• Open Access
    Review

    Focused ultrasound for treatment of peripheral brain tumors

    Phillip Mitchell Johansen 1
    Payton Yerke Hansen 1
    Ali A. Mohamed 1
    Sarah J. Girshfeld 1
    Marc Feldmann 2
    Brandon Lucke-Wold 3*

    Explor Drug Sci. 2023;1:107–125 DOI: https://doi.org/10.37349/eds.2023.00009

    Received: January 11, 2023 Accepted: April 13, 2023 Published: April 28, 2023

    Academic Editor: Rangasamy Jayakumar, Amrita Vishwa Vidyapeetham University, India

    Abstract

    Malignant brain tumors are the leading cause of cancer-related death in children and remain a significant cause of morbidity and mortality throughout all demographics. Central nervous system (CNS) tumors are classically treated with surgical resection and radiotherapy in addition to adjuvant chemotherapy. However, the therapeutic efficacy of chemotherapeutic agents is limited due to the blood-brain barrier (BBB). Magnetic resonance guided focused ultrasound (MRgFUS) is a new and promising intervention for CNS tumors, which has shown success in preclinical trials. High-intensity focused ultrasound (HIFU) has the capacity to serve as a direct therapeutic agent in the form of thermoablation and mechanical destruction of the tumor. Low-intensity focused ultrasound (LIFU) has been shown to disrupt the BBB and enhance the uptake of therapeutic agents in the brain and CNS. The authors present a review of MRgFUS in the treatment of CNS tumors. This treatment method has shown promising results in preclinical trials including minimal adverse effects, increased infiltration of the therapeutic agents into the CNS, decreased tumor progression, and improved survival rates.

    Keywords

    Low-intensity focused ultrasound, high-intensity focused ultrasound, magnetic resonance guided focused ultrasound, magnetic resonance-guided therapy, glioblastoma

    Introduction

    Over 260,000 people are diagnosed with primary malignant central nervous system (CNS) tumors annually. Malignant brain tumors are the leading cause of cancer-related death in children and the third largest cause of death in young adults [1, 2]. Currently, brain tumors are typically treated with surgical resection and radiotherapy in addition to chemotherapy or other therapeutic molecules [35]. However, the chemotherapeutic molecules must first cross the blood-brain barrier (BBB) to treat CNS tumors [5, 6]. The BBB is a semipermeable system comprised of capillary endothelial cells interlinked by tight junctions [68]. The BBB functions as a protective mechanism that defends the brain from exogenous and endogenous substances. Small lipid soluble molecules with a molecular weight of less than 400 Da can cross the BBB via passive diffusion or active transport channels [7, 8]. Large and hydrophilic molecules are blocked by the BBB [7].

    Unfortunately, the therapeutic efficacy of chemotherapeutic agents is limited due to the BBB [5, 6, 9, 10]. Treatment with therapeutic molecules is largely limited by the molecular size of the drugs (e.g., doxorubicin ∼540 Da and bevacizumab 149 kDa) [11]. In contrast, temozolomide (TMZ), a lipophilic molecule with a molecular size of 194 Da, is one of few chemotherapeutic agents that can cross the BBB and treat CNS tumors [12]. The concentration of TMZ in the brain and cerebrospinal fluid (CSF) can reach up to 20% of plasma concentrations. TMZ is limited by a half-life of 1.8 h and requires continuous administration to maintain therapeutic levels [13]. Additionally, TMZ has many adverse side effects, and some CNS tumors have been known to develop resistance to TMZ [6, 14]. To overcome these limitations, intra-arterial infusion of mannitol, direct injection, and convection enhanced delivery may be used. However, these approaches are invasive and non-targeted [11]. This poses a therapeutic dilemma as the efficacy of treatment does not outweigh the systemic effects.

    Focused ultrasound (FUS) is a promising intervention for CNS tumors, which has shown success in preclinical trials. The therapeutic capacity varies depending on the intensity of the ultrasound waves. High-intensity focused ultrasound (HIFU) has the capacity to serve as a direct therapeutic agent in the form of thermoablation and mechanical destruction of tumor cells. Mechanical destruction requires intravenously administered microbubbles (MBs), which have a gaseous core and are coated by a polymeric or lipid shell [15]. When targeted with ultrasound, MBs oscillate at the endothelium and disrupt the cell membrane thereby decreasing the required energy threshold and reducing the possible adverse effects seen in thermal ablation [16]. In comparison, low-intensity focused ultrasound (LIFU) enhanced with MBs has been shown to disrupt the BBB and enhance the uptake of chemotherapeutic agents in the brain and CNS. Additionally, LIFU has been used in liquid biopsies by disrupting the BBB thereby allowing for the release of tumor biomarkers into the peripheral circulation. We present a review of the mechanism of action of magnetic resonance guided focused ultrasound (MRgFUS), preclinical evidence, and current clinical studies in the treatment of CNS tumors.

    Mechanism of action

    Generally, FUS in the brain can be classified based on intensity. HIFU is a direct treatment approach that directly induces tissue necrosis (Figure 1), while LIFU induces BBB disruption for a more effective administration of pharmaceutical treatments (Figure 2) [17, 18]. The mechanism of HIFU treatment of peripheral brain tumors is through thermoablation and mechanical destruction while the mechanism of LIFU treatment of brain tumors is through the induction of functional and mechanical changes to the BBB and blood tumor barriers [19].

    Continuous HIFU waves produce a thermal effect that reaches the threshold for protein denaturation and subsequent cell death. Created with BioRender.com

    BBB opening for drug delivery: LIFU is used in conjunction with injected intravenous therapeutic agents. LIFU stimulates the MBs causing them to expand and contract. This leads to the opening of tight junctions and increased numbers of transcytotic vesicles in addition to decreased concentration of efflux pumps. Overall, it enhances the ability for therapeutic agents to cross the BBB. Created with BioRender.com

    Procedure

    For HIFU, pretreatment magnetic resonance imaging (MRI) volumetric computed tomography (CT) scans are taken for MRgFUS planning. MRI is used to identify the size of the target, and CT scan is used to determine bone density and thickness for phase aberration correction [20, 21]. Exact focusing metrics are determined, and the patient is prepared for the procedure. The head is shaved to prevent hair interference on ultrasound delivery and then immobilized by a stereotactic head frame. A silicon barrier seals cooled gasless water within the transducer cavity to prevent thermal damage and enhance ultrasound delivery [20]. The duration of the sonication, power, phase, and number of the array elements are adjusted throughout the treatment. HIFU is delivered as a single exposure of 2–10 s continuous waveform at 650 kHz with a short 2.3 mm wavelength to establish a tight 6–10 mm tissue ablation focus [22, 23]. Administered ultrasound intensity ranges from 100 W/cm2 to over 10,000 W/cm2 [24]. Passive cavitation detectors and MRI thermometry are used to monitor cavitation and thermal rise, respectively.

    Similar to HIFU, pretreatment MRIs are obtained for MRgFUS planning in LIFU, and a stereotactic frame is used to fix the ultrasound transducer to the scalp of the patient [18, 25]. Ultrasound attenuation is minimized by the application of degassed water between the scalp and the transducer [26]. MRI is used to identify the target tissue and magnetic resonance thermometry is used to detect and control tissue temperature at the target region [21, 26]. Contrast enhanced MRI and dye leakage is the most common method used to determine the extent of BBB or blood tumor barrier disruption to assess treatment efficacy [27]. Evans blue is a popular option for assessing barrier disruption. However, other proposed options include FITC-Dextran, Trypan blue, Nile blue, and sodium fluorescein [2834]. Because of the narrow and precise aperture of the converging LIFU wave, many different targeting methods have been developed to cover the entirety of the target tissue volume and associated areas [19]. This can be achieved using a grid system to target specific points that utilize the additive effect of merging waves to produce the intended therapeutic effect [3538]. Neuronavigation is another newly explored option for precise targeting that does not require MRI pretreatment planning [3941]. Ultrasonic pressure waves are applied in phases to induce size changes of exogenous MBs localized at the target zone. To establish the appropriate and safe degree of barrier disruption for the intended therapeutic effects, specific parameters must be established [42]. These include ultrasound frequency, acoustic pressure and duration, burst pulse repetition frequency, duty cycle, exposure duration, and MB type, size, and dosage [6, 18, 4244]. Unfortunately, there is a lack of consensus regarding the optimal parameters required for LIFU treatment [44, 45]. The discrepancy in the literature is likely attributed to ultrasound wave impendence, attenuation, distortion, scattering, reflection, and absorption when interacting with the varying thickness and density of the skull [18, 44, 4650] and hair [18]. The suggested delivery of LIFU is as 0.74% to 5% duty cycle for 50–150 s [22]. Frequencies of 220 kHz with a large 6.8 mm wavelength are administered for transient opening of the BBB [24]. Intensities range from 0.125–3.0 W/cm2 and a mechanical index of 0.48, 0.58, and 0.68 demonstrated transient opening of the BBB [24].

    HIFU: thermoablation

    Electrical signals are converted to ultrasonic waves and focused using a lens, phased array, or a concave/curved transducer to target a precise volume of tissue [18, 43]. Continuous waves of ultrasound are applied to produce a high-pressure thermal effect with a small target focus, which results in tissue destruction. This thermal effect is achieved when the local temperature reaches the threshold for protein denaturation and tissue damage and subsequent lesion formation [18, 51]. This thermoablative process further increases tumor sensitivity to radiation by damaging deoxyribonucleic acid (DNA) repair enzymes [2].

    Treatment of different volumes can be achieved by modulating volume dimension by transducer selection, ultrasound frequency, or repeated ultrasound exposure of multiple overlapping focal volumes [52, 53]. The administration of ultrasound mediated thermoablation is challenging because of skull induced disruption of ultrasound wave propagation [54, 55]. Historically, the administration of FUS treatment for thermoablation required a craniectomy because of the limitations introduced by the cranium [51, 56], which causes phase aberrations and beam distortions that can change the shape and location of focus [5759]. Current techniques allow for noninvasive transcranial administration of HIFU, reducing the risk of infection while simultaneously addressing factors that influence the efficacy of the treatment [21, 54, 60, 61].

    Real time MRI thermometry monitoring, MRgFUS, allows for the delivery of FUS transcranially with stereotactic precision. When administering HIFU with a phased array transducer, phase and amplitude can be adjusted to correct for skull introduced aberrations as well [18, 60, 61]. This is achieved by initiating a distorted wavefront that, upon disruption by the skull, will result in the desired shape and location of the focus. Phased arrays also allow for increased focal placement and accuracy via electronic focus steering [62]. Clinically, the use of both MRgFUS with electronic focus steering capacity of phased array transducers has demonstrated glioblastoma tumor ablation in a patient without adverse effects or neurological deficits [63].

    HIFU: mechanical destruction

    Ultrasound induced mechanical destruction occurs through pressure wave increases created by the charge components of the ultrasound mechanical wave [64]. Positive components induce compression and negative components induce expansion of gas filled crevices or bubbles. These bubbles increase in oscillation between compression and expansion as the ultrasound pressure wave increases. When the desired ultrasound threshold for treatment is reached, the bubbles explode and produce high velocity shock waves that induce cavitation of the focus [65, 66]. This phenomenon is described as inertial or acoustic cavitation (Figure 3). In addition, ultrasound waves produce a direct mechanical stress along the direction of the beam onto the target focus. This secondary mechanism of mechanical destruction can be induced when radiation forces are intense enough to cause tissue displacement and strain [67]. Shear forces induced by liquid movement also play a role in damaging focal tissue based on the presence of liquid [68]. This phenomenon is described as acoustic streaming.

    HIFU induces mechanical destruction of cells by creating pressure waves that oscillate the MBs. As the MBs expand and contract, they eventually reach a threshold where they explode, producing shock waves that disrupt cell membranes leading to cell death. Created with BioRender.com

    LIFU: mechanical effects

    Exogenous MBs are administered and allowed to circulate the blood vessels and localize to the treatment target zone [42]. Ultrasonic pressure waves are applied in phases to induce MB size changes. MBs expand during the compression phase and contract during the rarefaction phase. This process of stable cavitation induces linear and symmetric oscillations that, when combined with the morphological ultrasound induced changes in MB size, result in disruption of BBB and blood tumor barriers [17, 42]. The FUS induced alteration in size and shape of MBs apply physical stress on blood vessel walls. The changes in size and shape also create circumferential fluid streaming that places further stress on endothelial cell walls [6, 42, 69]. In addition, the ultrasound waves can directly increase the permeability of the BBB and blood tumor barriers by facilitating trans- and paracellular transport [45]. The stretching of cerebral endothelial cells along with sonopermeation temporarily alters the integrity of tight junctions by affecting adhesion molecules [17, 70]. The integrity of the barriers may also be disrupted through the release of substances by the neurovascular unit (cerebral endothelial unit, astrocytes, pericytes, nerve endings, and microglia) in response to treatment (Figure 2) [7173].

    LIFU: functional effects

    LIFU can increase permeability of the BBB and blood tumor barrier by inducing functional changes. The use of LIFU may constrain or otherwise modulate protein expression by elevating local endothelial cell temperatures [74]. Transcellular permeability is amplified through the increased expression of calcium activated potassium channels while pharmacological efflux is reduced through the decreased expression of P-glycoprotein [18, 7578].

    Preclinical studies

    HIFU

    While most pre-clinical research involving MRgFUS has focused on LIFU, recent studies have shown that HIFU can serve as a direct therapeutic agent through thermoablation [11]. One study analyzed RNA and protein expression changes in MRgFUS-induced BBB hyperpermeability using pulsed FUS with MB parameters. They used rat models to evaluate the thermoablation effects on tumor prognosis and found that proinflammatory cytokines and heat shock protein 70 (HSP70) concentrations significantly increased following MRgFUS. Furthermore, they found increased levels of ionized calcium-binding adapter molecule 1 (Iba1), which indicates microglial and macrophage activation [79]. A second study utilized a spherical transducer at a frequency of 551.5 kHz and delivered ultrasound waves in 10 ms bursts, beginning with a starting acoustic pressure of 0.128 MPa, and found that MRgFUS was associated with an upregulation in pro-inflammatory cytokine and chemokine genes in neuronal endothelial cells. Additionally, there was a significant downregulation of BBB transporter genes in the 24 h following MRgFUS exposure [80]. Both studies found increased concentrations of glial fibrillary acidic protein (GFAP), which is associated with astrocyte activation, indicating that HIFU activates the innate immune system to further degrade tumor cells [79, 80]. As a caveat to both preclinical HIFU and LIFU studies, preclinical animal models differ from their human counterparts. For example, the equipment used is often minimized to better facilitate the study resources and parameters which may drastically alter the clinical impact. Such key differences are often referenced as fundamental limitations in bridging preclinical studies to human trials.

    LIFU

    Multiple studies have evaluated the efficacy of LIFU to enhance drug delivery into the brain. Drugs that have been studied include bevacizumab [81], TMZ [82, 83], trastuzumab [84, 85], doxorubicin [8688], methotrexate [89], and carboplatin [90]. Additionally, this technique has been used to promote the migration of immunoglobulins [9195], viruses [96], and cells across the BBB (Table 1). In animal models, BBB disruption occurs immediately and resolves within 6–8 h. Additionally, multiple studies found that MRgFUS did not cause neuronal injury [6]. A summary of all preclinical trials reviewed is listed in Table 1.

    Summary of existing preclinical trials

    ModelFrequency (mHz)Burst length (ms)Repetition frequency (Hz)Exposure length (s)Acoustic pressure (MPa)Estimated acoustic power (W)MB typeTherapeutic agentObservation
    Mouse (Swiss Webster) [91]0.69101400.6–1.1-Optison™D4 receptor antibodyMinimal damage seen < 0.8 MPa. Damage observed > 0.8 MPa
    Mouse (Swiss Webster) [84]0.69101400.6 and 0.8-Optison™HerceptinGreater delivery at 0.8 MPa compared to 0.6 MPa
    Transgenic mice (B6C3-Tg and PDAPP) [92]0.6910140–450.67–0.80.28–0.4Optison™ or Definity®Anti-amyloid β antibodies3-fold increase in antibodies localized to plaques
    TgCRND8 mice [93]0.5581011200.3-Definity®Amyloid-β antibodiesMice treated with MRgFUS had a 12% reduction in plaque sizes
    non-Tg and TgCRND8 mice [94]0.51011200.3-Definity®Endogenous antibodiesReduced plaque sizes and activation of microglia
    pR5 mice [95]1101060.7-In-house lipid-shelledRN2N antibodiesMice treated with MRgFUS had an 11-fold increase in RN2N delivery to CNS. Reduced anxiety and tau phosphorylation
    nu/nu mice (intracranial U87mg cells) [81]0.4101600.4–0.84–18SonoVue®BevacizumabAnimals treated with MRgFUS had decreased tumor growth and vessel area as well as increased survival rates
    nu/nu mice (intracranial U87mg cells) [83]0.5101600.3–0.72–5SonoVue®TMZMRgFUS caused TMZ accumulation in the brain to increase and have a slower degradation rate compared to controls. MRgFUS also slowed tumor progression
    Male NOD-scid mice [88]1-160-2.86SonoVue®DoxorubicinMRgFUS increased oxorubicin concentrations in the brain by 2.35-fold compared with the control tumors
    C57BL/6J mice [96]0.5581011200.53–0.6-Definity®Virus serotype 9A dose of 2.5 × 109 VG/g allowed expression of the transgene in neurons, astrocytes, and oligodendrocytes in brain regions targeted with ultrasound. Nontargeted regions were minimally infected
    Athymic nude-Foxn1nu mice [90]1.0523.811200.3-SonoVue®CarboplatinAnimals who received MRgFUS had a 4.2-fold increase in carboplatin concentration in the brain. MRgFUS also enhanced survival and delayed tumor growth
    Nude rats (intracranial M.D. Anderson-metastatic breast (MDA-MB)-361 cells) [85]0.69101600.46–0.620.4–0.7Optison™Trastuzumab and pertuzumabAnimals treated with FUS had slower tumor growth rates and higher survival rates
    Sprague-Dawley rats (intracranial C6 glioma) [5]0.51001900.36–0.75 or 20SonoVue®IL-12Animals treated with MRgFUS had a 3-fold increase in IL-12 in the CNS. They also had higher CD8+/T-reg ratio and slower tumor growth
    Sprague-Dawley rats [97]0.55111200.24-Definity®GFP-tagged neural stem cellsMRgFUS allowed higher concentrations of neuronal stem cells in specifically targeted locations in the brain compared to controls
    Male Sprague-Dawley rats [98]1.7101.760–1201.2-Optison™DoxorubicinMRgFUS increased the antineoplastic efficacy of liposomal doxorubicin in the brain. Animals treated with FUS had better survival rates compared to controls
    Male Sprague-Dawley rats [80]0.551011200.128 and increased by a 0.008/s increment-Definity®GadovistMRgFUS induced a transient inflammatory response in microvessels
    Male Sprague-Dawley rats [87]1.5 or 1.7101300.36–2.50.06–3.0 Optison™DoxorubicinDoxorubicin concentrations were significantly higher in areas targeted by MRgFUS than nontargeted areas of the brain
    Athymic nude rat (intracranial MDA-MB-231 cells) [99]0.551011200.32–0.35-Definity®In HER2-specific NK-9210-fold increase in HER2-specific NK-92 cells regions targeted by MRgFUS
    Athymic nude rat (intracranial MDA-MB-231 cells) [100]0.55102120--Definity®In HER2-specific NK-92Rats with MRgFUS had significantly reduced tumor growth and higher survival rates compared to controls
    Male Fischer rats [82]0.5101600.63SonoVue®TMZMRgFUS increased the TMZ CSF/plasma ratio from 22.7% to 38.6%, reduced tumor progression, and increased survival rates
    Male Sprague-Dawley rats [62]0.69101600.55-Definity®DoxorubicinThe concentration of doxorubicin decreased by 32% when injected 10 min after MRgFUS compared to when doxorubicin is injected before sonification
    Female Sprague-Dawley rats [79]0.5891011200.3-Optison™-MRgFUS increased levels of proinflammatory, anti-inflammatory, trophic, neurotrophic and neurogenesis factors
    Rabbit (New Zealand white) [55]1.63 and 1.5100120-0.55 or 3 Optison™-Sonication as 0.55 W increased the number of endothelial vesicles and fenestrations on the luminal surface of endothelial cells. Damage occurred at 3 W
    Rabbit (New Zealand white) [89]1.1--10-6SonoVue®MethotrexateMRgFUS significantly increased methotrexate concentrations in the brain compared to controls
    Display full size

    GFP: green fluorescent protein; HER2: human epithelial growth factor receptor 2; NK: natural killer; NOD: non-obese diabetic; -: none

    TMZ

    Studies in mouse and rat models found that TMZ administered with MRgFUS had higher concentrations of the drug in the brain. Additionally, MRgFUS leads to slower tumor growth rates and prolonged survival [6]. Liu et al. [81] found that nude mice implanted with U87 human glioma cells had more TMZ accumulation in neuronal tissue and slower tumor progression. Additionally, they found that there were higher rates of TMZ degradation in the core of the tumor [83]. Furthermore, Wei et al. [82] used Fisher rat models with implanted 9-L glioma cells and found that MRgFUS facilitated (parameters included acoustic power  =  3 W; peak negative pressure  =  0.6 MPa; burst length  = 10 ms; pulse repetition frequency  =  1 Hz; exposure time  =  60 s) BBB opening and permitted higher concentrations of TMZ to enter the brain tissue. Overall, the rats with MRgFUS and TMZ had improved survival rates and elevated TMZ CNS/plasma ratios compared to the rats given only TMZ [82].

    Despite these findings, the therapeutic efficacy of TMZ is limited by its high rates of tumor resistance. TMZ has minimal effects on tumors with an unmethylated promotor region in the O6-methylguanine-DNA methyltransferase (MGMT) gene [101]. Therefore, MGMT gene modulation has become a growing interest [6, 102]. A study by Papachristodoulou et al. [103] used MB enhanced MRgFUS to deliver MGMT inactivator liposomal O6-(4-bromothenyl) guanine (O6BTG) in mice with TMZ-resistant tumors. They delivered ultrasonic waves in bursts of 10  ms at a repetition frequency of 1  Hz for a total duration of 180 s and found MRgFUS to be correlated with higher rates of MGMT resistance reversal and prolonged survival [103].

    Bevacizumab

    Bevacizumab is a monoclonal antibody that inhibits vascular endothelial growth factor (VEGF) [104]. While commonly used to improve peritumoral edema, it has not been known to improve prognosis [6]. However, a study using U87 glioma mouse models found that delivering MRgFUS with MB parameters increased bevacizumab trafficking into the brain. They found that the concentration of bevacizumab in the CNS increased by 57-fold [6, 81]. It was also noted that animals that received MRgFUS had a decrease in tumor vascular distribution and a slower tumor growth rate compared to controls leading to significantly improved survival rates [81].

    1,3-bis (2-chloroethyl)-1-nitrosourea

    1,3-bis (2-chloroethyl)-1-nitrosourea (BCNU) is a common second-line agent for recurrent gliomas. However, it lacks efficacy as a monotherapy [6]. Systemic administration of BCNU is highly toxic [6, 105]. However, the addition of MRgFUS enhanced BCNU concentrations in the CNS by 202% in rat C6 glioma models [6, 81]. Furthermore, MRgFUS slowed tumor growth and improved survival [81].

    Immunoglobulins

    Multiple studies have shown success in delivering antibodies into the CNS via MRgFUS. Chen et al. [5] found that Sprague-Dawley rats with implanted intracranial C6 gliomas had a three-fold increase in IL-12 concentrations in the CNS when exposed to MRgFUS [5]. Additionally, Nisbet et al. [95] found similar results with RN2N antibodies. They noted an 11-fold increase in RN2N antibodies with MRgFUS [95]. With the ability to introduce antibodies to the brain, treatment for malignant CNS tumors can become more targeted and less invasive. Furthermore, this treatment method can potentially be utilized in other diseases such as Alzheimer’s disease [92, 93]. Overall, this treatment approach offers a promising future that warrants further investigation.

    Viral therapy

    Although viruses fail to surpass the BBB, viral therapy remains a promising therapeutic technique for various brain tumors. Current strategies to overcome this include viral vector implantation through stereotactic or open surgery procedures [6]. Despite these approaches, viral therapy is less effective because the current delivery techniques result in uneven coverage throughout the CNS. However, recent studies have shown that MRgFUS combined with intravenous MBs facilitated the transmission of recombinant viral vectors into the brain parenchyma [106108].

    Cell therapy

    Chimeric antigen receptor T-cell therapy has been shown to decrease tumor progression. However, its efficacy is limited by T-cell migration into the CNS through the BBB. This can be overcome through direct intraventricular administration of the T-cell therapy or, as recent studies have shown, by utilizing MRgFUS to facilitate the trafficking of neuronal stem cells and immune cells past the BBB [97, 99, 100, 109].

    Targeted delivery

    While the use of carrier vehicles including MBs, liposomes, and nanoparticles to deliver chemotherapeutics offers protection from systemic side effects, it limits release of the agent at the target site (particularly in CNS tumors) and may result in subtherapeutic local drug levels [110]. Preclinical studies have demonstrated the ability of MRgFUS to increase the targeted release of therapeutic agents from drug delivery vehicles through hyperthermia, stable cavitation, and radiation forces [111113]. In addition to improving delivery of carrier vehicles through the BBB in rodents, MRgFUS has been successfully applied in a trans-skull model to generate controlled hyperthermia and effectively release thermosensitive drugs in glioma [110, 114]. Other studies have examined the frequency and duration of FUS for this purpose, with some results suggesting high intensity bursts may be optimal [115117].

    Liquid biopsy

    Blood-based biopsies are used to diagnose and monitor various other cancers. However, this approach is hindered in CNS malignancies due to the BBB blocking biomarker release into the peripheral circulation [6]. Recent studies have investigated the use of MRgFUS in tumor diagnostics. Zhu et al. [118] explored the effect of MRgFUS in tumor biomarkers in mice with orthotopic implanted murine glioma cells that were enhanced with green fluorescent protein (eGFP). They found that MRgFUS at 0.59 MPa resulted in significantly increased plasma eGFP mRNA levels. Furthermore, the levels of biomarker release were directly correlated to the extent of tumor expansion [118]. These findings offer a promising future for cancer diagnostics.

    Tumor subtypes

    Intracranial MRgFUS has historically been used to treat neurologic conditions such as essential tremor, epilepsy, and Parkinson’s disease. However, its indications are expanding and may now include intracranial neoplasms. LIFU and HIFU are valid options, and both may be used depending on the exact tumor present. LIFU has traditionally been used to open the BBB and allow for delivery of chemotherapeutic agents [6]. In contrast, HIFU uses FUS beams to thermally ablate or mechanically destroy target tissues [119]. Advancing technologies and new modalities of administering ultrasonic energy, such as needle-based therapeutic ultrasound, have improved the ability of HIFU to work through the calvarium [120]. Notwithstanding, much of the existing clinical literature surrounding HIFU for brain tumors surrounds centrally located brain tumors.

    A majority of current clinical trials regarding MRgFUS are investigating its use in treating glioblastoma multiforme (GBM). However, other types of tumors currently being investigated include astrocytomas, ependymomas, cranial nerve tumors, meningiomas, metastases, neuronal tumors, pineal tumors, and tumors in the sellar region [121]. Hersh et al. [2] found the reason for optimism in using both modalities to treat GBM [2]. However, limited sample sizes and outdated technologies have obscured the data. The same researchers found that LIFU, when used in conjunction with either chemotherapeutics (e.g., TMZ or carboplatin), nanoparticles (e.g., gold, iron, or platinum), or MBs, was able to locally disrupt the BBB and impair multi-drug efflux pumps, resulting in decreased GBM growth and improved overall survival, although further studies are needed to obtain more conclusive results [2, 22]. In contrast, they found that HIFU still faces technological obstacles to surpass calvarial attenuation and minimize collateral damage. Notwithstanding, current literature includes reports of HIFU and subsequent thermocoagulation resulting in partial high-grade glioma resection and improved treatment margins [63].

    Because most gliomas are infiltrative and difficult to distinguish from healthy brain tissue, gross total resection can be difficult, whether through MRgFUS or open surgery. In contrast, well-defined brain lesions, such as metastases or meningiomas, may be easier to ablate using HIFU while simultaneously monitoring temperature to ensure a safe and controllable procedure that limits damage to surrounding structures [121]. This may be most feasible when such benign tumors are in inaccessible or eloquent locations (e.g., adjacent to the brainstem or cranial nerves). In contrast, LIFU can be used to increase the permeability of the BBB for virtually any subtype of brain tumor for which chemotherapy is used, whether that be neoadjuvant or adjuvant [22]. These findings suggest that FUS may play a role in treating any CNS neoplasm, as some tumors may be ablated while others are treated with chemotherapy in conjunction with LIFU.

    Current level of evidence

    While LIFU disruption of the BBB for the delivery of therapeutic agents has been demonstrated to be safe and effective for the treatment of brain tumors [122, 123], there has been considerably less investigation into the use of HIFU to treat brain tumors via direct tissue destruction. Early attempts at HIFU for the treatment of brain tumors proved challenging, as ablative temperatures could not be safely reached without performing a craniectomy to provide an acoustic window [56, 124, 125]. However, with the advancement of phased array transducers, interest in transcranial MRgFUS thermoablation and histotripsy has been rapidly increasing [63, 125, 126]. In preclinical studies, both histotripsy and thermoablation have been shown effective through excised human skulls in vitro [127]. More recently, Lu et al. [128] demonstrated successful treatment of swine cerebral tissue through a human skull [128], and later demonstrated successful histotripsy ablation through a human skull in an in vivo swine model [129]. To date, clinical data are limited to case reports of 1–3 patients [54, 63]. Two phase I clinical trials evaluating the safety and efficacy of transcranial MRgFUS thermoablation for the treatment of either brain metastasis or recurrent glioma are ongoing [130, 131]. Though recruitment challenges delayed their timelines, selected early results have been promising and both trials concluded in December 2022 [20, 63, 130, 131]. A further exciting possibility is transcranial MRgFUS without the need for complex phased array devices. Sukovich et al. [132] in 2016 demonstrated targeted lesion generation in red blood cell agarose tissue through a human skullcap without aberration correction, which could simplify the procedure and open a new line of investigation [132].

    Implications

    In nearly every surgical subspecialty, the newest trends focus evermore on minimizing the invasiveness of the procedure. Regarding neurosurgery, endovascular and endoscopic procedures are vastly more common than they were in years past. MRgFUS presents the opportunity to eliminate the invasiveness of surgery altogether. MRgFUS provides a glimpse at what the future of tumor therapy might be, which utilizes MRI to locate the lesion and MRgFUS to ablate the lesion without requiring any truly intracranial operation. Such procedures may even be done in the outpatient setting with mild sedation rather than general anesthesia.

    FUS has the potential to drastically change how brain tumors are managed. The non-invasive approach would improve access to the deep brain structures while simultaneously decreasing the damage to the healthy surrounding brain tissue, reduce the likelihood of operative complications, and lessen the length of hospital stays and recovery time (and thus the overall cost of care) without compromising therapeutic efficacy or adding increased exposure to ionizing radiation. Additionally, and perhaps more importantly, LIFU has demonstrated clinical efficacy and is currently the modality with the most intrigue. LIFU can temporarily disrupt the BBB to enhance the effects of adjuvant therapies and improve the overall clinical impacts on chemotherapeutics. Such targeted increased permeability to the BBB can allow for maximal chemotherapeutic delivery to the target tissue in the brain with minimal adverse effects throughout the rest of the body.

    This treatment modality is faced with the challenge of parameter optimization. MRgFUS has demonstrated treatment efficacy in both LIFU and HIFU. Factors such as ultrasound wave impendence, attenuation, distortion, scattering, reflection, and absorption when interacting with the varying thickness and density of the skull and hair are key contributors to the discrepancies observed in the literature regarding the optimal parameters for treatment. Further clinical studies will be needed to consider these factors and determine the appropriate ultrasound frequency, acoustic pressure and duration, burst pulse repetition frequency, duty cycle, exposure duration, and MB type, size, and dosage. These are important considerations because of the function-mechanical effects of LIFU and thermo-mechanical effects of HIFU. In addition, a standardized monitoring approach of treatment efficacy will likely need to be established considering the variance in determining BBB disruption in LIFU and monitoring mechanical effects. Thus, future directions for this new treatment modality would be focused on controlling external factors, optimizing treatment parameters, and standardizing monitoring modalities.

    Conclusions

    FUS is an emerging treatment modality for CNS tumors that may be used in isolation or in conjunction with surgical resection and radiotherapy to improve overall outcomes. HIFU serves as a direct therapeutic agent in the form of thermoablation and mechanical destruction of tumor cells and their components while LIFU functionally and mechanically disrupts the BBB and enhances the uptake of chemotherapeutic agents in the CNS. Preclinical studies surrounding LIFU comprise a majority of the literature and have shown that MRgFUS significantly improved the transmission of multiple therapeutic agents such as chemotherapeutics, small molecules, and antibodies to the brain, leading to reduced tumor burden and improved outcomes. Additionally, LIFU was shown to play a role in monitoring neoplastic disease through liquid biopsy. Early preclinical studies with HIFU also showed promise, with evidence of tumor destruction and an improved host immune response. Both therapies have the potential to treat a broad array of CNS tumors, however, further clinical studies will likely need to be pursued before FUS becomes a routine treatment option for CNS tumors. Overall, MRgFUS has shown promising results throughout the existing literature, including minimal adverse effects, increased infiltration of therapeutic agents into the CNS, decreased tumor progression, and improved survival rates. As such, further investigation regarding MRgFUS is warranted.

    Abbreviations

    BBB:

    blood-brain barrier

    BCNU:

    1,3-bis (2-chloroethyl)-1-nitrosourea

    CNS:

    central nervous system

    FUS:

    focused ultrasound

    GBM:

    glioblastoma multiforme

    HIFU:

    high-intensity focused ultrasound

    LIFU:

    low-intensity focused ultrasound

    MBs:

    microbubbles

    MGMT :

    O6-methylguanine-deoxyribonucleic acid methyltransferase

    MRgFUS:

    magnetic resonance guided focused ultrasound

    MRI:

    magnetic resonance imaging

    TMZ:

    temozolomide

    Declarations

    Author contributions

    PMJ: Project administration, Investigation, Writing—original draft, Writing—review & editing. PYH, AAM, and SJG: Investigation, Writing—original draft, Writing—review & editing. MF: Writing—review & editing. BLW: Conceptualization, Writing—review & editing.

    Conflicts of interest

    The authors declare that they have no conflicts of interest.

    Ethical approval

    Not applicable.

    Consent to participate

    Not applicable.

    Consent to publication

    Not applicable.

    Availability of data and materials

    Not applicable.

    Funding

    Not applicable.

    Copyright

    © The Author(s) 2023.

    References

    Buckner JC, Brown PD, O’Neill BP, Meyer FB, Wetmore CJ, Uhm JH. Central nervous system tumors. Mayo Clin Proc. 2007;82:127186. [DOI] [PubMed]
    Hersh AM, Bhimreddy M, Weber-Levine C, Jiang K, Alomari S, Theodore N, et al. Applications of focused ultrasound for the treatment of glioblastoma: a new frontier. Cancers (Basel). 2022;14:4920. [DOI] [PubMed] [PMC]
    Grossman SA, Batara JF. Current management of glioblastoma multiforme. Semin Oncol. 2004;31:63544. [DOI] [PubMed]
    Galanis E, Buckner J. Chemotherapy for high-grade gliomas. Br J Cancer. 2000;82:137180. [DOI] [PubMed] [PMC]
    Chen PY, Hsieh HY, Huang CY, Lin CY, Wei KC, Liu HL. 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. [DOI] [PubMed] [PMC]
    Bunevicius A, McDannold NJ, Golby AJ. Focused ultrasound strategies for brain tumor therapy. Oper Neurosurg (Hagerstown). 2020;19:918. [DOI] [PubMed] [PMC]
    Abbott NJ, Patabendige AAK, Dolman DEM, Yusof SR, Begley DJ. Structure and function of the blood-brain barrier. Neurobiol Dis. 2010;37:1325. [DOI] [PubMed]
    Daneman R. The blood-brain barrier in health and disease. Ann Neurol. 2012;72:64872. [DOI] [PubMed]
    Achrol AS, Rennert RC, Anders C, Soffietti R, Ahluwalia MS, Nayak L, et al. Brain metastases. Nat Rev Dis Primers. 2019;5:5. [DOI] [PubMed]
    Deeken JF, Löscher W. The blood-brain barrier and cancer: transporters, treatment, and Trojan horses. Clin Cancer Res. 2007;13:166374. [DOI] [PubMed]
    Curley CT, Sheybani ND, Bullock TN, Price RJ. Focused ultrasound immunotherapy for central nervous system pathologies: challenges and opportunities. Theranostics. 2017;7:360823. [DOI] [PubMed] [PMC]
    Agarwala SS, Kirkwood JM. Temozolomide, a novel alkylating agent with activity in the central nervous system, may improve the treatment of advanced metastatic melanoma. Oncologist. 2000;5:14451. [DOI] [PubMed]
    Ostermann S, Csajka C, Buclin T, Leyvraz S, Lejeune F, Decosterd LA, et al. Plasma and cerebrospinal fluid population pharmacokinetics of temozolomide in malignant glioma patients. Clin Cancer Res. 2004;10:372836. [DOI] [PubMed]
    Berrocal A, Perez Segura P, Gil M, Balaña C, Garcia Lopez J, Yaya R, et al.; GENOM Cooperative Group. Extended-schedule dose-dense temozolomide in refractory gliomas. J Neurooncol. 2010;96:41722. [DOI] [PubMed] [PMC]
    Aryal M, Arvanitis CD, Alexander PM, McDannold N. Ultrasound-mediated blood-brain barrier disruption for targeted drug delivery in the central nervous system. Adv Drug Deliv Rev. 2014;72:94109. [DOI] [PubMed] [PMC]
    Arvanitis CD, Vykhodtseva N, Jolesz F, Livingstone M, McDannold N. Cavitation-enhanced nonthermal ablation in deep brain targets: feasibility in a large animal model. J Neurosurg. 2016;124:14509. [DOI] [PubMed] [PMC]
    Chen KT, Wei KC, Liu HL. Theranostic strategy of focused ultrasound induced blood-brain barrier opening for CNS disease treatment. Front Pharmacol. 2019;10:86. [DOI] [PubMed] [PMC]
    Meng Y, Hynynen K, Lipsman N. Applications of focused ultrasound in the brain: from thermoablation to drug delivery. Nat Rev Neurol. 2021;17:722. [DOI] [PubMed]
    Mungur R, Zheng J, Wang B, Chen X, Zhan R, Tong Y. Low-intensity focused ultrasound technique in glioblastoma multiforme treatment. Front Oncol. 2022;12:903059. [DOI] [PubMed] [PMC]
    Monteith S, Sheehan J, Medel R, Wintermark M, Eames M, Snell J, et al. Potential intracranial applications of magnetic resonance-guided focused ultrasound surgery. J Neurosurg. 2013;118:21521. [DOI] [PubMed]
    Hynynen K, McDannold N, Clement G, Jolesz FA, Zadicario E, Killiany R, et al. Pre-clinical testing of a phased array ultrasound system for MRI-guided noninvasive surgery of the brain—a primate study. Eur J Radiol. 2006;59:14956. [DOI] [PubMed]
    Baek H, Lockwood D, Mason EJ, Obusez E, Poturalski M, Rammo R, et al. Clinical intervention using focused ultrasound (FUS) stimulation of the brain in diverse neurological disorders. Front Neurol. 2022;13:880814. [DOI] [PubMed] [PMC]
    Haar >Gt, Coussios C. High intensity focused ultrasound: physical principles and devices. Int J Hyperthermia. 2007;23:89104. [DOI] [PubMed]
    Elhelf IAS, Albahar H, Shah U, Oto A, Cressman E, Almekkawy M. High intensity focused ultrasound: the fundamentals, clinical applications and research trends. Diagn Interv Imaging. 2018;99:34959. [DOI] [PubMed]
    Beccaria K, Sabbagh A, de Groot J, Canney M, Carpentier A, Heimberger AB. Blood-brain barrier opening with low intensity pulsed ultrasound for immune modulation and immune therapeutic delivery to CNS tumors. J Neurooncol. 2021;151:6573. [DOI] [PubMed]
    Ahmed N, Gandhi D, Melhem ER, Frenkel V. MRI guided focused ultrasound-mediated delivery of therapeutic cells to the brain: a review of the state-of-the-art methodology and future applications. Front Neurol. 2021;12:669449. [DOI] [PubMed] [PMC]
    Hynynen K, McDannold N, Vykhodtseva N, Jolesz FA. Noninvasive MR imaging-guided focal opening of the blood-brain barrier in rabbits. Radiology. 2001;220:6406. [DOI] [PubMed]
    Hugon G, Goutal S, Dauba A, Breuil L, Larrat B, Winkeler A, et al. [18F]2-fluoro-2-deoxy-sorbitol PET imaging for quantitative monitoring of enhanced blood-brain barrier permeability induced by focused ultrasound. Pharmaceutics. 2021;13:1752. [DOI] [PubMed] [PMC]
    Lea-Banks H, Hynynen K. Sub-millimetre precision of drug delivery in the brain from ultrasound-triggered nanodroplets. J Control Release. 2021;338:73141. [DOI] [PubMed]
    Shen Y, Pi Z, Yan F, Yeh CK, Zeng X, Diao X, et al. Enhanced delivery of paclitaxel liposomes using focused ultrasound with microbubbles for treating nude mice bearing intracranial glioblastoma xenografts. Int J Nanomedicine. 2017;12:561329. [DOI] [PubMed] [PMC]
    Zhang DY, Dmello C, Chen L, Arrieta VA, Gonzalez-Buendia E, Kane JR, 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;26:47786. [DOI] [PubMed] [PMC]
    Park J, Aryal M, Vykhodtseva N, Zhang YZ, McDannold N. Evaluation of permeability, doxorubicin delivery, and drug retention in a rat brain tumor model after ultrasound-induced blood-tumor barrier disruption. J Control Release. 2017;250:7785. [DOI] [PubMed] [PMC]
    MPdS, Della Giustina A, Petronilho F. Using evans blue dye to determine blood-brain barrier integrity in rodents. Curr Protoc Immunol. 2019;126:e83. [DOI] [PubMed]
    Xu Y, He Q, Wang M, Wang X, Gong F, Bai L, et al. Quantifying blood-brain-barrier leakage using a combination of evans blue and high molecular weight FITC-Dextran. J Neurosci Methods. 2019;325:108349. [DOI] [PubMed]
    Sulheim E, Mørch Y, Snipstad S, Borgos SE, Miletic H, Bjerkvig R, et al. Therapeutic effect of cabazitaxel and blood-brain barrier opening in a patient-derived glioblastoma model. Nanotheranostics. 2019;3:10312. [DOI] [PubMed] [PMC]
    Alli S, Figueiredo CA, Golbourn B, Sabha N, Wu MY, Bondoc A, et al. Brainstem blood brain barrier disruption using focused ultrasound: a demonstration of feasibility and enhanced doxorubicin delivery. J Control Release. 2018;281:2941. [DOI] [PubMed] [PMC]
    Wei HJ, Upadhyayula PS, Pouliopoulos AN, Englander ZK, Zhang X, Jan CI, 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;110:53950. [DOI] [PubMed] [PMC]
    Englander ZK, Wei HJ, Pouliopoulos AN, Bendau E, Upadhyayula P, Jan CI, et al. Focused ultrasound mediated blood-brain barrier opening is safe and feasible in a murine pontine glioma model. Sci Rep. 2021;11:6521. [DOI] [PubMed] [PMC]
    Wu SY, Aurup C, Sanchez CS, Grondin J, Zheng W, Kamimura H, et al. Efficient blood-brain barrier opening in primates with neuronavigation-guided ultrasound and real-time acoustic mapping. Sci Rep. 2018;8:7978. [DOI] [PubMed] [PMC]
    Pouliopoulos AN, Kwon N, Jensen G, Meaney A, Niimi Y, Burgess MT, et al. Safety evaluation of a clinical focused ultrasound system for neuronavigation guided blood-brain barrier opening in non-human primates. Sci Rep. 2021;11:15043. [DOI] [PubMed] [PMC]
    Pouliopoulos AN, Wu SY, Burgess MT, Karakatsani ME, Kamimura HAS, Konofagou EE. A clinical system for non-invasive blood-brain barrier opening using a neuronavigation-guided single-element focused ultrasound transducer. Ultrasound Med Biol. 2020;46:7389. [DOI] [PubMed] [PMC]
    McMahon D, O’Reilly MA, Hynynen K. Therapeutic agent delivery across the blood-brain barrier using focused ultrasound. Annu Rev Biomed Eng. 2021;23:89113. [DOI] [PubMed]
    Entzian K, Aigner A. Drug delivery by ultrasound-responsive nanocarriers for cancer treatment. Pharmaceutics. 2021;13:1135. [DOI] [PubMed] [PMC]
    Arsiwala TA, Sprowls SA, Blethen KE, Adkins CE, Saralkar PA, Fladeland RA, et al. Ultrasound-mediated disruption of the blood tumor barrier for improved therapeutic delivery. Neoplasia. 2021;23:67691. [DOI] [PubMed] [PMC]
    Snipstad S, Vikedal K, Maardalen M, Kurbatskaya A, Sulheim E, Davies CdL. Ultrasound and microbubbles to beat barriers in tumors: improving delivery of nanomedicine. Adv Drug Deliv Rev. 2021;177:113847. [DOI] [PubMed]
    Shriki J. Ultrasound physics. Crit Care Clin. 2014;30:124. [DOI] [PubMed]
    Coltrera MD. Ultrasound physics in a nutshell. Otolaryngol Clin North Am. 2010;43:114959. [DOI] [PubMed]
    Riis TS, Webb TD, Kubanek J. Acoustic properties across the human skull. Ultrasonics. 2022;119:106591. [DOI] [PubMed] [PMC]
    Zhang H, Zhang Y, Xu M, Song X, Chen S, Jian X, et al. The effects of the structural and acoustic parameters of the skull model on transcranial focused ultrasound. Sensors (Basel). 2021;21:5962. [DOI] [PubMed] [PMC]
    Dasgupta A, Liu M, Ojha T, Storm G, Kiessling F, Lammers T. Ultrasound-mediated drug delivery to the brain: principles, progress and prospects. Drug Discov Today Technol. 2016;20:418. [DOI] [PubMed] [PMC]
    Guthkelch AN, Carter LP, Cassady JR, Hynynen KH, Iacono RP, Johnson PC, et al. Treatment of malignant brain tumors with focused ultrasound hyperthermia and radiation: results of a phase I trial. J Neurooncol. 1991;10:27184. [DOI] [PubMed]
    Tempany CMC, McDannold NJ, Hynynen K, Jolesz FA. Focused ultrasound surgery in oncology: overview and principles. Radiology. 2011;259:3956. [DOI] [PubMed] [PMC]
    Kennedy JE. High-intensity focused ultrasound in the treatment of solid tumours. Nat Rev Cancer. 2005;5:3217. [DOI] [PubMed]
    McDannold N, Clement GT, Black P, Jolesz F, Hynynen K. Transcranial magnetic resonance imaging- guided focused ultrasound surgery of brain tumors: initial findings in 3 patients. Neurosurgery. 2010;66:32332. [DOI] [PubMed] [PMC]
    Sheikov N, McDannold N, Vykhodtseva N, Jolesz F, Hynynen K. Cellular mechanisms of the blood-brain barrier opening induced by ultrasound in presence of microbubbles. Ultrasound Med Biol. 2004;30:97989. [DOI] [PubMed]
    Ram Z, Cohen ZR, Harnof S, Tal S, Faibel M, Nass D, et al. Magnetic resonance imaging-guided, high-intensity focused ultrasound for brain tumor therapy. Neurosurgery. 2006;59:94956. [DOI] [PubMed]
    Fry FJ, Barger JE. Acoustical properties of the human skull. J Acoust Soc Am. 1978;63:157690. [DOI] [PubMed]
    Martin B, McElhaney JH. The acoustic properties of human skull bone. J Biomed Mater Res. 1971;5:32533.
    Pinton G, Aubry JF, Bossy E, Muller M, Pernot M, Tanter M. Attenuation, scattering, and absorption of ultrasound in the skull bone. Med Phys. 2012;39:299307. [DOI] [PubMed]
    Clement GT, Hynynen K. A non-invasive method for focusing ultrasound through the human skull. Phys Med Biol. 2002;47:1219. [DOI] [PubMed]
    Aubry JF, Tanter M. MR-guided transcranial focused ultrasound. In: Escoffre JM, Bouakaz A, editors. Therapeutic ultrasound. Cham: Springer International Publishing; 2016. pp. 97–111.
    Aryal M, Porter T. Emerging therapeutic strategies for brain tumors. Neuromolecular Med. 2022;24:2334. [DOI] [PubMed]
    Coluccia D, Fandino J, Schwyzer L, O’Gorman R, Remonda L, Anon J, et al. First noninvasive thermal ablation of a brain tumor with MR-guided focused ultrasound. J Ther Ultrasound. 2014;2:17. [DOI] [PubMed] [PMC]
    Dalecki D. Mechanical bioeffects of ultrasound. Annu Rev Biomed Eng. 2004;6:22948. [DOI] [PubMed]
    Krasovitski B, Frenkel V, Shoham S, Kimmel E. Intramembrane cavitation as a unifying mechanism for ultrasound-induced bioeffects. Proc Natl Acad Sci U S A. 2011;108:325863. [DOI] [PubMed] [PMC]
    Prada F, Kalani MYS, Yagmurlu K, Norat P, Del Bene M, DiMeco F, et al. Applications of focused ultrasound in cerebrovascular diseases and brain tumors. Neurotherapeutics. 2019;16:6787. [DOI] [PubMed] [PMC]
    Hancock HA, Smith LH, Cuesta J, Durrani AK, Angstadt M, Palmeri ML, et al. Investigations into pulsed high-intensity focused ultrasound-enhanced delivery: preliminary evidence for a novel mechanism. Ultrasound Med Biol. 2009;35:172236. [DOI] [PubMed] [PMC]
    Frenkel V, Gurka R, Liberzon A, Shavit U, Kimmel E. Preliminary investigations of ultrasound induced acoustic streaming using particle image velocimetry. Ultrasonics. 2001;39:1536. [DOI] [PubMed]
    VanBavel E. Effects of shear stress on endothelial cells: possible relevance for ultrasound applications. Prog Biophys Mol Biol. 2007;93:37483. [DOI] [PubMed]
    Wu P, Zhu M, Li Y, Ya Z, Yang Y, Yuan Y, et al. Cascade‐amplifying synergistic therapy for intracranial glioma via endogenous reactive oxygen species‐triggered “all‐in‐one” nanoplatform. Adv Funct Mater. 2021;31:2105786. [DOI]
    Arvanitis CD, Ferraro GB, Jain RK. The blood-brain barrier and blood-tumour barrier in brain tumours and metastases. Nat Rev Cancer. 2020;20:2641. [DOI] [PubMed] [PMC]
    Wilhelm I, Nyúl-Tóth Á, Suciu M, Hermenean A, Krizbai IA. Heterogeneity of the blood-brain barrier. Tissue Barriers. 2016;4:e1143544. [DOI] [PubMed] [PMC]
    Arif WM, Elsinga PH, Gasca-Salas C, Versluis M, Martínez-Fernández R, Dierckx RAJO, et al. Focused ultrasound for opening blood-brain barrier and drug delivery monitored with positron emission tomography. J Control Release. 2020;324:30316. [DOI] [PubMed]
    Meijering BDM, Juffermans LJM, van Wamel A, Henning RH, Zuhorn IS, Emmer M, et al. Ultrasound and microbubble-targeted delivery of macromolecules is regulated by induction of endocytosis and pore formation. Circ Res. 2009;104:67987. [DOI] [PubMed]
    Cho H, Lee HY, Han M, Choi JR, Ahn S, Lee T, et al. Localized down-regulation of P-glycoprotein by focused ultrasound and microbubbles induced blood-brain barrier disruption in rat brain. Sci Rep. 2016;6:31201. [DOI] [PubMed] [PMC]
    Aryal M, Fischer K, Gentile C, Gitto S, Zhang YZ, McDannold N. Effects on P-glycoprotein expression after blood-brain barrier disruption using focused ultrasound and microbubbles. PLoS One. 2017;12:e0166061. [DOI] [PubMed] [PMC]
    Beccaria K, Canney M, Bouchoux G, Desseaux C, Grill J, Heimberger AB, et al. Ultrasound-induced blood-brain barrier disruption for the treatment of gliomas and other primary CNS tumors. Cancer Lett. 2020;479:1322. [DOI] [PubMed]
    Zhang J, Liu H, Du X, Guo Y, Chen X, Wang S, et al. Increasing of blood-brain tumor barrier permeability through transcellular and paracellular pathways by microbubble-enhanced diagnostic ultrasound in a C6 glioma model. Front Neurosci. 2017;11:86. [DOI] [PubMed] [PMC]
    Kovacs ZI, Kim S, Jikaria N, Qureshi F, Milo B, Lewis BK, et al. Disrupting the blood-brain barrier by focused ultrasound induces sterile inflammation. Proc Natl Acad Sci U S A. 2016;114:E7584. [DOI] [PubMed] [PMC]
    McMahon D, Bendayan R, Hynynen K. Acute effects of focused ultrasound-induced increases in blood-brain barrier permeability on rat microvascular transcriptome. Sci Rep. 2017;7:45657. [DOI] [PubMed] [PMC]
    Liu HL, Hsu PH, Lin CY, Huang CW, Chai WY, Chu PC, et al. Focused ultrasound enhances central nervous system delivery of bevacizumab for malignant glioma treatment. Radiology. 2016;281:99108. [DOI] [PubMed]
    Wei KC, Chu PC, Wang HYJ, Huang CY, Chen PY, Tsai HC, et al. Focused ultrasound-induced blood-brain barrier opening to enhance temozolomide delivery for glioblastoma treatment: a preclinical study. PLoS One. 2013;8:e58995. [DOI] [PubMed] [PMC]
    Liu HL, Huang CY, Chen JY, Wang HYJ, Chen PY, Wei KC. Pharmacodynamic and therapeutic investigation of focused ultrasound-induced blood-brain barrier opening for enhanced temozolomide delivery in glioma treatment. PLoS One. 2014;9:e114311. [DOI] [PubMed] [PMC]
    Kinoshita M, McDannold N, Jolesz FA, Hynynen K. Noninvasive localized delivery of herceptin to the mouse brain by MRI-guided focused ultrasound-induced blood-brain barrier disruption. Proc Natl Acad Sci U S A. 2006;103:1171923. [DOI] [PubMed] [PMC]
    Kobus T, Zervantonakis IK, Zhang Y, McDannold NJ. Growth inhibition in a brain metastasis model by antibody delivery using focused ultrasound-mediated blood-brain barrier disruption. J Control Release. 2016;238:2818. [DOI] [PubMed] [PMC]
    Aryal M, Vykhodtseva N, Zhang YZ, McDannold N. Multiple sessions of liposomal doxorubicin delivery via focused ultrasound mediated blood-brain barrier disruption: a safety study. J Control Release. 2015;204:609. [DOI] [PubMed] [PMC]
    Treat LH, McDannold N, Vykhodtseva N, Zhang Y, Tam K, Hynynen K. Targeted delivery of doxorubicin to the rat brain at therapeutic levels using MRI-guided focused ultrasound. Int J Cancer. 2007;121:9017. [DOI] [PubMed]
    Lin YL, Wu MT, Yang FY. Pharmacokinetics of doxorubicin in glioblastoma multiforme following ultrasound-Induced blood-brain barrier disruption as determined by microdialysis. J Pharm Biomed Anal. 2018;149:4827. [DOI] [PubMed]
    Mei J, Cheng Y, Song Y, Yang Y, Wang F, Liu Y, et al. Experimental study on targeted methotrexate delivery to the rabbit brain via magnetic resonance imaging-guided focused ultrasound. J Ultrasound Med. 2009;28:87180. [DOI] [PubMed]
    Dréan A, Lemaire N, Bouchoux G, Goldwirt L, Canney M, Goli L, et al. Temporary blood-brain barrier disruption by low intensity pulsed ultrasound increases carboplatin delivery and efficacy in preclinical models of glioblastoma. J Neurooncol. 2019;144:3341. [DOI] [PubMed]
    Kinoshita M, McDannold N, Jolesz FA, Hynynen K. Targeted delivery of antibodies through the blood-brain barrier by MRI-guided focused ultrasound. Biochem Biophys Res Commun. 2006;340:108590. [DOI] [PubMed]
    Raymond SB, Treat LH, Dewey JD, McDannold NJ, Hynynen K, Bacskai BJ. Ultrasound enhanced delivery of molecular imaging and therapeutic agents in Alzheimer’s disease mouse models. PLoS One. 2008;3:e2175. [DOI] [PubMed] [PMC]
    Jordão JF, Ayala-Grosso CA, Markham K, Huang Y, Chopra R, McLaurin J, et al. Antibodies targeted to the brain with image-guided focused ultrasound reduces amyloid-β plaque load in the TgCRND8 mouse model of Alzheimer’s disease. PLoS One. 2010;5:e10549. [DOI] [PubMed] [PMC]
    Jordão JF, Thévenot E, Markham-Coultes K, Scarcelli T, Weng YQ, Xhima K, et al. Amyloid-β plaque reduction, endogenous antibody delivery and glial activation by brain-targeted, transcranial focused ultrasound. Exp Neurol. 2013;248:1629. [DOI] [PubMed] [PMC]
    Nisbet RM, Van der Jeugd A, Leinenga G, Evans HT, Janowicz PW, Götz J. Combined effects of scanning ultrasound and a tau-specific single chain antibody in a tau transgenic mouse model. Brain. 2017;140:122030. [DOI] [PubMed] [PMC]
    Thévenot E, Jordão JF, O’Reilly MA, Markham K, Weng YQ, Foust KD, et al. Targeted delivery of self-complementary adeno-associated virus serotype 9 to the brain, using magnetic resonance imaging-guided focused ultrasound. Hum Gene Ther. 2012;23:114455. [DOI] [PubMed] [PMC]
    Burgess A, Ayala-Grosso CA, Ganguly M, Jordão JF, Aubert I, Hynynen K. Targeted delivery of neural stem cells to the brain using MRI-guided focused ultrasound to disrupt the blood-brain barrier. PLoS One. 2011;6:e27877. [DOI] [PubMed] [PMC]
    Treat LH, McDannold N, Zhang Y, Vykhodtseva N, Hynynen K. Improved anti-tumor effect of liposomal doxorubicin after targeted blood-brain barrier disruption by MRI-guided focused ultrasound in rat glioma. Ultrasound Med Biol. 2012;38:171625. [DOI] [PubMed] [PMC]
    Alkins R, Burgess A, Ganguly M, Francia G, Kerbel R, Wels WS, et al. Focused ultrasound delivers targeted immune cells to metastatic brain tumors. Cancer Res. 2013;73:18929. [DOI] [PubMed] [PMC]
    Alkins R, Burgess A, Kerbel R, Wels WS, Hynynen K. Early treatment of HER2-amplified brain tumors with targeted NK-92 cells and focused ultrasound improves survival. Neuro Oncol. 2016;18:97481. [DOI] [PubMed] [PMC]
    Hegi ME, Diserens AC, Gorlia T, Hamou MF, de Tribolet N, Weller M, et al. MGMT gene silencing and benefit from temozolomide in glioblastoma. N Engl J Med. 2005;352:9971003. [DOI] [PubMed]
    Hegi ME, Liu L, Herman JG, Stupp R, Wick W, Weller M, et al. Correlation of O6-methylguanine methyltransferase (MGMT) promoter methylation with clinical outcomes in glioblastoma and clinical strategies to modulate MGMT activity. J Clin Oncol. 2008;26:418999. [DOI] [PubMed]
    Papachristodoulou A, Signorell RD, Werner B, Brambilla D, Luciani P, Cavusoglu M, et al. Chemotherapy sensitization of glioblastoma by focused ultrasound-mediated delivery of therapeutic liposomes. J Control Release. 2019;295:1309. [DOI] [PubMed]
    Norden AD, Drappatz J, Wen PY. Antiangiogenic therapies for high-grade glioma. Nat Rev Neurol. 2009;5:61020. [DOI] [PubMed]
    Walker MD, Green SB, Byar DP, Alexander E Jr, Batzdorf U, Brooks WH, et al. Randomized comparisons of radiotherapy and nitrosoureas for the treatment of malignant glioma after surgery. N Engl J Med. 1980;303:13239. [DOI] [PubMed]
    Peruzzi P, Chiocca EA. Viruses in cancer therapy - from benchwarmers to quarterbacks. Nat Rev Clin Oncol. 2018;15:6578. [DOI] [PubMed] [PMC]
    Lawler SE, Speranza MC, Cho CF, Chiocca EA. Oncolytic viruses in cancer treatment: a review. JAMA Oncol. 2017;3:8419. [DOI] [PubMed]
    Desjardins A, Gromeier M, Herndon JE 2nd, Beaubier N, Bolognesi DP, Friedman AH, et al. Recurrent glioblastoma treated with recombinant poliovirus. N Engl J Med. 2018;379:15061. [DOI] [PubMed] [PMC]
    Migliorini D, Dietrich PY, Stupp R, Linette GP, Posey AD Jr, June CH. CAR T-cell therapies in glioblastoma: a first look. Clin Cancer Res. 2018;24:53540. [DOI] [PubMed]
    Kim C, Guo Y, Velalopoulou A, Leisen J, Motamarry A, Ramajayam K, et al. Closed-loop trans-skull ultrasound hyperthermia leads to improved drug delivery from thermosensitive drugs and promotes changes in vascular transport dynamics in brain tumors. Theranostics. 2021;11:727693. [DOI] [PubMed] [PMC]
    Couture O, Foley J, Kassell NF, Larrat B, Aubry JF. Review of ultrasound mediated drug delivery for cancer treatment: Updates from pre-clinical studies. Transl Cancer Res. 2014;3:494511. [DOI]
    Oerlemans C, Deckers R, Storm G, Hennink WE, Nijsen JFW. Evidence for a new mechanism behind HIFU-triggered release from liposomes. J Control Release. 2013;168:32733. [DOI] [PubMed]
    Liang HD, Tang J, Halliwell M. Sonoporation, drug delivery, and gene therapy. Proc Inst Mech Eng H. 2010;224:34361. [DOI] [PubMed]
    Aryal M, Papademetriou I, Zhang YZ, Power C, McDannold N, Porter T. MRI monitoring and quantification of ultrasound-mediated delivery of liposomes dually labeled with gadolinium and fluorophore through the blood-brain barrier. Ultrasound Med Biol. 2019;45:173342. [DOI] [PubMed] [PMC]
    Santos MA, Goertz DE, Hynynen K. Focused ultrasound hyperthermia mediated drug delivery using thermosensitive liposomes and visualized with in vivo two-photon microscopy. Theranostics. 2017;7:271831. [DOI] [PubMed] [PMC]
    Grüll H, Langereis S. Hyperthermia-triggered drug delivery from temperature-sensitive liposomes using MRI-guided high intensity focused ultrasound. J Control Release. 2012;161:31727. [DOI] [PubMed]
    Morse SV, Mishra A, Chan TG, T M de Rosales R, Choi JJ. Liposome delivery to the brain with rapid short-pulses of focused ultrasound and microbubbles. J Control Release. 2022;341:60515. [DOI] [PubMed]
    Zhu L, Nazeri A, Pacia CP, Yue Y, Chen H. Focused ultrasound for safe and effective release of brain tumor biomarkers into the peripheral circulation. PLoS One. 2020;15:e0234182. [DOI] [PubMed] [PMC]
    Abe K, Taira T. Focused ultrasound treatment, present and future. Neurol Med Chir (Tokyo). 2017;57:38691. [DOI] [PubMed] [PMC]
    Szewczyk B, Tarasek M, Campwala Z, Trowbridge R, Zhao Z, Johansen PM, et al. What happens to brain outside the thermal ablation zones? An assessment of needle-based therapeutic ultrasound in survival swine. Int J Hyperthermia. 2022;39:128393. [DOI] [PubMed]
    Jolesz FA. MRI-guided focused ultrasound surgery. Annu Rev Med. 2009;60:41730. [DOI] [PubMed] [PMC]
    Etame AB, Diaz RJ, Smith CA, Mainprize TG, Hynynen K, Rutka JT. Focused ultrasound disruption of the blood-brain barrier: a new frontier for therapeutic delivery in molecular neurooncology. Neurosurg Focus. 2012;32:E3. [DOI] [PubMed] [PMC]
    Focused Ultrasound Foundation. 2022 state of the field [Internet]. c2022 [cited Apr 14 2023]. Available from: https://cdn.fusfoundation.org/2022/11/01111233/Focused-Ultrasound-Foundation_State-of-the-Field-Report-2022_Nov1.pdf
    Ghanouni P, Pauly KB, Elias WJ, Henderson J, Sheehan J, Monteith S, et al. Transcranial MRI-guided focused ultrasound: a review of the technologic and neurologic applications. AJR Am J Roentgenol. 2015;205:1509. [DOI] [PubMed] [PMC]
    McDannold N, Maier SE. Magnetic resonance acoustic radiation force imaging. Med Phys. 2008;35:374858. [DOI] [PubMed] [PMC]
    MacDonell J, Patel N, Rubino S, Ghoshal G, Fischer G, Burdette EC, et al. Magnetic resonance-guided interstitial high-intensity focused ultrasound for brain tumor ablation. Neurosurg Focus. 2018;44:E11. [DOI] [PubMed] [PMC]
    Kim Y, Hall T, Xu Z, Cain C. Transcranial histotripsy therapy: a feasibility study. IEEE Trans Ultrason Ferroelectr Freq Control. 2014;61:58293. [DOI]
    Lu N, Hall TL, Choi D, Gupta D, Daou BJ, Sukovich JR, et al. Transcranial MR-guided histotripsy system. IEEE Trans Ultrason Ferroelectr Freq Control. 2021;68:291729. [DOI] [PubMed] [PMC]
    Lu N, Gupta D, Daou BJ, Fox A, Choi D, Sukovich JR, et al. Transcranial magnetic resonance-guided histotripsy for brain surgery: pre-clinical investigation. Ultrasound Med Biol. 2022;48:98110. [DOI] [PubMed] [PMC]
    ClinicalTrials.gov. ExAblate (magnetic resonance-guided focused ultrasound surgery) treatment of brain tumors [Internet]. [cited Apr 14 2023]. Available from: https://clinicaltrials.gov/ct2/show/study/NCT01473485?term=HIFU+OR+%22focused+ultrasound%22&cond=brain+tumor&draw=2&rank=4
    ClinicalTrials.gov. MRI-guided focused ultrasound feasibility study for brain tumors [Internet]. Massachusetts, Washington: InSightec; [cited Apr 14 2023]. Available from: https://clinicaltrials.gov/ct2/show/study/NCT00147056?term=HIFU+OR+%22focused+ultrasound%22&cond=brain+tumor&draw=2&rank=1
    Sukovich JR, Xu Z, Kim Y, Cao H, Nguyen TS, Pandey AS, et al. Targeted lesion generation through the skull without aberration correction using histotripsy. IEEE Trans Ultrason Ferroelectr Freq Control. 2016;63:67182. [DOI] [PubMed] [PMC]