Magnetic resonance imaging-guided phase 1 trial of putaminal AADC gene therapy for Parkinson's disease

Chadwick W Christine, Krystof S Bankiewicz, Amber D Van Laar, R Mark Richardson, Bernard Ravina, Adrian P Kells, Brendon Boot, Alastair J Martin, John Nutt, Marin E Thompson, Paul S Larson, Chadwick W Christine, Krystof S Bankiewicz, Amber D Van Laar, R Mark Richardson, Bernard Ravina, Adrian P Kells, Brendon Boot, Alastair J Martin, John Nutt, Marin E Thompson, Paul S Larson

Abstract

Objective: To understand the safety, putaminal coverage, and enzyme expression of adeno-associated viral vector serotype-2 encoding the complementary DNA for the enzyme, aromatic L-amino acid decarboxylase (VY-AADC01), delivered using novel intraoperative monitoring to optimize delivery.

Methods: Fifteen subjects (three cohorts of 5) with moderately advanced Parkinson's disease and medically refractory motor fluctuations received VY-AADC01 bilaterally coadministered with gadoteridol to the putamen using intraoperative magnetic resonance imaging (MRI) guidance to visualize the anatomic spread of the infusate and calculate coverage. Cohort 1 received 8.3 × 1011 vg/ml and ≤450 μl per putamen (total dose, ≤7.5 × 1011 vg); cohort 2 received the same concentration (8.3 × 1011 vg/ml) and ≤900 μl per putamen (total dose, ≤1.5 × 1012 vg); and cohort 3 received 2.6 × 1012 vg/ml and ≤900 μl per putamen (total dose, ≤4.7 × 1012 vg). (18)F-fluoro-L-dihydroxyphenylalanine positron emission tomography (PET) at baseline and 6 months postprocedure assessed enzyme activity; standard assessments measured clinical outcomes.

Results: MRI-guided administration of ascending VY-AADC01 doses resulted in putaminal coverage of 21% (cohort 1), 34% (cohort 2), and 42% (cohort 3). Cohorts 1, 2, and 3 showed corresponding increases in enzyme activity assessed by PET of 13%, 56%, and 79%, and reductions in antiparkinsonian medication of -15%, -33%, and -42%, respectively, at 6 months. At 12 months, there were dose-related improvements in clinical outcomes, including increases in patient-reported ON-time without troublesome dyskinesia (1.6, 3.3, and 1.5 hours, respectively) and quality of life.

Interpretation: Novel intraoperative monitoring of administration facilitated targeted delivery of VY-AADC01 in this phase 1 study, which was well tolerated. Increases in enzyme expression and clinical improvements were dose dependent. ClinicalTrials.gov Identifier: NCT01973543 Ann Neurol 2019;85:704-714.

Conflict of interest statement

The authors report the following potential conflicts of interest during the conduct of this study: C.W.C. and J.N. received grants from Voyager Therapeutics, Inc.; K.S.B. received grants and personal fees from Voyager Therapeutics and holds a patent on the use of AAV2‐AADC for the treatment of Parkinson's disease licensed to Genzyme and Voyager Therapeutics, and a patent on the step cannula for gene delivery with royalties paid by MRI Interventions to UCSF; A.D.V.L., R.M.R., and A.J.M. received grants from Voyager Therapeutics; B.R. was an employee of Voyager Therapeutics and has a patent on AADC gene transfer in Parkinson's disease pending; A.P.K. was an employee of Voyager Therapeutics; B.B. was an employee of Voyager Therapeutics; M.E.T. received grants and nonfinancial support from Voyager Therapeutics; and P.S.L. received grants from Voyager Therapeutics and nonfinancial support from MRI Interventions, the company that makes the surgical delivery platform and step cannula used for gene delivery.

© 2019 The Authors. Annals of Neurology published by Wiley Periodicals, Inc. on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
Representative MP‐RAGE images during simultaneous bilateral administration of VY‐AADC01 in the putamen. Axial image (left) shows gadoteridol enhancement as a surrogate marker for vector distribution with two administrations in the posterior portion of each putamen. Coronal oblique image (right) in‐plane with the right infusion cannula during vector delivery; part of the skull mounted SmartFrame aiming device is visible above the burr hole (white arrow), as is the SmartFlow cannula (black arrow). MP‐RAGE = magnetization‐prepared rapid gradient echo.
Figure 2
Figure 2
Percentage increase from baseline in 18F‐dopa PET signal of AADC activity and corresponding reductions in levodopa equivalent dose at 6 months. The top of the figure shows the increase in 18F‐dopa PET assessment of AADC activity by cohort with increasing administration volume in cohort 2 and the same volume with an increase in genome concentration in cohort 3. The bottom of the figure shows corresponding reductions in levodopa equivalent dosing. LED = levodopa equivalent dose; PET = positron emission tomography.
Figure 3
Figure 3
Mean (±SE) change from baseline in UPDRS‐III assessed in the on‐ (A) and off‐medication (B) states. Change in UPDRS‐III scores in the on‐ (A) and off‐medication (B) states. Mean score reductions (improvements) were observed in cohorts 2 and 3 on medication and all three cohorts off medication through last follow‐up. Note that UPDRS data were not collected at month 18 for cohorts 1 and 2. UPDRS = Unified Parkinson's Disease Rating Scale.
Figure 4
Figure 4
Time‐action curve for UPDRS‐III for threshold and suprathreshold infusions of intravenous levodopa at baseline and approximately 6 months after administration of VY‐AADC01. The figure shows the UPDRS‐III time‐action curves following a threshold (0.6 mg/kg/h; left side) and suprathreshold (1.2 mg/kg/h; right side) intravenous dose of levodopa by cohort (A,B: cohort 1; C,D: cohort 2; E,F: cohort 3). Lower scores indicate improvement. Dashed lines in each figure show data before VY‐AADC01 administration and solid lines postadministration. The gray box shows the actual infusion time. Note in cohort 2 the time‐action curve for threshold (low‐dose) intravenous levodopa post‐VY‐AADC01 administration (C) appears similar to the suprathreshold (high‐dose) intravenous levodopa pre‐VY‐AADC01 administration (D). AUC = area under the curve; UPDRS = Unified Parkinson's Disease Rating Scale.
Figure 5
Figure 5
Mean diary ON and OFF time per 16‐hour waking day. Across cohorts, good quality ON time (green bars; ON time with no dyskinesia plus ON time with nontroublesome dyskinesia) increased over time, and both OFF time and ON time with troublesome dyskinesia decreased over time. NTD = nontroublesome dyskinesia; TD = troublesome dyskinesia.

References

    1. Fahn S, Oakes D, Shoulson I, et al. Levodopa and the progression of Parkinson's disease. N Engl J Med 2004;351:2498–2508.
    1. Kordower JH, Olanow CW, Dodiya HB, et al. Disease duration and the integrity of the nigrostriatal system in Parkinson's disease. Brain 2013;136(pt 8):2419–2431.
    1. Ciesielska A, Samaranch L, San Sebastian W, et al. Depletion of AADC activity in caudate nucleus and putamen of Parkinson's disease patients: implications for ongoing AAV2‐AADC gene therapy trial. PLoS One 2017;12:e0169965.
    1. Chan PL, Nutt JG, Holford NH. Pharmacokinetic and pharmacodynamic changes during the first four years of levodopa treatment in Parkinson's disease. J Pharmacokinet Pharmacodyn 2005;32:459–484.
    1. Gray R, Ives N, Rick C, et al. Long‐term effectiveness of dopamine agonists and monoamine oxidase B inhibitors compared with levodopa as initial treatment for Parkinson's disease (PD MED): a large, open‐label, pragmatic randomised trial. Lancet 2014;384:1196–1205.
    1. Voon V, Napier TC, Frank MJ, et al. Impulse control disorders and levodopa‐induced dyskinesias in Parkinson's disease: an update. Lancet Neurol 2017;16:238–250.
    1. Bankiewicz KS, Eberling JL, Kohutnicka M, et al. Convection‐enhanced delivery of AAV vector in parkinsonian monkeys: in vivo detection of gene expression and restoration of dopaminergic function using pro‐drug approach. Exp Neurol 2000;164:2–14.
    1. Sánchez‐Pernaute R, Harvey‐White J, Cunningham J, Bankiewicz KS. Functional effect of adeno‐associated virus mediated gene transfer of aromatic L‐amino acid decarboxylase into the striatum of 6‐OHDA‐lesioned rats. Mol Ther 2001;4:324–330.
    1. Su X, Kells AP, Salegio EA, et al. Real‐time MR imaging with Gadoteridol predicts distribution of transgenes after convection‐enhanced delivery of AAV2 vectors. Mol Ther 2010;18:1490–1495.
    1. 10.Richardson RM, Kells AP, Martin AJ, et al. Novel platform for MRI‐guided convection‐enhanced delivery of therapeutics: preclinical validation in nonhuman primate brain. Stereotact Funct Neurosurg 2011;89:141–151.
    1. Hadaczek P, Eberling JL, Pivirotto P, et al. Eight years of clinical improvement in MPTP‐lesioned primates after gene therapy with AAV2‐hAADC. Mol Ther 2010;18:1458–1461.
    1. Christine CW, Starr PA, Larson PS, et al. Safety and tolerability of putaminal AADC gene therapy for Parkinson disease. Neurology 2009;73:1662–1669.
    1. Muramatsu S, Fujimoto K, Kato S, et al. A phase I study of aromatic L‐amino acid decarboxylase gene therapy for Parkinson's disease. Mol Ther 2010;18:1731–1735.
    1. Dhawan V, Ma Y, Pillai V, et al. Comparative analysis of striatal FDOPA uptake in Parkinson's disease: ratio method versus graphical approach. J Nucl Med 2002;43:1324–1330.
    1. Hauser RA, Friedlander J, Zesiewicz TA, et al. A home diary to assess functional status in patients with Parkinson's disease with motor fluctuations and dyskinesia. Clin Neuropharmacol 2000;23:75–81.
    1. Tomlinson CL, Stowe R, Patel S, et al. Systematic review of levodopa dose equivalency reporting in Parkinson's disease. Mov Disord 2010;25:2649–2653.
    1. Espay AJ, Pagan FL, Walter BL, et al. Optimizing extended‐release carbidopa/levodopa in Parkinson disease: consensus on conversion from standard therapy. Neurol Clin Pract 2017;7:86–93.
    1. Brodsky MA, Park BS, Nutt JG. Effects of a dopamine agonist on the pharmacodynamics of levodopa in Parkinson disease. Arch Neurol 2010;67:27–32.
    1. Horváth K, Aschermann Z, Kovács M, et al. Changes in quality of life in Parkinson's disease: how large must they be to be relevant? Neuroepidemiology 2017;48:1–8.
    1. Bartus RT, Weinberg MS, Samulski RJ. Parkinson's disease gene therapy: success by design meets failure by efficacy. Mol Ther 2014;22:487–497.
    1. Forsayeth J, Bankiewicz KS. Transduction of antigen‐presenting cells in the brain by AAV9 warrants caution in preclinical studies. Mol Ther 2015;23:612.
    1. Shulman LM, Gruber‐Baldini AL, Anderson KE, et al. The clinically important difference on the Unified Parkinson's Disease Rating Scale. Arch Neurol 2010;67:64–70.
    1. Nutt JG. On‐off phenomenon: relation to levodopa pharmacokinetics and pharmacodynamics. Ann Neurol 1987;22:535–540.
    1. Forsayeth JR, Eberling JL, Sanftner LM, et al. A dose‐ranging study of AAV‐hAADC therapy in Parkinsonian monkeys. Mol Ther 2006;14:571–577.
    1. Bankiewicz KS, Forsayeth J, Eberling JL, et al. Long‐term clinical improvement in MPTP‐lesioned primates after gene therapy with AAV‐hAADC. Mol Ther 2006;14:564–570.
    1. Mittermeyer G, Christine CW, Rosenbluth KH, et al. Long‐term evaluation of a phase 1 study of AADC gene therapy for Parkinson's disease. Hum Gene Ther 2012;23:377–381.
    1. Sehara Y, Fujimoto KI, Ikeguchi K, et al. Persistent expression of dopamine‐synthesizing enzymes 15 years after gene transfer in a primate model of Parkinson's disease. Hum Gene Ther Clin Dev 2017;28:74–79.
    1. Lidstone SC. Great expectations: the placebo effect in Parkinson's disease. Handb Exp Pharmacol 2014;225:139–147.

Source: PubMed

3
S'abonner