBM to prevent recurrence and improve long-term outcome.
Collapse
Affiliation(s)
- Constantine
J. Georgiou
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada
| | - Zhongli Cai
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada
| | - Noor Alsaden
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada
| | - Hyungjun Cho
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - Minou Behboudi
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada
| | - Mitchell A. Winnik
- Department
of Chemistry, University of Toronto, 80 St. George Street, Toronto, OntarioM5S 3H6, Canada
| | - James T. Rutka
- Division
of Neurosurgery, The Hospital for Sick Children, 555 University Avenue, Toronto, OntarioM5G 1X8, Canada,Division
of Neurosurgery, Department of Surgery, Temerty Faculty of Medicine, University of Toronto, 149 College Street, Toronto, OntarioM5T 1P5, Canada
| | - Raymond M. Reilly
- Department
of Pharmaceutical Sciences, Leslie Dan Faculty of Pharmacy, University of Toronto, 144 College Street, Toronto, OntarioM5S 3M2, Canada,Department
of Medical Imaging, Temerty Faculty of Medicine, University of Toronto, Toronto, OntarioM5S 1A8, Canada,Joint Department
of Medical Imaging and Princess Margaret Cancer Centre, University Health Network, Toronto, OntarioM5G 2C1, Canada,
| |
Collapse
|
8
|
Wang J, Cai F, Lin Q, Zhao D, Zheng H. Acoustic radiation force dependence on properties of elastic spherical shells in standing waves. ULTRASONICS 2023; 127:106836. [PMID: 36174313 DOI: 10.1016/j.ultras.2022.106836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 08/20/2022] [Accepted: 08/29/2022] [Indexed: 06/16/2023]
Abstract
In this study, we theoretically investigate the acoustic radiation force acting on elastic spherical shells in standing waves in the dimensionless frequency range of 0<ka<0.7 (k is the wave number in the host fluid and a is the outer radius of the spherical shells), we show that in the low-frequency range (0<ka<0.2), the acoustic radiation force on core-shell spheres can vary from positive to negative by decreasing the effective density and effective bulk modulus of the spherical shells, while the effective density and bulk modulus are dependent on the hollow ratio of the core-shell spheres, the density and bulk modulus ratios of the shell to the core. Thus, the core-shell spheres cannot only be trapped from the pressure node to the pressure antinode by increasing the hollow ratio or decreasing the shell's density or longitudinal or shear velocity, but are also unresponsive to ultrasound waves with an optimal hollow ratio, density, or acoustic velocity of the shell. We further show that in the relatively low-frequency range of0<ka<0.7, the acoustic radiation force on core-shell spheres can be significantly enhanced around resonant frequencies. The acoustic radiation force could be enhanced at lower frequencies by increasing the hollow ratio or decreasing the shell's density or shear wave velocity. Our results pave the way for optimising the design of core-shell particles for efficient ultrasound-mediated drug delivery.
Collapse
Affiliation(s)
- Jinping Wang
- Paul C Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Feiyan Cai
- Paul C Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China.
| | - Qin Lin
- Paul C Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China; School of Biomedical Engineering, Guangdong Medical University, Dongguan 523808, China
| | - Degang Zhao
- Department of Physics, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Hairong Zheng
- Paul C Lauterbur Research Center for Biomedical Imaging, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| |
Collapse
|
9
|
Dahis D, Azagury DM, Obeid F, Dion MZ, Cryer AM, Riquelme MA, Dosta P, Abraham AW, Gavish M, Artzi N, Shamay Y, Azhari H. Focused Ultrasound Enhances Brain Delivery of Sorafenib Nanoparticles. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Affiliation(s)
- Daniel Dahis
- Department of Biomedical Engineering Technion Institute of Technology Haifa 3200003 Israel
- Department of Medicine Engineering of Medicine Division Brigham and Women's Hospital Harvard Medical School Cambridge 02115 MA USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
| | - Dana Meron Azagury
- Department of Biomedical Engineering Technion Institute of Technology Haifa 3200003 Israel
| | - Fadi Obeid
- The Ruth and Bruce Rappaport Faculty of Medicine Technion Institute of Technology Haifa 31096 Israel
| | - Michelle Z. Dion
- Department of Medicine Engineering of Medicine Division Brigham and Women's Hospital Harvard Medical School Cambridge 02115 MA USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
- Institute for Medical Engineering & Science MIT Cambridge 02139 MA USA
| | - Alexander M. Cryer
- Department of Medicine Engineering of Medicine Division Brigham and Women's Hospital Harvard Medical School Cambridge 02115 MA USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
- Institute for Medical Engineering & Science MIT Cambridge 02139 MA USA
| | - Mariana Alonso Riquelme
- Department of Medicine Engineering of Medicine Division Brigham and Women's Hospital Harvard Medical School Cambridge 02115 MA USA
| | - Pere Dosta
- Department of Medicine Engineering of Medicine Division Brigham and Women's Hospital Harvard Medical School Cambridge 02115 MA USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
- Institute for Medical Engineering & Science MIT Cambridge 02139 MA USA
| | - Ariel William Abraham
- Department of Medicine Engineering of Medicine Division Brigham and Women's Hospital Harvard Medical School Cambridge 02115 MA USA
| | - Moshe Gavish
- The Ruth and Bruce Rappaport Faculty of Medicine Technion Institute of Technology Haifa 31096 Israel
| | - Natalie Artzi
- Department of Medicine Engineering of Medicine Division Brigham and Women's Hospital Harvard Medical School Cambridge 02115 MA USA
- Wyss Institute for Biologically Inspired Engineering Harvard University Boston MA 02115 USA
- Broad Institute of Harvard and MIT Cambridge MA USA
| | - Yosi Shamay
- Department of Biomedical Engineering Technion Institute of Technology Haifa 3200003 Israel
| | - Haim Azhari
- Department of Biomedical Engineering Technion Institute of Technology Haifa 3200003 Israel
| |
Collapse
|
10
|
Antoniou A, Giannakou M, Georgiou E, Kleopa KA, Damianou C. Robotic device for transcranial focussed ultrasound applications in small animal models. Int J Med Robot 2022; 18:e2447. [PMID: 35924335 PMCID: PMC9786580 DOI: 10.1002/rcs.2447] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 07/22/2022] [Accepted: 07/23/2022] [Indexed: 12/30/2022]
Abstract
BACKGROUND Focussed Ultrasound (FUS) combined with microbubbles (MBs) was proven a promising modality for non-invasive blood brain barrier disruption (BBBD). Herein, two devices for FUS-mediated BBBD in rodents are presented. METHODS A two-axes robotic device was manufactured for navigating a single element FUS transducer of 1 MHz relative to the brain of rodents. A second more compact device featuring a single motorized vertical axis was also developed. Their performance was assessed in terms of motion accuracy, MRI compatibility and trans-skull BBBD in wild type mice using MBs in synergy with pulsed FUS. RESULTS Successful BBBD was evidenced by the Evans Blue dye method, as well as by Fibronectin and Fibrinogen immunostaining. BBB permeability was enhanced when the applied acoustic intensity was increased. CONCLUSIONS The proposed devices constitute a cost-effective and ergonomic solution for FUS-mediated BBBD in small animal models. Further experimentation is needed to examine the repeatability of results and optimise the therapeutic protocol.
Collapse
Affiliation(s)
- Anastasia Antoniou
- Department of Electrical Engineering, Computer Engineering, and InformaticsCyprus University of TechnologyLimassolCyprus
| | | | - Elena Georgiou
- Department of NeuroscienceThe Cyprus Institute of Neurology and GeneticsNicosiaCyprus
| | - Kleopas A. Kleopa
- Department of NeuroscienceThe Cyprus Institute of Neurology and GeneticsNicosiaCyprus
| | - Christakis Damianou
- Department of Electrical Engineering, Computer Engineering, and InformaticsCyprus University of TechnologyLimassolCyprus
| |
Collapse
|
11
|
Aryal M, Azadian MM, Hart AR, Macedo N, Zhou Q, Rosenthal EL, Airan RD. Noninvasive ultrasonic induction of cerebrospinal fluid flow enhances intrathecal drug delivery. J Control Release 2022; 349:434-442. [PMID: 35798095 DOI: 10.1016/j.jconrel.2022.06.067] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 06/15/2022] [Accepted: 06/30/2022] [Indexed: 10/17/2022]
Abstract
Intrathecal drug delivery is routinely used in the treatment and prophylaxis of varied central nervous system conditions, as doing so allows drugs to directly bypass the blood-brain barrier. However, the utility of this route of administration is limited by poor brain and spinal cord parenchymal drug uptake from the cerebrospinal fluid. We demonstrate that a simple noninvasive transcranial ultrasound protocol can significantly increase influx of cerebrospinal fluid into the perivascular spaces of the brain, to enhance the uptake of intrathecally administered drugs. Specifically, we administered small (~1 kDa) and large (~155 kDa) molecule agents into the cisterna magna of rats and then applied low, diagnostic-intensity focused ultrasound in a scanning protocol throughout the brain. Using real-time magnetic resonance imaging and ex vivo histologic analyses, we observed significantly increased uptake of small molecule agents into the brain parenchyma, and of both small and large molecule agents into the perivascular space from the cerebrospinal fluid. Notably, there was no evidence of brain parenchymal damage following this intervention. The low intensity and noninvasive approach of transcranial ultrasound in this protocol underscores the ready path to clinical translation of this technique. In this manner, this protocol can be used to directly bypass the blood-brain barrier for whole-brain delivery of a variety of agents. Additionally, this technique can potentially be used as a means to probe the causal role of the glymphatic system in the variety of disease and physiologic processes to which it has been correlated.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Departments of Engineering and Radiation Oncology, Loyola University Chicago, Chicago, IL, United States
| | - Matine M Azadian
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Alex R Hart
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Nicholas Macedo
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States
| | - Quan Zhou
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, United States; Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA, United States
| | - Eben L Rosenthal
- Department of Otolaryngology, Stanford University School of Medicine, Stanford, CA, United States; Stanford Cancer Center, Stanford Medical Center, Stanford, CA, United States; Department of Otolaryngology, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Raag D Airan
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, United States; Department of Materials Science and Engineering, Stanford University School of Medicine, Stanford, CA, United States; Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA, United States.
| |
Collapse
|
12
|
Zhang S, Zhang S, Luo S, Tang P, Wan M, Wu D, Gao W. Ultrasound-assisted brain delivery of nanomedicines for brain tumor therapy: advance and prospect. J Nanobiotechnology 2022; 20:287. [PMID: 35710426 PMCID: PMC9205090 DOI: 10.1186/s12951-022-01464-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 05/18/2022] [Indexed: 12/14/2022] Open
Abstract
Nowadays, brain tumors are challenging problems, and the key of therapy is ensuring therapeutic drugs cross the blood-brain barrier (BBB) effectively. Although the efficiency of drug transport across the BBB can be increased by innovating and modifying nanomedicines, they exert insufficient therapeutic effects on brain tumors due to the complex environment of the brain. It is worth noting that ultrasound combined with the cavitation effect of microbubbles can assist BBB opening and enhance brain delivery of nanomedicines. This ultrasound-assisted brain delivery (UABD) technology with related nanomedicines (UABD nanomedicines) can safely open the BBB, facilitate the entry of drugs into the brain, and enhance the therapeutic effect on brain tumors. UABD nanomedicines, as the main component of UABD technology, have great potential in clinical application and have been an important area of interest in the field of brain tumor therapy. However, research on UABD nanomedicines is still in its early stages despite the fact that they have been associated with many disciplines, including material science, brain science, ultrasound, biology, and medicine. Some aspects of UABD theory and technology remain unclear, especially the mechanisms of BBB opening, relationship between materials of nanomedicines and UABD technology, cavitation and UABD nanomedicines design theories. This review introduces the research status of UABD nanomedicines, investigates their properties and applications of brain tumor therapy, discusses the advantages and drawbacks of UABD nanomedicines for the treatment of brain tumors, and offers their prospects. We hope to encourage researchers from various fields to participate in this area and collaborate on developing UABD nanomedicines into powerful tools for brain tumor therapy.
Collapse
Affiliation(s)
- Shuo Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Shuai Zhang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Siyuan Luo
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Peng Tang
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Mingxi Wan
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China
| | - Daocheng Wu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, People's Republic of China.
| | - Wei Gao
- Department of Anesthesiology and Center for Brain Science and Center for Translational Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, People's Republic of China.
| |
Collapse
|
13
|
Hersh AM, Alomari S, Tyler BM. Crossing the Blood-Brain Barrier: Advances in Nanoparticle Technology for Drug Delivery in Neuro-Oncology. Int J Mol Sci 2022; 23:4153. [PMID: 35456971 PMCID: PMC9032478 DOI: 10.3390/ijms23084153] [Citation(s) in RCA: 95] [Impact Index Per Article: 47.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 04/01/2022] [Accepted: 04/07/2022] [Indexed: 12/10/2022] Open
Abstract
The blood-brain barrier (BBB) constitutes a microvascular network responsible for excluding most drugs from the brain. Treatment of brain tumors is limited by the impermeability of the BBB and, consequently, survival outcomes for malignant brain tumors remain poor. Nanoparticles (NPs) represent a potential solution to improve drug transport to brain tumors, given their small size and capacity to target tumor cells. Here, we review the unique physical and chemical properties of NPs that aid in BBB transport and discuss mechanisms of NP transport across the BBB, including paracellular transport, carrier-mediated transport, and adsorptive- and receptor-mediated transcytosis. The major types of NPs investigated for treatment of brain tumors are detailed, including polymeric NPs, liposomes, solid lipid NPs, dendrimers, metals, quantum dots, and nanogels. In addition to their role in drug delivery, NPs can be used as imaging contrast agents and can be conjugated with imaging probes to assist in visualizing tumors, demarcating lesion boundaries and margins, and monitoring drug delivery and treatment response. Multifunctional NPs can be designed that are capable of targeting tumors for both imaging and therapeutic purposes. Finally, limitations of NPs for brain tumor treatment are discussed.
Collapse
Affiliation(s)
| | | | - Betty M. Tyler
- Department of Neurosurgery, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; (A.M.H.); (S.A.)
| |
Collapse
|
14
|
Grudzenski S, Heger S, de Jonge A, Schipp J, Dumont E, Larrat B, Schad L, Platten M, Fatar M. Simulation, Implementation and Measurement of Defined Sound Fields for Blood-Brain Barrier Opening in Rats. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:422-436. [PMID: 34863589 DOI: 10.1016/j.ultrasmedbio.2021.10.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 09/28/2021] [Accepted: 10/04/2021] [Indexed: 06/13/2023]
Abstract
The blood-brain barrier (BBB) is the most important obstacle to delivery of therapeutics to the central nervous system. Low-intensity pulsed focused ultrasound (FUS) in combination with microbubbles applied under magnetic resonance imaging (MRI) control provides a non-invasive and safe technique for BBB opening (BBBo). In rodent models, however, settings and application protocols differ significantly. Depending on the strain and size, important variables include ultrasound attenuation and sound field distortion caused by the skull. We examined the ultrasound attenuation of the skull of Wistar rats using a targeted FUS system. By modifying the transducer elements and by varying and simulating the acoustic field of the FUS system, we measured a skull attenuation of about 60%. To evaluate potential application of the targeted FUS system in genetically modified animals with increased sensitivity to brain hemorrhage caused by vascular dysfunction, we assessed safety in healthy animals. Histological and MRI analyses of the central nervous system revealed an increase in the number and severity of hyperacute bleeds with focal pressure. At a pressure of 0.4 MPa, no bleeds were induced, albeit at the cost of a weaker hyperintense MRI signal post BBBo. These results indicate a relationship between pressure and the dimension of permeabilization.
Collapse
Affiliation(s)
- Saskia Grudzenski
- Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany.
| | - Stefan Heger
- Institute for Biomedical Engineering, Mannheim University, Mannheim, Germany
| | - Andreas de Jonge
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Julia Schipp
- Institute for Biomedical Engineering, Mannheim University, Mannheim, Germany
| | | | - Benoit Larrat
- Université Paris-Saclay, CEA, CNRS, Baobab, NeuroSpin, Gif-sur-Yvette, France
| | - Lothar Schad
- Computer Assisted Clinical Medicine, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Michael Platten
- Mannheimer Center of Translational Neuroscience (MCTN), Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Marc Fatar
- European Center of Angioscience (ECAS), Department of Neurology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| |
Collapse
|
15
|
Lee MJ, Lee I, Wang K. Recent Advances in RNA Therapy and Its Carriers to Treat the Single-Gene Neurological Disorders. Biomedicines 2022; 10:biomedicines10010158. [PMID: 35052837 PMCID: PMC8773368 DOI: 10.3390/biomedicines10010158] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 01/07/2022] [Accepted: 01/11/2022] [Indexed: 02/07/2023] Open
Abstract
The development of new sequencing technologies in the post-genomic era has accelerated the identification of causative mutations of several single gene disorders. Advances in cell and animal models provide insights into the underlining pathogenesis, which facilitates the development and maturation of new treatment strategies. The progress in biochemistry and molecular biology has established a new class of therapeutics—the short RNAs and expressible long RNAs. The sequences of therapeutic RNAs can be optimized to enhance their stability and translatability with reduced immunogenicity. The chemically-modified RNAs can also increase their stability during intracellular trafficking. In addition, the development of safe and high efficiency carriers that preserves the integrity of therapeutic RNA molecules also accelerates the transition of RNA therapeutics into the clinic. For example, for diseases that are caused by genetic defects in a specific protein, an effective approach termed “protein replacement therapy” can provide treatment through the delivery of modified translatable mRNAs. Short interference RNAs can also be used to treat diseases caused by gain of function mutations or restore the splicing aberration defects. Here we review the applications of newly developed RNA-based therapeutics and its delivery and discuss the clinical evidence supporting the potential of RNA-based therapy in single-gene neurological disorders.
Collapse
Affiliation(s)
- Ming-Jen Lee
- Department of Neurology, National Taiwan University Hospital, Taipei 10012, Taiwan;
- Department of Medical Genetics, National Taiwan University Hospital, Taipei 10012, Taiwan
| | - Inyoul Lee
- Institute for Systems Biology, Seattle, WA 98109, USA;
| | - Kai Wang
- Institute for Systems Biology, Seattle, WA 98109, USA;
- Correspondence: ; Tel.: +1-206-732-1336
| |
Collapse
|
16
|
Snipstad S, Vikedal K, Maardalen M, Kurbatskaya A, Sulheim E, Davies CDL. Ultrasound and microbubbles to beat barriers in tumors: Improving delivery of nanomedicine. Adv Drug Deliv Rev 2021; 177:113847. [PMID: 34182018 DOI: 10.1016/j.addr.2021.113847] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 06/18/2021] [Accepted: 06/22/2021] [Indexed: 12/18/2022]
Abstract
Successful delivery of drugs and nanomedicine to tumors requires a functional vascular network, extravasation across the capillary wall, penetration through the extracellular matrix, and cellular uptake. Nanomedicine has many merits, but penetration deep into the tumor interstitium remains a challenge. Failure of cancer treatment can be caused by insufficient delivery of the therapeutic agents. After intravenous administration, nanomedicines are often found in off-target organs and the tumor extracellular matrix close to the capillary wall. With circulating microbubbles, ultrasound exposure focused toward the tumor shows great promise in improving the delivery of therapeutic agents. In this review, we address the impact of focused ultrasound and microbubbles to overcome barriers for drug delivery such as perfusion, extravasation, and transport through the extracellular matrix. Furthermore, we discuss the induction of an immune response with ultrasound and delivery of immunotherapeutics. The review discusses mainly preclinical results and ends with a summary of ongoing clinical trials.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway.
| | - Krister Vikedal
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Matilde Maardalen
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Anna Kurbatskaya
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | | |
Collapse
|
17
|
Emerging Therapeutic Strategies for Brain Tumors. Neuromolecular Med 2021; 24:23-34. [PMID: 34406634 DOI: 10.1007/s12017-021-08681-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 07/10/2021] [Indexed: 12/14/2022]
Abstract
Nearly thirty thousand incidences of primary and 300 thousand incidences of metastatic brain cancer are diagnosed in the USA each year. It has a high mortality rate and is often unresponsive to the standard of care, which includes surgical resection, radiation, and chemotherapy. These treatment strategies are also hindered by their invasiveness and toxic effects on healthy cells and tissues. Furthermore, the blood-brain/tumor barrier severely limits delivery of anti-cancer therapeutics administered intravenously to brain tumors, resulting in poor tumor response to the treatment. There is a critical need to develop new approaches to brain cancer therapy that can overcome these limitations. Focused ultrasound has emerged as a modality that addresses many of these limitations and has the potential to alter the treatment paradigm for brain cancer. Ultrasound transmitted through the skull can be focused on tumors and used for targeted ablation or opening the vascular barriers for drug delivery. This review provides insight on the current status of these unique ultrasound techniques, different strategies of using this technique for brain cancer, experience in preclinical models, and potential for clinical translation. We also debate the safety perspective of these techniques and discuss potential avenues for future work in noninvasive planning, monitoring, and evaluation of the ultrasonic neurointervention.
Collapse
|
18
|
Satapathy MK, Yen TL, Jan JS, Tang RD, Wang JY, Taliyan R, Yang CH. Solid Lipid Nanoparticles (SLNs): An Advanced Drug Delivery System Targeting Brain through BBB. Pharmaceutics 2021; 13:1183. [PMID: 34452143 PMCID: PMC8402065 DOI: 10.3390/pharmaceutics13081183] [Citation(s) in RCA: 75] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 12/12/2022] Open
Abstract
The blood-brain barrier (BBB) plays a vital role in the protection and maintenance of homeostasis in the brain. In this way, it is an interesting target as an interface for various types of drug delivery, specifically in the context of the treatment of several neuropathological conditions where the therapeutic agents cannot cross the BBB. Drug toxicity and on-target specificity are among some of the limitations associated with current neurotherapeutics. In recent years, advances in nanodrug delivery have enabled the carrier system containing the active therapeutic drug to target the signaling pathways and pathophysiology that are closely linked to central nervous system (CNS) disorders such as Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), multiple sclerosis (MS), brain tumor, epilepsy, ischemic stroke, and neurodegeneration. At present, among the nano formulations, solid lipid nanoparticles (SLNs) have emerged as a putative drug carrier system that can deliver the active therapeutics (drug-loaded SLNs) across the BBB at the target site of the brain, offering a novel approach with controlled drug delivery, longer circulation time, target specificity, and higher efficacy, and more importantly, reducing toxicity in a biomimetic way. This paper highlights the synthesis and application of SLNs as a novel nontoxic formulation strategy to carry CNS drugs across the BBB to improve the use of therapeutics agents in treating major neurological disorders in future clinics.
Collapse
Affiliation(s)
- Mantosh Kumar Satapathy
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
| | - Ting-Lin Yen
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
- Department of Medical Research, Cathay General Hospital, Taipei 22174, Taiwan
| | - Jing-Shiun Jan
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
| | - Ruei-Dun Tang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan;
| | - Jia-Yi Wang
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan;
- Department of Neurosurgery, Taipei Medical University Hospital, Taipei 110, Taiwan
- Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
| | - Rajeev Taliyan
- Department of Pharmacy, Neuropsychopharmacology Division, Birla Institute of Technology and Science, Pilani 333031, India;
| | - Chih-Hao Yang
- Department of Pharmacology, School of Medicine, College of Medicine, Taipei Medical University, No. 250, Wu Hsing St., Taipei 110, Taiwan; (M.K.S.); (T.-L.Y.); (J.-S.J.); (R.-D.T.)
- Neuroscience Research Center, Taipei Medical University, Taipei 110, Taiwan
| |
Collapse
|
19
|
Kouhi A, Pachipulusu V, Kapenstein T, Hu P, Epstein AL, Khawli LA. Brain Disposition of Antibody-Based Therapeutics: Dogma, Approaches and Perspectives. Int J Mol Sci 2021; 22:ijms22126442. [PMID: 34208575 PMCID: PMC8235515 DOI: 10.3390/ijms22126442] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022] Open
Abstract
Due to their high specificity, monoclonal antibodies have been widely investigated for their application in drug delivery to the central nervous system (CNS) for the treatment of neurological diseases such as stroke, Alzheimer’s, and Parkinson’s disease. Research in the past few decades has revealed that one of the biggest challenges in the development of antibodies for drug delivery to the CNS is the presence of blood–brain barrier (BBB), which acts to restrict drug delivery and contributes to the limited uptake (0.1–0.2% of injected dose) of circulating antibodies into the brain. This article reviews the various methods currently used for antibody delivery to the CNS at the preclinical stage of development and the underlying mechanisms of BBB penetration. It also describes efforts to improve or modulate the physicochemical and biochemical properties of antibodies (e.g., charge, Fc receptor binding affinity, and target affinity), to adapt their pharmacokinetics (PK), and to influence their distribution and disposition into the brain. Finally, a distinction is made between approaches that seek to modify BBB permeability and those that use a physiological approach or antibody engineering to increase uptake in the CNS. Although there are currently inherent difficulties in developing safe and efficacious antibodies that will cross the BBB, the future prospects of brain-targeted delivery of antibody-based agents are believed to be excellent.
Collapse
|
20
|
Quantitative analysis of in-vivo microbubble distribution in the human brain. Sci Rep 2021; 11:11797. [PMID: 34083642 PMCID: PMC8175375 DOI: 10.1038/s41598-021-91252-w] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2020] [Accepted: 05/21/2021] [Indexed: 02/04/2023] Open
Abstract
Microbubbles (MB) are widely used as contrast agents to perform contrast-enhanced ultrasound (CEUS) imaging and as acoustic amplifiers of mechanical bioeffects incited by therapeutic-level ultrasound. The distribution of MBs in the brain is not yet fully understood, thereby limiting intra-operative CEUS guidance or MB-based FUS treatments. In this paper we describe a robust platform for quantification of MB distribution in the human brain, allowing to quantitatively discriminate between tumoral and normal brain tissues and we provide new information regarding real-time cerebral MBs distribution. Intraoperative CEUS imaging was performed during surgical tumor resection using an ultrasound machine (MyLab Twice, Esaote, Italy) equipped with a multifrequency (3-11 MHz) linear array probe (LA332) and a specific low mechanical index (MI < 0.4) CEUS algorithm (CnTi, Esaote, Italy; section thickness, 0.245 cm) for non-destructive continuous MBs imaging. CEUS acquisition is started by enabling the CnTI PEN-M algorithm automatically setting the MI at 0.4 with a center frequency of 2.94 MHz-10 Hz frame rate at 80 mm-allowing for continuous non-destructive MBs imaging. 19 ultrasound image sets of adequate length were selected and retrospectively analyzed using a custom image processing software for quantitative analysis of echo power. Regions of interest (ROIs) were drawn on key structures (artery-tumor-white matter) by a blinded neurosurgeon, following which peak enhancement and time intensity curves (TICs) were quantified. CEUS images revealed clear qualitative differences in MB distribution: arteries showed the earliest and highest enhancement among all structures, followed by tumor and white matter regions, respectively. The custom software built for quantitative analysis effectively captured these differences. Quantified peak intensities showed regions containing artery, tumor or white matter structures having an average MB intensity of 0.584, 0.436 and 0.175 units, respectively. Moreover, the normalized area under TICs revealed the time of flight for MB to be significantly lower in brain tissue as compared with tumor tissue. Significant heterogeneities in TICs were also observed within different regions of the same brain lesion. In this study, we provide the most comprehensive strategy for accurate quantitative analysis of MBs distribution in the human brain by means of CEUS intraoperative imaging. Furthermore our results demonstrate that CEUS imaging quantitative analysis enables discernment between different types of brain tumors as well as regions and structures within the brain. Similar considerations will be important for the planning and implementation of MB-based imaging or treatments in the future.
Collapse
|
21
|
Olsman M, Sereti V, Mühlenpfordt M, Johnsen KB, Andresen TL, Urquhart AJ, Davies CDL. Focused Ultrasound and Microbubble Treatment Increases Delivery of Transferrin Receptor-Targeting Liposomes to the Brain. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1343-1355. [PMID: 33608142 DOI: 10.1016/j.ultrasmedbio.2021.01.014] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 01/06/2021] [Accepted: 01/13/2021] [Indexed: 05/15/2023]
Abstract
The blood-brain barrier (BBB) is a major obstacle to treating several brain disorders. Focused ultrasound (FUS) in combination with intravascular microbubbles increases BBB permeability by opening tight junctions, creating endothelial cell openings, improving endocytosis and increasing transcytosis. Here we investigated whether combining FUS and microbubbles with transferrin receptor-targeting liposomes would result in enhanced delivery to the brain of post-natal rats compared with liposomes lacking the BBB-targeting moiety. For all animals, increased BBB permeability was observed after FUS treatment. A 40% increase in accumulation of transferrin receptor-targeting liposomes was observed in the FUS-treated hemisphere, whereas the isotype immunoglobulin G liposomes showed no increased accumulation. Confocal laser scanning microscopy of brain sections revealed that both types of liposomes were mainly observed in endothelial cells in the FUS-treated hemisphere. The results demonstrate that FUS and microbubble treatment combined with BBB-targeting liposomes could be a promising approach to enhance drug delivery to the brain.
Collapse
Affiliation(s)
- Marieke Olsman
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Viktoria Sereti
- Department of Health Technology, Technical University Denmark, Kongens Lyngby, Denmark
| | - Melina Mühlenpfordt
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kasper Bendix Johnsen
- Department of Health Technology, Technical University Denmark, Kongens Lyngby, Denmark
| | - Thomas Lars Andresen
- Department of Health Technology, Technical University Denmark, Kongens Lyngby, Denmark
| | - Andrew James Urquhart
- Department of Health Technology, Technical University Denmark, Kongens Lyngby, Denmark
| | | |
Collapse
|
22
|
Ung C, Tsoli M, Liu J, Cassano D, Pocoví-Martínez S, Upton DH, Ehteda A, Mansfeld FM, Failes TW, Farfalla A, Katsinas C, Kavallaris M, Arndt GM, Vittorio O, Cirillo G, Voliani V, Ziegler DS. Doxorubicin-Loaded Gold Nanoarchitectures as a Therapeutic Strategy against Diffuse Intrinsic Pontine Glioma. Cancers (Basel) 2021; 13:1278. [PMID: 33805713 PMCID: PMC7999568 DOI: 10.3390/cancers13061278] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2021] [Revised: 03/09/2021] [Accepted: 03/09/2021] [Indexed: 01/11/2023] Open
Abstract
Diffuse Intrinsic Pontine Gliomas (DIPGs) are highly aggressive paediatric brain tumours. Currently, irradiation is the only standard treatment, but is palliative in nature and most patients die within 12 months of diagnosis. Novel therapeutic approaches are urgently needed for the treatment of this devastating disease. We have developed non-persistent gold nano-architectures (NAs) functionalised with human serum albumin (HSA) for the delivery of doxorubicin. Doxorubicin has been previously reported to be cytotoxic in DIPG cells. In this study, we have preclinically evaluated the cytotoxic efficacy of doxorubicin delivered through gold nanoarchitectures (NAs-HSA-Dox). We found that DIPG neurospheres were equally sensitive to doxorubicin and doxorubicin-loaded NAs. Colony formation assays demonstrated greater potency of NAs-HSA-Dox on colony formation compared to doxorubicin. Western blot analysis indicated increased apoptotic markers cleaved Parp, cleaved caspase 3 and phosphorylated H2AX in NAs-HSA-Dox treated DIPG neurospheres. Live cell content and confocal imaging demonstrated significantly higher uptake of NAs-HSA-Dox into DIPG neurospheres compared to doxorubicin alone. Despite the potency of the NAs in vitro, treatment of an orthotopic model of DIPG showed no antitumour effect. This disparate outcome may be due to the integrity of the blood-brain barrier and highlights the need to develop therapies to enhance penetration of drugs into DIPG.
Collapse
Affiliation(s)
- Caitlin Ung
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
| | - Maria Tsoli
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Jie Liu
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
| | - Domenico Cassano
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy; (D.C.); (S.P.-M.); (V.V.)
| | - Salvador Pocoví-Martínez
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy; (D.C.); (S.P.-M.); (V.V.)
| | - Dannielle H. Upton
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
| | - Anahid Ehteda
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
| | - Friederike M. Mansfeld
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Monash University, Royal Parade, Parkville, VIC 3052, Australia
| | - Timothy W. Failes
- ACRF Drug Discovery Centre, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (T.W.F.); (G.M.A.)
| | - Annafranca Farfalla
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy; (A.F.); (G.C.)
| | - Christopher Katsinas
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
| | - Maria Kavallaris
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Greg M. Arndt
- ACRF Drug Discovery Centre, Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (T.W.F.); (G.M.A.)
| | - Orazio Vittorio
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- ARC Centre of Excellence in Convergent Bio-Nano Science and Technology, Australian Centre for NanoMedicine, University of New South Wales, Kensington, NSW 2052, Australia
| | - Giuseppe Cirillo
- Department of Pharmacy Health and Nutritional Science, University of Calabria, 87036 Rende, Italy; (A.F.); (G.C.)
| | - Valerio Voliani
- Center for Nanotechnology Innovation, Istituto Italiano di Tecnologia, Piazza San Silvestro 12, 56127 Pisa, Italy; (D.C.); (S.P.-M.); (V.V.)
| | - David S. Ziegler
- Children’s Cancer Institute, Lowy Cancer Research Centre, University of New South Wales, High Street, Randwick, NSW 2052, Australia; (C.U.); (J.L.); (D.H.U.); (A.E.); (F.M.M.); (C.K.); (M.K.); (O.V.)
- School of Women’s and Children’s Health, University of New South Wales, Kensington, NSW 2052, Australia
- Kids Cancer Centre, Sydney Children’s Hospital, Randwick, NSW 2052, Australia
| |
Collapse
|
23
|
Sarmah D, Banerjee M, Datta A, Kalia K, Dhar S, Yavagal DR, Bhattacharya P. Nanotechnology in the diagnosis and treatment of stroke. Drug Discov Today 2021; 26:585-592. [PMID: 33242696 DOI: 10.1016/j.drudis.2020.11.018] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 11/05/2020] [Accepted: 11/16/2020] [Indexed: 01/28/2023]
Abstract
Increasing developments in the field of nanotechnology have ignited its use in stroke diagnosis and treatment. The benefits of structural modification, ease of synthesis, and biocompatibility support the use of nanomaterials in the clinic. The pathophysiology of stroke is complex, involving different brain regions; hence, therapeutic agents are required to be delivered to specific regions. Nanoparticles (NPs) can be engineered to help improve the delivery and release of therapeutic agents in a localized manner, especially in the penumbra. This contributes not only to therapy, but also to neurosurgery and neuroimaging. Nanomaterials also offer high efficacy with few adverse effects. In this review, we provide a concise summary of the caveats associated with nanotechnology with respect to stroke therapy and diagnosis.
Collapse
Affiliation(s)
- Deepaneeta Sarmah
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Mainak Banerjee
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Aishika Datta
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Kiran Kalia
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India
| | - Shanta Dhar
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL, USA; Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Dileep R Yavagal
- Department of Neurology and Neurosurgery, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Pallab Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Ahmedabad, Gandhinagar, Gujarat 382355, India.
| |
Collapse
|
24
|
Hwang CH. Targeted Delivery of Erythropoietin Hybridized with Magnetic Nanocarriers for the Treatment of Central Nervous System Injury: A Literature Review. Int J Nanomedicine 2020; 15:9683-9701. [PMID: 33311979 PMCID: PMC7726550 DOI: 10.2147/ijn.s287456] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2020] [Accepted: 11/21/2020] [Indexed: 12/15/2022] Open
Abstract
Although the incidence of central nervous system injuries has continued to rise, no promising treatments have been elucidated. Erythropoietin plays an important role in neuroprotection and neuroregeneration as well as in erythropoiesis. Moreover, the current worldwide use of erythropoietin in the treatment of hematologic diseases allows for its ready application in patients with central nervous system injuries. However, erythropoietin has a very short therapeutic time window (within 6–8 hours) after injury, and it has both hematopoietic and nonhematopoietic receptors, which exhibit heterogenic and phylogenetic differences. These differences lead to limited amounts of erythropoietin binding to in situ erythropoietin receptors. The lack of high-quality evidence for clinical use and the promising results of in vitro/in vivo models necessitate fast targeted delivery agents such as nanocarriers. Among current nanocarriers, noncovalent polymer-entrapping or polymer-adsorbing erythropoietin obtained by nanospray drying may be the most promising. With the incorporation of magnetic nanocarriers into an erythropoietin polymer, spatiotemporal external magnetic navigation is another area of great interest for targeted delivery within the therapeutic time window. Intravenous administration is the most readily used route. Manufactured erythropoietin nanocarriers should be clearly characterized using bioengineering analyses of the in vivo size distribution and the quality of entrapment or adsorption. Further preclinical trials are required to increase the therapeutic bioavailability (in vivo biological identity alteration, passage through the lung capillaries or the blood brain barrier, and timely degradation followed by removal of the nanocarriers from the body) and decrease the adverse effects (hematological complications, neurotoxicity, and cytotoxicity), especially of the nanocarrier.
Collapse
Affiliation(s)
- Chang Ho Hwang
- Department of Physical and Rehabilitation Medicine, Chungnam National University Sejong Hospital, Chungnam National University College of Medicine, Sejong, Republic of Korea
| |
Collapse
|
25
|
Ohta S, Kikuchi E, Ishijima A, Azuma T, Sakuma I, Ito T. Investigating the optimum size of nanoparticles for their delivery into the brain assisted by focused ultrasound-induced blood-brain barrier opening. Sci Rep 2020; 10:18220. [PMID: 33106562 PMCID: PMC7588485 DOI: 10.1038/s41598-020-75253-9] [Citation(s) in RCA: 100] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2020] [Accepted: 09/30/2020] [Indexed: 02/07/2023] Open
Abstract
The blood–brain barrier (BBB) has hampered the efficiency of nanoparticle delivery into the brain via conventional strategies. The widening of BBB tight junctions via focused ultrasound (FUS) offers a promising approach for enhancing the delivery of nanoparticles into the brain. However, there is currently an insufficient understanding of how nanoparticles pass through the opened BBB gaps. Here we investigated the size-dependence of nanoparticle delivery into the brain assisted by FUS-induced BBB opening, using gold nanoparticles (AuNPs) of 3, 15, and 120 nm diameter. For 3- and 15-nm AuNPs, FUS exposure significantly increased permeation across an in vitro BBB model by up to 9.5 times, and the permeability was higher with smaller diameter. However, in vivo transcranial FUS exposure in mice demonstrated that smaller particles were not necessarily better for delivery into the brain. Medium-sized (15 nm) AuNPs showed the highest delivery efficiency (0.22% ID), compared with 3- and 120-nm particles. A computational model suggested that this optimum size was determined by the competition between their permeation through opened BBB gaps and their excretion from blood. Our results would greatly contribute to designing nanoparticles for their delivery into the brain for the treatment of central nervous system diseases.
Collapse
Affiliation(s)
- Seiichi Ohta
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan. .,Institute of Engineering Innovation, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| | - Emi Kikuchi
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Ayumu Ishijima
- Department of Precision Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Takashi Azuma
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Ichiro Sakuma
- Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.,Department of Precision Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| | - Taichi Ito
- Center for Disease Biology and Integrative Medicine, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8655, Japan.,Department of Bioengineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan
| |
Collapse
|
26
|
Lee LKC, Leong LI, Liu Y, Luo M, Chan HYE, Choi CHJ. Preclinical Nanomedicines for Polyglutamine-Based Neurodegenerative Diseases. Mol Pharm 2020; 18:610-626. [PMID: 32584043 DOI: 10.1021/acs.molpharmaceut.0c00506] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Polyglutamine (polyQ) diseases, such as Huntington's disease and several types of spinocerebellar ataxias, are dominantly inherited progressive neurodegenerative disorders and characterized by the presence of expanded CAG trinucleotide repeats in the respective disease locus of the patient genomes. Patients with polyQ diseases currently need to rely on symptom-relieving treatments because disease-modifying therapeutic interventions remain scarce. Many disease-modifying therapeutic agents are now under clinical testing for treating polyQ diseases, but their delivery to the brain is often too invasive (e.g., intracranial injection) or inefficient, owing to in vivo degradation and clearance by physiological barriers (e.g., oral and intravenous administration). Nanoparticles provide a feasible solution for improving drug delivery to the brain, as evidenced by an increasing number of preclinical studies that document the efficacy of nanomedicines for polyQ diseases over the past 5-6 years. In this review, we present the pathogenic mechanisms of polyQ diseases, the common animal models of polyQ diseases for evaluating the efficacy of nanomedicines, and the common administration routes for delivering nanoparticles to the brain. Next, we summarize the recent preclinical applications of nanomedicines for treating polyQ diseases and improving neurological conditions in vivo, placing emphasis on antisense oligonucleotides, small peptide inhibitors, and small molecules as the disease-modifying agents. We conclude with our perspectives of the burgeoning field of "nanomedicines for polyQ diseases", including the use of inorganic nanoparticles and potential drugs as next-generation nanomedicines, development of higher-order animal models of polyQ diseases, and importance of "brain-nano" interactions.
Collapse
Affiliation(s)
| | | | | | - Meihua Luo
- Monash Institute of Pharmaceutics Science, Monash University, Parkville, Victoria 3052, Australia
| | | | | |
Collapse
|
27
|
Nowak M, Brown TD, Graham A, Helgeson ME, Mitragotri S. Size, shape, and flexibility influence nanoparticle transport across brain endothelium under flow. Bioeng Transl Med 2020; 5:e10153. [PMID: 32440560 PMCID: PMC7237148 DOI: 10.1002/btm2.10153] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/28/2019] [Accepted: 12/11/2019] [Indexed: 12/19/2022] Open
Abstract
Nanoparticle-based therapeutic formulations are being increasingly explored for the treatment of various ailments. Despite numerous advances, the success of nanoparticle-based technologies in treating brain diseases has been limited. Translational hurdles of nanoparticle therapies are attributed primarily to their limited ability to cross the blood-brain barrier (BBB), which is one of the body's most exclusive barriers. Several efforts have been focused on developing affinity-based agents and using them to increase nanoparticle accumulation at the brain endothelium. Very little is known about the role of fundamental physical parameters of nanoparticles such as size, shape, and flexibility in determining their interactions with and penetration across the BBB. Using a three-dimensional human BBB microfluidic model (μHuB), we investigate the impact of these physical parameters on nanoparticle penetration across the BBB. To gain insights into the dependence of transport on nanoparticle properties, two separate parameters were measured: the number of nanoparticles that fully cross the BBB and the number that remain associated with the endothelium. Association of nanoparticles with the brain endothelium was substantially impacted by their physical characteristics. Hard particles associate more with the endothelium compared to soft particles, as do small particles compared to large particles, and spherical particles compared to rod-shaped particles. Transport across the BBB also exhibited a dependence on nanoparticle properties. A nonmonotonic dependence on size was observed, where 200 nm particles exhibited higher BBB transport compared to 100 and 500 nm spheres. Rod-shaped particles exhibited higher BBB transport when normalized by endothelial association and soft particles exhibited comparable transport to hard particles when normalized by endothelial association. Tuning nanoparticles' physical parameters could potentially enhance their ability to cross the BBB for therapeutic applications.
Collapse
Affiliation(s)
- Maksymilian Nowak
- John A. Paulson School of Engineering and Applied SciencesHarvard University29 Oxford St. CambridgeMA02138
- Wyss Institute of Biologically Inspired EngineeringHarvard University3 Blackfan CircleBostonMA02115
| | - Tyler D. Brown
- John A. Paulson School of Engineering and Applied SciencesHarvard University29 Oxford St. CambridgeMA02138
- Wyss Institute of Biologically Inspired EngineeringHarvard University3 Blackfan CircleBostonMA02115
| | - Adam Graham
- Center for Nanoscale SystemsHarvard University11 Oxford St. CambridgeMA02138
| | - Matthew E. Helgeson
- Department of Chemical EngineeringUniversity of California, Santa BarbaraSanta BarbaraCA93106
| | - Samir Mitragotri
- John A. Paulson School of Engineering and Applied SciencesHarvard University29 Oxford St. CambridgeMA02138
- Wyss Institute of Biologically Inspired EngineeringHarvard University3 Blackfan CircleBostonMA02115
| |
Collapse
|
28
|
Paris JL, Vallet-Regí M. Ultrasound-Activated Nanomaterials for Therapeutics. BULLETIN OF THE CHEMICAL SOCIETY OF JAPAN 2020. [DOI: 10.1246/bcsj.20190346] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Juan L. Paris
- Department of Life Sciences, Nano4Health Unit, Nanomedicine Group. International Iberian Nanotechnology Laboratory (INL), Av. Mestre José Veiga s/n, 4715-330 Braga, Portugal
| | - María Vallet-Regí
- Dpto. Química en Ciencias Farmacéuticas (Unidad Docente de Química Inorgánica y Bioinorgánica), Facultad de Farmacia, Universidad Complutense de Madrid, Instituto de Investigación Sanitaria Hospital 12 de Octubre (imas12), 28040-Madrid, Spain
- Centro de Investigación Biomédica en Red de Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Spain
| |
Collapse
|
29
|
Srinivasan VM, Lang FF, Chen SR, Chen MM, Gumin J, Johnson J, Burkhardt JK, Kan P. Advances in endovascular neuro-oncology: endovascular selective intra-arterial (ESIA) infusion of targeted biologic therapy for brain tumors. J Neurointerv Surg 2020; 12:197-203. [PMID: 31676690 DOI: 10.1136/neurintsurg-2019-015137] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2019] [Revised: 10/12/2019] [Accepted: 10/15/2019] [Indexed: 02/03/2023]
Abstract
BACKGROUND Malignant gliomas continue to have a poor clinical outcome with available therapies. In the past few years, new targeted biologic therapies have been studied, with promising results. However, owing to problems with ineffective IV delivery of these newer agents, an alternative, more direct delivery mechanism is needed. Simultaneously, advancements in neuroendovascular technology have allowed endovascular selective intra-arterial approaches to delivery. This method has the potential to increase drug delivery and selectively target tumor vasculature. OBJECTIVE To review the history of IA therapy for brain tumors, prior failures and successes, the emergence of new technologies and therapies, and the future direction of this young field. METHODS A comprehensive literature search of two databases (PubMed, Ovid Medline) was performed for several terms including 'brain tumor', 'glioma', and 'endovascular intra-arterial'. Forty-five relevant articles were identified via a systematic review following PRISMA guidelines. Additional relevant articles were selected for further in-depth review. Emphasis was given to articles discussing selective intra-arterial intracranial delivery using microcatheters. RESULTS Endovascular intra-arterial therapy with chemotherapy has had mixed results, with currently active trials using temozolomide, cetuximab, and bevacizumab. Prior attempts at IA chemotherapy with older-generation medications did not surpass the efficacy of IV administration. Advances in neuro-oncology have brought to the forefront new targeted biologic therapies. CONCLUSIONS In this review, we discuss the emerging field of endovascular neuro-oncology, a field that applies modern neuroendovascular techniques to the delivery of new therapeutic agents to brain tumors. The development of targeted therapies for brain tumors has been concurrent with the development of microcatheter technology, which has made superselective distal intracranial arterial access feasible and safe.
Collapse
Affiliation(s)
| | - Frederick F Lang
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Stephen R Chen
- Department of Radiology, Baylor College of Medicine, Houston, Texas, USA.,Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Melissa M Chen
- Department of Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Joy Gumin
- Department of Neurosurgery, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Jeremiah Johnson
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Jan-Karl Burkhardt
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| | - Peter Kan
- Department of Neurosurgery, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
30
|
Lombardo SM, Schneider M, Türeli AE, Günday Türeli N. Key for crossing the BBB with nanoparticles: the rational design. BEILSTEIN JOURNAL OF NANOTECHNOLOGY 2020; 11:866-883. [PMID: 32551212 PMCID: PMC7277618 DOI: 10.3762/bjnano.11.72] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/08/2020] [Accepted: 05/08/2020] [Indexed: 05/15/2023]
Abstract
Central nervous system diseases are a heavy burden on society and health care systems. Hence, the delivery of drugs to the brain has gained more and more interest. The brain is protected by the blood-brain barrier (BBB), a selective barrier formed by the endothelial cells of the cerebral microvessels, which at the same time acts as a bottleneck for drug delivery by preventing the vast majority of drugs to reach the brain. To overcome this obstacle, drugs can be loaded inside nanoparticles that can carry the drug through the BBB. However, not all particles are able to cross the BBB and a multitude of factors needs to be taken into account when developing a carrier system for this purpose. Depending on the chosen pathway to cross the BBB, nanoparticle material, size and surface properties such as functionalization and charge should be tailored to fit the specific route of BBB crossing.
Collapse
Affiliation(s)
- Sonia M Lombardo
- MyBiotech GmbH; Industriestraße 1B, 66802 Überherrn, Germany
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, 66123 Saarbrücken, Germany
| | - Marc Schneider
- Department of Pharmacy, Biopharmaceutics and Pharmaceutical Technology, Saarland University, Campus C4 1, 66123 Saarbrücken, Germany
| | - Akif E Türeli
- MyBiotech GmbH; Industriestraße 1B, 66802 Überherrn, Germany
| | | |
Collapse
|
31
|
Overview of Current Drug Delivery Methods Across the Blood-Brain Barrier for the Treatment of Primary Brain Tumors. CNS Drugs 2020; 34:1121-1131. [PMID: 32965590 PMCID: PMC7658069 DOI: 10.1007/s40263-020-00766-w] [Citation(s) in RCA: 66] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/05/2020] [Indexed: 01/05/2023]
Abstract
Existing drug delivery methods have not led to a significant increase in survival for patients with malignant primary brain tumors. While the combination of conventional therapies consisting of surgery, radiotherapy, and chemotherapy has improved survival for some types of brain tumors (e.g., WNT medulloblastoma), other types of brain tumors (e.g., glioblastoma and diffuse midline glioma) still have a poor prognosis. The reason for the differences in response can be largely attributed to the blood-brain barrier (BBB), a specialized structure at the microvasculature level that regulates the transport of molecules across the blood vessels into the brain parenchyma. This structure hampers the delivery of most chemotherapeutic agents for the treatment of primary brain tumors. Several drug delivery methods such as nanoparticles, convection enhanced delivery, focused ultrasound, intranasal delivery, and intra-arterial delivery have been developed to overcome the BBB in primary brain tumors. However, prognosis of most primary brain tumors still remains poor. The heterogeneity of the BBB in primary brain tumors and the distinct vasculature of tumors make it difficult to design a drug delivery method that targets the entire tumor. Drug delivery methods that combine strategies such as focused ultrasound and nanoparticles might be a more successful approach. However, more research is needed to optimize and develop new drug delivery techniques to improve survival of patients with primary brain tumors.
Collapse
|
32
|
Bidram E, Esmaeili Y, Ranji-Burachaloo H, Al-Zaubai N, Zarrabi A, Stewart A, Dunstan DE. A concise review on cancer treatment methods and delivery systems. J Drug Deliv Sci Technol 2019. [DOI: 10.1016/j.jddst.2019.101350] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
|
33
|
Sheybani ND, Price RJ. Perspectives on Recent Progress in Focused Ultrasound Immunotherapy. Am J Cancer Res 2019; 9:7749-7758. [PMID: 31695798 PMCID: PMC6831458 DOI: 10.7150/thno.37131] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 07/28/2019] [Indexed: 02/06/2023] Open
Abstract
Immunotherapy holds tremendous promise as a strategy for eradicating solid tumors. However, poor T cell infiltration and persistence within most solid tumor microenvironments, as well as mechanisms of adaptive resistance, continue to severely limit the accessibility of most immunotherapies to a broad patient population. This limitation perpetuates the demand for allied therapeutic strategies. Among such strategies is focused ultrasound (FUS), a non-invasive, non-ionizing technique for precisely targeted acoustic energy deposition into tissues. FUS has gained remarkable attention over recent years as a modality for elicitation of immune mechanisms in cancer and other pathologies. In 2017, we published a comprehensive review paper detailing existing evidence for immune modulation and therapy with FUS, as well as impending challenges and opportunities of consideration for the field. Over the last two years, a multitude of clinical trials have come online to explore safety, feasibility, and efficacy of FUS for cancers of the brain and periphery - including the first clinical trial to combine FUS with immunotherapy. Moreover, the last two years have seen a surge in FUS immunotherapy presentations at therapeutic ultrasound scientific meetings. Given the burst of activity in this field, we submit that an update on FUS immunotherapy progress is timely. In this review, we offer an updated overview and perspectives on scientific and clinical development in the FUS immunotherapy domain.
Collapse
|
34
|
Nowak M, Helgeson ME, Mitragotri S. Delivery of Nanoparticles and Macromolecules across the Blood–Brain Barrier. ADVANCED THERAPEUTICS 2019. [DOI: 10.1002/adtp.201900073] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/01/2023]
Affiliation(s)
- Maksymilian Nowak
- School of Engineering and Applied Sciences Harvard University 29 Oxford St. Cambridge MA 02318 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University 3 Blackfan Circle Boston MA 02115 USA
| | - Matthew E. Helgeson
- Department of Chemical Engineering University of California Santa Barbara Santa Barbara CA 93106 USA
| | - Samir Mitragotri
- School of Engineering and Applied Sciences Harvard University 29 Oxford St. Cambridge MA 02318 USA
- Wyss Institute of Biologically Inspired Engineering Harvard University 3 Blackfan Circle Boston MA 02115 USA
| |
Collapse
|
35
|
Focused Ultrasonography-Mediated Blood-Brain Barrier Disruption in the Enhancement of Delivery of Brain Tumor Therapies. World Neurosurg 2019; 131:65-75. [PMID: 31323404 DOI: 10.1016/j.wneu.2019.07.096] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 07/09/2019] [Accepted: 07/10/2019] [Indexed: 01/06/2023]
Abstract
Glioblastoma is the most common intracranial malignancy in adults and carries a poor prognosis. Chemotherapeutic treatment figures prominently in the management of primary and recurrent disease. However, the blood-brain barrier presents a significant and formidable impediment to the entry of oncotherapeutic compounds to target tumor tissue. Several strategies have been developed to effect disruption of the blood-brain barrier and in turn enhance the efficacy of cytotoxic chemotherapy, as well as newly developed biologic agents. Focused ultrasonography is one such treatment modality, using acoustic cavitation of parenterally administered microbubbles to mechanically effect disruption of the vascular endothelium. We review and discuss the preclinical and clinical studies evaluating the biophysical basis for, and efficacy of, focused ultrasonography in the enhancement of oncotherapeutic agent delivery. Further, we provide some perspectives regarding future directions for the role of focused ultrasound in facilitating and improving the safe and effective delivery of oncotherapeutic agents in the treatment of glioblastoma.
Collapse
|
36
|
Aryal M, Papademetriou I, Zhang YZ, Power C, McDannold N, Porter T. MRI Monitoring and Quantification of Ultrasound-Mediated Delivery of Liposomes Dually Labeled with Gadolinium and Fluorophore through the Blood-Brain Barrier. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:1733-1742. [PMID: 31010598 PMCID: PMC6555669 DOI: 10.1016/j.ultrasmedbio.2019.02.024] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/25/2018] [Revised: 01/11/2019] [Accepted: 02/28/2019] [Indexed: 05/15/2023]
Abstract
Magnetic resonance image-guided focused ultrasound has emerged as a viable non-invasive technique for the treatment of central nervous system-related diseases/disorders. Application of mechanical and thermal effects associated with focused transcranial ultrasound has been studied extensively in pre-clinical models, which has paved the way for clinical trials. However, in vivo treatment evaluation techniques on drug delivery application via blood-brain barrier opening has not been fully explored. Current treatment evaluation techniques via magnetic resonance imaging are hindered by systemic toxicity resulting from free gadolinium delivery. Here we propose a novel treatment evaluation strategy to overcome limitations by (i) synthesizing liposomes that are dually labeled with gadolinium, a magnetic resonance imaging (MRI) contrast agent, and rhodamine, a fluorophore; (ii) applying a focused ultrasound (FUS)-mediated BBB opening technique to deliver the liposomes across vascular barriers, achieving local gadolinium enhancement while reducing systemic and unwanted regional toxic effects associated with free gadolinium; and (iii) utilizing the MRI modality to confirm the delivery as it is already included in the FUS treatment in clinic. Liposomes were secondarily labeled with a fluorescent marker to confirm results obtained by MRI quantification postmortem. Two different sizes, 77.5 nm (group A) and 140 nm (group B), of gadolinium- and fluorescence-labeled liposomes were fabricated using thin-film hydration followed by extrusion methods and determined their stability up to 6 h under physiologic conditions. Gadolinium signal was detected on contrast-enhanced T1-weighted MRI 5 h after the delivery of liposomes via the BBB opening approach with an ultrasound pulse of 0.42 MPa (estimate in water) combined with microbubbles. MRI contrast was enhanced significantly in sonicated regions compared with non-sonicated regions of the brain. This was due to the accumulation of labeled liposomes, which was confirmed by detection of rhodamine fluorescence in histologic sections. The relative increase in MRI signal intensity was greater for smaller liposomes (mean diameter = 77.5 nm) than larger liposomes (mean diameter = 140 nm), which suggested a greater accumulation of the smaller liposomes in the brain after ultrasound-mediated opening of the BBB. Our findings suggest that the dual-labeled nanocarrier platform can be established, the FUS-mediated BBB opening approach can be used to deliver it through vascular barriers and MRI can be used to evaluate the extent of nanocarrier delivery.
Collapse
Affiliation(s)
- Muna Aryal
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA.
| | - Iason Papademetriou
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts, USA
| | - Yong-Zhi Zhang
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Chanikarn Power
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Nathan McDannold
- Department of Radiology, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | - Tyrone Porter
- Department of Mechanical Engineering, Boston University, Boston, Massachusetts, USA; Department of Biomedical Engineering, Boston University, Boston, Massachusetts, USA
| |
Collapse
|
37
|
A Monte Carlo study on the radio-sensitization effect of gold nanoparticles in brachytherapy of prostate by 103Pd seeds. POLISH JOURNAL OF MEDICAL PHYSICS AND ENGINEERING 2019. [DOI: 10.2478/pjmpe-2019-0012] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Abstract
103Pd seed is being used for prostate brachytherapy. Additionally, the dose enhancement effect of gold nanoparticles (GNP) has been reported in previous studies. The aim of this study was to characterize the dosimetric effect of gold nanoparticles in brachytherapy with a 103Pd source. Two brachytherapy seeds including 103 Pd source was simulated using MCNPX Monte Carlo code. The seeds’ models were validated by comparing the MC with reported results. Then, GNPs (10 nm in diameter) with a concentration of 7mg Au/g were simulated uniformly inside the prostate of a humanoid computational phantom. Additionally, the dose enhancement factor (DEF) of nanoparticles was calculated for both modeled brachytherapy seeds. A good agreement was found between the MC calculated and the reported dosimetric parameters. For both seeds, an average DEF of 23% was obtained in tumor volume for prostate brachytherapy. The application of GNPs in conjunction with 103Pd seed in brachytherapy can enhance the delivered dose to the tumor and consequently leads to better treatment outcome.
Collapse
|
38
|
|
39
|
Jones RM, Hynynen K. Advances in acoustic monitoring and control of focused ultrasound-mediated increases in blood-brain barrier permeability. Br J Radiol 2019; 92:20180601. [PMID: 30507302 DOI: 10.1259/bjr.20180601] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
Transcranial focused ultrasound (FUS) combined with intravenously circulating microbubbles can transiently and selectively increase blood-brain barrier permeability to enable targeted drug delivery to the central nervous system, and is a technique that has the potential to revolutionize the way neurological diseases are managed in medical practice. Clinical testing of this approach is currently underway in patients with brain tumors, early Alzheimer's disease, and amyotrophic lateral sclerosis. A major challenge that needs to be addressed in order for widespread clinical adoption of FUS-mediated blood-brain barrier permeabilization to occur is the development of systems and methods for real-time treatment monitoring and control, to ensure that safe and effective acoustic exposure levels are maintained throughout the procedures. This review gives a basic overview of the oscillation dynamics, acoustic emissions, and biological effects associated with ultrasound-stimulated microbubbles in vivo, and provides a summary of recent advances in acoustic-based strategies for detecting, controlling, and mapping microbubble activity in the brain. Further development of next-generation clinical FUS brain devices tailored towards microbubble-mediated applications is warranted and required for translation of this potentially disruptive technology into routine clinical practice.
Collapse
Affiliation(s)
- Ryan M Jones
- 1 Physical Sciences Platform, Sunnybrook Research Institute , Toronto, ON , Canada
| | - Kullervo Hynynen
- 1 Physical Sciences Platform, Sunnybrook Research Institute , Toronto, ON , Canada.,2 Department of Medical Biophysics, University of Toronto , Toronto, ON , Canada.,3 Institute of Biomaterials and Biomedical Engineering, University of Toronto , Toronto, ON , Canada
| |
Collapse
|
40
|
Nanodelivery systems for overcoming limited transportation of therapeutic molecules through the blood-brain barrier. Future Med Chem 2018; 10:2659-2674. [PMID: 30499740 DOI: 10.4155/fmc-2018-0208] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Due to the impermeable structure and barrier function of the blood-brain barrier (BBB), the delivery of therapeutic molecules into the CNS is extremely limited. Nanodelivery systems are regarded as the most effective and versatile carriers for the CNS, as they can transport cargo molecules across the BBB via various mechanisms. This review emphasizes the multi-functionalization strategies of nanodelivery systems and combinatorial approaches for the delivery of therapeutic drugs and genes into the CNS. The characteristics and functions of the BBB and underlying mechanisms of molecular translocation across the BBB are also described.
Collapse
|
41
|
Poellmann MJ, Bu J, Hong S. Would antioxidant-loaded nanoparticles present an effective treatment for ischemic stroke? Nanomedicine (Lond) 2018; 13:2327-2340. [DOI: 10.2217/nnm-2018-0084] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Ischemic stroke is a leading cause of death and disability worldwide and is in urgent need of new treatment options. The only approved treatment for stroke restores blood flow to the brain, but much of the tissue damage occurs during the subsequent reperfusion. Antioxidant therapies that directly address ischemia-reperfusion injury have shown promise in preclinical results. In this review, we discuss that reformulating antioxidant therapies as nanomedicine can potentially overcome the barriers that have kept these therapies from succeeding in the clinic. We begin by reviewing the pathophysiology of ischemic stroke with a focus on the effects of reperfusion injury. Next, we review nanotherapeutic systems designed to treat the disease with a focus on those addressing reperfusion injury. Mechanisms of passive and active transport required to traverse a blood–brain barrier are discussed. Finally, we conclude by outlining design parameters for potentially successful nanomedicines as front-line therapeutics for ischemic stroke.
Collapse
Affiliation(s)
- Michael J Poellmann
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Jiyoon Bu
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
| | - Seungpyo Hong
- Pharmaceutical Sciences Division, School of Pharmacy, University of Wisconsin, Madison, WI 53705, USA
- Carbone Cancer Center, School of Medicine & Public Health, University of Wisconsin, Madison, WI 53792, USA
- Yonsei Frontier Lab & Department of Pharmacy, Yonsei University, Seoul 03722, Korea
| |
Collapse
|
42
|
McMahon D, Mah E, Hynynen K. Angiogenic response of rat hippocampal vasculature to focused ultrasound-mediated increases in blood-brain barrier permeability. Sci Rep 2018; 8:12178. [PMID: 30111814 PMCID: PMC6093874 DOI: 10.1038/s41598-018-30825-8] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 08/06/2018] [Indexed: 01/12/2023] Open
Abstract
Focused ultrasound (FUS) and circulating microbubbles can induce a targeted and transient increase in blood-brain barrier permeability. While preclinical research has demonstrated the utility of FUS for efficacious drug deliver to the brain, there remain gaps in our knowledge regarding the long-term response of brain vasculature to this intervention. Previous work has demonstrated transcriptional changes in hippocampal microvessels following sonication that are indicative of the initiation of angiogenic processes. Moreover, blood vessel growth has been reported in skeletal muscle following application of FUS and microbubbles. The current study demonstrates that blood vessel density in the rat hippocampus is modestly elevated at 7 and 14 d post-FUS compared to the contralateral hemisphere (7 d: 10.9 ± 6.0%, p = 0.02; 14 d: 12.1 ± 3.2%, p < 0.01), but returns to baseline by 21 d (5.9 ± 2.6%, p = 0.12). Concurrently, relative newborn endothelial cell density and frequency of small blood vessel segments were both elevated in the sonicated hippocampus. While further work is required to determine the mechanisms driving these changes, the findings presented here may have relevance to the optimal frequency of repeated treatments.
Collapse
Affiliation(s)
- Dallan McMahon
- Sunnybrook Research Institute, Toronto, M4N 3M5, Canada. .,University of Toronto, Department of Medical Biophysics, Toronto, M4N 3M5, Canada.
| | - Ethan Mah
- Sunnybrook Research Institute, Toronto, M4N 3M5, Canada
| | - Kullervo Hynynen
- Sunnybrook Research Institute, Toronto, M4N 3M5, Canada.,University of Toronto, Department of Medical Biophysics, Toronto, M4N 3M5, Canada.,University of Toronto, Institute of Biomaterials and Biomedical Engineering, Toronto, M5S 3G9, Canada
| |
Collapse
|
43
|
Zhou M, Tang M, Li S, Peng L, Huang H, Fang Q, Liu Z, Xie P, Li G, Zhou J. Effective lock-in strategy for proteomic analysis of corona complexes bound to amino-free ligands of gold nanoparticles. NANOSCALE 2018; 10:12413-12423. [PMID: 29926046 DOI: 10.1039/c8nr01077c] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
For specific applications, gold nanoparticles (GNPs) are commonly functionalized with various biological ligands, including amino-free ligands such as amino acids, peptides, proteins, and nucleic acids. Upon entering a biological fluid, the protein corona that forms around GNPs can conceal the targeting ligands and sterically hinder the functional properties. The protein corona is routinely prepared by standard centrifugation or sucrose cushion centrifugation. However, such methodologies are not applicable to the exclusive analysis of a ligand-binding protein corona. In this study, we first proposed a lock-in strategy based on a combination of rapid crosslinking and stringent washing. Cysteine was used as a model of amino-free ligands and attached to GNPs. After corona formation in the human plasma, GNP cysteine and corona proteins were quickly fixed by 5 s of crosslinking with 7.5% formaldehyde. After stringent washing using SDS buffer with sonication, the cysteine-bound proteins were effectively separated from unbound proteins. Qualitative and quantitative analyses using a mass spectrometry-based proteomics approach indicated that the protein composition of the cysteine-binding corona from the new method was significantly different from the composition of the whole corona from the two conventional methods. Furthermore, network and formaldehyde-linked site analyses of cysteine-binding proteins provided useful information toward a better knowledge of the behavior of protein-ligand and protein-protein interactions. Collectively, our new strategy has the capability to particularly characterize the protein composition of a cysteine-binding corona. The presented methodology in principal provides a generic way to analyze a nanoparticle corona bound to amino-free ligands and has the potential to decipher corona-masked ligand functions.
Collapse
Affiliation(s)
- Mi Zhou
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing 400016, China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Ye D, Sultan D, Zhang X, Yue Y, Heo GS, Kothapalli SVVN, Luehmann H, Tai YC, Rubin JB, Liu Y, Chen H. Focused ultrasound-enabled delivery of radiolabeled nanoclusters to the pons. J Control Release 2018; 283:143-150. [PMID: 29864474 DOI: 10.1016/j.jconrel.2018.05.039] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2017] [Revised: 03/15/2018] [Accepted: 05/31/2018] [Indexed: 01/17/2023]
Abstract
The goal of this study was to establish the feasibility of integrating focused ultrasound (FUS)-mediated delivery of 64Cu-integrated gold nanoclusters (64Cu-AuNCs) to the pons for in vivo quantification of the nanocluster brain uptake using positron emission tomography (PET) imaging. FUS was targeted at the pons for the blood-brain barrier (BBB) disruption in the presence of systemically injected microbubbles, followed by the intravenous injection of 64Cu-AuNCs. The spatiotemporal distribution of the 64Cu-AuNCs in the brain was quantified using in vivo microPET/CT imaging at different time points post injection. Following PET imaging, the accumulation of radioactivity in the pons was further confirmed using autoradiography and gamma counting, and the gold concentration was quantified using inductively coupled plasma-mass spectrometry (ICP-MS). We found that the noninvasive and localized BBB opening by the FUS successfully delivered the 64Cu-AuNCs to the pons. We also demonstrated that in vivo real-time microPET/CT imaging was a reliable method for monitoring and quantifying the brain uptake of 64Cu-AuNCs delivered by the FUS. This drug delivery platform that integrates FUS, radiolabeled nanoclusters, and PET imaging provides a new strategy for noninvasive and localized nanoparticle delivery to the pons with concurrent in vivo quantitative imaging to evaluate delivery efficiency. The long-term goal is to apply this drug delivery platform to the treatment of pontine gliomas.
Collapse
Affiliation(s)
- Dezhuang Ye
- Department of Mechanical Engineering and Material Science, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Deborah Sultan
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Xiaohui Zhang
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yimei Yue
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Gyu Seong Heo
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Satya V V N Kothapalli
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA
| | - Hannah Luehmann
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yuan-Chuan Tai
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Joshua B Rubin
- Department of Pediatrics, Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Yongjian Liu
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| | - Hong Chen
- Department of Biomedical Engineering, Washington University in St. Louis, Saint Louis, MO 63130, USA; Department of Radiation Oncology, Washington University School of Medicine, Saint Louis, MO 63108, USA.
| |
Collapse
|
45
|
Mathew RK, Rutka JT. Diffuse Intrinsic Pontine Glioma : Clinical Features, Molecular Genetics, and Novel Targeted Therapeutics. J Korean Neurosurg Soc 2018; 61:343-351. [PMID: 29742880 PMCID: PMC5957322 DOI: 10.3340/jkns.2018.0008] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2018] [Accepted: 01/21/2018] [Indexed: 12/18/2022] Open
Abstract
Diffuse intrinsic pontine glioma (DIPG) is a deadly paediatric brain cancer. Transient response to radiation, ineffective chemotherapeutic agents and aggressive biology result in rapid progression of symptoms and a dismal prognosis. Increased availability of tumour tissue has enabled the identification of histone gene aberrations, genetic driver mutations and methylation changes, which have resulted in molecular and phenotypic subgrouping. However, many of the underlying mechanisms of DIPG oncogenesis remain unexplained. It is hoped that more representative in vitro and preclinical models–using both xenografted material and genetically engineered mice–will enable the development of novel chemotherapeutic agents and strategies for targeted drug delivery. This review provides a clinical overview of DIPG, the barriers to progress in developing effective treatment, updates on drug development and preclinical models, and an introduction to new technologies aimed at enhancing drug delivery.
Collapse
Affiliation(s)
- Ryan K Mathew
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada.,Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK.,Department of Neurosurgery, Leeds General Infirmary, Leeds, UK
| | - James T Rutka
- Arthur and Sonia Labatt Brain Tumour Research Centre, The Hospital for Sick Children, Toronto, Canada.,Division of Neurosurgery, Department of Surgery, University of Toronto, Toronto, Canada
| |
Collapse
|
46
|
Gorick CM, Sheybani ND, Curley CT, Price RJ. Listening in on the Microbubble Crowd: Advanced Acoustic Monitoring for Improved Control of Blood-Brain Barrier Opening with Focused Ultrasound. Theranostics 2018; 8:2988-2991. [PMID: 29897053 PMCID: PMC5996352 DOI: 10.7150/thno.26025] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2018] [Accepted: 03/12/2018] [Indexed: 01/21/2023] Open
Abstract
Non-invasive drug and gene delivery to the brain to treat central nervous system pathologies has long been inhibited by the blood-brain barrier. The activation of microbubbles with focused ultrasound has emerged as a promising non-invasive approach to circumvent this obstacle, by transiently disrupting the blood-brain barrier and permitting passage of systemically administered therapeutics into the tissue. Clinical trials are underway to evaluate the safety of this technique; however, concerns remain regarding the potential for the treatment to induce sterile inflammation or petechiae. In this issue of Theranostics, Jones et al.[1] address these concerns through the development of an advanced three-dimensional imaging system for monitoring acoustic emissions from oscillating microbubbles. When subharmonic emissions are detected with this system, focused ultrasound pressure is reduced by 50% for the remainder of the treatment. This serves to transiently open the blood-brain barrier without generating adverse effects. While the ideal configuration of the transducer array for treatment and monitoring still presents an area for further optimization, the approach indicates that the acoustic signature of microbubble behavior within the skull can be used to ensure safe and effective blood-brain barrier opening using focused ultrasound.
Collapse
Affiliation(s)
| | | | | | - Richard J. Price
- Department of Biomedical Engineering, University of Virginia, Charlottesville, VA
| |
Collapse
|
47
|
Li N, Jie MM, Yang M, Tang L, Chen SY, Sun XM, Tang B, Yang SM. Magnetic Gold Nanoparticle-Labeled Heparanase Monoclonal Antibody and its Subsequent Application for Tumor Magnetic Resonance Imaging. NANOSCALE RESEARCH LETTERS 2018; 13:106. [PMID: 29671088 PMCID: PMC5906410 DOI: 10.1186/s11671-018-2518-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 04/05/2018] [Indexed: 05/03/2023]
Abstract
Heparanase (HPA) is ubiquitously expressed in various metastatic malignant tumors; previous studies have demonstrated that HPA was a potential tumor-associated antigen (TAA) for tumor immunotherapy. We sought to evaluate the feasibility of HPA as a common TAA for magnetic resonance imaging (MRI) of tumor metastasis and its potential application in tumor molecular imaging. We prepared a targeted probe based on magnetic gold nanoparticles coupled with an anti-HPA antibody for the specific detection of HPA by MRI. The specificity of the targeted probe was validated in vitro by incubation of the probe with various tumor cells, and the probe was able to selectively detect HPA (+) cells. We found the probes displayed significantly reduced signal intensity in several tumor cells, and the signal intensity decreased significantly after the targeted probe was injected in tumor-bearing nude mice. In the study, we demonstrated that the HPA&GoldMag probe had excellent physical and chemical properties and immune activities and could specifically target many tumor cell tissues both in vitro and in vivo. This may provide an experimental base for molecular imaging of tumor highly expressing heparanase using HPA mAbs.
Collapse
Affiliation(s)
- Ning Li
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400030 China
- Department of Gastroenterology, Institute of Surgery research, Daping Hospital, Third Military Medical University (Army Medical University), Chongqing, 400042 China
| | - Meng-Meng Jie
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400030 China
| | - Min Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400030 China
| | - Li Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400030 China
| | - Si-Yuan Chen
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400030 China
| | - Xue-Mei Sun
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400030 China
| | - Bo Tang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400030 China
| | - Shi-Ming Yang
- Department of Gastroenterology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, 400030 China
| |
Collapse
|
48
|
Coluccia D, Figueiredo CA, Wu MY, Riemenschneider AN, Diaz R, Luck A, Smith C, Das S, Ackerley C, O'Reilly M, Hynynen K, Rutka JT. Enhancing glioblastoma treatment using cisplatin-gold-nanoparticle conjugates and targeted delivery with magnetic resonance-guided focused ultrasound. NANOMEDICINE-NANOTECHNOLOGY BIOLOGY AND MEDICINE 2018; 14:1137-1148. [PMID: 29471172 DOI: 10.1016/j.nano.2018.01.021] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 01/26/2018] [Accepted: 01/29/2018] [Indexed: 01/25/2023]
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor resulting in high rates of morbidity and mortality. A strategy to increase the efficacy of available drugs and enhance the delivery of chemotherapeutics through the blood brain barrier (BBB) is desperately needed. We investigated the potential of Cisplatin conjugated gold nanoparticle (GNP-UP-Cis) in combination with MR-guided Focused Ultrasound (MRgFUS) to intensify GBM treatment. Viability assays demonstrated that GNP-UP-Cis greatly inhibits the growth of GBM cells compared to free cisplatin and shows marked synergy with radiation therapy. Additionally, increased DNA damage through γH2AX phosphorylation was observed in GNP-UP-Cis treated cells, along with enhanced platinum concentrations. In vivo, GNP-UP-Cis greatly reduced the growth of GBM tumors and MRgFUS led to increased BBB permeability and GNP-drug delivery in brain tissue. Our studies suggest that GNP-Cis conjugates and MRgFUS can be used to focally enhance the delivery of targeted chemotherapeutics to brain tumors.
Collapse
Affiliation(s)
- Daniel Coluccia
- Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumor Research Centre
| | - Carlyn A Figueiredo
- Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumor Research Centre; Division of Laboratory Medicine and Pathobiology, the Hospital For Sick Children
| | - Megan YiJun Wu
- Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumor Research Centre
| | | | - Roberto Diaz
- Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumor Research Centre
| | - Amanda Luck
- Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumor Research Centre
| | - Christian Smith
- Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumor Research Centre
| | - Sunit Das
- Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumor Research Centre; Division of Laboratory Medicine and Pathobiology, the Hospital For Sick Children
| | - Cameron Ackerley
- Division of Laboratory Medicine and Pathobiology, the Hospital For Sick Children
| | - Meaghan O'Reilly
- Sunnybrook Health Sciences Centre Research Institute, the University of Toronto
| | - Kullervo Hynynen
- Sunnybrook Health Sciences Centre Research Institute, the University of Toronto
| | - James T Rutka
- Division of Neurosurgery, the Arthur and Sonia Labatt Brain Tumor Research Centre; Division of Laboratory Medicine and Pathobiology, the Hospital For Sick Children.
| |
Collapse
|
49
|
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
|
50
|
McLaughlan JR, Cowell DMJ, Freear S. Gold nanoparticle nucleated cavitation for enhanced high intensity focused ultrasound therapy. Phys Med Biol 2017; 63:015004. [PMID: 29098986 DOI: 10.1088/1361-6560/aa97e9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
High intensity focused ultrasound (HIFU) or focused ultrasound surgery is a non-invasive technique for the treatment of cancerous tissue, which is limited by difficulties in getting real-time feedback on treatment progress and long treatment durations. The formation and activity of acoustic cavitation, specifically inertial cavitation, during HIFU exposures has been demonstrated to enhance heating rates. However, without the introduction of external nuclei its formation an activity can be unpredictable, and potentially counter-productive. In this study, a combination of pulse laser illumination (839 nm), HIFU exposures (3.3 MHz) and plasmonic gold nanorods (AuNR) was demonstrated as a new approach for the guidance and enhancement of HIFU treatments. For imaging, short duration HIFU pulses (10 μs) demonstrated broadband acoustic emissions from AuNR nucleated cavitation with a signal-to-noise ranging from 5-35 dB for peak negative pressures between 1.19-3.19 ± 0.01 MPa. In the absence of either AuNR or laser illumination these emissions were either not present or lower in magnitude (e.g. 5 dB for 3.19 MPa). Continuous wave (CW) HIFU exposures for 15 s, were then used to generate thermal lesions for peak negative pressures from 0.2-2.71 ± 0.01 MPa at a fluence of 3.4 mJ [Formula: see text]. Inertial cavitation dose (ICD) was monitored during all CW exposures, where exposures combined with both laser illumination and AuNRs resulted in the highest level of detectable emissions. This parameter was integrated over the entire exposure to give a metric to compare with measured thermal lesion area, where it was found that a minimum total ICD of [Formula: see text] a.u. was correlated with the formation of thermal lesions in gel phantoms. Furthermore, lesion area (mm2) was increased for equivalent exposures without either AuNRs or laser illumination. Once combined with cancer targeting AuNRs this approach could allow for the future theranostic use of HIFU, such as providing the ability to identify and treat small multi-focal cancerous regions with minimal damage to surrounding healthy tissue.
Collapse
Affiliation(s)
- J R McLaughlan
- School of Electronic and Electrical Engineering, University of Leeds, Leeds, LS2 9JT, United Kingdom. Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | | | | |
Collapse
|