Focused Ultrasound-Mediated Blood–Brain Barrier Opening for Neurological Disorders: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Chanho Kong.

Several therapeutic agents for neurological disorders are usually not delivered to the brain owing to the presence of the blood–brain barrier (BBB), a special structure present in the central nervous system (CNS). Focused ultrasound (FUS) combined with microbubbles can reversibly and temporarily open the BBB, enabling the application of various therapeutic agents in patients with neurological disorders. 

  • focused ultrasound
  • blood–brain barrier
  • neurological disorders
  • drug delivery

1. Alzheimer’s Disease

The incidence of Alzheimer’s disease (AD), the most representative neurodegenerative brain disease, is steadily increasing as the aging population increases. However, only drugs that can alleviate and delay symptoms are currently being used, and no specific treatment methods or therapeutic agents [19][1] have been developed yet. Over the past decades, several clinical trials have been conducted with various targets, focusing on these two clinical indications: amyloid beta plaques and neurofibrillary tau tangles [20][2]. However, all clinical trials have failed; only Aducanumab, which targets amyloid-β (Aβ) plaque removal, has shown a therapeutic effect, but it is controversial due to side effects [21,22][3][4]. Although the amyloid hypothesis remains controversial, since the accumulation of Aβ is a representative pathological hallmark of AD, numerous therapeutic studies targeting Aβ have been conducted.
The first preclinical study on FUS for AD aimed to deliver anti-Aβ antibodies targeting amyloid plaques into the brain by a BBB opening. Consequently, anti-Aβ antibodies bound to the Aβ plaques and rapidly reduced the plaque pathology [23][5]. Subsequently, research on delivering therapeutic agents through FUS-mediated BBB opening in patients with AD has gained attention [24,25,26,27,28,29,30][6][7][8][9][10][11][12]. Interestingly, studies have reported that amyloid pathology [24,31,32,33,34][6][13][14][15][16] and phosphorylated tau [35,36][17][18] are reduced only by FUS-induced BBB opening without specific drug delivery. Treatment delivery via FUS-mediated BBB opening also affected memory recovery in AD animal models [32,37,38,39,40][14][19][20][21][22]. Research studies on various biological changes by FUS-mediated BBB opening are ongoing. However, for FUS to be a promising non-pharmacological treatment delivery method for AD, further research is needed on why amyloid is reduced and cognitive function is restored. FUS induces the activation of microglia and astrocytes, which may increase phagocytosis of the amyloid plaques [31,32,41][13][14][23]. Recently, a study confirming the therapeutic effect in an AD mouse model (5×FAD) by combining FUS and Aducanumab was reported [42][24]. Aducanumab, a monoclonal antibody targeting fibril forms and beta-amyloid oligomer, has been proven effective since receiving FDA approval in 2021. However, due to side effects, debate continues as to whether or not it should be used.
In conclusion, combined treatment with FUS and Aducanumab reduced amyloid plaque levels, increased hippocampal neurogenesis, and restored cognitive function. Here, FUS activated phagocytic microglia and increased the number of astrocytes associated with amyloid plaques. This suggests that FUS can induce a reduction in amyloid plaques through phagocytosis. In addition, an RNA sequencing analysis showed that the combined treatment with FUS and Aducanumab upregulated neuroinflammation signaling, phagosome formation, reelin signaling, and CREB signaling [42][24]. The immunomodulatory effect of FUS, such as the activation of various innate immune cells, plays a vital role in reducing amyloid plaques [37][19]. Regarding the recovery of cognitive function by FUS, the increase in hippocampal neurogenesis [43,44,45,46][25][26][27][28] or synaptic plasticity [45,47][27][29] may play a role here, but further research is needed on this topic. The researchers summarized the most relevant preclinical studies on FUS-mediated BBB opening in AD (Table 1).
Table 1.
Recent preclinical studies on focused ultrasound-mediated blood–brain barrier opening in Alzheimer’s disease.
(Table 2).
Table 2.
Recent preclinical studies on focused ultrasound-mediated blood–brain barrier opening in Parkinson’s disease.

2. Parkinson’s Disease

Parkinson’s disease (PD) is a neurodegenerative brain disease accompanied by motor dysfunction due to the loss of dopaminergic neurons. PD is neuropathologically characterized by proteinaceous inclusions called Lewy bodies [56][38]. Notably, as many studies have reported that α-synuclein plays a direct role in disease development, PD is classified as α-synucleinopathies [57][39]. Currently, there are no clear treatments to slow or alleviate the progression of neurodegenerative diseases such as PD. Treatment with glial-derived neurotrophic factor (GDNF) is considered appropriate for PD due to its neuroprotective and neurotrophic effects [58,59,60][40][41][42]. The overexpression of neuroprotective genes that induce dopamine regeneration in activated neurons can delay disease progression [61][43]. The potential benefit of GDNF with regard to recovery and the functional improvement of dopaminergic neurons has been confirmed [61,62,63][43][44][45]; however, one study was discontinued due to safety concerns in clinical trials [64][46]. Animal studies of GDNF gene delivery by FUS began in PD and have highlighted the possibility of effective gene therapy [65,66,67][47][48][49].
Since neurturin has been found to have neuroprotective and neuro-regenerative effects on dopaminergic neurons [68][50], the FUS-based delivery of neurturin has been studied to find an alternative to GDNF [69,70][51][52]. Recently, recombinant adeno-associated viral (rAAV) vectors have received much attention as a tool for gene delivery to the brain. The technology of delivering rAAV using FUS-mediated BBB permeability and expressing the delivered gene has already been examined [71][53]. Accordingly, recent studies on PD models using FUS mainly involve gene delivery using rAAV. While there are many studies on delaying disease symptoms by delivering various therapeutic agents using FUS, there is a lack of preclinical research studies on α-synuclein-based PD models. We summarized tThe most relevant preclinical studies on FUS-mediated BBB opening in PD were summarized

3. Brain Tumor

Glioblastoma is the most aggressive brain tumor with a high recurrence rate and poor prognosis despite treatments such as resection, radiotherapy, and chemotherapy [77][59]. The blood–tumor barrier (BTB) is created by the often heterogeneous disruption of the BBB within the tumor due to aberrant angiogenic signaling. As the delivery of anticancer drugs is limited despite the irregular leakiness of the BTB, quantitative drug delivery through FUS-mediated BBB opening is required [78][60]. Many previous studies on drug delivery by FUS have involved patients with brain tumors. Doxorubicin is a chemotherapeutic agent that inhibits cell growth and induces apoptosis in malignant glioma cells; however, it is not commonly used because it cannot cross the BBB. In 2007, Treat et al. delivered doxorubicin to a tumor in the brain via FUS-mediated BBB opening, indicating that this drug could be a viable treatment option [79][61]. Until now, various therapeutic agents have been used to treat glioblastomas, and FUS-mediated BBB opening technology is being developed. In the early days of FUS research, unencapsulated drugs such as the common anticancer drug temozolomide (TMZ) [80,81][62][63], carmustine (BCNU) [82][64], and immunostimulatory interleukin-12 (IL-12) [83,84][65][66] were mainly used.
Brain metastasis represents an important predictor of mortality for various non-brain cancers such as breast cancer. Like primary brain tumors, brain metastases do not have an intact BBB, but most therapeutics still have lower intra-tumoral bioavailability than non-brain tumors [85][67]. FUS studies have continued to treat metastatic brain tumors as well as primary brain tumors. In 2012, there was a study confirming the therapeutic effect by delivering Trastuzumab based on FUS-BBB opening in a breast cancer brain metastases model [86][68]. Additional research reported in 2016 demonstrated that the administration of trastuzumab and pertuzumab in a brain metastasis mouse model of breast cancer inhibited the growth of brain metastasis when used with FUS, compared to chemotherapy alone [87][69].
Whether it is a primary brain tumor or a metastatic brain tumor, the critical factor in the tumor microenvironment is to what extent the anticancer drugs could be delivered into the target region. It has been reported that the delivery of chemotherapeutic agents with small molecular weights to the brain tumor microenvironment is approximately 3.9-fold higher under FUS-mediated BBB opening conditions [88][70]. This enhanced delivery rate has been shown to increase median survival by approximately 30% compared to chemotherapy alone.
However, efflux transporters such as Pgp are overexpressed in cancer cells and prevent the uptake of anticancer drugs into the cells, resulting in resistance to them. FUS-mediated BBB opening temporarily inhibits Pgp expression, thereby preventing drug efflux and interfering with functional components of the BBB [89][71]. Additional research is needed on efflux transporter inhibitors targeting cancer cells. In addition to unencapsulated drugs, studies have reported that tumors (metastatic breast cancer) can be effectively controlled by delivering natural killer cells under BBB opening [90][72]. Furthermore, studies on suppressing brain tumors by delivering patient-specific antibodies or complexes loaded on short-hairpin RNA-liposomes have also been previously reported [91][73]. Since then, several studies have been conducted to enhance the safety and efficiency of tumor treatment by delivering encapsulated therapeutics through the conjugation of existing drugs or genes with improved MB, virus, and nanoparticles [92,93,94,95][74][75][76][77]. As immunotherapy is a critical issue in neuro-oncology, additional research on immunotherapy using FUS-mediated BBB opening is expected to become more active in the future. We summarized tThe most relevant preclinical studies on FUS-mediated BBB opening in brain tumors   were summarized (Table 3).
Table 3.
Recent preclinical studies on focused ultrasound-mediated blood–brain barrier opening in brain tumors.
Authors, Year of Publication Animal

Model
FUS Parameters Target Region Main Results
McDannold (2020) [96][78] Sprague-Dawley rats

F98 glioma
CF:230 kHz

PRF:1.1 Hz

TD:55 s

AP:119–186 kPa
Striatum

(Tumor)
It was confirmed that the ExAblate Neuro low-frequency clinical TcMRgFUS system could stably open the BBB in a rat model. Although delivery of irinotecan to the brain was not neurotoxic, it was not effective in prolonging survival or reducing the growth of gliomas.
Curley (2020) [93][75] athymic nude mice

U87 GBM
CF:1.1 MHz

DC:0.5%

TD:120 s

AP:0.45–0.55 MPa
Striatum

(Tumor)
Interstitial fluid transport in brain tumors is increased by FUS. FUS increased the dispersion of directly injected brain-penetrating nanoparticles through tumor tissue by >100%.
Englander (2021) [97][79] B6 mice

PDGF-B + PTEN−/−p53−/− murine glioma
CF:1.5 MHz

PRF:5 Hz

TD:120 s

AP:0.7 MPa
Pons

(Tumor)
FUS increased the delivery rate of etoposide into the tumor site more than five times compared to the control group, but there was no difference in survival rate or inflammation.
Sheybani (2021) [98][80] C57BL/6 mice

GL261 glioma
CF:1.1 MHz

DC:0.5%

TD:120 s

AP:0.4 MPa
Striatum

(Tumor)
[89Zr]-mCD47 (phagocytic immunotherapy) delivery with repeated FUS can significantly constrain tumor outgrowth and extend survival rate.
Ye (2021) [99][81] Swiss-Webster mice

GL261 glioma
CF:1.5 MHz

PRF:5 Hz

TD:60 s

AP:0.43 MPa
Brain stem

(Tumor)
FUS-mediated intranasal delivery increased the delivery rate of anti-PD-L1 antibodies to the brain stem by 4.03-fold.
Chen (2021) [100][82] Fisher rats

C6 glioma
CF:400 kHz

PRF:1 Hz

TD:120 s

AP:0.81 MPa
Caudate putamen

(Tumor)
CD4+ (helper TILs) and CD8+ (cytotoxic TILs) immunogenic responses were significantly increased after 7 days of FUS treatment.
Moon (2022) [101][83] BALB/c nude mice

U87 GBM
CF:1 MHz

PRF:1 Hz

TD:60 s

AP:1 W/cm2
Cerebral hemisphere Sonosensitive liposome-encapsulating doxorubicin enhances permeability by FUS-mediated BBB opening. The GBM cytotoxicity of IMP301-DC was significantly increased.
Sheybani (2022) [102][84] C57BL/6 mice

GL261 glioma
CF:1.1 MHz

PRF:1 Hz

TD:120 s

AP:0.4–0.6 MPa
Striatum

(Tumor)
FUS-mediated BBB opening in gliomas transiently induces inflammatory effects.
AP, acoustic pressure; BBB, blood–brain barrier; CF, center frequency; DC, duty cycle; FUS, focused ultrasound; GBM, glioblastoma; PRF, pulse repetition frequency; TD, train duration; TILs, tumor-infiltrating lymphocytes.

References

  1. Geerts, H.; Grossberg, G.T. Pharmacology of acetylcholinesterase inhibitors and n-methyl-d-aspartate receptors for combination therapy in the treatment of Alzheimer’s disease. J. Clin. Pharmacol. 2006, 46, 8S–16S.
  2. Asher, S.; Priefer, R. Alzheimer’s disease failed clinical trials. Life Sci. 2022, 306, 120861.
  3. Sevigny, J.; Chiao, P.; Bussière, T.; Weinreb, P.H.; Williams, L.; Maier, M.; Dunstan, R.; Salloway, S.; Chen, T.; Ling, Y. The antibody aducanumab reduces aβ plaques in Alzheimer’s disease. Nature 2016, 537, 50–56.
  4. Walsh, S.; Merrick, R.; Milne, R.; Brayne, C. Aducanumab for Alzheimer’s disease? BMJ 2021, 374, n1682.
  5. Jordão, J.F.; Ayala-Grosso, C.A.; Markham, K.; Huang, Y.; Chopra, R.; McLaurin, J.; 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.
  6. 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, 2257–2270.
  7. Xhima, K.; Markham-Coultes, K.; Nedev, H.; Heinen, S.; Saragovi, H.; Hynynen, K.; Aubert, I. Focused ultrasound delivery of a selective trka agonist rescues cholinergic function in a mouse model of Alzheimer’s disease. Sci. Adv. 2020, 6, eaax6646.
  8. 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, 12882.
  9. Xhima, K.; Markham-Coultes, K.; Hahn Kofoed, R.; Saragovi, H.U.; Hynynen, K.; Aubert, I. Ultrasound delivery of a trka agonist confers neuroprotection to Alzheimer-associated pathologies. Brain 2022, 145, 2806–2822.
  10. Dubey, S.; Heinen, S.; Krantic, S.; McLaurin, J.; Branch, D.R.; Hynynen, K.; Aubert, I. Clinically approved ivig delivered to the hippocampus with focused ultrasound promotes neurogenesis in a model of Alzheimer’s disease. Proc. Natl. Acad. Sci. USA 2020, 117, 32691–32700.
  11. Mi, X.; Du, H.; Guo, X.; Wu, Y.; Shen, L.; Luo, Y.; Wang, D.; Su, Q.; Xiang, R.; Yue, S.; et al. Asparagine endopeptidase-targeted ultrasound-responsive nanobubbles alleviate tau cleavage and amyloid-β deposition in an Alzheimer’s disease model. Acta Biomater. 2022, 141, 388–397.
  12. Zhu, Q.; Xu, X.; Chen, B.; Liao, Y.; Guan, X.; He, Y.; Cui, H.; Rong, Y.; Liu, Z.; Xu, Y. Ultrasound-targeted microbubbles destruction assists dual delivery of beta-amyloid antibody and neural stem cells to restore neural function in transgenic mice of Alzheimer’s disease. Med. Phys. 2022, 49, 1357–1367.
  13. Jordão, J.F.; Thévenot, E.; Markham-Coultes, K.; Scarcelli, T.; Weng, Y.-Q.; Xhima, K.; O’Reilly, M.; Huang, Y.; McLaurin, J.; Hynynen, K. Amyloid-β plaque reduction, endogenous antibody delivery and glial activation by brain-targeted, transcranial focused ultrasound. Exp. Neurol. 2013, 248, 16–29.
  14. Leinenga, G.; Götz, J. Scanning ultrasound removes amyloid-β and restores memory in an Alzheimer’s disease mouse model. Sci. Transl. Med. 2015, 7, 278ra233.
  15. Leinenga, G.; Koh, W.K.; Götz, J. Scanning ultrasound in the absence of blood-brain barrier opening is not sufficient to clear β-amyloid plaques in the app23 mouse model of Alzheimer’s disease. Brain Res. Bull. 2019, 153, 8–14.
  16. Poon, C.T.; Shah, K.; Lin, C.; Tse, R.; Kim, K.K.; Mooney, S.; Aubert, I.; Stefanovic, B.; Hynynen, K. Time course of focused ultrasound effects on β-amyloid plaque pathology in the tgcrnd8 mouse model of Alzheimer’s disease. Sci. Rep. 2018, 8, 14061.
  17. Karakatsani, M.E.; Kugelman, T.; Ji, R.; Murillo, M.; Wang, S.; Niimi, Y.; Small, S.A.; Duff, K.E.; Konofagou, E.E. Unilateral focused ultrasound-induced blood-brain barrier opening reduces phosphorylated tau from the rtg4510 mouse model. Theranostics 2019, 9, 5396.
  18. Pandit, R.; Leinenga, G.; Götz, J. Repeated ultrasound treatment of tau transgenic mice clears neuronal tau by autophagy and improves behavioral functions. Theranostics 2019, 9, 3754–3767.
  19. Burgess, A.; Dubey, S.; Yeung, S.; Hough, O.; Eterman, N.; Aubert, I.; Hynynen, K. Alzheimer disease in a mouse model: Mr imaging–guided focused ultrasound targeted to the hippocampus opens the blood-brain barrier and improves pathologic abnormalities and behavior. Radiology 2014, 273, 736.
  20. Leinenga, G.; Götz, J. Safety and efficacy of scanning ultrasound treatment of aged app23 mice. Front. Neurosci. 2018, 12, 55.
  21. Shin, J.; Kong, C.; Cho, J.S.; Lee, J.; Koh, C.S.; Yoon, M.-S.; Na, Y.C.; Chang, W.S.; Chang, J.W. Focused ultrasound–mediated noninvasive blood-brain barrier modulation: Preclinical examination of efficacy and safety in various sonication parameters. Neurosurg. Focus 2018, 44, E15.
  22. Shen, Y.; Hua, L.; Yeh, C.K.; Shen, L.; Ying, M.; Zhang, Z.; Liu, G.; Li, S.; Chen, S.; Chen, X.; et al. Ultrasound with microbubbles improves memory, ameliorates pathology and modulates hippocampal proteomic changes in a triple transgenic mouse model of Alzheimer’s disease. Theranostics 2020, 10, 11794–11819.
  23. Bard, F.; Cannon, C.; Barbour, R.; Burke, R.-L.; Games, D.; Grajeda, H.; Guido, T.; Hu, K.; Huang, J.; Johnson-Wood, K. Peripherally administered antibodies against amyloid β-peptide enter the central nervous system and reduce pathology in a mouse model of Alzheimer disease. Nat. Med. 2000, 6, 916–919.
  24. Kong, C.; Yang, E.-J.; Shin, J.; Park, J.; Kim, S.-H.; Park, S.-W.; Chang, W.S.; Lee, C.-H.; Kim, H.; Kim, H.-S. Enhanced delivery of a low dose of aducanumab via fus in 5× fad mice, an ad model. Transl. Neurodegener. 2022, 11, 57.
  25. Scarcelli, T.; Jordão, J.F.; O’reilly, M.A.; Ellens, N.; Hynynen, K.; Aubert, I. Stimulation of hippocampal neurogenesis by transcranial focused ultrasound and microbubbles in adult mice. Brain Stimul. 2014, 7, 304–307.
  26. Shin, J.; Kong, C.; Lee, J.; Choi, B.Y.; Sim, J.; Koh, C.S.; Park, M.; Na, Y.C.; Suh, S.W.; Chang, W.S. Focused ultrasound-induced blood-brain barrier opening improves adult hippocampal neurogenesis and cognitive function in a cholinergic degeneration dementia rat model. Alzheimer Res. Ther. 2019, 11, 110.
  27. Blackmore, D.G.; Turpin, F.; Palliyaguru, T.; Evans, H.T.; Chicoteau, A.; Lee, W.; Pelekanos, M.; Nguyen, N.; Song, J.; Sullivan, R.K. Low-intensity ultrasound restores long-term potentiation and memory in senescent mice through pleiotropic mechanisms including nmdar signaling. Mol. Psychiatry 2021, 26, 6975–6991.
  28. Mooney, S.J.; Shah, K.; Yeung, S.; Burgess, A.; Aubert, I.; Hynynen, K. Focused ultrasound-induced neurogenesis requires an increase in blood-brain barrier permeability. PLoS ONE 2016, 11, e0159892.
  29. Niu, X.; Yu, K.; He, B. Transcranial focused ultrasound induces sustained synaptic plasticity in rat hippocampus. Brain Stimul. 2022, 15, 352–359.
  30. Deng, Z.; Wang, J.; Xiao, Y.; Li, F.; Niu, L.; Liu, X.; Meng, L.; Zheng, H. Ultrasound-mediated augmented exosome release from astrocytes alleviates amyloid-β-induced neurotoxicity. Theranostics 2021, 11, 4351.
  31. Wang, F.; Wei, X.-X.; Chang, L.-S.; Dong, L.; Wang, Y.-L.; Li, N.-N. Ultrasound combined with microbubbles loading bdnf retrovirus to open bloodbrain barrier for treatment of Alzheimer’s disease. Front. Pharmacol. 2021, 12, 615104.
  32. Leinenga, G.; Koh, W.K.; Götz, J. A comparative study of the effects of aducanumab and scanning ultrasound on amyloid plaques and behavior in the app23 mouse model of Alzheimer disease. Alzheimer’s Res. Ther. 2021, 13, 76.
  33. Poon, C.; Pellow, C.; Hynynen, K. Neutrophil recruitment and leukocyte response following focused ultrasound and microbubble mediated blood-brain barrier treatments. Theranostics 2021, 11, 1655.
  34. Sun, T.; Shi, Q.; Zhang, Y.; Power, C.; Hoesch, C.; Antonelli, S.; Schroeder, M.K.; Caldarone, B.J.; Taudte, N.; Schenk, M. Focused ultrasound with anti-pglu3 aβ enhances efficacy in Alzheimer’s disease-like mice via recruitment of peripheral immune cells. J. Control. Release 2021, 336, 443–456.
  35. Luo, K.; Wang, Y.; Chen, W.-S.; Feng, X.; Liao, Y.; Chen, S.; Liu, Y.; Liao, C.; Chen, M.; Ao, L. Treatment combining focused ultrasound with gastrodin alleviates memory deficit and neuropathology in an Alzheimer’s disease-like experimental mouse model. Neural Plast. 2022, 2022, 5241449.
  36. Bathini, P.; Sun, T.; Schenk, M.; Schilling, S.; McDannold, N.J.; Lemere, C.A. Acute effects of focused ultrasound-induced blood-brain barrier opening on anti-pyroglu3 abeta antibody delivery and immune responses. Biomolecules 2022, 12, 951.
  37. Bajracharya, R.; Cruz, E.; Götz, J.; Nisbet, R.M. Ultrasound-mediated delivery of novel tau-specific monoclonal antibody enhances brain uptake but not therapeutic efficacy. J. Control. Release 2022, 349, 634–648.
  38. Rodrigues e Silva, A.M.; Geldsetzer, F.; Holdorff, B.; Kielhorn, F.W.; Balzer-Geldsetzer, M.; Oertel, W.H.; Hurtig, H.; Dodel, R. Who was the man who discovered the “lewy bodies”? Mov. Disord. 2010, 25, 1765–1773.
  39. Appel-Cresswell, S.; Vilarino-Guell, C.; Encarnacion, M.; Sherman, H.; Yu, I.; Shah, B.; Weir, D.; Thompson, C.; Szu-Tu, C.; Trinh, J. Alpha-synuclein p. H50q, a novel pathogenic mutation for Parkinson’s disease. Mov. Disord. 2013, 28, 811–813.
  40. Choi-Lundberg, D.L.; Lin, Q.; Chang, Y.-N.; Chiang, Y.L.; Hay, C.M.; Mohajeri, H.; Davidson, B.L.; Bohn, M.C. Dopaminergic neurons protected from degeneration by gdnf gene therapy. Science 1997, 275, 838–841.
  41. Kearns, C.M.; Gash, D.M. Gdnf protects nigral dopamine neurons against 6-hydroxydopamine in vivo. Brain Res. 1995, 672, 104–111.
  42. Burke, R.E. GDNF as a candidate striatal target-derived neurotrophic factor for the development of substantia nigra dopamine neurons. J. Neural. Transm. Suppl. 2006, 70, 41–45.
  43. Kordower, J.H.; Emborg, M.E.; Bloch, J.; Ma, S.Y.; Chu, Y.; Leventhal, L.; McBride, J.; Chen, E.-Y.; Palfi, S.; Roitberg, B.Z. Neurodegeneration prevented by lentiviral vector delivery of gdnf in primate models of Parkinson’s disease. Science 2000, 290, 767–773.
  44. Gash, D.M.; Zhang, Z.; Ovadia, A.; Cass, W.A.; Yi, A.; Simmerman, L.; Russell, D.; Martin, D.; Lapchak, P.A.; Collins, F. Functional recovery in Parkinsonian monkeys treated with gdnf. Nature 1996, 380, 252–255.
  45. Grondin, R.; Zhang, Z.; Yi, A.; Cass, W.A.; Maswood, N.; Andersen, A.H.; Elsberry, D.D.; Klein, M.C.; Gerhardt, G.A.; Gash, D.M. Chronic, controlled gdnf infusion promotes structural and functional recovery in advanced Parkinsonian monkeys. Brain 2002, 125, 2191–2201.
  46. Lang, A.E.; Gill, S.; Patel, N.K.; Lozano, A.; Nutt, J.G.; Penn, R.; Brooks, D.J.; Hotton, G.; Moro, E.; Heywood, P. Randomized controlled trial of intraputamenal glial cell line–derived neurotrophic factor infusion in Parkinson disease. Ann. Neurol. 2006, 59, 459–466.
  47. Lin, C.Y.; Hsieh, H.Y.; Chen, C.M.; Wu, S.R.; Tsai, C.H.; Huang, C.Y.; Hua, M.Y.; Wei, K.C.; Yeh, C.K.; Liu, H.L. 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.
  48. Fan, C.-H.; Ting, C.-Y.; Lin, C.Y.; Chan, H.-L.; Chang, Y.-C.; Chen, Y.-Y.; Liu, H.-L.; Yeh, C.-K. Noninvasive, targeted and non-viral ultrasound-mediated gdnf-plasmid delivery for treatment of Parkinson’s disease. Sci. Rep. 2016, 6, 19579.
  49. Yue, P.; Miao, W.; Gao, L.; Zhao, X.; Teng, J. 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.
  50. Grondin, R.; Zhang, Z.; Ai, Y.; Ding, F.; Walton, A.; Surgener, S.; Gerhardt, G.; Gash, D. Intraputamenal infusion of exogenous neurturin protein restores motor and dopaminergic function in the globus pallidus of mptp-lesioned rhesus monkeys. Cell Transplant. 2008, 17, 373–381.
  51. Samiotaki, G.; Acosta, C.; Wang, S.; Konofagou, E.E. 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, 611–622.
  52. Karakatsani, M.E.; Wang, S.; Samiotaki, G.; Kugelman, T.; Olumolade, O.O.; Acosta, C.; Sun, T.; Han, Y.; Kamimura, H.A.; Jackson-Lewis, V. Amelioration of the nigrostriatal pathway facilitated by ultrasound-mediated neurotrophic delivery in early Parkinson’s disease. J. Control. Release 2019, 303, 289–301.
  53. Noroozian, Z.; Xhima, K.; Huang, Y.; Kaspar, B.K.; Kügler, S.; Hynynen, K.; Aubert, I. Mri-guided focused ultrasound for targeted delivery of raav to the brain. In Adeno-Associated Virus Vectors: Design and Delivery; Springer: Berlin/Heidelberg, Germany, 2019; pp. 177–197.
  54. Ji, R.; Smith, M.; Niimi, Y.; Karakatsani, M.E.; Murillo, M.F.; Jackson-Lewis, V.; Przedborski, S.; Konofagou, E.E. Focused ultrasound enhanced intranasal delivery of brain derived neurotrophic factor produces neurorestorative effects in a Parkinson’s disease mouse model. Sci. Rep. 2019, 9, 19402.
  55. Lin, C.-Y.; Lin, Y.-C.; Huang, C.-Y.; Wu, S.-R.; Chen, C.-M.; Liu, H.-L. Ultrasound-responsive neurotrophic factor-loaded microbubble-liposome complex: Preclinical investigation for Parkinson’s disease treatment. J. Control. Release 2020, 321, 519–528.
  56. Yan, Y.; Chen, Y.; Liu, Z.; Cai, F.; Niu, W.; Song, L.; Liang, H.; Su, Z.; Yu, B.; Yan, F. Brain delivery of curcumin through low-intensity ultrasound-induced blood–brain barrier opening via lipid-plga nanobubbles. Int. J. Nanomed. 2021, 16, 7433.
  57. Wang, Y.; Luo, K.; Li, J.; Liao, Y.; Liao, C.; Chen, W.-S.; Chen, M.; Ao, L. Focused ultrasound promotes the delivery of gastrodin and enhances the protective effect on dopaminergic neurons in a mouse model of Parkinson’s disease. Front. Cell. Neurosci. 2022, 16, 884788.
  58. Trinh, D.; Nash, J.; Goertz, D.; Hynynen, K.; Bulner, S.; Iqbal, U.; Keenan, J. Microbubble drug conjugate and focused ultrasound blood brain barrier delivery of aav-2 sirt-3. Drug Deliv. 2022, 29, 1176–1183.
  59. DeCordova, S.; Shastri, A.; Tsolaki, A.G.; Yasmin, H.; Klein, L.; Singh, S.K.; Kishore, U. Molecular heterogeneity and immunosuppressive microenvironment in glioblastoma. Front. Immunol. 2020, 11, 1402.
  60. Bunevicius, A.; McDannold, N.J.; Golby, A.J. Focused ultrasound strategies for brain tumor therapy. Oper. Neurosurg. 2020, 19, 9–18.
  61. Treat, L.H.; McDannold, N.; Vykhodtseva, N.; Zhang, Y.; Tam, K.; Hynynen, K. Targeted delivery of doxorubicin to the rat brain at therapeutic levels using mri-guided focused ultrasound. Int. J. Cancer 2007, 121, 901–907.
  62. Wei, K.-C.; Chu, P.-C.; Wang, H.-Y.J.; Huang, C.-Y.; Chen, P.-Y.; Tsai, H.-C.; Lu, Y.-J.; Lee, P.-Y.; Tseng, I.-C.; Feng, L.-Y. Focused ultrasound-induced blood–brain barrier opening to enhance temozolomide delivery for glioblastoma treatment: A preclinical study. PLoS ONE 2013, 8, e58995.
  63. Liu, H.-L.; Huang, C.-Y.; Chen, J.-Y.; Wang, H.-Y.J.; Chen, P.-Y.; Wei, K.-C. Pharmacodynamic and therapeutic investigation of focused ultrasound-induced blood-brain barrier opening for enhanced temozolomide delivery in glioma treatment. PLoS ONE 2014, 9, e114311.
  64. Liu, H.-L.; Hua, M.-Y.; Chen, P.-Y.; Chu, P.-C.; Pan, C.-H.; Yang, H.-W.; Huang, C.-Y.; Wang, J.-J.; Yen, T.-C.; Wei, K.-C. Blood-brain barrier disruption with focused ultrasound enhances delivery of chemotherapeutic drugs for glioblastoma treatment. Radiology 2010, 255, 415–425.
  65. Chen, P.-Y.; Hsieh, H.-Y.; Huang, C.-Y.; Lin, C.-Y.; Wei, K.-C.; Liu, H.-L. Focused ultrasound-induced blood–brain barrier opening to enhance interleukin-12 delivery for brain tumor immunotherapy: A preclinical feasibility study. J. Transl. Med. 2015, 13, 93.
  66. Chen, P.-Y.; Wei, K.-C.; Liu, H.-L. Neural immune modulation and immunotherapy assisted by focused ultrasound induced blood-brain barrier opening. Hum. Vaccines Immunother. 2015, 11, 2682–2687.
  67. Yonemori, K.; Tsuta, K.; Ono, M.; Shimizu, C.; Hirakawa, A.; Hasegawa, T.; Hatanaka, Y.; Narita, Y.; Shibui, S.; Fujiwara, Y. Disruption of the blood brain barrier by brain metastases of triple-negative and basal-type breast cancer but not her2/neu-positive breast cancer. Cancer Interdiscip. Int. J. Am. Cancer Soc. 2010, 116, 302–308.
  68. Park, E.J.; Zhang, Y.Z.; Vykhodtseva, N.; McDannold, N. Ultrasound-mediated blood-brain/blood-tumor barrier disruption improves outcomes with trastuzumab in a breast cancer brain metastasis model. J. Control. Release 2012, 163, 277–284.
  69. Kobus, T.; Zervantonakis, I.K.; Zhang, Y.; McDannold, N.J. Growth inhibition in a brain metastasis model by antibody delivery using focused ultrasound-mediated blood-brain barrier disruption. J. Control. Release 2016, 238, 281–288.
  70. Schoen, S.; Kilinc, M.S.; Lee, H.; Guo, Y.; Degertekin, F.L.; Woodworth, G.F.; Arvanitis, C. Towards controlled drug delivery in brain tumors with microbubble-enhanced focused ultrasound. Adv. Drug Deliv. Rev. 2022, 180, 114043.
  71. Aryal, M.; Fischer, K.; Gentile, C.; Gitto, S.; Zhang, Y.-Z.; McDannold, N. Effects on p-glycoprotein expression after blood-brain barrier disruption using focused ultrasound and microbubbles. PLoS ONE 2017, 12, e0166061.
  72. Alkins, R.; Burgess, A.; Kerbel, R.; Wels, W.S.; Hynynen, K. Early treatment of her2-amplified brain tumors with targeted nk-92 cells and focused ultrasound improves survival. Neuro-Oncology 2016, 18, 974–981.
  73. Zhao, G.; Huang, Q.; Wang, F.; Zhang, X.; Hu, J.; Tan, Y.; Huang, N.; Wang, Z.; Wang, Z.; Cheng, Y. Targeted shrna-loaded liposome complex combined with focused ultrasound for blood brain barrier disruption and suppressing glioma growth. Cancer Lett. 2018, 418, 147–158.
  74. Sun, T.; Zhang, Y.; Power, C.; Alexander, P.M.; Sutton, J.T.; Aryal, M.; Vykhodtseva, N.; Miller, E.L.; McDannold, N.J. Closed-loop control of targeted ultrasound drug delivery across the blood–brain/tumor barriers in a rat glioma model. Proc. Natl. Acad. Sci. USA 2017, 114, E10281–E10290.
  75. Curley, C.T.; Mead, B.P.; Negron, K.; Kim, N.; Garrison, W.J.; Miller, G.W.; Kingsmore, K.M.; Thim, E.A.; Song, J.; Munson, J.M. Augmentation of brain tumor interstitial flow via focused ultrasound promotes brain-penetrating nanoparticle dispersion and transfection. Sci. Adv. 2020, 6, eaay1344.
  76. Zhang, D.Y.; Dmello, C.; Chen, L.; Arrieta, V.A.; Gonzalez-Buendia, E.; Kane, J.R.; Magnusson, L.P.; Baran, A.; James, C.D.; Horbinski, C. Ultrasound-mediated delivery of paclitaxel for glioma: A comparative study of distribution, toxicity, and efficacy of albumin-bound versus cremophor formulationsus-delivered abx extends survival in gbm pdx mouse model. Clin. Cancer Res. 2020, 26, 477–486.
  77. Yang, Q.; Zhou, Y.; Chen, J.; Huang, N.; Wang, Z.; Cheng, Y. Gene therapy for drug-resistant glioblastoma via lipid-polymer hybrid nanoparticles combined with focused ultrasound. Int. J. Nanomed. 2021, 16, 185.
  78. McDannold, N.; Zhang, Y.; Supko, J.G.; Power, C.; Sun, T.; Vykhodtseva, N.; Golby, A.J.; Reardon, D.A. Blood-brain barrier disruption and delivery of irinotecan in a rat model using a clinical transcranial mri-guided focused ultrasound system. Sci. Rep. 2020, 10, 8766.
  79. Englander, Z.K.; Wei, H.-J.; Pouliopoulos, A.N.; Bendau, E.; Upadhyayula, P.; Jan, C.-I.; Spinazzi, E.F.; Yoh, N.; Tazhibi, M.; McQuillan, N.M. Focused ultrasound mediated blood–brain barrier opening is safe and feasible in a murine pontine glioma model. Sci. Rep. 2021, 11, 6521.
  80. Sheybani, N.D.; Breza, V.R.; Paul, S.; McCauley, K.S.; Berr, S.S.; Miller, G.W.; Neumann, K.D.; Price, R.J. Immunopet-informed sequence for focused ultrasound-targeted mcd47 blockade controls glioma. J. Control. Release 2021, 331, 19–29.
  81. Ye, D.; Yuan, J.; Yue, Y.; Rubin, J.B.; Chen, H. Focused ultrasound-enhanced delivery of intranasally administered anti-programmed cell death-ligand 1 antibody to an intracranial murine glioma model. Pharmaceutics 2021, 13, 190.
  82. Chen, K.-T.; Chai, W.-Y.; Lin, Y.-J.; Lin, C.-J.; Chen, P.-Y.; Tsai, H.-C.; Huang, C.-Y.; Kuo, J.S.; Liu, H.-L.; Wei, K.-C. Neuronavigation-guided focused ultrasound for transcranial blood-brain barrier opening and immunostimulation in brain tumors. Sci. Adv. 2021, 7, eabd0772.
  83. Moon, H.; Hwang, K.; Nam, K.M.; Kim, Y.-S.; Ko, M.J.; Kim, H.R.; Lee, H.J.; Kim, M.J.; Kim, T.H.; Kang, K.-S. Enhanced delivery to brain using sonosensitive liposome and microbubble with focused ultrasound. Biomater. Adv. 2022, 141, 213102.
  84. Sheybani, N.D.; Witter, A.R.; Garrison, W.J.; Miller, G.W.; Price, R.J.; Bullock, T.N. Profiling of the immune landscape in murine glioblastoma following blood brain/tumor barrier disruption with mr image-guided focused ultrasound. J. Neuro-Oncol. 2022, 156, 109–122.
More