1
|
Snipstad S, Sulheim E, Åslund AKO, Hyldbakk A, Wågbø AM, Klinkenberg G, Mørch Y. Nanoparticle-loaded microbubbles for treatment of lung cancer. Eur J Pharm Sci 2024; 199:106804. [PMID: 38763448 DOI: 10.1016/j.ejps.2024.106804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 03/20/2024] [Accepted: 05/16/2024] [Indexed: 05/21/2024]
Abstract
Lung cancer is one of the most common cancers and a leading cause of death, with poor prognosis and high unmet clinical need. Chemotherapy is a common part of the treatment, either alone or in combination with other treatment modalities, but with limited efficacy and severe side effects. Encapsulation of drugs into nanoparticles can enable a more targeted delivery with reduced off-target toxicity. Delivery to the lungs is however often insufficient due to various biological barriers in the body and in the tumor microenvironment. Here we demonstrate that by incorporating drug-loaded nanoparticles into air-filled microbubbles, a more effective targeting to the lungs can be achieved. Fluorescence imaging and mass spectrometry revealed that the microbubbles could significantly improve accumulation of drug in the lungs of mice, compared to injecting either the free drug by itself or only the drug-loaded nanoparticles. Therapeutic efficacy was verified in a preclinical mouse model with non-small cell lung cancer, monitoring tumor growth by luminescence.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olavs Hospital, Trondheim, Norway.
| | - Einar Sulheim
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Institute for Research in Biomedicine, Bellinzona, Switzerland
| | - Andreas K O Åslund
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ane Marit Wågbø
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Geir Klinkenberg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; NaDeNo Nanoscience AS, Trondheim, Norway
| |
Collapse
|
2
|
Nawijn CL, Segers T, Lajoinie G, Berg S, Snipstad S, Davies CDL, Versluis M. High-Speed Optical Characterization of Protein-and-Nanoparticle-Stabilized Microbubbles for Ultrasound-Triggered Drug Release. ULTRASOUND IN MEDICINE & BIOLOGY 2024; 50:1099-1107. [PMID: 38851940 DOI: 10.1016/j.ultrasmedbio.2024.03.011] [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: 01/03/2024] [Revised: 03/11/2024] [Accepted: 03/21/2024] [Indexed: 06/10/2024]
Abstract
OBJECTIVE Ultrasound-triggered bubble-mediated local drug delivery has shown potential to increase therapeutic efficacy and reduce systemic side effects, by loading drugs into the microbubble shell and triggering delivery of the payload on demand using ultrasound. Understanding the behavior of the microbubbles in response to ultrasound is crucial for efficient and controlled release. METHODS In this work, the response of microbubbles with a coating consisting of poly(2-ethyl-butyl cyanoacrylate) (PEBCA) nanoparticles and denatured casein was characterized. High-speed recordings were taken of single microbubbles, in both bright field and fluorescence. RESULTS The nanoparticle-loaded microbubbles show resonance behavior, but with a large variation in response, revealing a substantial interbubble variation in mechanical shell properties. The probability of shell rupture and the probability of nanoparticle release were found to strongly depend on microbubble size, and the most effective size was inversely proportional to the driving frequency. The probabilities of both rupture and release increased with increasing driving pressure amplitude. Rupture of the microbubble shell occurred after fewer cycles of ultrasound as the driving pressure amplitude or driving frequency was increased. CONCLUSION The results highlight the importance of careful selection of the driving frequency, driving pressure amplitude and duration of ultrasound to achieve the most efficient ultrasound-triggered shell rupture and nanoparticle release of protein-and-nanoparticle-stabilized microbubbles.
Collapse
Affiliation(s)
- Charlotte L Nawijn
- Physics of Fluids Group, Technical Medical (TechMed) Center, University of Twente, Enschede, The Netherlands.
| | - Tim Segers
- BIOS/Lab on a Chip Group, Max Planck Center Twente for Complex Fluid Dynamics, MESA+ Institute for Nanotechnology, University of Twente, Enschede, The Netherlands
| | - Guillaume Lajoinie
- Physics of Fluids Group, Technical Medical (TechMed) Center, University of Twente, Enschede, The Netherlands
| | - Sigrid Berg
- Department of Health Research, SINTEF Digital, Trondheim, Norway
| | - Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | | | - Michel Versluis
- Physics of Fluids Group, Technical Medical (TechMed) Center, University of Twente, Enschede, The Netherlands
| |
Collapse
|
3
|
Nistor M, Rugina D, Diaconeasa Z, Socaciu C, Socaciu MA. Pentacyclic Triterpenoid Phytochemicals with Anticancer Activity: Updated Studies on Mechanisms and Targeted Delivery. Int J Mol Sci 2023; 24:12923. [PMID: 37629103 PMCID: PMC10455110 DOI: 10.3390/ijms241612923] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 08/27/2023] Open
Abstract
Pentacyclic triterpenoids (TTs) represent a unique family of phytochemicals with interesting properties and pharmacological effects, with some representatives, such as betulinic acid (BA) and betulin (B), being mainly investigated as potential anticancer molecules. Considering the recent scientific and preclinical investigations, a review of their anticancer mechanisms, structure-related activity, and efficiency improved by their insertion in nanolipid vehicles for targeted delivery is presented. A systematic literature study about their effects on tumor cells in vitro and in vivo, as free molecules or encapsulated in liposomes or nanolipids, is discussed. A special approach is given to liposome-TTs and nanolipid-TTs complexes to be linked to microbubbles, known as contrast agents in ultrasonography. The production of such supramolecular conjugates to deliver the drugs to target cells via sonoporation represents a new scientific and applicative direction to improve TT efficiency, considering that they have limited availability as lipophilic molecules. Relevant and recent examples of in vitro and in vivo studies, as well as the challenges for the next steps towards the application of these complex delivery systems to tumor cells, are discussed, as are the challenges for the next steps towards the application of targeted delivery to tumor cells, opening new directions for innovative nanotechnological solutions.
Collapse
Affiliation(s)
- Madalina Nistor
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Dumitrita Rugina
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Zorita Diaconeasa
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Carmen Socaciu
- Department of Biochemistry, University of Agricultural Sciences and Veterinary Medicine, 400372 Cluj-Napoca, Romania; (M.N.); (D.R.); (Z.D.)
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
| | - Mihai Adrian Socaciu
- Department of Biotechnology, BIODIATECH—Research Centre for Applied Biotechnology in Diagnosis and Molecular Therapy, 400478 Cluj-Napoca, Romania
- Department of Radiology, Imaging & Nuclear Medicine, Faculty of Medicine, University of Medicine and Pharmacy “Iuliu Hatieganu”, 400347 Cluj-Napoca, Romania
| |
Collapse
|
4
|
Edwards IA, De Carlo F, Sitta J, Varner W, Howard CM, Claudio PP. Enhancing Targeted Therapy in Breast Cancer by Ultrasound-Responsive Nanocarriers. Int J Mol Sci 2023; 24:ijms24065474. [PMID: 36982548 PMCID: PMC10053544 DOI: 10.3390/ijms24065474] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 03/04/2023] [Accepted: 03/08/2023] [Indexed: 03/17/2023] Open
Abstract
Currently, the response to cancer treatments is highly variable, and severe side effects and toxicity are experienced by patients receiving high doses of chemotherapy, such as those diagnosed with triple-negative breast cancer. The main goal of researchers and clinicians is to develop new effective treatments that will be able to specifically target and kill tumor cells by employing the minimum doses of drugs exerting a therapeutic effect. Despite the development of new formulations that overall can increase the drugs’ pharmacokinetics, and that are specifically designed to bind overexpressed molecules on cancer cells and achieve active targeting of the tumor, the desired clinical outcome has not been reached yet. In this review, we will discuss the current classification and standard of care for breast cancer, the application of nanomedicine, and ultrasound-responsive biocompatible carriers (micro/nanobubbles, liposomes, micelles, polymeric nanoparticles, and nanodroplets/nanoemulsions) employed in preclinical studies to target and enhance the delivery of drugs and genes to breast cancer.
Collapse
Affiliation(s)
- Isaiah A. Edwards
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Flavia De Carlo
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Juliana Sitta
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - William Varner
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Candace M. Howard
- Department of Radiology, University of Mississippi Medical Center, Jackson, MS 39216, USA
| | - Pier Paolo Claudio
- Department of Pharmacology and Toxicology, Cancer Center and Research Institute, University of Mississippi Medical Center, Jackson, MS 39216, USA
- Correspondence:
| |
Collapse
|
5
|
Moradi Kashkooli F, Jakhmola A, Hornsby TK, Tavakkoli JJ, Kolios MC. Ultrasound-mediated nano drug delivery for treating cancer: Fundamental physics to future directions. J Control Release 2023; 355:552-578. [PMID: 36773959 DOI: 10.1016/j.jconrel.2023.02.009] [Citation(s) in RCA: 32] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 02/03/2023] [Accepted: 02/06/2023] [Indexed: 02/13/2023]
Abstract
The application of biocompatible nanocarriers in medicine has provided several benefits over conventional treatment methods. However, achieving high treatment efficacy and deep penetration of nanocarriers in tumor tissue is still challenging. To address this, stimuli-responsive nano-sized drug delivery systems (DDSs) are an active area of investigation in delivering anticancer drugs. While ultrasound is mainly used for diagnostic purposes, it can also be applied to affect cellular function and the delivery/release of anticancer drugs. Therapeutic ultrasound (TUS) has shown potential as both a stand-alone anticancer treatment and a method to induce targeted drug release from nanocarrier systems. TUS approaches have been used to overcome various physiological obstacles, including endothelial barriers, the tumor microenvironment (TME), and immunological hurdles. Combining nanomedicine and ultrasound as a smart DDS can increase in situ drug delivery and improve access to impermeable tissues. Furthermore, smart DDSs can perform targeted drug release in response to distinctive TMEs, external triggers, or dual/multi-stimulus. This results in enhanced treatment efficacy and reduced damage to surrounding healthy tissue or organs at risk. Integrating DDSs and ultrasound is still in its early stages. More research and clinical trials are required to fully understand ultrasound's underlying physical mechanisms and interactions with various types of nanocarriers and different types of cells and tissues. In the present review, ultrasound-mediated nano-sized DDS, specifically focused on cancer treatment, is presented and discussed. Ultrasound interaction with nanoparticles (NPs), drug release mechanisms, and various types of ultrasound-sensitive NPs are examined. Additionally, in vitro, in vivo, and clinical applications of TUS are reviewed in light of the critical challenges that need to be considered to advance TUS toward an efficient, secure, straightforward, and accessible cancer treatment. This study also presents effective TUS parameters and safety considerations for this treatment modality and gives recommendations about system design and operation. Finally, future perspectives are considered, and different TUS approaches are examined and discussed in detail. This review investigates drug release and delivery through ultrasound-mediated nano-sized cancer treatment, both pre-clinically and clinically.
Collapse
Affiliation(s)
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Jahangir Jahan Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, Ontario, Canada; Institute for Biomedical Engineering, Science and Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, Ontario, Canada.
| |
Collapse
|
6
|
Fleten KG, Hyldbakk A, Einen C, Benjakul S, Strand BL, Davies CDL, Mørch Ý, Flatmark K. Alginate Microsphere Encapsulation of Drug-Loaded Nanoparticles: A Novel Strategy for Intraperitoneal Drug Delivery. Mar Drugs 2022; 20:744. [PMID: 36547891 PMCID: PMC9782800 DOI: 10.3390/md20120744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Alginate hydrogels have been broadly investigated for use in medical applications due to their biocompatibility and the possibility to encapsulate cells, proteins, and drugs. In the treatment of peritoneal metastasis, rapid drug clearance from the peritoneal cavity is a major challenge. Aiming to delay drug absorption and reduce toxic side effects, cabazitaxel (CAB)-loaded poly(alkyl cyanoacrylate) (PACA) nanoparticles were encapsulated in alginate microspheres. The PACAlg alginate microspheres were synthesized by electrostatic droplet generation and the physicochemical properties, stability, drug release kinetics, and mesothelial cytotoxicity were analyzed before biodistribution and therapeutic efficacy were studied in mice. The 450 µm microspheres were stable at in vivo conditions for at least 21 days after intraperitoneal implantation in mice, and distributed evenly throughout the peritoneal cavity without aggregation or adhesion. The nanoparticles were stably retained in the alginate microspheres, and nanoparticle toxicity to mesothelial cells was reduced, while the therapeutic efficacy of free CAB was maintained or improved in vivo. Altogether, this work presents the alginate encapsulation of drug-loaded nanoparticles as a promising novel strategy for the treatment of peritoneal metastasis that can improve the therapeutic ratio between toxicity and therapeutic efficacy.
Collapse
Affiliation(s)
- Karianne Giller Fleten
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
| | - Astrid Hyldbakk
- Department of Biotechnology and Nanomedicine, SINTEF Industry, 7034 Trondheim, Norway
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Caroline Einen
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Sopisa Benjakul
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Berit Løkensgard Strand
- Department of Biotechnology and Food Science, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Catharina de Lange Davies
- Department of Physics, Norwegian University of Science and Technology (NTNU), 7034 Trondheim, Norway
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, 7034 Trondheim, Norway
| | - Kjersti Flatmark
- Department of Tumor Biology, Institute for Cancer Research, Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
- Faculty of Medicine, Institute of Clinical Medicine, University of Oslo, 0372 Oslo, Norway
- Department of Gastroenterological Surgery, The Norwegian Radium Hospital, Oslo University Hospital, 0379 Oslo, Norway
| |
Collapse
|
7
|
Chapla R, Huynh KT, Schutt CE. Microbubble–Nanoparticle Complexes for Ultrasound-Enhanced Cargo Delivery. Pharmaceutics 2022; 14:pharmaceutics14112396. [PMID: 36365214 PMCID: PMC9698658 DOI: 10.3390/pharmaceutics14112396] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 10/05/2022] [Accepted: 10/08/2022] [Indexed: 11/09/2022] Open
Abstract
Targeted delivery of therapeutics to specific tissues is critically important for reducing systemic toxicity and optimizing therapeutic efficacy, especially in the case of cytotoxic drugs. Many strategies currently exist for targeting systemically administered drugs, and ultrasound-controlled targeting is a rapidly advancing strategy for externally-stimulated drug delivery. In this non-invasive method, ultrasound waves penetrate through tissue and stimulate gas-filled microbubbles, resulting in bubble rupture and biophysical effects that power delivery of attached cargo to surrounding cells. Drug delivery capabilities from ultrasound-sensitive microbubbles are greatly expanded when nanocarrier particles are attached to the bubble surface, and cargo loading is determined by the physicochemical properties of the nanoparticles. This review serves to highlight and discuss current microbubble–nanoparticle complex component materials and designs for ultrasound-mediated drug delivery. Nanocarriers that have been complexed with microbubbles for drug delivery include lipid-based, polymeric, lipid–polymer hybrid, protein, and inorganic nanoparticles. Several schemes exist for linking nanoparticles to microbubbles for efficient nanoparticle delivery, including biotin–avidin bridging, electrostatic bonding, and covalent linkages. When compared to unstimulated delivery, ultrasound-mediated cargo delivery enables enhanced cell uptake and accumulation of cargo in target organs and can result in improved therapeutic outcomes. These ultrasound-responsive delivery complexes can also be designed to facilitate other methods of targeting, including bioactive targeting ligands and responsivity to light or magnetic fields, and multi-level targeting can enhance therapeutic efficacy. Microbubble–nanoparticle complexes present a versatile platform for controlled drug delivery via ultrasound, allowing for enhanced tissue penetration and minimally invasive therapy. Future perspectives for application of this platform are also discussed in this review.
Collapse
Affiliation(s)
- Rachel Chapla
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
| | - Katherine T. Huynh
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
| | - Carolyn E. Schutt
- Cancer Early Detection Advanced Research Center, Oregon Health and Science University, Portland, OR 97201, USA
- Department of Biomedical Engineering, Oregon Health and Science University, Portland, OR 97239, USA
- Correspondence:
| |
Collapse
|
8
|
Vidallon MLP, Teo BM, Bishop AI, Tabor RF. Next-Generation Colloidal Materials for Ultrasound Imaging Applications. ULTRASOUND IN MEDICINE & BIOLOGY 2022; 48:1373-1396. [PMID: 35641393 DOI: 10.1016/j.ultrasmedbio.2022.04.001] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 03/31/2022] [Accepted: 04/03/2022] [Indexed: 06/15/2023]
Abstract
Ultrasound has important applications, predominantly in the field of diagnostic imaging. Presently, colloidal systems such as microbubbles, phase-change emulsion droplets and particle systems with acoustic properties and multiresponsiveness are being developed to address typical issues faced when using commercial ultrasound contrast agents, and to extend the utility of such systems to targeted drug delivery and multimodal imaging. Current technologies and increasing research data on the chemistry, physics and materials science of new colloidal systems are also leading to the development of more complex, novel and application-specific colloidal assemblies with ultrasound contrast enhancement and other properties, which could be beneficial for multiple biomedical applications, especially imaging-guided treatments. In this article, we review recent developments in new colloids with applications that use ultrasound contrast enhancement. This work also highlights the emergence of colloidal materials fabricated from or modified with biologically derived and bio-inspired materials, particularly in the form of biopolymers and biomembranes. Challenges, limitations, potential developments and future directions of these next-generation colloidal systems are also presented and discussed.
Collapse
Affiliation(s)
| | - Boon Mian Teo
- School of Chemistry, Monash University, Clayton, Victoria, Australia
| | - Alexis I Bishop
- School of Physics and Astronomy, Monash University, Clayton, Victoria, Australia
| | - Rico F Tabor
- School of Chemistry, Monash University, Clayton, Victoria, Australia.
| |
Collapse
|
9
|
Perra E, Hayward N, Pritzker KPH, Nieminen HJ. An ultrasonically actuated needle promotes the transport of nanoparticles and fluids. THE JOURNAL OF THE ACOUSTICAL SOCIETY OF AMERICA 2022; 152:251. [PMID: 35931509 DOI: 10.1121/10.0012190] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Accepted: 06/13/2022] [Indexed: 06/15/2023]
Abstract
Non-invasive therapeutic ultrasound (US) methods, such as high-intensity focused ultrasound (HIFU), have limited access to tissue targets shadowed by bones or presence of gas. This study demonstrates that an ultrasonically actuated medical needle can be used to translate nanoparticles and fluids under the action of nonlinear phenomena, potentially overcoming some limitations of HIFU. A simulation study was first conducted to study the delivery of a tracer with an ultrasonically actuated needle (33 kHz) inside a porous medium acting as a model for soft tissue. The model was then validated experimentally in different concentrations of agarose gel showing a close match with the experimental results, when diluted soot nanoparticles (diameter < 150 nm) were employed as delivered entity. An additional simulation study demonstrated a threefold increase in the volume covered by the delivered agent in liver under a constant injection rate, when compared to without US. This method, if developed to its full potential, could serve as a cost effective way to improve safety and efficacy of drug therapies by maximizing the concentration of delivered entities within, e.g., a small lesion, while minimizing exposure outside the lesion.
Collapse
Affiliation(s)
- Emanuele Perra
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, 02150, Finland
| | - Nick Hayward
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, 02150, Finland
| | - Kenneth P H Pritzker
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, M5S 1A8, Canada
| | - Heikki J Nieminen
- Medical Ultrasonics Laboratory (MEDUSA), Department of Neuroscience and Biomedical Engineering, Aalto University, Espoo, 02150, Finland
| |
Collapse
|
10
|
Kotopoulis S, Lam C, Haugse R, Snipstad S, Murvold E, Jouleh T, Berg S, Hansen R, Popa M, Mc Cormack E, Gilja OH, Poortinga A. Formulation and characterisation of drug-loaded antibubbles for image-guided and ultrasound-triggered drug delivery. ULTRASONICS SONOCHEMISTRY 2022; 85:105986. [PMID: 35358937 PMCID: PMC8967728 DOI: 10.1016/j.ultsonch.2022.105986] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/14/2022] [Accepted: 03/21/2022] [Indexed: 06/13/2023]
Abstract
The aim of this study was to develop high load-capacity antibubbles that can be visualized using diagnostic ultrasound and the encapsulated drug can be released and delivered using clinically translatable ultrasound. The antibubbles were developed by optimising a silica nanoparticle stabilised double emulsion template. We produced an emulsion with a mean size diameter of 4.23 ± 1.63 µm where 38.9 ± 3.1% of the droplets contained a one or more cores. Following conversion to antibubbles, the mean size decreased to 2.96 ± 1.94 µm where 99% of antibubbles were <10 µm. The antibubbles had a peak attenuation of 4.8 dB/cm at 3.0 MHz at a concentration of 200 × 103 particles/mL and showed distinct attenuation spikes at frequencies between 5.5 and 13.5 MHz. No increase in subharmonic response was observed for the antibubbles in contrast to SonoVue®. High-speed imaging revealed that antibubbles can release their cores at MIs of 0.6. In vivo imaging indicated that the antibubbles have a long half-life of 68.49 s vs. 40.02 s for SonoVue®. The antibubbles could be visualised using diagnostic ultrasound and could be disrupted at MIs of ≥0.6. The in vitro drug delivery results showed that antibubbles can significantly improve drug delivery (p < 0.0001) and deliver the drug within the antibubbles. In conclusion antibubbles are a viable concept for ultrasound guided drug delivery.
Collapse
Affiliation(s)
- Spiros Kotopoulis
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Neoety AS, Kløfta, Norway.
| | - Christina Lam
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Ragnhild Haugse
- Department of Clinical Science, University of Bergen, Bergen, Norway; Department of Quality and Development, Hospital Pharmacies Enterprise in Western Norway, Bergen, Norway
| | - 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
| | - Elisa Murvold
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway; KinN Therapeutics, Bergen, Norway
| | - Tæraneh Jouleh
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Sigrid Berg
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Rune Hansen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway
| | - Mihaela Popa
- Department of Clinical Science, University of Bergen, Bergen, Norway; CCBIO, Department of Clinical Science, University of Bergen, Norway
| | - Emmet Mc Cormack
- Department of Clinical Science, University of Bergen, Bergen, Norway; KinN Therapeutics, Bergen, Norway
| | - Odd Helge Gilja
- Department of Clinical Medicine, University of Bergen, Bergen, Norway; National Centre for Ultrasound in Gastroenterology, Haukeland University Hospital, Bergen, Norway
| | - Albert Poortinga
- Polymer Technology, Eindhoven University of Technology, Eindhoven, the Netherlands
| |
Collapse
|
11
|
Li X, Montague EC, Pollinzi A, Lofts A, Hoare T. Design of Smart Size-, Surface-, and Shape-Switching Nanoparticles to Improve Therapeutic Efficacy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104632. [PMID: 34936204 DOI: 10.1002/smll.202104632] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Revised: 11/04/2021] [Indexed: 05/21/2023]
Abstract
Multiple biological barriers must be considered in the design of nanomedicines, including prolonged blood circulation, efficient accumulation at the target site, effective penetration into the target tissue, selective uptake of the nanoparticles into target cells, and successful endosomal escape. However, different particle sizes, surface chemistries, and sometimes shapes are required to achieve the desired transport properties at each step of the delivery process. In response, this review highlights recent developments in the design of switchable nanoparticles whose size, surface chemistry, shape, or a combination thereof can be altered as a function of time, a disease-specific microenvironment, and/or via an externally applied stimulus to enable improved optimization of nanoparticle properties in each step of the delivery process. The practical use of such nanoparticles in chemotherapy, bioimaging, photothermal therapy, and other applications is also discussed.
Collapse
Affiliation(s)
- Xiaoyun Li
- Department of Chemical Engineering, McMaster University, 1280 Main Street, Hamilton, ON L8S 4L8, Canada
- State Key Laboratory of Pulp & Paper Engineering, South China University of Technology, 381 Wushan Road, Guangzhou, Guangdong, 510640, China
| | - E Coulter Montague
- Department of Chemical Engineering, McMaster University, 1280 Main Street, Hamilton, ON L8S 4L8, Canada
| | - Angela Pollinzi
- Department of Chemical Engineering, McMaster University, 1280 Main Street, Hamilton, ON L8S 4L8, Canada
| | - Andrew Lofts
- School of Biomedical Engineering, McMaster University, 1280 Main Street, Hamilton, ON L8S 4L8, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, 1280 Main Street, Hamilton, ON L8S 4L8, Canada
- School of Biomedical Engineering, McMaster University, 1280 Main Street, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
12
|
Sønstevold T, Engedal N, Torgersen ML. Perturbation of Cellular Redox Homeostasis Dictates Divergent Effects of Polybutyl Cyanoacrylate (PBCA) Nanoparticles on Autophagy. Cells 2021; 10:3432. [PMID: 34943939 PMCID: PMC8699722 DOI: 10.3390/cells10123432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2021] [Revised: 11/20/2021] [Accepted: 12/03/2021] [Indexed: 01/18/2023] Open
Abstract
Nanoparticles (NPs) are used in our everyday life, including as drug delivery vehicles. However, the effects of NPs at the cellular level and their impacts on autophagy are poorly understood. Here, we demonstrate that the NP drug delivery vehicle poly(butyl cyanoacrylate) (PBCA) perturbs redox homeostasis in human epithelial cells, and that the degree of redox perturbation dictates divergent effects of PBCA on autophagy. Specifically, PBCA promoted functional autophagy at low concentrations, whereas it inhibited autophagy at high concentrations. Both effects were completely abolished by the antioxidant N-acetyl cysteine (NAC). High concentrations of PBCA inhibited MAP1LC3B/GABARAP lipidation and LC3 flux, and blocked bulk autophagic cargo flux induced by mTOR inhibition. These effects were mimicked by the redox regulator H2O2. In contrast, low concentrations of PBCA enhanced bulk autophagic cargo flux in a Vps34-, ULK1/2- and ATG13-dependent manner, yet interestingly, without an accompanying increase in LC3 lipidation or flux. PBCA activated MAP kinase signaling cascades in a redox-dependent manner, and interference with individual signaling components revealed that the autophagy-stimulating effect of PBCA required the action of the JNK and p38-MK2 pathways, whose activities converged on the pro-autophagic protein Beclin-1. Collectively, our results reveal that PBCA exerts a dual effect on autophagy depending on the severity of the NP insult and the resulting perturbation of redox homeostasis. Such a dual autophagy-modifying effect may be of general relevance for redox-perturbing NPs and have important implications in nanomedicine.
Collapse
Affiliation(s)
- Tonje Sønstevold
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway;
| | - Nikolai Engedal
- Department of Tumor Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway;
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Montebello, N-0379 Oslo, Norway;
| |
Collapse
|
13
|
Walsh AP, Gordon HN, Peter K, Wang X. Ultrasonic particles: An approach for targeted gene delivery. Adv Drug Deliv Rev 2021; 179:113998. [PMID: 34662671 PMCID: PMC8518240 DOI: 10.1016/j.addr.2021.113998] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 09/24/2021] [Accepted: 10/05/2021] [Indexed: 02/07/2023]
Abstract
Gene therapy has been widely investigated for the treatment of genetic, acquired, and infectious diseases. Pioneering work utilized viral vectors; however, these are suspected of causing serious adverse events, resulting in the termination of several clinical trials. Non-viral vectors, such as lipid nanoparticles, have attracted significant interest, mainly due to their successful use in vaccines in the current COVID-19 pandemic. Although they allow safe delivery, they come with the disadvantage of off-target delivery. The application of ultrasound to ultrasound-sensitive particles allows for a direct, site-specific transfer of genetic materials into the organ/site of interest. This process, termed ultrasound-targeted gene delivery (UTGD), also increases cell membrane permeability and enhances gene uptake. This review focuses on the advances in ultrasound and the development of ultrasonic particles for UTGD across a range of diseases. Furthermore, we discuss the limitations and future perspectives of UTGD.
Collapse
Affiliation(s)
- Aidan P.G. Walsh
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia
| | - Henry N. Gordon
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Biochemistry and Pharmacology, University of Melbourne, VIC, Australia
| | - Karlheinz Peter
- Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia
| | - Xiaowei Wang
- Molecular Imaging and Theranostics Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Atherothrombosis and Vascular Biology Laboratory, Baker Heart and Diabetes Institute, Melbourne, VIC, Australia,Department of Medicine, Monash University, Melbourne, VIC, Australia,Department of Cardiometabolic Health, University of Melbourne, VIC, Australia,La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC, Australia,Corresponding author at: Baker Heart and Diabetes Institute, 75 Commercial Road, Melbourne, VIC 3004, Australia
| |
Collapse
|
14
|
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
|
15
|
Sulheim E, Hanson I, Snipstad S, Vikedal K, Mørch Y, Boucher Y, Davies CDL. Sonopermeation with Nanoparticle‐Stabilized Microbubbles Reduces Solid Stress and Improves Nanomedicine Delivery to Tumors. ADVANCED THERAPEUTICS 2021. [DOI: 10.1002/adtp.202100147] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Einar Sulheim
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
- Cancer Clinic St.Olavs Hospital Trondheim 7030 Norway
| | - Ingunn Hanson
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| | - Sofie Snipstad
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
- Cancer Clinic St.Olavs Hospital Trondheim 7030 Norway
| | - Krister Vikedal
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine SINTEF AS Trondheim 7034 Norway
| | - Yves Boucher
- Edwin L. Steele Laboratory for Tumor Biology Massachusetts General Hospital Boston MA 02114 USA
| | - Catharina de Lange Davies
- Department of Physics Norwegian University of Science and Technology (NTNU) Trondheim NO‐7491 Norway
| |
Collapse
|
16
|
Drug Delivery by Ultrasound-Responsive Nanocarriers for Cancer Treatment. Pharmaceutics 2021; 13:pharmaceutics13081135. [PMID: 34452096 PMCID: PMC8397943 DOI: 10.3390/pharmaceutics13081135] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/28/2021] [Accepted: 06/29/2021] [Indexed: 12/13/2022] Open
Abstract
Conventional cancer chemotherapies often exhibit insufficient therapeutic outcomes and dose-limiting toxicity. Therefore, there is a need for novel therapeutics and formulations with higher efficacy, improved safety, and more favorable toxicological profiles. This has promoted the development of nanomedicines, including systems for drug delivery, but also for imaging and diagnostics. Nanoparticles loaded with drugs can be designed to overcome several biological barriers to improving efficiency and reducing toxicity. In addition, stimuli-responsive nanocarriers are able to release their payload on demand at the tumor tissue site, preventing premature drug loss. This review focuses on ultrasound-triggered drug delivery by nanocarriers as a versatile, cost-efficient, non-invasive technique for improving tissue specificity and tissue penetration, and for achieving high drug concentrations at their intended site of action. It highlights aspects relevant for ultrasound-mediated drug delivery, including ultrasound parameters and resulting biological effects. Then, concepts in ultrasound-mediated drug delivery are introduced and a comprehensive overview of several types of nanoparticles used for this purpose is given. This includes an in-depth compilation of the literature on the various in vivo ultrasound-responsive drug delivery systems. Finally, toxicological and safety considerations regarding ultrasound-mediated drug delivery with nanocarriers are discussed.
Collapse
|
17
|
Poon K, Lu Z, De Deene Y, Ramaswamy Y, Zreiqat H, Singh G. Tuneable manganese oxide nanoparticle based theranostic agents for potential diagnosis and drug delivery. NANOSCALE ADVANCES 2021; 3:4052-4061. [PMID: 36132835 PMCID: PMC9419237 DOI: 10.1039/d0na00991a] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 06/04/2021] [Indexed: 06/16/2023]
Abstract
Among various magnetic nanoparticles, manganese oxide nanoparticles are considered as established T 1 magnetic resonance imaging (MRI) contrast agents for preclinical research. The implications of their degradation properties and use as therapeutic carriers in drug delivery systems have not been explored. In addition, how the chemical composition and size of manganese oxide nanoparticles, as well as the surrounding environment, influence their degradation and MRI contrast properties (T 1 vs. T 2) have not been studied in great detail. A fundamental understanding of their characteristic properties, such as degradation, is highly desirable for developing simultaneous diagnosis and therapeutic solutions. Here, we demonstrate how the precursor type and reaction environment affect the size and chemical composition of manganese oxide nanoparticles and evaluate their influence on the nanoparticle degradability and release of the drug l-3,4-dihydroxyphenylalanine (l-dopa). The results show that the degradation rate (and the associated release of drug l-dopa molecules) of manganese oxide nanoparticles depends on their size, composition and the surrounding environment (aqueous or biometric fluid). The dependence of MRI relaxivities of manganese oxide nanoparticles on the size, chemical composition and nanoparticle degradation in water is also established. A preliminary cell viability study reveals the cytocompatible properties of l-dopa functionalized manganese oxide nanoparticles. Overall, this work provides new insights into smartly designed manganese oxide nanoparticles with multitasking capabilities to target bioimaging and therapeutic applications.
Collapse
Affiliation(s)
- Kingsley Poon
- ARC Centre for Innovative BioEngineering, Tissue Engineering and Biomaterials Research Unit, Sydney Nano Institute, School of Biomedical Engineering, The University of Sydney NSW 2008 Australia
| | - Zufu Lu
- ARC Centre for Innovative BioEngineering, Tissue Engineering and Biomaterials Research Unit, Sydney Nano Institute, School of Biomedical Engineering, The University of Sydney NSW 2008 Australia
| | - Yves De Deene
- Department of Engineering, The Biomedical Engineering Laboratory, Macquarie University Sydney 2109 Australia
| | - Yogambha Ramaswamy
- ARC Centre for Innovative BioEngineering, Tissue Engineering and Biomaterials Research Unit, Sydney Nano Institute, School of Biomedical Engineering, The University of Sydney NSW 2008 Australia
| | - Hala Zreiqat
- ARC Centre for Innovative BioEngineering, Tissue Engineering and Biomaterials Research Unit, Sydney Nano Institute, School of Biomedical Engineering, The University of Sydney NSW 2008 Australia
| | - Gurvinder Singh
- ARC Centre for Innovative BioEngineering, Tissue Engineering and Biomaterials Research Unit, Sydney Nano Institute, School of Biomedical Engineering, The University of Sydney NSW 2008 Australia
| |
Collapse
|
18
|
Øverbye A, Torgersen ML, Sønstevold T, Iversen TG, Mørch Ý, Skotland T, Sandvig K. Cabazitaxel-loaded poly(alkyl cyanoacrylate) nanoparticles: toxicity and changes in the proteome of breast, colon and prostate cancer cells. Nanotoxicology 2021; 15:865-884. [PMID: 34047629 DOI: 10.1080/17435390.2021.1924888] [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: 12/23/2022]
Abstract
Nanoparticles composed of poly(alkyl cyanoacrylate) (PACA) have shown great promise due to their biodegradability and high drug loading capacity. Development of optimal PACA nanocarriers requires detailed analysis of the overall cellular impact exerted by PACA variants. We here perform a comprehensive comparison of cabazitaxel (CBZ)-loaded nanocarriers composed of three different PACA monomers, i.e. poly(n-butyl cyanoacrylate) (PBCA), poly(2-ethylbutyl cyanoacrylate) (PEBCA) and poly(octyl cyanoacrylate) (POCA). The cytotoxicity of drug-loaded and empty PACA nanoparticles were compared to that of free CBZ across a panel of nine cancer cell lines by assessing cellular metabolism, proliferation and protein synthesis. The analyses revealed that the cytotoxicity of all CBZ-loaded PACAs was similar to that of free CBZ for all cell lines tested, whereas the empty PACAs exerted much lower toxicity. To increase our understanding of the toxic effects of these treatments comprehensive MS-based proteomics were performed with HCT116, MDA-MB-231 and PC3 cells incubated with PACA-CBZ variants or free CBZ. Interestingly, PACA-CBZ specifically led to decreased levels of proteins involved in focal adhesion and stress fibers in all cell lines. Since we recently demonstrated that encapsulation of CBZ within PEBCA nanoparticles significantly improved the therapeutic effect of CBZ on a patient derived xenograft model in mice, we investigated the effects of this PACA variant more closely by immunoblotting. Interestingly, we detected several changes in the protein expression and degree of phosphorylation of SRC-pathway proteins that can be relevant for the therapeutic effects of these substances.
Collapse
Affiliation(s)
- Anders Øverbye
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Maria Lyngaas Torgersen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Tonje Sønstevold
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| | - Tore Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim, Norway
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, The Norwegian Radium Hospital, Oslo, Norway.,Department of Biosciences, University of Oslo, Oslo, Norway
| |
Collapse
|
19
|
Snipstad S, Mørch Ý, Sulheim E, Åslund A, Pedersen A, Davies CDL, Hansen R, Berg S. Sonopermeation Enhances Uptake and Therapeutic Effect of Free and Encapsulated Cabazitaxel. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:1319-1333. [PMID: 33549379 DOI: 10.1016/j.ultrasmedbio.2020.12.026] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 09/18/2020] [Accepted: 12/27/2020] [Indexed: 06/12/2023]
Abstract
Delivery of drugs and nanomedicines to tumors is often heterogeneous and insufficient and, thus, of limited efficacy. Microbubbles in combination with ultrasound have been found to improve delivery to tumors, enhancing accumulation and penetration. We used a subcutaneous prostate cancer xenograft model in mice to investigate the effect of free and nanoparticle-encapsulated cabazitaxel in combination with ultrasound and microbubbles with a lipid shell or a shell of nanoparticles. Sonopermeation reduced tumor growth and prolonged survival (26%-100%), whether the free drug was co-injected with lipid-shelled microbubbles or the nanoformulation was co-injected with lipid-shelled or nanoparticle-shelled microbubbles. Coherently with the improved therapeutic response, we found enhanced uptake of nanoparticles directly after ultrasound treatment that lasted several weeks (2.3 × -15.8 × increase). Neither cavitation dose nor total accumulation of nanoparticles could explain the variation within treatment groups, emphasizing the need for a better understanding of the tumor biology and mechanisms involved in ultrasound-mediated treatment.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway.
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Einar Sulheim
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Cancer Clinic, St. Olav's Hospital, Trondheim, Norway
| | - Andreas Åslund
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - André Pedersen
- Department of Health Research, SINTEF Digital, Trondheim, Norway
| | | | - Rune Hansen
- Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway; Department of Circulation and Medical imaging, Norwegian University of Science and Technology, Trondheim, Norway
| |
Collapse
|
20
|
Deprez J, Lajoinie G, Engelen Y, De Smedt SC, Lentacker I. Opening doors with ultrasound and microbubbles: Beating biological barriers to promote drug delivery. Adv Drug Deliv Rev 2021; 172:9-36. [PMID: 33705877 DOI: 10.1016/j.addr.2021.02.015] [Citation(s) in RCA: 101] [Impact Index Per Article: 33.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 02/01/2021] [Accepted: 02/17/2021] [Indexed: 12/13/2022]
Abstract
Apart from its clinical use in imaging, ultrasound has been thoroughly investigated as a tool to enhance drug delivery in a wide variety of applications. Therapeutic ultrasound, as such or combined with cavitating nuclei or microbubbles, has been explored to cross or permeabilize different biological barriers. This ability to access otherwise impermeable tissues in the body makes the combination of ultrasound and therapeutics very appealing to enhance drug delivery in situ. This review gives an overview of the most important biological barriers that can be tackled using ultrasound and aims to provide insight on how ultrasound has shown to improve accessibility as well as the biggest hurdles. In addition, we discuss the clinical applicability of therapeutic ultrasound with respect to the main challenges that must be addressed to enable the further progression of therapeutic ultrasound towards an effective, safe and easy-to-use treatment tailored for drug delivery in patients.
Collapse
Affiliation(s)
- J Deprez
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - G Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology and Technical Medical (TechMed) Center, University of Twente, P.O. Box 217, 7500 AE Enschede, Netherlands
| | - Y Engelen
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| | - S C De Smedt
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium.
| | - I Lentacker
- Ghent Research Group on Nanomedicines, Department of Pharmaceutics, Ghent University, Ottergemsesteenweg 460, 9000 Ghent, Belgium; Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
21
|
Snipstad S, Hanstad S, Bjørkøy A, Mørch Ý, de Lange Davies C. Sonoporation Using Nanoparticle-Loaded Microbubbles Increases Cellular Uptake of Nanoparticles Compared to Co-Incubation of Nanoparticles and Microbubbles. Pharmaceutics 2021; 13:640. [PMID: 33946327 PMCID: PMC8146007 DOI: 10.3390/pharmaceutics13050640] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/15/2021] [Accepted: 04/26/2021] [Indexed: 12/19/2022] Open
Abstract
Therapeutic agents can benefit from encapsulation in nanoparticles, due to improved pharmacokinetics and biodistribution, protection from degradation, increased cellular uptake and sustained release. Microbubbles in combination with ultrasound have been shown to improve the delivery of nanoparticles and drugs to tumors and across the blood-brain barrier. Here, we evaluate two different microbubbles for enhancing the delivery of polymeric nanoparticles to cells in vitro: a commercially available lipid microbubble (Sonazoid) and a microbubble with a shell composed of protein and nanoparticles. Various ultrasound parameters are applied and confocal microscopy is employed to image cellular uptake. Ultrasound enhanced cellular uptake depending on the pressure and duty cycle. The responsible mechanisms are probably sonoporation and sonoprinting, followed by uptake, and to a smaller degree enhanced endocytosis. The use of commercial Sonazoid microbubbles leads to significantly lower uptake than when using nanoparticle-loaded microbubbles, suggesting that proximity between cells, nanoparticles and microbubbles is important, and that mainly nanoparticles in the shell are taken up, rather than free nanoparticles in solution.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Sem Sælandsvei 2A, 7034 Trondheim, Norway;
- Cancer Clinic, St. Olav’s Hospital, Prinsesse Kristinas Gate 1, 7030 Trondheim, Norway
| | - Sigurd Hanstad
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| | - Ýrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Sem Sælandsvei 2A, 7034 Trondheim, Norway;
| | - Catharina de Lange Davies
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway; (S.H.); (A.B.); (C.d.L.D.)
| |
Collapse
|
22
|
Wei P, Cornel EJ, Du J. Ultrasound-responsive polymer-based drug delivery systems. Drug Deliv Transl Res 2021; 11:1323-1339. [PMID: 33761101 PMCID: PMC7989687 DOI: 10.1007/s13346-021-00963-0] [Citation(s) in RCA: 62] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/11/2021] [Indexed: 02/06/2023]
Abstract
Ultrasound-responsive polymeric materials have received a tremendous amount of attention from scientists for several decades. Compared to other stimuli-responsive materials (such as UV-, thermal-, and pH-responsive materials), these smart materials are more applicable since they allow more efficient drug delivery and targeted treatment by fairly non-invasive means. This review describes the recent advances of such ultrasound-responsive polymer-based drug delivery systems and illustrates various applications. More specifically, the mechanism of ultrasound-induced drug delivery, typical formulations, and biomedical applications (tumor therapy, disruption of blood-brain barrier, fighting infectious diseases, transdermal drug delivery, and enhanced thrombolysis) are summarized. Finally, a perspective on the future research directions for the development of ultrasound-responsive polymeric materials to facilitate a clinical translation is given.
Collapse
Affiliation(s)
- Ping Wei
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
| | - Erik Jan Cornel
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai, 201804, China
| | - Jianzhong Du
- Department of Polymeric Materials, School of Materials Science and Engineering, Key Laboratory of Advanced Civil Engineering Materials of Ministry of Education, Tongji University, 4800 Caoan Road, Shanghai, 201804, China. .,Department of Orthopedics, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
23
|
Lovmo MK, Yemane PT, Bjorkoy A, Hansen R, Cleveland RO, Angelsen BA, de Lange Davies C. Effect of Acoustic Radiation Force on Displacement of Nanoparticles in Collagen Gels. IEEE TRANSACTIONS ON ULTRASONICS, FERROELECTRICS, AND FREQUENCY CONTROL 2021; 68:416-431. [PMID: 32746200 DOI: 10.1109/tuffc.2020.3006762] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Penetration of nanoscale therapeutic agents into the extracellular matrix (ECM) of a tumor is a limiting factor for the sufficient delivery of drugs in tumors. Ultrasound (US) in combination with microbubbles causing cavitation is reported to improve delivery of nanoparticles (NPs) and drugs to tumors. Acoustic radiation force (ARF) could also enhance the penetration of NPs in tumor ECM. In this work, a collagen gel was used as a model for tumor ECM to study the effects of ARF on the penetration of NPs as well as the deformation of collagen gels applying different US parameters (varying pressure and duty cycle). The collagen gel was characterized, and the diffusion of water and NPs was measured. The penetration of NPs into the gel was measured by confocal laser scanning microscopy and numerical simulations were performed to determine the ARF and to estimate the penetration distance and extent of deformation. ARF had no effect on the penetration of NPs into the collagen gels for the US parameters and gel used, whereas a substantial deformation was observed. The width of the deformation on the collagen gel surface corresponded to the US beam. Comparing ARF caused by attenuation within the gel and Langevin pressure caused by reflection at the gel-water surface, ARF was the prevalent mechanism for the gel deformation. The experimental and theoretical results were consistent both with respect to the NP penetration and the gel deformation.
Collapse
|
24
|
Barmin RA, Rudakovskaya PG, Chernyshev VS, Guslyakova OI, Belcov PA, Obukhova EN, Gayer AV, Shirshin EA, Gorin DA. Optoacoustic/Fluorescent/Acoustic Imaging Probe Based on Air-Filled Bubbles Functionalized with Gold Nanorods and Fluorescein Isothiocyanate. ACS OMEGA 2021; 6:3809-3821. [PMID: 33585760 PMCID: PMC7876831 DOI: 10.1021/acsomega.0c05518] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Accepted: 12/28/2020] [Indexed: 05/08/2023]
Abstract
Liquid/surfactant/gas interfaces are promising objects for nanoengineered multimodal contrasts, which can be used for biomedical imaging in preclinical and clinical applications. Microbubbles with the gaseous core and shell made of lipids/proteins have already acted as ultrasound (US) contrast agents for angiography. In the present work, microbubbles with a shell composed of Span 60 and Tween 80 surfactants functionalized with fluorescein isothiocyanate and gold nanorods to achieve a multimodal combination of US, fluorescence, and optoacoustic imaging are described. Optimal conditions for microbubble generation by studying the surface tension of the initial solutions and analyzing the size, stability, and charge of the resulting bubbles were found. By controlling and modifying bubbles' surface properties, an increase in stability and storage time can be achieved. The functionalization of bubbles with gold nanoparticles and a dye by using an optimally selected sonication protocol was performed. The biomedical application's potential in imaging modalities of functionalized microbubbles using a medical US device with a frequency of 50 MHz, fluorescence tomography, and raster-scanning optoacoustic mesoscopy measurements was evaluated. The obtained results are important for optimum stabilization and functionalization of gas/liquid interfaces and the following applications in the multimodal biomedical imaging.
Collapse
Affiliation(s)
- Roman A. Barmin
- Skolkovo
Institute of Science and Technology, 3 Nobelya Str., Moscow 121205, Russia
| | | | | | - Olga I. Guslyakova
- Saratov
State University, 83 Astrakhanskaya Str., Saratov 410012, Russia
| | - Pavel A. Belcov
- Anta-Med
Premium, LLC, 11 Derbenevskaya
Naberezhnaya, Moscow 115114, Russia
| | | | - Alexey V. Gayer
- Lomonosov
Moscow State University, 1/2 Leninskie Gory, Moscow 119991, Russia
| | - Evgeny A. Shirshin
- Lomonosov
Moscow State University, 1/2 Leninskie Gory, Moscow 119991, Russia
- Institute
of Spectroscopy of the Russian Academy of Sciences, 5 Fizicheskaya Str., Troitsk, Moscow 108840, Russia
- Institute
for Regenerative Medicine, Sechenov First
Moscow State Medical University, Trubetskaya 8-2, Moscow 119048, Russia
| | - Dmitry A. Gorin
- Skolkovo
Institute of Science and Technology, 3 Nobelya Str., Moscow 121205, Russia
| |
Collapse
|
25
|
Versluis M, Stride E, Lajoinie G, Dollet B, Segers T. Ultrasound Contrast Agent Modeling: A Review. ULTRASOUND IN MEDICINE & BIOLOGY 2020; 46:2117-2144. [PMID: 32546411 DOI: 10.1016/j.ultrasmedbio.2020.04.014] [Citation(s) in RCA: 97] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 04/11/2020] [Accepted: 04/14/2020] [Indexed: 05/21/2023]
Abstract
Ultrasound is extensively used in medical imaging, being safe and inexpensive and operating in real time. Its scope of applications has been widely broadened by the use of ultrasound contrast agents (UCAs) in the form of microscopic bubbles coated by a biocompatible shell. Their increased use has motivated a large amount of research to understand and characterize their physical properties as well as their interaction with the ultrasound field and their surrounding environment. Here we review the theoretical models that have been proposed to study and predict the behavior of UCAs. We begin with a brief introduction on the development of UCAs. We then present the basics of free-gas-bubble dynamics upon which UCA modeling is based. We review extensively the linear and non-linear models for shell elasticity and viscosity and present models for non-spherical and asymmetric bubble oscillations, especially in the presence of surrounding walls or tissue. Then, higher-order effects such as microstreaming, shedding and acoustic radiation forces are considered. We conclude this review with promising directions for the modeling and development of novel agents.
Collapse
Affiliation(s)
- Michel Versluis
- Physics of Fluids Group, MESA+ Institute for Nanotechnology, Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands.
| | - Eleanor Stride
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Guillaume Lajoinie
- Physics of Fluids Group, MESA+ Institute for Nanotechnology, Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| | - Benjamin Dollet
- Centre National de la Recherche Scientifique (CNRS), Laboratoire Interdisciplinaire de Physique (LIPhy), Université Grenoble Alpes, Grenoble, France
| | - Tim Segers
- Physics of Fluids Group, MESA+ Institute for Nanotechnology, Technical Medical (TechMed) Center, University of Twente, Enschede, the Netherlands
| |
Collapse
|
26
|
Sultana S, Alzahrani N, Alzahrani R, Alshamrani W, Aloufi W, Ali A, Najib S, Siddiqui NA. Stability issues and approaches to stabilised nanoparticles based drug delivery system. J Drug Target 2020; 28:468-486. [PMID: 31984810 DOI: 10.1080/1061186x.2020.1722137] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
Nanoparticles form the fundamental building blocks for many exciting applications in various scientific disciplines due to its unique features such as large surface to mass ratio, targeting potential, ability to adsorbed and carry other compound which makes them suitable for biomedical applications. However, the problem of the large-scale synthesis of nanoparticles remains challenging due to physical instability associated with nanoparticles which lead to generation of aggregates particles with high polydispersity index (PDI) indicating low particle homogeneity and eventually loss of their special nanoscale properties. The stabilisation concept can be generated by repulsive electrostatic force, which nanoparticles experience, when they are surrounded by a double layer of electric charges. Selection of proper stabiliser will govern the stability of NPs and ultimately development of optimised drug delivery system. This review summarises mechanism of physical instability issues likely to be encountered during the development of nanoformulations. It also discusses potential stabilising agents used so far and their mechanism in achieving stable nanosystems.
Collapse
Affiliation(s)
| | | | | | | | - Waad Aloufi
- Pharmaceutics, Taif University, Taif, Saudi Arabia
| | - Amena Ali
- Pharmaceutical Chemistry, Taif University, Taif, Saudi Arabia
| | - Shehla Najib
- Pharmacognosy and Phytochemistry, King Khalid University, Abha, Saudi Arabia
| | | |
Collapse
|
27
|
Abstract
Ultrasound and magneto-responsive nanosized drug delivery systems have been designed as novel carriers for controlled release. Colloidal bubbles (CBs) could be designed to incorporate different materials, such as protein, lipid, polymer, surfactants, and even nanoparticles in their shell, which makes them suitable for a wide range of drug delivery applications. The interior of CBs may be filled with different gases, which is essential for conferring the characteristics of an ultrasounds contrasting agent. Manipulating the core of CBs enhances features such as stability and duration of the echogenic effect. Thus CBs derivatized with nanoparticles combine functional properties of CBs and NPs to yield a versatile theranostics platform technology.
Collapse
|
28
|
Fang B, Nakagawa K. Modification of casein aggregate microstructures under frozen conditions: A study using tunable resistive pulse sensing. Food Hydrocoll 2020. [DOI: 10.1016/j.foodhyd.2019.105259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
29
|
Yemane PT, Åslund AKO, Snipstad S, Bjørkøy A, Grendstad K, Berg S, Mørch Y, Torp SH, Hansen R, Davies CDL. Effect of Ultrasound on the Vasculature and Extravasation of Nanoscale Particles Imaged in Real Time. ULTRASOUND IN MEDICINE & BIOLOGY 2019; 45:3028-3041. [PMID: 31474384 DOI: 10.1016/j.ultrasmedbio.2019.07.683] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/31/2019] [Accepted: 07/31/2019] [Indexed: 06/10/2023]
Abstract
Ultrasound and microbubbles have been found to improve the delivery of drugs and nanoparticles to tumor tissue. To obtain new knowledge on the influence of vascular parameters on extravasation and to elucidate the effect of acoustic pressure on extravasation and penetration of nanoscale particles into the extracellular matrix, real-time intravital multiphoton microscopy was performed during sonication of tumors growing in dorsal window chambers. The impact of vessel diameter, vessel structure and blood flow was characterized. Fluorescein isothiocyanate-dextran (2 MDa) was injected to visualize blood vessels. Mechanical indexes (MI) of 0.2-0.8 and in-house-made, nanoparticle-stabilized microbubbles or Sonovue were applied. The rate and extent of penetration into the extracellular matrix increased with increasing MI. However, to achieve extravasation, smaller vessels required MIs (0.8) higher than those of blood vessels with larger diameters. Ultrasound changed the blood flow rate and direction. Interestingly, the majority of extravasations occurred at vessel branching points.
Collapse
Affiliation(s)
- Petros T Yemane
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Andreas K O Åslund
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway; Stroke Unit, Department of Internal Medicine, St. Olav's Hospital, Trondheim, Norway
| | - 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
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Kristin Grendstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sigrid Berg
- Cancer Clinic, St. Olav's Hospital, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF Industry, Trondheim, Norway
| | - Sverre H Torp
- Department of Pathology, St. Olav's Hospital, Trondheim, Norway; Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology, Trondheim, Norway
| | - Rune Hansen
- Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway; Department of Health Research, SINTEF Digital, Trondheim, Norway
| | | |
Collapse
|
30
|
Xie Y, Wang J, Wang J, Hu Z, Hariri A, Tu N, Krug KA, Burkart MD, Gianneschi NC, Jokerst JV, Rinehart JD. Tuning the ultrasonic and photoacoustic response of polydopamine-stabilized perfluorocarbon contrast agents. J Mater Chem B 2019; 7:4833-4842. [PMID: 31389967 PMCID: PMC6690494 DOI: 10.1039/c9tb00928k] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Contrast-enhanced ultrasound (CEUS) offers the exciting prospect of retaining the ease of ultrasound imaging while enhancing imaging clarity, diagnostic specificity, and theranostic capability. To advance the capabilities of CEUS, the synthesis and understanding of new ultrasound contrast agents (UCAs) is a necessity. Many UCAs are nano- or micro-scale materials composed of a perfluorocarbon (PFC) and stabilizer that synergistically induce an ultrasound response that is both information-rich and easily differentiated from natural tissue. In this work, we probe the extent to which CEUS is modulated through variation in a PFC stabilized with fluorine-modified polydopamine nanoparticles (PDA NPs). The high level of synthetic tunability in this system allows us to study signal as a function of particle aggregation and PFC volatility in a systematic manner. Separation of aggregated and non-aggregated nanoparticles lead to a fundamentally different signal response, and for this system, PFC volatility has little effect on CEUS intensity despite a range of over 50 °C in boiling point. To further explore the imaging tunability and multimodality, Fe3+-chelation was employed to generate an enhanced photoacoustic (PA) signal in addition to the US signal. In vitro and in vivo results demonstrate that PFC-loaded PDA NPs show stronger PA signal than the non-PFC ones, indicating that the PA signal can be used for in situ differentiation between PFC-loading levels. In sum, these data evince the rich role synthetic chemistry can play in guiding new directions of development for UCAs.
Collapse
Affiliation(s)
- Yijun Xie
- Department of Chemistry and Biochemistry, University of California, San Diego, La Jolla, CA 92093, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
31
|
Szwed M, Sønstevold T, Øverbye A, Engedal N, Grallert B, Mørch Ý, Sulheim E, Iversen TG, Skotland T, Sandvig K, Torgersen ML. Small variations in nanoparticle structure dictate differential cellular stress responses and mode of cell death. Nanotoxicology 2019; 13:761-782. [PMID: 30760074 DOI: 10.1080/17435390.2019.1576238] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
For optimal exploitation of nanoparticles (NPs) in biomedicine, and to predict nanotoxicity, detailed knowledge of the cellular responses to cell-bound or internalized NPs is imperative. The final outcome of NP-cell interaction is dictated by the type and magnitude of the NP insult and the cellular response. Here, this has been systematically studied by using poly(alkylcyanoacrylate) (PACA) particles differing only in their alkyl side chains; butyl (PBCA), ethylbutyl (PEBCA), or octyl (POCA), respectively. Surprisingly, these highly similar NPs induced different stress responses and modes of cell death in human cell lines. The POCA particles generally induced endoplasmic reticulum stress and apoptosis. In contrast, PBCA and PEBCA particles induced oxidative stress and lipid peroxidation depending on the level of the glutathione precursor cystine and transcription of the cystine transporter SLC7A11. The latter was induced as a protective response by the transcription factors ATF4 and Nrf2. PBCA particles strongly activated ATF4 downstream of the eIF2α kinase HRI, whereas PEBCA particles more potently induced Nrf2 antioxidant responses. Intriguingly, PBCA particles activated the cell death mechanism ferroptosis; a promising option for targeting multidrug-resistant cancers. Our findings highlight that even minor differences in NP composition can severely impact the cellular response to NPs. This may have important implications in therapeutic settings.
Collapse
Affiliation(s)
- Marzena Szwed
- a Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway
| | - Tonje Sønstevold
- a Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,b Faculty of Mathematics and Natural Sciences, Department of Biosciences , University of Oslo , Oslo , Norway
| | - Anders Øverbye
- a Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway
| | - Nikolai Engedal
- c Centre for Molecular Medicine Norway (NCMM), Nordic EMBL Partnership, University of Oslo , Oslo , Norway
| | - Beata Grallert
- d Department of Radiation Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway
| | - Ýrr Mørch
- e Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway
| | - Einar Sulheim
- e Department of Biotechnology and Nanomedicine , SINTEF AS , Trondheim , Norway.,f Faculty of Natural Sciences, Department of Physics , The Norwegian University of Science and Technology (NTNU) , Trondheim , Norway
| | - Tore-Geir Iversen
- a Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway
| | - Tore Skotland
- a Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway
| | - Kirsten Sandvig
- a Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway.,b Faculty of Mathematics and Natural Sciences, Department of Biosciences , University of Oslo , Oslo , Norway
| | - Maria L Torgersen
- a Department of Molecular Cell Biology , Institute for Cancer Research, Oslo University Hospital , Oslo , Norway
| |
Collapse
|
32
|
Sulheim E, Mørch Y, Snipstad S, Borgos SE, Miletic H, Bjerkvig R, Davies CDL, Åslund AK. Therapeutic Effect of Cabazitaxel and Blood-Brain Barrier opening in a Patient-Derived Glioblastoma Model. Nanotheranostics 2019; 3:103-112. [PMID: 30899638 PMCID: PMC6427936 DOI: 10.7150/ntno.31479] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2018] [Accepted: 02/05/2019] [Indexed: 01/21/2023] Open
Abstract
Treatment of glioblastoma and other diseases in the brain is especially challenging due to the blood-brain barrier, which effectively protects the brain parenchyma. In this study we show for the first time that cabazitaxel, a semi-synthetic derivative of docetaxel can cross the blood-brain barrier and give a significant therapeutic effect in a patient-derived orthotopic model of glioblastoma. We show that the drug crosses the blood-brain barrier more effectively in the tumor than in the healthy brain due to reduced expression of p-glycoprotein efflux pumps in the vasculature of the tumor. Surprisingly, neither ultrasound-mediated blood-brain barrier opening (sonopermeation) nor drug formulation in polymeric nanoparticles could increase either accumulation of the drug in the brain or therapeutic effect. This indicates that for hydrophobic drugs, sonopermeation of the blood brain barrier might not be sufficient to achieve improved drug delivery. Nonetheless, our study shows that cabazitaxel is a promising drug for the treatment of brain tumors.
Collapse
Affiliation(s)
- Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim Norway
- Cancer Clinic, St.Olav's University Hospital, Trondheim Norway
| | - Yrr Mørch
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim Norway
| | - Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim Norway
- Cancer Clinic, St.Olav's University Hospital, Trondheim Norway
| | - Sven Even Borgos
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim Norway
| | - Hrvoje Miletic
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- Department of Biomedicine, University of Bergen, Norway
| | - Rolf Bjerkvig
- Department of Biomedicine, University of Bergen, Norway
- Department of Oncology, Luxembourg Institute of Health, Luxembourg
| | | | - Andreas K.O. Åslund
- Department of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Biotechnology and Nanomedicine, SINTEF AS, Trondheim Norway
- Stroke Unit, Department of internal medicine, St. Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
33
|
Valadbaigi P, Ettelaie R, Kulak AN, Murray BS. Generation of ultra-stable Pickering microbubbles via poly alkylcyanoacrylates. J Colloid Interface Sci 2019; 536:618-627. [DOI: 10.1016/j.jcis.2018.10.004] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 10/01/2018] [Accepted: 10/03/2018] [Indexed: 12/16/2022]
|
34
|
Yoon YI, Pang X, Jung S, Zhang G, Kong M, Liu G, Chen X. Smart Gold Nanoparticle-Stabilized Ultrasound Microbubbles as Cancer Theranostics. J Mater Chem B 2018; 6:3235-3239. [PMID: 30420913 PMCID: PMC6226101 DOI: 10.1039/c8tb00368h] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Smart gold nanoparticle-stabilized microbubbles (SAuMBs) composed of a gas-filled core and shell including smart gold nanoparticles (SAuNPs) which can be aggregated in tumors were applied as ultrasound-mediated cancer theranostics. The gas core in the microstructure enabled the detection of tumors using ultrasound and facilitated the delivery of SAuNPs by sonoporation. The SAuNPs spontaneously aggregated in tumors, which allowed photoacoustic (PA) monitoring and photothermal treatment (PTT) of tumors.
Collapse
Affiliation(s)
- Young Il Yoon
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
- IT and Medical Research Team, Korea Textile Development Institute (KTDI), 136 Gukchaebosangro, Seo-gu, Daegu, 41842, South Korea
| | - Xin Pang
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Sungwook Jung
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Guofeng Zhang
- Trans-NIH Shared Resource on Biomedical Engineering and Physical Science, National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| | - Minsuk Kong
- Laboratory of Molecular Biology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, 20892, USA
| | - Gang Liu
- State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics & Center for Molecular Imaging and Translational Medicine, School of Public Health, Xiamen University, Xiamen, 361102, China
| | - Xiaoyuan Chen
- Laboratory of Molecular Imaging and Nanomedicine (LOMIN), National Institute of Biomedical Imaging and Bioengineering (NIBIB), National Institutes of Health (NIH), Bethesda, Maryland, 20892, USA
| |
Collapse
|
35
|
Heider S, Muzard J, Zaruba M, Metzner C. Integrated Method for Purification and Single-Particle Characterization of Lentiviral Vector Systems by Size Exclusion Chromatography and Tunable Resistive Pulse Sensing. Mol Biotechnol 2018; 59:251-259. [PMID: 28567687 PMCID: PMC5486506 DOI: 10.1007/s12033-017-0009-8] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Elements derived from lentiviral particles such as viral vectors or virus-like particles are commonly used for biotechnological and biomedical applications, for example in mammalian protein expression, gene delivery or therapy, and vaccine development. Preparations of high purity are necessary in most cases, especially for clinical applications. For purification, a wide range of methods are available, from density gradient centrifugation to affinity chromatography. In this study we have employed size exclusion columns specifically designed for the easy purification of extracellular vesicles including exosomes. In addition to viral marker protein and total protein analysis, a well-established single-particle characterization technology, termed tunable resistive pulse sensing, was employed to analyze fractions of highest particle load and purity and characterize the preparations by size and surface charge/electrophoretic mobility. With this study, we propose an integrated platform combining size exclusion chromatography and tunable resistive pulse sensing for monitoring production and purification of viral particles.
Collapse
Affiliation(s)
- Susanne Heider
- Institute of Virology, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria.,Biological Physics, Department of Physics, Chalmers University of Technology, 412 96, Gothenburg, Sweden
| | - Julien Muzard
- Izon Science, 8C Homersham Place, PO Box 39168, Burnside, Christchurch, 8053, New Zealand
| | - Marianne Zaruba
- Institute of Virology, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria
| | - Christoph Metzner
- Institute of Virology, University of Veterinary Medicine, Veterinärplatz 1, 1210, Vienna, Austria.
| |
Collapse
|
36
|
Baghirov H, Snipstad S, Sulheim E, Berg S, Hansen R, Thorsen F, Mørch Y, Davies CDL, Åslund AKO. Ultrasound-mediated delivery and distribution of polymeric nanoparticles in the normal brain parenchyma of a metastatic brain tumour model. PLoS One 2018; 13:e0191102. [PMID: 29338016 PMCID: PMC5770053 DOI: 10.1371/journal.pone.0191102] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2017] [Accepted: 12/28/2017] [Indexed: 01/12/2023] Open
Abstract
The treatment of brain diseases is hindered by the blood-brain barrier (BBB) preventing most drugs from entering the brain. Focused ultrasound (FUS) with microbubbles can open the BBB safely and reversibly. Systemic drug injection might induce toxicity, but encapsulation into nanoparticles reduces accumulation in normal tissue. Here we used a novel platform based on poly(2-ethyl-butyl cyanoacrylate) nanoparticle-stabilized microbubbles to permeabilize the BBB in a melanoma brain metastasis model. With a dual-frequency ultrasound transducer generating FUS at 1.1 MHz and 7.8 MHz, we opened the BBB using nanoparticle-microbubbles and low-frequency FUS, and applied high-frequency FUS to generate acoustic radiation force and push nanoparticles through the extracellular matrix. Using confocal microscopy and image analysis, we quantified nanoparticle extravasation and distribution in the brain parenchyma. We also evaluated haemorrhage, as well as the expression of P-glycoprotein, a key BBB component. FUS and microbubbles distributed nanoparticles in the brain parenchyma, and the distribution depended on the extent of BBB opening. The results from acoustic radiation force were not conclusive, but in a few animals some effect could be detected. P-glycoprotein was not significantly altered immediately after sonication. In summary, FUS with our nanoparticle-stabilized microbubbles can achieve accumulation and displacement of nanoparticles in the brain parenchyma.
Collapse
Affiliation(s)
- Habib Baghirov
- Department of Physics, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sofie Snipstad
- Department of Physics, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- SINTEF Materials and Chemistry, Trondheim, Norway
| | - Sigrid Berg
- SINTEF Medical Technology, Trondheim, Norway
- Department of Circulation and Medical Imaging, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Rune Hansen
- SINTEF Medical Technology, Trondheim, Norway
- Department of Circulation and Medical Imaging, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Frits Thorsen
- Molecular Imaging Center and Kristian Gerhard Jebsen Brain Tumour Research Centre, Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Yrr Mørch
- SINTEF Materials and Chemistry, Trondheim, Norway
| | - Catharina de Lange Davies
- Department of Physics, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- * E-mail:
| | - Andreas K. O. Åslund
- Department of Physics, The Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| |
Collapse
|
37
|
Sulheim E, Iversen TG, To Nakstad V, Klinkenberg G, Sletta H, Schmid R, Hatletveit AR, Wågbø AM, Sundan A, Skotland T, Sandvig K, Mørch Ý. Cytotoxicity of Poly(Alkyl Cyanoacrylate) Nanoparticles. Int J Mol Sci 2017; 18:ijms18112454. [PMID: 29156588 PMCID: PMC5713421 DOI: 10.3390/ijms18112454] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 11/13/2017] [Accepted: 11/15/2017] [Indexed: 12/17/2022] Open
Abstract
Although nanotoxicology has become a large research field, assessment of cytotoxicity is often reduced to analysis of one cell line only. Cytotoxicity of nanoparticles is complex and should, preferentially, be evaluated in several cell lines with different methods and on multiple nanoparticle batches. Here we report the toxicity of poly(alkyl cyanoacrylate) nanoparticles in 12 different cell lines after synthesizing and analyzing 19 different nanoparticle batches and report that large variations were obtained when using different cell lines or various toxicity assays. Surprisingly, we found that nanoparticles with intermediate degradation rates were less toxic than particles that were degraded faster or more slowly in a cell-free system. The toxicity did not vary significantly with either the three different combinations of polyethylene glycol surfactants or with particle size (range 100–200 nm). No acute pro- or anti-inflammatory activity on cells in whole blood was observed.
Collapse
Affiliation(s)
- Einar Sulheim
- SINTEF Materials and Chemistry, Sem Sælands vei 2A, 7034 Trondheim, Norway.
- Department of Physics, Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway.
| | - Tore-Geir Iversen
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway.
- Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway.
| | - Vu To Nakstad
- SINTEF Materials and Chemistry, Sem Sælands vei 2A, 7034 Trondheim, Norway.
| | - Geir Klinkenberg
- SINTEF Materials and Chemistry, Sem Sælands vei 2A, 7034 Trondheim, Norway.
| | - Håvard Sletta
- SINTEF Materials and Chemistry, Sem Sælands vei 2A, 7034 Trondheim, Norway.
| | - Ruth Schmid
- SINTEF Materials and Chemistry, Sem Sælands vei 2A, 7034 Trondheim, Norway.
| | | | - Ane Marit Wågbø
- SINTEF Materials and Chemistry, Sem Sælands vei 2A, 7034 Trondheim, Norway.
| | - Anders Sundan
- Department of Cancer Research and Molecular Medicine, Norwegian University of Science and Technology, 8905 MH, 7491 Trondheim, Norway.
| | - Tore Skotland
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway.
- Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway.
| | - Kirsten Sandvig
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital-The Norwegian Radium Hospital, 0379 Oslo, Norway.
- Center for Cancer Biomedicine, Faculty of Medicine, University of Oslo, 0379 Oslo, Norway.
- Department of Biosciences, Faculty of Mathematics and Natural Sciences, University of Oslo, 0316 Oslo, Norway.
| | - Ýrr Mørch
- SINTEF Materials and Chemistry, Sem Sælands vei 2A, 7034 Trondheim, Norway.
| |
Collapse
|
38
|
Snipstad S, Berg S, Mørch Ý, Bjørkøy A, Sulheim E, Hansen R, Grimstad I, van Wamel A, Maaland AF, Torp SH, Davies CDL. Ultrasound Improves the Delivery and Therapeutic Effect of Nanoparticle-Stabilized Microbubbles in Breast Cancer Xenografts. ULTRASOUND IN MEDICINE & BIOLOGY 2017; 43:2651-2669. [PMID: 28781149 DOI: 10.1016/j.ultrasmedbio.2017.06.029] [Citation(s) in RCA: 66] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2017] [Revised: 06/16/2017] [Accepted: 06/29/2017] [Indexed: 05/19/2023]
Abstract
Compared with conventional chemotherapy, encapsulation of drugs in nanoparticles can improve efficacy and reduce toxicity. However, delivery of nanoparticles is often insufficient and heterogeneous because of various biological barriers and uneven tumor perfusion. We investigated a unique multifunctional drug delivery system consisting of microbubbles stabilized by polymeric nanoparticles (NPMBs), enabling ultrasound-mediated drug delivery. The aim was to examine mechanisms of ultrasound-mediated delivery and to determine if increased tumor uptake had a therapeutic benefit. Cellular uptake and toxicity, circulation and biodistribution were characterized. After intravenous injection of NPMBs into mice, tumors were treated with ultrasound of various pressures and pulse lengths, and distribution of nanoparticles was imaged on tumor sections. No effects of low pressures were observed, whereas complete bubble destruction at higher pressures improved tumor uptake 2.3 times, without tissue damage. An enhanced therapeutic effect was illustrated in a promising proof-of-concept study, in which all tumors exhibited regression into complete remission.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway.
| | - Sigrid Berg
- SINTEF Technology and Society, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ýrr Mørch
- SINTEF Materials and Chemistry, Trondheim, Norway
| | - Astrid Bjørkøy
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Einar Sulheim
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway; SINTEF Materials and Chemistry, Trondheim, Norway
| | - Rune Hansen
- SINTEF Technology and Society, Trondheim, Norway; Department of Circulation and Medical Imaging, Norwegian University of Science and Technology, Trondheim, Norway
| | - Ingeborg Grimstad
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Annemieke van Wamel
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Astri F Maaland
- Department of Physics, Norwegian University of Science and Technology, Trondheim, Norway
| | - Sverre H Torp
- Department of Laboratory Medicine, Children's and Women's Health, Norwegian University of Science and Technology, Trondheim, Norway; Department of Pathology, St. Olav's University Hospital, Trondheim, Norway
| | | |
Collapse
|
39
|
Laser-Activated Polymeric Microcapsules for Ultrasound Imaging and Therapy: In Vitro Feasibility. Biophys J 2017; 112:1894-1907. [PMID: 28494960 DOI: 10.1016/j.bpj.2017.03.033] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Revised: 03/16/2017] [Accepted: 03/27/2017] [Indexed: 11/23/2022] Open
Abstract
Polymeric microcapsules with a light-absorbing dye incorporated in their shell can generate vapor microbubbles that can be spatiotemporally controlled by pulsed laser irradiation. These contrast agents of 6-8 μm in diameter can circulate through the vasculature, offering possibilities for ultrasound (molecular) imaging and targeted therapies. Here, we study the impact of such vapor bubbles on human endothelial cells in terms of cell poration and cell viability to establish the imaging and therapeutic windows. Two capsule formulations were used: the first one consisted of a high boiling point oil (hexadecane), whereas the second was loaded with a low boiling point oil (perfluoropentane). Poration probability was already 40% for the smallest bubbles that were formed (<7.5 μm diameter), and reached 100% for the larger bubbles. The hexadecane-loaded capsules also produced bubbles while their shell remained intact. These encapsulated bubbles could therefore be used for noninvasive ultrasound imaging after laser activation without inducing any cell damage. The controlled and localized cell destruction achieved by activation of both capsule formulations may provide an innovative approach for specifically inducing cell death in vivo, e.g., for cancer therapy.
Collapse
|
40
|
Mullick Chowdhury S, Lee T, Willmann JK. Ultrasound-guided drug delivery in cancer. Ultrasonography 2017; 36:171-184. [PMID: 28607323 PMCID: PMC5494871 DOI: 10.14366/usg.17021] [Citation(s) in RCA: 122] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/23/2017] [Accepted: 05/01/2017] [Indexed: 12/14/2022] Open
Abstract
Recent advancements in ultrasound and microbubble (USMB) mediated drug delivery technology has shown that this approach can improve spatially confined delivery of drugs and genes to target tissues while reducing systemic dose and toxicity. The mechanism behind enhanced delivery of therapeutics is sonoporation, the formation of openings in the vasculature, induced by ultrasound-triggered oscillations and destruction of microbubbles. In this review, progress and challenges of USMB mediated drug delivery are summarized, with special focus on cancer therapy.
Collapse
Affiliation(s)
| | - Taehwa Lee
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jürgen K. Willmann
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| |
Collapse
|
41
|
Abstract
The fields of biomedical nanotechnology and theranostics have enjoyed exponential growth in recent years. The "Molecular Imaging in Nanotechnology and Theranostics" (MINT) Interest Group of the World Molecular Imaging Society (WMIS) was created in order to provide a more organized and focused forum on these topics within the WMIS and at the World Molecular Imaging Conference (WMIC). The interest group was founded in 2015 and was officially inaugurated during the 2016 WMIC. The overarching goal of MINT is to bring together the many scientists who work on molecular imaging approaches using nanotechnology and those that work on theranostic agents. MINT therefore represents scientists, labs, and institutes that are very diverse in their scientific backgrounds and areas of expertise, reflecting the wide array of materials and approaches that drive these fields. In this short review, we attempt to provide a condensed overview over some of the key areas covered by MINT. Given the breadth of the fields and the given space constraints, we have limited the coverage to the realm of nanoconstructs, although theranostics is certainly not limited to this domain. We will also focus only on the most recent developments of the last 3-5 years, in order to provide the reader with an intuition of what is "in the pipeline" and has potential for clinical translation in the near future.
Collapse
Affiliation(s)
- Chrysafis Andreou
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Suchetan Pal
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Lara Rotter
- Department of Neurology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Jiang Yang
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - Moritz F Kircher
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Center for Molecular Imaging and Nanotechnology (CMINT), Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
- Department of Radiology, Weill Cornell Medical College, New York, NY, 10065, USA.
| |
Collapse
|
42
|
Appold L, Shi Y, Rütten S, Kühne A, Pich A, Kiessling F, Lammers T. Physicochemical Characterization of the Shell Composition of PBCA-Based Polymeric Microbubbles. Macromol Biosci 2017; 17. [PMID: 28371270 DOI: 10.1002/mabi.201700002] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2017] [Revised: 02/16/2017] [Indexed: 12/31/2022]
Abstract
Microbubbles (MB) are routinely used as contrast agents for ultrasound (US) imaging. In recent years, MB have also attracted interest as drug delivery systems. Soft-shelled lipidic MB tend to be more advantageous for US imaging, while hard-shelled polymeric MB appear to be more suitable for drug delivery purposes because of their thicker shell and the resulting higher drug loading capacity. The physicochemical composition of the shell of polymeric MB, however, remains largely unknown. This study sets out to evaluate the molecular weight and polydispersity of the building blocks constituting the shell of poly(butyl cyanoacrylate) (PBCA) MB. Several different PBCA MB were synthesized, varying preparation parameters such as pH, surfactant, stirring speed, and stirring time. Using gel permeation chromatography, it is found that the number average molecular weight (M n ) of the polymer chains in the shell of PBCA MB is 4 kDa, and that >99% of the polymer chains are below 40 kDa. This demonstrates that virtually all polymeric building blocks in the shell of PBCA MB have a size which allows for renal excretion, thereby supporting their use for drug delivery applications.
Collapse
Affiliation(s)
- Lia Appold
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Yang Shi
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Stephan Rütten
- Electron Microscopic Facility, University Hospital RWTH, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Alexander Kühne
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forckenbeckstrasse 50, 52056, Aachen, Germany
| | - Andrij Pich
- DWI-Leibniz Institute for Interactive Materials, RWTH Aachen University, Forckenbeckstrasse 50, 52056, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Pauwelsstrasse 30, 52074, Aachen, Germany
| | - Twan Lammers
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Clinic and Helmholtz Institute for Biomedical Engineering, Pauwelsstrasse 30, 52074, Aachen, Germany
| |
Collapse
|
43
|
Åslund AKO, Sulheim E, Snipstad S, von Haartman E, Baghirov H, Starr N, Kvåle Løvmo M, Lelú S, Scurr D, Davies CDL, Schmid R, Mørch Ý. Quantification and Qualitative Effects of Different PEGylations on Poly(butyl cyanoacrylate) Nanoparticles. Mol Pharm 2017; 14:2560-2569. [DOI: 10.1021/acs.molpharmaceut.6b01085] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Andreas K. O. Åslund
- Department
of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Einar Sulheim
- Department
of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- SINTEF Materials and Chemistry, Trondheim, Norway
| | - Sofie Snipstad
- Department
of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Eva von Haartman
- Pharmaceutical
Sciences Laboratory, Åbo Akademi University, Turku, Finland
| | - Habib Baghirov
- Department
of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Nichola Starr
- School
of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | - Mia Kvåle Løvmo
- Department
of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Sylvie Lelú
- Department
of Physics, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - David Scurr
- School
of Pharmacy, The University of Nottingham, Nottingham NG7 2RD, United Kingdom
| | | | - Ruth Schmid
- SINTEF Materials and Chemistry, Trondheim, Norway
| | - Ýrr Mørch
- SINTEF Materials and Chemistry, Trondheim, Norway
| |
Collapse
|
44
|
Baghirov H, Melikishvili S, Mørch Y, Sulheim E, Åslund AK, Hianik T, de Lange Davies C. The effect of poly(ethylene glycol) coating and monomer type on poly(alkyl cyanoacrylate) nanoparticle interactions with lipid monolayers and cells. Colloids Surf B Biointerfaces 2017; 150:373-383. [DOI: 10.1016/j.colsurfb.2016.10.051] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Revised: 10/21/2016] [Accepted: 10/29/2016] [Indexed: 12/17/2022]
|
45
|
Fan Q, Qi F, Miao C, Yue H, Gong F, Wu J, Ma G, Su Z. Direct and controllable preparation of uniform PLGA particles with various shapes and surface morphologies. Colloids Surf A Physicochem Eng Asp 2016. [DOI: 10.1016/j.colsurfa.2016.04.028] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
46
|
Snipstad S, Hak S, Baghirov H, Sulheim E, Mørch Ý, Lélu S, von Haartman E, Bäck M, Nilsson KPR, Klymchenko AS, de Lange Davies C, Åslund AKO. Labeling nanoparticles: Dye leakage and altered cellular uptake. Cytometry A 2016; 91:760-766. [DOI: 10.1002/cyto.a.22853] [Citation(s) in RCA: 68] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/02/2016] [Accepted: 03/17/2016] [Indexed: 12/12/2022]
Affiliation(s)
- Sofie Snipstad
- Department of Physics; Norwegian University of Science and Technology; Trondheim Norway
| | - Sjoerd Hak
- Department of Physics; Norwegian University of Science and Technology; Trondheim Norway
- Department of Circulation and Medical Imaging; Norwegian University of Science and Technology; Trondheim Norway
| | - Habib Baghirov
- Department of Physics; Norwegian University of Science and Technology; Trondheim Norway
| | - Einar Sulheim
- Department of Physics; Norwegian University of Science and Technology; Trondheim Norway
- SINTEF Materials and Chemistry; Trondheim Norway
| | - Ýrr Mørch
- SINTEF Materials and Chemistry; Trondheim Norway
| | - Sylvie Lélu
- Department of Physics; Norwegian University of Science and Technology; Trondheim Norway
| | - Eva von Haartman
- Pharmaceutical Sciences Laboratory; Åbo Akademi University; Turku Finland
- Laboratory of Physical Chemistry; Åbo Akademi University; Turku Finland
| | - Marcus Bäck
- Department of Physics; Chemistry and Biology, Linköping University; Linköping Sweden
| | - K. Peter R. Nilsson
- Department of Physics; Chemistry and Biology, Linköping University; Linköping Sweden
| | - Andrey S. Klymchenko
- Laboratoire de Biophotonique et Pharmacologie, UMR CNRS 7213, Université de Strasbourg; Strasbourg France
| | | | - Andreas K. O. Åslund
- Department of Physics; Norwegian University of Science and Technology; Trondheim Norway
| |
Collapse
|
47
|
Sulheim E, Baghirov H, von Haartman E, Bøe A, Åslund AKO, Mørch Y, Davies CDL. Cellular uptake and intracellular degradation of poly(alkyl cyanoacrylate) nanoparticles. J Nanobiotechnology 2016; 14:1. [PMID: 26743777 PMCID: PMC4705582 DOI: 10.1186/s12951-015-0156-7] [Citation(s) in RCA: 75] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2015] [Accepted: 12/29/2015] [Indexed: 11/10/2022] Open
Abstract
Background
Poly(alkyl cyanoacrylate) (PACA) nanoparticles have shown promise as drug carriers both to solid tumors and across the blood–brain barrier. Efficient drug delivery requires both high cellular uptake of the nanoparticles and release of the drug from the nanoparticles. Release of hydrophobic drugs from PACA nanoparticles is primarily governed by nanoparticle degradation, and this process has been poorly studied at the cellular level. Here we use the hydrophobic model drug Nile Red 668 (NR668) to investigate intracellular degradation of PACA nanoparticles by measuring changes in NR668 fluorescence emission and lifetime, as the spectral properties of NR668 depend on the hydrophobicity of the dye environment. We also assess the potential of poly(butyl cyanoacrylate) (PBCA) and poly(octyl cyanoacrylate) (POCA) nanoparticles for intracellular drug delivery in the prostate cancer cell line PC3 and rat brain endothelial cell line RBE4 and the role of endocytosis pathways in PACA nanoparticle uptake in those cell lines. Results Fluorescence lifetime imaging, emission spectra analysis and Förster resonance energy transfer indicated that the intracellular degradation was in line with the degradation found by direct methods such as gas chromatography and scanning electron microscopy, showing that PBCA has a faster degradation rate compared to POCA. The combined P(BCA/OCA) nanoparticles had an intermediate degradation rate. The uptake of POCA and PBCA nanoparticles was much higher in RBE4 than in PC3 cells. Endocytosis inhibition studies showed that both clathrin- and caveolin-mediated endocytosis were involved in PACA nanoparticle uptake, and that the former played a predominant role, particularly in PC3 cells. Conclusions In the present study, we used three different optical techniques to show that within a 24-hour period PBCA nanoparticles degraded significantly inside cells, releasing their payload into the cytosol, while POCA nanoparticles remained intact. This indicates that it is possible to tune the intracellular drug release rate by choosing appropriate monomers from the PACA family or by using hybrid PACA nanoparticles containing different monomers. In addition, we showed that the uptake of PACA nanoparticles depends not only on the monomer material, but also on the cell type, and that different cell lines can use different internalization pathways. Electronic supplementary material The online version of this article (doi:10.1186/s12951-015-0156-7) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Einar Sulheim
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| | - Habib Baghirov
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| | - Eva von Haartman
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway. .,Pharmaceutical Sciences Laboratory, Faculty of Natural Sciences and Technology, Åbo Akademi University, Turku, Finland.
| | - Andreas Bøe
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| | - Andreas K O Åslund
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| | - Yrr Mørch
- SINTEF Materials and Chemistry, Trondheim, Norway.
| | - Catharina de Lange Davies
- Department of Physics, The Norwegian University of Science and Technology, NTNU, Høgskoleringen 5, 7491, Trondheim, Norway.
| |
Collapse
|
48
|
Snipstad S, Westrøm S, Mørch Y, Afadzi M, Åslund AK, de Lange Davies C. Contact-mediated intracellular delivery of hydrophobic drugs from polymeric nanoparticles. Cancer Nanotechnol 2014; 5:8. [PMID: 25774230 PMCID: PMC4355425 DOI: 10.1186/s12645-014-0008-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2014] [Accepted: 11/18/2014] [Indexed: 12/25/2022] Open
Abstract
Encapsulation of drugs in nanoparticles can enhance the accumulation of drugs in tumours, reduce toxicity toward healthy tissue, and improve pharmacokinetics compared to administration of free drug. To achieve efficient delivery and release of drugs at the target site, mechanisms of interaction between the nanoparticles and cells and the mechanism of delivery of the encapsulated drug are crucial to understand. Our aim was to determine the mechanisms for cellular uptake of a fluorescent hydrophobic model drug from poly(butylcyanoacrylate) nanoparticles. Prostate adenocarcinoma cells were incubated with Nile Red-loaded nanoparticles or free Nile Red. Uptake and intracellular distribution were evaluated by flow cytometry and confocal laser scanning microscopy. The nanoparticles mediated a higher intracellular level and more rapid uptake of encapsulated Nile Red compared to model drug administered alone. The main mechanism for delivery was not by endocytosis of nanoparticles but by nanoparticle-cell contact-mediated transfer directly to the cytosol and, to a smaller extent, release of payload from nanoparticles into the medium followed by diffusion into cells. The payload thus avoids entering the endocytic pathway, evading lysosomal degradation and instead gains direct access to intracellular targets. The nanoparticles are promising tools for efficient intracellular delivery of hydrophobic anticancer drugs; therefore, they are clinically relevant for improved cancer therapy.
Collapse
Affiliation(s)
- Sofie Snipstad
- Department of Physics, The Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway
| | - Sara Westrøm
- Department of Physics, The Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway
| | - Yrr Mørch
- SINTEF Materials and Chemistry, Trondheim, Norway
| | - Mercy Afadzi
- Department of Physics, The Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway
| | - Andreas Ko Åslund
- Department of Physics, The Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway
| | - Catharina de Lange Davies
- Department of Physics, The Norwegian University of Science and Technology, Høgskoleringen 5, 7491 Trondheim, Norway
| |
Collapse
|