Next Article in Journal
A 5-Year Follow-Up after Endovascular Treatment of 402 Intracranial Aneurysms—A Single-Centre Experience
Next Article in Special Issue
Hyperbaric Oxygen Therapy as a Novel Approach to Modulating Macrophage Polarization for the Treatment of Glioblastoma
Previous Article in Journal
The Role of Lipoprotein(a) in Peripheral Artery Disease
Previous Article in Special Issue
High PGC-1α Expression as a Poor Prognostic Indicator in Intracranial Glioma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Microbubble-Enhanced Focused Ultrasound for Infiltrating Gliomas

by
Alexandra A. Seas
1,
Adarsha P. Malla
1,
Nima Sharifai
2,3,
Jeffrey A. Winkles
1,2,
Graeme F. Woodworth
1,2 and
Pavlos Anastasiadis
1,2,*
1
Department of Neurosurgery, University of Maryland School of Medicine, Baltimore, MD 21201, USA
2
Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland Medical Center, Baltimore, MD 21201, USA
3
Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
*
Author to whom correspondence should be addressed.
Biomedicines 2024, 12(6), 1230; https://doi.org/10.3390/biomedicines12061230
Submission received: 3 May 2024 / Revised: 20 May 2024 / Accepted: 28 May 2024 / Published: 1 June 2024
(This article belongs to the Special Issue Gliomas: Signaling Pathways, Molecular Mechanisms and Novel Therapies)

Abstract

:
Infiltrating gliomas are challenging to treat, as the blood-brain barrier significantly impedes the success of therapeutic interventions. While some clinical trials for high-grade gliomas have shown promise, patient outcomes remain poor. Microbubble-enhanced focused ultrasound (MB-FUS) is a rapidly evolving technology with demonstrated safety and efficacy in opening the blood-brain barrier across various disease models, including infiltrating gliomas. Initially recognized for its role in augmenting drug delivery, the potential of MB-FUS to augment liquid biopsy and immunotherapy is gaining research momentum. In this review, we will highlight recent advancements in preclinical and clinical studies that utilize focused ultrasound to treat gliomas and discuss the potential future uses of image-guided precision therapy using focused ultrasound.

1. Introduction

Gliomas are primary brain tumors derived from neuroglial precursor cells and are commonly stratified by histological and molecular features into astrocytomas, oligodendrogliomas, ependymomas, and other/rare types [1]. They are further assigned WHO grades 1–4 based on clinical phenotype and prognosis [2]. Gliomas of WHO grades 3 and 4 are classified as high-grade gliomas (HGGs) and include glioblastoma (GBM) and pediatric gliomas such as diffuse intrinsic pontine glioma (DIPG) [3]. GBM is the most common primary adult malignant glioma, accounting for >50% of all gliomas, and is the most aggressive and heterogeneous glioma—it is uniformly fatal, with a median survival of less than 18 months [1,4,5].
Gliomas are typically diagnosed by tissue biopsy after the onset of neurological symptoms such as seizures or cognitive disorders [6]. Conventional magnetic resonance imaging (MRI) is a critical tool in glioma diagnosis and disease monitoring, as it provides robust structural information. Imaging features such as contrast enhancement, vascularity, and tumor mass effect may be indicators of HGGs; however, these are not specific markers. More advanced imaging techniques such as diffusion-weighted imaging, diffusion tensor imaging, connectomics, spectroscopy, and positron emission tomography have been utilized in clinical practice and clinical trials—these techniques can potentially elucidate functional properties of gliomas [7,8,9]. Despite these advancements, conventional MRI remains the core radiologic diagnostic tool for gliomas.
Standard care for patients with high-grade gliomas is a maximally safe surgical resection with adjuvant radiotherapy targeting the tumor margin and invasive rim. Treatment of GBM often includes adjuvant chemotherapy with the alkylating agent temozolomide (TMZ). While adding TMZ confers some survival benefits for GBM compared to adjuvant radiation alone, it does not result in significant progression-free survival [10,11]. Moreover, GBM is notorious for intrinsic and acquired resistance to chemotherapy and radiation, further limiting the effectiveness of these therapeutic interventions. In recent years, there has been a significant push towards developing novel therapies to enhance survival and methods for improving the drug delivery of currently available therapies [12].
Gliomas, particularly GBM, are challenging to treat for several reasons. First, their infiltrative nature makes complete surgical removal nearly impossible, even with supramarginal resection. Surgical treatment is further complicated by the fact that these tumors frequently invade eloquent brain areas such as the motor and somatosensory cortices, making gross total resection of just the contrast-enhancing components high-risk for neurological deficits [13,14]. Secondly, while immunotherapies have shown promise in treating other malignancies, the unique immune landscape of the brain poses challenges for immunotherapy applications in glioma treatment [15,16]. Finally, the blood-brain barrier (BBB) is a significant impediment to effective glioma treatment. The BBB is a neuro-vascular interface between the systemic blood circulation and the brain parenchyma. The BBB plays a critical role in protecting the central nervous system (CNS) from systemic insults and preserving homeostasis in the brain. However, it also hinders the trafficking of exogenous molecules (e.g., therapeutics) into the brain [17]. BBB function depends on the neurovascular unit comprised of multiple cell types: microvascular endothelial cells, pericytes, and astrocytes (specifically the foot processes). The movement of molecules across the BBB is primarily regulated by tight junctions formed between endothelial cells, which block paracellular transport. Pericytes line this border, forming part of the extracellular matrix. Astrocyte foot processes form a complex network outside the pericytes, which connect the vascular compartment and the neuroglial network. The foot processes significantly maintain ionic and metabolic homeostasis at the BBB [18]. The BBB also contributes to the tight regulation of brain–immune interactions. It shields the brain from circulating toxins and protects fragile brain parenchyma from the potentially deleterious effects of robust systemic immune responses [16,19].
Due to the restrictive BBB and an immunosuppressive tumor microenvironment, a large portion of CNS tumors are shielded from immune surveillance; that is, the host immune system is unable to identify the tumor as foreign failing to mount an effective immune response, even when it is stimulated with pharmaceutic agents [20,21]. Several methods have been developed to bypass the BBB, both to enhance the delivery of therapeutic agents and to allow for immune surveillance of the brain. Focused ultrasound (FUS) is one such method that has garnered significant attention in neuro-oncology over the past two decades.

2. Focused Ultrasound (FUS)

FUS is a rapidly evolving technique in which acoustic waves are targeted to a specific area. FUS was first described as a method for therapeutic tissue heating or thermoablation [22]. Tissue heating is thought to be helpful in the context of treatment-resistant tumors and has shown efficacy in neurological and movement disorders such as Parkinson’s disease. FUS has also been explored as a potential treatment for chronic pain and carcinomatosis and has been utilized as an alternative to lobotomy [23].
The history of FUS for brain pathologies dates back to the 1950s, when the brothers William and Francis Fry developed the first FUS system for creating focal lesions in feline brains. Their novel technique did not affect surrounding tissues or delicate vasculature, mitigating the central issue associated with invasive surgical ablation [24,25]. The Fry brothers also established the reversibility of low-power FUS-mediated neuroanatomical changes [26] and determined threshold ultrasound doses for varying levels of neuromodulation in mammalian brains [27]. These discoveries led the Fry brothers to spearhead clinical studies of FUS to treat brain pathologies [28]. However, their work was limited by the need for a cranial window to limit the phase distortion of the ultrasound waves. The development of phased array systems in the late 1990s enabled the use of phase corrections to overcome aberrations caused by the skull without the need for cranial windows [29], ultimately leading to a substantial increase in neurological research using FUS (Figure 1). A landmark trial applying FUS for thermal ablation in Essential Tremor led to the FDA approval of the first FUS brain device in 2016 [30].
More recently, attention has been given to the potential of FUS for BBB opening (BBBO) in neuro-oncology, specifically with the addition of microbubbles (MB). MBs are micron-sized gas-filled bubbles originally developed as imaging contrast agents. Upon interaction with an acoustic field, MBs undergo oscillations, which, in turn, cause mechanical perturbation of endothelial-cell tight junctions, resulting in transient BBBO (Figure 2). MBs are injected intravascularly into the bloodstream as a single bolus or a continuous infusion during FUS treatments. The interactions between MBs and FUS are tunable depending on the frequency and power of FUS beams and the dose of MBs. Furthermore, the extent of BBBO can be customized depending on the expected result and safety controls.
Landmark studies of microbubble-enhanced FUS (MB-FUS) for BBBO used MRI guidance to treat only certain brain areas—termed MR-guided FUS (MRgFUS). Successful BBBO was confirmed by new signal intensity in the brain parenchyma on contrast-enhancing MRI [34]. This effect was proportional to the MB dose and FUS intensity—a higher dose was analogous to greater signal intensity. Follow-up MRI scans in preclinical and clinical studies have shown that the BBB returns to its baseline within several hours [34,40,41]. Recent studies have indicated the potential of MB-FUS for radiation sensitization in glioma, further expanding the utility of this technology [42].
One of the most common adverse effects of MB-FUS is the formation of microhemorrhages in the brain. This can be seen on MRI as hypointense (dark) puncta on susceptibility-weighted sequences [43]. Microhemorrhages are seen more commonly with increasing ultrasound intensity and increased MB dose, corresponding to increased degree of BBBO. However, studies have shown that despite several areas of brain microhemorrhage, there are no effects on long-term cognition and neurological function [44,45,46]. Comparative MRI imaging shows that these microhemorrhages often resolve; thus, repeated FUS treatments can be performed without permanently damaging brain tissue [47].
Following initial studies indicating reversible BBBO using FUS and MBs, research shifted towards establishing the safety and efficacy of this process. Histological studies showed the expected vascular effects of FUS without ischemia or apoptosis of neuronal cells up to several weeks after treatment, further indicating that FUS can achieve targeted BBBO without damaging the brain [48]. Furthermore, groups have aimed to establish parameters to induce BBBO without tissue heating by altering ultrasound burst length, pulse repetition frequency, and MB dose [49,50]. Kinetic studies with dynamic contrast-enhanced MRI confirmed increased BBB permeability as a function of the ultrasound strength and MB dose compared to untreated groups [51]. Several other studies laid the groundwork for characterizing MB-FUS BBBO by analyzing the effects of phase distortion, cavitation response, pharmacological delivery, MB size, and MB acoustic emissions on BBBO [31,52,53,54,55,56,57].

3. Preclinical Applications of FUS

3.1. Drug Delivery

Given the safety and reversibility of BBBO with FUS, it has been investigated as a potential avenue for enhancing GBM drug delivery. Studies have shown that MB-FUS can increase small-molecule-drug transmembrane transport when it is injected just before or immediately after treatment [58]. Drug delivery across the BBB also depends on the size of the agent to be delivered; particularly small and particularly large agents do not cross the BBB as easily as moderately sized ones (tens of nanometers in diameter) [59]. Thus, FUS provides the potential for glioma therapy with small molecules (nanometers in size), nanoparticles, and even chimeric antigen-receptor T cells [60,61].

3.1.1. Drug Loaded MBs

As previously discussed, BBBO with MB-FUS depends on gas-filled MBs, which oscillate upon interacting with the acoustic energy field (Figure 2). MBs are gas–liquid emulsions with a gaseous core that is stabilized by a surrounding shell comprising lipids, proteins, or other biocompatible molecules. The size of MBs allows for the loading of sufficient drugs of interest and the conjugation of targeting moieties for an additional dimension of targeted delivery [62]. Additionally, drugs can be dissolved within the oil layer of the shell or directly incorporated into the shell, which can protect the cargo from degradation and clearance, as well as minimize off-target toxicity. However, a technical concern exists with drug-loaded MBs in that the disruption of the MB structure is required for drug release, which would only be secondary to inertial cavitation induced by the application of higher ultrasound pressures. This could raise concerns about the unwanted effects of MB cavitation, such as microhemorrhages or gliosis. The majority of these studies apply drug-loaded MBs in conjunction with FUS-BBBO by loading these microspheres with chemotherapeutics such as carmustine (BCNU) [63,64], doxorubicin [65,66], or cabazitaxel [67]. Few of these studies also applied an MB-based targeting strategy in conjunction with drug loading. Most studies found that MB-FUS BBBO combined with drug-loaded MBs increased drug accumulation within the targeted tissue and extended animal survival without inducing unstable microbubble cavitation or hemorrhage.

3.1.2. Free Drugs

With the establishment of MB-FUS to increase drug delivery, initial efforts focused on delivering compounds with known pharmacologic properties across the BBB. Seminal studies utilized TMZ, the standard-of-care chemotherapy for GBM and other gliomas. These studies showed that FUS treatment before drug administration improved the accumulation and retention of TMZ in the tumor space, reduced tumor progression, and enhanced survival in animals [68,69,70]. MB-FUS has also opened a door for exploring pharmacological agents that previously were ineffective at treating gliomas. These include platinum agents such as carboplatin, anthracyclines such as doxorubicin, and several others [71,72,73,74,75]. These trials indicated that administration of MB-FUS treatment, as well as administration of the drug of choice, enhanced survival and reduced progression of gliomas, further confirming the challenge of using the BBB to treat CNS tumors. Given promising results in preclinical trials, several of these drugs are currently being explored in clinical trials of FUS for glioma treatment [76,77,78].

3.1.3. Nanomedicines

Nanomaterials have been explored in drug delivery for several years and have been utilized for varying purposes, including improving drug solubility and biodistribution, decreasing degradation, and targeting drugs to areas of interest. These highly customizable systems allow for more robust control of drug release into the tumor environment [79]. Nanostructures were previously underutilized in the treatment of glioma because standard formulations are too large or possess morphological features that limit their ability to bypass the BBB regardless of targeting strategies [80]. The development of MB-FUS BBBO expanded the utility of nanomaterials for glioma treatment. Many trials have focused on novel methods of packaging known and verified anticancer drugs to increase their stability and provide a method of controlled and prolonged drug release—this includes liposomal, polymeric, and peptide encapsulation. Studies using these methods showed increased antitumor efficacy and prolonged survival in animal glioma models [81,82,83]. Other than drug encapsulation, nanostructures can also be used as drug conjugates to increase penetration and targeting to the brain. Promising conjugates in glioma treatment include inorganic compounds such as gold nanoparticles and organic conjugates such as polymers of hyaluronic acid, albumin, and engineered DNA structures [84,85,86,87].

3.1.4. CAR-T Cells

Adoptive immunotherapies, such as CAR-T cells, have revolutionized cancer treatment for certain hematological malignancies and solid tumors. However, the success of CAR-T cells for primary brain tumors has been minimal due to several challenges associated with GBMs, such as the BBB and an immunosuppressive microenvironment. Only one study to date has reported the effects of MB-FUS on CAR-T cell trafficking, persistence, and efficacy [88]. This study directly compared CAR-T cell delivery and persistence with and without MB-FUS BBBO and observed increased delivery with MB-FUS BBBO at 24 and 72 h after administration. Additionally, in tissues treated with MB-FUS BBBO, they observed an enrichment of the CAR-T cells 15 days after treatment. This improved delivery also had biological significance, as CAR-T cells delivered with MB-FUS BBBO extended median survival by >50 days compared to the no-MB-FUS BBBO control group. Still, as has been observed in humans and preclinical studies with CAR-T cells, neurotoxicity, and immune-related adverse effects are serious concerns. Additional preclinical studies will be critical to determine whether MB-FUS can enhance the delivery and efficacy of this immunotherapy approach while reducing off-target effects.

3.2. Sono-Liquid Biopsy

FUS has been explored more recently in the context of augmenting liquid biopsy (LBx). This is a method that involves the collection of patient blood and analysis for biomarkers. The use of LBx in oncology has expanded dramatically in the last 10 years for early disease detection, therapy guidance, residual disease, and outcome monitoring [89]. In brain tumors, the utility of LBx has been impeded by the BBB, which prevents tumor DNA and tumor-specific biomarkers from entering the circulation. Recent studies have shown that MB-FUS BBBO treatment can increase plasma levels of brain-specific biomarkers and tumor-associated DNA in mouse and porcine GBM models [90,91,92]. Data from these studies support the clinical translation of MB-FUS BBBO-mediated LBx to diagnosis or disease monitoring [93], and a prospective clinical trial has proven the safety and efficacy of this process, as well as proving that sonobiopsy increased levels of circulating tumor DNA in HGG patients [94].

3.3. Immune System Modulation

The effects of FUS on the immune microenvironment of the brain have also opened new avenues for research in the context of immune-system modulation. It was previously understood that brain tumors such as gliomas are generally shielded from the immune system [95]. This limits the use of immunotherapies that have been impactful in other areas of oncology, such as immune checkpoint inhibitors, adoptive T-cell therapy, and therapeutic vaccines [96]. MB-FUS BBBO treatments have been shown to induce mild and transient neuroinflammation in animal models, indicating that the process of opening the BBB may allow for immune surveillance of the brain tissue [97]. This may convert the previously immunosuppressive tumor microenvironment to an immune-activated one [98]. In addition to its transient inflammatory effects, FUS treatment in a mouse glioma model has been shown to upregulate specific markers of innate and adaptive immunity within the tumor microenvironment, even showing dependence on FUS intensity and MB dose [99,100]. When combined with known immunotherapies such as anti-PD1, IL12, and anti-CD47, FUS improved antitumor immune response and, in some cases, extended survival in glioma animal models [88,101,102].

3.4. Sonodynamic Therapy

A novel application of FUS is selective acoustic activation of therapeutics (sonodynamic therapy (SDT). SDT utilizes sonosensitizing agents, which are delivered to the tumor microenvironment and which, when activated by FUS waves, have therapeutic effects. This has the potential to limit the off-target toxicities of some glioma treatments and provide a different avenue for noninvasive treatment of gliomas [103,104]. Early studies have focused on using 5-aminolevulinic acid, fluorescein, and TMZ as sonosensitizers in murine and porcine glioma models. These have yielded promising preliminary data for increased accumulation of the sonosensitizer within the tumor microenvironment, as well as evidence of safety—animals were not found to have damage to surrounding brain tissue following SDT [105,106,107,108,109].

4. Clinical Applications of FUS

Currently, 24 interventional clinical trials registered with ClinicalTrials.gov utilize MB-enhanced focused or unfocused ultrasound to treat gliomas. Most of these trials use one of four FDA-approved clinical-grade ultrasound systems (Table 1). Each system offers a fully customizable spectrum of treatment for each patient, and each is unique in its application.

4.1. Comparison of Clinical FUS Systems

InSightec’s Exablate model 4000 Type II system comprises a phased array helmet-like apparatus with 1024 transducers, which can be tuned with MRI-guidance for transcranial sonication of foci in the brain. While providing anatomical guidance, MRI thermometry is also utilized to track temperature in the focal region for treatment adjustment [110]. In addition to being studied for BBB disruption, the Exablate Type I (650 kHz) system has also been FDA-approved for the treatment of essential tremor and Parkinson’s disease [111,112]. A similar experimental system was first utilized in clinical trials for BBBO in non-human primates and ALS patients, wherein MRgFUS BBB disruption was validated radiographically by gadolinium contrast, and this disruption was resolved mainly within 24 h [44,113]. These studies elucidated the potential role of MRgFUS with the Exablate system for BBB opening and subsequent drug delivery [44]. In the context of glioma, the Exablate Type II (220 kHz) system is being utilized in several clinical trials of GBM and recurrent glioma for safety and feasibility, as well as for the potential increase in drug delivery for these patients following maximal bulk tumor resection.
The NaviFUS system and Cordance Medical NeuroAccess devices utilize multiple elements to facilitate FUS treatment. Both use pre-treatment CT/MRI images with patented protocols to localize the area of interest and upload it to the device. The NaviFUS system uses a multi-channel hemispherical phased array ultrasound connected to a flexible arm and intra-procedure neuronavigation tracking for focal guidance. Furthermore, real-time passive cavitation data are collected to monitor energy levels and patient outcomes [114]. The NeuroAccess system, on the other hand, utilizes the ‘Cordance Cap’, a multi-transducer helmet that is specially fitted to each patient and connected to a monitor. NeuroAccess is also the first system indicated for enhancing liquid biopsy in those with brain pathologies [94,115].
The CarThera Sonocloud-9 device is unique to the other three systems in that it is implanted into a skull window, harkening back to early FUS methods used to bypass the mechanical barrier of the intact skull. It comprises 9 ultrasound transducers and utilizes fixed low-intensity pulsed ultrasound (LIPU). Phase 1 studies have shown that when it was activated in conjunction with circulating MBs, the Sonocloud-9 device was able to facilitate transient BBBO and increase parenchymal delivery of albumin-bound paclitaxel (Abraxane) and carboplatin. Additionally, imaging showed that the BBB opening diminished within one hour following LIPU treatment, highlighting the transient nature of MB and ultrasound-mediated BBB opening [78].

4.2. Review of Clinical Trials

Current FUS Phase 0 and Phase I clinical trials primarily focus on the safety and efficacy of ultrasound, both in conjunction with standard-of-care therapies and other experimental treatments (Table 2). In each trial, ultrasound treatment is given directly before or following maximal surgical resection with or without adjuvant chemoradiation with alkylating agent TMZ, as well as other therapeutics, including etoposide (inhibits DNA synthesis), panobinostat (causes cell cycle arrest and apoptosis), doxorubicin (anthracycline), carboplatin (alkylating agent), and pembrolizumab (immune checkpoint inhibitor), among others [116].

4.3. Clinical versus Preclinical Advancements

FUS has proven to be a promising technology for enhancing the treatment of gliomas in animal models; however, challenges remain in clinical translation. There is a disconnect between preclinical and clinical glioma research: preclinical models typically focus on treating the primary tumor, whereas the clinical need lies more in post-resection therapies to limit recurrence and enhance adjuvant treatment. Additionally, given the significant degree of cellular and molecular heterogeneity of gliomas, it is challenging to accurately mimic the tumor microenvironment in cell or animal models [119]. Another challenge in HGG treatment and research is that the success of an experimental therapy in preclinical studies is often not predictive of success in human patients. The primary experimental therapies currently being explored for GBM include immunotherapies, such as checkpoint inhibitors and CAR-T cell therapy, viral therapy, angiogenesis inhibitors, and gene therapies. While many experimental therapies have shown promise in preclinical models, few have achieved strong safety or efficacy results in human patients [120].

5. Conclusions and Future Directions

Technological innovations and new FUS systems have continued to advance the applications and implementation of FUS-based therapies for infiltrating gliomas. The existence of a multitude of FUS systems and protocols introduces new challenges in the context of the standardization of treatment settings and efficacy metrics. In the initial clinical MB-FUS trials, the primary focus has been the safety and feasibility of treating larger volumes and repeated cycles, as well as early assessments of drug delivery. As FUS technologies evolve toward clinical implementation, it will be essential to develop methods for treatment standardization and correlate treatments with the desired therapeutic effects. Despite the tremendous progress in the field, challenges continue to exist in the translatability and comparability of preclinical studies. The development of preclinical tools and models reflective of clinical conditions will also be highly valuable in the predictive testing of novel therapeutic combinations.

Author Contributions

Conceptualization, P.A. and A.A.S.; writing—original draft preparation, A.A.S., A.P.M. and P.A.; writing—review and editing, A.A.S., A.P.M., J.A.W., G.F.W., P.A. and N.S.; supervision, P.A. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported in part by the National Cancer Institute of the National Institutes of Health Award Number T32CA154274 (A.P.M.), Seed Funds from the Department of Neurosurgery, University of Maryland School of Medicine, and the American Cancer Society’s Institutional Research Grant IRG-18-160-16 (P.A.).

Acknowledgments

The authors acknowledge the contributions of Tina Wang, Biomedical Illustrator at the Department of Neurosurgery, University of Maryland School of Medicine.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Ostrom, Q.T.; Price, M.; Neff, C.; Cioffi, G.; Waite, K.A.; Kruchko, C.; Barnholtz-Sloan, J.S. CBTRUS Statistical Report: Primary Brain and Other Central Nervous System Tumors Diagnosed in the United States in 2015-2019. Neuro Oncol. 2022, 24, v1–v95. [Google Scholar] [CrossRef] [PubMed]
  2. Louis, D.N.; Perry, A.; Wesseling, P.; Brat, D.J.; Cree, I.A.; Figarella-Branger, D.; Hawkins, C.; Ng, H.K.; Pfister, S.M.; Reifenberger, G.; et al. The 2021 WHO Classification of Tumors of the Central Nervous System: A summary. Neuro Oncol. 2021, 23, 1231–1251. [Google Scholar] [CrossRef] [PubMed]
  3. Panditharatna, E.; Yaeger, K.; Kilburn, L.B.; Packer, R.J.; Nazarian, J. Clinicopathology of diffuse intrinsic pontine glioma and its redefined genomic and epigenomic landscape. Cancer Genet. 2015, 208, 367–373. [Google Scholar] [CrossRef] [PubMed]
  4. Mitchel, S.B.; Michael, W. Gliomas; Elsevier: Amsterdam, The Netherlands, 2016. [Google Scholar]
  5. Mohammed, S.; Dinesan, M.; Ajayakumar, T. Survival and quality of life analysis in glioblastoma multiforme with adjuvant chemoradiotherapy: A retrospective study. Rep. Pract. Oncol. Radiother. 2022, 27, 1026–1036. [Google Scholar] [CrossRef] [PubMed]
  6. Posti, J.P.; Bori, M.; Kauko, T.; Sankinen, M.; Nordberg, J.; Rahi, M.; Frantzén, J.; Vuorinen, V.; Sipilä, J.O.T. Presenting symptoms of glioma in adults. Acta Neurol. Scand. 2015, 131, 88–93. [Google Scholar] [CrossRef]
  7. Baehring, J.M.; Bi, W.L.; Bannykh, S.; Piepmeier, J.M.; Fulbright, R.K. Diffusion MRI in the early diagnosis of malignant glioma. J. Neuro-Oncol. 2007, 82, 221–225. [Google Scholar] [CrossRef] [PubMed]
  8. Drake, L.R.; Hillmer, A.T.; Cai, Z. Approaches to PET Imaging of Glioblastoma. Molecules 2020, 25, 568. [Google Scholar] [CrossRef]
  9. Wang, S.; Kim, S.J.; Poptani, H.; Woo, J.H.; Mohan, S.; Jin, R.; Voluck, M.R.; O’Rourke, D.M.; Wolf, R.L.; Melhem, E.R.; et al. Diagnostic utility of diffusion tensor imaging in differentiating glioblastomas from brain metastases. AJNR Am. J. Neuroradiol. 2014, 35, 928–934. [Google Scholar] [CrossRef]
  10. Lee, J.H.; Wee, C.W. Treatment of Adult Gliomas: A Current Update. Brain Neurorehabil. 2022, 15, e24. [Google Scholar] [CrossRef]
  11. Stupp, R.; Mason, W.P.; van den Bent, M.J.; Weller, M.; Fisher, B.; Taphoorn, M.J.B.; Belanger, K.; Brandes, A.A.; Marosi, C.; Bogdahn, U.; et al. Radiotherapy plus Concomitant and Adjuvant Temozolomide for Glioblastoma. N. Engl. J. Med. 2005, 352, 987–996. [Google Scholar] [CrossRef]
  12. Stupp, R.; Taillibert, S.; Kanner, A.; Read, W.; Steinberg, D.; Lhermitte, B.; Toms, S.; Idbaih, A.; Ahluwalia, M.S.; Fink, K.; et al. Effect of Tumor-Treating Fields Plus Maintenance Temozolomide vs Maintenance Temozolomide Alone on Survival in Patients With Glioblastoma: A Randomized Clinical Trial. JAMA 2017, 318, 2306–2316. [Google Scholar] [CrossRef] [PubMed]
  13. Esmaeili, M.; Stensjøen, A.L.; Berntsen, E.M.; Solheim, O.; Reinertsen, I. The Direction of Tumour Growth in Glioblastoma Patients. Sci. Rep. 2018, 8, 1199. [Google Scholar] [CrossRef] [PubMed]
  14. Wei, Y.; Li, C.; Cui, Z.; Mayrand, R.C.; Zou, J.; Wong, A.L.K.C.; Sinha, R.; Matys, T.; Schönlieb, C.-B.; Price, S.J. Structural connectome quantifies tumour invasion and predicts survival in glioblastoma patients. Brain 2022, 146, 1714–1727. [Google Scholar] [CrossRef] [PubMed]
  15. Castellani, G.; Croese, T.; Peralta Ramos, J.M.; Schwartz, M. Transforming the understanding of brain immunity. Science 2023, 380, eabo7649. [Google Scholar] [CrossRef] [PubMed]
  16. Engelhardt, B.; Vajkoczy, P.; Weller, R.O. The movers and shapers in immune privilege of the CNS. Nat. Immunol. 2017, 18, 123–131. [Google Scholar] [CrossRef] [PubMed]
  17. Arvanitis, C.D.; Ferraro, G.B.; Jain, R.K. The blood–brain barrier and blood–tumour barrier in brain tumours and metastases. Nat. Rev. Cancer 2020, 20, 26–41. [Google Scholar] [CrossRef] [PubMed]
  18. Abbott, N.J.; Patabendige, A.A.; Dolman, D.E.; Yusof, S.R.; Begley, D.J. Structure and function of the blood-brain barrier. Neurobiol. Dis. 2010, 37, 13–25. [Google Scholar] [CrossRef] [PubMed]
  19. Forrester, J.V.; McMenamin, P.G.; Dando, S.J. CNS infection and immune privilege. Nat. Rev. Neurosci. 2018, 19, 655–671. [Google Scholar] [CrossRef] [PubMed]
  20. Liu, E.K.; Sulman, E.P.; Wen, P.Y.; Kurz, S.C. Novel Therapies for Glioblastoma. Curr. Neurol. Neurosci. Rep. 2020, 20, 19. [Google Scholar] [CrossRef]
  21. Noorani, I.; Sidlauskas, K.; Pellow, S.; Savage, R.; Norman, J.L.; Chatelet, D.S.; Fabian, M.; Grundy, P.; Ching, J.; Nicoll, J.A.R.; et al. Clinical impact of anti-inflammatory microglia and macrophage phenotypes at glioblastoma margins. Brain Commun. 2023, 5, fcad176. [Google Scholar] [CrossRef]
  22. Lynn, J.G.; Zwemer, R.L.; Chick, A.J.; Miller, A.E. A new method for the generation and use of focused ultrasound in experimental biology. J. Gen. Physiol. 1942, 26, 179–193. [Google Scholar] [CrossRef] [PubMed]
  23. Lindstrom, P.A. Prefrontal ultrasonic irradiation—A substitute for lobotomy. A.M.A. Arch. Neurol. Psychiatry 1954, 72, 399–425. [Google Scholar] [CrossRef]
  24. Fry, W.J.; Mosberg, W.H.; Barnard, J.W.; Fry, F.J. Production of Focal Destructive Lesions in the Central Nervous System With Ultrasound. J. Neurosurg. 1954, 11, 471–478. [Google Scholar] [CrossRef] [PubMed]
  25. Wulff, V.J.; Fry, W.J.; Tucker, D.; Fry, F.J.; Melton, C. Effects of Ultrasonic Vibrations on Nerve Tissues. Exp. Biol. Med. 1951, 76, 361–366. [Google Scholar] [CrossRef] [PubMed]
  26. Fry, F.J.; Ades, H.W.; Fry, W.J. Production of Reversible Changes in the Central Nervous System by Ultrasound. Science 1958, 127, 83–84. [Google Scholar] [CrossRef] [PubMed]
  27. Fry, F.J.; Kossoff, G.; Eggleton, R.C.; Dunn, F. Threshold Ultrasonic Dosages for Structural Changes in the Mammalian Brain. J. Acoust. Soc. Am. 1970, 48, 1413–1417. [Google Scholar] [CrossRef]
  28. Fry, F.J.; Johnson, L.K. Tumor irradiation with intense ultrasound. Ultrasound Med. Biol. 1978, 4, 337–341. [Google Scholar] [CrossRef] [PubMed]
  29. Daum, D.R.; Buchanan, M.T.; Fjield, T.; Hynynen, K. Design and evaluation of a feedback based phased array system for ultrasound surgery. IEEE Trans. Ultrason. Ferroelectr. Freq. Control 1998, 45, 431–438. [Google Scholar] [CrossRef]
  30. Elias, W.J.; Lipsman, N.; Ondo, W.G.; Ghanouni, P.; Kim, Y.G.; Lee, W.; Schwartz, M.; Hynynen, K.; Lozano, A.M.; Shah, B.B.; et al. A Randomized Trial of Focused Ultrasound Thalamotomy for Essential Tremor. N. Engl. J. Med. 2016, 375, 730–739. [Google Scholar] [CrossRef]
  31. Anastasiadis, P.; Gandhi, D.; Guo, Y.; Ahmed, A.K.; Bentzen, S.M.; Arvanitis, C.; Woodworth, G.F. Localized blood-brain barrier opening in infiltrating gliomas with MRI-guided acoustic emissions-controlled focused ultrasound. Proc. Natl. Acad. Sci. USA 2021, 118, e2103280118. [Google Scholar] [CrossRef]
  32. Guthkelch, A.N.; Carter, L.P.; Cassady, J.R.; Hynynen, K.H.; Iacono, R.P.; Johnson, P.C.; Obbens, E.A.; Roemer, R.B.; Seeger, J.F.; Shimm, D.S.; et al. Treatment of malignant brain tumors with focused ultrasound hyperthermia and radiation: Results of a phase I trial. J. Neuro-Oncol. 1991, 10, 271–284. [Google Scholar] [CrossRef]
  33. Hynynen, K.; Freund, W.R.; Cline, H.E.; Chung, A.H.; Watkins, R.D.; Vetro, J.P.; Jolesz, F.A. A clinical, noninvasive, MR imaging-monitored ultrasound surgery method. Radiographics 1996, 16, 185–195. [Google Scholar] [CrossRef] [PubMed]
  34. Hynynen, K.; McDannold, N.; Vykhodtseva, N.; Jolesz, F.A. Noninvasive MR imaging-guided focal opening of the blood-brain barrier in rabbits. Radiology 2001, 220, 640–646. [Google Scholar] [CrossRef] [PubMed]
  35. Idbaih, A.; Canney, M.; Belin, L.; Desseaux, C.; Vignot, A.; Bouchoux, G.; Asquier, N.; Law-Ye, B.; Leclercq, D.; Bissery, A.; et al. Safety and Feasibility of Repeated and Transient Blood-Brain Barrier Disruption by Pulsed Ultrasound in Patients with Recurrent Glioblastoma. Clin. Cancer Res. 2019, 25, 3793–3801. [Google Scholar] [CrossRef]
  36. Lynn, J.G.; Putnam, T.J. Histology of Cerebral Lesions Produced by Focused Ultrasound. Am. J. Pathol. 1944, 20, 637–649. [Google Scholar] [PubMed]
  37. Ram, Z.; Cohen, Z.R.; Harnof, S.; Tal, S.; Faibel, M.; Nass, D.; Maier, S.E.; Hadani, M.; Mardor, Y. Magnetic resonance imaging-guided, high-intensity focused ultrasound for brain tumor therapy. Neurosurgery 2006, 59, 949–955. [Google Scholar] [CrossRef] [PubMed]
  38. Tobias, J.; Hynynen, K.; Roemer, R.; Guthkelch, A.N.; Fleischer, A.S.; Shively, J. An ultrasound window to perform scanned, focused ultrasound hyperthermia treatments of brain tumors. Med. Phys. 1987, 14, 228–234. [Google Scholar] [CrossRef] [PubMed]
  39. Zheng, H.; Benjamin, I.J.; Basu, S.; Li, Z. Heat shock factor 1-independent activation of dendritic cells by heat shock: Implication for the uncoupling of heat-mediated immunoregulation from the heat shock response. Eur. J. Immunol. 2003, 33, 1754–1762. [Google Scholar] [CrossRef] [PubMed]
  40. Conti, A.; Mériaux, S.; Larrat, B. About the Marty model of blood-brain barrier closure after its disruption using focused ultrasound. Phys. Med. Biol. 2019, 64, 14NT02. [Google Scholar] [CrossRef]
  41. Rezai, A.R.; Ranjan, M.; D’Haese, P.F.; Haut, M.W.; Carpenter, J.; Najib, U.; Mehta, R.I.; Chazen, J.L.; Zibly, Z.; Yates, J.R.; et al. Noninvasive hippocampal blood-brain barrier opening in Alzheimer’s disease with focused ultrasound. Proc. Natl. Acad. Sci. USA 2020, 117, 9180–9182. [Google Scholar] [CrossRef]
  42. Fletcher, S.-M.P.; Chisholm, A.; Lavelle, M.; Guthier, R.; Zhang, Y.; Power, C.; Berbeco, R.; McDannold, N. A study combining microbubble-mediated focused ultrasound and radiation therapy in the healthy rat brain and a F98 glioma model. Sci. Rep. 2024, 14, 4831. [Google Scholar] [CrossRef] [PubMed]
  43. Liu, H.-L.; Wai, Y.-Y.; Chen, W.-S.; Chen, J.-C.; Hsu, P.-H.; Wu, X.-Y.; Huang, W.-C.; Yen, T.-C.; Wang, J.-J. Hemorrhage Detection During Focused-Ultrasound Induced Blood-Brain-Barrier Opening by Using Susceptibility-Weighted Magnetic Resonance Imaging. Ultrasound Med. Biol. 2008, 34, 598–606. [Google Scholar] [CrossRef] [PubMed]
  44. Abrahao, A.; Meng, Y.; Llinas, M.; Huang, Y.; Hamani, C.; Mainprize, T.; Aubert, I.; Heyn, C.; Black, S.E.; Hynynen, K.; et al. First-in-human trial of blood-brain barrier opening in amyotrophic lateral sclerosis using MR-guided focused ultrasound. Nat. Commun. 2019, 10, 4373. [Google Scholar] [CrossRef] [PubMed]
  45. Lipsman, N.; Meng, Y.; Bethune, A.J.; Huang, Y.; Lam, B.; Masellis, M.; Herrmann, N.; Heyn, C.; Aubert, I.; Boutet, A.; et al. Blood–brain barrier opening in Alzheimer’s disease using MR-guided focused ultrasound. Nat. Commun. 2018, 9, 2336. [Google Scholar] [CrossRef] [PubMed]
  46. Mainprize, T.; Lipsman, N.; Huang, Y.; Meng, Y.; Bethune, A.; Ironside, S.; Heyn, C.; Alkins, R.; Trudeau, M.; Sahgal, A.; et al. Blood-Brain Barrier Opening in Primary Brain Tumors with Non-invasive MR-Guided Focused Ultrasound: A Clinical Safety and Feasibility Study. Sci. Rep. 2019, 9, 321. [Google Scholar] [CrossRef] [PubMed]
  47. Kobus, T.; Vykhodtseva, N.; Pilatou, M.; Zhang, Y.; McDannold, N. Safety Validation of Repeated Blood-Brain Barrier Disruption Using Focused Ultrasound. Ultrasound Med. Biol. 2016, 42, 481–492. [Google Scholar] [CrossRef]
  48. McDannold, N.; Vykhodtseva, N.; Raymond, S.; Jolesz, F.A.; Hynynen, K. MRI-guided targeted blood-brain barrier disruption with focused ultrasound: Histological findings in rabbits. Ultrasound Med. Biol. 2005, 31, 1527–1537. [Google Scholar] [CrossRef] [PubMed]
  49. Choi, J.J.; Selert, K.; Gao, Z.; Samiotaki, G.; Baseri, B.; Konofagou, E.E. Noninvasive and localized blood-brain barrier disruption using focused ultrasound can be achieved at short pulse lengths and low pulse repetition frequencies. J. Cereb. Blood Flow. Metab. 2011, 31, 725–737. [Google Scholar] [CrossRef]
  50. McDannold, N.; Vykhodtseva, N.; Hynynen, K. Effects of Acoustic Parameters and Ultrasound Contrast Agent Dose on Focused-Ultrasound Induced Blood-Brain Barrier Disruption. Ultrasound Med. Biol. 2008, 34, 930–937. [Google Scholar] [CrossRef]
  51. Vlachos, F.; Tung, Y.S.; Konofagou, E.E. Permeability assessment of the focused ultrasound-induced blood-brain barrier opening using dynamic contrast-enhanced MRI. Phys. Med. Biol. 2010, 55, 5451–5466. [Google Scholar] [CrossRef] [PubMed]
  52. Choi, J.J.; Feshitan, J.A.; Baseri, B.; Wang, S.; Tung, Y.S.; Borden, M.A.; Konofagou, E.E. Microbubble-size dependence of focused ultrasound-induced blood-brain barrier opening in mice in vivo. IEEE Trans. Biomed. Eng. 2010, 57, 145–154. [Google Scholar] [CrossRef] [PubMed]
  53. Choi, J.J.; Pernot, M.; Small, S.A.; Konofagou, E.E. Noninvasive, transcranial and localized opening of the blood-brain barrier using focused ultrasound in mice. Ultrasound Med. Biol. 2007, 33, 95–104. [Google Scholar] [CrossRef]
  54. Choi, J.J.; Wang, S.; Tung, Y.S.; Morrison, B., III; Konofagou, E.E. Molecules of various pharmacologically-relevant sizes can cross the ultrasound-induced blood-brain barrier opening in vivo. Ultrasound Med. Biol. 2010, 36, 58–67. [Google Scholar] [CrossRef] [PubMed]
  55. McDannold, N.; Vykhodtseva, N.; Hynynen, K. Blood-Brain Barrier Disruption Induced by Focused Ultrasound and Circulating Preformed Microbubbles Appears to Be Characterized by the Mechanical Index. Ultrasound Med. Biol. 2008, 34, 834–840. [Google Scholar] [CrossRef]
  56. Tung, Y.S.; Choi, J.J.; Baseri, B.; Konofagou, E.E. Identifying the inertial cavitation threshold and skull effects in a vessel phantom using focused ultrasound and microbubbles. Ultrasound Med. Biol. 2010, 36, 840–852. [Google Scholar] [CrossRef] [PubMed]
  57. Tung, Y.S.; Vlachos, F.; Choi, J.J.; Deffieux, T.; Selert, K.; Konofagou, E.E. In vivo transcranial cavitation threshold detection during ultrasound-induced blood-brain barrier opening in mice. Phys. Med. Biol. 2010, 55, 6141–6155. [Google Scholar] [CrossRef]
  58. Arvanitis, C.D.; Askoxylakis, V.; Guo, Y.; Datta, M.; Kloepper, J.; Ferraro, G.B.; Bernabeu, M.O.; Fukumura, D.; McDannold, N.; Jain, R.K. Mechanisms of enhanced drug delivery in brain metastases with focused ultrasound-induced blood-tumor barrier disruption. Proc. Natl. Acad. Sci. USA 2018, 115, E8717–E8726. [Google Scholar] [CrossRef] [PubMed]
  59. Ohta, S.; Kikuchi, E.; Ishijima, A.; Azuma, T.; Sakuma, I.; Ito, T. Investigating the optimum size of nanoparticles for their delivery into the brain assisted by focused ultrasound-induced blood-brain barrier opening. Sci. Rep. 2020, 10, 18220. [Google Scholar] [CrossRef]
  60. Wu, Y.; Liu, Y.; Huang, Z.; Wang, X.; Jin, Z.; Li, J.; Limsakul, P.; Zhu, L.; Allen, M.; Pan, Y.; et al. Control of the activity of CAR-T cells within tumours via focused ultrasound. Nat. Biomed. Eng. 2021, 5, 1336–1347. [Google Scholar] [CrossRef]
  61. Zhang, S.; Zhang, S.; Luo, S.; Tang, P.; Wan, M.; Wu, D.; Gao, W. Ultrasound-assisted brain delivery of nanomedicines for brain tumor therapy: Advance and prospect. J. Nanobiotechnology 2022, 20, 287. [Google Scholar] [CrossRef]
  62. Lentacker, I.; De Smedt, S.; Sanders, N. Drug loaded microbubble design for ultrasound triggered delivery. Soft Matter 2009, 5, 2161–2170. [Google Scholar] [CrossRef]
  63. Fan, C.H.; Ting, C.Y.; Chang, Y.C.; Wei, K.C.; Liu, H.L.; Yeh, C.K. Drug-loaded bubbles with matched focused ultrasound excitation for concurrent blood-brain barrier opening and brain-tumor drug delivery. Acta Biomater. 2015, 15, 89–101. [Google Scholar] [CrossRef] [PubMed]
  64. Ting, C.Y.; Fan, C.H.; Liu, H.L.; Huang, C.Y.; Hsieh, H.Y.; Yen, T.C.; Wei, K.C.; Yeh, C.K. Concurrent blood-brain barrier opening and local drug delivery using drug-carrying microbubbles and focused ultrasound for brain glioma treatment. Biomaterials 2012, 33, 704–712. [Google Scholar] [CrossRef] [PubMed]
  65. Chan, M.H.; Chen, W.; Li, C.H.; Fang, C.Y.; Chang, Y.C.; Wei, D.H.; Liu, R.S.; Hsiao, M. An Advanced In Situ Magnetic Resonance Imaging and Ultrasonic Theranostics Nanocomposite Platform: Crossing the Blood-Brain Barrier and Improving the Suppression of Glioblastoma Using Iron-Platinum Nanoparticles in Nanobubbles. ACS Appl. Mater. Interfaces 2021, 13, 26759–26769. [Google Scholar] [CrossRef] [PubMed]
  66. Fan, C.H.; Cheng, Y.H.; Ting, C.Y.; Ho, Y.J.; Hsu, P.H.; Liu, H.L.; Yeh, C.K. Ultrasound/Magnetic Targeting with SPIO-DOX-Microbubble Complex for Image-Guided Drug Delivery in Brain Tumors. Theranostics 2016, 6, 1542–1556. [Google Scholar] [CrossRef] [PubMed]
  67. Sulheim, E.; Mørch, Y.; Snipstad, S.; Borgos, S.E.; Miletic, H.; Bjerkvig, R.; Davies, C.L.; Åslund, A.K.O. Therapeutic Effect of Cabazitaxel and Blood-Brain Barrier opening in a Patient-Derived Glioblastoma Model. Nanotheranostics 2019, 3, 103–112. [Google Scholar] [CrossRef] [PubMed]
  68. Dong, Q.; He, L.; Chen, L.; Deng, Q. Opening the Blood-Brain Barrier and Improving the Efficacy of Temozolomide Treatments of Glioblastoma Using Pulsed, Focused Ultrasound with a Microbubble Contrast Agent. Biomed. Res. Int. 2018, 2018, 6501508. [Google Scholar] [CrossRef] [PubMed]
  69. Liu, H.L.; Huang, C.Y.; Chen, J.Y.; Wang, H.Y.; Chen, P.Y.; Wei, K.C. Pharmacodynamic and therapeutic investigation of focused ultrasound-induced blood-brain barrier opening for enhanced temozolomide delivery in glioma treatment. PLoS ONE 2014, 9, e114311. [Google Scholar] [CrossRef]
  70. Wei, K.C.; Chu, P.C.; Wang, H.Y.; Huang, C.Y.; Chen, P.Y.; Tsai, H.C.; Lu, Y.J.; Lee, P.Y.; Tseng, I.C.; Feng, L.Y.; et al. Focused ultrasound-induced blood-brain barrier opening to enhance temozolomide delivery for glioblastoma treatment: A preclinical study. PLoS ONE 2013, 8, e58995. [Google Scholar] [CrossRef]
  71. Dréan, A.; Lemaire, N.; Bouchoux, G.; Goldwirt, L.; Canney, M.; Goli, L.; Bouzidi, A.; Schmitt, C.; Guehennec, J.; Verreault, M.; et al. Temporary blood-brain barrier disruption by low intensity pulsed ultrasound increases carboplatin delivery and efficacy in preclinical models of glioblastoma. J. Neuro-Oncol. 2019, 144, 33–41. [Google Scholar] [CrossRef]
  72. Ishida, J.; Alli, S.; Bondoc, A.; Golbourn, B.; Sabha, N.; Mikloska, K.; Krumholtz, S.; Srikanthan, D.; Fujita, N.; Luck, A.; et al. MRI-guided focused ultrasound enhances drug delivery in experimental diffuse intrinsic pontine glioma. J. Control Release 2021, 330, 1034–1045. [Google Scholar] [CrossRef] [PubMed]
  73. Kovacs, Z.; Werner, B.; Rassi, A.; Sass, J.O.; Martin-Fiori, E.; Bernasconi, M. Prolonged survival upon ultrasound-enhanced doxorubicin delivery in two syngenic glioblastoma mouse models. J. Control Release 2014, 187, 74–82. [Google Scholar] [CrossRef] [PubMed]
  74. McDannold, N.; Zhang, Y.; Supko, J.G.; Power, C.; Sun, T.; Peng, C.; Vykhodtseva, N.; Golby, A.J.; Reardon, D.A. Acoustic feedback enables safe and reliable carboplatin delivery across the blood-brain barrier with a clinical focused ultrasound system and improves survival in a rat glioma model. Theranostics 2019, 9, 6284–6299. [Google Scholar] [CrossRef] [PubMed]
  75. Wei, H.J.; Upadhyayula, P.S.; Pouliopoulos, A.N.; Englander, Z.K.; Zhang, X.; Jan, C.I.; Guo, J.; Mela, A.; Zhang, Z.; Wang, T.J.C.; et al. Focused Ultrasound-Mediated Blood-Brain Barrier Opening Increases Delivery and Efficacy of Etoposide for Glioblastoma Treatment. Int. J. Radiat. Oncol. Biol. Phys. 2021, 110, 539–550. [Google Scholar] [CrossRef] [PubMed]
  76. Park, S.H.; Kim, M.J.; Jung, H.H.; Chang, W.S.; Choi, H.S.; Rachmilevitch, I.; Zadicario, E.; Chang, J.W. One-Year Outcome of Multiple Blood-Brain Barrier Disruptions With Temozolomide for the Treatment of Glioblastoma. Front. Oncol. 2020, 10, 1663. [Google Scholar] [CrossRef]
  77. Park, S.H.; Kim, M.J.; Jung, H.H.; Chang, W.S.; Choi, H.S.; Rachmilevitch, I.; Zadicario, E.; Chang, J.W. Safety and feasibility of multiple blood-brain barrier disruptions for the treatment of glioblastoma in patients undergoing standard adjuvant chemotherapy. J. Neurosurg. 2020, 134, 475–483. [Google Scholar] [CrossRef] [PubMed]
  78. Sonabend, A.M.; Gould, A.; Amidei, C.; Ward, R.; Schmidt, K.A.; Zhang, D.Y.; Gomez, C.; Bebawy, J.F.; Liu, B.P.; Bouchoux, G.; et al. Repeated blood–brain barrier opening with an implantable ultrasound device for delivery of albumin-bound paclitaxel in patients with recurrent glioblastoma: A phase 1 trial. Lancet Oncol. 2023, 24, 509–522. [Google Scholar] [CrossRef] [PubMed]
  79. Mitchell, M.J.; Billingsley, M.M.; Haley, R.M.; Wechsler, M.E.; Peppas, N.A.; Langer, R. Engineering precision nanoparticles for drug delivery. Nat. Rev. Drug Discov. 2021, 20, 101–124. [Google Scholar] [CrossRef]
  80. Li, J.; Zhao, J.; Tan, T.; Liu, M.; Zeng, Z.; Zeng, Y.; Zhang, L.; Fu, C.; Chen, D.; Xie, T. Nanoparticle Drug Delivery System for Glioma and Its Efficacy Improvement Strategies: A Comprehensive Review. Int. J. Nanomed. 2020, 15, 2563–2582. [Google Scholar] [CrossRef]
  81. Li, Y.; Wu, M.; Zhang, N.; Tang, C.; Jiang, P.; Liu, X.; Yan, F.; Zheng, H. Mechanisms of enhanced antiglioma efficacy of polysorbate 80-modified paclitaxel-loaded PLGA nanoparticles by focused ultrasound. J. Cell. Mol. Med. 2018, 22, 4171–4182. [Google Scholar] [CrossRef]
  82. Song, Z.; Huang, X.; Wang, J.; Cai, F.; Zhao, P.; Yan, F. Targeted Delivery of Liposomal Temozolomide Enhanced Anti-Glioblastoma Efficacy through Ultrasound-Mediated Blood-Brain Barrier Opening. Pharmaceutics 2021, 13, 1270. [Google Scholar] [CrossRef] [PubMed]
  83. Timbie, K.F.; Afzal, U.; Date, A.; Zhang, C.; Song, J.; Wilson Miller, G.; Suk, J.S.; Hanes, J.; Price, R.J. MR image-guided delivery of cisplatin-loaded brain-penetrating nanoparticles to invasive glioma with focused ultrasound. J. Control Release 2017, 263, 120–131. [Google Scholar] [CrossRef] [PubMed]
  84. Coluccia, D.; Figueiredo, C.A.; Wu, M.Y.; Riemenschneider, A.N.; Diaz, R.; Luck, A.; Smith, C.; Das, S.; Ackerley, C.; O’Reilly, M.; et al. Enhancing glioblastoma treatment using cisplatin-gold-nanoparticle conjugates and targeted delivery with magnetic resonance-guided focused ultrasound. Nanomedicine 2018, 14, 1137–1148. [Google Scholar] [CrossRef]
  85. Shen, Y.; Hu, M.; Li, W.; Chen, Y.; Xu, Y.; Sun, L.; Liu, D.; Chen, S.; Gu, Y.; Ma, Y.; et al. Delivery of DNA octahedra enhanced by focused ultrasound with microbubbles for glioma therapy. J. Control. Release 2022, 350, 158–174. [Google Scholar] [CrossRef] [PubMed]
  86. Sun, T.; Krishnan, V.; Pan, D.C.; Filippov, S.K.; Ravid, S.; Sarode, A.; Kim, J.; Zhang, Y.; Power, C.; Aday, S.; et al. Ultrasound-mediated delivery of flexibility-tunable polymer drug conjugates for treating glioblastoma. Bioeng. Transl. Med. 2023, 8, e10408. [Google Scholar] [CrossRef] [PubMed]
  87. Zhang, D.Y.; Dmello, C.; Chen, L.; Arrieta, V.A.; Gonzalez-Buendia, E.; Kane, J.R.; Magnusson, L.P.; Baran, A.; James, C.D.; Horbinski, C.; et al. Ultrasound-mediated Delivery of Paclitaxel for Glioma: A Comparative Study of Distribution, Toxicity, and Efficacy of Albumin-bound Versus Cremophor Formulations. Clin. Cancer Res. 2020, 26, 477–486. [Google Scholar] [CrossRef]
  88. Sabbagh, A.; Beccaria, K.; Ling, X.; Marisetty, A.; Ott, M.; Caruso, H.; Barton, E.; Kong, L.Y.; Fang, D.; Latha, K.; et al. Opening of the Blood-Brain Barrier Using Low-Intensity Pulsed Ultrasound Enhances Responses to Immunotherapy in Preclinical Glioma Models. Clin. Cancer Res. 2021, 27, 4325–4337. [Google Scholar] [CrossRef] [PubMed]
  89. Xie, W.; Suryaprakash, S.; Wu, C.; Rodriguez, A.; Fraterman, S. Trends in the use of liquid biopsy in oncology. Nat. Rev. Drug Discov. 2023, 22, 612–613. [Google Scholar] [CrossRef]
  90. Pacia, C.P.; Zhu, L.; Yang, Y.; Yue, Y.; Nazeri, A.; Michael Gach, H.; Talcott, M.R.; Leuthardt, E.C.; Chen, H. Feasibility and safety of focused ultrasound-enabled liquid biopsy in the brain of a porcine model. Sci. Rep. 2020, 10, 7449. [Google Scholar] [CrossRef]
  91. Zhu, L.; Cheng, G.; Ye, D.; Nazeri, A.; Yue, Y.; Liu, W.; Wang, X.; Dunn, G.P.; Petti, A.A.; Leuthardt, E.C.; et al. Focused Ultrasound-enabled Brain Tumor Liquid Biopsy. Sci. Rep. 2018, 8, 6553. [Google Scholar] [CrossRef]
  92. Zhu, L.; Nazeri, A.; Pacia, C.P.; Yue, Y.; Chen, H. Focused ultrasound for safe and effective release of brain tumor biomarkers into the peripheral circulation. PLoS ONE 2020, 15, e0234182. [Google Scholar] [CrossRef] [PubMed]
  93. Pacia, C.P.; Yuan, J.; Yue, Y.; Xu, L.; Nazeri, A.; Desai, R.; Gach, H.M.; Wang, X.; Talcott, M.R.; Chaudhuri, A.A.; et al. Sonobiopsy for minimally invasive, spatiotemporally-controlled, and sensitive detection of glioblastoma-derived circulating tumor DNA. Theranostics 2022, 12, 362–378. [Google Scholar] [CrossRef] [PubMed]
  94. Yuan, J.; Xu, L.; Chien, C.Y.; Yang, Y.; Yue, Y.; Fadera, S.; Stark, A.H.; Schwetye, K.E.; Nazeri, A.; Desai, R.; et al. First-in-human prospective trial of sonobiopsy in high-grade glioma patients using neuronavigation-guided focused ultrasound. NPJ Precis. Oncol. 2023, 7, 92. [Google Scholar] [CrossRef] [PubMed]
  95. Rustenhoven, J.; Kipnis, J. Brain borders at the central stage of neuroimmunology. Nature 2022, 612, 417–429. [Google Scholar] [CrossRef]
  96. Waldman, A.D.; Fritz, J.M.; Lenardo, M.J. A guide to cancer immunotherapy: From T cell basic science to clinical practice. Nat. Rev. Immunol. 2020, 20, 651–668. [Google Scholar] [CrossRef] [PubMed]
  97. Jung, O.; Thomas, A.; Burks, S.R.; Dustin, M.L.; Frank, J.A.; Ferrer, M.; Stride, E. Neuroinflammation associated with ultrasound-mediated permeabilization of the blood–brain barrier. Trends Neurosci. 2022, 45, 459–470. [Google Scholar] [CrossRef]
  98. Chen, K.T.; Chai, W.Y.; Lin, Y.J.; Lin, C.J.; Chen, P.Y.; Tsai, H.C.; Huang, C.Y.; Kuo, J.S.; Liu, H.L.; Wei, K.C. Neuronavigation-guided focused ultrasound for transcranial blood-brain barrier opening and immunostimulation in brain tumors. Sci. Adv. 2021, 7, eabd0772. [Google Scholar] [CrossRef]
  99. McMahon, D.; Hynynen, K. Acute Inflammatory Response Following Increased Blood-Brain Barrier Permeability Induced by Focused Ultrasound is Dependent on Microbubble Dose. Theranostics 2017, 7, 3989–4000. [Google Scholar] [CrossRef] [PubMed]
  100. Sheybani, N.D.; Witter, A.R.; Garrison, W.J.; Miller, G.W.; Price, R.J.; Bullock, T.N.J. Profiling of the immune landscape in murine glioblastoma following blood brain/tumor barrier disruption with MR image-guided focused ultrasound. J. Neuro-Oncol. 2022, 156, 109–122. [Google Scholar] [CrossRef]
  101. Chen, P.Y.; Hsieh, H.Y.; Huang, C.Y.; Lin, C.Y.; Wei, K.C.; Liu, H.L. Focused ultrasound-induced blood-brain barrier opening to enhance interleukin-12 delivery for brain tumor immunotherapy: A preclinical feasibility study. J. Transl. Med. 2015, 13, 93. [Google Scholar] [CrossRef]
  102. Sheybani, N.D.; Breza, V.R.; Paul, S.; McCauley, K.S.; Berr, S.S.; Miller, G.W.; Neumann, K.D.; Price, R.J. ImmunoPET-informed sequence for focused ultrasound-targeted mCD47 blockade controls glioma. J. Control Release 2021, 331, 19–29. [Google Scholar] [CrossRef] [PubMed]
  103. Bonosi, L.; Marino, S.; Benigno, U.E.; Musso, S.; Buscemi, F.; Giardina, K.; Gerardi, R.; Brunasso, L.; Costanzo, R.; Iacopino, D.G.; et al. Sonodynamic therapy and magnetic resonance-guided focused ultrasound: New therapeutic strategy in glioblastoma. J. Neuro-Oncol. 2023, 163, 219–238. [Google Scholar] [CrossRef] [PubMed]
  104. Keenlyside, A.; Marples, T.; Gao, Z.; Hu, H.; Nicely, L.G.; Nogales, J.; Li, H.; Landgraf, L.; Solth, A.; Melzer, A.; et al. Development and optimisation of in vitro sonodynamic therapy for glioblastoma. Sci. Rep. 2023, 13, 20215. [Google Scholar] [CrossRef] [PubMed]
  105. Huang, C.Y.; Li, J.C.; Chen, K.T.; Lin, Y.J.; Feng, L.Y.; Liu, H.L.; Wei, K.C. Evaluation the Effect of Sonodynamic Therapy with 5-Aminolevulinic Acid and Sodium Fluorescein by Preclinical Animal Study. Cancers 2024, 16, 253. [Google Scholar] [CrossRef] [PubMed]
  106. Raspagliesi, L.; D’Ammando, A.; Gionso, M.; Sheybani, N.D.; Lopes, M.B.; Moore, D.; Allen, S.; Gatesman, J.; Porto, E.; Timbie, K.; et al. Intracranial Sonodynamic Therapy With 5-Aminolevulinic Acid and Sodium Fluorescein: Safety Study in a Porcine Model. Front. Oncol. 2021, 11, 679989. [Google Scholar] [CrossRef] [PubMed]
  107. Suehiro, S.; Ohnishi, T.; Yamashita, D.; Kohno, S.; Inoue, A.; Nishikawa, M.; Ohue, S.; Tanaka, J.; Kunieda, T. Enhancement of antitumor activity by using 5-ALA-mediated sonodynamic therapy to induce apoptosis in malignant gliomas: Significance of high-intensity focused ultrasound on 5-ALA-SDT in a mouse glioma model. J. Neurosurg. 2018, 129, 1416–1428. [Google Scholar] [CrossRef] [PubMed]
  108. Wu, S.K.; Santos, M.A.; Marcus, S.L.; Hynynen, K. MR-guided Focused Ultrasound Facilitates Sonodynamic Therapy with 5-Aminolevulinic Acid in a Rat Glioma Model. Sci. Rep. 2019, 9, 10465. [Google Scholar] [CrossRef]
  109. Zhou, Y.; Jiao, J.; Yang, R.; Wen, B.; Wu, Q.; Xu, L.; Tong, X.; Yan, H. Temozolomide-based sonodynamic therapy induces immunogenic cell death in glioma. Clin. Immunol. 2023, 256, 109772. [Google Scholar] [CrossRef]
  110. Huang, Y.; Meng, Y.; Pople, C.B.; Bethune, A.; Jones, R.M.; Abrahao, A.; Hamani, C.; Kalia, S.K.; Kalia, L.V.; Lipsman, N.; et al. Cavitation Feedback Control of Focused Ultrasound Blood-Brain Barrier Opening for Drug Delivery in Patients with Parkinson’s Disease. Pharmaceutics 2022, 14, 2607. [Google Scholar] [CrossRef]
  111. Jones, R.M.; Huang, Y.; Meng, Y.; Scantlebury, N.; Schwartz, M.L.; Lipsman, N.; Hynynen, K. Echo-Focusing in Transcranial Focused Ultrasound Thalamotomy for Essential Tremor: A Feasibility Study. Mov. Disord. 2020, 35, 2327–2333. [Google Scholar] [CrossRef]
  112. Jung, N.Y.; Park, C.K.; Kim, M.; Lee, P.H.; Sohn, Y.H.; Chang, J.W. The efficacy and limits of magnetic resonance-guided focused ultrasound pallidotomy for Parkinson’s disease: A Phase I clinical trial. J. Neurosurg. 2018, 130, 1853–1861. [Google Scholar] [CrossRef] [PubMed]
  113. Chien, C.Y.; Xu, L.; Pacia, C.P.; Yue, Y.; Chen, H. Blood-brain barrier opening in a large animal model using closed-loop microbubble cavitation-based feedback control of focused ultrasound sonication. Sci. Rep. 2022, 12, 16147. [Google Scholar] [CrossRef] [PubMed]
  114. Chen, K.T.; Lin, Y.J.; Chai, W.Y.; Lin, C.J.; Chen, P.Y.; Huang, C.Y.; Kuo, J.S.; Liu, H.L.; Wei, K.C. Neuronavigation-guided focused ultrasound (NaviFUS) for transcranial blood-brain barrier opening in recurrent glioblastoma patients: Clinical trial protocol. Ann. Transl. Med. 2020, 8, 673. [Google Scholar] [CrossRef] [PubMed]
  115. Ramamurthy, B. Ultrasound Guided Opening of Blood-Brain Barrier. US 2023/0128189 A1, 2023. [Google Scholar]
  116. Zhong, L.; Li, Y.; Xiong, L.; Wang, W.; Wu, M.; Yuan, T.; Yang, W.; Tian, C.; Miao, Z.; Wang, T.; et al. Small molecules in targeted cancer therapy: Advances, challenges, and future perspectives. Signal Transduct. Target. Ther. 2021, 6, 201. [Google Scholar] [CrossRef] [PubMed]
  117. Meng, Y.; Pople, C.B.; Suppiah, S.; Llinas, M.; Huang, Y.; Sahgal, A.; Perry, J.; Keith, J.; Davidson, B.; Hamani, C.; et al. MR-guided focused ultrasound liquid biopsy enriches circulating biomarkers in patients with brain tumors. Neuro Oncol. 2021, 23, 1789–1797. [Google Scholar] [CrossRef] [PubMed]
  118. Dmello, C.; Sonabend, A.; Arrieta, V.A.; Zhang, D.Y.; Kanojia, D.; Chen, L.; Gould, A.; Zhang, J.; Kang, S.J.; Winter, J.; et al. Translocon-associated Protein Subunit SSR3 Determines and Predicts Susceptibility to Paclitaxel in Breast Cancer and Glioblastoma. Clin. Cancer Res. 2022, 28, 3156–3169. [Google Scholar] [CrossRef]
  119. Brighi, C.; Salimova, E.; de Veer, M.; Puttick, S.; Egan, G. Translation of focused ultrasound for blood-brain barrier opening in glioma. J. Control. Release 2022, 345, 443–463. [Google Scholar] [CrossRef]
  120. Sener, U.; Ruff, M.W.; Campian, J.L. Immunotherapy in Glioblastoma: Current Approaches and Future Perspectives. Int. J. Mol. Sci. 2022, 23, 7046. [Google Scholar] [CrossRef]
Figure 1. A brief timeline of FUS BBBO history. This timeline shows critical events that have led to current research and clinical trials using FUS. Adapted from the Focused Ultrasound Foundation Timeline of Focused Ultrasound. Created with BioRender.com [24,27,30,31,32,33,34,35,36,37,38,39].
Figure 1. A brief timeline of FUS BBBO history. This timeline shows critical events that have led to current research and clinical trials using FUS. Adapted from the Focused Ultrasound Foundation Timeline of Focused Ultrasound. Created with BioRender.com [24,27,30,31,32,33,34,35,36,37,38,39].
Biomedicines 12 01230 g001
Figure 2. Effects of MB-FUS on the BBB. The schematic depicts the BBB with the brain parenchyma above and the blood vessel with circulating MBs below (left). Note the cell types of the neurovascular unit: endothelial cells, pericytes, and astrocytic endfeet. MBs oscillate as they enter the acoustic field. At low MB-FUS intensities, typical for MB-FUS BBBO, the MBs oscillate stably, leading to the transient disruption of endothelial tight junctions, which then, in turn, lead to BBBO (right). MB-FUS BBBO allows small molecules to pass through the BBB for enhanced drug delivery (purple arrow) or tumor-associated DNA to enter the bloodstream for liquid biopsy (blue arrow).
Figure 2. Effects of MB-FUS on the BBB. The schematic depicts the BBB with the brain parenchyma above and the blood vessel with circulating MBs below (left). Note the cell types of the neurovascular unit: endothelial cells, pericytes, and astrocytic endfeet. MBs oscillate as they enter the acoustic field. At low MB-FUS intensities, typical for MB-FUS BBBO, the MBs oscillate stably, leading to the transient disruption of endothelial tight junctions, which then, in turn, lead to BBBO (right). MB-FUS BBBO allows small molecules to pass through the BBB for enhanced drug delivery (purple arrow) or tumor-associated DNA to enter the bloodstream for liquid biopsy (blue arrow).
Biomedicines 12 01230 g002
Table 1. Clinical FUS systems.
Table 1. Clinical FUS systems.
NameCompanyMethod of Operation
Exablate model 4000 Type 2InSightec (Tirat Carmel, Israel)Multi-helmet—MRgFUS
NaviFUS®NaviFUS (Taipei, Taiwan)Multi-element FUS
NeuroAccessCordance Medical (Mountain View, CA, USA)Multi-element FUS
Sonocloud9CarThera (Paris, France)Implanted device
Table 2. Registered clinical trials using FUS for glioma are separated by glioma type. An overview of ongoing clinical trials for treating HGGs.
Table 2. Registered clinical trials using FUS for glioma are separated by glioma type. An overview of ongoing clinical trials for treating HGGs.
Study NameNCT NumberConditionsInterventions
The Use of Focused Ultrasound and DCE K-trans Imaging to Evaluate Permeability of the Blood-Brain BarrierNCT04063514Low-grade gliomaDWL doppler sonography
Brainsonix Pulsed LIFU
Study of Sonodynamic Therapy in Participants With Recurrent High-Grade GliomaNCT04559685High-grade gliomaaminolevulinic acid (ALA)
InSightec ExAblate system
Safety Study of the Repeated Opening of the Blood-brain Barrier With the SonoCloud® Device to Treat Malignant Brain Tumors in Pediatric Patients (SONOKID)NCT05293197GliomaCarThera SonoCloud9
ExAblate (Magnetic Resonance-guided Focused Ultrasound Surgery) Treatment of Brain TumorsNCT01473485GliomaInSightec ExAblate system
Assessment of Safety and Feasibility of ExAblate Blood-Brain Barrier (BBB) DisruptionNCT03551249Glioma
GBM
InSightec ExAblate system
Assessment of Safety and Feasibility of ExAblate Blood-Brain Barrier (BBB) Disruption in GBM PatientsNCT04998864GBMInSightec ExAblate system
Blood-Brain Barrier Disruption (BBBD) for Liquid Biopsy in Subjects With Glioblastoma Brain Tumor NCT05383872GBMInSightec ExAblate system
Assessment of Safety and Feasibility of ExAblate Blood-Brain Barrier (BBB) Disruption for Treatment of Glioma [117]NCT03616860GBMInSightec ExAblate system
Safety of BBB Disruption Using NaviFUS System in Recurrent Glioblastoma Multiforme (GBM) PatientsNCT03626896GBMNaviFUS System
Sonodynamic Therapy With ExAblate System in Glioblastoma Patients (Sonic ALA)NCT04845919GBM5-ALA
InSightec ExAblate system
ExAblate Blood-Brain Barrier Disruption for Glioblastoma in Patients Undergoing Standard Chemotherapy [76,77]NCT03712293GBMTemozolomide
InSightec ExAblate system
Safety and Efficacy of Transient Opening of the Blood-brain Barrier (BBB) With the SonoCloud-9 (SC9-GBM-01) [78]NCT03744026GBMCarboplatin
CarThera SonoCloud9
Phase 2a Immune Modulation With Ultrasound for Newly Diagnosed GlioblastomaNCT05864534GBM
Giosarcoma
Balstilimab
Botensilimab
Liposomal Doxorubicin
CarThera Sonocloud-9
Exablate Blood-Brain Barrier Disruption With Carboplatin for the Treatment of rGBMNCT04440358
NCT04417088
Recurrent GBMCarboplatin
InSightec ExAblate system
Sonodynamic Therapy in Patients With Recurrent GBM (GBM 001)NCT06039709Recurrent GBM5-ALA
Neuro-navigation guided LIFU
Efficacy and Safety of NaviFUS System add-on Bevacizumab (BEV) in Recurrent GBM PatientsNCT04446416Recurrent GBMBevacizumab
NaviFUS system
Evaluate the Safety and Preliminary Efficacy of the Combination of NaviFUS System With Re-irradiation for rGBM PatientsNCT04988750Recurrent GBMNaviFUS sysem
Sonocloud-9 in Association With Carboplatin Versus Standard-of-Care Chemotherapies (CCNU or TMZ) in Recurrent GBM (SONOBIRD)NCT05902169Recurrent GBMCarboplatin
Lomustine
Temozolomide
CarThera SonoCloud9
Randomized Study of Neo-adjuvant and Adjuvant Pembrolizumab With and Without Targeted Blood Brain Barrier Opening Using Exablate MRI-guided Focused Ultrasound (Exablate MRgFUS) for Recurrent GlioblastomaNCT05879120Recurrent GBMPembrolizumab
InSightec ExAblate system
Ultrasound-based Blood-brain Barrier Opening and Albumin-bound Paclitaxel and Carboplatin for Recurrent Glioblastoma [78,118]NCT04528680Recurrent GBMalbumin-bound paclitaxel
carboplatin
CarThera Sonocloud-9
FUS Etoposide for DMG—A Feasibility StudyNCT05762419Diffuse Midline Glioma (DMG)Etoposide
Neuro-navigator controlled FUS
Noninvasive Focused Ultrasound (FUS) With Oral Panobinostat in Children With Progressive Diffuse Midline Glioma (DMG)NCT04804709DMGPanobinostat
Neuro-navigator controlled FUS
Blood Brain Barrier (BBB) Disruption Using Exablate Focused Ultrasound With Doxorubicin for Treatment of Pediatric DIPGNCT05630209
NCT05615623
Diffuse Intrinsic Pontine Glioma (DIPG)Doxorubicin
InSightec ExAblate system
A Phase 2 Study of Sonodynamic Therapy Using SONALA-001 and Exablate 4000 Type 2.0 in Patients with DIPGNCT05123534DIPGALA
InSightec ExAblate system
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Seas, A.A.; Malla, A.P.; Sharifai, N.; Winkles, J.A.; Woodworth, G.F.; Anastasiadis, P. Microbubble-Enhanced Focused Ultrasound for Infiltrating Gliomas. Biomedicines 2024, 12, 1230. https://doi.org/10.3390/biomedicines12061230

AMA Style

Seas AA, Malla AP, Sharifai N, Winkles JA, Woodworth GF, Anastasiadis P. Microbubble-Enhanced Focused Ultrasound for Infiltrating Gliomas. Biomedicines. 2024; 12(6):1230. https://doi.org/10.3390/biomedicines12061230

Chicago/Turabian Style

Seas, Alexandra A., Adarsha P. Malla, Nima Sharifai, Jeffrey A. Winkles, Graeme F. Woodworth, and Pavlos Anastasiadis. 2024. "Microbubble-Enhanced Focused Ultrasound for Infiltrating Gliomas" Biomedicines 12, no. 6: 1230. https://doi.org/10.3390/biomedicines12061230

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop