1
|
Bader KB, Padilla F, Haworth KJ, Ellens N, Dalecki D, Miller DL, Wear KA. Overview of Therapeutic Ultrasound Applications and Safety Considerations: 2024 Update. JOURNAL OF ULTRASOUND IN MEDICINE : OFFICIAL JOURNAL OF THE AMERICAN INSTITUTE OF ULTRASOUND IN MEDICINE 2025; 44:381-433. [PMID: 39526313 PMCID: PMC11796337 DOI: 10.1002/jum.16611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/11/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
A 2012 review of therapeutic ultrasound was published to educate researchers and physicians on potential applications and concerns for unintended bioeffects (doi: 10.7863/jum.2012.31.4.623). This review serves as an update to the parent article, highlighting advances in therapeutic ultrasound over the past 12 years. In addition to general mechanisms for bioeffects produced by therapeutic ultrasound, current applications, and the pre-clinical and clinical stages are outlined. An overview is provided for image guidance methods to monitor and assess treatment progress. Finally, other topics relevant for the translation of therapeutic ultrasound are discussed, including computational modeling, tissue-mimicking phantoms, and quality assurance protocols.
Collapse
Affiliation(s)
| | - Frederic Padilla
- Gene Therapy ProgramFocused Ultrasound FoundationCharlottesvilleVirginiaUSA
- Department of RadiologyUniversity of Virginia Health SystemCharlottesvilleVirginiaUSA
| | - Kevin J. Haworth
- Department of PediatricsUniversity of CincinnatiCincinnatiOhioUnited States
- Department of Internal MedicineUniversity of CincinnatiCincinnatiOhioUSA
- Department of Biomedical EngineeringUniversity of CincinnatiCincinnatiOhioUSA
| | | | - Diane Dalecki
- Department of Biomedical EngineeringUniversity of RochesterRochesterNew YorkUSA
| | - Douglas L. Miller
- Department of RadiologyUniversity of Michigan Health SystemAnn ArborMichiganUSA
| | - Keith A. Wear
- Center for Devices and Radiological HealthU.S. Food and Drug AdministrationSilver SpringMarylandUSA
| |
Collapse
|
2
|
Heo D, Kim H, Katagiri W, Yoon C, Lim HG, Kim C, Choi HS, Kashiwagi S, Kim HH. Enhanced model antigen retention in tissue through topical high-frequency ultrasound treatment. Biomed Eng Lett 2025; 15:273-282. [PMID: 40026896 PMCID: PMC11871201 DOI: 10.1007/s13534-024-00445-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 11/05/2024] [Accepted: 11/08/2024] [Indexed: 03/05/2025] Open
Abstract
Reproducible control of tissue delivery and retention of a therapeutic agent facilitates the maximization of the efficacy of pharmacotherapy. Despite the proposal of various chemical and physical methods for modulating drug delivery and retention, the choice of the physical modality for this purpose has been limited in clinical practice. Thus, this study proposes a novel strategy for modulating the retention of a model antigen in tissues by using non-tissue-damaging high-frequency ultrasoundAfter the injection of a fluorescently labeled model antigen, followed by brief treatment with high-frequency ultrasound (5-20 MHz), the clearance of the antigen was monitored using a real-time near-infrared (NIR) fluorescence imaging system in vivo. Further, ultrasound treatment increased tissue retention at the site of model antigen administration. The results suggested that high-frequency ultrasound could change the response to a macromolecule injected into the tissue, such as a vaccine, thereby modulating the immune response. The proposed ultrasound-based technology is translatable to clinical settings and benefits from well-established ultrasound technologies that have been employed in various medical applications for decades. Moreover, this approach can be broadly applied to enhance the therapeutic effects of current and future immune-mediated therapeutic systems.
Collapse
Affiliation(s)
- Dasom Heo
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
| | - Hyunhee Kim
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
| | - Wataru Katagiri
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129 USA
| | - Changhan Yoon
- Department of Biomedical Engineering, Inje University, Gimhae, 50834 Republic of Korea
| | - Hae Gyun Lim
- Department of Biomedical Engineering, Pukyong National University, Busan, 48513 Republic of Korea
| | - Chulhong Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Department of Electrical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Graduate School of Convergence Science and Technology, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
| | - Hak Soo Choi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129 USA
| | - Satoshi Kashiwagi
- Gordon Center for Medical Imaging, Department of Radiology, Massachusetts General Hospital, 149 13th Street, Boston, MA 02129 USA
| | - Hyung Ham Kim
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Department of Electrical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
- Graduate School of Convergence Science and Technology, Pohang University of Science and Technology (POSTECH), Pohang, 37673 Republic of Korea
| |
Collapse
|
3
|
Belyaev IB, Griaznova OY, Yaremenko AV, Deyev SM, Zelepukin IV. Beyond the EPR effect: Intravital microscopy analysis of nanoparticle drug delivery to tumors. Adv Drug Deliv Rev 2025:115550. [PMID: 40021012 DOI: 10.1016/j.addr.2025.115550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/18/2025] [Accepted: 02/18/2025] [Indexed: 03/03/2025]
Abstract
Delivery of nanoparticles (NPs) to solid tumors has long relied on enhanced permeability and retention (EPR) effect, involving permeation of NPs through a leaky vasculature with prolonged retention by reduced lymphatic drainage in tumor. Recent research studies and clinical data challenge EPR concept, revealing alternative pathways and approaches of NP delivery. The area was significantly impacted by the implementation of intravital optical microscopy, unraveling delivery mechanisms at cellular level in vivo. This review presents analysis of the reasons for EPR heterogeneity in tumors and describes non-EPR based concepts for drug delivery, which can supplement the current paradigm. One of the approaches is targeting tumor endothelium by NPs with subsequent intravascular drug release and gradient-driven drug transport to tumor interstitium. Others exploit various immune cells for tumor infiltration and breaking endothelial barriers. Finally, we discuss the involvement of active transcytosis through endothelial cells in NP delivery. This review aims to inspire further understanding of the process of NP extravasation in tumors and provide insights for developing next-generation nanomedicines with improved delivery.
Collapse
Affiliation(s)
- Iaroslav B Belyaev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Eindhoven University of Technology, Eindhoven 5600 MB, the Netherlands
| | - Olga Yu Griaznova
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | | | - Sergey M Deyev
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia
| | - Ivan V Zelepukin
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry of the Russian Academy of Sciences, Moscow, Russia; Department of Immunology, Genetics and Pathology, Uppsala University 75123 Uppsala, Sweden.
| |
Collapse
|
4
|
Kaovasia TP, Duclos S, Gupta D, Kalayeh K, Fabiilli M, Noll DC, Sukovich J, Pandey A, Xu Z, Hall TL. A pre-clinical MRI-guided all-in-one focused ultrasound system for murine brain studies. Sci Rep 2025; 15:144. [PMID: 39747938 PMCID: PMC11696467 DOI: 10.1038/s41598-024-84078-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 12/19/2024] [Indexed: 01/04/2025] Open
Abstract
This paper describes the design and initial proof-of-concept of a single pre-clinical transcranial focused ultrasound (FUS) system capable of performing histotripsy (mechanical ablation), hyperthermia, blood-brain barrier opening (BBBO), sonodynamic therapy, or neuromodulation in a murine brain. We have termed it the All-in-One FUS system for murine brain studies, which is the first FUS system of its kind. The 1.5 MHz ultrasound transducer was fabricated and driven using a custom electronic driver to produce 3-cycle pulses with a focal peak-negative pressure (P-) of up to 87 MPa at a low duty cycle (< 0.1%) for histotripsy as well as 50% duty cycle pulsed-ultrasound with a spatial-peak temporal-average intensity (Ispta) of up to 251 W/cm2 for the other FUS modalities. This All-in-One system can be guided by MRI or stereotactically to maximize its flexibility. To validate the design of the system, histotripsy, BBBO, and hyperthermia were performed in naïve brains of two mice for each modality. Histotripsy and BBBO were performed using MRI-based stereotactic co-registration. The therapeutic effect was confirmed using T2-weighted MR-images for histotripsy, and T1-weighted Gadolinium contrast-enhanced MR-images for BBBO. For hyperthermia, an MRI-compatible insert was designed to fit inside the 80 mm imaging coil of a 7-Tesla small-animal MRI-system, with T2-weighted MR-images used to confirm targeting, and MR-thermometry used to monitor the thermal dose delivered.
Collapse
Affiliation(s)
| | - Sarah Duclos
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Dinank Gupta
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Kourosh Kalayeh
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Mario Fabiilli
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Douglas C Noll
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
| | - Jonathan Sukovich
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Aditya Pandey
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Zhen Xu
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI, USA
| | - Timothy L Hall
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
5
|
Price SEN, Gjennestad MA, Kjelstrup S, Hansen R. The effect of temperature constraints on the treatment of tumors using focused ultrasound-induced acoustic streaming. Sci Rep 2025; 15:49. [PMID: 39747331 PMCID: PMC11697381 DOI: 10.1038/s41598-024-83782-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 12/17/2024] [Indexed: 01/04/2025] Open
Abstract
The transport of drugs into tumor cells near the center of the tumor is known to be severely hindered due to the high interstitial pressure and poor vascularization. The aim of this work is to investigate the possibility to induce acoustic streaming in a tumor. Two tumor cases (breast and abdomen) are simulated to find the acoustic streaming and temperature rise, while varying the focused ultrasound transducer radius, frequency, and power for a constant duty cycle (1%). In the absence of perfusion, the simulated rise in temperature, despite the low duty cycle, never reaches a steady state and is fitted to a logarithmic equation, enabling predictions of the temperature for long treatment times. Higher frequencies and larger probe radii are found to result in shorter treatment times relative to the temperature rise, at the cost of a smaller treated area. Results from the simulations indicate that it may be possible to achieve reasonable acoustic streaming values in tumor without the temperature exceeding 50 °C. Treatment times for streaming a distance of 50 μm in the breast case are shown to range from less than one and a half hour to 93 h, depending on the probe settings.
Collapse
Affiliation(s)
- Sebastian E N Price
- Porelab and Department of Chemistry, The Norwegian University of Science and Technology NTNU, 7491, Trondheim, Norway.
| | | | - Signe Kjelstrup
- Porelab and Department of Chemistry, The Norwegian University of Science and Technology NTNU, 7491, Trondheim, Norway
| | - Rune Hansen
- SINTEF, Department of Health Research and Department of Circulation and Medical Imaging, The Norwegian University of Science and Technology NTNU, 7491, Trondheim, Norway
| |
Collapse
|
6
|
Yazdan M, Naghib SM. Smart Ultrasound-responsive Polymers for Drug Delivery: An Overview on Advanced Stimuli-sensitive Materials and Techniques. Curr Drug Deliv 2025; 22:283-309. [PMID: 38288800 DOI: 10.2174/0115672018283792240115053302] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2023] [Revised: 12/11/2023] [Accepted: 12/28/2023] [Indexed: 04/11/2025]
Abstract
In recent years, a notable advancement has occurred in the domain of drug delivery systems via the integration of intelligent polymers that respond to ultrasound. The implementation of this groundbreaking methodology has significantly revolutionised the controlled and precise delivery of therapeutic interventions. An in-depth investigation is conducted into the most recent developments in ultrasonic stimulus-responsive materials and techniques for the purpose of accomplishing precise medication administration. The investigation begins with an exhaustive synopsis of the foundational principles underlying drug delivery systems that react to ultrasonic stimuli, focusing specifically on the complex interplay between polymers and ultrasound waves. Significant attention is devoted to the development of polymers that demonstrate tailored responsiveness to ultrasound, thereby exemplifying their versatility in generating controlled drug release patterns. Numerous classifications of intelligent polymers are examined in the discussion, including those that react to variations in temperature, pH, and enzymes. When coupled with ultrasonic stimuli, these polymers offer a sophisticated framework for the precise manipulation of drug release in terms of both temporal and spatial dimensions. The present study aims to examine the synergistic effects of responsive polymers and ultrasound in overcoming biological barriers such as the blood-brain barrier and the gastrointestinal tract. By doing so, it seeks to shed light on the potential applications of these materials in intricate clinical scenarios. The issues and future prospects of intelligent ultrasound-responsive polymers in the context of drug delivery are critically analysed in this article. The objective of this study is to offer valuable perspectives on the challenges that must be overcome to enable the effective implementation of these technologies. The primary objective of this comprehensive review is to furnish researchers, clinicians, and pharmaceutical scientists with a wealth of information that will serve as a guide for forthcoming developments in the development and enhancement of intelligent drug delivery systems that employ ultrasound-responsive polymers to attain superior therapeutic outcomes.
Collapse
Affiliation(s)
- Mostafa Yazdan
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| | - Seyed Morteza Naghib
- Department of Nanotechnology, School of Advanced Technologies, Iran University of Science and Technology (IUST), Tehran, 1684613114, Iran
| |
Collapse
|
7
|
Zubair M, Uddin I, Dickinson R, Diederich CJ. Delivering Volumetric Hyperthermia to Head and Neck Cancer Patient-Specific Models Using an Ultrasound Spherical Random Phased Array Transducer. Bioengineering (Basel) 2024; 12:14. [PMID: 39851288 PMCID: PMC11760898 DOI: 10.3390/bioengineering12010014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Revised: 12/10/2024] [Accepted: 12/23/2024] [Indexed: 01/26/2025] Open
Abstract
In exploring adjuvant therapies for head and neck cancer, hyperthermia (40-45 °C) has shown efficacy in enhancing chemotherapy and radiation, as well as the delivery of liposomal drugs. Current hyperthermia treatments, however, struggle to reach large deep tumors uniformly and non-invasively. This study investigates the feasibility of delivering targeted uniform hyperthermia deep into the tissue using a non-invasive ultrasound spherical random phased array transducer. Simulations in 3D patient-specific models for thyroid and oropharyngeal cancers assessed the transducer's proficiency. The transducer consisting of 256 elements randomly positioned on a spherical shell, operated at a frequency of 1 MHz with various phasing schemes and power modulations to analyze 40, 41, and 43 °C isothermal volumes and the penetration depth of the heating volume, along with temperature uniformity within the target area using T10, T50, and T90 temperatures, across different tumor models. Intensity distributions and volumetric temperature contours were calculated to define moderate hyperthermia boundaries. The results indicated the array's ability to produce controlled heating volumes from 1 to 48 cm3 at 40 °C, 0.35 to 27 cm3 at 41 °C, and 0.1 to 8 cm3 at 43 °C. The heating depths ranged from 7 to 39 mm minimum and 52 to 59 mm maximum, measured from the skin's inner surface. The transducer, with optimal phasing and water-cooled bolus, confined the heating to the targeted regions effectively. Multifocal sonications also improved the heating homogeneity, reducing the length-to-diameter ratio by 38% when using eight foci versus a single one. This approach shows potential for treating a range of tumors, notably deep-seated and challenging oropharyngeal cancers.
Collapse
Affiliation(s)
- Muhammad Zubair
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94115, USA
- Department of Neurology and Neurological Sciences, Stanford University, Stanford, CA 94305, USA
| | - Imad Uddin
- Department of Cardiology, Hayat Abad Medical Complex, Peshawar 23301, Pakistan
| | - Robert Dickinson
- Department of Bioengineering, Imperial College London, London SW7 2AZ, UK
| | - Chris J. Diederich
- Department of Radiation Oncology, University of California, San Francisco, San Francisco, CA 94115, USA
| |
Collapse
|
8
|
Li X, Lai Y, Wan G, Zou J, He W, Yang P. Approved natural products-derived nanomedicines for disease treatment. Chin J Nat Med 2024; 22:1100-1116. [PMID: 39725511 DOI: 10.1016/s1875-5364(24)60726-0] [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: 08/12/2024] [Indexed: 12/28/2024]
Abstract
In recent years, there has been an increasing emphasis on exploring innovative drug delivery approaches due to the limitations of conventional therapeutic strategies, such as inadequate drug targeting, insufficient therapeutic efficacy, and significant adverse effects. Nanomedicines have emerged as a promising solution with notable advantages, including extended drug circulation, targeted delivery, and improved bioavailability, potentially enhancing the clinical treatment of various diseases. Natural products/materials-derived nanomedicines, characterized by their natural therapeutic efficacy, superior biocompatibility, and safety profile, play a crucial role in nanomedicine-based treatments. This review provides a comprehensive overview of currently approved natural products-derived nanomedicines, emphasizing the essential properties of natural products-derived drug carriers, their applications in clinical diagnosis and treatment, and the current therapeutic potential and challenges. The aim is to offer guidance for the application and further development of these innovative therapeutic approaches.
Collapse
Affiliation(s)
- Xiaotong Li
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Yaoyao Lai
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Guanghan Wan
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Jiahui Zou
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China
| | - Wei He
- School of Pharmacy, China Pharmaceutical University, Nanjing 2111198, China.
| | - Pei Yang
- School of Science, China Pharmaceutical University, Nanjing 2111198, China.
| |
Collapse
|
9
|
Monck C, Elani Y, Ceroni F. Genetically programmed synthetic cells for thermo-responsive protein synthesis and cargo release. Nat Chem Biol 2024; 20:1380-1386. [PMID: 38969863 PMCID: PMC11427347 DOI: 10.1038/s41589-024-01673-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 06/06/2024] [Indexed: 07/07/2024]
Abstract
Synthetic cells containing genetic programs and protein expression machinery are increasingly recognized as powerful counterparts to engineered living cells in the context of biotechnology, therapeutics and cellular modelling. So far, genetic regulation of synthetic cell activity has been largely confined to chemical stimuli; to unlock their potential in applied settings, engineering stimuli-responsive synthetic cells under genetic regulation is imperative. Here we report the development of temperature-sensitive synthetic cells that control protein production by exploiting heat-responsive mRNA elements. This is achieved by combining RNA thermometer technology, cell-free protein expression and vesicle-based synthetic cell design to create cell-sized capsules able to initiate synthesis of both soluble proteins and membrane proteins at defined temperatures. We show that the latter allows for temperature-controlled cargo release phenomena with potential implications for biomedicine. Platforms like the one presented here can pave the way for customizable, genetically programmed synthetic cells under thermal control to be used in biotechnology.
Collapse
Affiliation(s)
- Carolina Monck
- Department of Chemical Engineering, Imperial College London, London, UK
- Imperial College Centre for Synthetic Biology, London, UK
- fabriCELL, Imperial College London, London, UK
| | - Yuval Elani
- Department of Chemical Engineering, Imperial College London, London, UK.
- Imperial College Centre for Synthetic Biology, London, UK.
- fabriCELL, Imperial College London, London, UK.
| | - Francesca Ceroni
- Department of Chemical Engineering, Imperial College London, London, UK.
- Imperial College Centre for Synthetic Biology, London, UK.
| |
Collapse
|
10
|
Lafond M, Payne A, Lafon C. Therapeutic ultrasound transducer technology and monitoring techniques: a review with clinical examples. Int J Hyperthermia 2024; 41:2389288. [PMID: 39134055 PMCID: PMC11375802 DOI: 10.1080/02656736.2024.2389288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 07/02/2024] [Accepted: 08/01/2024] [Indexed: 09/07/2024] Open
Abstract
The exponential growth of therapeutic ultrasound applications demonstrates the power of the technology to leverage the combinations of transducer technology and treatment monitoring techniques to effectively control the preferred bioeffect to elicit the desired clinical effect.Objective: This review provides an overview of the most commonly used bioeffects in therapeutic ultrasound and describes existing transducer technologies and monitoring techniques to ensure treatment safety and efficacy.Methods and materials: Literature reviews were conducted to identify key choices that essential in terms of transducer design, treatment parameters and procedure monitoring for therapeutic ultrasound applications. Effective combinations of these options are illustrated through descriptions of several clinical indications, including uterine fibroids, prostate disease, liver cancer, and brain cancer, that have been successful in leveraging therapeutic ultrasound to provide effective patient treatments.Results: Despite technological constraints, there are multiple ways to achieve a desired bioeffect with therapeutic ultrasound in a target tissue. Visualizations of the interplay of monitoring modality, bioeffect, and applied acoustic parameters are presented that demonstrate the interconnectedness of the field of therapeutic ultrasound. While the clinical indications explored in this review are at different points in the clinical evaluation path, based on the ever expanding research being conducted in preclinical realms, it is clear that additional clinical applications of therapeutic ultrasound that utilize a myriad of bioeffects will continue to grow and improve in the coming years.Conclusions: Therapeutic ultrasound will continue to improve in the next decades as the combination of transducer technology and treatment monitoring techniques will continue to evolve and be translated in clinical settings, leading to more personalized and efficient therapeutic ultrasound mediated therapies.
Collapse
Affiliation(s)
- Maxime Lafond
- LabTAU, INSERM, Centre Léon Bérard, Université, Lyon, France
| | - Allison Payne
- Department of Radiology and Imaging Sciences, University of UT, Salt Lake City, UT, USA
| | - Cyril Lafon
- LabTAU, INSERM, Centre Léon Bérard, Université, Lyon, France
| |
Collapse
|
11
|
Campea MA, Macdonald C, Hoare T. Supramolecularly-Cross-Linked Nanogel Assemblies for On-Demand, Ultrasound-Triggered Chemotherapy. Biomacromolecules 2024; 25:4697-4714. [PMID: 38995854 DOI: 10.1021/acs.biomac.3c01432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2024]
Abstract
Stimulating the release of small nanoparticles (NPs) from a larger NP via the application of an exogenous stimulus offers the potential to address the different size requirements for circulation versus penetration that hinder chemotherapeutic drug delivery. Herein, we report a size-switching nanoassembly-based drug delivery system comprised of ultrasmall starch nanoparticles (SNPs, ∼20-50 nm major size fraction) encapsulated in a poly(oligo(ethylene glycol) methyl ether methacrylate) nanogel (POEGMA, ∼150 nm major size fraction) cross-linked via supramolecular PEG/α-cyclodextrin (α-CD) interactions. Upon heating the nanogel using a non-invasive, high-intensity focused ultrasound (HIFU) trigger, the thermoresponsive POEGMA-CD nanoassemblies are locally de-cross-linked, inducing in situ release of the highly penetrative drug-loaded SNPs. HIFU triggering increased the release of nanoassembly-loaded DOX from 17 to 37% after 3 h, a result correlated with significantly more effective tumor killing relative to nanoassemblies in the absence of HIFU or drug alone. Furthermore, 1.5× more total fluorescence was observed inside a tumor spheroid when nanoassemblies prepared with fluorophore-labeled SNPs were triggered with HIFU relative to the absence of HIFU. We anticipate this strategy holds promise for delivering tunable doses of chemotherapeutic drugs both at and within a tumor site using a non-invasive triggering approach.
Collapse
Affiliation(s)
- Matthew A Campea
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Cameron Macdonald
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Todd Hoare
- Department of Chemical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
12
|
Adamou P, Harkou E, Villa A, Constantinou A, Dimitratos N. Ultrasonic reactor set-ups and applications: A review. ULTRASONICS SONOCHEMISTRY 2024; 107:106925. [PMID: 38810367 PMCID: PMC11157283 DOI: 10.1016/j.ultsonch.2024.106925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2024] [Revised: 05/01/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Sonochemistry contributes to green science as it uses less hazardous solvents and methods to carry out a reaction. In this review, different reactor designs are discussed in detail providing the necessary knowledge for implementing various processes. The main characteristics of ultrasonic batch systems are their low cost and enhanced mixing; however, they still have immense drawbacks such as their scalability. Continuous flow reactors offer enhanced production yields as the limited cognition which governs the design of these sonoreactors, renders them unusable in industry. In addition, microstructured sonoreactors show improved heat and mass transfer phenomena due to their small size but suffer though from clogging. The optimisation of various conditions of regulations, such as temperature, frequency of ultrasound, intensity of irradiation, sonication time, pressure amplitude and reactor design, it is also discussed to maximise the production rates and yields of reactions taking place in sonoreactors. The optimisation of operating parameters and the selection of the reactor system must be considered to each application's requirements. A plethora of different applications that ultrasound waves can be implemented are in the biochemical and petrochemical engineering, the chemical synthesis of materials, the crystallisation of organic and inorganic substances, the wastewater treatment, the extraction processes and in medicine. Sonochemistry must overcome challenges that consider the scalability of processes and its embodiment into commercial applications, through extensive studies for understanding the designs and the development of computational tools to implement timesaving and efficient theoretical studies.
Collapse
Affiliation(s)
- Panayiota Adamou
- Department of Chemical Engineering Cyprus University of Technology, 57 Corner of Athinon and Anexartisias, 3036 Limassol, Cyprus
| | - Eleana Harkou
- Department of Chemical Engineering Cyprus University of Technology, 57 Corner of Athinon and Anexartisias, 3036 Limassol, Cyprus
| | - Alberto Villa
- Dipartimento di Chimica, Universitá degli Studi di Milano, via Golgi, 20133 Milan, Italy
| | - Achilleas Constantinou
- Department of Chemical Engineering Cyprus University of Technology, 57 Corner of Athinon and Anexartisias, 3036 Limassol, Cyprus.
| | - Nikolaos Dimitratos
- Department of Industrial Chemistry "Toso Montanari", University of Bologna, viale Risorgimento 4, 40136 Bologna, Italy; Center for Chemical Catalysis - C3, University of Bologna, viale Risorgimento 4, 40136 Bologna, Italy.
| |
Collapse
|
13
|
Wang C, Lan X, Zhu L, Wang Y, Gao X, Li J, Tian H, Liang Z, Xu W. Construction Strategy of Functionalized Liposomes and Multidimensional Application. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2309031. [PMID: 38258399 DOI: 10.1002/smll.202309031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 12/30/2023] [Indexed: 01/24/2024]
Abstract
Liposomes are widely used in the biological field due to their good biocompatibility and surface modification properties. With the development of biochemistry and material science, many liposome structures and their surface functional components have been modified and optimized one by one, pushing the liposome platform from traditional to functionalized and intelligent, which will better satisfy and expand the needs of scientific research. However, a main limiting factor effecting the efficiency of liposomes is the complicated environmental conditions in the living body. Currently, in order to overcome the above problem, functionalized liposomes have become a very promising strategy. In this paper, binding strategies of liposomes with four main functional elements, namely nucleic acids, antibodies, peptides, and stimuli-responsive motif have been summarized for the first time. In addition, based on the construction characteristics of functionalized liposomes, such as drug-carrying, targeting, long-circulating, and stimulus-responsive properties, a comprehensive overview of their features and respective research progress are presented. Finally, the paper critically presents the limitations of these functionalized liposomes in the current applications and also prospectively suggests the future development directions, aiming to accelerate realization of their industrialization.
Collapse
Affiliation(s)
- Chengyun Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, 071000, China
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Xinyue Lan
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Longjiao Zhu
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Yanhui Wang
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Xinru Gao
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
| | - Jie Li
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| | - Hongtao Tian
- College of Food Science and Technology, Hebei Agricultural University, Baoding, Hebei, 071000, China
| | - Zhihong Liang
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
| | - Wentao Xu
- College of Food Science and Nutritional Engineering, China Agricultural University, No. 17, Qinghua East Road, Beijing, 100083, China
- Key Laboratory of Precision Nutrition and Food Quality, Beijing Laboratory for Food Quality and Safety, Department of Nutrition and Health, China Agricultural University, Beijing, 100191, China
| |
Collapse
|
14
|
Lammers T. Nanomedicine Tumor Targeting. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2312169. [PMID: 38361435 DOI: 10.1002/adma.202312169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Revised: 01/24/2024] [Indexed: 02/17/2024]
Abstract
Nanomedicines are extensively explored for cancer therapy. By delivering drug molecules more efficiently to pathological sites and by attenuating their accumulation in healthy organs and tissues, nanomedicine formulations aim to improve the balance between drug efficacy and toxicity. More than 20 cancer nanomedicines are approved for clinical use, and hundreds of formulations are in (pre)clinical development. Over the years, several key pitfalls have been identified as bottlenecks in nanomedicine tumor targeting and translation. These go beyond materials- and production-related issues, and particularly also encompass biological barriers and pathophysiological heterogeneity. In this manuscript, the author describes the most important principles, progress, and products in nanomedicine tumor targeting, delineates key current problems and challenges, and discusses the most promising future prospects to create clinical impact.
Collapse
Affiliation(s)
- Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, Center for Biohyhrid Medical Systems, University Hospital RWTH Aachen, Forckenbeckstrasse 55, 52074, Aachen, Germany
| |
Collapse
|
15
|
Luan S, Ji Y, Liu Y, Zhu L, Zhou H, Ouyang J, Yang X, Zhao H, Zhu B. Real-Time Reconstruction of HIFU Focal Temperature Field Based on Deep Learning. BME FRONTIERS 2024; 5:0037. [PMID: 38515637 PMCID: PMC10956737 DOI: 10.34133/bmef.0037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 02/07/2024] [Indexed: 03/23/2024] Open
Abstract
Objective and Impact Statement: High-intensity focused ultrasound (HIFU) therapy is a promising noninvasive method that induces coagulative necrosis in diseased tissues through thermal and cavitation effects, while avoiding surrounding damage to surrounding normal tissues. Introduction: Accurate and real-time acquisition of the focal region temperature field during HIFU treatment marked enhances therapeutic efficacy, holding paramount scientific and practical value in clinical cancer therapy. Methods: In this paper, we initially designed and assembled an integrated HIFU system incorporating diagnostic, therapeutic, and temperature measurement functionalities to collect ultrasound echo signals and temperature variations during HIFU therapy. Furthermore, we introduced a novel multimodal teacher-student model approach, which utilizes the shared self-expressive coefficients and the deep canonical correlation analysis layer to aggregate each modality data, then through knowledge distillation strategies, transfers the knowledge from the teacher model to the student model. Results: By investigating the relationship between the phantoms, in vitro, and in vivo ultrasound echo signals and temperatures, we successfully achieved real-time reconstruction of the HIFU focal 2D temperature field region with a maximum temperature error of less than 2.5 °C. Conclusion: Our method effectively monitored the distribution of the HIFU temperature field in real time, providing scientifically precise predictive schemes for HIFU therapy, laying a theoretical foundation for subsequent personalized treatment dose planning, and providing efficient guidance for noninvasive, nonionizing cancer treatment.
Collapse
Affiliation(s)
- Shunyao Luan
- School of Integrated Circuits, Laboratory for Optoelectronics,
Huazhong University of Science and Technology, Wuhan, China
| | - Yongshuo Ji
- HIFU Center of Oncology Department,
Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Yumei Liu
- HIFU Center of Oncology Department,
Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Linling Zhu
- HIFU Center of Oncology Department,
Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Haoyu Zhou
- School of Integrated Circuits, Laboratory for Optoelectronics,
Huazhong University of Science and Technology, Wuhan, China
| | - Jun Ouyang
- School of Integrated Circuits, Laboratory for Optoelectronics,
Huazhong University of Science and Technology, Wuhan, China
| | - Xiaofei Yang
- School of Integrated Circuits, Laboratory for Optoelectronics,
Huazhong University of Science and Technology, Wuhan, China
| | - Hong Zhao
- HIFU Center of Oncology Department,
Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Benpeng Zhu
- School of Integrated Circuits, Laboratory for Optoelectronics,
Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Paun RA, Jurchuk S, Tabrizian M. A landscape of recent advances in lipid nanoparticles and their translational potential for the treatment of solid tumors. Bioeng Transl Med 2024; 9:e10601. [PMID: 38435821 PMCID: PMC10905562 DOI: 10.1002/btm2.10601] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/30/2023] [Accepted: 09/05/2023] [Indexed: 03/05/2024] Open
Abstract
Lipid nanoparticles (LNPs) are biocompatible drug delivery systems that have found numerous applications in medicine. Their versatile nature enables the encapsulation and targeting of various types of medically relevant molecular cargo, including oligonucleotides, proteins, and small molecules for the treatment of diseases, such as cancer. Cancers that form solid tumors are particularly relevant for LNP-based therapeutics due to the enhanced permeation and retention effect that allows nanoparticles to accumulate within the tumor tissue. Additionally, LNPs can be formulated for both locoregional and systemic delivery depending on the tumor type and stage. To date, LNPs have been used extensively in the clinic to reduce systemic toxicity and improve outcomes in cancer patients by encapsulating chemotherapeutic drugs. Next-generation lipid nanoparticles are currently being developed to expand their use in gene therapy and immunotherapy, as well as to enable the co-encapsulation of multiple drugs in a single system. Other developments include the design of targeted LNPs to specific cells and tissues, and triggerable release systems to control cargo delivery at the tumor site. This review paper highlights recent developments in LNP drug delivery formulations and focuses on the treatment of solid tumors, while also discussing some of their current translational limitations and potential opportunities in the field.
Collapse
Affiliation(s)
- Radu A. Paun
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Sarah Jurchuk
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
| | - Maryam Tabrizian
- Department of Biomedical Engineering, Faculty of Medicine and Health SciencesMcGill UniversityMontrealQuebecCanada
- Faculty of Dentistry and Oral Health SciencesMcGill UniversityMontrealQuebecCanada
| |
Collapse
|
17
|
Gray MD, Spiers L, Coussios CC. Sound speed and attenuation of human pancreas and pancreatic tumors and their influence on focused ultrasound thermal and mechanical therapies. Med Phys 2024; 51:809-825. [PMID: 37477551 DOI: 10.1002/mp.16622] [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: 12/30/2022] [Revised: 06/06/2023] [Accepted: 06/20/2023] [Indexed: 07/22/2023] Open
Abstract
BACKGROUND There is increasing interest in using ultrasound for thermal ablation, histotripsy, and thermal or cavitational enhancement of drug delivery for the treatment of pancreatic cancer. Ultrasonic and thermal modelling conducted as part of the treatment planning process requires acoustic property values for all constituent tissues, but the literature contains no data for the human pancreas. PURPOSE This study presents the first acoustic property measurements of human pancreatic samples and provides examples of how these properties impact a broad range of ultrasound therapies. METHODS Data were collected on human pancreatic tissue samples at physiological temperature from 23 consented patients in cooperation with a hospital pathology laboratory. Propagation of ultrasound over the 2.1-4.5 MHz frequency range through samples of various thicknesses and pathologies was measured using a set of custom-built ultrasonic calipers, with the data processed to estimate sound speed and attenuation. The results were used in acoustic and thermal simulations to illustrate the impacts on extracorporeal ultrasound therapies for mild hyperthermia, thermal ablation, and histotripsy implemented with a CE-marked clinical system operating at 0.96 MHz. RESULTS The mean sound speed and attenuation coefficient values for human samples were well below the range of values in the literature for non-human pancreata, while the human attenuation power law exponents were substantially higher. The simulated impacts on ultrasound mediated therapies for the pancreas indicated that when using the human data instead of the literature average, there was a 30% reduction in median temperature elevation in the treatment volume for mild hyperthermia and 43% smaller volume within a 60°C contour for thermal ablation, all driven by attenuation. By comparison, impacts on boiling and intrinsic threshold histotripsy were minor, with peak pressures changing by less than 15% (positive) and 1% (negative) as a consequence of the counteracting effects of attenuation and sound speed. CONCLUSION This study provides the most complete set of speed of sound and attenuation data available for the human pancreas, and it reiterates the importance of acoustic material properties in the planning and conduct of ultrasound-mediated procedures, particularly thermal therapies.
Collapse
Affiliation(s)
- Michael D Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Laura Spiers
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | | |
Collapse
|
18
|
Moradi Kashkooli F, Hornsby TK, Kolios MC, Tavakkoli JJ. Ultrasound-mediated nano-sized drug delivery systems for cancer treatment: Multi-scale and multi-physics computational modeling. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1913. [PMID: 37475577 DOI: 10.1002/wnan.1913] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 05/18/2023] [Accepted: 05/30/2023] [Indexed: 07/22/2023]
Abstract
Computational modeling enables researchers to study and understand various complex biological phenomena in anticancer drug delivery systems (DDSs), especially nano-sized DDSs (NSDDSs). The combination of NSDDSs and therapeutic ultrasound (TUS), that is, focused ultrasound and low-intensity pulsed ultrasound, has made significant progress in recent years, opening many opportunities for cancer treatment. Multiple parameters require tuning and optimization to develop effective DDSs, such as NSDDSs, in which mathematical modeling can prove advantageous. In silico computational modeling of ultrasound-responsive DDS typically involves a complex framework of acoustic interactions, heat transfer, drug release from nanoparticles, fluid flow, mass transport, and pharmacodynamic governing equations. Owing to the rapid development of computational tools, modeling the different phenomena in multi-scale complex problems involved in drug delivery to tumors has become possible. In the present study, we present an in-depth review of recent advances in the mathematical modeling of TUS-mediated DDSs for cancer treatment. A detailed discussion is also provided on applying these computational models to improve the clinical translation for applications in cancer treatment. This article is categorized under: Nanotechnology Approaches to Biology > Nanoscale Systems in Biology Therapeutic Approaches and Drug Discovery > Nanomedicine for Oncologic Disease.
Collapse
Affiliation(s)
| | - Tyler K Hornsby
- Department of Physics, Toronto Metropolitan University, 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
| | - 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
| |
Collapse
|
19
|
Grutman T, Ilovitsh T. Dense speed-of-sound shift imaging for ultrasonic thermometry. Phys Med Biol 2023; 68:215004. [PMID: 37774710 DOI: 10.1088/1361-6560/acfec3] [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: 06/30/2023] [Accepted: 09/29/2023] [Indexed: 10/01/2023]
Abstract
Objective. Develop a dense algorithm for calculating the speed-of-sound shift between consecutive acoustic acquisitions as a noninvasive means to evaluating temperature change during thermal ablation.Methods. An algorithm for dense speed-of-sound shift imaging (DSI) was developed to simultaneously incorporate information from the entire field of view using a combination of dense optical flow and inverse problem regularization, thus speeding up the calculation and introducing spatial agreement between pixels natively. Thermal ablation monitoring consisted of two main steps: pixel shift tracking using Farneback optical flow, and mathematical modeling of the relationship between the pixel displacement and temperature change as an inverse problem to find the speed-of-sound shift. A calibration constant translates from speed-of-sound shift to temperature change. The method performance was tested inex vivosamples and compared to standard thermal strain imaging (TSI) methods.Main results. Thermal ablation at a frequency of 2 MHz was applied to an agarose phantom that created a speed-of-sound shift measured by an L12-5 imaging transducer. A focal spot was reconstructed by solving the inverse problem. Next, a thermocouple measured the temperature rise during thermal ablation ofex vivochicken breast to calibrate the setup. Temperature changes between 3 °C and 15 °C was measured with high thermometry precision of less than 2 °C error for temperature changes as low as 8 °C. The DSI method outperformed standard TSI in both spatial coherence and runtime in high-intensity focused ultrasound-induced hyperthermia.Significance. Dense ultrasonic speed-of-sound shift imaging can successfully monitor the speed-of-sound shift introduced by thermal ablation. This technique is faster and more robust than current methods, and therefore can be used as a noninvasive, real time and cost-effective thermometry method, with high clinical applicability.
Collapse
Affiliation(s)
- Tal Grutman
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
| | - Tali Ilovitsh
- Department of Biomedical Engineering, Tel Aviv University, Tel Aviv 6997801, Israel
- The Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 6997801, Israel
| |
Collapse
|
20
|
Spiers L, Gray M, Lyon P, Sivakumar S, Bekkali N, Scott S, Collins L, Carlisle R, Wu F, Middleton M, Coussios C. Clinical trial protocol for PanDox: a phase I study of targeted chemotherapy delivery to non-resectable primary pancreatic tumours using thermosensitive liposomal doxorubicin (ThermoDox®) and focused ultrasound. BMC Cancer 2023; 23:896. [PMID: 37741968 PMCID: PMC10517508 DOI: 10.1186/s12885-023-11228-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2022] [Accepted: 07/24/2023] [Indexed: 09/25/2023] Open
Abstract
BACKGROUND The dense stroma of pancreatic ductal adenocarcinomas is a major barrier to drug delivery. To increase the local drug diffusion gradient, high doses of chemotherapeutic agent doxorubicin can be released from thermally-sensitive liposomes (ThermoDox®) using ultrasound-mediated hyperthermia at the tumour target. PanDox is designed as a Phase 1 single centre study to investigate enhancing drug delivery to adult patients with non-operable pancreatic ductal adenocarcinomas. The study compares a single cycle of either conventional doxorubicin alone or ThermoDox® with focused ultrasound-induced hyperthermia for targeted drug release. METHODS Adults with non-resectable pancreatic ductal adenocarcinoma are allocated to receive a single cycle of either doxorubicin alone (Arm A) or ThermoDox® with focused ultrasound-induced hyperthermia (Arm B), based on patient- and tumour-specific safety conditions. Participants in Arm B will undergo a general anaesthetic and pre-heating of the tumour by extra-corporal focused ultrasound (FUS). Rather than employing invasive thermometry, ultrasound parameters are derived from a patient-specific treatment planning model to reach the 41 °C target temperature for drug release. ThermoDox® is then concurrently infused with further ultrasound exposure. Tumour biopsies at the targeted site from all patients are analysed post-treatment using high performance liquid chromatography to quantify doxorubicin delivered to the tumour. The primary endpoint is defined as a statistically significant enhancement in concentration of total intra-tumoural doxorubicin, comparing samples from patients receiving liposomal drug with FUS to free drug alone. Participants are followed for 21 days post-treatment to assess secondary endpoints, including radiological assessment to measure changes in tumour activity by Positron Emission Tomography Response Criteria in Solid Tumours (PERCIST) criteria, adverse events and patient-reported symptoms. DISCUSSION This early phase study builds on previous work targeting tumours in the liver to investigate whether enhancement of chemotherapy delivery using ultrasound-mediated hyperthermia can be translated to the stroma-dense environment of pancreatic ductal adenocarcinoma. If successful, it could herald a new approach towards managing these difficult-to-treat tumours. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT04852367 . Registered 21st April 2022. EudraCT number: 2019-003950-10 (Registered 2019) Iras Project ID: 272253 (Registered 2019) Ethics Number: 20/EE/0284.
Collapse
Affiliation(s)
- Laura Spiers
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
- NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Michael Gray
- NIHR Oxford Biomedical Research Centre, Oxford, UK
- Institute of Biomedical Engineering, University of Oxford, Marcella Wing, Botnar Research Centre, Old Rd, Headington, Oxford, OX3 7LD, UK
| | - Paul Lyon
- Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Shivan Sivakumar
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
- Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Noor Bekkali
- Oxford University Hospitals NHS Foundation Trust, Oxford, OX3 7LE, UK
| | - Shaun Scott
- Nuffield Department of Anaesthetics, John Radcliffe Hospital, Oxford, OX3 7LE, UK
| | - Linda Collins
- Department of Oncology, Oncology Clinical Trials Office (OCTO), University of Oxford, Oxford, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, University of Oxford, Marcella Wing, Botnar Research Centre, Old Rd, Headington, Oxford, OX3 7LD, UK
| | - Feng Wu
- Nuffield Department of Surgery, Churchill Hospital, Oxford, OX3 7LE, UK
| | - Mark Middleton
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Roosevelt Drive, Oxford, OX3 7DQ, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, University of Oxford, Marcella Wing, Botnar Research Centre, Old Rd, Headington, Oxford, OX3 7LD, UK.
| |
Collapse
|
21
|
Lee JY, Oh DY, Lee KH, Lee SH, Lee DH, Kang K, Kang SY, Park DH. Combination of chemotherapy and focused ultrasound for the treatment of unresectable pancreatic cancer: a proof-of-concept study. Eur Radiol 2023; 33:2620-2628. [PMID: 36482217 DOI: 10.1007/s00330-022-09271-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 08/19/2022] [Accepted: 10/25/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To investigate the safety and preliminary efficacy of the combined treatment of focused ultrasound (FUS) and chemotherapy (nab-paclitaxel plus gemcitabine, nPac/Gem) for patients with unresectable pancreatic cancer. METHODS Patients pathologically diagnosed with unresectable pancreatic cancer were included. Low (Isppa = 1.5 kW/cm2), intermediate (2.0 kW/cm2), and high (2.5 kW/cm2) FUS intensity treatment groups were predefined. A 1% duty cycle and the 3+3 scheme were used. Six combined treatments were performed, and adverse events were assessed. Changes in tumor size and tumor response, CA 19-9 level, and patient-reported outcomes at the immediate follow-up (F/U) and/or at the 3-month F/U and survival were evaluated. RESULTS Three participants were enrolled in each intensity group. No adverse device effect or dose-limiting toxicity occurred in any of the participants. Seven of the nine participants experienced a >15% tumor size decrease at the immediate F/U CT and at the 3-month F/U CT. The CA 19-9 level decreased in all of the participants at the immediate F/U. All participants in the intermediate-intensity treatment group showed a > 30% tumor size decrease, partial response, and a significant decrease in the CA 19-9 level at 3-month F/U and longer survival (p < 0.05). CONCLUSION FUS with an intensity of 1.5 to 2.5 kW/cm2 was safe in the combined treatment of FUS and nPac/Gem. Considering the results of the change in tumor size, the change in CA 19-9 level, tumor response, and survival, these FUS parameters can be used for subsequent clinical trials. KEY POINTS • No adverse device effect or dose-limiting toxicity occurred in any of the participants when focused ultrasound with an intensity of 1.5-2.5 kW/cm2 and a low duty cycle of 1% was combined with chemotherapy. • The intermediate-intensity group showed a >30% tumor size decrease, partial response, and a significant decrease in CA 19-9 in all of the participants at the 3-month follow-up and the longest survival. • Any focused ultrasound setting used in this study could be safe and optimal for subsequent clinical trials.
Collapse
Affiliation(s)
- Jae Young Lee
- Department of Radiology and the Institute of Radiation Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Do-Youn Oh
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Cancer Research Institute, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea.
- Integrated Major in Innovative Medical Science, Seoul National University Graduate School, Seoul, 03080, Republic of Korea.
| | - Kyung-Hun Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Sang Hyub Lee
- Department of Gastroenterology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Dong Ho Lee
- Department of Radiology and the Institute of Radiation Medicine, Seoul National University College of Medicine, 103 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Kookjin Kang
- Alpinion Medical Systems, 1F, New Building, 77, Heungan-daero 81beon-gil, Dongan-gu, Anyang-si, Gyeonggi-do, 14117, Republic of Korea
| | - Soo Yeon Kang
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| | - Dong Hyuk Park
- Department of Radiology, Seoul National University Hospital, 101 Daehak-ro, Jongno-gu, Seoul, 03080, Republic of Korea
| |
Collapse
|
22
|
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: 59] [Impact Index Per Article: 29.5] [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
|
23
|
Sharma S, Mahajan SD, Chevli K, Schwartz SA, Aalinkeel R. Nanotherapeutic Approach to Delivery of Chemo- and Gene Therapy for Organ-Confined and Advanced Castration-Resistant Prostate Cancer. Crit Rev Ther Drug Carrier Syst 2023; 40:69-100. [PMID: 37075068 PMCID: PMC11007628 DOI: 10.1615/critrevtherdrugcarriersyst.2022043827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Treatments for late-stage prostate cancer (CaP) have not been very successful. Frequently, advanced CaP progresses to castration-resistant prostate cancer (CRPC), with 50#37;-70% of patients developing bone metastases. CaP with bone metastasis-associated clinical complications and treatment resistance presents major clinical challenges. Recent advances in the formulation of clinically applicable nanoparticles (NPs) have attracted attention in the fields of medicine and pharmacology with applications to cancer and infectious and neurological diseases. NPs have been rendered biocompatible, pose little to no toxicity to healthy cells and tissues, and are engineered to carry large therapeutic payloads, including chemo- and genetic therapies. Additionally, if required, targeting specificity can be achieved by chemically coupling aptamers, unique peptide ligands, or monoclonal antibodies to the surface of NPs. Encapsulating toxic drugs within NPs and delivering them specifically to their cellular targets overcomes the problem of systemic toxicity. Encapsulating highly labile genetic therapeutics such as RNA within NPs provides a protective environment for the payload during parenteral administration. The loading efficiencies of NPs have been maximized while the controlled their therapeutic cargos has been released. Theranostic ("treat and see") NPs have developed combining therapy with imaging capabilities to provide real-time, image-guided monitoring of the delivery of their therapeutic payloads. All of these NP accomplishments have been applied to the nanotherapy of late-stage CaP, offering a new opportunity for a previously dismal prognosis. This article gives an update on current developments in the use of nanotechnology for treating late-stage, castration-resistant CaP.
Collapse
Affiliation(s)
- Satish Sharma
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Supriya D. Mahajan
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Kent Chevli
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Stanley A. Schwartz
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| | - Ravikumar Aalinkeel
- Department of Urology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
- Department of Medicine, Division of Allergy, Immunology and Rheumatology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY
| |
Collapse
|
24
|
Gray M, Spiers L, Coussios C. Effects of human tissue acoustic properties, abdominal wall shape, and respiratory motion on ultrasound-mediated hyperthermia for targeted drug delivery to pancreatic tumors. Int J Hyperthermia 2022; 39:918-934. [PMID: 35853611 PMCID: PMC9612938 DOI: 10.1080/02656736.2022.2091799] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022] Open
Abstract
Background PanDox is a Phase-1 trial of chemotherapeutic drug delivery to pancreatic tumors using ultrasound-mediated hyperthermia to release doxorubicin from thermally sensitive liposomes. This report describes trial-related hyperthermia simulations featuring: (i) new ultrasonic properties of human pancreatic tissues, (ii) abdomen deflections imposed by a water balloon, and (iii) respiration-driven organ motion. Methods Pancreas heating simulations were carried out using three patient body models. Pancreas acoustic properties were varied between values found in the literature and those determined from our human tissue study. Acoustic beam distortion was assessed with and without balloon-induced abdomen deformation. Target heating was assessed for static, normal respiratory, and jet-ventilation-controlled pancreas motion. Results Human pancreatic tumor attenuation is 63% of the literature values, so that pancreas treatments require commensurately higher input intensity to achieve adequate hyperthermia. Abdominal wall deformation decreased the peak field pressure by as much as 3.5 dB and refracted the focal spot by as much as 4.5 mm. These effects were thermally counteracted by sidelobe power deposition, so the net impact on achieving mild hyperthermia was small. Respiratory motion during moving beam hyperthermia produced localized regions overheated by more than 8.0 °C above the 4.0 °C volumetric goal. The use of jet ventilation reduced this excess to 0.7 °C and yielded temperature field uniformity that was nearly identical to having no respiratory motion. Conclusion Realistic modeling of the ultrasonic propagation environment is critical to achieving adequate mild hyperthermia without the use of real time thermometry for targeted drug delivery in pancreatic cancer patients.
Collapse
Affiliation(s)
- Michael Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| | - Laura Spiers
- NIHR Oxford Biomedical Research Centre, Oxford, UK.,Department of Oncology, University of Oxford, Oxford, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford, UK
| |
Collapse
|
25
|
Lee J, Um W, Moon H, Joo H, Song Y, Park M, Yoon B, Kim HR, Park JH. Evading Doxorubicin-Induced Systemic Immunosuppression Using Ultrasound-Responsive Liposomes Combined with Focused Ultrasound. Pharmaceutics 2022; 14:pharmaceutics14122603. [PMID: 36559097 PMCID: PMC9784431 DOI: 10.3390/pharmaceutics14122603] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 11/11/2022] [Accepted: 11/24/2022] [Indexed: 11/29/2022] Open
Abstract
Doxorubicin (DOX) is a representative anticancer drug with a unique ability to induce immunogenic cell death of cancer cells. However, undesired toxicity on immune cells has remained a significant challenge, hindering the usage of DOX in cancer immunotherapy. Here, we report a combined therapy to avoid the off-target toxicity of DOX by adapting ultrasound-responsive liposomal doxorubicin and focused ultrasound exposure. Histological analysis demonstrated that the combined therapy induced less hemosiderosis of splenocytes and improved tumor infiltration of cytotoxic T lymphocytes. Additionally, in vivo therapeutic evaluation results indicate that the combined therapy achieved higher efficacy when combined with PD-1 immune-checkpoint blockade therapy by improving immunogenicity.
Collapse
Affiliation(s)
- Jeongjin Lee
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Seoul 06351, Republic of Korea
| | - Wooram Um
- Department of Biotechnology, Pukyong National University, 45 Yongso-ro, Busan 48513, Republic of Korea
| | - Hyungwon Moon
- R&D Center, IMGT Co., Ltd., 172 Dolma-ro, Seongnam 13605, Republic of Korea
| | - Hyeyeon Joo
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea
| | - Yeari Song
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea
| | - Minsung Park
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea
| | - Been Yoon
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea
| | - Hyun-Ryoung Kim
- R&D Center, IMGT Co., Ltd., 172 Dolma-ro, Seongnam 13605, Republic of Korea
- Correspondence: (H.-R.K.); (J.H.P.)
| | - Jae Hyung Park
- Department of Health Sciences and Technology, SAIHST, Sungkyunkwan University, 81 Irwon-ro, Seoul 06351, Republic of Korea
- School of Chemical Engineering, Sungkyunkwan University, 2066 Seobu-ro, Suwon 16419, Republic of Korea
- Correspondence: (H.-R.K.); (J.H.P.)
| |
Collapse
|
26
|
Barzegar-Fallah A, Gandhi K, Rizwan SB, Slatter TL, Reynolds JNJ. Harnessing Ultrasound for Targeting Drug Delivery to the Brain and Breaching the Blood–Brain Tumour Barrier. Pharmaceutics 2022; 14:pharmaceutics14102231. [PMID: 36297666 PMCID: PMC9607160 DOI: 10.3390/pharmaceutics14102231] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 10/11/2022] [Accepted: 10/17/2022] [Indexed: 11/16/2022] Open
Abstract
Despite significant advances in developing drugs to treat brain tumours, achieving therapeutic concentrations of the drug at the tumour site remains a major challenge due to the presence of the blood–brain barrier (BBB). Several strategies have evolved to enhance brain delivery of chemotherapeutic agents to treat tumours; however, most approaches have several limitations which hinder their clinical utility. Promising studies indicate that ultrasound can penetrate the skull to target specific brain regions and transiently open the BBB, safely and reversibly, with a high degree of spatial and temporal specificity. In this review, we initially describe the basics of therapeutic ultrasound, then detail ultrasound-based drug delivery strategies to the brain and the mechanisms by which ultrasound can improve brain tumour therapy. We review pre-clinical and clinical findings from ultrasound-mediated BBB opening and drug delivery studies and outline current therapeutic ultrasound devices and technologies designed for this purpose.
Collapse
Affiliation(s)
- Anita Barzegar-Fallah
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Kushan Gandhi
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
| | - Shakila B. Rizwan
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- School of Pharmacy, University of Otago, Dunedin 9016, New Zealand
| | - Tania L. Slatter
- Department of Pathology, Dunedin School of Medicine, University of Otago, Dunedin 9016, New Zealand
| | - John N. J. Reynolds
- Department of Anatomy, School of Biomedical Sciences, University of Otago, Dunedin 9016, New Zealand
- Brain Health Research Centre, University of Otago, Dunedin 9016, New Zealand
- Correspondence: ; Tel.: +64-3-479-5781; Fax: +64-3-479-7254
| |
Collapse
|
27
|
Amin M, Lammers T, Ten Hagen TLM. Temperature-sensitive polymers to promote heat-triggered drug release from liposomes: Towards bypassing EPR. Adv Drug Deliv Rev 2022; 189:114503. [PMID: 35998827 DOI: 10.1016/j.addr.2022.114503] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 06/14/2022] [Accepted: 08/17/2022] [Indexed: 01/24/2023]
Abstract
Heat-triggered drug release from temperature-sensitive nanocarriers upon the application of mild hyperthermia is a promising approach to achieve site-specific delivery of drugs. The combination of mild hyperthermia (41-42 °C) and temperature-sensitive liposomes (TSL) that undergo lipid phase-transition and drug release has been studied extensively and has shown promising therapeutic outcome in a variety of animal tumor models as well as initial indications of success in humans. Sensitization of liposomes to mild hyperthermia by means of exploiting the thermal behavior of temperature-sensitive polymers (TSP) provides novel opportunities. Recently, TSP-modified liposomes (TSPL) have shown potential for enhancing tumor-directed drug delivery, either by triggered drug release or by triggered cell interactions in response to heat. In this review, we describe different classes of TSPL, and analyze and discuss the mechanisms and kinetics of content release from TSPL in response to local heating. In addition, the impact of lipid composition, polymer and copolymer characteristics, serum components and PEGylation on the mechanism of content release and TSPL performance is addressed. This is done from the perspective of rationally designing TSPL, with the overall goal of conceiving efficient strategies to increase the efficacy of TSPL plus hyperthermia to improve the outcome of targeted anticancer therapy.
Collapse
Affiliation(s)
- Mohamadreza Amin
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging, RWTH Aachen University Clinic, Center for Biohybrid Medical Systems, Aachen, Germany.
| | - Timo L M Ten Hagen
- Laboratory of Experimental Oncology (LEO), Department of Pathology, Erasmus Medical Center, Rotterdam, The Netherlands; Nanomedicine Innovation Center Erasmus (NICE), Erasmus Medical Center, Rotterdam, The Netherlands.
| |
Collapse
|
28
|
Chen Y, Du M, Yuan Z, Chen Z, Yan F. Spatiotemporal control of engineered bacteria to express interferon-γ by focused ultrasound for tumor immunotherapy. Nat Commun 2022; 13:4468. [PMID: 35918309 PMCID: PMC9345953 DOI: 10.1038/s41467-022-31932-x] [Citation(s) in RCA: 83] [Impact Index Per Article: 27.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 07/11/2022] [Indexed: 11/15/2022] Open
Abstract
Bacteria-based tumor therapy has recently attracted wide attentions due to its unique capability in targeting tumors and preferentially colonizing the core area of the tumor. Various therapeutic genes are also harbored into these engineering bacteria to enhance their anti-tumor efficacy. However, it is difficult to spatiotemporally control the expression of these inserted genes in the tumor site. Here, we engineer an ultrasound-responsive bacterium (URB) which can induce the expression of exogenous genes in an ultrasound-controllable manner. Owing to the advantage of ultrasound in tissue penetration, an acoustic remote control of bacterial gene expression can be realized by designing a temperature-actuated genetic switch. Cytokine interferon-γ (IFN-γ), an important immune regulatory molecule that plays a significant role in tumor immunotherapy, is used to test the system. Our results show that brief hyperthermia induced by focused ultrasound promotes the expression of IFN-γ gene, improving anti-tumor efficacy of URB in vitro and in vivo. Our study provides an alternative strategy for bacteria-mediated tumor immunotherapy.
Collapse
Affiliation(s)
- Yuhao Chen
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Faculty of Health Sciences, Centre for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR, China
| | - Meng Du
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China
| | - Zhen Yuan
- Faculty of Health Sciences, Centre for Cognitive and Brain Sciences, University of Macau, Taipa, Macau SAR, China
| | - Zhiyi Chen
- The First Affiliated Hospital, Medical Imaging Centre, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
- Institute of Medical Imaging, Hengyang Medical School, University of South China, Hengyang, Hunan, 421001, China.
| | - Fei Yan
- Center for Cell and Gene Circuit Design, CAS Key Laboratory of Quantitative Engineering Biology, Shenzhen Institute of Synthetic Biology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
29
|
Vesicular and Planar Membranes of Archaea Lipids: Unusual Physical Properties and Biomedical Applications. Int J Mol Sci 2022; 23:ijms23147616. [PMID: 35886964 PMCID: PMC9319432 DOI: 10.3390/ijms23147616] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 06/27/2022] [Accepted: 06/27/2022] [Indexed: 12/12/2022] Open
Abstract
Liposomes and planar membranes made of archaea or archaea-like lipids exhibit many unusual physical properties compared to model membranes composed of conventional diester lipids. Here, we review several recent findings in this research area, which include (1) thermosensitive archaeosomes with the capability to drastically change the membrane surface charge, (2) MthK channel's capability to insert into tightly packed tetraether black lipid membranes and exhibit channel activity with surprisingly high calcium sensitivity, and (3) the intercalation of apolar squalane into the midplane space of diether bilayers to impede proton permeation. We also review the usage of tetraether archaeosomes as nanocarriers of therapeutics and vaccine adjuvants, as well as the biomedical applications of planar archaea lipid membranes. The discussion on archaeosomal therapeutics is focused on partially purified tetraether lipid fractions such as the polar lipid fraction E (PLFE) and glyceryl caldityl tetraether (GCTE), which are the main components of PLFE with the sugar and phosphate removed.
Collapse
|
30
|
Zhang L, Lin Z, Zeng L, Zhang F, Sun L, Sun S, Wang P, Xu M, Zhang J, Liang X, Ge H. Ultrasound-induced biophysical effects in controlled drug delivery. SCIENCE CHINA. LIFE SCIENCES 2022; 65:896-908. [PMID: 34453275 DOI: 10.1007/s11427-021-1971-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2021] [Accepted: 06/27/2021] [Indexed: 12/30/2022]
Abstract
Ultrasound is widely used in biomedical engineering and has applications in conventional diagnosis and drug delivery. Recent advances in ultrasound-induced drug delivery have been summarized previously in several reviews that have primarily focused on the fabrication of drug delivery carriers. This review discusses the mechanisms underlying ultrasound-induced drug delivery and factors affecting delivery efficiency, including the characteristics of drug delivery carriers and ultrasound parameters. Firstly, biophysical effects induced by ultrasound, namely thermal effects, cavitation effects, and acoustic radiation forces, are illustrated. Secondly, the use of these biophysical effects to enhance drug delivery by affecting drug carriers and corresponding tissues is clarified in detail. Thirdly, recent advances in ultrasound-triggered drug delivery are detailed. Safety issues and optimization strategies to improve therapeutic outcomes and reduce side effects are summarized. Finally, current progress and future directions are discussed.
Collapse
Affiliation(s)
- Lulu Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Zhuohua Lin
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Lan Zeng
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Fan Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Lihong Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Suhui Sun
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Ping Wang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Menghong Xu
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Jinxia Zhang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China
| | - Xiaolong Liang
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| | - Huiyu Ge
- Department of Ultrasound, Peking University Third Hospital, Beijing, 100191, China.
| |
Collapse
|
31
|
Hashemi M, abbasiazam A, Oraee-Yazdani S, Lenzer J. Response of human glioblastoma cells to hyperthermia: Cellular apoptosis and molecular events. Tissue Cell 2022; 75:101751. [DOI: 10.1016/j.tice.2022.101751] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 02/03/2022] [Indexed: 10/19/2022]
|
32
|
Alphandéry E. Ultrasound and nanomaterial: an efficient pair to fight cancer. J Nanobiotechnology 2022; 20:139. [PMID: 35300712 PMCID: PMC8930287 DOI: 10.1186/s12951-022-01243-w] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Accepted: 01/02/2022] [Indexed: 01/12/2023] Open
Abstract
Ultrasounds are often used in cancer treatment protocols, e.g. to collect tumor tissues in the right location using ultrasound-guided biopsy, to image the region of the tumor using more affordable and easier to use apparatus than MRI and CT, or to ablate tumor tissues using HIFU. The efficacy of these methods can be further improved by combining them with various nano-systems, thus enabling: (i) a better resolution of ultrasound imaging, allowing for example the visualization of angiogenic blood vessels, (ii) the specific tumor targeting of anti-tumor chemotherapeutic drugs or gases attached to or encapsulated in nano-systems and released in a controlled manner in the tumor under ultrasound application, (iii) tumor treatment at tumor site using more moderate heating temperatures than with HIFU. Furthermore, some nano-systems display adjustable sizes, i.e. nanobubbles can grow into micro-bubbles. Such dual size is advantageous since it enables gathering within the same unit the targeting properties of nano bubbles via EPR effect and the enhanced ultrasound contrasting properties of micro bubbles. Interestingly, the way in which nano-systems act against a tumor could in principle also be adjusted by accurately selecting the nano-system among a large choice and by tuning the values of the ultrasound parameters, which can lead, due to their mechanical nature, to specific effects such as cavitation that are usually not observed with purely electromagnetic waves and can potentially help destroying the tumor. This review highlights the clinical potential of these combined treatments that can improve the benefit/risk ratio of current cancer treatments.
Collapse
Affiliation(s)
- Edouard Alphandéry
- Sorbonne Université, Muséum National d'Histoire Naturelle, UMR CNRS, 7590, IRD, Institut de Minéralogie, de Physique des Matériaux et de. Cosmochimie, IMPMC, 75005, Paris, France. .,Nanobacterie SARL, 36 boulevard Flandrin, 75116, Paris, France. .,Institute of Anatomy, UZH University of Zurich, Instiute of Anatomy, Winterthurerstrasse 190, 8057, Zurich, Switzerland.
| |
Collapse
|
33
|
Mahmoud K, Swidan S, El-Nabarawi M, Teaima M. Lipid based nanoparticles as a novel treatment modality for hepatocellular carcinoma: a comprehensive review on targeting and recent advances. J Nanobiotechnology 2022; 20:109. [PMID: 35248080 PMCID: PMC8898455 DOI: 10.1186/s12951-022-01309-9] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2022] [Accepted: 02/12/2022] [Indexed: 12/12/2022] Open
Abstract
Liver cancer is considered one of the deadliest diseases with one of the highest disease burdens worldwide. Among the different types of liver cancer, hepatocellular carcinoma is considered to be the most common type. Multiple conventional approaches are being used in treating hepatocellular carcinoma. Focusing on drug treatment, regular agents in conventional forms fail to achieve the intended clinical outcomes. In order to improve the treatment outcomes, utilizing nanoparticles-specifically lipid based nanoparticles-are considered to be one of the most promising approaches being set in motion. Multiple forms of lipid based nanoparticles exist including liposomes, solid lipid nanoparticles, nanostructured lipid carriers, microemulsion, nanoemulsion, phytosomes, lipid coated nanoparticles, and nanoassemblies. Multiple approaches are used to enhance the tumor uptake as well tumor specificity such as intratumoral injection, passive targeting, active targeting, and stimuli responsive nanoparticles. In this review, the effect of utilizing lipidic nanoparticles is being discussed as well as the different tumor uptake enhancement techniques used.
Collapse
Affiliation(s)
- Khaled Mahmoud
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt
| | - Shady Swidan
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt.
- The Center for Drug Research and Development (CDRD), Faculty of Pharmacy, The British University in Egypt, El-Sherouk City, Cairo, 11837, Egypt.
| | - Mohamed El-Nabarawi
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt.
| | - Mahmoud Teaima
- Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, Cairo, 11562, Egypt
| |
Collapse
|
34
|
Mouratidis PXE, ter Haar G. Latest Advances in the Use of Therapeutic Focused Ultrasound in the Treatment of Pancreatic Cancer. Cancers (Basel) 2022; 14:638. [PMID: 35158903 PMCID: PMC8833696 DOI: 10.3390/cancers14030638] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2021] [Revised: 01/17/2022] [Accepted: 01/19/2022] [Indexed: 02/07/2023] Open
Abstract
Traditional oncological interventions have failed to improve survival for pancreatic cancer patients significantly. Novel treatment modalities able to release cancer-specific antigens, render immunologically "cold" pancreatic tumours "hot" and disrupt or reprogram the pancreatic tumour microenvironment are thus urgently needed. Therapeutic focused ultrasound exerts thermal and mechanical effects on tissue, killing cancer cells and inducing an anti-cancer immune response. The most important advances in therapeutic focused ultrasound use for initiation and augmentation of the cancer immunity cycle against pancreatic cancer are described. We provide a comprehensive review of the use of therapeutic focused ultrasound for the treatment of pancreatic cancer patients and describe recent studies that have shown an ultrasound-induced anti-cancer immune response in several tumour models. Published studies that have investigated the immunological effects of therapeutic focused ultrasound in pancreatic cancer are described. This article shows that therapeutic focused ultrasound has been deemed to be a safe technique for treating pancreatic cancer patients, providing pain relief and improving survival rates in pancreatic cancer patients. Promotion of an immune response in the clinic and sensitisation of tumours to the effects of immunotherapy in preclinical models of pancreatic cancer is shown, making it a promising candidate for use in the clinic.
Collapse
Affiliation(s)
- Petros X. E. Mouratidis
- Department of Physics, Division of Radiotherapy and Imaging, The Institute of Cancer Research: Royal Marsden Hospital, Sutton, London SM25NG, UK;
| | | |
Collapse
|
35
|
McLean K, Zhan W. Mathematical modelling of nanoparticle-mediated topical drug delivery to skin tissue. Int J Pharm 2022; 611:121322. [PMID: 34848364 DOI: 10.1016/j.ijpharm.2021.121322] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 11/14/2021] [Accepted: 11/24/2021] [Indexed: 12/11/2022]
Abstract
Nanoparticles have been extensively studied to improve drug delivery outcomes, however, their use in topical delivery remains controversial. Although the feasibility to cross the human skin barrier has been demonstrated in experiments, the risk of low drug concentration in deep tissue still limits the application. In this study, mathematical modelling is employed to examine the performance of nanoparticle-mediated topical delivery for sending drugs into the deep skin tissue. The pharmacokinetic effect is evaluated based on the drug exposure over time. As compared to the delivery using plain drugs, nanoparticle-mediated topical delivery has the potential to significantly improve the drug exposure in deep skin tissue. Modelling predictions denote that the importance of sufficient long-term drug-skin contact in achieving effective drug deposition in the deep skin tissue. The delivery outcomes are highly sensitive to the release rate. Accelerating the release from nanoparticles in stratum corneum is able to improve the drug exposure in stratum corneum and viable epidermis while resulting in the reductions in dermis and blood. The release rate in stratum corneum and viable epidermis should be well-designed below a threshold for generating effective drug accumulation in dermis and blood. A more localised drug accumulation can be achieved in the capillary-rich region of dermis by increasing the local release rate. The release rate in dermis needs to be optimised to increase the drug exposure in the dermis region where there are fewer blood and lymphatics capillaries. Results from this study can be used to improve the regimen of topical delivery for localised treatment.
Collapse
Affiliation(s)
- Kevin McLean
- School of Engineering, King's College, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom
| | - Wenbo Zhan
- School of Engineering, King's College, University of Aberdeen, Aberdeen AB24 3UE, United Kingdom.
| |
Collapse
|
36
|
Holman R, Lorton O, Guillemin PC, Desgranges S, Santini F, Preso DB, Farhat M, Contino-Pépin C, Salomir R. Perfluorocarbon emulsion enhances MR-ARFI displacement and temperature in vitro: Evaluating the response with MRI, NMR, and hydrophone. Front Oncol 2022; 12:1025481. [PMID: 36713528 PMCID: PMC9880467 DOI: 10.3389/fonc.2022.1025481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 11/18/2022] [Indexed: 01/15/2023] Open
Abstract
Sonosensitive perfluorocarbon F8TAC18-PFOB emulsion is under development to enhance heating, increase thermal contrast, and reduce treatment times during focused ultrasound tumor ablation of highly perfused tissue. The emulsion previously showed enhanced heating during ex vivo and in vitro studies. Experiments were designed to observe the response in additional scenarios by varying focused ultrasound conditions, emulsion concentrations, and surfactants. Most notably, changes in acoustic absorption were assessed with MR-ARFI. Phantoms were developed to have thermal, elastic, and relaxometry properties similar to those of ex vivo pig tissue. The phantoms were embedded with varying amounts of F8TAC18-PFOB emulsion or lecithin-PFOB emulsion, between about 0.0-0.3% v:w, in 0.05% v:w increments. MR-ARFI measurements were performed using a FLASH-ARFI-MRT sequence to obtain simultaneous displacement and temperature measurements. A Fabry-Perot hydrophone was utilized to observe the acoustic emissions. Susceptibility-weighted imaging and relaxometry mapping were performed to observe concentration-dependent effects. 19F diffusion-ordered spectroscopy NMR was used to measure the diffusion coefficient of perfluorocarbon droplets in a water emulsion. Increased displacement and temperature were observed with higher emulsion concentration. In semi-rigid MR-ARFI phantoms, a linear response was observed with low-duty cycle MR-ARFI sonications and a mono-exponential saturating response was observed with sustained sonications. The emulsifiers did not have a significant effect on acoustic absorption in semi-rigid gels. Stable cavitation might also contribute to enhanced heating.
Collapse
Affiliation(s)
- Ryan Holman
- Image Guided Interventions Laboratory (GR-949), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Orane Lorton
- Image Guided Interventions Laboratory (GR-949), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Pauline C Guillemin
- Image Guided Interventions Laboratory (GR-949), Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Stéphane Desgranges
- Avignon Université, Equipe Systèmes Amphiphiles bioactifs et Formulations Eco-compatibles, Unité Propre de Recherche et d'Innovation (UPRI), Avignon, France
| | - Francesco Santini
- Department of Biomedical Engineering, University of Basel, Basel, Switzerland
| | - Davide Bernardo Preso
- Institute of Mechanical Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Mohamed Farhat
- Institute of Mechanical Engineering, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Christiane Contino-Pépin
- Avignon Université, Equipe Systèmes Amphiphiles bioactifs et Formulations Eco-compatibles, Unité Propre de Recherche et d'Innovation (UPRI), Avignon, France
| | - Rares Salomir
- Image Guided Interventions Laboratory (GR-949), Faculty of Medicine, University of Geneva, Geneva, Switzerland.,Radiology Department, University Hospitals of Geneva, Geneva, Switzerland
| |
Collapse
|
37
|
Helbert A, von Wronski M, Mestas JL, Tardy I, Bettinger T, Lafon C, Hyvelin JM, Padilla F. Ultrasound Molecular Imaging for the Guidance of Ultrasound-Triggered Release of Liposomal Doxorubicin and Its Treatment Monitoring in an Orthotopic Prostatic Tumor Model in Rat. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:3420-3434. [PMID: 34503895 DOI: 10.1016/j.ultrasmedbio.2021.07.022] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 07/22/2021] [Accepted: 07/25/2021] [Indexed: 06/13/2023]
Abstract
Liposome encapsulation of drugs is an interesting approach in cancer therapy to specifically release the encapsulated drug at the desired treatment site. In addition to thermo-, pH-, light-, enzyme- or redox-responsive liposomes, which have had promising results in (pre-) clinical studies, ultrasound-triggered sonosensitive liposomes represent an exciting alternative to locally trigger the release from these cargos. Localized drug release requires precise tumor visualization to produce a targeted and ultrasound stimulus. We used ultrasound molecular imaging (USMI) with BR55, a vascular endothelial growth factor receptor 2 (VEGFR2)-targeted ultrasound contrast agent, to guide ultrasound-triggered release of sonosensitive liposomes encapsulating doxorubicin (L-DXR) in an orthotopic prostatic rodent tumor model. Forty-eight hours after L-DXR injection, local release of doxorubicin was triggered with a confocal ultrasound device with two focused transducers, 1.1-MHz center frequency, and peak positive and negative pressures of 20.5 and 13 MPa at focus. Tumor size decreased by 20% in 2 wk with L-DXR alone (n = 9) and by 70% after treatment with L-DXR and confocal ultrasound (n = 7) (p < 0.01). The effect of doxorubicin on perfusion/vascularity and VEGFR2 expression was evaluated by USMI and immunohistochemistry of CD31 and VEGFR2 and did not reveal differences in perfusion or VEGFR2 expression in the absence or after the triggered release of liposomes. USMI can provide precise guidance for ultrasound-triggered release of liposomal doxorubicin mediated by a confocal ultrasound device; moreover, the combination of B-mode imaging and USMI can help to follow the response of the tumor to the therapy.
Collapse
Affiliation(s)
- Alexandre Helbert
- Bracco Suisse SA, Bracco Global Research & Development, Geneva, Switzerland.
| | - Mathew von Wronski
- Bracco Suisse SA, Bracco Global Research & Development, Geneva, Switzerland
| | - Jean-Louis Mestas
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, F-69003, Lyon, France
| | - Isabelle Tardy
- Bracco Suisse SA, Bracco Global Research & Development, Geneva, Switzerland
| | - Thierry Bettinger
- Bracco Suisse SA, Bracco Global Research & Development, Geneva, Switzerland
| | - Cyril Lafon
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, F-69003, Lyon, France
| | | | - Frédéric Padilla
- LabTAU, INSERM, Centre Léon Bérard, Université Lyon 1, Univ Lyon, F-69003, Lyon, France; FUS Foundation, Charlottesville, Virginia, USA; Department of Radiology, University of Virginia School of Medicine, Charlottesville, Virginia, USA
| |
Collapse
|
38
|
Wanigasekara J, de Carvalho AMA, Cullen PJ, Tiwari B, Curtin JF. Converging technologies: targeting the hallmarks of cancer using ultrasound and microbubbles. Trends Cancer 2021; 7:886-890. [PMID: 34426143 DOI: 10.1016/j.trecan.2021.07.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 07/26/2021] [Accepted: 07/26/2021] [Indexed: 10/20/2022]
Abstract
Various complex biological effects occur when ultrasonic compression waves travel through biological material. The myriad of biological outcomes instigated by ultrasound are evident when viewed through the lens of the hallmarks of cancer. Herein, we summarise the therapeutic potential of ultrasound, enhanced by microbubbles, for the treatment of cancer.
Collapse
Affiliation(s)
- Janith Wanigasekara
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland; Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland; Environmental Sustainability & Health Institute (ESHI), Technological University Dublin, Dublin, Ireland
| | - Andressa Maria Aguiar de Carvalho
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland; Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland; Environmental Sustainability & Health Institute (ESHI), Technological University Dublin, Dublin, Ireland
| | - Patrick J Cullen
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland; University of Sydney, School of Chemical and Biomolecular Engineering, Sydney, Australia
| | - Brijesh Tiwari
- Department of Food Biosciences, Teagasc Food Research Centre, Ashtown, Dublin, Ireland
| | - James F Curtin
- BioPlasma Research Group, School of Food Science and Environmental Health, Technological University Dublin, Dublin, Ireland; Environmental Sustainability & Health Institute (ESHI), Technological University Dublin, Dublin, Ireland; FOCAS Research Institute, Technological University Dublin, Dublin, Ireland.
| |
Collapse
|
39
|
Zubair M, Adams MS, Diederich CJ. Deployable ultrasound applicators for endoluminal delivery of volumetric hyperthermia. Int J Hyperthermia 2021; 38:1188-1204. [PMID: 34376103 DOI: 10.1080/02656736.2021.1936216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
PURPOSE To investigate the design of an endoluminal deployable ultrasound applicator for delivering volumetric hyperthermia to deep tissue sites as a possible adjunct to radiation and chemotherapy. METHOD This study considers an ultrasound applicator consisting of two tubular transducers situated at the end of a catheter assembly, encased within a distensible conical shaped balloon-based reflector that redirects acoustic energy distally into the tissue. The applicator assembly can be inserted endoluminally or laparoscopically in a compact form and expanded after delivery to the target site. Comprehensive acoustic and biothermal simulations and parametric studies were employed in generalized 3D and patient-specific pancreatic head and body tumor models to characterize the acoustic performance and evaluate heating capabilities of the applicator by investigating the device at a range of operating frequencies, tissue acoustic and thermal properties, transducer configurations, power modulation, applicator positioning, and by analyzing the resultant 40, 41, and 43 °C isothermal volumes and penetration depth of the heating volume. Intensity distributions and volumetric temperature contours were calculated to define moderate hyperthermia boundaries. RESULTS Parametric studies demonstrated the frequency selection to control volume and depth of therapeutic heating from 62 to 22 cm3 and 4 to 2.6 cm as frequency ranges from 1 MHz to 4.7 MHz, respectively. Width of the heating profile tracks closely with the aperture. Water cooling within the reflector balloon was effective in controlling temperature to 37 °C maximum within the luminal wall. Patient-specific studies indicated that applicators with extended OD in the range of 3.6-6.2 cm with 0.5-1 cm long and 1 cm OD transducers can heat volumes of 1.1-7 cm3, 3-26 cm3, and 3.3-37.4 cm3 of pancreatic body and head tumors above 43, 41, and 40 °C, respectively. CONCLUSION In silico studies demonstrated the feasibility of combining endoluminal ultrasound with an integrated expandable balloon reflector for delivering volumetric hyperthermia in regions adjacent to body lumens and cavities.
Collapse
Affiliation(s)
- Muhammad Zubair
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Matthew S Adams
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| | - Chris J Diederich
- Thermal Therapy Research Group, Department of Radiation Oncology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
40
|
Hydralazine augmented ultrasound hyperthermia for the treatment of hepatocellular carcinoma. Sci Rep 2021; 11:15553. [PMID: 34330960 PMCID: PMC8324788 DOI: 10.1038/s41598-021-94323-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Accepted: 07/01/2021] [Indexed: 12/11/2022] Open
Abstract
This study investigates the use of hydralazine to enhance ultrasound hyperthermia for the treatment of hepatocellular carcinoma (HCC) by minimizing flow-mediated heat loss from the tumor. Murine HCC tumors were treated with a continuous mode ultrasound with or without an intravenous administration of hydralazine (5 mg/kg). Tumor blood flow and blood vessels were evaluated by contrast-enhanced ultrasound (CEUS) imaging and histology, respectively. Hydralazine markedly enhanced ultrasound hyperthermia through the disruption of tumor blood flow in HCC. Ultrasound treatment with hydralazine significantly reduced peak enhancement (PE), perfusion index (PI), and area under the curve (AUC) of the CEUS time-intensity curves by 91.9 ± 0.9%, 95.7 ± 0.7%, and 96.6 ± 0.5%, compared to 71.4 ± 1.9%, 84.7 ± 1.1%, and 85.6 ± 0.7% respectively without hydralazine. Tumor temperature measurements showed that the cumulative thermal dose delivered by ultrasound treatment with hydralazine (170.8 ± 11.8 min) was significantly higher than that without hydralazine (137.7 ± 10.7 min). Histological assessment of the ultrasound-treated tumors showed that hydralazine injection formed larger hemorrhagic pools and increased tumor vessel dilation consistent with CEUS observations illustrating the augmentation of hyperthermic effects by hydralazine. In conclusion, we demonstrated that ultrasound hyperthermia can be enhanced significantly by hydralazine in murine HCC tumors by modulating tumor blood flow. Future studies demonstrating the safety of the combined use of ultrasound and hydralazine would enable the clinical translation of the proposed technique.
Collapse
|
41
|
Bienia A, Wiecheć-Cudak O, Murzyn AA, Krzykawska-Serda M. Photodynamic Therapy and Hyperthermia in Combination Treatment-Neglected Forces in the Fight against Cancer. Pharmaceutics 2021; 13:1147. [PMID: 34452108 PMCID: PMC8399393 DOI: 10.3390/pharmaceutics13081147] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/26/2021] [Accepted: 07/16/2021] [Indexed: 12/24/2022] Open
Abstract
Cancer is one of the leading causes of death in humans. Despite the progress in cancer treatment, and an increase in the effectiveness of diagnostic methods, cancer is still highly lethal and very difficult to treat in many cases. Combination therapy, in the context of cancer treatment, seems to be a promising option that may allow minimizing treatment side effects and may have a significant impact on the cure. It may also increase the effectiveness of anti-cancer therapies. Moreover, combination treatment can significantly increase delivery of drugs to cancerous tissues. Photodynamic therapy and hyperthermia seem to be ideal examples that prove the effectiveness of combination therapy. These two kinds of therapy can kill cancer cells through different mechanisms and activate various signaling pathways. Both PDT and hyperthermia play significant roles in the perfusion of a tumor and the network of blood vessels wrapped around it. The main goal of combination therapy is to combine separate mechanisms of action that will make cancer cells more sensitive to a given therapeutic agent. Such an approach in treatment may contribute toward increasing its effectiveness, optimizing the cancer treatment process in the future.
Collapse
Affiliation(s)
| | | | | | - Martyna Krzykawska-Serda
- Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, 30-387 Kraków, Poland; (A.B.); (O.W.-C.); (A.A.M.)
| |
Collapse
|
42
|
Dimitriou P, Li J, Tornillo G, McCloy T, Barrow D. Droplet Microfluidics for Tumor Drug-Related Studies and Programmable Artificial Cells. GLOBAL CHALLENGES (HOBOKEN, NJ) 2021; 5:2000123. [PMID: 34267927 PMCID: PMC8272004 DOI: 10.1002/gch2.202000123] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 03/19/2021] [Indexed: 05/11/2023]
Abstract
Anticancer drug development is a crucial step toward cancer treatment, that requires realistic predictions of malignant tissue development and sophisticated drug delivery. Tumors often acquire drug resistance and drug efficacy, hence cannot be accurately predicted in 2D tumor cell cultures. On the other hand, 3D cultures, including multicellular tumor spheroids (MCTSs), mimic the in vivo cellular arrangement and provide robust platforms for drug testing when grown in hydrogels with characteristics similar to the living body. Microparticles and liposomes are considered smart drug delivery vehicles, are able to target cancerous tissue, and can release entrapped drugs on demand. Microfluidics serve as a high-throughput tool for reproducible, flexible, and automated production of droplet-based microscale constructs, tailored to the desired final application. In this review, it is described how natural hydrogels in combination with droplet microfluidics can generate MCTSs, and the use of microfluidics to produce tumor targeting microparticles and liposomes. One of the highlights of the review documents the use of the bottom-up construction methodologies of synthetic biology for the formation of artificial cellular assemblies, which may additionally incorporate both target cancer cells and prospective drug candidates, as an integrated "droplet incubator" drug assay platform.
Collapse
Affiliation(s)
- Pantelitsa Dimitriou
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - Jin Li
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - Giusy Tornillo
- Hadyn Ellis BuildingCardiff UniversityMaindy RoadCardiffCF24 4HQUK
| | - Thomas McCloy
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| | - David Barrow
- Applied Microfluidic LaboratorySchool of EngineeringCardiff UniversityCardiffCF24 3AAUK
| |
Collapse
|
43
|
Zhao X, Ye Y, Ge S, Sun P, Yu P. Cellular and Molecular Targeted Drug Delivery in Central Nervous System Cancers: Advances in Targeting Strategies. Curr Top Med Chem 2021; 20:2762-2776. [PMID: 32851962 DOI: 10.2174/1568026620666200826122402] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 10/10/2019] [Accepted: 10/16/2019] [Indexed: 12/12/2022]
Abstract
Central nervous system (CNS) cancers are among the most common and treatment-resistant diseases. The main reason for the low treatment efficiency of the disorders is the barriers against targeted delivery of anticancer agents to the site of interest, including the blood-brain barrier (BBB) and blood-brain tumor barrier (BBTB). BBB is a strong biological barrier separating circulating blood from brain extracellular fluid that selectively and actively prevents cytotoxic agents and majority of anticancer drugs from entering the brain. BBB and BBTB are the major impediments against targeted drug delivery into CNS tumors. Nanotechnology and its allied modalities offer interesting and effective delivery strategies to transport drugs across BBB to reach brain tissue. Integrating anticancer drugs into different nanocarriers improves the delivery performance of the resultant compounds across BBB. Surface engineering of nanovehicles using specific ligands, antibodies and proteins enhances the BBB crossing efficacy as well as selective and specific targeting to the target cancerous tissues in CNS tumors. Multifunctional nanoparticles (NPs) have brought revolutionary advances in targeted drug delivery to brain tumors. This study reviews the main anatomical, physiological and biological features of BBB and BBTB in drug delivery and the recent advances in targeting strategies in NPs-based drug delivery for CNS tumors. Moreover, we discuss advances in using specific ligands, antibodies, and surface proteins for designing and engineering of nanocarriers for targeted delivery of anticancer drugs to CNS tumors. Finally, the current clinical applications and the perspectives in the targeted delivery of therapeutic molecules and genes to CNS tumors are discussed.
Collapse
Affiliation(s)
- Xin Zhao
- Department of Pharmacy, Beilun People's Hospital, Ningbo 315800, Zhejiang Province, China
| | - Yun Ye
- Department of Pharmacy, Beilun People's Hospital, Ningbo 315800, Zhejiang Province, China
| | - Shuyu Ge
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| | - Pingping Sun
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| | - Ping Yu
- Department of Pharmacy, Tongde Hospital of Zhejiang Province, Hangzhou 310012, Zhejiang Province, China
| |
Collapse
|
44
|
Cheng B, Bing C, Staruch RM, Shaikh S, Wodzak Staruch M, Szczepanski D, Williams NS, Laetsch TW, Chopra R. The effect of injected dose on localized tumor accumulation and cardiac uptake of doxorubicin in a Vx2 rabbit tumor model using MR-HIFU mild hyperthermia and thermosensitive liposomes. Int J Hyperthermia 2021; 37:1052-1059. [PMID: 32892667 DOI: 10.1080/02656736.2020.1812737] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
PURPOSE When doxorubicin (DOX) is administered via lyso-thermosensitive liposomes (LTLD), mild hyperthermia enhances localized delivery to heated vs. unheated tumors. The optimal LTLD dose and the impact of different doses on systemic drug distribution are unknown. Materials and methods: In this study, we evaluated local and systemic DOX delivery with three LTLD doses (0.1, 0.5, and 2.5 mg/kg) in a Vx2 rabbit tumor model. Temporally and spatially accurate controlled hyperthermia was achieved using a clinical MR-HIFU system for the intended heating duration (40 min). Results: DOX concentration in tissues delivered from LTLD combined with MR-HIFU mild hyperthermia are dose-dependent, including heated/unheated tumor, heart, and other healthy organs. Higher DOX accumulation and tumor-to-heart drug concentration ratio, defined as the ratio of DOX delivered into the tumor vs the heart, were observed in heated tumors compared to unheated tumors in all three tested doses. The DOX uptake efficiency for each mg/kg of LTLD injected IV of heated tumor was significantly higher than that of unheated tumor and heart within the tested dose range (0.1-2.5 mg/kg). The DOX uptake for the heart linearly scaled up as a function of dose while that for the heated tumor showed some evidence of saturation at the high dose of 2.5 mg/kg. Conclusions: These results provide guidance on clinical protocol design of hyperthermia-triggered drug delivery.
Collapse
Affiliation(s)
- Bingbing Cheng
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Chenchen Bing
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Robert M Staruch
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA.,Profound Medical, Mississauga, Canada
| | - Sumbul Shaikh
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Debra Szczepanski
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Noelle S Williams
- Department of Biochemistry, UT Southwestern Medical Center, Dallas, TX, USA
| | - Theodore W Laetsch
- Children's Health, Dallas, TX, USA.,Department of Pediatrics, Division of Hematology-Oncology and Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Rajiv Chopra
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA.,Advanced Imaging Research Center, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
45
|
Tsirkin S, Goldbart R, Traitel T, Kost J. Tailor-Made Single-Core PLGA Microbubbles as Acoustic Cavitation Enhancers for Therapeutic Applications. ACS APPLIED MATERIALS & INTERFACES 2021; 13:25748-25758. [PMID: 34048218 DOI: 10.1021/acsami.1c04770] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/12/2023]
Abstract
Microbubbles (MBs), being gas bubbles encapsulated inside a solid shell, have been investigated extensively in the field of therapeutic ultrasound as acoustic cavitation enhancers. Hard-shell MBs have an advantage over soft-shell MBs due to their improved stability. Poly(lactic-co-glycolic acid) (PLGA) is one of the most attractive polymers for hard-shell MB synthesis; however, very little is known regarding the effect of synthesis parameters on the acoustic cavitation activity of PLGA MBs and the tunability of this activity. In this study, by manipulating the synthesis parameters, we were able to control the characteristics of the MBs, such as their internal structure, gas core, size distribution, and shell thickness, which significantly affect the total acoustic cavitation activity that they exhibit (i.e., their cavitation dose). We showed that single-core MBs filled with C3F8 gas can produce cavitation effects for extended periods under continuous circulation. These MBs exhibited high stability, and their cavitation activity was not affected by prior circulation in the system. Preliminary in vivo results demonstrated that intravenously injected MBs did not cause adverse effects and produced cavitation activity that increased the permeability of the pig blood-brain barrier. Although more tests should be performed to evaluate the MB long-term safety and activity in vivo, these encouraging results suggest that our PLGA MBs have potential for future therapeutic applications as cavitation enhancers.
Collapse
Affiliation(s)
- Shani Tsirkin
- Department of Chemical Engineering, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel
| | - Riki Goldbart
- Department of Chemical Engineering, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel
| | - Tamar Traitel
- Department of Chemical Engineering, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel
| | - Joseph Kost
- Department of Chemical Engineering, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel
- Ilse Katz Institute for Nanoscale Science and Technology, Ben-Gurion University of the Negev, 8410501 Beer Sheva, Israel
| |
Collapse
|
46
|
Dahaghin A, Emadiyanrazavi S, Haghpanahi M, Salimibani M, Bahreinizad H, Eivazzadeh-Keihan R, Maleki A. A comparative study on the effects of increase in injection sites on the magnetic nanoparticles hyperthermia. J Drug Deliv Sci Technol 2021. [DOI: 10.1016/j.jddst.2021.102542] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
47
|
Awad N, Paul V, AlSawaftah NM, ter Haar G, Allen TM, Pitt WG, Husseini GA. Ultrasound-Responsive Nanocarriers in Cancer Treatment: A Review. ACS Pharmacol Transl Sci 2021; 4:589-612. [PMID: 33860189 PMCID: PMC8033618 DOI: 10.1021/acsptsci.0c00212] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Indexed: 12/13/2022]
Abstract
The safe and effective delivery of anticancer agents to diseased tissues is one of the significant challenges in cancer therapy. Conventional anticancer agents are generally cytotoxins with poor pharmacokinetics and bioavailability. Nanocarriers are nanosized particles designed for the selectivity of anticancer drugs and gene transport to tumors. They are small enough to extravasate into solid tumors, where they slowly release their therapeutic load by passive leakage or biodegradation. Using smart nanocarriers, the rate of release of the entrapped therapeutic(s) can be increased, and greater exposure of the tumor cells to the therapeutics can be achieved when the nanocarriers are exposed to certain internally (enzymes, pH, and temperature) or externally (light, magnetic field, and ultrasound) applied stimuli that trigger the release of their load in a safe and controlled manner, spatially and temporally. This review gives a comprehensive overview of recent research findings on the different types of stimuli-responsive nanocarriers and their application in cancer treatment with a particular focus on ultrasound.
Collapse
Affiliation(s)
- Nahid
S. Awad
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Vinod Paul
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Nour M. AlSawaftah
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| | - Gail ter Haar
- Joint
Department of Physics, The Institute of
Cancer Research and The Royal Marsden NHS Foundation Trust, London SM2 5NG, U.K.
| | - Theresa M. Allen
- Department
of Pharmacology, University of Alberta, Edmonton, Alberta T6G 2R3, Canada
| | - William G. Pitt
- Department
of Chemical Engineering, Brigham Young University, Provo, Utah 84602, United States
| | - Ghaleb A. Husseini
- Department
of Chemical Engineering, American University
of Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
48
|
Lyon PC, Mannaris C, Gray M, Carlisle R, Gleeson FV, Cranston D, Wu F, Coussios CC. Large-Volume Hyperthermia for Safe and Cost-Effective Targeted Drug Delivery Using a Clinical Ultrasound-Guided Focused Ultrasound Device. ULTRASOUND IN MEDICINE & BIOLOGY 2021; 47:982-997. [PMID: 33451816 DOI: 10.1016/j.ultrasmedbio.2020.12.008] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/03/2020] [Accepted: 12/10/2020] [Indexed: 06/12/2023]
Abstract
Lyso-thermosensitive liposomes (LTSLs) are specifically designed to release chemotherapy agents under conditions of mild hyperthermia. Preclinical studies have indicated that magnetic resonance (MR)-guided focused ultrasound (FUS) systems can generate well-controlled volumetric hyperthermia using real-time thermometry. However, high-throughput clinical translation of these approaches for drug delivery is challenging, not least because of the significant cost overhead of MR guidance and the much larger volumes that need to be heated clinically. Using an ultrasound-guided extracorporeal clinical FUS device (Chongqing HAIFU, JC200) with thermistors in a non-perfused ex vivo bovine liver tissue model with ribs, we present an optimised strategy for rapidly inducing (5-15 min) and sustaining (>30 min) mild hyperthermia (ΔT <+4°C) in large tissue volumes (≤92 cm3). We describe successful clinical translation in a first-in-human clinical trial of targeted drug delivery of LTSLs (TARDOX: a phase I study to investigate drug release from thermosensitive liposomes in liver tumours), in which targeted tumour hyperthermia resulted in localised chemo-ablation. The heating strategy is potentially applicable to other indications and ultrasound-guided FUS devices.
Collapse
Affiliation(s)
- Paul Christopher Lyon
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK; Nuffield Department of Surgical Sciences, Oxford, UK; Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK.
| | | | - Michael Gray
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, University of Oxford, Oxford, UK
| | - Fergus V Gleeson
- Department of Radiology, Oxford University Hospitals NHS Foundation Trust, Oxford, UK
| | | | - Feng Wu
- Nuffield Department of Surgical Sciences, Oxford, UK
| | | |
Collapse
|
49
|
Lyon PC, Suomi V, Jakeman P, Campo L, Coussios C, Carlisle R. Quantifying cell death induced by doxorubicin, hyperthermia or HIFU ablation with flow cytometry. Sci Rep 2021; 11:4404. [PMID: 33623089 PMCID: PMC7902827 DOI: 10.1038/s41598-021-83845-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2020] [Accepted: 02/04/2021] [Indexed: 12/31/2022] Open
Abstract
Triggered release and targeted drug delivery of potent anti-cancer agents using hyperthermia-mediated focused-ultrasound (FUS) is gaining momentum in the clinical setting. In early phase studies, tissue biopsy samples may be harvested to assess drug delivery efficacy and demonstrate lack of instantaneous cell death due to FUS exposure. We present an optimised tissue cell recovery method and a cell viability assay, compatible with intra-cellular doxorubicin. Flow cytometry was used to determine levels of cell death with suspensions comprised of: (i) HT29 cell line exposed to hyperthermia (30 min at 47 °C) and/or doxorubicin, or ex-vivo bovine liver tissue exposed to (ii) hyperthermia (up to 2 h at 45 °C), or (iii) ablative high intensity FUS (HIFU). Flow cytometric analysis revealed maximal cell death in HT29 receiving both heat and doxorubicin insults and increases in both cell granularity (p < 0.01) and cell death (p < 0.01) in cells recovered from ex-vivo liver tissue exposed to hyperthermia and high pressures of HIFU (8.2 MPa peak-to-peak free-field at 1 MHz) relative to controls. Ex-vivo results were validated with microscopy using pan-cytokeratin stain. This rapid, sensitive and highly quantitative cell-viability method is applicable to the small masses of liver tissue typically recovered from a standard core biopsy (5-20 mg) and may be applied to tissues of other histological origins including immunostaining.
Collapse
Affiliation(s)
- Paul Christopher Lyon
- Nuffield Department of Surgical Sciences, John Radcliffe Hospital, University of Oxford, Oxford, OX3 9DU, UK.
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK.
| | - Visa Suomi
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Philip Jakeman
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Leticia Campo
- Department of Oncology, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Constantin Coussios
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| | - Robert Carlisle
- Institute of Biomedical Engineering, University of Oxford, Old Road Campus Research Building, Oxford, OX3 7DQ, UK
| |
Collapse
|
50
|
Zhang H, Tang WL, Kheirolomoom A, Fite BZ, Wu B, Lau K, Baikoghli M, Raie MN, Tumbale SK, Foiret J, Ingham ES, Mahakian LM, Tam SM, Cheng RH, Borowsky AD, Ferrara KW. Development of thermosensitive resiquimod-loaded liposomes for enhanced cancer immunotherapy. J Control Release 2021; 330:1080-1094. [PMID: 33189786 PMCID: PMC7906914 DOI: 10.1016/j.jconrel.2020.11.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2020] [Revised: 11/01/2020] [Accepted: 11/08/2020] [Indexed: 12/12/2022]
Abstract
Resiquimod (R848) is a toll-like receptor 7 and 8 (TLR7/8) agonist with potent antitumor and immunostimulatory activity. However, systemic delivery of R848 is poorly tolerated because of its poor solubility in water and systemic immune activation. In order to address these limitations, we developed an intravenously-injectable formulation with R848 using thermosensitive liposomes (TSLs) as a delivery vehicle. R848 was remotely loaded into TSLs composed of DPPC: DSPC: DSPE-PEG2K (85:10:5, mol%) with 100 mM FeSO4 as the trapping agent inside. The final R848 to lipid ratio of the optimized R848-loaded TSLs (R848-TSLs) was 0.09 (w/w), 10-fold higher than the previously-reported values. R848-TSLs released 80% of R848 within 5 min at 42 °C. These TSLs were then combined with αPD-1, an immune checkpoint inhibitor, and ultrasound-mediated hyperthermia in a neu deletion (NDL) mouse mammary carcinoma model (Her2+, ER/PR negative). Combined with αPD-1, local injection of R848-TSLs showed superior efficacy with complete NDL tumor regression in both treated and abscopal sites achieved in 8 of 11 tumor bearing mice over 100 days. Immunohistochemistry confirmed enhanced CD8+ T cell infiltration and accumulation by R848-TSLs. Systemic delivery of R848-TSLs, combined with local hyperthermia and αPD-1, inhibited tumor growth and extended median survival from 28 days (non-treatment control) to 94 days. Upon re-challenge with reinjection of tumor cells, none of the previously cured mice developed tumors, as compared with 100% of age-matched control mice. The dose of R848 (10 μg for intra-tumoral injection or 6 mg/kg for intravenous injection delivered up to 4 times) was well-tolerated without weight loss or organ hypertrophy. In summary, we developed R848-TSLs that can be administered locally or systematically, resulting in tumor regression and enhanced survival when combined with αPD-1 in mouse models of breast cancer.
Collapse
Affiliation(s)
- Hua Zhang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Wei-Lun Tang
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Azadeh Kheirolomoom
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA; Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Brett Z Fite
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Bo Wu
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Kenneth Lau
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Mo Baikoghli
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | - Marina Nura Raie
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Spencer K Tumbale
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Josquin Foiret
- Department of Radiology, Stanford University, Palo Alto, CA 94304, USA
| | - Elizabeth S Ingham
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Lisa M Mahakian
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - Sarah M Tam
- Department of Biomedical Engineering, University of California, Davis, CA 95616, USA
| | - R Holland Cheng
- Department of Molecular and Cellular Biology, University of California, Davis, CA 95616, USA
| | | | - Katherine W Ferrara
- Molecular Imaging Program, Department of Radiology, Stanford University, 3165 Porter Drive, Palo Alto, CA 94304, USA.
| |
Collapse
|