Abstract
Parkinson’s disease has many symptomatic treatments but there is no neuroprotective therapy currently available. The evolution of this disease is inexorably progressive and halting or stopping the neurodegenerative process is a major unmet need. Parkinson’s disease motor features at onset are typically limited to one body segment, i.e. focal signs, and the nigrostriatal degeneration is highly asymmetrical and mainly present in the caudal putamen. Thus, clinically and neurobiologically the process is fairly limited early in its evolution. Tentatively, this would allow the possibility of intervening to halt neurodegeneration at the most vulnerable site. The recent use of new technologies such as focused ultrasound provides interesting prospects. In particular, the possibility of transiently opening the blood brain barrier to facilitate penetrance of putative neuroprotective agents is a highly attractive approach that could be readily applied to Parkinson’s disease. However, because there are currently effective treatments available (i.e. dopaminergic pharmacological therapy), more experimental evidence is needed to construct a feasible and practical therapeutic approach to be tested early in the evolution of Parkinson’s disease patients. In this review, we provide the current evidence for the application of blood brain barrier opening in experimental models of Parkinson’s disease and discuss its potential clinical applicability.
Keywords: Parkinson’s disease, Blood-Brain Barrier, Focused Ultrasound, Alpha-Synuclein, Therapy
Introduction
Early treatment for Parkinson’s disease (PD) remains a challenge. PD has several symptomatic treatments but there is no neuroprotective therapy currently available. We still do not know its etiology, and we cannot stop the progression of the disease1. At onset, there are very few regions which have obvious neuronal loss, and motor symptoms are typically highly asymmetrical and limited to one limb2. In fact, it is clear that PD motor features are closely associated with the degeneration of the dopaminergic neurons in the substantia nigra pars compacta (SNc), especially those in the ventrolateral part, and with the loss of dopaminergic terminals predominantly in the posterior putamen2. The process thus seems to be fairly limited clinically and anatomically in the evolution of the disease.
The other pathological hallmark of PD is the presence of proteinaceous inclusions that are rich in fibrillary forms of alpha-synuclein (α-syn), commonly named Lewy pathology (LP)3. LP is observed more widely in the brain but to variable degrees. However, it is commonly accepted that LP generally occurs in selected nuclei including the olfactory bulb, dorsal motor nucleus of the vagus nerve, locus coeruleus, SNc, raphe nuclei, amygdala or cortex, leaving many other brain regions unaffected2. Therefore, developing therapeutic strategies aiming to delay the clinical onset of PD in the most vulnerable regions of the brain looks reasonable.
Unfortunately, a number of difficulties, particularly the inaccessibility of the degenerating brain, have made this impossible so far. In this regard, new technologies such as focused ultrasound (FUS) provide an interesting opportunity. In particular, the possibility of transiently opening the blood brain barrier (BBB) to facilitate the penetrance of putative neuroprotective agents is a highly attractive approach, which could be applied readily to PD and other neurodegenerative diseases4. Gene delivery is a clear example of a promising therapeutic modality that would benefit5,6. However, because there are currently effective treatments for PD patients available (i.e. dopaminergic pharmacological therapy) more experimental evidence is needed in order to construct a feasible and practical therapeutic approach to be tested in the early symptomatic phase of PD patients. In this Review, we provide current evidence of the application of BBB opening for the delivery of therapeutic agents to the brain in experimental models of PD and discuss its potential clinical applicability in the future.
The blood-brain barrier
The BBB is a term used to describe the unique properties of the microvasculature of the central nervous system (CNS). Blood vessels are made up of two main cell types: endothelial cells that form the walls of the blood vessels, and mural cells that sit on the ablumenal surface of the endothelial cell layer. Interactions with mural cells, immune cells, astrocytes, pericites and neural cells in the capillary basement membrane maintain intact the properties of the BBB7.
Importantly, the BBB prevents the brain from the entry of neurotoxic plasma components, blood cells, and pathogens in normal conditions. The BBB also serves to regulate the transport of different molecules though the CNS, maintaining control of the environment and homeostasis of the brain required for correct neuronal functioning. However, at the same time the BBB represents an obstacle for drug delivery to the CNS, and thus major efforts have been made to generate methods to bypass the BBB for delivery of therapeutic compounds and drugs8.
The blood-brain barrier opening with focused ultrasound
The treatment of CNS diseases involves the synergistic action of the BBB to transport therapeutic agents. The BBB hinders the transcellular diffusion path, which is confined only to lipid soluble compounds smaller than 400 Da with fewer than nine hydrogen bonds crossing via lipid-mediated transport. To overcome this obstacle, current treatment strategies involve transcranial injection or infusion, and employment of medicinal chemistry to chemically alter the nature of the compound so it can cross the BBB through carrier-mediated, receptor-mediated or active efflux transport9. However, all of these methods are either invasive, non-targeted and/or involve alteration of the drug composition. Direct injection, convection enhanced delivery and osmotic BBB disruption are some examples of targeted but invasive techniques while biological and chemical approaches and intranasal drug delivery are non-invasive but non-targeted methods10,11.
FUS technology has emerged as a promising alternative in delivering pharmacological agents into the brain by overcoming the impermeable BBB and concurrently the adverse events associated with excessive drug administration. FUS coupled with the administration of microbubbles has been proposed as the only noninvasive technique to transiently, locally and reversibly disrupt the BBB, allowing a temporal and spatial window for molecules to cross to the brain parenchyma12,13 (Figure 1).
Figure 1:

Mechanism of BBB opening disruption. (a) The microbubbles (white) and the therapeutic agent (purple) follow the circulation after intravenous injection. (b) The microbubble oscillate when reaching the focus of the beam that exerts mechanical forces to the endothelial cells and loosens the tight junctions. (c) The therapeutic agent diffuses through the disrupted barrier into the brain parenchyma.
The microbubbles described herein are perfluorocarbon-filled, lipid-coated microspheres on the order of a few microns in diameter, characterized by slow solubility and dissolution kinetics, attributed to their shell composition14. Their stability in the blood vessels has been improved by increasing the hydrocarbon chain-length of the coat-constituent lipids, improving their physicochemical properties and their overall efficiency14–17.
Ultrasonic energy focused at the geometrical center can be tightly deposited deeply within the brain tissue while minimizing skull energy absorption18. During focused ultrasound, application of the transmitted acoustic pulse generates a radiation force that drives the expansion and contraction (or collapse) of the microbubbles14, characterized as acoustic cavitation. Controlled oscillation of the microbubbles results in increased vascular permeability, while rapture of the bubbles19 has been associated with increased risk for damaging the surrounding microenvironment20,21 (Figure 1).
Hence, the size-dependent resonance behavior of the microbubbles and their response to the acoustic field varies according primarily to the center frequency, the applied pressure and the pulse length14. The relatively low frequencies combined with various pressures have been shown to successfully induce BBB openings of different sizes depending on the weight of the deliverable agent22. The effect of the ultrasound parameters on the vascular permeability is linearly dependent on the microbubble concentration, whereas the functional outcomes are more predictable for narrower size distributions of microbubbles23.
The closing timeline and the reversibility of the BBB opening have been extensively investigated to assess the safety profile of the intervention. The time necessary for the barrier to be fully restored has been found proportionally related to the opening volume assuming the induction of a single opening24. The decoupling of this dependence has been achieved by substituting large openings with small multifoci openings decreasing the necessary time for the barrier to be restored25,26. Longitudinal studies on rodents and primates have shown that repeated ultrasound-induced BBB opening in the absence of vascular damage is a transient and reversible application27,28. Restoration of the barrier was not only macroscopically evaluated by MRI but also neurologically by visual, cognitive, motivational and motor function behavioral testing27,28.
Although the interaction of systemically-administered microbubbles with the capillary walls has been proposed to drive the disruption of the BBB with FUS, the mechanism is not entirely clear as not fully understood are the downstream bioeffects. Disassembling of the tight junctional (TJs) molecular structure has been placed at the beginning of the induced biological cascade, explaining the paracellular passage of molecules that has been reported29,30. Moreover, transcriptomic analysis in the acute stages following sonication revealed a transient upregulation in proinflammatory cytokines and chemokines including CCL2, CCL3, and Tnf that have been found to promote migration, proliferation, differentiation and survival of neural progenitor cells favoring neurogenesis31. The prominent presence of BrdU-positive cells in animals that survived for a week after the last of six sonications has been linked to enhanced neurogenesis attributed to the expression of trophic factors such as brain-derived neurotrophic factor in the targeted brain32. Concurrent with the overexpression of inflammatory markers that mostly resolved within 24 hours, was the increase in angiogenetic-related genes and astrocytic activation31.
Despite the positive impact of the technique, reports on elevated microtubule-associated protein tau, phosphorylated at the Thr231 epitope32, and activated Nf-kB pathway33 following repeated sonications, alarmed the ultrasound field and surfaced important unmet needs. Although the uncontrolled phosphorylation of tau protein has been linked to Alzheimer’s disease, phosphorylation has to occur at specific epitopes and proven to lead to pathological outcomes34. Tau phosphorylation at the Thr231 epitope is associated with both physiological and pathological processes34, and further experimentation is required to assess association with Alzheimer’s disease as the upregulation alone does not suffice. Regarding the initiation of the Nf-kB pathway, several reports argue on whether it is a byproduct of the sonication regime33 or it is completely dissociated from the intervention31. Contradictory findings and ambiguous interpretations signify the need to fully characterize the biological changes specific to the selected sonication protocol.
So far, FUS has been studied extensively in a multitude of experiments involving the safe disruption of the BBB of various animal species (including rabbits35,36 mice37, rats38 and primates39,40) and in different PD animal models (Table 1). The integrity of the BBB is restored within hours and it remains intact24 depending on the ultrasound parameters, regardless of the pathological state of the brain at least for early stages41. FUS-mediated BBB opening has been proven indifferent in terms of energy requirements to achieve permeability and closing timeline between transgenic and wild type mice41.
Table1:
Summary of studies undertaking BBB Opening in animal models of Parkinsońs disease.
| Animal Model | Deliverable | Delivery vehicle | Delivery method | FUS applications | Staining | Behavioral | References |
|---|---|---|---|---|---|---|---|
| MPTP mice | GDNF | AAV | IV | 1 | Yes | Yes | Karakatsani et al. 2019 |
| MPTP mice | GDNF | gene-liposome-microbubbles | IV | 1 | Yes | Yes | Lin et al. 2016 |
| 6-OHDA rats | GDNF | brain-penetrating nanoparticles | IV | 1 | Yes | Yes | Mead et al. 2017 |
| 6-OHDA rats | GDNF | Cationic microbubbles | IV | 1 | Yes | Yes | Fan et al. 2017 |
| Wild type mice | NTN | Direct | IV | 1 | Yes | No | Samiotaki et al. 2015 |
| MPTP mice | NTN | Direct | IV | 3 | Yes | No | Karakatsani et al. 2019 |
| Wild type mice | BDNF | Direct | IN | 1 | Yes | No | Chen et al. 2016 |
| MPTP mice | BDNF | Direct | IN | 3 | Yes | Yes | Ji et al. 2018 |
| A53T α-syn mice | anti α-syn antibody | Direct | IV | 3 | Yes | No | Zhang et al. 2018 |
| WT α-syn mice | α-syn shRNA | AAV | IV | 1 | Yes | No | Xhima et al. 2018 |
This intervention has shown efficacy in delivering various compounds of different molecular weights into the brain parenchyma including contrast agents42, sugars43, antibodies44, chemotherapeutics45 and neurotrophic factors6,25,43,46–49. Aside from direct delivery of the pharmacological agent, FUS-facilitated viral and non-viral vector-based gene delivery has been proven feasible to promote long term expression of endogenous proteins6,46,49.
Focused ultrasound-induced BBB opening is an innovative and non-invasive approach to achieve drug delivery within the CNS by providing significant advantages compared to other approached in terms of targeting, non-invasiveness, and reversibility. The recent advances in technical optimization and preclinical validation support the immense potential of the intervention as the drug-delivery technique of choice in neurodegeneration models18.
Focused ultrasound-induced BBB opening in experimental models of Parkinson’s disease
When considering opening the BBB in experimental models of PD, we have to consider i. the animal model of choice and ii. what we are going to deliver. One obvious option is to target the dopaminergic system, preferentially at the level of the SNc or striatum. So far, the toxin-based models (MPTP and 6-OHDA) are the best choice for those studies designed to test neuroprotection, since there is clear dopaminergic neuronal loss in the SNc and dopamine loss in the striatum50. Looking ahead, the other therapeutic option to consider in PD patients is to modulate α-syn aggregation in the brain. Unfortunately, these models do not recapitulate the α-syn aggregation observed clinically in PD patients50. In this case we have the following different options: i. intra-parenchymal inoculations of exogenous α-syn (e.g., synthetic α-syn fibrils), ii. transgenic mice, and iii. animals in which α-syn overexpression is induced by viral vector injections51.
Targeting the dopaminergic system: trophic factors
Despite highly positive evidence from pre-clinical studies52, clinical trials of PD testing the delivery of different neurotrophic factors have been largely ineffective for several reasons including dosage, poor distribution in the brain, poor retrograde transport and late time points of delivery53. FUS can partially address some of these issues primarily by improving the distribution of the deliverable molecule in the targeted location43,54 (Figure 2) and thus adjusting the dosage to balance sufficient deposition and saturation, making trophic factors a possible alternative for PD treatment55.
Figure 2:

Figure adjusted from Samiotaki et al. (# 39). Horizontal section at the striatum, outlined by the red dotted lines, immunostained against Neurturin (brown color) and counterstained with hematoxylin (purple color): (a) The sonicated striatum developed higher intensity of the anti-Neurturin antibody suggesting higher concentration of the trophic factor compared to the contralateral side. (b–c) Higher magnification at sonicated and contralateral side, respectively, (d–e) Extraction of the brown color only corresponding to Neurturin for b and c, respectively.
Different studies have tried to restore the integrity of the dopaminergic system by combination of FUS with trophic factors in combination with different particles or vectors. Normally, this approach is thought to be either neuroprotective, delaying the death of the dopaminergic neurons in the SNc, or restorative, by restoring or improving the capacity of the brain to produce dopamine again. Glial-derived-neurotrophic-factor (GDNF) is usually the first choice, but neurturin (NTN) and brain-derived-neurotrophic-factor (BDNF) have also been tested.
Following the initial feasibility study, Karakatsani and Wang et al.47 delivered adeno-associated virus (AAV) – GDNF (AAV1-CAG-eGFP-GDNF) in the left striatum and midbrain of subacute MPTP treated mice. Dopaminergic neuronal cell bodies demonstrated a 58.4% upregulation with more than a twofold increase in their projections’ density after the administration of AAV-GDNF and its diffusion through the FUS-induced BBB opening, as evidenced by tyrosine hydroxylase (TH) immunostaining. Similar upregulation was observed in the striatum where the evidenced upregulation of the terminal density reached 30%. Dopamine-related behavioral changes demonstrated by amphetamine-elicited unilateral rotations, confirmed the physiological advances of the FUS-facilitated viral delivery (Figure 3).
Figure 3:

Figure adjusted from Karakatsani et al. (# 61). The nigrostriatal pathway includes the Substantia Nigra (SN), where the dopaminergic neuronal cells (SNc) and dendrites (SNr) lie and the Striatum, where the neuronal terminals can be found. The nigrostriatal pathway is downregulated upon MPTP induction in a “dying back” regime, from the terminals to the cell bodies. Application of focused ultrasound coupled with the administration of microbubbles results in increased BBB permeability allowing the diffusion of neurotrophic factors. Single administration of Neurturin resulted in a 19% increase in the TH expression at the striatal site while 4% and 20% in the SPc and SNr sites respectively. Triple administration of NTN with the equivalent number of FUS applications resulted in an increased effect compared to the single treatment on the order of 50%, 17% and 22% for the striatum, SNc and SNr respectively. Diffusion of AAV-GDNF resulted in a similar to the triple treatment effect on the order of 32%, 14% and 76% for the corresponding structures.
Similarly, Lin et al.48 delivered gene-carrying liposomes in combination with FUS to improve the GDNF gene-delivery efficiency in the MPTP-mice model48. The FUS-induced BBB opening was verified by contrast-enhanced MRI, and gene expression was verified by in vivo imaging. The focal delivery of gene-liposome complexes successfully served as gene carrier and BBB-opening catalyst. Immunoblotting and histological staining confirmed the expression of reporter genes in neuronal cells leading to reduced expression and progression of motor abnormalities. Post-mortem analysis confirmed preserved dopaminergic metabolism associated with the improvement of motor abnormalities.
In another study, Mead and colleagues49 used FUS in combination with brain-penetrating nanoparticles to induce widespread and focal GDNF transgene expression in the brain following systemic administration in 6-OHDA-treated rats. After only a single treatment, this strategy led to therapeutically relevant levels of GDNF protein content in the striatum that lasted at least for 10 weeks. This strategy restored both dopamine levels at the striatum and dopaminergic TH neuron density in the SNc and reversed behavioral abnormalities with no evidence of local or systemic toxicity.
Fan et al.5 loaded cationic microbubbles with GDNF and FUS was used to allow transient gene permeation and induce local GDNF expression. In this study, FUS made is possible to achieve higher titer GDNF genes than with intracerebral injections. The combination of GDNF-loaded microbubbles and FUS resulted in restored behavioral motor deficits and ameliorated neuronal death in the SNc and dopamine loss in the striatum of 6-OHDA-trated rats. In another similar approach the delivery of GDNF alone or in combination with Nurr1 with PEGylated liposomes-coupled microbubbles using FUS alleviated the behavioral deficits and neuron loss in the 6-OHDA rats56,57. The multistep process involved in gene transfection requires successful delivery, translation, and release followed by receptor ligation. It is therefore subjected to limited efficacy, shifting the scientific interest towards direct protein delivery.
Samiotaki et al.25 demonstrated enhanced non-invasive local FUS delivery of NTN in wild-type mice at the level of striatum and midbrain, confirmed by immunostaining25 (Figure 2). Briefly, the area of NTN bioavailability was 5.07 ± 0.64 mm2 in the striatum and 2.25 ± 1.14 mm2 in the midbrain, with NTN being present across the entire ultrasound-treated brain region in contrast to the relatively smaller region reached by direct injection (Figure 2). Furthermore, NTN bioactivity was evaluated by tracing the local activation of the downstream signaling pathway through the detection of increased phosphorylation of the Ret receptor, cytoplasmic kinase Erk 1 and 2 and CREB transcription factor in structures associated with their abundance. This finding was particularly significant since the nigrostriatal pathway, which connects the ventral midbrain region with the striatum, is the most severely affected dopaminergic pathway in PD43.
To determine the potential value of using FUS to improve brain penetrance of bioactive molecules of the compromised neurons in the neurodegenerated brain, Karakatsani et al.47 compared TH-based parameters between hemispheres of MPTP-injected mice that received single or triple systemic NTN injections coupled with the equivalent number of FUS applications (Figure 3). Upregulation of TH expression in the midbrain was initiated by both single and triple administrations, hence the 20–22% denser dendritic network, with only multiple exposures achieving restoration of neurotransmission, evidenced by a 50% increase in the immunoreactivity of the innervating dopaminergic neurons at the striatal level. Significantly increased dopamine levels in the treated ventral midbrain were confirmed with HPLC analysis, strengthening the relevance of the two techniques in achieving functional outcomes47.
Another promising, non-invasive drug delivery approach that has been developed and evaluated in animal models and clinical trials is intranasal delivery (IN), which circumvents the impermeable BBB by employing the olfactory epithelium to reach the central nervous system11. Low delivery specificity, the major drawback of this method, was resolved by coupling it with FUS. Chen et al.43 administered BDNF to wild type mice through the nostrils, before exposing their left striatum to ultrasound43. Immunohistochemical findings revealed the increased bioavailability of BDNF in the exposed striatum compared to the contralateral side, showing its immense potential as a surrogate for intravenous delivery especially when systemic exposure to pharmacological agents needs to remain contained.
To explore the translational potential of intranasal BDNF delivery, Ji et al.58 has been investigating the functional outcomes of this delivery approach in subacute MPTP mice employing multiple administrations and exposures, following the rationale of extended dopaminergic upregulation achieved by multiple treatments shown by Karakatsani et al.47. Expectedly, the fiber density in the ventral midbrain was increased by 13% as evidenced by TH-immunopositivity, whereas the projections of the dopaminergic neurons in the striatum were upregulated by 20% after the experimental procedure. Animals tested in the circular open-field task after amphetamine injection showed a significant preference towards ipsilateral-rotations, suggesting an amelioration of the pathology in the treated hemisphere.
Similar approaches with molecules other than trophic factors have been adopted with similar results. For example, delivery of nuclear factor E2-related factor 2 (Nrf2, NFE2L2) with FUS and microbubbles in the SNc of 6-OHDA rats results in reduced reactive oxygen species levels, thereby protecting dopaminergic neurons59.
The therapeutic alternatives explored so far have shown a gradual increase in beneficial outcome with direct protein delivery proven efficient in upregulating neuronal function. However, translation to clinical practice entails multiple applications that would significantly improve neuronal integrity along the entire nigrostriatal pathway. Gene therapy comes as an alternative to multiple applications allowing for constant release of the neurotrophic factor with only one ultrasound session.
Targeting α-synuclein aggregation
Several studies have shown the effectiveness of the use of FUS in animal models of AD targeting both β-amyloid and tau4,44,60–62. Indeed, in some of these cases there was a clear beneficial influence of BBB opening alone (in the absence of any therapeutic agent) on the clearance of β-amyloid plaques4,44,60 or tau61. On the other hand, FUS enhanced antibody delivery, increasing clearance of proteins61. Subsequently to these experiments, BBB opening with FUS was started in five AD patients with early to moderate AD in a phase I safety trial63. In all patients, the BBB within the target volume was safely, reversibly, and repeatedly opened. However, despite the encouraging preliminary results in AD, to date FUS-facilitated therapy against the abnormal α-syn accumulation remains uncharted. Only a few studies have targeted this so far, all of them with positive results.
In a pioneering study in cell culture, Karmacharya et al.64 show that FUS decreases α-syn aggregation by attenuation of mitochondrial reactive oxygen species in MPP(+)-treated PC12 cells. Regarding in vivo studies, Zhang et al.65 designed a study on A53T transgenic mice that overexpress human α-syn with prominent inclusions by the pre-symptomatic age of 9 months. The animals that received three exposures to FUS coupled with the administration of anti α-syn antibody experienced a 1.5 fold decrease in the α-syn load in the treated hemisphere compared to the untreated brains one month after the delivery completion. These preliminary findings suggest the feasibility of such an approach in clearing accumulated proteins from the degenerated brain.
In a more recent study, transgenic mice expressing WT human α-syn were subjected to MRI-guided FUS focally in different brain regions susceptible to α-syn aggregation (hippocampus, SNc, olfactory bulb, and dorsal motor nucleus) in tandem with intravenous microbubbles and an AAV9 bearing a shRNA targeting α-syn66. One month after treatment, α-syn immunoreactivity was decreased, whereas other neuronal markers such as synaptophysin or TH were unchanged, and cell death and glial activation remained at baseline levels. These results demonstrate that FUS can effectively deliver viral vectors targeting α-syn to multiple brain areas. This approach might be useful to alter the progression of LP in PD patients particularly in those diagnosed early, thus improving the evolution of the disease.
Limitations and Prospect
The inconsistency in the pathological outcomes stemming from different sonication protocols surfaces the necessity to establish standardized methods to evaluate the sonication regime. Aside the ultrasonic parametric space, microbubble dosing and distribution have to be fully characterized and brain-structure susceptibility to ultrasound is crucial in understanding the biological effects that occur during drug delivery and fairly compare findings67. Furthermore, a larger number of molecules should be tested given that so far, only GDNF has been widely tested in several independent laboratories. Further studies are needed to assess the effects of antibody administration and other drug types such as anti-inflammatory compounds or drugs regulating genes/enzymes related to PD (i.e. glucocerebrosidase), alone or in combination with FUS, to the brain. In this regard it is important to remember that none of the available PD animal models recapitulates all the pathologic abnormalities observed in PD clinical cases (i.e. absence of LP in toxin-based models or absence of extensive neurodegeneration in α-syn models)68. For example, up to now, experiments aimed to reduce α-syn expression with FUS have not addressed if there is neuroprotection. There are a lot of promising candidates, some of which are “old friends” such as GDNF, but others that are new, like GBA modulators, iron-chelators, antibodies against α-syn or anti-inflammatory drugs69, which can benefit a lot from the FUS-facilitated delivery. More gene therapy experiments should be attempted. For example, recent clinical trials involved invasive putaminal injections53,70. AAV-AADC delivery proved to improve motor function70 whereas AAV-GDNF did not meet its primary endpoint and did not provide clinical benefits53. These approaches could be more effective (and safer) in PD patients if the viral vectors are delivered focally with FUS.
Finally, moving towards a possible clinical application in PD, several important questions remain unresolved. These include defining regions of the brain that should be targeted, how many times this procedure can be performed safely in patients and the duration of action of the delivered agent(s) to have a significant effect against the neurodegenerative process. Another fundamental underlying issue is the interaction between the described increased permeability of the BBB in the SNc and striatum in PD patients71–74 and the impact of BBB opening via ultrasound. Indeed, one could question the need for opening the BBB if it is already deranged. However, we would argue that such abnormalities would not guarantee local and sufficient delivery of putative neuroprotective/restorative agents. In any case, this remains a topic in need for further investigation.
Conclusion
Currently the use of FUS for ablative purposes shows some promise for the treatment of PD. This includes targeting either the ventral intermediate thalamus, the subthalamic nucleus or the internal pallidum75–78). Phase I trials exploring both of these indications are currently underway (NCT03608553). However, the use of FUS to disrupt the BBB in a focal and temporary way and facilitate the entry of different compounds such as GDNF, anti-inflammatory drugs or antibodies seems to be an extremely promising therapeutic option for the treatment of PD not only as a symptomatic treatment but also to impact on the mechanisms underlying neurodegeneration. This would also hold for other neurodegenerative disease including AD or ALS63,79,80. Here we have reviewed several studies that display encouraging results regarding the possible application of this technology for PD. However, more experimental evidence is needed before clinical applications of these approaches can be developed.
In sum, FUS-facilitated drug delivery is a highly promising therapeutic approach, but probably still in need of further experimental work before reaching clinical trials. Thus, different protocols for FUS exposure must be explored in valid animal models in the near future.
Funding sources:
The author M.E.K. has been supported by the National Institutes of Health (AG038961, EB009041) and the Focused Ultrasound Foundation. The author J.B. is currently funded by grant S2017/BMD-3700 (NEUROMETAB-CM) from Comunidad de Madrid co-financed with the Structural Funds of the European Union, Fundación BBVA and Fundación Tatiana Pérez de Guzmán el Bueno.
References
- 1.Del Rey NL-G, Quiroga-Varela A, Garbayo E, et al. Advances in Parkinson’s Disease: 200 Years Later. Front. Neuroanat 2018;12:113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Surmeier DJ, Obeso JA, Halliday GM. Selective neuronal vulnerability in Parkinson disease. Nat. Rev. Neurosci 2017;18(2):101–113. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Goedert M, Spillantini MGG, Tredici K Del, et al. 100 years of Lewy pathology. Nat. Rev. Neurol 2013;9(1):13–24. [DOI] [PubMed] [Google Scholar]
- 4.Burgess A, Shah K, Hough O, Hynynen K. Focused ultrasound-mediated drug delivery through the blood–brain barrier. Expert Rev. Neurother 2015;15(5):477–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Fan C-H, Lin C-Y, Liu H-L, Yeh C-K. Ultrasound targeted CNS gene delivery for Parkinson’s disease treatment. J. Control. Release 2017;261:246–262. [DOI] [PubMed] [Google Scholar]
- 6.Wang S, Olumolade OO, Sun T, et al. Noninvasive, neuron-specific gene therapy can be facilitated by focused ultrasound and recombinant adeno-associated virus.. Gene Ther. 2015;22(1):104–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Daneman R, Prat A. The Blood–Brain Barrier. Cold Spring Harb. Perspect. Biol 2015;7(1):a020412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8.He Q, Liu J, Liang J, et al. Towards Improvements for Penetrating the Blood–Brain Barrier—Recent Progress from a Material and Pharmaceutical Perspective. Cells 2018;7(4):24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Pardridge WM. BLOOD-BRAIN BARRIER DRUG TARGETING: THE FUTURE OF BRAIN DRUG DEVELOPMENT. Mol. Interv 2003;3(2):90–105. [DOI] [PubMed] [Google Scholar]
- 10.Spencer BJ, Verma IM. Targeted delivery of proteins across the blood-brain barrier. Proc. Natl. Acad. Sci. U. S. A 2007;104(18):7594–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Stockwell J, Abdi N, Lu X, et al. Novel Central Nervous System Drug Delivery Systems. Chem. Biol. Drug Des 2014;83(5):507–520. [DOI] [PubMed] [Google Scholar]
- 12.Hynynen K, McDannold N, Vykhodtseva N, Jolesz FA. Noninvasive MR Imaging–guided Focal Opening of the Blood-Brain Barrier in Rabbits. Radiology 2001;220(3):640–646. [DOI] [PubMed] [Google Scholar]
- 13.Choi JJ, Pernot M, Brown TR, et al. Spatio-temporal analysis of molecular delivery through the blood–brain barrier using focused ultrasound. Phys. Med. Biol 2007;52(18):5509–5530. [DOI] [PubMed] [Google Scholar]
- 14.Ferrara K, Pollard R, Borden M. Ultrasound Microbubble Contrast Agents: Fundamentals and Application to Gene and Drug Delivery. Annu. Rev. Biomed. Eng 2006;9(1):415–447. [DOI] [PubMed] [Google Scholar]
- 15.Borden MA, Longo ML. Dissolution behavior of lipid monolayer-coated, air-filled microbubbles: Effect of lipid hydrophobic chain length. Langmuir 2002;18(24):9225–9233. [Google Scholar]
- 16.Borden MA, Kruse DE, Caskey CF, et al. Influence of lipid shell physicochemical properties on ultrasound-induced microbubble destruction. IEEE Trans. Ultrason. Ferroelectr. Freq. Control 2005;52(11):1992–2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Wu SY, Chen CC, Tung YS, et al. Effects of the microbubble shell physicochemical properties on ultrasound-mediated drug delivery to the brain. J. Control. Release 2015;212:30–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Chen K-T, Wei K-C, Liu H-L. Theranostic Strategy of Focused Ultrasound Induced Blood-Brain Barrier Opening for CNS Disease Treatment. Front. Pharmacol 2019;10(February) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Caskey CF, Stieger SM, Qin S, et al. Direct observations of ultrasound microbubble contrast agent interaction with the microvessel wall. J. Acoust. Soc. Am 2007;122(2):1191–1200. [DOI] [PubMed] [Google Scholar]
- 20.Qin S, Ferrara KW. Acoustic response of compliable microvessels containing ultrasound contrast agents. Phys. Med. Biol 2006;51(20):5065–5088. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Tung YS, Choi JJ, Baseri B, Konofagou EE. Identifying the inertial cavitation threshold and skull effects in a vessel phantom using focused ultrasound and microbubbles. Ultrasound Med. Biol 2010;36(5):840–852. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Chen H, Konofagou EE. The size of blood-brain barrier opening induced by focused ultrasound is dictated by the acoustic pressure. J. Cereb. Blood Flow Metab 2014;34(7):1197–1204. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Song KH, Harvey BK, Borden MA. State-of-the-art of microbubble-assisted blood-brain barrier disruption. Theranostics 2018;8(16):4393–4408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Samiotaki G, Konofagou EE. Dependence of the Reversibility of Focused- Length In Vivo. 2013;60(11):2257–2265. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Samiotaki G, Acosta C, Wang S, Konofagou EE. Enhanced delivery and bioactivity of the neurturin neurotrophic factor through focused ultrasound-mediated blood--brain barrier opening in vivo. J. Cereb. Blood Flow Metab 2015;35(4):611–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.O’Reilly MA, Hough O, Hynynen K. Blood-Brain Barrier Closure Time After Controlled Ultrasound-Induced Opening Is Independent of Opening Volume. J. Ultrasound Med 2017;36(3):475–483. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Downs ME, Buch A, Sierra C, et al. Correction: Long-term safety of repeated blood-brain barrier opening via focused ultrasound with microbubbles in non-human primates performing a cognitive task. PLoS One 2015;10(6) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Olumolade OO, Wang S, Samiotaki G, Konofagou EE. Longitudinal Motor and Behavioral Assessment of Blood–Brain Barrier Opening with Transcranial Focused Ultrasound. Ultrasound Med. Biol 2016;42(9):2270–2282. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Vykhodtseva N, McDannold N, Hynynen K. Progress and problems in the application of focused ultrasound for blood-brain barrier disruption. Ultrasonics 2008;48(4):279–296. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Han M, Hur Y, Hwang J, Park J. Biological effects of blood-brain barrier disruption using a focused ultrasound. Biomed. Eng. Lett 2017;7(2):115–120. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.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] [PMC free article] [PubMed] [Google Scholar]
- 32.Kovacs ZI, Tu T-W, Sundby M, et al. MRI and histological evaluation of pulsed focused ultrasound and microbubbles treatment effects in the brain. Theranostics 2018;8(17):4837–4855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kovacs ZI, Kim S, Jikaria N, et al. Disrupting the blood-brain barrier by focused ultrasound induces sterile inflammation. Proc. Natl. Acad. Sci. U. S. A 2017;114(1):E75–E84 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Simic G, Babic Leko M, Wray S, et al. Tau Protein Hyperphosphorylation and Aggregation in Alzheimer’s Disease and Other Tauopathies, and Possible Neuroprotective Strategies. Biomolecules 2016;6(1):6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Hynynen K, McDannold N, Sheikov NA, et al. Local and reversible blood-brain barrier disruption by noninvasive focused ultrasound at frequencies suitable for trans-skull sonications. Neuroimage 2005;24(1):12–20. [DOI] [PubMed] [Google Scholar]
- 36.McDannold N, Vykhodtseva N, Raymond S, et al. MRI-guided targeted blood-brain barrier disruption with focused ultrasound: histological findings in rabbits. Ultrasound Med. Biol 2005;31(11):1527–37. [DOI] [PubMed] [Google Scholar]
- 37.Choi JJ, Pernot M, Small SA, Konofagou EE. Noninvasive, transcranial and localized opening of the blood-brain barrier using focused ultrasound in mice. Ultrasound Med. Biol 2007;33(1):95–104. [DOI] [PubMed] [Google Scholar]
- 38.O’Reilly MA, Waspe AC, Chopra R, Hynynen K. MRI-guided Disruption of the Blood-brain Barrier using Transcranial Focused Ultrasound in a Rat Model. J. Vis. Exp 2012; (61) [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Marquet F, Tung Y-S, Teichert T, et al. Noninvasive, transient and selective blood-brain barrier opening in non-human primates in vivo. PLoS One 2011;6(7):e22598. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Downs ME, Buch A, Karakatsani ME, et al. Blood-Brain Barrier Opening in Behaving Non-Human Primates via Focused Ultrasound with Systemically Administered Microbubbles. Sci. Rep 2015;5:15076. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Choi JJ, Wang S, Brown TR, et al. Noninvasive and transient blood-brain barrier opening in the hippocampus of Alzheimer’s double transgenic mice using focused ultrasound. Ultrason. Imaging 2008;30(3):189–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Samiotaki G, Vlachos F, Tung YS, Konofagou EE. A quantitative pressure and microbubble-size dependence study of focused ultrasound-induced blood-brain barrier opening reversibility in vivo using MRI. Magn. Reson. Med 2012;67(3):769–777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chen H, Yang GZX, Getachew H, et al. Focused ultrasound-enhanced intranasal brain delivery of brain-derived neurotrophic factor. Sci. Rep 2016;6(1):28599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Jordão JF, Thévenot E, Markham-Coultes 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Park J, Aryal M, Vykhodtseva N, et al. 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;250:77–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Fan C-H, Ting C-Y, Lin C, et al. Noninvasive, Targeted and Non-Viral Ultrasound-Mediated GDNF-Plasmid Delivery for Treatment of Parkinson’s Disease. Sci. Rep 2016;6(1):19579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Karakatsani ME, Wang S, Samiotaki G, et al. Amelioration of the nigrostriatal pathway facilitated by ultrasound-mediated neurotrophic delivery in early Parkinson’s disease. J. Control. Release 2019; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Lin C-Y, Hsieh H-Y, Chen C-M, et al. Non-invasive, neuron-specific gene therapy by focused ultrasound-induced blood-brain barrier opening in Parkinson’s disease mouse model. J. Control. Release 2016;235:72–81. [DOI] [PubMed] [Google Scholar]
- 49.Mead BP, Kim N, Miller GW, et al. Novel Focused Ultrasound Gene Therapy Approach Noninvasively Restores Dopaminergic Neuron Function in a Rat Parkinson’s Disease Model. Nano Lett. 2017;17(6):3533–3542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Blesa J, Przedborski S. Parkinson’s Disease: animal models and dopaminergic cell vulnerability. Front. Neuroanat 2014;8:155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Recasens A, Ulusoy A, Kahle PJ, et al. In vivo models of alpha-synuclein transmission and propagation. Cell Tissue Res. 2017; [DOI] [PubMed] [Google Scholar]
- 52.Kordower JH, Burke RE. Disease Modification for Parkinson’s Disease: Axonal Regeneration and Trophic Factors. Mov. Disord 2018;33(5):678–683. [DOI] [PubMed] [Google Scholar]
- 53.Whone A, Luz M, Boca M, et al. Randomized trial of intermittent intraputamenal glial cell line-derived neurotrophic factor in Parkinson’s disease. Brain 2019;142(3):512–525. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Samiotaki G, Karakatsani ME, Buch A, et al. Pharmacokinetic analysis and drug delivery efficiency of the focused ultrasound-induced blood-brain barrier opening in non-human primates. Magn. Reson. Imaging 2017;37 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Price RJ, Fisher DG, Suk JS, et al. Parkinson’s disease gene therapy: Will focused ultrasound and nanovectors be the next frontier? Mov. Disord 2019;mds.27675. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Yue P, Miao W, Gao L, et al. Ultrasound-Triggered Effects of the Microbubbles Coupled to GDNF Plasmid-Loaded PEGylated Liposomes in a Rat Model of Parkinson’s Disease. Front. Neurosci 2018;12:222. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Yue P, Gao L, Wang X, et al. Ultrasound-triggered effects of the microbubbles coupled to GDNF- and Nurr1-loaded PEGylated liposomes in a rat model of Parkinson’s disease. J. Cell. Biochem 2018;119(6):4581–4591. [DOI] [PubMed] [Google Scholar]
- 58.Ji R, Konofagou E. Determining a cavitation threshold for focused ultrasound enhanced intranasal drug delivery. In: 2017 IEEE International Ultrasonics Symposium (IUS) 2017. p. 1. [Google Scholar]
- 59.Long L, Cai X, Guo R, et al. Treatment of Parkinson’s disease in rats by Nrf2 transfection using MRI-guided focused ultrasound delivery of nanomicrobubbles. Biochem. Biophys. Res. Commun 2017;482(1):75–80. [DOI] [PubMed] [Google Scholar]
- 60.Leinenga G, Gotz J, Götz J. Scanning ultrasound removes amyloid- and restores memory in an Alzheimer’s disease mouse model. Sci. Transl. Med 2015;7(278):278ra33–278ra33. [DOI] [PubMed] [Google Scholar]
- 61.Nisbet RM, Van der Jeugd A, Leinenga G, et al. Combined effects of scanning ultrasound and a tau-specific single chain antibody in a tau transgenic mouse model. Brain 2017;140(5):1220–1230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Alecou T, Giannakou M, Damianou C. Amyloid β Plaque Reduction With Antibodies Crossing the Blood-Brain Barrier, Which Was Opened in 3 Sessions of Focused Ultrasound in a Rabbit Model. J. Ultrasound Med 2017;36(11):2257–2270. [DOI] [PubMed] [Google Scholar]
- 63.Lipsman N, Meng Y, Bethune AJ, et al. Blood-brain barrier opening in Alzheimer’s disease using MR-guided focused ultrasound. Nat. Commun 2018;9(1):2336. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Karmacharya MB, Hada B, Park SR, Choi BH. Low-Intensity Ultrasound Decreases α-Synuclein Aggregation via Attenuation of Mitochondrial Reactive Oxygen Species in MPP(+)-Treated PC12 Cells. Mol. Neurobiol 2017;54(8):6235–6244. [DOI] [PubMed] [Google Scholar]
- 65.Zhang H, Sierra C, Kwon N, et al. Focused-ultrasound Mediated Anti-Alpha-Synuclein Antibody Delivery for the Treatment of Parkinson’s Disease In: IEEE International Ultrasonics Symposium, IUS; 2018 [Google Scholar]
- 66.Xhima K, Nabbouh F, Hynynen K, et al. Noninvasive delivery of an α-synuclein gene silencing vector with magnetic resonance-guided focused ultrasound. Mov. Disord 2018;33(10):1567–1579. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Song K-H, Harvey BK, Borden MA. State-of-the-art of microbubble-assisted blood-brain barrier disruption. Theranostics 2018;8(16):4393–4408. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Trigo-Damas I, Del Rey NL-G, Blesa J. Novel models for Parkinson’s disease and their impact on future drug discovery. Expert Opin. Drug Discov 2018;13(3):229–239. [DOI] [PubMed] [Google Scholar]
- 69.Elkouzi A, Vedam-Mai V, Eisinger RS, Okun MS. Emerging therapies in Parkinson disease — repurposed drugs and new approaches. Nat. Rev. Neurol 2019;15(4):204–223. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Kojima K, Nakajima T, Taga N, et al. Gene therapy improves motor and mental function of aromatic l-amino acid decarboxylase deficiency. Brain 2019;142(2):322–333. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Brochard V, Combadière B, Prigent A, et al. Infiltration of CD4+ lymphocytes into the brain contributes to neurodegeneration in a mouse model of Parkinson disease. J. Clin. Invest 2008; [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.Kortekaas R, Leenders KL, van Oostrom JCH, et al. Blood-brain barrier dysfunction in parkinsonian midbrain in vivo. Ann. Neurol 2005;57(2):176–179. [DOI] [PubMed] [Google Scholar]
- 73.Pisani V, Stefani A, Pierantozzi M, et al. Increased blood-cerebrospinal fluid transfer of albumin in advanced Parkinson’s disease. J. Neuroinflammation 2012;9(1):670. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Gray MT, Woulfe JM. Striatal Blood–Brain Barrier Permeability in Parkinson’S Disease. J. Cereb. Blood Flow Metab 2015;35(5):747–750. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Bond AE, Shah BB, Elias WJ. Assessing Tremor and Adverse Events in Patients With Tremor-Dominant Parkinson Disease Undergoing Focused Ultrasound Thalamotomy—Reply. JAMA Neurol. 2018;75(5):633. [DOI] [PubMed] [Google Scholar]
- 76.Martínez-Fernández R, Rodríguez-Rojas R, del Álamo M, et al. Focused ultrasound subthalamotomy in patients with asymmetric Parkinson’s disease: a pilot study. Lancet. Neurol 2018;17(1):54–63. [DOI] [PubMed] [Google Scholar]
- 77.Jung NY, Park CK, Kim M, et al. The efficacy and limits of magnetic resonance-guided focused ultrasound pallidotomy for Parkinson’s disease: a Phase I clinical trial. J. Neurosurg 2018;1–9. [DOI] [PubMed] [Google Scholar]
- 78.Moosa S, Martinez-Fernandez R, Elias WJ, et al. The role of high-intensity focused ultrasound as a symptomatic treatment for Parkinson’s disease. Mov. Disord 2019;34(9):1243–1251. [DOI] [PubMed] [Google Scholar]
- 79.Leinenga G, Langton C, Nisbet R, Götz J. Ultrasound treatment of neurological diseases - current and emerging applications. Nat. Rev. Neurol 2016;12(3):161–74. [DOI] [PubMed] [Google Scholar]
- 80.Karakatsani ME, Kugelman T, Wang S, et al. Unilateral focused ultrasound-induced blood-brain barrier opening alters spatial profile of hyperphosphorylated tau in an Alzheimer’s mouse model In: IEEE International Ultrasonics Symposium, IUS; 2017 [Google Scholar]
