Intravenous arylsulfatase A in metachromatic leukodystrophy: a phase 1/2 study

Christine Í Dali, Samuel Groeschel, Mihai Moldovan, Mohamed H Farah, Ingeborg Krägeloh-Mann, Margaret Wasilewski, Jing Li, Norman Barton, Christian Krarup, Christine Í Dali, Samuel Groeschel, Mihai Moldovan, Mohamed H Farah, Ingeborg Krägeloh-Mann, Margaret Wasilewski, Jing Li, Norman Barton, Christian Krarup

Abstract

Objective: Metachromatic leukodystrophy (MLD) is an autosomal recessive lysosomal storage disease caused by deficient activity of arylsulfatase A (ASA), resulting in severe motor and cognitive dysfunction. This phase 1/2 study evaluated the safety and efficacy of intravenous (IV) recombinant human ASA (rhASA; HGT-1111, previously known as Metazym) in children with MLD.

Methods: Thirteen children with MLD (symptom onset < 4 years of age) were enrolled in an open-label, nonrandomized, dose-escalation trial and received IV rhASA at 50, 100, or 200 U/kg body weight every 14 (± 4) days for 52 weeks (NCT00418561; NCT00633139). Eleven children continued to receive rhASA at 100 or 200 U/kg during a 24-month extension period (NCT00681811). Outcome measures included safety observations, changes in motor and cognitive function, and changes in nerve conduction and morphometry.

Results: There were no serious adverse events considered related to IV rhASA. Motor function and developmental testing scores declined during the study in all dose groups; no significant differences were observed between groups. Nerve conduction studies and morphometric analysis indicated that peripheral nerve pathology did not worsen during the study in any dose group.

Interpretation: IV rhASA was generally well tolerated. There was no evidence of efficacy in preventing motor and cognitive deterioration, suggesting that IV rhASA may not cross the blood-brain barrier in therapeutic quantities. The relative stability of peripheral nerve function during the study indicates that rhASA may be beneficial if delivered to the appropriate target site and supports the development of rhASA for intrathecal administration in MLD.

Conflict of interest statement

CíD reports personal fees from University Hospital Copenhagen Rigshospitalet during the conduct of the study. MM has nothing to disclose. MHF has nothing to disclose. IKM reports grants from Shire (a Takeda company) during the conduct of the study. MW reports fees for consulting services from ID Remedies LLC. In addition, Dr Wasilewski has a patent pending: Wasilewski, Margaret, Wijatyk, Anna; c/o Shire Human Genetic Therapies, Inc., 300 Shire Way, Lexington, MA 02421 (US) PCT/US20 17/0 18440, Methods and Compositions for CNS Delivery of Arylsulfatase A. 17 February 2017. She was also an employee of Shire (a Takeda company) from July 2015 to August 2018 and is a Takeda stock owner. JL is an employee of Shire (a Takeda company). NB is an employee of Shire (a Takeda company). SG reports an institutional research grant from Shire (a Takeda company) outside of the submitted work. He serves as an advisor for trials in metachromatic leukodystrophy (Shire [a Takeda company], Orchard, Bioclinica, Homology Medicine) but receives no personal payment related to this role. CK reports grants and personal fees from Shire (a Takeda company) and grants from Danish Medical Research during the conduct of the study.

© 2020 The Authors. Annals of Clinical and Translational Neurology published by Wiley Periodicals LLC on behalf of American Neurological Association.

Figures

Figure 1
Figure 1
Study design and patient characteristics. aMedian age at baseline (the start of Study‐048).
Figure 2
Figure 2
Sulfatide measurements, lysosulfatide measurements, and light microscopic measurements of the right sural nerve at baseline and the left sural nerve at 26 weeks. (A) Sulfatide concentrations in all patients with available data (n = 12 at baseline; n = 11 at 26 weeks). (B) Lysosulfatide concentrations in all patients with available data (n = 11 at baseline and 26 weeks). (C) Total number of large fibers> 7 µm in diameter (cross‐section of whole nerve) in all patients with available data (n = 12 at baseline; n = 11 at 26 weeks). (D) Total number of small fibers < 7 µm in diameter (cross‐section of whole nerve) in all patients with available data (n = 12 at baseline; n = 11 at 26 weeks). (E) Examples of transverse light microscopic sections of the sural nerves in two patients. (F) Fiber diameter distributions for sural nerves from the patients in (E). The external diameters of fibers were traced (as shown in E) and the diameter of the fiber was calculated from the area enclosed by the trace. The diameters were compared using paired nonparametric tests.
Figure 3
Figure 3
Sensory nerve conduction studies of the right and left sural nerves at baseline and the left sural nerve at 10 and 26 weeks. (A) SNAP amplitudes in all patients with available data (n = 12 at baseline; n = 11 at 10 weeks and 26 weeks). Values measured as zero are plotted at 0.01 to enable visualization on the logarithmic scale. (B) SNCVs in all patients with available data (n = 11 at baseline and 10 weeks; n = 10 at 26 weeks). (C) Example SNAP recordings in two patients, one with less severe disease and one with severe disease. The values above the traces refer to SNAP amplitudes (μV) and SNCVs (m/s). Comparisons of the parameters were carried out using nonparametric tests. L, left; R, right; SNAP, sensory nerve action potential; SNCV, sensory nerve conduction velocity.
Figure 4
Figure 4
Motor nerve conduction studies of the right median nerve at baseline and 10, 26, 52, and 78 weeks. (A) CMAP amplitudes in the APB in all patients with available data (n = 13 at baseline; n = 12 at 10 weeks and 26 weeks; n = 11 at 52 weeks; n = 9 at 78 weeks). (B) DMLs in the APB in all patients with available data (n = 13 at baseline; n = 12 at 10 weeks and 26 weeks; n = 11 at 52 weeks; n = 9 at 78 weeks). (C) MNCVs of the median nerve in all patients with available data (n = 13 at baseline; n = 12 at 10 weeks and 26 weeks; n = 11 at 52 weeks; n = 9 at 78 weeks). (D) Example recordings of median nerve action potentials in an individual patient. The values above the traces refer to CMAP amplitudes (mV), DMLs (ms), and MNCVs (m/s). Comparisons of the parameters were carried out using nonparametric tests. APB, abductor pollicis brevis; CMAP, compound muscle action potential; DML, distal motor latency; MNCV, motor nerve conduction velocity.
Figure 5
Figure 5
Individual MRI total scores and MRS‐derived NAA values from baseline to 52 weeks by patient age. (A) MRI total scores. (B) MRS‐derived NAA values from the occipital cortex. (C) MRS‐derived NAA values from white matter, parieto‐occipital area. (D) MRS‐derived NAA values from white matter, frontal area. IV, intravenous; MRI, magnetic resonance imaging; MRS, magnetic resonance spectroscopy; NAA, N‐acetylaspartate; rhASA, recombinant human arylsulfatase A.

References

    1. Gieselmann V. Metachromatic leukodystrophy: genetics, pathogenesis and therapeutic options. Acta Paediatr Suppl 2008;97:15–21.
    1. Gieselmann V, Krägeloh‐Mann I. Metachromatic leukodystrophy – an update. Neuropediatrics 2010;41:1–6.
    1. Gieselmann V, Krägeloh‐Mann I, et al. Chapter 148: Metachromatic Leukodystrophy In: Valle D, Beaudet AL, Vogelstein B, eds. The Online Metabolic and Molecular Bases of Inherited Disease. New York: McGraw‐Hill, 2014.
    1. van Rappard DF, Boelens JJ, Wolf NI. Metachromatic leukodystrophy: disease spectrum and approaches for treatment. Best Pract Res Clin Endocrinol Metab 2015;29:261–273.
    1. Cesani M, Lorioli L, Grossi S, et al. Mutation update of ARSA and PSAP genes causing metachromatic leukodystrophy. Hum Mutat 2016;37:16–27.
    1. Kehrer C, Blumenstock G, Gieselmann V, Krägeloh‐Mann I. The natural course of gross motor deterioration in metachromatic leukodystrophy. Dev Med Child Neurol 2011;53:850–855.
    1. Kehrer C, Groeschel S, Kustermann‐Kuhn B, et al. Language and cognition in children with metachromatic leukodystrophy: onset and natural course in a nationwide cohort. Orphanet J Rare Dis 2014;9:18.
    1. Harrington M, Whalley D, Twiss J, et al. Insights into the natural history of metachromatic leukodystrophy from interviews with caregivers. Orphanet J Rare Dis 2019;14:89.
    1. Groeschel S, Kuhl JS, Bley AE, et al. Long‐term Outcome of Allogeneic hematopoietic stem cell transplantation in patients with juvenile metachromatic leukodystrophy compared with nontransplanted control patients. JAMA Neurol 2016;73:1133–1140.
    1. van Rappard DF, Boelens JJ, van Egmond ME, et al. Efficacy of hematopoietic cell transplantation in metachromatic leukodystrophy: the Dutch experience. Blood 2016;127:3098–3101.
    1. Biffi A, Montini E, Lorioli L, et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science 2013;341:1233158.
    1. Sessa M, Lorioli L, Fumagalli F, et al. Lentiviral haemopoietic stem‐cell gene therapy in early‐onset metachromatic leukodystrophy: an ad‐hoc analysis of a non‐randomised, open‐label, phase 1/2 trial. Lancet 2016;388:476–487.
    1. Baldo BA. Enzymes approved for human therapy: indications, mechanisms and adverse effects. BioDrugs 2015;29:31–55.
    1. Giugliani R, Federhen A, Vairo F, et al. Emerging drugs for the treatment of mucopolysaccharidoses. Expert Opin Emerg Drugs 2016;21:9–26.
    1. Su K, Donaldson E, Sharma R. Novel treatment options for lysosomal acid lipase deficiency: critical appraisal of sebelipase alfa. Appl Clin Genet 2016;9:157–167.
    1. Li M. Enzyme replacement therapy: a review and its role in treating lysosomal storage diseases. Pediatr Ann 2018;47:e191–e197.
    1. Whiteman DA, Kimura A. Development of idursulfase therapy for mucopolysaccharidosis type II (Hunter syndrome): the past, the present and the future. Drug Des Devel Ther 2017;11:2467–2480.
    1. Begley DJ, Pontikis CC, Scarpa M. Lysosomal storage diseases and the blood‐brain barrier. Curr Pharm Des 2008;14:1566–1580.
    1. Matzner U, Lullmann‐Rauch R, Stroobants S, et al. Enzyme replacement improves ataxic gait and central nervous system histopathology in a mouse model of metachromatic leukodystrophy. Mol Ther 2009;17:600–606.
    1. Matthes F, Stroobants S, Gerlach D, et al. Efficacy of enzyme replacement therapy in an aggravated mouse model of metachromatic leukodystrophy declines with age. Hum Mol Genet 2012;21:2599–2609.
    1. í Dali C, Barton NW, Farah MH, et al. Sulfatide levels correlate with severity of neuropathy in metachromatic leukodystrophy. Ann Clin Transl Neurol 2015;2:518–533.
    1. í Dali C, Hanson LG, Barton NW, et al. Brain N‐acetylaspartate levels correlate with motor function in metachromatic leukodystrophy. Neurology 2010;75:1896–1903.
    1. Russell DJ, Rosenbaum PL, Avery LM, Lane M. Gross Motor Function Measure (GMFM‐66 and GMFM‐88) User's Manual: Mac Keith Press. London UK: 2002.
    1. Rosenbaum PL, Palisano RJ, Bartlett DJ, et al. Development of the Gross Motor Function Classification System for cerebral palsy. Dev Med Child Neurol 2008;50:249–253.
    1. Russell DJ, Rosenbaum PL, Cadman DT, et al. The gross motor function measure: a means to evaluate the effects of physical therapy. Dev Med Child Neurol 1989;31:341–352.
    1. Mullen EM. Mullen Scales of Early Learning Manual: American Guidance Service. Circle Pines, MN, 1995.
    1. Farmer C, Golden C, Thurm A. Concurrent validity of the differential ability scales, second edition with the Mullen Scales of Early Learning in young children with and without neurodevelopmental disorders. Child Neuropsychol 2016;22:556–569.
    1. Davidsson P, Fredman P, Mansson JE, Svennerholm L. Determination of gangliosides and sulfatide in human cerebrospinal fluid with a microimmunoaffinity technique. Clin Chim Acta 1991;197:105–115.
    1. Kristjansdottir R, Uvebrant P, Lekman A, Mansson JE. Cerebrospinal fluid markers in children with cerebral white matter abnormalities. Neuropediatrics 2001;32:176–182.
    1. Bonadio W. Pediatric lumbar puncture and cerebrospinal fluid analysis. J Emerg Med 2014;46:141–150.
    1. Behrman RE, Kliegman RM, Nelson JHB. Textbook of Pediatrics, 16th ed Philadelphia: Saunders, 2000.
    1. Eeg‐Olofsson O, Link H, Wigertz A. Concentrations of CSF proteins as a measure of blood brain barrier function and synthesis of IgG within the CNS in 'normal' subjects from the age of 6 months to 30 years. Acta Paediatr Scand 1981;70:167–170.
    1. Tibbling G, Link H, Ohman S. Principles of albumin and IgG analyses in neurological disorders. I. Establishment of reference values. Scand J Clin Lab Invest 1977;37:385–390.
    1. Eichler F, Grodd W, Grant E, et al. Metachromatic leukodystrophy: a scoring system for brain MR imaging observations. AJNR Am J Neuroradiol 2009;30:1893–1897.
    1. Moffett JR, Ross B, Arun P, et al. N‐Acetylaspartate in the CNS: from neurodiagnostics to neurobiology. Prog Neurogibol 2007;81:89–131.
    1. Palisano R, Rosenbaum P, Walter S, et al. Development and reliability of a system to classify gross motor function in children with cerebral palsy. Dev Med Child Neurol 1997;39:214–223.
    1. Wagner AL, Buchthal F. Motor and sensory conduction in infancy and childhood: reappraisal. Dev Med Child Neurol 1972;14:189–216.
    1. Groeschel S, í Dali C, Clas P, et al. Cerebral gray and white matter changes and clinical course in metachromatic leukodystrophy. Neurology 2012;79:1662–1670.
    1. Beerepoot S, Nierkens S, Boelens JJ, et al. Peripheral neuropathy in metachromatic leukodystrophy: current status and future perspective. Orphanet J Rare Dis 2019;14:240.
    1. O'Brown NM, Pfau SJ, Gu C. Bridging barriers: a comparative look at the blood‐brain barrier across organisms. Genes Dev 2018;32:466–478.
    1. Urayama A. Toward the successful delivery of lysosomal enzymes across the blood‐brain barrier. Clin Exp Neuroimmunol 2013;4:228–238.
    1. Krägeloh‐Mann I, Groeschel S. Therapies of lysosomal storage disorders targeting the brain. Lancet 2016;388:440–442.
    1. Stroobants S, Gerlach D, Matthes F, et al. Intracerebroventricular enzyme infusion corrects central nervous system pathology and dysfunction in a mouse model of metachromatic leukodystrophy. Hum Mol Genet 2011;20:2760–2769.
    1. Calias P, Banks WA, Begley D, et al. Intrathecal delivery of protein therapeutics to the brain: a critical reassessment. Pharmacol Ther 2014;144:114–122.
    1. í Dali C, Sevin C, Krageloh‐Mann I, et al. Safety of intrathecal delivery of recombinant human arylsulfatase A in children with metachromatic leukodystrophy: Results from a phase 1/2 clinical trial. Mol Genet Metab 2020. 10.1016/j.ymgme.2020.07.002
    1. Simonis H, Yaghootfam C, Sylvester M, et al. Evolutionary redesign of the lysosomal enzyme arylsulfatase A increases efficacy of enzyme replacement therapy for metachromatic leukodystrophy. Hum Mol Genet 2019;28(11):1810–1821.

Source: PubMed

3
Abonner