1
|
Johansen PM, Hansen PY, Mohamed AA, Girshfeld SJ, Feldmann M, Lucke-Wold B. Focused ultrasound for treatment of peripheral brain tumors. EXPLORATION OF DRUG SCIENCE 2023; 1:107-125. [PMID: 37171968 PMCID: PMC10168685 DOI: 10.37349/eds.2023.00009] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/11/2023] [Accepted: 04/13/2023] [Indexed: 05/14/2023]
Abstract
Malignant brain tumors are the leading cause of cancer-related death in children and remain a significant cause of morbidity and mortality throughout all demographics. Central nervous system (CNS) tumors are classically treated with surgical resection and radiotherapy in addition to adjuvant chemotherapy. However, the therapeutic efficacy of chemotherapeutic agents is limited due to the blood-brain barrier (BBB). Magnetic resonance guided focused ultrasound (MRgFUS) is a new and promising intervention for CNS tumors, which has shown success in preclinical trials. High-intensity focused ultrasound (HIFU) has the capacity to serve as a direct therapeutic agent in the form of thermoablation and mechanical destruction of the tumor. Low-intensity focused ultrasound (LIFU) has been shown to disrupt the BBB and enhance the uptake of therapeutic agents in the brain and CNS. The authors present a review of MRgFUS in the treatment of CNS tumors. This treatment method has shown promising results in preclinical trials including minimal adverse effects, increased infiltration of the therapeutic agents into the CNS, decreased tumor progression, and improved survival rates.
Collapse
Affiliation(s)
| | - Payton Yerke Hansen
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Ali A. Mohamed
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Sarah J. Girshfeld
- Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton, FL 33431, USA
| | - Marc Feldmann
- College of Medicine, University of Florida, Gainesville, FL 32611, USA
| | - Brandon Lucke-Wold
- Department of Neurosurgery, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
2
|
Lim Kee Chang W, Chan TG, Raguseo F, Mishra A, Chattenton D, de Rosales RTM, Long NJ, Morse SV. Rapid short-pulses of focused ultrasound and microbubbles deliver a range of agent sizes to the brain. Sci Rep 2023; 13:6963. [PMID: 37117169 PMCID: PMC10147927 DOI: 10.1038/s41598-023-33671-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/17/2023] [Indexed: 04/30/2023] Open
Abstract
Focused ultrasound and microbubbles can non-invasively and locally deliver therapeutics and imaging agents across the blood-brain barrier. Uniform treatment and minimal adverse bioeffects are critical to achieve reliable doses and enable safe routine use of this technique. Towards these aims, we have previously designed a rapid short-pulse ultrasound sequence and used it to deliver a 3 kDa model agent to mouse brains. We observed a homogeneous distribution in delivery and blood-brain barrier closing within 10 min. However, many therapeutics and imaging agents are larger than 3 kDa, such as antibody fragments and antisense oligonucleotides. Here, we evaluate the feasibility of using rapid short-pulses to deliver higher-molecular-weight model agents. 3, 10 and 70 kDa dextrans were successfully delivered to mouse brains, with decreasing doses and more heterogeneous distributions with increasing agent size. Minimal extravasation of endogenous albumin (66.5 kDa) was observed, while immunoglobulin (~ 150 kDa) and PEGylated liposomes (97.9 nm) were not detected. This study indicates that rapid short-pulses are versatile and, at an acoustic pressure of 0.35 MPa, can deliver therapeutics and imaging agents of sizes up to a hydrodynamic diameter between 8 nm (70 kDa dextran) and 11 nm (immunoglobulin). Increasing the acoustic pressure can extend the use of rapid short-pulses to deliver agents beyond this threshold, with little compromise on safety. This study demonstrates the potential for deliveries of higher-molecular-weight therapeutics and imaging agents using rapid short-pulses.
Collapse
Affiliation(s)
- William Lim Kee Chang
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Tiffany G Chan
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Federica Raguseo
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Aishwarya Mishra
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SW1 7EH, UK
| | - Dani Chattenton
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK
- Division of Radiotherapy and Imaging, The Institute of Cancer Research, Sutton, London, SM2 5NG, UK
| | - Rafael T M de Rosales
- School of Biomedical Engineering and Imaging Sciences, King's College London, St Thomas' Hospital, London, SW1 7EH, UK
| | - Nicholas J Long
- Department of Chemistry, Imperial College London, Molecular Sciences Research Hub, White City, London, W12 0BZ, UK
| | - Sophie V Morse
- Department of Bioengineering, Imperial College London, South Kensington, London, SW7 2BP, UK.
| |
Collapse
|
3
|
Engineered Graphene Quantum Dots as a Magnetic Resonance Signal Amplifier for Biomedical Imaging. Molecules 2023; 28:molecules28052363. [PMID: 36903608 PMCID: PMC10005761 DOI: 10.3390/molecules28052363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 02/19/2023] [Accepted: 02/24/2023] [Indexed: 03/08/2023] Open
Abstract
The application of magnetic resonance imaging (MRI) nano-contrast agents (nano-CAs) has increasingly attracted scholarly interest owing to their size, surface chemistry, and stability. Herein, a novel T1 nano-CA (Gd(DTPA)-GQDs) was successfully prepared through the functionalization of graphene quantum dots with poly(ethylene glycol) bis(amine) and their subsequent incorporation into Gd-DTPA. Remarkably, the resultant as-prepared nano-CA displayed an exceptionally high longitudinal proton relaxivity (r1) of 10.90 mM-1 s-1 (R2 = 0.998), which was significantly higher than that of commercial Gd-DTPA (4.18 mM-1 s-1, R2 = 0.996). The cytotoxicity studies indicated that the Gd(DTPA)-GQDs were not cytotoxic by themselves. The results of the hemolysis assay and the in vivo safety evaluation demonstrate the outstanding biocompatibility of Gd(DTPA)-GQDs. The in vivo MRI study provides evidence that Gd(DTPA)-GQDs exhibit exceptional performance as T1-CAs. This research constitutes a viable approach for the development of multiple potential nano-CAs with high-performance MR imaging capabilities.
Collapse
|
4
|
Tranos J, Das A, Zhang J, Hafeez S, Arvanitakis GN, Thomson SAJ, Khan S, Pandya N, Kim SG, Wadghiri YZ. Rapid In Vitro Quantification of a Sensitized Gadolinium Chelate via Photoinduced Triplet Harvesting. ACS OMEGA 2023; 8:2907-2914. [PMID: 36713694 PMCID: PMC9878670 DOI: 10.1021/acsomega.2c05040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Accepted: 11/18/2022] [Indexed: 06/18/2023]
Abstract
Gadolinium (Gd) based contrast agents (GBCAs) are widely used in magnetic resonance imaging (MRI) and are paramount to cancer diagnostics and tumor pharmacokinetic analysis. Accurate quantification of gadolinium concentration is essential to monitoring the biodistribution, clearance, and pharmacodynamics of GBCAs. However, current methods of quantifying gadolinium in blood or plasma (biological media) are both low throughput and clinically unavailable. Here, we have demonstrated the use of a sensitized gadolinium chelate, Gd[DTPA-cs124], as an MRI contrast agent that can be used to measure the concentration of gadolinium via luminescence quantification in biological media following transmetalation with a terbium salt. Gd[DTPA-cs124] was synthesized by conjugating carbostyril-124 (cs124) to diethylenetriaminepentaacetic acid (DTPA) and chelating to gadolinium. We report increases in both stability and relaxivity compared to the clinically approved analog Gd[DTPA] (gadopentetic acid or Magnevist). In vivo MRI experiments were conducted using C57BL6 mice in order to further illustrate the performance of Gd[DTPA-cs124] as an MRI contrast agent in comparison to Magnevist. Our results indicate that similar chemical modification to existing clinically approved GBCA may likewise provide favorable property changes, with the ability to be used in a gadolinium quantification assay. Furthermore, our assay provides a straightforward and high-throughput method of measuring gadolinium in biological media using a standard laboratory plate reader.
Collapse
Affiliation(s)
- James
A. Tranos
- Center
for Biomedical Imaging (CBI), Center for Advanced Imaging Innovation
and Research (CAI2R), Department of Radiology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Ayesha Das
- Department
of Radiology, Weill Cornell Medical College, New York, New York 10065, United States
| | - Jin Zhang
- Department
of Radiology, Weill Cornell Medical College, New York, New York 10065, United States
| | - Sonia Hafeez
- Center
for Biomedical Imaging (CBI), Center for Advanced Imaging Innovation
and Research (CAI2R), Department of Radiology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | | | | | - Suleiman Khan
- Center
for Biomedical Imaging (CBI), Center for Advanced Imaging Innovation
and Research (CAI2R), Department of Radiology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Neelam Pandya
- Center
for Biomedical Imaging (CBI), Center for Advanced Imaging Innovation
and Research (CAI2R), Department of Radiology, NYU Grossman School of Medicine, New York, New York 10016, United States
| | - Sungheon Gene Kim
- Department
of Radiology, Weill Cornell Medical College, New York, New York 10065, United States
| | - Youssef Z. Wadghiri
- Center
for Biomedical Imaging (CBI), Center for Advanced Imaging Innovation
and Research (CAI2R), Department of Radiology, NYU Grossman School of Medicine, New York, New York 10016, United States
| |
Collapse
|
5
|
Magnetic resonance imaging analysis predicts nanoparticle concentration delivered to the brain parenchyma. Commun Biol 2022; 5:964. [PMID: 36109574 PMCID: PMC9477799 DOI: 10.1038/s42003-022-03881-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 08/23/2022] [Indexed: 12/03/2022] Open
Abstract
Ultrasound in combination with the introduction of microbubbles into the vasculature effectively opens the blood brain barrier (BBB) to allow the passage of therapeutic agents. Increased permeability of the BBB is typically demonstrated with small-molecule agents (e.g., 1-nm gadolinium salts). Permeability to small-molecule agents, however, cannot reliably predict the transfer of remarkably larger molecules (e.g., monoclonal antibodies) required by numerous therapies. To overcome this issue, we developed a magnetic resonance imaging analysis based on the ΔR2* physical parameter that can be measured intraoperatively for efficient real-time treatment management. We demonstrate successful correlations between ΔR2* values and parenchymal concentrations of 3 differently sized (18 nm–44 nm) populations of liposomes in a rat model. Reaching an appropriate ΔR2* value during treatment can reflect the effective delivery of large therapeutic agents. This prediction power enables the achievement of desirable parenchymal drug concentrations, which is paramount to obtaining effective therapeutic outcomes. ΔR2* values from MRI analysis correlate with concentrations of liposomes in the size range of 18–44 nm in a rat model.
Collapse
|
6
|
Cheruku RR, Turowski SG, Durrani FA, Tabaczynski WA, Cacaccio J, Missert JR, Curtin L, Sexton S, Alberico R, Hendler CM, Spernyak JA, Grossman Z, Pandey RK. Tumor-Avid 3-(1'-Hexyloxy)ethyl-3-devinylpyrpyropheophorbide-a (HPPH)-3Gd(III)tetraxetan (DOTA) Conjugate Defines Primary Tumors and Metastases. J Med Chem 2022; 65:9267-9280. [PMID: 35763292 DOI: 10.1021/acs.jmedchem.2c00547] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
3-(1'-Hexyloxyethyl)-3-devinylpyropheophorbide-a (HPPH or Photochlor), a tumor-avid chlorophyll a derivative currently undergoing human clinical trials, was conjugated with mono-, di-, and tri-Gd(III)tetraxetan (DOTA) moieties. The T1/T2 relaxivity and in vitro PDT efficacy of these conjugates were determined. The tumor specificity of the most promising conjugate was also investigated at various time points in mice and rats bearing colon tumors, as well as rabbits bearing widespread metastases from VX2 systemic arterial disseminated metastases. All the conjugates showed significant T1 and T2 relaxivities. However, the conjugate containing 3-Gd(III)-aminoethylamido-DOTA at position 17 of HPPH demonstrated great potential for tumor imaging by both MR and fluorescence while maintaining its PDT efficacy. At an MR imaging dose (10 μmol/kg), HPPH-3Gd(III)DOTA did not cause any significant organ toxicity in mice, indicating its potential as a cancer imaging (MR and fluorescence) agent with an option to treat cancer by photodynamic therapy (PDT).
Collapse
Affiliation(s)
- Ravindra R Cheruku
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Steven G Turowski
- Translational Imaging Shared Resource, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Farukh A Durrani
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Walter A Tabaczynski
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Joseph Cacaccio
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Joseph R Missert
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Leslie Curtin
- Department of Laboratory Animal Resources (DLAR), Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Sandra Sexton
- Department of Laboratory Animal Resources (DLAR), Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Ronald Alberico
- Department of Radiology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Craig M Hendler
- Department of Radiology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Joseph A Spernyak
- Translational Imaging Shared Resource, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Zachary Grossman
- Department of Radiology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| | - Ravindra K Pandey
- Photodynamic Therapy Center, Cell Stress Biology, Roswell Park Comprehensive Cancer Center, Buffalo, New York 14263, United States
| |
Collapse
|
7
|
Yogesh Kumar K, Prashanth MK, Parashuram L, Palanivel B, Alharti FA, Jeon BH, Raghu MS. Gadolinium sesquisulfide anchored N-doped reduced graphene oxide for sensitive detection and degradation of carbendazim. CHEMOSPHERE 2022; 296:134030. [PMID: 35189195 DOI: 10.1016/j.chemosphere.2022.134030] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 01/29/2022] [Accepted: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Agriculture is having a major role in solving issues associated with food shortages across the globe. Carbendazim (CZM) is one of the fungicides which is commonly used in agriculture to grow crops in large quantities and fast. Monitoring CZM content is in high demand for environmental remediation. The present work deals with the synthesis of gadolinium sesquisulfide anchored Nitrogen-doped reduced graphene oxide (Gd2S3/NRGO) through a simple microwave-assisted method. X-ray diffraction and morphological studies confirm the formation of the nanocomposite. Gd2S3/NRGO showed enhanced activity both in electrochemical detection and light-driven degradation of CZM compared to Gd2S3 and NRGO. Gd2S3/NRGO modified glassy carbon electrode (GCE) exhibit a wide linear range of 0.01-450 μM CZM with 0.009 μM LOD using differential pulse voltammetry (DPV). Gd2S3/NRGO@GCE showed good selectivity, stability, and recovery (98.13-99.10%) in the river water sample. In addition, Gd2S3/NRGO has been explored towards the visible-light-induced degradation of CZM. The reactions conditions were optimized to achieve maximum efficiency. 94% of CZM was degraded within 90 min in presence of Gd2S3/NRGO. Mechanism of electrochemical redox reaction and degradation of CZM in presence of Gd2S3/NRGO has been explored to the maximum extent possible. Degradation intermediates were identified using LC-MS.
Collapse
Affiliation(s)
- K Yogesh Kumar
- Department of Chemistry, Faculty of Engineering and Technology, Jain University, Bangalore, 562112, India
| | - M K Prashanth
- Department of Chemistry, BNM Institute of Technology, Banashankari, Bangalore, 560070, India
| | - L Parashuram
- Department of Chemistry, New Horizon College of Engineering, Outer Ring Road, Bangalore, 560103, India
| | - Baskaran Palanivel
- Department of Physics, King Engineering College, Sriperumbudur, Kancheepuram, 602117, India
| | - Fahad A Alharti
- Department of Chemistry, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Byong-Hun Jeon
- Department of Earth Resources and Environmental Engineering, Hanyang University, 222, Wangsimni-ro, Seongdong-gu, Seoul, 04763, Republic of Korea.
| | - M S Raghu
- Department of Chemistry, New Horizon College of Engineering, Outer Ring Road, Bangalore, 560103, India.
| |
Collapse
|
8
|
Mukhopadhyay D, Sano C, AlSawaftah N, El-Awady R, Husseini GA, Paul V. Ultrasound-Mediated Cancer Therapeutics Delivery using Micelles and Liposomes: A Review. Recent Pat Anticancer Drug Discov 2021; 16:498-520. [PMID: 34911412 DOI: 10.2174/1574892816666210706155110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 03/02/2021] [Accepted: 03/21/2021] [Indexed: 11/22/2022]
Abstract
BACKGROUND Existing cancer treatment methods have many undesirable side effects that greatly reduce the quality of life of cancer patients. OBJECTIVE This review will focus on the use of ultrasound-responsive liposomes and polymeric micelles in cancer therapy. METHODS This review presents a survey of the literature regarding ultrasound-triggered micelles and liposomes using articles recently published in various journals, as well as some new patents in this field. RESULTS Nanoparticles have proven promising as cancer theranostic tools. Nanoparticles are selective in nature, have reduced toxicity, and controllable drug release patterns making them ideal carriers for anticancer drugs. Numerous nanocarriers have been designed to combat malignancies, including liposomes, micelles, dendrimers, solid nanoparticles, quantum dots, gold nanoparticles, and, more recently, metal-organic frameworks. The temporal and spatial release of therapeutic agents from these nanostructures can be controlled using internal and external triggers, including pH, enzymes, redox, temperature, magnetic and electromagnetic waves, and ultrasound. Ultrasound is an attractive modality because it is non-invasive, can be focused on the diseased site, and has a synergistic effect with anticancer drugs. CONCLUSION The functionalization of micellar and liposomal surfaces with targeting moieties and the use of ultrasound as a triggering mechanism can help improve the selectivity and enable the spatiotemporal control of drug release from nanocarriers.
Collapse
Affiliation(s)
- Debasmita Mukhopadhyay
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Catherine Sano
- Department of Chemical Engineering, University of Virginia, Charlottesville, VA, United States
| | - Nour AlSawaftah
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Raafat El-Awady
- College of Pharmacy, University of Sharjah, Sharjah, United Arab Emirates
| | - Ghaleb A Husseini
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| | - Vinod Paul
- Department of Chemical Engineering, American University of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
9
|
Liposome delivery to the brain with rapid short-pulses of focused ultrasound and microbubbles. J Control Release 2021; 341:605-615. [PMID: 34896448 DOI: 10.1016/j.jconrel.2021.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 10/13/2021] [Accepted: 12/04/2021] [Indexed: 12/12/2022]
Abstract
Liposomes are clinically used drug carriers designed to improve the delivery of drugs to specific tissues while minimising systemic distribution. However, liposomes are unable to cross the blood-brain barrier (BBB) and enter the brain, mostly due to their large size (ca. 100 nm). A noninvasive and localised method of delivering liposomes across the BBB is to intravenously inject microbubbles and apply long pulses of ultrasound (pulse length: >1 ms) to a targeted brain region. Recently, we have shown that applying rapid short pulses (RaSP) (pulse length: 5 μs) can deliver drugs with an improved efficacy and safety profile. However, this was tested with a relatively smaller 3-kDa molecule (dextran). In this study, we examine whether RaSP can deliver liposomes to the murine brain in vivo. Fluorescent DiD-PEGylated liposomes were synthesized and injected intravenously alongside microbubbles. The left hippocampus of mice was then sonicated with either a RaSP sequence (5 μs at 1.25 kHz in groups of 10 ms at 0.5 Hz) or a long pulse sequence (10 ms at 0.5 Hz), with each pulse having a 1-MHz centre frequency (0.35 and 0.53 MPa). The delivery and distribution of the fluorescently-labelled liposomes were assessed by fluorescence imaging of the brain sections. The safety profile of the sonicated brains was assessed by histological staining. RaSP was shown to locally deliver liposomes across the BBB at 0.53 MPa with a more diffused and safer profile compared to the long pulse ultrasound sequence. Cellular uptake of liposomes was observed in neurons and microglia, while no uptake within astrocytes was observed in both RaSP and long pulse-treated brains. This study shows that RaSP allows a targeted and safe delivery of liposomal drugs into the murine brain with potential to deliver drugs into neuronal and glial targets.
Collapse
|
10
|
The Role of Ultrasound as a Diagnostic and Therapeutic Tool in Experimental Animal Models of Stroke: A Review. Biomedicines 2021; 9:biomedicines9111609. [PMID: 34829837 PMCID: PMC8615437 DOI: 10.3390/biomedicines9111609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 10/25/2021] [Accepted: 11/01/2021] [Indexed: 11/18/2022] Open
Abstract
Ultrasound is a noninvasive technique that provides real-time imaging with excellent resolution, and several studies demonstrated the potential of ultrasound in acute ischemic stroke monitoring. However, only a few studies were performed using animal models, of which many showed ultrasound to be a safe and effective tool also in therapeutic applications. The full potential of ultrasound application in experimental stroke is yet to be explored to further determine the limitations of this technique and to ensure the accuracy of translational research. This review covers the current status of ultrasound applied to monitoring and treatment in experimental animal models of stroke and examines the safety, limitations, and future perspectives.
Collapse
|
11
|
Fatima A, Ahmad MW, Al Saidi AKA, Choudhury A, Chang Y, Lee GH. Recent Advances in Gadolinium Based Contrast Agents for Bioimaging Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2021; 11:2449. [PMID: 34578765 PMCID: PMC8465722 DOI: 10.3390/nano11092449] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/15/2021] [Accepted: 09/17/2021] [Indexed: 12/12/2022]
Abstract
Gadolinium (Gd) based contrast agents (CAs) (Gd-CAs) represent one of the most advanced developments in the application of Gd for magnetic resonance imaging (MRI). Current challenges with existing CAs generated an urgent requirement to develop multimodal CAs with good biocompatibility, low toxicity, and prolonged circulation time. This review discussed the Gd-CAs used in bioimaging applications, addressing their advantages and limitations. Future research is required to establish the safety, efficacy and theragnostic capabilities of Gd-CAs. Nevertheless, these Gd-CAs offer extraordinary potential as imaging CAs and promise to benefit bioimaging applications significantly.
Collapse
Affiliation(s)
- Atiya Fatima
- Department of Chemical Engineering, College of Engineering, Dhofar University, P.O. Box 2509, Salalah 211, Sultanate of Oman;
| | - Md. Wasi Ahmad
- Department of Chemical Engineering, College of Engineering, Dhofar University, P.O. Box 2509, Salalah 211, Sultanate of Oman;
| | - Abdullah Khamis Ali Al Saidi
- Department of Chemistry, College of Natural Sciences, Kyungpook National University (KNU), Taegu 702-701, Korea;
| | - Arup Choudhury
- Department of Chemical Engineering, Birla Institute of Technology, Ranchi 835215, India
| | - Yongmin Chang
- Department of Molecular Medicine and Medical & Biological Engineering, School of Medicine, Kyungpook National University (KNU), Taegu 702-701, Korea;
| | - Gang Ho Lee
- Department of Chemistry, College of Natural Sciences, Kyungpook National University (KNU), Taegu 702-701, Korea;
| |
Collapse
|
12
|
Ultrasound-Enabled Therapeutic Delivery and Regenerative Medicine: Physical and Biological Perspectives. ACS Biomater Sci Eng 2021; 7:4371-4387. [PMID: 34460238 DOI: 10.1021/acsbiomaterials.1c00276] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The role of ultrasound in medicine and biological sciences is expanding rapidly beyond its use in conventional diagnostic imaging. Numerous studies have reported the effects of ultrasound on cellular and tissue physiology. Advances in instrumentation and electronics have enabled successful in vivo applications of therapeutic ultrasound. Despite path breaking advances in understanding the biophysical and biological mechanisms at both microscopic and macroscopic scales, there remain substantial gaps. With the progression of research in this area, it is important to take stock of the current understanding of the field and to highlight important areas for future work. We present herein key developments in the biological applications of ultrasound especially in the context of nanoparticle delivery, drug delivery, and regenerative medicine. We conclude with a brief perspective on the current promise, limitations, and future directions for interfacing ultrasound technology with biological systems, which could provide guidance for future investigations in this interdisciplinary area.
Collapse
|
13
|
Bernal A, Calcagno C, Mulder WJM, Pérez-Medina C. Imaging-guided nanomedicine development. Curr Opin Chem Biol 2021; 63:78-85. [PMID: 33735814 DOI: 10.1016/j.cbpa.2021.01.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Revised: 01/18/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022]
Abstract
Nanomedicine research is an active field that produces thousands of studies every year. However, translation of nanotherapeutics to the clinic has yet to catch up with such a vast output. In recent years, the need to better understand nanomedicines' in vivo behavior has been identified as one of the major challenges for efficient clinical translation. In this context, noninvasive imaging offers attractive solutions to provide valuable information about nanomedicine biodistribution, pharmacokinetics, stability, or therapeutic efficacy. Here, we review the latest imaging approaches used in the development of therapeutic nanomedicines, discuss why these strategies bring added value along the translational pipeline, and give a perspective on future advances in the field.
Collapse
Affiliation(s)
- Aurora Bernal
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain
| | - Claudia Calcagno
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Willem J M Mulder
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA; Department of Chemical Biology, Eindhoven University of Technology, Eindhoven, the Netherlands
| | - Carlos Pérez-Medina
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Madrid, Spain; BioMedical Engineering and Imaging Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
14
|
Recent Advances on Ultrasound Contrast Agents for Blood-Brain Barrier Opening with Focused Ultrasound. Pharmaceutics 2020; 12:pharmaceutics12111125. [PMID: 33233374 PMCID: PMC7700476 DOI: 10.3390/pharmaceutics12111125] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Revised: 11/16/2020] [Accepted: 11/17/2020] [Indexed: 12/14/2022] Open
Abstract
The blood-brain barrier is the primary obstacle to efficient intracerebral drug delivery. Focused ultrasound, in conjunction with microbubbles, is a targeted and non-invasive way to disrupt the blood-brain barrier. Many commercially available ultrasound contrast agents and agents specifically designed for therapeutic purposes have been investigated in ultrasound-mediated blood-brain barrier opening studies. The new generation of sono-sensitive agents, such as liquid-core droplets, can also potentially disrupt the blood-brain barrier after their ultrasound-induced vaporization. In this review, we describe the different compositions of agents used for ultrasound-mediated blood-brain barrier opening in recent studies, and we discuss the challenges of the past five years related to the optimal formulation of agents.
Collapse
|
15
|
Gondim BLC, da Silva Catarino J, de Sousa MAD, de Oliveira Silva M, Lemes MR, de Carvalho-Costa TM, de Lima Nascimento TR, Machado JR, Rodrigues V, Oliveira CJF, Cançado Castellano LR, da Silva MV. Nanoparticle-Mediated Drug Delivery: Blood-Brain Barrier as the Main Obstacle to Treating Infectious Diseases in CNS. Curr Pharm Des 2020; 25:3983-3996. [PMID: 31612822 DOI: 10.2174/1381612825666191014171354] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Accepted: 09/19/2019] [Indexed: 12/25/2022]
Abstract
BACKGROUND Parasitic infections affecting the central nervous system (CNS) present high morbidity and mortality rates and affect millions of people worldwide. The most important parasites affecting the CNS are protozoans (Plasmodium sp., Toxoplasma gondii, Trypanosoma brucei), cestodes (Taenia solium) and free-living amoebae (Acantamoeba spp., Balamuthia mandrillaris and Naegleria fowleri). Current therapeutic regimens include the use of traditional chemicals or natural compounds that have very limited access to the CNS, despite their elevated toxicity to the host. Improvements are needed in drug administration and formulations to treat these infections and to allow the drug to cross the blood-brain barrier (BBB). METHODS This work aims to elucidate the recent advancements in the use of nanoparticles as nanoscaled drug delivery systems (NDDS) for treating and controlling the parasitic infections that affect the CNS, addressing not only the nature and composition of the polymer chosen, but also the mechanisms by which these nanoparticles may cross the BBB and reach the infected tissue. RESULTS There is a strong evidence in the literature demonstrating the potential usefulness of polymeric nanoparticles as functional carriers of drugs to the CNS. Some of them demonstrated the mechanisms by which drugloaded nanoparticles access the CNS and control the infection by using in vivo models, while others only describe the pharmacological ability of these particles to be utilized in in vitro environments. CONCLUSION The scarcity of the studies trying to elucidate the compatibility as well as the exact mechanisms by which NDDS might be entering the CNS infected by parasites reveals new possibilities for further exploratory projects. There is an urgent need for new investments and motivations for applying nanotechnology to control parasitic infectious diseases worldwide.
Collapse
Affiliation(s)
- Brenna Louise Cavalcanti Gondim
- Human Immunology Research and Education Group-GEPIH, Technical School of Health, Federal University of Paraiba, Joao Pessoa, Paraiba, Brazil.,Post-Graduation Program in Dentistry, Department of Dentistry, State University of Paraíba, Campina Grande, Paraíba, Brazil
| | - Jonatas da Silva Catarino
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | - Mariana de Oliveira Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Marcela Rezende Lemes
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba, Minas Gerais, Brazil
| | | | - Tatiana Rita de Lima Nascimento
- Human Immunology Research and Education Group-GEPIH, Technical School of Health, Federal University of Paraiba, Joao Pessoa, Paraiba, Brazil
| | - Juliana Reis Machado
- Department of Pathology, Genetics and Evolution, Federal University of Triangulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Virmondes Rodrigues
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Carlo José Freire Oliveira
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba, Minas Gerais, Brazil
| | - Lúcio Roberto Cançado Castellano
- Human Immunology Research and Education Group-GEPIH, Technical School of Health, Federal University of Paraiba, Joao Pessoa, Paraiba, Brazil
| | - Marcos Vinicius da Silva
- Department of Microbiology, Immunology and Parasitology, Federal University of Triangulo Mineiro, Uberaba, Minas Gerais, Brazil
| |
Collapse
|
16
|
Omata D, Hagiwara F, Munakata L, Shima T, Kageyama S, Suzuki Y, Azuma T, Takagi S, Seki K, Maruyama K, Suzuki R. Characterization of Brain-Targeted Drug Delivery Enhanced by a Combination of Lipid-Based Microbubbles and Non-Focused Ultrasound. J Pharm Sci 2020; 109:2827-2835. [PMID: 32534883 DOI: 10.1016/j.xphs.2020.06.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2020] [Revised: 06/01/2020] [Accepted: 06/04/2020] [Indexed: 12/20/2022]
Abstract
The combination of focused ultrasound (FUS) and microbubbles, an ultrasound (US) contrast agent, has attracted much attention for its ability to open the blood brain barrier (BBB) and deliver drugs to the brain parenchyma. FUS can concentrate US energy in a restricted space, whereas non-focused US can affect a wide area of tissue. Non-focused US is also promising for drug delivery to the brain and other tissues. We have previously developed lipid-based microbubbles (LBs), and demonstrated that non-focused US and LBs have potential for drug delivery to tumor tissues. In this study, to achieve efficient and safe brain-targeted drug delivery, we evaluated the characteristics of BBB opening using non-focused US and LBs. Our results indicated that LBs could induce BBB opening with non-focused US. US frequency and intensity affected the efficiency of BBB opening and brain damage, and showed that the dose of LBs was also related to the efficiency of BBB opening. Furthermore, the combination of non-focused US and LBs could deliver macromolecules at 2000 kDa to the brain, and the induction of BBB opening was found to be reversible. These results suggest that the combination of non-focused US and LBs has potential as a brain-targeted drug delivery system.
Collapse
Affiliation(s)
- Daiki Omata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Fumiko Hagiwara
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Lisa Munakata
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Tadamitsu Shima
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Saori Kageyama
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Yuno Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Takashi Azuma
- Department of Mechanical Engineering, The University of Tokyo, Tokyo, Japan
| | - Shu Takagi
- Department of Mechanical Engineering, The University of Tokyo, Tokyo, Japan
| | - Kazuhiko Seki
- Department of Neurophysiology, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Tokyo, Japan
| | - Kazuo Maruyama
- Laboratory of Theranostics, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan
| | - Ryo Suzuki
- Laboratory of Drug and Gene Delivery Research, Faculty of Pharma-Science, Teikyo University, Tokyo, Japan.
| |
Collapse
|
17
|
Abstract
Iron oxide nanoparticles have been extensively utilised as negative (T2) contrast agents in magnetic resonance imaging. In the past few years, researchers have also exploited their application as positive (T1) contrast agents to overcome the limitation of traditional Gd3+ contrast agents. To provide T1 contrast, these particles must present certain physicochemical properties with control over the size, morphology and surface of the particles. In this review, we summarise the reported T1 iron oxide nanoparticles and critically revise their properties, synthetic protocols and application, not only in MRI but also in multimodal imaging. In addition, we briefly summarise the most important nanoparticulate Gd and Mn agents to evaluate whether T1 iron oxide nanoparticles can reach Gd/Mn contrast capabilities.
Collapse
|
18
|
Morse SV, Boltersdorf T, Harriss BI, Chan TG, Baxan N, Jung HS, Pouliopoulos AN, Choi JJ, Long NJ. Neuron labeling with rhodamine-conjugated Gd-based MRI contrast agents delivered to the brain via focused ultrasound. Am J Cancer Res 2020; 10:2659-2674. [PMID: 32194827 PMCID: PMC7052893 DOI: 10.7150/thno.42665] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/01/2020] [Indexed: 12/14/2022] Open
Abstract
Gadolinium-based magnetic resonance imaging contrast agents can provide information regarding neuronal function, provided that these agents can cross the neuronal cell membrane. Such contrast agents are normally restricted to extracellular domains, however, by attaching cationic fluorescent dyes, they can be made cell-permeable and allow for both optical and magnetic resonance detection. To reach neurons, these agents also need to cross the blood-brain barrier. Focused ultrasound combined with microbubbles has been shown to enhance the permeability of this barrier, allowing molecules into the brain non-invasively, locally and transiently. The goal of this study was to investigate whether combining fluorescent rhodamine with a gadolinium complex would form a dual-modal contrast agent that could label neurons in vivo when delivered to the mouse brain with focused ultrasound and microbubbles. Methods: Gadolinium complexes were combined with a fluorescent, cationic rhodamine unit to form probes with fluorescence and relaxivity properties suitable for in vivo applications. The left hemisphere of female C57bl/6 mice (8-10 weeks old; 19.07 ± 1.56 g; n = 16) was treated with ultrasound (centre frequency: 1 MHz, peak-negative pressure: 0.35 MPa, pulse length: 10 ms, repetition frequency: 0.5 Hz) while intravenously injecting SonoVue microbubbles and either the 1 kDa Gd(rhodamine-pip-DO3A) complex or a conventionally-used lysine-fixable Texas Red® 3 kDa dextran. The opposite right hemisphere was used as a non-treated control region. Brains were then extracted and either sectioned and imaged via fluorescence or confocal microscopy or imaged using a 9.4 T magnetic resonance imaging scanner. Brain slices were stained for neurons (NeuN), microglia (Iba1) and astrocytes (GFAP) to investigate the cellular localization of the probes. Results: Rhodamine fluorescence was detected in the left hemisphere of all ultrasound treated mice, while none was detected in the right control hemisphere. Cellular uptake of Gd(rhodamine-pip-DO3A) was observed in all the treated regions with a uniform distribution (coefficient of variation = 0.4 ± 0.05). Uptake was confirmed within neurons, whereas the probe did not co-localize with microglia and astrocytes. Compared to the dextran molecule, Gd(rhodamine-pip-DO3A) distributed more homogeneously and was less concentrated around blood vessels. Furthermore, the dextran molecule was found to accumulate unselectively in microglia as well as neurons, whereas our probe was only taken up by neurons. Gd(rhodamine-pip-DO3A) was detected via magnetic resonance imaging ex vivo in similar regions to where fluorescence was detected. Conclusion: We have introduced a method to image neurons with a dual-modal imaging agent delivered non-invasively and locally to the brain using focused ultrasound and microbubbles. When delivered to the mouse brain, the agent distributed homogeneously and was only uptaken by neurons; in contrast, conventionally used dextran distributed heterogeneously and was uptaken by microglia as well as neurons. This result indicates that our probe labels neurons without microglial involvement and in addition the probe was found to be detectable via both ex vivo MRI and fluorescence. Labeling neurons with such dual-modal agents could facilitate the study of neuronal morphology and physiology using the advantages of both imaging modalities.
Collapse
|