Pilot study of repeated blood-brain barrier disruption in patients with mild Alzheimer's disease with an implantable ultrasound device

Stéphane Epelbaum, Ninon Burgos, Michael Canney, Dawn Matthews, Marion Houot, Mathieu D Santin, Carole Desseaux, Guillaume Bouchoux, Sebastian Stroer, Cyril Martin, Marie-Odile Habert, Marcel Levy, Aicha Bah, Karine Martin, Benoît Delatour, Maximilien Riche, Bruno Dubois, Lisa Belin, Alexandre Carpentier, Stéphane Epelbaum, Ninon Burgos, Michael Canney, Dawn Matthews, Marion Houot, Mathieu D Santin, Carole Desseaux, Guillaume Bouchoux, Sebastian Stroer, Cyril Martin, Marie-Odile Habert, Marcel Levy, Aicha Bah, Karine Martin, Benoît Delatour, Maximilien Riche, Bruno Dubois, Lisa Belin, Alexandre Carpentier

Abstract

Background: Temporary disruption of the blood-brain barrier (BBB) using pulsed ultrasound leads to the clearance of both amyloid and tau from the brain, increased neurogenesis, and mitigation of cognitive decline in pre-clinical models of Alzheimer's disease (AD) while also increasing BBB penetration of therapeutic antibodies. The goal of this pilot clinical trial was to investigate the safety and efficacy of this approach in patients with mild AD using an implantable ultrasound device.

Methods: An implantable, 1-MHz ultrasound device (SonoCloud-1) was implanted under local anesthesia in the skull (extradural) of 10 mild AD patients to target the left supra-marginal gyrus. Over 3.5 months, seven ultrasound sessions in combination with intravenous infusion of microbubbles were performed twice per month to temporarily disrupt the BBB. 18F-florbetapir and 18F-fluorodeoxyglucose positron emission tomography (PET) imaging were performed on a combined PET/MRI scanner at inclusion and at 4 and 8 months after the initiation of sonications to monitor the brain metabolism and amyloid levels along with cognitive evaluations. The evolution of cognitive and neuroimaging features was compared to that of a matched sample of control participants taken from the Alzheimer's Disease Neuroimaging Initiative (ADNI).

Results: A total of 63 BBB opening procedures were performed in nine subjects. The procedure was well-tolerated. A non-significant decrease in amyloid accumulation at 4 months of - 6.6% (SD = 7.2%) on 18F-florbetapir PET imaging in the sonicated gray matter targeted by the ultrasound transducer was observed compared to baseline in six subjects that completed treatments and who had evaluable imaging scans. No differences in the longitudinal change in the glucose metabolism were observed compared to the neighboring or contralateral regions or to the change observed in the same region in ADNI participants. No significant effect on cognition evolution was observed in comparison with the ADNI participants as expected due to the small sample size and duration of the trial.

Conclusions: These results demonstrate the safety of ultrasound-based BBB disruption and the potential of this technology to be used as a therapy for AD patients. Research of this technique in a larger clinical trial with a device designed to sonicate larger volumes of tissue and in combination with disease-modifying drugs may further enhance the effects observed.

Trial registration: ClinicalTrials.gov, NCT03119961.

Keywords: Alzheimer’s disease; Amyloid; Blood-brain barrier; Clinical trial; Florbetapir; Magnetic resonance imaging; Position emission tomography; Ultrasound.

Conflict of interest statement

Michael Canney, Carole Desseaux, Guillaume Bouchoux, and Cyril Martin are employees of Carthera and have an ownership interest in the company. Alexandre Carpentier is a paid consultant to Carthera and has an ownership interest in Carthera.

© 2022. The Author(s).

Figures

Fig. 1
Fig. 1
The SonoCloud-1 is a 1-MHz implantable ultrasound device that is implanted within the skull thickness (extradural) and activated at each treatment cycle by connecting it using a transdermal needle to an external radiofrequency generator. In this study, the SonoCloud-1 was implanted to target and temporarily disrupt the BBB above the left supramarginal gyrus
Fig. 2
Fig. 2
Region of interest (ROI) definition to measure the changes in PET amyloid levels in gray matter targeted by the implant (ROI1) as compared to similar tissue in the opposite (ROI2) and same hemispheres (ROI3)
Fig. 3
Fig. 3
Trial overview
Fig. 4
Fig. 4
PET amyloid (florbetapir) change observed in the region targeted by the SonoCloud-1 device (ROI1). SUVRs referenced to a ROI similar to the target ROI but in the opposite hemisphere (ROI2) showed decreases from the baseline of − 6.6% (SD = 7.2%) at 4 months and − 5.7% (SD = 6.2%) at 8 months
Fig. 5
Fig. 5
FDG PET changes in the target ROI (ROI1) referenced to the whole cerebellum. Overall, a blunting or plateau of decline was observed in the target region referenced to the whole cerebellum after 4 months

References

    1. Cummings J, Lee G, Ritter A, Sabbagh M, Zhong K. Alzheimer’s disease drug development pipeline: 2020. Alzheimer’s Dement Transl Res Clin Interv. 2020;6(1):e12050.
    1. Tustison NJ, Avants BB, Cook PA, Zheng Y, Egan A, Yushkevich PA, et al. N4ITK: improved N3 bias correction. IEEE Trans Med Imaging. 2010;29(6):1310–1320.
    1. Modat M, Cash DM, Daga P, Winston GP, Duncan JS, Ourselin S. Global image registration using a symmetric block-matching approach. J Med Imaging. 2014;1(2):24003.
    1. Cardoso MJ, Modat M, Wolz R, Melbourne A, Cash D, Rueckert D, et al. Geodesic information flows: spatially-variant graphs and their application to segmentation and fusion. IEEE Trans Med Imaging. 2015;34(9):1976–1988.
    1. Rasmussen JM, Lakatos A, van Erp TGM, Kruggel F, Keator DB, Fallon JT, et al. Empirical derivation of the reference region for computing diagnostic sensitive 18fluorodeoxyglucose ratios in Alzheimer’s disease based on the ADNI sample. Biochim Biophys Acta (BBA)-Molecular Basis Dis. 2012;1822(3):457–466.
    1. Landau SM, Fero A, Baker SL, Koeppe R, Mintun M, Chen K, et al. Measurement of longitudinal β-amyloid change with 18F-florbetapir PET and standardized uptake value ratios. J Nucl Med. 2015;56(4):567–574.
    1. Howard R, Liu KY. Questions EMERGE as Biogen claims aducanumab turnaround. Nat Rev Neurol. 2020;16(2):63–4. 10.1038/s41582-019-0295-9.
    1. Schneider L. A resurrection of aducanumab for Alzheimer’s disease. Lancet Neurol. 2020;19(2):111–2. 10.1016/S1474-4422(19)30480-6.
    1. Sevigny J, Chiao P, Bussière T, Weinreb PH, Williams L, Maier M, et al. The antibody aducanumab reduces Aβ plaques in Alzheimer’s disease. Nature. 2016;537(7618):50–56.
    1. Banks WA, Terrell B, Farr SA, Robinson SM, Nonaka N, Morley JE. Passage of amyloid β protein antibody across the blood–brain barrier in a mouse model of Alzheimer’s disease. Peptides. 2002;23(12):2223–2226.
    1. Pardridge WM. Targeted delivery of protein and gene medicines through the blood-brain barrier. Clin Pharmacol Ther. 2015;97(4):347–361.
    1. 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.
    1. Dréan A, Lemaire N, Bouchoux G, Goldwirt L, Canney M, Goli L, et al. Temporary blood–brain barrier disruption by low intensity pulsed ultrasound increases carboplatin delivery and efficacy in preclinical models of glioblastoma. J Neurooncol. 2019;144(1).
    1. Zhang DY, Dmello C, Chen L, Arrieta VA, Gonzalez-Buendia E, Robert Kane J, et al. Ultrasound-mediated delivery of paclitaxel for glioma: a comparative study of distribution, toxicity, and efficacy of albumin-bound versus cremophor formulations. Clin Cancer Res. 2020;26(2):477–486.
    1. Leinenga G, Koh WK, 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. bioRxiv. 2021:2001–21.
    1. Jordão JF, Thévenot E, Markham-Coultes K, Scarcelli T, Weng YQ, Xhima K, et al. Amyloid-β plaque reduction, endogenous antibody delivery and glial activation by brain-targeted, transcranial focused ultrasound. Exp Neurol. 2013;248:16–29.
    1. 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(13):3754–67.
    1. Poon C, Pellow C, Hynynen K. Neutrophil recruitment and leukocyte response following focused ultrasound and microbubble mediated blood-brain barrier treatments. Theranostics. 2021;11(4):1655.
    1. Kinoshita M, McDannold N, Jolesz FA, Hynynen K. Targeted delivery of antibodies through the blood-brain barrier by MRI-guided focused ultrasound. Biochem Biophys Res Commun. 2006;340(4):1085–1090.
    1. Alkins R, Burgess A, Ganguly M, Francia G, Kerbel R, Wels WS, et al. Focused ultrasound delivers targeted immune cells to metastatic brain tumors. Cancer Res. 2013;73(6):1892–1899.
    1. Mainprize T, Lipsman N, Huang Y, Meng Y, Bethune A, Ironside S, et al. Blood-brain barrier opening in primary brain tumors with non-invasive MR-guided focused ultrasound: a clinical safety and feasibility study. Sci Rep. 2019;9(1):321.
    1. Carpentier A, Canney M, Vignot A, Reina V, Beccaria K, Horodyckid C, et al. Clinical trial of blood-brain barrier disruption by pulsed ultrasound. Sci Transl Med. 2016;8(343):343re2.
    1. Idbaih A, Canney M, Belin L, Desseaux C, Vignot A, Bouchoux G, 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(13).
    1. Park SH, Kim MJ, Jung HH, Chang WS, Choi HS, Rachmilevitch I, et al. One-year outcome of multiple blood–brain barrier disruptions with temozolomide for the treatment of glioblastoma. Front Oncol. 2020;10:1663.
    1. Rezai AR, Ranjan M, D’Haese PF, Haut MW, Carpenter J, Najib U, et al. Noninvasive hippocampal blood−brain barrier opening in Alzheimer’s disease with focused ultrasound. Proc Natl Acad Sci U S A. 2020; [cited 2020 Oct 26];117(17):9180–2. Available from: .
    1. Abrahao A, Meng Y, Llinas M, Huang Y, Hamani C, Mainprize T, et al. First-in-human trial of blood–brain barrier opening in amyotrophic lateral sclerosis using MR-guided focused ultrasound. Nat Commun. 2019;10(1):1–9.
    1. Rezai AR, Ranjan M, D’Haese P-F, Haut MW, Carpenter J, Najib U, et al. Noninvasive hippocampal blood−brain barrier opening in Alzheimer’s disease with focused ultrasound. Proc Natl Acad Sci. 2020;117(17):9180–9182.
    1. D’Haese P-F, Ranjan M, Song A, Haut MW, Carpenter J, Dieb G, et al. β-Amyloid plaque reduction in the hippocampus after focused ultrasound-induced blood–brain barrier opening in Alzheimer’s disease. Front Hum Neurosci. 2020;14:422.
    1. Dubois B, Feldman HH, Jacova C, Hampel H, Molinuevo JL, Blennow K, et al. Advancing research diagnostic criteria for Alzheimer’s disease: the IWG-2 criteria. Lancet Neurol. 2014;13(6):614–629.
    1. Teichmann M, Epelbaum S, Samri D, Nogueira ML, Michon A, Hampel H, et al. Free and Cued Selective Reminding Test–accuracy for the differential diagnosis of Alzheimer’s and neurodegenerative diseases: a large-scale biomarker-characterized monocenter cohort study (ClinAD) Alzheimer’s Dement. 2017;13(8):913–923.
    1. Asquier N, Bouchoux G, Canney M, Martin C, Law-Ye B, Leclercq D, et al. Blood-brain barrier disruption in humans using an implantable ultrasound device: quantification with MR images and correlation with local acoustic pressure. J Neurosurg. 2019. 10.3171/2018.9.JNS182001 in press.
    1. Folstein MF, Folstein SE, McHugh PR. “Mini-mental state”: a practical method for grading the cognitive state of patients for the clinician. J Psychiatr Res. 1975;12(3):189–198.
    1. Berg L, Miller JP, Baty J, Rubin EH, Morris JC, Figiel G. Mild senile dementia of the Alzheimer type. 4. Evaluation of intervention. Ann Neurol Off J Am Neurol Assoc Child Neurol Soc. 1992;31(3):242–249.
    1. O’Bryant SE, Waring SC, Cullum CM, Hall J, Lacritz L, Massman PJ, et al. Staging dementia using Clinical Dementia Rating Scale Sum of Boxes scores: a Texas Alzheimer’s research consortium study. Arch Neurol. 2008;65(8):1091–1095.
    1. Dubois B, Slachevsky A, Litvan I, Pillon B. The FAB: a frontal assessment battery at bedside. Neurology. 2000;55(11):1621–1626.
    1. Grober E, Buschke H, Crystal H, Bang S, Dresner R. Screening for dementia by memory testing. Neurology. 1988;38(6):900.
    1. Reitan RM. Validity of the Trail Making Test as an indicator of organic brain damage. Percept Mot Skills. 1958;8(3):271–276.
    1. Cardebat D, Doyon B, Puel M, Goulet P, Joanette Y. Formal and semantic lexical evocation in normal subjects. Performance and dynamics of production as a function of sex, age and educational level. Acta Neurol Belg. 1990;90(4):207–217.
    1. Mahieux-Laurent F, Fabre C, Galbrun E, Dubrulle A, Moroni C. Validation of a brief screening scale evaluating praxic abilities for use in memory clinics. Evaluation in 419 controls, 127 mild cognitive impairment and 320 demented patients. Rev Neurol (Paris) 2009;165(6–7):560–567.
    1. Rey A. L’examen psychologique dans les cas d’encéphalopathie traumatique. (Les problems.). Arch Psychol (Geneve). 1941;28:215–85.
    1. Spielberger CD. Manual for the State-Trait Anxiety Inventory: STAI (Form Y) Palo Alto: Consulting Psychologists Press; 1983.
    1. Montgomery S, Asberg M. A new depression scale designed to be sensitive to change. Br J Psychiatry. 1979;134(4):382–389.
    1. Wiesinger F, Sacolick LI, Menini A, Kaushik SS, Ahn S, Veit-Haibach P, et al. Zero TE MR bone imaging in the head. Magn Reson Med. 2016;75(1):107–114.
    1. Shen Y, Goerner FL, Snyder C, Morelli JN, Hao D, Hu D, et al. T1 relaxivities of gadolinium-based magnetic resonance contrast agents in human whole blood at 1.5, 3, and 7 T. Invest Radiol. 2015;50(5):330–338.
    1. Chen K, Roontiva A, Thiyyagura P, Lee W, Liu X, Ayutyanont N, et al. Improved power for characterizing longitudinal amyloid-β PET changes and evaluating amyloid-modifying treatments with a cerebral white matter reference region. J Nucl Med. 2015;56(4):560–566.
    1. Strother S, Oder A, Spring R, Grady C. Proceedings of COMPSTAT’2010. Springer; 2010. The NPAIRS computational statistics framework for data analysis in neuroimaging; pp. 111–120.
    1. Spiegel R, Berres M, Miserez AR, Monsch AU. For debate: substituting placebo controls in long-term Alzheimer’s prevention trials. Alzheimers Res Ther. 2011;3(2):1–11.
    1. Jack CR, Jr, Barnes J, Bernstein MA, Borowski BJ, Brewer J, Clegg S, et al. Magnetic resonance imaging in Alzheimer’s disease neuroimaging initiative 2. Alzheimer’s Dement. 2015;11(7):740–756.
    1. Jagust WJ, Landau SM, Koeppe RA, Reiman EM, Chen K, Mathis CA, et al. The Alzheimer’s disease neuroimaging initiative 2 PET core: 2015. Alzheimer’s Dement. 2015;11(7):757–771.
    1. Dréan A, Lemaire N, Bouchoux G, Goldwirt L, Canney M, Goli L, et al. Temporary blood–brain barrier disruption by low intensity pulsed ultrasound increases carboplatin delivery and efficacy in preclinical models of glioblastoma. J Neurooncol. 2019;144:33–41.
    1. Park SH, Baik K, Jeon S, Chang WS, Ye BS, Chang JW. Extensive frontal focused ultrasound mediated blood–brain barrier opening for the treatment of Alzheimer’s disease: a proof-of-concept study. Transl Neurodegener. 2021;10(1):1–11.
    1. Jeong H, Im JJ, Park J-S, Na S-H, Lee W, Yoo S-S, et al. A pilot clinical study of low-intensity transcranial focused ultrasound in Alzheimer’s disease. Ultrasonography. 2021;40(4):512–19.
    1. Mintun MA, Lo AC, Duggan Evans C, Wessels AM, Ardayfio PA, Andersen SW, et al. Donanemab in early Alzheimer’s disease. N Engl J Med. 2021;384(18):1691–1704.
    1. Blackmore DG, Turpin F, Palliyaguru T, Evans HT, Chicoteau A, Lee W, et al. Low-intensity ultrasound restores long-term potentiation and memory in senescent mice through pleiotropic mechanisms including NMDAR signaling. Mol Psychiatry. 2021:1–17.

Source: PubMed

3
Tilaa