Gene therapy for aromatic L-amino acid decarboxylase deficiency by MR-guided direct delivery of AAV2-AADC to midbrain dopaminergic neurons

Toni S Pearson, Nalin Gupta, Waldy San Sebastian, Jill Imamura-Ching, Amy Viehoever, Ana Grijalvo-Perez, Alex J Fay, Neha Seth, Shannon M Lundy, Youngho Seo, Miguel Pampaloni, Keith Hyland, Erin Smith, Gardenia de Oliveira Barbosa, Jill C Heathcock, Amy Minnema, Russell Lonser, J Bradley Elder, Jeffrey Leonard, Paul Larson, Krystof S Bankiewicz, Toni S Pearson, Nalin Gupta, Waldy San Sebastian, Jill Imamura-Ching, Amy Viehoever, Ana Grijalvo-Perez, Alex J Fay, Neha Seth, Shannon M Lundy, Youngho Seo, Miguel Pampaloni, Keith Hyland, Erin Smith, Gardenia de Oliveira Barbosa, Jill C Heathcock, Amy Minnema, Russell Lonser, J Bradley Elder, Jeffrey Leonard, Paul Larson, Krystof S Bankiewicz

Abstract

Aromatic L-amino acid decarboxylase (AADC) deficiency is a rare genetic disorder characterized by deficient synthesis of dopamine and serotonin. It presents in early infancy, and causes severe developmental disability and lifelong motor, behavioral, and autonomic symptoms including oculogyric crises (OGC), sleep disorder, and mood disturbance. We investigated the safety and efficacy of delivery of a viral vector expressing AADC (AAV2-hAADC) to the midbrain in children with AADC deficiency (ClinicalTrials.gov Identifier NCT02852213). Seven (7) children, aged 4-9 years underwent convection-enhanced delivery (CED) of AAV2-hAADC to the bilateral substantia nigra (SN) and ventral tegmental area (VTA) (total infusion volume: 80 µL per hemisphere) in 2 dose cohorts: 1.3 × 1011 vg (n = 3), and 4.2 × 1011 vg (n = 4). Primary aims were to demonstrate the safety of the procedure and document biomarker evidence of restoration of brain AADC activity. Secondary aims were to assess clinical improvement in symptoms and motor function. Direct bilateral infusion of AAV2-hAADC was safe, well-tolerated and achieved target coverage of 98% and 70% of the SN and VTA, respectively. Dopamine metabolism was increased in all subjects and FDOPA uptake was enhanced within the midbrain and the striatum. OGC resolved completely in 6 of 7 subjects by Month 3 post-surgery. Twelve (12) months after surgery, 6/7 subjects gained normal head control and 4/7 could sit independently. At 18 months, 2 subjects could walk with 2-hand support. Both the primary and secondary endpoints of the study were met. Midbrain gene delivery in children with AADC deficiency is feasible and safe, and leads to clinical improvements in symptoms and motor function.

Conflict of interest statement

N.G. reports relationships with Oscine Therapeutics (consulting) and Y-mAbs Therapeutics (consulting). K.H. reports that he is employed by Medical Neurogenetics Laboratories, a company that provides commercial diagnostic testing for aromatic l-amino acid decarboxylase deficiency. P.L. reports relationships with Axovant (Advisory Board), Neurocrine Biosciences (research funding, consulting), UniQure (research funding), Voyager Therapeutics (research funding), and Clearpoint Neuro (consulting). K.S.B. is the founder and equity holder of Brain Neurotherapy Bio. The remaining authors have no competing interests to disclose.

© 2021. The Author(s).

Figures

Fig. 1. MR-guided delivery of AAV2-hAADC into…
Fig. 1. MR-guided delivery of AAV2-hAADC into the midbrain, baseline DaTscan and changes in FDOPA PET biomarker after gene delivery.
Coronal (A) and axial (C) MR images at the conclusion of the vector infusions into SN and VTA regions (white arrows). Bright signal corresponds to gadoteridol admixed with AAV2-hAADC. Infusions are performed sequentially while imaging, starting with right SN. Please, note accurate targeting and coverage in respective anatomical regions. B Coverage (volume of distribution, Vd) of all infusions performed into the SN and VTA in 7 subjects (n = 2 independent infusion sites (left and right) examined per participant (n = 7) for each target structure (SNc and VTA), for a total of n = 14 independent infusions per target structure). SN infusion (50 µL) achieved coverage of ~160 ± 60 mm3 (mean ± SD, n = 14 infusions (two infusions per participant)) with one suboptimal infusion due to leakage along the perivascular space. VTA infusion (30 µL) resulted in coverage of 103 ± 22 mm3. Coverage volume of gadoteridol (Vd) suggests almost 80% anatomical coverage of both SN and VTA in all subjects (except single SN in one patient). D DaTscan imaging of the striatum at baseline confirmed a normal pattern of dopaminergic innervation in all study subjects, indicative of preserved nigrostriatal pathway. E, F Baseline FDOPA imaging of the midbrain regions (SN and VTA, black arrows in E) and nigrostriatal projection (caudate nucleus and putamen, dotted line in F). Lack of signal in both regions represents impaired conversion of FDOPA to F-dopamine due to absent AADC activity. G, H Increased FDOPA PET uptake 3 months after AAV2-hAADC administration in the midbrain and striatum, respectively. Images for Subject 4 are shown as representative of the group; see Supplementary Fig. S1 for images for each individual subject. Source data are provided as a Source Data file.
Fig. 2. CSF Neurotransmitter metabolites.
Fig. 2. CSF Neurotransmitter metabolites.
Concentrations of CSF metabolites measured at 2 separate baseline (BL) timepoints, Month 3, Month 6, and Month 12, in individual subjects in Cohort 1 (low-dose, black markers); and Cohort 2 (high-dose, white markers) and summarized at each timepoint as the group median (bars). A Homovanillic acid (HVA), the dopamine metabolite, was significantly higher at each post-operative timepoint compared to the baseline mean (inverted bracket; (*p = 0.0078 at Months 3 and 6, p = 0.0313 at Month 12, one-tailed Wilcoxon signed-rank test). Lower limit of normal range: 218 nmol/L (dotted line). B 5-hydroxyindoleacetic acid (5-HIAA), the serotonin metabolite, did not change after gene delivery. Lower limit of normal range: 66 nmol/L (dotted line). C 3-O-methyldopa (3-OMD) was elevated in all subjects at all timepoints. Normal: <100 nmol/L (dotted line). Source data can be found in Table 2.
Fig. 3. Changes in Oculogyric Crises (OGC)…
Fig. 3. Changes in Oculogyric Crises (OGC) and motor function after gene delivery.
A The OGC score was calculated monthly, and represents the weekly average of the duration of episodes (hours per week), weighted by severity (grade 1–3: 1 = mild/eye deviation only; 2 = moderate/eye deviation + dystonia or dyskinesia of the face and/or neck; 3 = severe/dystonia or dyskinesia involving the trunk and/or limbs). By Month 3, OGCs completely resolved in 6/7 subjects. Inset (black arrow): Subject 1 had residual episodes throughout the 24-month study period; the severity decreased after surgery (red arrow). B Gross Motor Function Measure-88 (GMFM-88) scores for Cohorts 1 (red) and 2 (blue). Data are shown through Month 24 for Subjects 1–3, Month 18 for Subjects 4 and 5, and Month 6 for Subjects 6 and 7. Baseline GMFM scores for all subjects (circled) were ≤10, consistent with severe motor impairment. Inset: changes in GMFM score between Baseline and Month 12. An increase of ≥7 points (dotted line, representing a clinically meaningful positive change) was observed in 6/7 subjects by Month 12. Source data are provided as a Source Data file.
Fig. 4. Non-motor symptoms before and after…
Fig. 4. Non-motor symptoms before and after gene delivery.
A The percentage of subjects (n = 7) who, by caregiver report, experienced irritability at baseline compared to the last available follow-up timepoint (Month 24 for Subjects 1–3, Month 18 for Subjects 4 and 5, and Month 6 for Subjects 6 and 7). B The number of hours or irritability per day recorded in caregiver diaries increased slightly immediately after surgery, and then improved on average (mean ± SE). C Prevalence and severity of insomnia before and after gene delivery by caregiver report. D The mean number of hours (±SE) of night-time sleep recorded in caregiver diaries improved slightly across subjects after gene delivery. For Subject 2 (blue triangles), the improvement was dramatic. EG Prevalence and severity of other non-motor symptoms before and after gene delivery by caregiver report (see details in A, above). Major (black): frequent and/or severe, with significant impact on comfort or function; Minor (gray): infrequent and/or mild. Source data are provided as a Source Data file.
Fig. 5. Time course of dyskinesia after…
Fig. 5. Time course of dyskinesia after gene transfer.
Dyskinesia score, adapted from the Abnormal Involuntary Movement Scale (AIMS). Involuntary movements in each of 7 body regions were scored from 0 (none) to 4 (severe), for a maximum possible score of 28. A total score of 14 (dotted line) may correspond with either mild generalized dyskinesia, or moderate-severe involuntary movements in a more focal distribution. Dyskinesia peaked in severity between 1 and 3 months after surgery, and gradually improved thereafter. At Month 24, dyskinesia had resolved completely in all 3 subjects in Cohort 1.

References

    1. Hyland K, Clayton PT. Aromatic amino acid decarboxylase deficiency in twins. J. Inherit. Metab. Dis. 1990;13:301–304. doi: 10.1007/BF01799380.
    1. Wassenberg T, et al. Consensus guideline for the diagnosis and treatment of aromatic l-amino acid decarboxylase (AADC) deficiency. Orphanet J. Rare Dis. 2017;12:12. doi: 10.1186/s13023-016-0522-z.
    1. Brun L, et al. Clinical and biochemical features of aromatic L-amino acid decarboxylase deficiency. Neurology. 2010;75:64–71. doi: 10.1212/WNL.0b013e3181e620ae.
    1. Pearson, T. S. et al. AADC deficiency from infancy to adulthood: symptoms and developmental outcome in an international cohort of 63 patients. J. Inherit. Metab. Dis. (2020) 10.1002/jimd.12247.
    1. Hwu, W. L., Chien, Y. H., Lee, N. C. & Li, M. H. Natural history of aromatic L-amino acid decarboxylase deficiency in Taiwan. JIMD Rep. (2017) 10.1007/8904_2017_54.
    1. Limousin P, Foltynie T. Long-term outcomes of deep brain stimulation in Parkinson disease. Nat. Rev. Neurol. 2019;15:234–242. doi: 10.1038/s41582-019-0145-9.
    1. Parmar M, Grealish S, Henchcliffe C. The future of stem cell therapies for Parkinson disease. Nat. Rev. Neurosci. 2020;21:103–115. doi: 10.1038/s41583-019-0257-7.
    1. Hitti FL, Yang AI, Gonzalez-Alegre P, Baltuch GH. Human gene therapy approaches for the treatment of Parkinson’s disease: an overview of current and completed clinical trials. Parkinsonism Relat. Disord. 2019;66:16–24. doi: 10.1016/j.parkreldis.2019.07.018.
    1. Kordower JH, et al. Delivery of neurturin by AAV2 (CERE-120)-mediated gene transfer provides structural and functional neuroprotection and neurorestoration in MPTP-treated monkeys. Ann. Neurol. 2006;60:706–715. doi: 10.1002/ana.21032.
    1. Heiss JD, et al. Trial of magnetic resonance-guided putaminal gene therapy for advanced Parkinson’s disease. Mov. Disord. 2019;34:1073–1078. doi: 10.1002/mds.27724.
    1. Christine CW, et al. Safety and tolerability of putaminal AADC gene therapy for Parkinson disease. Neurology. 2009;73:1662–1669. doi: 10.1212/WNL.0b013e3181c29356.
    1. Christine CW, et al. Magnetic resonance imaging-guided phase 1 trial of putaminal AADC gene therapy for Parkinson’s disease. Ann. Neurol. 2019;85:704–714. doi: 10.1002/ana.25450.
    1. Bankiewicz KS, 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. doi: 10.1006/exnr.2000.7408.
    1. Lee WT, Weng WC, Peng SF, Tzen KY. Neuroimaging findings in children with paediatric neurotransmitter diseases. J. Inherit. Metab. Dis. 2009;32:361–370. doi: 10.1007/s10545-009-1106-z.
    1. Hwu WL, et al. Gene therapy for aromatic L-amino acid decarboxylase deficiency. Sci. Transl. Med. 2012;4:134ra61. doi: 10.1126/scitranslmed.3003640.
    1. Chien Y-H, et al. Efficacy and safety of AAV2 gene therapy in children with aromatic L-amino acid decarboxylase deficiency: an open-label, phase 1/2 trial. Lancet Child Adolesc. Health. 2017;1:265–273. doi: 10.1016/S2352-4642(17)30125-6.
    1. Kojima K, et al. Gene therapy improves motor and mental function of aromatic l-amino acid decarboxylase deficiency. Brain J. Neurol. 2019;142:322–333. doi: 10.1093/brain/awy331.
    1. Kells AP, et al. Efficient gene therapy-based method for the delivery of therapeutics to primate cortex. Proc. Natl Acad. Sci. USA. 2009;106:2407–2411. doi: 10.1073/pnas.0810682106.
    1. San Sebastian, W. et al. Safety and tolerability of MRI-guided infusion of AAV2-hAADC into the mid-brain of non-human primate. Mol. Ther. Methods Clin. Dev. 3, 14049 (2014).
    1. Richardson RM, et al. Interventional MRI-guided putaminal delivery of AAV2-GDNF for a planned clinical trial in Parkinson’s disease. Mol. Ther. 2011;19:1048–1057. doi: 10.1038/mt.2011.11.
    1. Richardson RM, 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. doi: 10.1159/000323544.
    1. Richardson RM, et al. T2 imaging in monitoring of intraparenchymal real-time convection-enhanced delivery. Neurosurgery. 2011;69:154–163. doi: 10.1227/NEU.0b013e318217217e.
    1. Su X, et al. Real-time MR imaging with Gadoteridol predicts distribution of transgenes after convection-enhanced delivery of AAV2 vectors. Mol. Ther. J. Am. Soc. Gene Ther. 2010;18:1490–1495. doi: 10.1038/mt.2010.114.
    1. Yin D, et al. Cannula placement for effective convection-enhanced delivery in the nonhuman primate thalamus and brainstem: implications for clinical delivery of therapeutics. J. Neurosurg. 2010;113:240–248. doi: 10.3171/2010.2.JNS091744.
    1. Russell DJ, et al. The gross motor function measure: a means to evaluate the effects of physical therapy. Dev. Med. Child Neurol. 1989;31:341–352. doi: 10.1111/j.1469-8749.1989.tb04003.x.
    1. Matsuda W, et al. Single nigrostriatal dopaminergic neurons form widely spread and highly dense axonal arborizations in the neostriatum. J. Neurosci. J. Soc. Neurosci. 2009;29:444–453. doi: 10.1523/JNEUROSCI.4029-08.2009.
    1. Muramatsu S, et al. A phase I study of aromatic L-amino acid decarboxylase gene therapy for Parkinson’s disease. Mol. Ther. J. Am. Soc. Gene Ther. 2010;18:1731–1735. doi: 10.1038/mt.2010.135.
    1. Hadaczek P, et al. Eight years of clinical improvement in MPTP-lesioned primates after gene therapy with AAV2-hAADC. Mol. Ther. J. Am. Soc. Gene Ther. 2010;18:1458–1461. doi: 10.1038/mt.2010.106.
    1. Sehara Y, 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. doi: 10.1089/humc.2017.010.
    1. Mittermeyer G, 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. doi: 10.1089/hum.2011.220.
    1. Sparrow, S., Cicchetti, D. & Balla, D. Vineland Adaptive Behavior Scales, Survey Interview Form/Caregiver Rating Form (Pearson Assessments, 2005).
    1. Hyland K, et al. Cerebrospinal fluid concentrations of pterins and metabolites of serotonin and dopamine in a pediatric reference population. Pediatr. Res. 1993;34:10–14. doi: 10.1203/00006450-199307000-00003.
    1. Hyland K, Clayton PT. Aromatic L-amino acid decarboxylase deficiency: diagnostic methodology. Clin. Chem. 1992;38:2405–2410. doi: 10.1093/clinchem/38.12.2405.

Source: PubMed

3
Se inscrever