Next Article in Journal
AEG-1 Regulates TWIK-1 Expression as an RNA-Binding Protein in Astrocytes
Next Article in Special Issue
Clinical Efficacy and Safety of Silicone Elastomer Sheet during Decompressive Craniectomy: Anti-Adhesive Role in Cranioplasty
Previous Article in Journal
Clinical Outcomes of Interlaminar Percutaneous Endoscopic Decompression for Degenerative Lumbar Spondylolisthesis with Spinal Stenosis
Previous Article in Special Issue
Common Challenges and Solutions Associated with the Preparation of Silicone-Injected Human Head and Neck Vessels for Anatomical Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Opinion

Focused Ultrasound in Neuroscience. State of the Art and Future Perspectives

by Giuseppe Roberto Giammalva 1, Cesare Gagliardo 2, Salvatore Marrone 1, Federica Paolini 1, Rosa Maria Gerardi 1, Giuseppe Emmanuele Umana 3,*, Kaan Yağmurlu 4, Bipin Chaurasia 5, Gianluca Scalia 6, Federico Midiri 2, Ludovico La Grutta 7, Luigi Basile 1, Carlo Gulì 1, Domenico Messina 1, Maria Angela Pino 1, Francesca Graziano 6, Silvana Tumbiolo 8, Domenico Gerardo Iacopino 1 and Rosario Maugeri 1
1
Neurosurgery Unit, Department of Biomedicine, Neurosciences & Advanced Diagnostics, School of Medicine, University of Palermo, 90127 Palermo, Italy
2
Section of Radiological Sciences, Department of Biomedicine, Neurosciences & Advanced Diagnostics, School of Medicine, University of Palermo, 90127 Palermo, Italy
3
Department of Neurosurgery, Cannizzaro Hospital, 95126 Catania, Italy
4
Departments of Neuroscience and Neurosurgery, University of Virginia Health System, Charlottesville, VA 22903, USA
5
Department of Neurosurgery, Neurosurgery Clinic, Birgunj 44300, Nepal
6
Department of Neurosurgery, Highly Specialized Hospital of National Importance “Garibaldi”, 95122 Catania, Italy
7
Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties-ProMISE, University of Palermo, 90127 Palermo, Italy
8
Division of Neurosurgery, Villa Sofia Hospital, 90146 Palermo, Italy
*
Author to whom correspondence should be addressed.
Brain Sci. 2021, 11(1), 84; https://doi.org/10.3390/brainsci11010084
Submission received: 21 December 2020 / Revised: 4 January 2021 / Accepted: 5 January 2021 / Published: 10 January 2021
(This article belongs to the Special Issue Neurosurgery and Neuroanatomy)

Abstract

:
Transcranial MR-guided Focused ultrasound (tcMRgFUS) is a surgical procedure that adopts focused ultrasounds beam towards a specific therapeutic target through the intact skull. The convergence of focused ultrasound beams onto the target produces tissue effects through released energy. Regarding neurosurgical applications, tcMRgFUS has been successfully adopted as a non-invasive procedure for ablative purposes such as thalamotomy, pallidotomy, and subthalamotomy for movement disorders. Several studies confirmed the effectiveness of tcMRgFUS in the treatment of several neurological conditions, ranging from motor disorders to psychiatric disorders. Moreover, using low-frequencies tcMRgFUS systems temporarily disrupts the blood–brain barrier, making this procedure suitable in neuro-oncology and neurodegenerative disease for controlled drug delivery. Nowadays, tcMRgFUS represents one of the most promising and fascinating technologies in neuroscience. Since it is an emerging technology, tcMRgFUS is still the subject of countless disparate studies, even if its effectiveness has been already proven in many experimental and therapeutic fields. Therefore, although many studies have been carried out, many others are still needed to increase the degree of knowledge of the innumerable potentials of tcMRgFUS and thus expand the future fields of application of this technology.

1. Introduction

The employment of acoustic energy for diagnostic purposes represented 20th century medicine as the achievement of a great goal. The US allowed bridging the complicated gap between semiotics and pathology, defined by the need for more precise diagnostics tools. Sound waves are elastic mechanical waves that can only propagate in a “non-empty” space or a medium. The human ear has a limited ability to perceive sound waves between 20 and 20,000 Hz, and higher frequency waves are called ultrasounds. Ultrasounds are created and received by transducers; they are constituted of piezoelectric crystals that produce ultrasound beams when electrically charged. Some physical parameters are characteristic of a specific medium influence ultrasound delivery; these are propagation speed (which depends on the density and compressibility of the medium) and the acoustic impedance (which measures the forces that oppose the transmission of waves). These parameters vary from medium to medium and, therefore, from tissue to tissue. From the interaction between ultrasound beams and cells, four main physical phenomena occur reflection, refraction, diffusion, absorption. In the last few decades, many studies showed how ultrasounds could be used not only for clinical or intraoperative diagnostic purposes also for therapeutics [1].
Using ultrasounds for therapeutic purposes dates back to the early 1940s when Lynn and Putnam first performed targeted, well-demarcated and controlled ablations in cortical/subcortical areas of mammals [2]. In the 1950s, the Fry brothers used high-intensity ultrasounds to produce targeted necrotic lesions into brain basal ganglia of previously craniotomized animals without affecting the neighboring brain parenchyma [3]. At this former stage, a craniotomy was inevitable to convey focused ultrasound directly into brain parenchyma. With the development of devices consisting of multiple phased-array transducers, it was possible to deliver enough thermal energy in a small and sharp focus through the intact skull directly to deep brain structures to induce a controlled thermal ablation [4]. The introduction of MRI proton resonance frequency (PRF) shift thermometry, which is employed for monitoring the energy release in the targeted area and surrounding tissues, along with the possibility to precisely modulate the delivery of acoustic energy, made tcMRgFUS brain ablation a more accurate and safe technique, paving the way for the such called non-invasive functional neurosurgery [5].
During tcMRgFUS treatments for functional neurological disorders, the patient is awake and compliant in order to receive quick feedback on his health state and perform a neurological evaluation at the end of each sonication. Although tcMRgFUS procedures were initially conducted with three Tesla MRI scanners, today it has been demonstrated that they can be safely and effectively also performed with widely used 1.5 Tesla MRI scanners [4,6,7], which enables a high-resolution intraoperative imaging thanks to a dedicated MR coil still not available for 3T integrated tcMRgFUS systems [8,9].
The procedure is performed by applying a stereotactic frame to the patient’s head after a full scalp shaving. During tcMRgFUS ablative procedures, the patient lays inside the MRI scanner [10]. Pre-operative and intra-operative brain MRI allows for a precise calculation of stereotactic coordinates of the target and to evaluate the lesion appearance made by FUS. Initial low-energy sonications are used to assess the thermal map, confirming the absence of areas of unwanted overheating outside the target point, and to ensure the focusing of ultrasound beams into the target area [11,12]. Once the focusing is confirmed, the energy is gradually increased. During this stage, the patient is continuously monitored and neurologically evaluated after every sonication. Once the target is even clinically confirmed, with the best neurological improvement without adverse events, sonication parameters (temperature (C/F), frequency (Hz), application duration (sec), power (W), energy (J) are adjusted to reach a peak temperature above 52 °C, which makes a permanent necrotic lesion [12,13].
Unlike other ablative techniques such as stereotaxic radiosurgery, tcMRgFUS does not require radiations, thus avoiding the possible side effects such as brain edema, headache, aseptic meningitis, actinic keratosis, and alopecia. Moreover, it is performed over a short hospitalization, and it could even be repeated [14,15,16,17,18].
In recent years, tcMRgFUS has caused a great reverberation within the scientific community for its remarkable applications in treating movement disorders such as drug-resistant parkinsonian tremor and essential tremor, a psychiatric disorder such as major depressive disorder (MDD) and obsessive-compulsive disorder (OCD) through non-invasive ablative procedures. Moreover, recent interest has been inspired by the capability of focused ultrasounds to produce targeted transient alterations of the blood-brain barrier (BBB) and the subsequent possible employment in the treatment of oncological pathologies by improving the permeability of BBB to specific chemotherapy and by altering some of their biomolecular mechanisms [19,20,21].
The purpose of this study is to investigate potential future applications of tcMRgFUS and to enlighten the prospective field of employment and procedures in which tcMRgFUS has just been adopted.

2. Materials and Methods

An extensive search of English literature was performed on PubMed (https://pubmed.ncbi.nlm.nih.gov) using the following keywords and their combinations: FUS, Blood-Brain-Barrier, thalamotomy, essential tremor, Parkinsonisms, movement disorders, capsulotomy, psychiatric disorders, trigeminal neuralgia, Guillan-Barrè Syndrome (GBS), neuro-oncology. Preclinical and clinical studies of the last four years were carefully reviewed, focusing on new perspectives in focused ultrasounds. Publication time was restricted to the last four years to achieve a careful insight on future perspectives of tcMRgFUS. English publications with available full text were included along with their most meaningful references. Studies dealing with the comparison between tcMRgFUS and different treatments in the same pathology were also included.
Exclusion criteria were: DBS and gamma-knife radiosurgery procedures exclusively treated publication dated over ten years, unavailability of full text, non-English publications, publications in which previously mentioned queries were not related to tcMRgFUS, publications in which clinical conditions were not treated by tcMRgFUS, or publications in with previously mentioned clinical conditions without any comparison to tcMRgFUS. Studies focusing on essential tremor were excluded since the effectiveness of this treatment has been already confirmed and accepted. Inclusion and exclusion criteria are summarized in Table 1.
Then, the results of our queries were screened according to the PRISMA statement. Recent tgMRgFUS applications in neurosurgery were described from the included studies, and they were systematically organized and grouped in perspective.

3. Results

From the literature search performed on PubMed with the abovementioned queries, we identified 96 unique articles. These were later screened for relevance: 42 studies were excluded according to our exclusion criteria, and 54 were included according to our inclusion criteria. Full-text was available for all of 54 included studies, which were included in our qualitative analysis.

4. Discussion

TcMRgFUS is a novel technology that has been recently introduced in neurosurgery for the treatment of movement disorders. Beyond its proven effectiveness in functional neurosurgery, tcMRgFUS has recently shown several potential employment fields, and other prospective applications have just been being explored. For this review, recent and upcoming applications of tcMRgFUS have been summarized from the available literature and grouped for fields of applications.

4.1. Neuromodulation

Since 1962, it has been shown that mechanical stimulation of a peripheral axon can trigger action potentials, and it can interfere with the state of membrane-cell channels [22]. It has recently been demonstrated that ultrasonic stimulation onto the peripheral nerve can produce muscular contraction through action potentials’ modulation [23]. This phenomenon may be attributed to a transient deformation of the axon sheath by mechanical pressure of sound waves and the subsequent ionic stream through channels in Ranviers’ nodes. In fact, it has been demonstrated that focused ultrasound may produce transient focal alterations of brain circuits that are also noticeable through electroencephalography [24,25] or focal neural disconnection by the delivery of neurotoxin through permeabilized BBB [26]. In this way, low intensity focused ultrasounds (LIFU) (1–100 mW/cm2) may be used to excite or inhibit targeted clusters of neurons, to modulate nerve conduction, to modulate ionic stream, to promote neuroplasticity, to stimulate and further to non-invasively map the cerebral cortex, the deep brain nuclei and the white matter tracts [27,28,29,30,31,32,33,34,35,36,37].

4.2. Drug-Resistant Epilepsy

Recently tcMRgFUS has shown potential effectiveness in the treatment of drug-resistant epilepsy. About 20–40% of epileptic patients are affected by drug-resistant epilepsy, and they often undergo surgical treatment. Non-resective neuromodulatory treatments are performed by deep brain stimulation (DBS), vagus nerve stimulation (VNS), or responsive neurostimulation (RNS) [38].
Regarding the ablative surgery, it has been reported the first case of a female patient affected by mesial temporal lobe epilepsy undergone tcMRgFUS ablation. After the treatment, the patient was seizure-free and improved her quality of life at 1-year follow-up. Besides ablative applications, tcMRgFUS can modulate neurological functions by using low-frequency stimulation. Experiences are limited to preclinical studies; few clinical trials are still ongoing [39].

4.3. Drug-Resistant Trigeminal Neuralgia

Drug-resistant trigeminal neuralgia is a debilitating pain condition that often is treated by surgery. In such cases, recently, it has been shown that tcMRgFUS bilateral central-lateral thalamotomy effectively treats patients affected by drug-resistant trigeminal neuralgia, with 63% of good outcomes at three months, 88% at 1 year and 100% at last follow-up with no adverse events [40].

4.4. Chronic Neuropathic Pain

There is a lowering of the excitability threshold in chronic pain. Lower intensity stimulations suffice to activate spikes and transmit the painful signal along with dedicated neurological pathways.
It has been recently shown in a preclinical study that pulsed FUS on dorsal root ganglia may be an effective treatment of chronic neuropathic pain since it can determine a temporary reduction of mechanical allodynia secondary to an iatrogenic lesion of rat peroneal nerve [41]. The physiological mechanisms which could justify temporary analgesia are still unclear. However, it could be associated with the mechanical change of the ionic channels induced by acoustic waves and improved microcirculation and nerve conduction velocity. It has been found in a preclinical model of diabetic neuropathy [42].
Regarding clinical applications of focused ultrasound, it has been recently demonstrated that tcMRgFUS postero-central lateral thalamotomy is an effective treatment of drug-resistant chronic neuropathic pain and trigeminal neuralgia [40]. In contrast, local MRgFUS ablation effectively treats pain related to lumbar bone metastases [43,44].

4.5. Psychiatric Disorders

Patients affected by obsessive-compulsive disorder (OCD) and major depressive disorder (MDD) may be refractory to pharmacotherapy and cognitive-behavioral treatment. In case of refractory OCD, capsulotomy is the most effective procedure to control OCD symptoms. Capsulotomy can be performed by several techniques, such as radiofrequency, radiosurgery, and DBS. Among these, tcMRgFUS is an effective procedure aimed to disrupt hyperactivated circuits between the dorsal thalamus and prefrontal cortex. It is possible through precise targeting guided by MRI real-time monitoring, avoiding the exposure to ionizing radiation and radiation side effects [45,46,47,48].
In 2014, for the first time, four patients affected by medically refractory OCD were successfully treated by tcMRgFUS; no side effect was encountered after a six-month follow up [49]. As regards MDD, several brain structures may be identified as therapeutic targets. Drug refractory MDD is treated by anterior cingulotomy, subcaudate tractotomy, limbic leucotomy, and anterior capsulotomy [50]. It has recently been reported that tcMRgFUS bilateral anterior capsulotomy is an effective treatment of drug-resistant MDD in a 56-years-old patient, capable of alleviating symptoms and improving neuropsychological tests and quality of life [51].

4.6. Ischemic and Hemorrhagic Stroke

Despite the incidence of ischemic and hemorrhagic stroke, only a fraction of patients benefit from medical (thrombolysis) and surgical treatment (clot evacuation/decompressive craniectomy) [52].
The implementation of ultrasounds in the treatment of ischemic stroke dates back to 2000, when transcranial doppler ultrasonography was used to monitor and improve the rate of medical thrombolysis [53]. It has been hypothesized that TcMrgFUS could further enhance the treatment of ischemic and hemorrhagic strokes through the targeted delivery of acoustic energy to the clot, thus performing the so-called “sonothrombolysis”. In 2013 and then in 2014, the effectiveness of tcMRgFUS sonotrhombolysis was demonstrated in the in-vivo porcine model and ex-vivo human model of intracranial hemorrhage. In these models, sonications were used to liquefy clot while MRI controlled treatment targeting and temperature monitoring. Clot liquefaction was achieved in both models between 2 and 24 h after clot formation [54,55]. Recently, promising results have been obtained by the adjunct of microbubbles to tMRgFUS sonotrombolysis in ischemic stroke, even the effectiveness needs to be confirmed by further in-vivo studies [56].

4.7. Modulation of Blood-Brain Barrier Permeability

In the 1970s, it has been described as the possibility of altering the BBB permeability by hypertonic solutions [57]. In recent years, it has been explored the potential to modulate BBB permeability by the use of pulsatile LIFU. In fact, while high-intensity ultrasound (1000 W/cm2) can induce coagulative necrotic lesions in targeted regions of the brain, pulsatile low-intensity ultrasound can interfere with BBB permeability without causing permanent lesion. Oscillations induced by pulsed LIFU result from the compression-release cycle and they would be able to alter the permeability of BBB tight junctions, transiently undocking tight bonds between endotheliocytes and then open that portion of the barrier (lasting about 4–6 h) [57]. One clinical study performed pulsated LIFU on 21 GBM patients who are under carboplatin treatment and concluded that pulsated LIFU might increase the drug therapies without inducing neurotoxicity [58]. In particular, BBB permeability would be mostly influenced by short pulse lengths and low pulse repetition frequencies of focused ultrasounds [59]. The evidence that LIFU can alter BBB permeability recently paved the way to new diagnostic and therapeutic possibilities. LIFU has been used for diagnostic purposes to obtain liquid biopsy in a mouse model of glioblastoma (GBM) [60,61]. In this case, the localized BBB opening induced by LIFU could increase vascular permeability and, therefore, the outflow of tumoral markers into the bloodstream and thus their detection [60,61]. Moreover, it has been demonstrated in a rat model of GBM that the adjunct of sonosensitizers such as 5-aminolevulinic acid could determine localized cytotoxic effects even in absence of thermal effects with the so called sonodynamic therapy [60,61].
Regarding the therapeutic application of LIFU, several other studies have been conducted in oncological and neurodegenerative diseases. In particular, it has been demonstrated that LIFU-induced BBB permeabilization could be enhanced by the administration of microbubbles [62], and this technique may be effective in improving chemotherapy delivery through BBB for the treatment of brain tumors [63,64,65]. The evidence of biochemical dyes extravasation confirmed the effectiveness and safety of microbubble administration in increasing BBB permeability during LIFU treatment from one to 96 h after treatment in the absence of lesions. This evidence paves the way to obtaining a microbubble-mediate chemical treatment by inserting drug moles inside nanoparticles during LIFU treatment to obtain the localized emission of the compound to the targeted tissue, thus maximizing the local concentration and reducing systemic toxicity. This technique has been named “sonopermeation” [62,66]. The combination of LIFU and high intensity focused ultrasounds (HIFU) recently implemented microbubble BBB permeabilization to alter BBB’s permeability and subsequently convey poly (2-ethyl-butyl cyanoacrylate) nanoparticle-stabilized microbubbles through the extracellular matrix of a rat model of melanoma brain metastasis. Further microscopic studies underlined in some animals a directly proportional correlation between the rate of BBB permeabilization and the distribution of nanoparticles [67]. This technique presents several limitations. One of them is related to the range of effective sonication parameters used to obtain a therapeutic effect and acceptable impact on tissue health, avoiding complications such as hemorrhage, edema, extensive inflammation, ischemia, apoptosis, prolonged BBB permeabilization. Another important limitation is related to the correct dose of microbubble to infuse. Several factors that can influence how microbubbles respond to ultrasounds, and there are no standard protocols in these procedures. Moreover, inhomogeneities in BBB can lead to different effects related to a different location. In particular, transient enhancing in BBB permeability can lead to increased complication rate [62,66].
While clinical applications of tcMRgFUS for localized chemotherapy delivery through BBB sonopermeation is still controversial [68], several studies have already enlightened a localized pro-inflammatory activity of tcMRgFUS due to the stimulation of innate and acquired immune responses through local immunomodulation and gene-regulation [69,70]. This paves the way to novel frontiers to treat brain tumors and the treatment of neurodegenerative diseases through gene-delivery therapy. Sonoselective gene-delivery has recently been possible through endothelial-sonoselective transfection of the cerebral blood vessels without opening BBB without inducing sterile inflammation. This novel technique could promote angiogenesis, releasing nerve growth factors, and promoting neurogenesis through the recruitment of stem cells [71].
Regarding new perspectives in the treatment of neurodegenerative diseases, the delivery of a gene-liposome system containing glial cell line-derived neurotrophic factor has been recently possible by applying pulsed FUS with microbubbles in a transgenic mouse model of Huntington disease. This technique has been possible to obtain an effective gene therapy with reduced polyglutamine-expanded aggregates and improved motor function [72].
Based on the evidence of BBB dysfunction since the early stage of neurodegenerative diseases [73,74], it has been recently possible to demonstrate that a targeted gene and protein therapy in combination with FUS sonopermeation and microbubbles is capable of attenuating the nigrostriatal pathway degeneration since the early stage in a mouse model of Parkinson Disease (PD) [75].
As regards current clinical scenarios, tcMRgFUS sonopermeation has recently been used to induce a transitory BBB permeation localized at the primary motor cortex of some ALS volunteers, without clinically and electrophysiologically noticeable complications, opening up the possibility of targeted therapy such as localized delivery of superoxide dismutase (SOD) inhibitors [76]. Previously, the possibility of reducing up to 20% of beta-amyloid plaques has been already described in a rat model of Alzheimer Disease (AD) undergone tcMRgSUS sonopermeation for targeted delivery of exogenous antibodies [77,78]. The local effect of sonopermeation on AD beta-amyloid plaques would be further increased by the BBB crossing of endogenous antibodies from the systemic bloodstream and the local activation of microglia [79]. Since microglial activation, the more significant beta-amyloid could be internalized in lysosomes, leading to improved memory functions in mouse models of AD [80]. This virtuous circle could be probably sustained also by enhanced kinase inhibitors (i.e., GSK-3 inhibitor) delivery through a permeabilized BBB [81].
In conclusion, the whole backdrop of FUS application in BBB modulation and neurodegenerative disease treatment is a very prospective study field. Nowadays, several other applications are still to be tested.

4.8. Progression in Neuro-Oncology

The neuro-oncological field experiences are limited to small studies in patients with no surgical treatment possibility [82]. The thermal effect of HIFU, according to temperature, can achieve both ablative effect (55–60 °C or 240 CEM 43°, cumulative equivalent minutes at 43 °C) and radiosensitization induced by mild hyperthermia [83].
Moreover, the release of tumor antigens induced by necrosis can enhance antitumor immunity. Heat generation arises from sonication and cavitation. Cavitation also has a mechanical effect, essential for BBB disruption [84].
In this regard, the first study was a Phase I/II study with untreatable patients affected by recurrent malignant gliomas. They underwent a craniectomy and a biopsy of the tumor; 7–10 days later, a tcMrgFUS treatment was performed. The first patient had a tumor volume of 9 cm3; sonications did not achieve ablation temperature because of attenuation by synthetic dural substitute implanted during previous operations. The second patient had a successful ablation of 50% of tumor volume; after the procedure, no radiological or neurological adverse events were noticed, apart from a transient worsening of her pre-existing dysphasia that improved with steroid therapy. She has undergone surgery to complete removal, and histological analysis showed necrosis induced by FUS. The third patient had a complete ablation of all 5cm3 tumor volume; a 24 h MRI showed a new lesion in the right cerebral peduncle, with no clinical findings, resolving spontaneously. He had a recurrence after four months [85]. The first successful tcMRgFUS ablation of brain tumor was reported in a 17-year-old female affected by non-surgical anaplastic astrocytoma, whose volume decreased six months after tcMRgFUS treatment [86].
First sonic ablations without previous craniectomy were attempted in 2010. Treatments were performed in three patients with glioblastoma, not eligible for the surgical procedure, who previously underwent chemotherapy and radiotherapy. Unfortunately, they did not reach sufficient US power to achieve ablation due to skull characteristics [87].
The first successful non-invasive thermal ablation using tcMrgFUS was performed in 2014. The procedure allowed physicians to ablate 10% of tumor volume; however, time consumption and overheating of the skull represented two of the most critical limits of the procedure [88].
Based on these studies, tcMRgFUS seems to be a feasible treatment option for brain tumor ablation, but other studies and clinical experiences are still needed. In the attempt to achieve a total ablation of lesions, three Phase I trials are still ongoing [89,90,91].

5. Conclusions

MRgFUS is a versatile and promising technique whit several and still undiscovered applications in diagnostic and therapeutic fields. However, since its several areas of applications, more preclinical and clinical studies are still needed. To sum up, tcMRgFUS shows several ambitious applications and new potential capabilities that deserve continuous meticulous investigations. On the other hand, nowadays, tcMRgFUS procedures for functional neurological disorders affect the field as a valid non-invasive, safe, and effective therapeutic option.

Author Contributions

Conceptualization, G.R.G., R.M., G.E.U. and D.G.I.; methodology, G.R.G., C.G. (Cesare Gagliardo) and S.M.; software, F.P., R.M.G., G.E.U. and B.C.; validation, G.S., F.M., L.L.G. and L.B.; formal analysis, C.G. (Carlo Gulì), D.M., M.A.P. and F.G.; investigation, S.T., D.G.I., K.Y. and R.M.; resources, D.G.I.; data curation, R.M.; writing—original draft preparation, G.R.G., C.G. (Cesare Gagliardo) and S.M.; writing—review and editing, D.G.I., K.Y., R.M. and G.E.U.; visualization, D.G.I.; supervision, K.Y., R.M. and G.E.U.; project administration, D.G.I.; funding acquisition, K.Y. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Informed Consent Statement

Informed consent was obtained from all subjects involved in the study.

Acknowledgments

We thank the reviewer and the editor for revising this paper and making it successful.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Pino, M.A.; Imperato, A.; Musca, I.; Maugeri, R.; Giammalva, G.R.; Costantino, G.; Graziano, F.; Meli, F.; Francaviglia, N.; Iacopino, D.G.; et al. New hope in brain glioma surgery: The role of intraoperative ultrasound. A review. Brain Sci. 2018, 8, 202. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Lynn, J.G.; Putnam, T.J. Histology of cerebral lesions produced by focused ultrasound. Am. J. Pathol. 1944, 20, 637–649. [Google Scholar] [PubMed]
  3. Fry, F.J. Precision high intensity focusing ultrasonic machines for surgery. Am. J. Phys. Med. 1958, 37, 152–156. [Google Scholar] [CrossRef] [PubMed]
  4. Iacopino, D.G.; Gagliardo, C.; Giugno, A.; Giammalva, G.R.; Napoli, A.; Maugeri, R.; Graziano, F.; Valentino, F.; Cosentino, G.; D’Amelio, M.; et al. Preliminary experience with a transcranial magnetic resonance–guided focused ultrasound surgery system integrated with a 1.5-T MRI unit in a series of patients with essential tremor and Parkinson’s disease. Neurosurg. Focus 2018, 44, E7. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Quadri, S.A.; Waqas, M.; Khan, I.; Khan, M.A.; Suriya, S.S.; Farooqui, M.; Fiani, B. High-intensity focused ultrasound: Past, present, and future in neurosurgery. Neurosurg. Focus 2018, 44. [Google Scholar] [CrossRef] [Green Version]
  6. Giammalva, G.R.; Gagliardo, C.; Maugeri, R.; Midiri, M.; Iacopino, D.G. Transcranial MRgFUS for movement disorder: Toward a wider and affordable employment for functional neurosurgery through 1.5-T MRI? J. Neurosurg. 2018, 129, 843. [Google Scholar] [CrossRef] [Green Version]
  7. Iacopino, D.G.; Giugno, A.; Maugeri, R.; Gagliardo, C.; Franzini, A.; Catalano, C.; Midiri, M. Is there still a role for lesioning in functional neurosurgery? Preliminary Italian (and world-first) experience with a transcranial MRI-guided focused ultrasound surgery system operating at 1.5 Tesla. J. Neurosurg. Sci. 2017, 61, 681–683. [Google Scholar]
  8. Gagliardo, C.; Midiri, M.; Cannella, R.; Napoli, A.; Wragg, P.; Collura, G.; Marrale, M.; Vincenzo Bartolotta, T.; Catalano, C.; Lagalla, R. Transcranial magnetic resonance-guided focused ultrasound surgery at 1.5T: A technical note. Neuroradiol. J. 2019, 32, 132–138. [Google Scholar] [CrossRef] [Green Version]
  9. Gagliardo, C.; Cannella, R.; Quarrella, C.; D’Amelio, M.; Napoli, A.; Bartolotta, T.V.; Catalano, C.; Midiri, M.; Lagalla, R. Intraoperative imaging findings in transcranial MR imaging-guided focused ultrasound treatment at 1.5T may accurately detect typical lesional findings correlated with sonication parameters. Eur. Radiol. 2020. [Google Scholar] [CrossRef]
  10. Fishman, P.S.; Frenkel, V. Focused ultrasound: An emerging therapeutic modality for neurologic disease. Neurotherapeutics 2017, 14, 393–404. [Google Scholar] [CrossRef] [Green Version]
  11. Fishman, P.S.; Frenkel, V. Treatment of movement disorders with focused ultrasound. J. Cent. Nerv. Syst. Dis. 2017, 9. [Google Scholar] [CrossRef] [PubMed]
  12. Gagliardo, C.; Marrale, M.; D’Angelo, C.; Cannella, R.; Collura, G.; Iacopino, G.; D’Amelio, M.; Napoli, A.; Bartolotta, T.V.; Catalano, C.; et al. Transcranial magnetic resonance imaging-guided focused ultrasound treatment at 1.5 T: A retrospective study on treatment and patient-related parameters obtained from 52 procedures. Front. Phys. 2020, 7, 223. [Google Scholar] [CrossRef]
  13. Bond, A.E.; Elias, W.J. Predicting lesion size during focused ultrasound thalamotomy: A review of 63 lesions over 3 clinical trials. Neurosurg. Focus 2018, 44, 1–6. [Google Scholar] [CrossRef] [PubMed]
  14. Dallapiazza, R.F.; Lee, D.J.; De Vloo, P.; Fomenko, A.; Hamani, C.; Hodaie, M.; Kalia, S.K.; Fasano, A.; Lozano, A.M. Outcomes from stereotactic surgery for essential tremor. J. Neurol. Neurosurg. Psychiatry 2019, 90, 474–482. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Valentino, F.; Cosentino, G.; Maugeri, R.; Giammalva, R.; Iacopino, G.D.; Marrale, M.; Bartolotta, T.V.; Gagliardo, C. Is transcranial magnetic resonance imaging-guided focused ultrasound a repeatable treatment option? Case report of a retreated patient with tremor combined with Parkinsonism. Oper. Neurosurg. 2020, 18, 557–582. [Google Scholar] [CrossRef]
  16. Fasano, A.; De Vloo, P.; Llinas, M.; Hlasny, E.; Kucharczyk, W.; Hamani, C.; Lozano, A.M. Magnetic resonance imaging-guided focused ultrasound thalamotomy in Parkinson tremor: Reoperation after benefit decay. Mov. Disord. 2018, 33, 848–849. [Google Scholar] [CrossRef]
  17. Weidman, E.K.; Kaplitt, M.G.; Strybing, K.; Levi Chazen, J. Repeat magnetic resonance imaging-guided focused ultrasound thalamotomy for recurrent essential tremor: Case report and review of MRI findings. J. Neurosurg. 2020, 132, 211–216. [Google Scholar] [CrossRef]
  18. Umana, G.E.; Raudino, G.; Alberio, N.; Inserra, F.; Giovinazzo, G.; Fricia, M.; Chiriatti, S.; Nicoletti, G.F.; Cicero, S.; Scalia, G. Slit-like hypertensive hydrocephalus: Report of a late, complex, and multifactorial complication in an oncologic patient. Surg. Neurol. Int. 2020, 11. [Google Scholar] [CrossRef]
  19. Konofagou, E.E.; Tunga, Y.-S.; Choia, J.; Deffieuxa, T.; Baseria, B.; Vlachosa, F. Ultrasound-induced blood-brain barrier opening. Curr. Pharm. Biotechnol. 2012, 13, 1332–1345. [Google Scholar] [CrossRef]
  20. Han, M.; Hur, Y.; Hwang, J.; Park, J. Biological effects of blood–brain barrier disruption using a focused ultrasound. Biomed. Eng. Lett. 2017, 7, 115–120. [Google Scholar] [CrossRef]
  21. Greene, C.; Campbell, M. Tight junction modulation of the blood brain barrier: CNS delivery of small molecules. Tissue Barriers 2016, 4, e1138017. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Julian, F.J.; Goldman, D.E. The effects of mechanical stimulation on some electrical properties of axons. J. Gen. Physiol. 1962, 46, 297–313. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Downs, M.E.; Lee, S.A.; Yang, G.; Kim, S.; Wang, Q.; Konofagou, E.E. Non-invasive peripheral nerve stimulation via focused ultrasound in vivo. Phys. Med. Biol. 2018, 63. [Google Scholar] [CrossRef] [PubMed]
  24. Mueller, J.; Legon, W.; Opitz, A.; Sato, T.F.; Tyler, W.J. Transcranial focused ultrasound modulates intrinsic and evoked EEG dynamics. Brain Stimul. 2014, 7, 900–908. [Google Scholar] [CrossRef] [Green Version]
  25. Crusio, W.E.; Dong, H.; Radeke, H.H.; Rezaei, N.; Xiao, J. MR-guided transcranial focused ultrasound. In Advances in Experimental Medicine and Biology; Aubry, J.F., Tanter, M., Eds.; Springer New York LLC: New York, NY, USA, 2016; Volume 880, pp. 97–111. [Google Scholar]
  26. Zhang, Y.; Tan, H.; Bertram, E.H.; Aubry, J.F.; Lopes, M.B.; Roy, J.; Dumont, E.; Xie, M.; Zuo, Z.; Klibanov, A.L.; et al. Non-invasive, focal disconnection of brain circuitry using magnetic resonance-guided low-intensity focused ultrasound to deliver a neurotoxin. Ultrasound Med. Biol. 2016, 42, 2261–2269. [Google Scholar] [CrossRef]
  27. Adrianov, O.S.; Vykhodtseva, N.I.; Fokin, V.F.; Avirom, V.M. Method of local application of focused ultrasound to deep brain structures of an unrestrained unanesthetized animal. Bull. Exp. Biol. Med. 1984, 98, 992–994. [Google Scholar] [CrossRef]
  28. Juan, E.J.; González, R.; Albors, G.; Ward, M.P.; Irazoqui, P. Vagus nerve modulation using focused pulsed ultrasound: Potential applications and preliminary observations in a rat. Int. J. Imaging Syst. Technol. 2014, 24, 67–71. [Google Scholar] [CrossRef] [Green Version]
  29. Verhagen, L.; Gallea, C.; Folloni, D.; Constans, C.; Jensen, D.E.A.; Ahnine, H.; Roumazeilles, L.; Santin, M.; Ahmed, B.; Lehericy, S.; et al. Offline impact of transcranial focused ultrasound on cortical activation in primates. Elife 2019, 8. [Google Scholar] [CrossRef]
  30. Tufail, Y.; Matyushov, A.; Baldwin, N.; Tauchmann, M.L.; Georges, J.; Yoshihiro, A.; Tillery, S.I.H.; Tyler, W.J. Transcranial pulsed ultrasound stimulates intact brain circuits. Neuron 2010, 66, 681–694. [Google Scholar] [CrossRef] [Green Version]
  31. King, R.L.; Brown, J.R.; Pauly, K.B. Localization of ultrasound-induced invivo neurostimulation in the mouse model. Ultrasound Med. Biol. 2014, 40, 1512–1522. [Google Scholar] [CrossRef]
  32. Deffieux, T.; Younan, Y.; Wattiez, N.; Tanter, M.; Pouget, P.; Aubry, J.F. Low-intensity focused ultrasound modulates monkey visuomotor behavior. Curr. Biol. 2013, 23, 2430–2433. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  33. Kubanek, J. Neuromodulation with transcranial focused ultrasound. Neurosurg. Focus 2018, 44, E14. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Dallapiazza, R.F.; Timbie, K.F.; Holmberg, S.; Gatesman, J.; Lopes, M.B.; Price, R.J.; Miller, G.W.; Elias, W.J. Noninvasive neuromodulation and thalamic mapping with low-intensity focused ultrasound. J. Neurosurg. 2018, 128, 875–884. [Google Scholar] [CrossRef] [PubMed]
  35. Legon, W.; Bansal, P.; Tyshynsky, R.; Ai, L.; Mueller, J.K. Transcranial focused ultrasound neuromodulation of the human primary motor cortex. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef] [Green Version]
  36. Fini, M.; Tyler, W.J. Transcranial focused ultrasound: A new tool for non-invasive neuromodulation. Int. Rev. Psychiatry 2017, 29, 168–177. [Google Scholar] [CrossRef]
  37. Tyler, W.J.; Lani, S.W.; Hwang, G.M. Ultrasonic modulation of neural circuit activity. Curr. Opin. Neurobiol. 2018, 50, 222–231. [Google Scholar] [CrossRef]
  38. Ranjan, M.; Boutet, A.; Bhatia, S.; Wilfong, A.; Hader, W.; Lee, M.R.; Rezai, A.R.; Adelson, P.D. Neuromodulation beyond neurostimulation for epilepsy: Scope for focused ultrasound. Expert Rev. Neurother. 2019, 19, 937–943. [Google Scholar] [CrossRef]
  39. Abe, K.; Yamaguchi, T.; Hori, H.; Sumi, M.; Horisawa, S.; Taira, T.; Hori, T. Magnetic resonance-guided focused ultrasound for mesial temporal lobe epilepsy: A case report. BMC Neurol. 2020, 20. [Google Scholar] [CrossRef]
  40. Gallay, M.N.; Moser, D.; Jeanmonod, D. MR-guided focused ultrasound central lateral thalamotomy for trigeminal neuralgia. Single center experience. Front. Neurol. 2020, 11. [Google Scholar] [CrossRef]
  41. Prabhala, T.; Hellman, A.; Walling, I.; Maietta, T.; Qian, J.; Burdette, C.; Neubauer, P.; Shao, M.; Stapleton, A.; Thibodeau, J.; et al. External focused ultrasound treatment for neuropathic pain induced by common peroneal nerve injury. Neurosci. Lett. 2018, 684, 145–151. [Google Scholar] [CrossRef]
  42. Tan, J.S.; Lin, C.C.; Chen, G.S. Vasomodulation of peripheral blood flow by focused ultrasound potentiates improvement of diabetic neuropathy. BMJ Open Diabetes Res. Care 2020, 8, e001004. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Huisman, M.; ter Haar, G.; Napoli, A.; Hananel, A.; Ghanouni, P.; Lövey, G.; Nijenhuis, R.J.; Van Den Bosch, M.A.A.J.; Rieke, V.; Majumdar, S.; et al. International consensus on use of focused ultrasound for painful bone metastases: Current status and future directions. Int. J. Hyperth. 2015, 31, 251–259. [Google Scholar] [CrossRef] [PubMed]
  44. Nicoletti, G.F.; Umana, G.E.; Graziano, F.; Cal, A.; Fricia, M.; Cicero, S.; Scalia, G. Cauda equina syndrome caused by lumbar leptomeningeal metastases from lung adenocarcinoma mimicking a schwannoma. Surg. Neurol. Int. 2020. [Google Scholar] [CrossRef] [PubMed]
  45. Rück, C.; Karlsson, A.; Steele, J.D.; Edman, G.; Meyerson, B.A.; Ericson, K.; Nyman, H.; Åsberg, M.; Svanborg, P. Capsulotomy for obsessive-compulsive disorder: Long-term follow-up of 25 patients. Arch. Gen. Psychiatry 2008, 65, 914–922. [Google Scholar] [CrossRef] [PubMed]
  46. Pepper, J.; Zrinzo, L.; Hariz, M. Anterior capsulotomy for obsessive-compulsive disorder: A review of old and new literature. J. Neurosurg. 2019, 1–10. [Google Scholar] [CrossRef]
  47. Jung, H.H.; Chang, W.S.; Rachmilevitch, I.; Tlusty, T.; Zadicario, E.; Chang, J.W. Different magnetic resonance imaging patterns after transcranial magnetic resonance-guided focused ultrasound of the ventral intermediate nucleus of the thalamus and anterior limb of the internal capsule in patients with essential tremor or obsessive-comp. J. Neurosurg. 2015, 122, 162–168. [Google Scholar] [CrossRef]
  48. Siragusa, M.A.; Réméniéras, J.P.; Bouakaz, A.; Escoffre, J.M.; Patat, F.; Dujardin, P.A.; Brizard, B.; Belzung, C.; Camus, V.; El-Hage, W.; et al. A systematic review of ultrasound imaging and therapy in mental disorders. Prog. Neuropsychopharmacol. Biol. Psychiatry 2020, 101, 109919. [Google Scholar] [CrossRef]
  49. Jung, H.H.; Kim, S.J.; Roh, D.; Chang, J.G.; Chang, W.S.; Kweon, E.J.; Kim, C.-H.; Chang, J.W. Bilateral thermal capsulotomy with MR-guided focused ultrasound for patients with treatment-refractory obsessive-compulsive disorder: A proof-of-concept study. Mol. Psychiatry 2015, 20, 1205–1211. [Google Scholar] [CrossRef] [Green Version]
  50. Volpini, M.; Giacobbe, P.; Cosgrove, G.R.; Levitt, A.; Lozano, A.M.; Lipsman, N. The history and future of ablative neurosurgery for major depressive disorder. Stereotact. Funct. Neurosurg. 2017, 95, 216–228. [Google Scholar] [CrossRef]
  51. Kim, M.; Kim, C.H.; Jung, H.H.; Kim, S.J.; Chang, J.W. Treatment of major depressive disorder via magnetic resonance-guided Focused Ultrasound Surgery. Biol. Psychiatry 2018, 83, e17–e18. [Google Scholar] [CrossRef]
  52. Ilyas, A.; Chen, C.J.; Ding, D.; Romeo, A.; Buell, T.J.; Wang, T.R.; Kalani, M.Y.S.; Park, M.S. Magnetic resonance-guided, high-intensity focused ultrasound sonolysis: Potential applications for stroke. Neurosurg. Focus 2018, 44. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Alexandrov, A.V.; Demchuk, A.M.; Felberg, R.A.; Christou, I.; Barber, P.A.; Burgin, W.S.; Malkoff, M.; Wojner, A.W.; Grotta, J.C. High rate of complete recanalization and dramatic clinical recovery during tPA infusion when continuously monitored with 2-MHz transcranial Doppler monitoring. Stroke 2000, 31, 610–614. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Monteith, S.J.; Harnof, S.; Medel, R.; Popp, B.; Wintermark, M.; Lopes, M.B.S.; Kassell, N.F.; Elias, W.J.; Snell, J.; Eames, M.; et al. Minimally invasive treatment of intracerebral hemorrhage with magnetic resonance-guided focused ultrasound: Laboratory investigation. J. Neurosurg. 2013, 118, 1035–1045. [Google Scholar] [CrossRef] [PubMed]
  55. Harnof, S.; Zibly, Z.; Hananel, A.; Monteith, S.; Grinfeld, J.; Schiff, G.; Kulbatski, I.; Kassell, N. Potential of magnetic resonance-guided focused ultrasound for intracranial hemorrhage: An in vivo feasibility study. J. Stroke Cerebrovasc. Dis. 2014, 23, 1585–1591. [Google Scholar] [CrossRef] [PubMed]
  56. Auboire, L.; Sennoga, C.A.; Hyvelin, J.M.; Ossant, F.; Escoffre, J.M.; Tranquart, F.; Bouakaz, A. Microbubbles combined with ultrasound therapy in ischemic stroke: A systematic review of in-vivo preclinical studies. PLoS ONE 2018, 13, e0191788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Neuwelt, E.A.; Maravilla, K.R.; Frenkel, E.P.; Rapaport, S.I.; Hill, S.A.; Barnett, P.A. Osmotic blood-brain barrier disruption. Computerized tomographic monitoring of chemotherapeutic agent delivery. J. Clin. Investig. 1979, 64, 684–688. [Google Scholar] [CrossRef] [Green Version]
  58. 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] [Green Version]
  59. 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]
  60. 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. [Google Scholar] [CrossRef]
  61. 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. [Google Scholar] [CrossRef] [Green Version]
  62. McMahon, D.; Poon, C.; Hynynen, K. Evaluating the safety profile of focused ultrasound and microbubble-mediated treatments to increase blood-brain barrier permeability. Expert Opin. Drug Deliv. 2019, 16, 129–142. [Google Scholar] [CrossRef] [PubMed]
  63. Carpentier, A.; Canney, M.; Vignot, A.; Reina, V.; Beccaria, K.; Horodyckid, C.; Karachi, C.; Leclercq, D.; Lafon, C.; Chapelon, J.Y.; et al. Clinical trial of blood-brain barrier disruption by pulsed ultrasound. Sci. Transl. Med. 2016, 8. [Google Scholar] [CrossRef] [PubMed]
  64. Beccaria, K.; Canney, M.; Bouchoux, G.; Puget, S.; Grill, J.; Carpentier, A. Blood-brain barrier disruption with low-intensity pulsed ultrasound for the treatment of pediatric brain tumors: A review and perspectives. Neurosurg. Focus 2020, 48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. 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. Neurooncol. 2019, 144, 33–41. [Google Scholar] [CrossRef] [Green Version]
  66. Snipstad, S.; Sulheim, E.; de Lange Davies, C.; Moonen, C.; Storm, G.; Kiessling, F.; Schmid, R.; Lammers, T. Sonopermeation to improve drug delivery to tumors: From fundamental understanding to clinical translation. Expert Opin. Drug Deliv. 2018, 15, 1249–1261. [Google Scholar] [CrossRef]
  67. Baghirov, H.; Snipstad, S.; Sulheim, E.; Berg, S.; Hansen, R.; Thorsen, F.; Mørch, Y.; De Lange Davies, C.; Åslund, A.K.O. Ultrasound-mediated delivery and distribution of polymeric nanoparticles in the normal brain parenchyma of a metastatic brain tumour model. PLoS ONE 2018, 13, e0191102. [Google Scholar] [CrossRef] [Green Version]
  68. Sulheim, E.; Mørch, Y.; Snipstad, S.; Borgos, S.E.; Miletic, H.; Bjerkvig, R.; de Lange Davies, C.; 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] [Green Version]
  69. Liu, H.L.; Hsieh, H.Y.; Lu, L.A.; Kang, C.W.; Wu, M.F.; Lin, C.Y. Low-pressure pulsed focused ultrasound with microbubbles promotes an anticancer immunological response. J. Transl. Med. 2012, 10. [Google Scholar] [CrossRef] [Green Version]
  70. Kovacsa, Z.I.; Kima, S.; Jikariaa, N.; Qureshia, F.; Miloa, B.; Lewisa, B.K.; Breslera, M.; Burksa, S.R.; Franka, J.A. Disrupting the blood-brain barrier by focused ultrasound induces sterile inflammation. Proc. Natl. Acad. Sci. USA 2017, 114, E75–E84. [Google Scholar] [CrossRef] [Green Version]
  71. Gorick, C.M.; Mathew, A.S.; Garrison, W.J.; Andrew Thim, E.; Fisher, D.G.; Copeland, C.A.; Song, J.; Klibanov, A.L.; Wilson Miller, G.; Price, R.J. Sonoselective transfection of cerebral vasculature without blood-brain barrier disruption. Proc. Natl. Acad. Sci. USA 2020, 117, 5644–5654. [Google Scholar] [CrossRef] [Green Version]
  72. Lin, C.Y.; Tsai, C.H.; Feng, L.Y.; Chai, W.Y.; Lin, C.J.; Huang, C.Y.; Wei, K.C.; Yeh, C.K.; Chen, C.M.; Liu, H.L. Focused ultrasound-induced blood brain-barrier opening enhanced vascular permeability for GDNF delivery in Huntington’s disease mouse model. Brain Stimul. 2019, 12, 1143–1150. [Google Scholar] [CrossRef] [PubMed]
  73. Maiuolo, J.; Gliozzi, M.; Musolino, V.; Scicchitano, M.; Carresi, C.; Scarano, F.; Bosco, F.; Nucera, S.; Ruga, S.; Zito, M.C.; et al. The “frail” brain blood barrier in neurodegenerative diseases: Role of early disruption of endothelial cell-to-cell connections. Int. J. Mol. Sci. 2018, 19, 2693. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  74. Burgess, A.; Nhan, T.; Moffatt, C.; Klibanov, A.L.; Hynynen, K. Analysis of focused ultrasound-induced blood-brain barrier permeability in a mouse model of Alzheimer’s disease using two-photon microscopy. J. Control. Release 2014, 192, 243–248. [Google Scholar] [CrossRef] [PubMed]
  75. Karakatsani, M.E.; Wang, S.; Samiotaki, G.; Kugelman, T.; Olumolade, O.O.; Acosta, C.; Sun, T.; Han, Y.; Kamimura, H.A.S.; Jackson-Lewis, V.; et al. Amelioration of the nigrostriatal pathway facilitated by ultrasound-mediated neurotrophic delivery in early Parkinson’s disease. J. Control. Release 2019, 303, 289–301. [Google Scholar] [CrossRef]
  76. 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. [Google Scholar] [CrossRef] [Green Version]
  77. Jordão, J.F.; Ayala-Grosso, C.A.; Markham, K.; Huang, Y.; Chopra, R.; McLaurin, J.A.; Hynynen, K.; Aubert, I. 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. [Google Scholar] [CrossRef] [Green Version]
  78. Burgess, A.; Hynynen, K. Drug delivery across the blood-brain barrier using focused ultrasound. Expert Opin. Drug Deliv. 2014, 11, 711–721. [Google Scholar] [CrossRef] [Green Version]
  79. Jordão, J.F.; Thévenot, E.; Markham-Coultes, K.; Scarcelli, T.; Weng, Y.Q.; Xhima, K.; O’Reilly, M.; Huang, Y.; McLaurin, J.A.; Hynynen, K.; et al. Amyloid-β plaque reduction, endogenous antibody delivery and glial activation by brain-targeted, transcranial focused ultrasound. Exp. Neurol. 2013, 248, 16–29. [Google Scholar] [CrossRef] [Green Version]
  80. Leinenga, G.; Götz, J. Scanning ultrasound removes amyloid-b and restores memory in an Alzheimer’s disease mouse model. Sci. Transl. Med. 2015, 7. [Google Scholar] [CrossRef] [Green Version]
  81. Hsu, P.H.; Lin, Y.T.; Chung, Y.H.; Lin, K.J.; Yang, L.Y.; Yen, T.C.; Liu, H.L. Focused ultrasound-induced blood-brain barrier opening enhances GSK-3 inhibitor delivery for amyloid-beta plaque reduction. Sci. Rep. 2018, 8. [Google Scholar] [CrossRef]
  82. Toccaceli, G.; Delfini, R.; Colonnese, C.; Raco, A.; Peschillo, S. Emerging strategies and future perspective in neuro-oncology using transcranial focused ultrasonography technology. World Neurosurg. 2018, 117, 84–91. [Google Scholar] [CrossRef] [PubMed]
  83. Schneider, C.S.; Woodworth, G.F.; Vujaskovic, Z.; Mishra, M. V Radiosensitization of high-grade gliomas through induced hyperthermia: Review of clinical experience and the potential role of MR-guided focused ultrasound. Radiother. Oncol. 2020, 142, 43–51. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Lamsam, L.; Johnson, E.; Connolly, I.D.; Wintermark, M.; Gephart, M.H. A review of potential applications of MR-guided focused ultrasound for targeting brain tumor therapy. Neurosurg. Focus 2018, 44, E10. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. 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]
  86. Park, J.W.; Jung, S.; Jung, T.Y.; Lee, M.C. Focused ultrasound surgery for the treatment of recurrent anaplastic astrocytoma: A preliminary report. AIP Conf. Proc. 2006, 829, 238–240. [Google Scholar]
  87. McDannold, N.; Clement, G.T.; 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, 323–332. [Google Scholar] [CrossRef] [Green Version]
  88. Coluccia, D.; Fandino, J.; Schwyzer, L.; O’Gorman, R.; Remonda, L.; Anon, J.; Martin, E.; Werner, B. First noninvasive thermal ablation of a brain tumor with MR-guided focused ultrasound. J. Ther. Ultrasound 2014, 2. [Google Scholar] [CrossRef] [Green Version]
  89. Prada, F.; Kalani, M.Y.S.; Yagmurlu, K.; Norat, P.; Del Bene, M.; DiMeco, F.; Kassell, N.F. Applications of focused ultrasound in cerebrovascular diseases and brain tumors. Neurotherapeutics 2019, 16, 67–87. [Google Scholar] [CrossRef] [Green Version]
  90. A Study to Evaluate the Safety and Feasibility of Transcranial MRI-Guided Focused Ultrasound Surgery in the Treatment of Brain Tumors. Available online: https://clinicaltrials.gov/ct2/show/NCT00147056 (accessed on 9 January 2021).
  91. Magnetic Resonance (MR) Guided Focused Ultrasound in the Treatment of Brain Tumors (FUS-Tumor). Available online: https://clinicaltrials.gov/ct2/show/NCT01698437 (accessed on 9 January 2021).
Table 1. Inclusion and exclusion criteria.
Table 1. Inclusion and exclusion criteria.
Inclusion CriteriaExclusion Criteria
Publications of the last four years and their most meaningful referencesPublications dated over four years
New perspectives in pathology treatmentStudies focusing on Essential Tremor
Availability of full textUnavailability of full text
English publicationsNon-English publications
Comparison between tcMRgFUS and different treatments in the same pathologyStudies without any comparison to tcMRgFUS
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Giammalva, G.R.; Gagliardo, C.; Marrone, S.; Paolini, F.; Gerardi, R.M.; Umana, G.E.; Yağmurlu, K.; Chaurasia, B.; Scalia, G.; Midiri, F.; et al. Focused Ultrasound in Neuroscience. State of the Art and Future Perspectives. Brain Sci. 2021, 11, 84. https://doi.org/10.3390/brainsci11010084

AMA Style

Giammalva GR, Gagliardo C, Marrone S, Paolini F, Gerardi RM, Umana GE, Yağmurlu K, Chaurasia B, Scalia G, Midiri F, et al. Focused Ultrasound in Neuroscience. State of the Art and Future Perspectives. Brain Sciences. 2021; 11(1):84. https://doi.org/10.3390/brainsci11010084

Chicago/Turabian Style

Giammalva, Giuseppe Roberto, Cesare Gagliardo, Salvatore Marrone, Federica Paolini, Rosa Maria Gerardi, Giuseppe Emmanuele Umana, Kaan Yağmurlu, Bipin Chaurasia, Gianluca Scalia, Federico Midiri, and et al. 2021. "Focused Ultrasound in Neuroscience. State of the Art and Future Perspectives" Brain Sciences 11, no. 1: 84. https://doi.org/10.3390/brainsci11010084

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