Ambroxol for the Treatment of Patients With Parkinson Disease With and Without Glucocerebrosidase Gene Mutations: A Nonrandomized, Noncontrolled Trial

Stephen Mullin, Laura Smith, Katherine Lee, Gayle D'Souza, Philip Woodgate, Josh Elflein, Jenny Hällqvist, Marco Toffoli, Adam Streeter, Joanne Hosking, Wendy E Heywood, Rajeshree Khengar, Philip Campbell, Jason Hehir, Sarah Cable, Kevin Mills, Henrik Zetterberg, Patricia Limousin, Vincenzo Libri, Tom Foltynie, Anthony H V Schapira, Stephen Mullin, Laura Smith, Katherine Lee, Gayle D'Souza, Philip Woodgate, Josh Elflein, Jenny Hällqvist, Marco Toffoli, Adam Streeter, Joanne Hosking, Wendy E Heywood, Rajeshree Khengar, Philip Campbell, Jason Hehir, Sarah Cable, Kevin Mills, Henrik Zetterberg, Patricia Limousin, Vincenzo Libri, Tom Foltynie, Anthony H V Schapira

Abstract

Importance: Mutations of the glucocerebrosidase gene, GBA1 (OMIM 606463), are the most important risk factor for Parkinson disease (PD). In vitro and in vivo studies have reported that ambroxol increases β-glucocerebrosidase (GCase) enzyme activity and reduces α-synuclein levels. These observations support a potential role for ambroxol therapy in modifying a relevant pathogenetic pathway in PD.

Objective: To assess safety, tolerability, cerebrospinal fluid (CSF) penetration, and target engagement of ambroxol therapy with GCase in patients with PD with and without GBA1 mutations.

Interventions: An escalating dose of oral ambroxol to 1.26 g per day.

Design, setting, and participants: This single-center open-label noncontrolled clinical trial was conducted between January 11, 2017, and April 25, 2018, at the Leonard Wolfson Experimental Neuroscience Centre, a dedicated clinical research facility and part of the University College London Queen Square Institute of Neurology in London, United Kingdom. Participants were recruited from established databases at the Royal Free London Hospital and National Hospital for Neurology and Neurosurgery in London. Twenty-four patients with moderate PD were evaluated for eligibility, and 23 entered the study. Of those, 18 patients completed the study; 1 patient was excluded (failed lumbar puncture), and 4 patients withdrew (predominantly lumbar puncture-related complications). All data analyses were performed from November 1 to December 14, 2018.

Main outcomes and measures: Primary outcomes at 186 days were the detection of ambroxol in the CSF and a change in CSF GCase activity.

Results: Of the 18 participants (15 men [83.3%]; mean [SD] age, 60.2 [9.7] years) who completed the study, 17 (8 with GBA1 mutations and 9 without GBA1 mutations) were included in the primary analysis. Between days 0 and 186, a 156-ng/mL increase in the level of ambroxol in CSF (lower 95% confidence limit, 129 ng/mL; P < .001) was observed. The CSF GCase activity decreased by 19% (0.059 nmol/mL per hour; 95% CI, -0.115 to -0.002; P = .04). The ambroxol therapy was well tolerated, with no serious adverse events. An increase of 50 pg/mL (13%) in the CSF α-synuclein concentration (95% CI, 14-87; P = .01) and an increase of 88 ng/mol (35%) in the CSF GCase protein levels (95% CI, 40-137; P = .002) were observed. Mean (SD) scores on part 3 of the Movement Disorders Society Unified Parkinson Disease Rating Scale decreased (ie, improved) by 6.8 (7.1) points (95% CI, -10.4 to -3.1; P = .001). These changes were observed in patients with and without GBA1 mutations.

Conclusions and relevance: The study results suggest that ambroxol therapy was safe and well tolerated; CSF penetration and target engagement of ambroxol were achieved, and CSF α-synuclein levels were increased. Placebo-controlled clinical trials are needed to examine whether ambroxol therapy is associated with changes in the natural progression of PD.

Trial registration: ClinicalTrials.gov identifier: NCT02941822; EudraCT identifier: 2015-002571-24.

Conflict of interest statement

Conflict of Interest Disclosures: Dr Foltynie reported having received honoraria for speaking at meetings supported by Profile Pharma, BIAL, AbbVie; having served on advisory boards for BIAL, Oxford Biomedica, Living Cell Technologies, and Peptron. Dr Zetterberg reported having served at scientific advisory boards for Roche Diagnostics, Wave, Samumed, and CogRx; having given lectures in symposia sponsored by Alzecure and Biogen; and reported being a cofounder of Brain Biomarker Solutions in Gothenburg AB, a GU Ventures-based platform company at the University of Gothenburg. Dr Limousin reported having received travel support and honoraria from Boston Scientific and Medtronic. Dr Heywood reported having received funding and travel support from Shire Pharmaceuticals; and reported having received honoraria from Freeline Therapeutics. Dr Mills reported having received honoraria from Freeline Therapeutics; and having received travel support from Actelion and Genzyme Sanofi. Dr Libri reported having served on Advisory Boards for Biogen and Nova Laboratories Ltd. Dr Streeter reported being a consultant to Sanofi, Prevail, Inflazome, and Kyowa. No other disclosures were reported.

Figures

Figure 1.. Flow Diagram of Participant Recruitment…
Figure 1.. Flow Diagram of Participant Recruitment and Retention
CSF indicates cerebrospinal fluid; GBA1-, negative glucocerebrosidase gene; GBA1+, positive glucocerebrosidase gene; and LP, lumbar puncture.
Figure 2.. Biochemical Changes After Ambroxol Administration
Figure 2.. Biochemical Changes After Ambroxol Administration
A, Box plot with superimposed data points at baseline (median, 0.321; interquartile range [IQR], 0.142) and 186 days (median, 0.216; IQR, 0.221). Analysis included 17 participants (8 GBA1+ and 9 GBA1–). Mean (SE) change of 0.059 (0.026) nmol/mL per hour (95% CI, –0.115 to –0.002; 2-sided paired t test, P = .04) represents a 19% decrease. B, Box plot (median and IQR) with superimposed data points at baseline and 186 days. Analysis included 17 participants (8 GBA1+ and 9 GBA1–). Mean (SE) change of 50 (17) pg/mL (95% CI, 14-87; 2-sided paired t test, P = .01) represents a 13% increase. C, Box plot (median and IQR) with superimposed data points at baseline and 186 days. Analysis included 17 participants (8 GBA1+ and 9 GBA1–). Mean (SE) change of 88 (22) pmol/L (95% CI, 40-137; 2-sided paired t test, P = .002) represents a 35% increase. All data points are horizontally offset for ease of interpretation. D, Error bars indicate SE of the mean. Analysis included 18 participants (8 GBA1+ and 10 GBA1–). Mean (SE) change between baseline and 186 days of 1.0 (1.4) nmol/mg per hour (95% CI, –2.0 to 4.0; P = .48) represents a 9% increase. CSF indicates cerebrospinal fluid; GBA1-, negative glucocerebrosidase gene; GBA1+, positive glucocerebrosidase gene; and GCase, glucocerebrosidase enzyme.

References

    1. Grabowski GA, Zimran A, Ida H. Gaucher disease types 1 and 3. Am J Hematol. 2015;90(suppl 1):S12-S18.
    1. Sidransky E, Nalls MA, Aasly JO, et al. . Multicenter analysis of glucocerebrosidase mutations in Parkinson’s disease. N Engl J Med. 2009;361(17):1651-1661.
    1. Rana HQ, Balwani M, Bier L, Alcalay RN. Age-specific Parkinson disease risk in GBA mutation carriers. Genet Med. 2013;15(2):146-149.
    1. Anheim M, Elbaz A, Lesage S, et al. ; French Parkinson Disease Genetic Group . Penetrance of Parkinson disease in glucocerebrosidase gene mutation carriers. Neurology. 2012;78(6):417-420.
    1. Zhang Y, Shu L, Sun Q, et al. . Integrated genetic analysis of racial differences of common GBA variants in Parkinson’s disease: a meta-analysis. Front Mol Neurosci. 2018;11:43.
    1. Yang SY, Beavan M, Chau KY, Taanman JW, Schapira AHV. A human neural crest stem cell–derived dopaminergic neuronal model recapitulates biochemical abnormalities in GBA1 mutation carriers. Stem Cell Reports. 2017;8(3):728-742.
    1. Migdalska-Richards A, Daly L, Bezard E, Schapira AH. Ambroxol effects in glucocerebrosidase and α-synuclein transgenic mice. Ann Neurol. 2016;80(5):766-775.
    1. Migdalska-Richards A, Ko WKD, Li Q, Bezard E, Schapira AHV. Oral ambroxol increases brain glucocerebrosidase activity in a nonhuman primate. Synapse. 2017;71(7):e21967.
    1. Magalhaes J, Gegg ME, Migdalska-Richards A, Schapira AH. Effects of ambroxol on the autophagy-lysosome pathway and mitochondria in primary cortical neurons. Sci Rep. 2018;8(1):1385.
    1. Mazzulli JR, Zunke F, Tsunemi T, et al. . Activation of β-glucocerebrosidase reduces pathological α-synuclein and restores lysosomal function in Parkinson’s patient midbrain neurons. J Neurosci. 2016;36(29):7693-7706.
    1. McNeill A, Magalhaes J, Shen C, et al. . Ambroxol improves lysosomal biochemistry in glucocerebrosidase mutation-linked Parkinson disease cells. Brain. 2014;137(pt 5):1481-1495.
    1. Gegg ME, Burke D, Heales SJ, et al. . Glucocerebrosidase deficiency in substantia nigra of Parkinson disease brains. Ann Neurol. 2012;72(3):455-463.
    1. Murphy KE, Gysbers AM, Abbott SK, et al. . Reduced glucocerebrosidase is associated with increased α-synuclein in sporadic Parkinson’s disease. Brain. 2014;137(pt 3):834-848.
    1. Parnetti L, Chiasserini D, Persichetti E, et al. . Cerebrospinal fluid lysosomal enzymes and alpha-synuclein in Parkinson’s disease. Mov Disord. 2014;29(8):1019-1027.
    1. Sardi SP, Clarke J, Viel C, et al. . Augmenting CNS glucocerebrosidase activity as a therapeutic strategy for parkinsonism and other Gaucher-related synucleinopathies. Proc Natl Acad Sci U S A. 2013;110(9):3537-3542.
    1. Mullin S, Hughes D, Mehta A, Schapira AHV. Neurological effects of glucocerebrosidase gene mutations. Eur J Neurol. 2019;26(3):388-e29.
    1. Narita A, Shirai K, Itamura S, et al. . Ambroxol chaperone therapy for neuronopathic Gaucher disease. Ann Clin Transl Neurol. 2016;3(3):200-215.
    1. Maegawa GH, Tropak MB, Buttner JD, et al. . Identification and characterization of ambroxol as an enzyme enhancement agent for Gaucher disease. J Biol Chem. 2009;284(35):23502-23516.
    1. Bendikov-Bar I, Maor G, Filocamo M, Horowitz M. Ambroxol as a pharmacological chaperone for mutant glucocerebrosidase. Blood Cells Mol Dis. 2013;50(2):141-145.
    1. Luan Z, Li L, Higaki K, Nanba E, Suzuki Y, Ohno K. The chaperone activity and toxicity of ambroxol on Gaucher cells and normal mice. Brain Dev. 2013;35(4):317-322.
    1. Xilouri M, Brekk OR, Stefanis L. α-Synuclein and protein degradation systems: a reciprocal relationship. Mol Neurobiol. 2013;47(2):537-551.
    1. Yap TL, Gruschus JM, Velayati A, et al. . Alpha-synuclein interacts with glucocerebrosidase providing a molecular link between Parkinson and Gaucher diseases. J Biol Chem. 2011;286(32):28080-28088.
    1. Fois G, Hobi N, Felder E, et al. . A new role for an old drug. Cell Calcium. 2015;58(6):628-637.
    1. Maor G, Rencus-Lazar S, Filocamo M, Steller H, Segal D, Horowitz M. Unfolded protein response in Gaucher disease: from human to drosophila. Orphanet J Rare Dis. 2013;8:140.
    1. Babajani G, Tropak MB, Mahuran DJ, Kermode AR. Pharmacological chaperones facilitate the post-ER transport of recombinant N370S mutant β-glucocerebrosidase in plant cells. Mol Genet Metab. 2012;106(3):323-329.
    1. World Medical Association World Medical Association Declaration of Helsinki: ethical principles for medical research involving human subjects. JAMA. 2013;310(20):2191-2194.
    1. International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use ICH Harmonised Tripartite Guideline: Guideline for Good Clinical Practice E6 (R1). . Published June 10, 1996. Accessed December 5, 2019.
    1. Hughes AJ, Daniel SE, Kilford L, Lees AJ. Accuracy of clinical diagnosis of idiopathic Parkinson’s disease. J Neurol Neurosurg Psychiatry. 1992;55(3):181-184.
    1. Rocha EM, Smith GA, Park E, et al. . Sustained systemic glucocerebrosidase inhibition induces brain α-synuclein aggregation, microglia and complement C1q activation in mice. Antioxid Redox Signal. 2015;23(6):550-564.
    1. Schirinzi T, Sancesario GM, Di Lazzaro G, et al. . CSF α-synuclein inversely correlates with non-motor symptoms in a cohort of PD patients. Parkinsonism Relat Disord. 2019;61:203-206.

Source: PubMed

3
Abonnere