Efficacy and Safety of a Krabbe Disease Gene Therapy

Juliette Hordeaux, Brianne A Jeffrey, Jinlong Jian, Gourav R Choudhury, Kristofer Michalson, Thomas W Mitchell, Elizabeth L Buza, Jessica Chichester, Cecilia Dyer, Jessica Bagel, Charles H Vite, Allison M Bradbury, James M Wilson, Juliette Hordeaux, Brianne A Jeffrey, Jinlong Jian, Gourav R Choudhury, Kristofer Michalson, Thomas W Mitchell, Elizabeth L Buza, Jessica Chichester, Cecilia Dyer, Jessica Bagel, Charles H Vite, Allison M Bradbury, James M Wilson

Abstract

Krabbe disease is a lysosomal storage disease caused by mutations in the gene that encodes galactosylceramidase, in which galactosylsphingosine (psychosine) accumulation drives demyelination in the central and peripheral nervous systems, ultimately progressing to death in early childhood. Gene therapy, alone or in combination with transplant, has been developed for almost two decades in mouse models, with increasing therapeutic benefit paralleling the improvement of next-generation adeno-associated virus (AAV) vectors. This effort has recently shown remarkable efficacy in the canine model of the disease by two different groups that used either systemic or cerebrospinal fluid (CSF) administration of AAVrh10 or AAV9. Building on our experience developing CSF-delivered, AAV-based drug products for a variety of neurodegenerative disorders, we conducted efficacy, pharmacology, and safety studies of AAVhu68 delivered to the CSF in two relevant natural Krabbe animal models, and in nonhuman primates. In newborn Twitcher mice, the highest dose (1 × 1011 genome copies [GC]) of AAVhu68.hGALC injected into the lateral ventricle led to a median survival of 130 days compared to 40.5 days in vehicle-treated mice. When this dose was administered intravenously, the median survival was 49 days. A single intracisterna magna injection of AAVhu68.cGALC at 3 × 1013 GC into presymptomatic Krabbe dogs increased survival for up to 85 weeks compared to 12 weeks in controls. It prevented psychosine accumulation in the CSF, preserved peripheral nerve myelination, ambulation, and decreased brain neuroinflammation and demyelination, although some regions remained abnormal. In a Good Laboratory Practice-compliant toxicology study, we administered the clinical candidate into the cisterna magna of 18 juvenile rhesus macaques at 3 doses that displayed efficacy in mice. We observed no dose-limiting toxicity and sporadic minimal degeneration of dorsal root ganglia (DRG) neurons. Our studies demonstrate the efficacy, scalability, and safety of a single cisterna magna AAVhu68 administration to treat Krabbe disease. ClinicalTrials.Gov ID: NCT04771416.

Keywords: AAV; Krabbe; Twitcher mouse; cisterna magna; demyelination; galactosylceramidase (GALC); galactosylceramide; lysosomal storage disease; psychosine.

Conflict of interest statement

J.M.W. is a paid advisor to and holds equity in iECURE, Scout Bio, Passage Bio, and the Center for Breakthrough Medicines (CBM). He also holds equity in the G2 Bio-associated asset companies. He has sponsored research agreements with Amicus Therapeutics, Biogen, CBM, Elaaj Bio, FA212, G2 Bio, G2 Bio-associated asset companies, iECURE, Janssen, Passage Bio, and Scout Bio, which are licensees of Penn technology. J.M.W. and J.H. are inventors on patents that have been licensed to various biopharmaceutical companies and for which they may receive payments.

A.M.B. is a beneficiary of a licensing agreement with Axovant Gene Therapies (royalties); has received income from Neurogene (consulting and honorarium); and is an inventor on a patent application (Gray SJ, Lykken E, Vite CH, Bradbury AM. Optimized GALC Genes and Expression Cassettes and Their Use. PCT/US2019/067727). All other authors have no competing financial interests exist.

Figures

Figure 1.
Figure 1.
Proof-of-concept efficacy in neonatal Twitcher mice. (A) Survival curve of Twitcher mice injected intravenously on PND0 with 50 μL of PBS (vehicle, n = 8; median survival 40.5 days) or with 1 × 1011 GC of AAVhu68.CB7.hGALCco.rBG diluted in 50 μL PBS (n = 6; median survival 49 days). ***p < 0.001 Log-rank (Mantel-Cox) test. (B) Survival curve of Twitcher mice injected in the lateral cerebral ventricle (ICV) on PND0 with 2 μL of PBS (vehicle, n = 8; median survival 43 days), with 2 × 1010 GC of AAVhu68.CB7.hGALCco.rBG in 2 μL PBS (low dose, n = 10; median survival 62 days), with 5 × 1010 GC of AAVhu68.CB7.hGALCco.rBG in 2 μL PBS (mid dose, n = 12; median survival 99 days), or with 1 × 1011 GC of AAVhu68.CB7.hGALCco.rBG in 2 × 2 μL PBS (bilateral ICV, high dose, n = 12; median survival 130 days). ****p < 0.0001 Log-rank (Mantel-Cox) test. (C) Neuromotor function assessed by the accelerated rotarod test on PND35 in mice treated via neonatal ICV administration [same animals as in (B)]. **p < 0.01, ***p < 0.001, ****p < 0.0001, Kruskal-Wallis test followed by post hoc Dunn's multiple comparison test, alpha = 0.05, comparison to vehicle Twitcher mice. (D) GALC enzyme activity in brain lysate from tissue obtained at humane endpoint in animals administered IV or ICV [same animals as in (A, B)]. *p < 0.05, ****p < 0.0001, Kruskal-Wallis test followed by post hoc Dunn's multiple comparison test, alpha = 0.05, comparison to vehicle Twitcher mice. GALC, galactosylceramidase; GC, genome copies; ICV, intracerebroventricular; IV, intravenous; PBS, phosphate-buffered saline; PND, postnatal day.
Figure 2.
Figure 2.
MED study in juvenile postdisease onset Twitcher mice. (A) Compound clinical severity score in Twitcher mice treated on PND12–14 ICV with either 4 μL of artificial CSF (vehicle, n = 17), or 4 μL of AAVhu68.CB7.hGALCco.rBG at the following doses: 6.8 × 109 GC (n = 16), 2 × 1010 GC (n = 17), 6.8 × 1010 (n = 17), or 2 × 1011 GC (n = 16). WT littermates were treated ICV with 4 μL of artificial CSF (n = 17). The operator was blinded to the mice genotype and treatment. ****p < 0.0001 linear mixed-effect modeling comparing the clinical score change over time compared to the vehicle Twitcher group, alpha = 0.05. (B) Neuromotor function assessed by the accelerated rotarod test on PND35 in same mice as in (A) **p < 0.01, ***p < 0.001, ****p < 0.0001, Kruskal-Wallis test followed by post hoc Dunn's multiple comparison test, alpha = 0.05, comparison to vehicle Twitcher mice. (C) GALC enzyme activity in brain (sagittal half), heart (sagittal half), and liver (half of the left lobe) lysate from tissue obtained either at scheduled necropsy (PND40, half of the animals) or humane endpoint (half of the animals) in the same animals as in (A) *p < 0.05, ****p < 0.0001, Kruskal-Wallis test followed by post hoc Dunn's multiple comparison test, alpha = 0.05, comparison to vehicle Twitcher mice. (D) Neuroinflammation quantification in the cerebral cortex, spinal cord, and sciatic nerve measured by mean object area of IBA1-positive cells stained by immunohistochemistry in tissues collected at the fixed necropsy time PND40. ***p < 0.001, ****p < 0.0001, Kruskal-Wallis test followed by post hoc Dunn's multiple comparison test, alpha = 0.05, comparison to vehicle-treated Twitcher mice. Representative images of IBA1 IHC from the vehicle groups (Twitcher and WT mice) and the HD group (2 × 1011 GC, ICV PND12–14). CSF, cerebrospinal fluid; MED, minimum effective dose; WT, wild type.
Figure 3.
Figure 3.
ICM efficacy study in Krabbe dogs, nerve-conduction studies, CSF biomarkers, and MRI. (A) Nerve-conduction velocities in the radial (sensory), sciatic (motor), ulnar (motor), and tibial (motor) nerves in 2- to 3-week-old Krabbe dogs treated ICM with either (1) 1 mL of artificial CSF (vehicle, n = 2); or (2) 3 × 1013 GC of AAVhu68.CB7.cGAMCco.rBG in 1 mL (n = 4). A WT littermate that received 1 mL of artificial CSF ICM was used as a control. Two treated dogs were sacrificed at the scheduled timepoint of 6 months postinjection for tissue collection. Two treated dogs and the vehicle-treated Krabbe dogs were followed until humane endpoint. (B) Quantification of CSF psychosine and GalCer levels and CSF GALC enzyme activity. The dotted line represent the average GALC activity value from the serial CSF timepoints of the WT control (K928). (C) Brain MRI WM intensity semiquantitative scoring. (D) Brain MRI examples from one Krabbe vehicle dog (8 weeks), one Krabbe dog treated with AAV (10 weeks), and one WT control littermate (10 weeks). A dark hypointense WM represents normal myelination (stars); a grey isointense signal represents mild demyelination (arrowhead); a white hyperintense signal represents marked demyelination (arrows). AAV, adeno-associated virus; GalCer, galactosylceramide; ICM, intracisterna magna; MRI, magnetic resonance imaging; WM, white matter.
Figure 4.
Figure 4.
ICM efficacy study in Krabbe dogs, neuroinflammation and GALC levels. (A) Representative pictures of IBA1 staining by IHC illustrating the macrophage/microglial neuroinflammation in Krabbe dogs treated with vehicle control or with AAV ICM. (B) Quantification of the mean area of the IBA1-positive signal in different neuroanatomical regions of the brain, spinal cord, and sciatic nerve of Krabbe dogs treated with AAV or vehicle. Each bar represents an animal. Error bars when present (cerebral cortical WM, corpus callosum, centrum semiovale, and internal capsule), represent the standard deviation from quantification of multiple slides when the neuroanatomical region was present on more than one brain section. (C) GALC activity in tissue lysate (50 μg protein per reaction, 2 h incubation) relative to the WT control (dotted line). Each bar represents one animal. WM, white matter.
Figure 5.
Figure 5.
GLP toxicology study in rhesus macaques, safety. (A) Fluoroscopic images of the neck region, before and after contrast ICM administration, in juvenile rhesus macaques (animal 18–167, mid-dose group). Contrast injection was used to confirm needle placement before AAV administration. Arrows show the diffusion of the contrast in the cisterna magna and spinal canal. (B) WBC counts in the CSF of juvenile rhesus macaques treated ICM with artificial CSF (vehicle, n = 2) or AAVhu68.CB7.hGALCco.rBG at the dose of 4.5 × 1012 GC (low dose, n = 6), 1.5 × 1013 GC (mid dose, n = 6), or 4.5 × 1013 (high dose, n = 6). Half of the animals were euthanized for tissue collection 3 months postdosing; the other half were euthanized 6 months postdosing. Dotted line represents the upper limit of normal (C) Histopathological analysis, DRG neuronal degeneration, and spinal cord dorsal column axonopathy severity grades (0 = normal, 1 = minimal, 2 = mild, 3 = moderate, 4 = marked, and 5 = severe). Three and six months cohorts are included, each data point represents 1 segment per animal (cervical, thoracic, and lumbar). *p < 0.05 Mann-Whitney rank test, alpha = 0.05. (D) Median nerve SNAP amplitudes at BL (before dosing), 28-, 60-, and 90-day postdosing in the 3 months cohorts. BL, baseline; DRG, dorsal root ganglia; GLP, Good Laboratory Practice; SNAP, sensory nerve action potential; WBC, white blood cell.

References

    1. Wenger DA, Rafi MA, Luzi P, et al. . Krabbe disease: genetic aspects and progress toward therapy. Mol Genet Metab 2000;70:1–9.
    1. Svennerholm L, Vanier MT, Mansson JE. Krabbe disease: a galactosylsphingosine (psychosine) lipidosis. J Lipid Res 1980;21:53–64.
    1. Escolar ML, Kiely BT, Shawgo E, et al. . Psychosine, a marker of Krabbe phenotype and treatment effect. Mol Genet Metab 2017;121:271–278.
    1. Voccoli V, Tonazzini I, Signore G, et al. . Role of extracellular calcium and mitochondrial oxygen species in psychosine-induced oligodendrocyte cell death. Cell Death Dis 2014;5:e1529.
    1. Potter GB, Santos M, Davisson MT, et al. . Missense mutation in mouse GALC mimics human gene defect and offers new insights into Krabbe disease. Hum Mol Genet 2013;22:3397–3414.
    1. D'Auria L, Reiter C, Ward E, et al. . Psychosine enhances the shedding of membrane microvesicles: implications in demyelination in Krabbe's disease. PLoS One 2017;12:e0178103.
    1. Smith B, Galbiati F, Castelvetri LC, et al. . Peripheral neuropathy in the Twitcher mouse involves the activation of axonal caspase 3. ASN Neuro 2011;3:e00066.
    1. Weinstock NI, Shin D, Dhimal N, et al. Macrophages expressing GALC improve peripheral Krabbe disease by a mechanism independent of cross-correction. Neuron 2020;107:65.e9–81.e9.
    1. Suzuki K. Globoid cell leukodystrophy (Krabbe's disease): update. J Child Neurol 2003;18:595–603.
    1. Nicaise AM, Bongarzone ER, Crocker SJ. A microglial hypothesis of globoid cell leukodystrophy pathology. J Neurosci Res 2016;94:1049–1061.
    1. Duffner PK, Barczykowski A, Jalal K, et al. . Early infantile Krabbe disease: results of the World-Wide Krabbe Registry. Pediatr Neurol 2011;45:141–148.
    1. Duffner PK, Barczykowski A, Kay DM, et al. . Later onset phenotypes of Krabbe disease: results of the world-wide registry. Pediatr Neurol 2012;46:298–306.
    1. Duffner PK, Jalal K, Carter RL. The Hunter's Hope Krabbe family database. Pediatr Neurol 2009;40:13–18.
    1. Abdelhalim AN, Alberico RA, Barczykowski AL, et al. . Patterns of magnetic resonance imaging abnormalities in symptomatic patients with Krabbe disease correspond to phenotype. Pediatr Neurol 2014;50:127–134.
    1. Escolar ML, Poe MD, Martin HR, et al. . A staging system for infantile Krabbe disease to predict outcome after unrelated umbilical cord blood transplantation. Pediatrics 2006;118:e879–e889.
    1. Escolar ML, West T, Dallavecchia A, et al. . Clinical management of Krabbe disease. J Neurosci Res 2016;94:1118–1125.
    1. Escolar ML, Poe MD, Provenzale JM, et al. . Transplantation of umbilical-cord blood in babies with infantile Krabbe's disease. N Engl J Med 2005;352:2069–2081.
    1. Wright MD, Poe MD, DeRenzo A, et al. . Developmental outcomes of cord blood transplantation for Krabbe disease: a 15-year study. Neurology 2017;89:1365–1372.
    1. Wasserstein MP, Andriola M, Arnold G, et al. . Clinical outcomes of children with abnormal newborn screening results for Krabbe disease in New York State. Genet Med 2016;18:1235–1243.
    1. Kobayashi T, Yamanaka T, Jacobs JM, et al. . The Twitcher mouse: an enzymatically authentic model of human globoid cell leukodystrophy (Krabbe disease). Brain Res 1980;202:479–483.
    1. Wenger DA, Victoria T, Rafi MA, et al. . Globoid cell leukodystrophy in cairn and West Highland white terriers. J Hered 1999;90:138–142.
    1. Borda JT, Alvarez X, Mohan M, et al. . Clinical and immunopathologic alterations in rhesus macaques affected with globoid cell leukodystrophy. Am J Pathol 2008;172:98–111.
    1. Lee WC, Courtenay A, Troendle FJ, et al. . Enzyme replacement therapy results in substantial improvements in early clinical phenotype in a mouse model of globoid cell leukodystrophy. FASEB J 2005;19:1549–1551.
    1. Lee WC, Tsoi YK, Troendle FJ, et al. . Single-dose intracerebroventricular administration of galactocerebrosidase improves survival in a mouse model of globoid cell leukodystrophy. FASEB J 2007;21:2520–2527.
    1. Rafi MA, Zhi Rao H, Passini MA, et al. . AAV-mediated expression of galactocerebrosidase in brain results in attenuated symptoms and extended life span in murine models of globoid cell leukodystrophy. Mol Ther 2005;11:734–744.
    1. Lin D, Donsante A, Macauley S, et al. . Central nervous system-directed AAV2/5-mediated gene therapy synergizes with bone marrow transplantation in the murine model of globoid-cell leukodystrophy. Mol Ther 2007;15:44–52.
    1. Rafi MA, Rao HZ, Luzi P, et al. . Extended normal life after AAVrh10-mediated gene therapy in the mouse model of Krabbe disease. Mol Ther 2012;20:2031–2042.
    1. Rafi MA, Rao HZ, Luzi P, et al. . Long-term improvements in lifespan and pathology in CNS and PNS after BMT plus one intravenous injection of AAVrh10-GALC in Twitcher mice. Mol Ther 2015;23:1681–1690.
    1. Rafi MA, Luzi P, Wenger DA. Can early treatment of twitcher mice with high dose AAVrh10-GALC eliminate the need for BMT? Bioimpacts 2021;11:135–146.
    1. Karumuthil-Melethil S, Marshall MS, Heindel C, et al. . Intrathecal administration of AAV/GALC vectors in 10-11-day-old twitcher mice improves survival and is enhanced by bone marrow transplant. J Neurosci Res 2016;94:1138–1151.
    1. Bradbury AM, Rafi MA, Bagel JH, et al. . AAVrh10 gene therapy ameliorates central and peripheral nervous system disease in canine globoid cell leukodystrophy (Krabbe disease). Hum Gene Ther 2018;29:785–801.
    1. Bradbury AM, Bagel JH, Nguyen D, et al. . Krabbe disease successfully treated via monotherapy of intrathecal gene therapy. J Clin Invest 2020;130:4906–4920.
    1. Li Y, Miller CA, Shea LK, et al. . Enhanced efficacy and increased long-term toxicity of CNS-directed, AAV-based combination therapy for Krabbe disease. Mol Ther 2021;29:691–701.
    1. Gurda BL, De Guilhem De Lataillade A, Bell P, et al. . Evaluation of AAV-mediated gene therapy for central nervous system disease in canine mucopolysaccharidosis VII. Mol Ther 2016;24:206–216.
    1. Hinderer C, Bell P, Gurda BL, et al. . Intrathecal gene therapy corrects CNS pathology in a feline model of mucopolysaccharidosis I. Mol Ther 2014;22:2018–2027.
    1. Hinderer C, Bell P, Katz N, et al. . Evaluation of intrathecal routes of administration for adeno-associated viral vectors in large animals. Hum Gene Ther 2018;29:15–24.
    1. Hinderer C, Bell P, Louboutin JP, et al. . Neonatal tolerance induction enables accurate evaluation of gene therapy for MPS I in a canine model. Mol Genet Metab 2016;119:124–130.
    1. Hinderer C, Bell P, Vite CH, et al. . Widespread gene transfer in the central nervous system of cynomolgus macaques following delivery of AAV9 into the cisterna magna. Mol Ther Methods Clin Dev 2014;1:14051.
    1. Hinderer C, Katz N, Dyer C, et al. . Translational feasibility of lumbar puncture for intrathecal AAV administration. Mol Ther Methods Clin Dev 2020;17:969–974.
    1. Haurigot V, Marco S, Ribera A, et al. . Whole body correction of mucopolysaccharidosis IIIA by intracerebrospinal fluid gene therapy. J Clin Invest 2013;123:3254–3271.
    1. Ribera A, Haurigot V, Garcia M, et al. . Biochemical, histological and functional correction of mucopolysaccharidosis type IIIB by intra-cerebrospinal fluid gene therapy. Hum Mol Genet 2015;24:2078–2095.
    1. Gray SJ, Nagabhushan Kalburgi S, McCown TJ, et al. . Global CNS gene delivery and evasion of anti-AAV-neutralizing antibodies by intrathecal AAV administration in non-human primates. Gene Ther 2013;20:450–459.
    1. Bucher T, Dubreil L, Colle MA, et al. . Intracisternal delivery of AAV9 results in oligodendrocyte and motor neuron transduction in the whole central nervous system of cats. Gene Ther 2014;21:522–528.
    1. Passini MA, Bu J, Richards AM, et al. . Translational fidelity of intrathecal delivery of self-complementary AAV9-survival motor neuron 1 for spinal muscular atrophy. Hum Gene Ther 2014;25:619–630.
    1. Samaranch L, Salegio EA, San Sebastian W, et al. . Adeno-associated virus serotype 9 transduction in the central nervous system of nonhuman primates. Hum Gene Ther 2012;23:382–389.
    1. Hinderer C, Katz N, Buza EL, et al. . Severe toxicity in nonhuman primates and piglets following high-dose intravenous administration of an adeno-associated virus vector expressing human SMN. Hum Gene Ther 2018;29:285–298.
    1. Wilson JM, Flotte TR. Moving forward after two deaths in a gene therapy trial of myotubular myopathy. Hum Gene Ther 2020;31:695–696.
    1. Hordeaux J, Buza EL, Dyer C, et al. . Adeno-associated virus-induced dorsal root ganglion pathology. Hum Gene Ther 2020;31:808–818.
    1. Hordeaux J, Hinderer C, Buza EL, et al. . Safe and sustained expression of human iduronidase after intrathecal administration of adeno-associated virus serotype 9 in infant rhesus monkeys. Hum Gene Ther 2019;30:957–966.
    1. Hordeaux J, Hinderer C, Goode T, et al. . Toxicology study of intra-cisterna magna adeno-associated virus 9 expressing iduronate-2-sulfatase in rhesus macaques. Mol Ther Methods Clin Dev 2018;10:68–78.
    1. Hordeaux J, Hinderer C, Goode T, et al. . Toxicology study of intra-cisterna magna adeno-associated virus 9 expressing human alpha-L-iduronidase in rhesus macaques. Mol Ther Methods Clin Dev 2018;10:79–88.
    1. Chand DH, Zaidman C, Arya K, et al. . Thrombotic microangiopathy following onasemnogene abeparvovec for spinal muscular atrophy: a case series. J Pediatr 2021;231:265–268.
    1. Lock M, Alvira M, Vandenberghe LH, et al. . Rapid, simple, and versatile manufacturing of recombinant adeno-associated viral vectors at scale. Hum Gene Ther 2010;21:1259–1271.
    1. Lock M, Alvira MR, Chen SJ, et al. . Absolute determination of single-stranded and self-complementary adeno-associated viral vector genome titers by droplet digital PCR. Hum Gene Ther Methods 2014;25:115–125.
    1. Katz N, Goode T, Hinderer C, et al. . Standardized method for intra-cisterna magna delivery under fluoroscopic guidance in nonhuman primates. Hum Gene Ther Methods 2018;29:212–219.
    1. Corado CR, Pinkstaff J, Jiang X, et al. . Cerebrospinal fluid and serum glycosphingolipid biomarkers in canine globoid cell leukodystrophy (Krabbe Disease). Mol Cell Neurosci 2020;102:103451.
    1. Calcedo R, Chichester JA, Wilson JM. Assessment of humoral, innate, and T-cell immune responses to adeno-associated virus vectors. Hum Gene Ther Methods 2018;29:86–95.
    1. Hinderer C, Bell P, Louboutin JP, et al. . Neonatal systemic AAV induces tolerance to CNS gene therapy in MPS I dogs and nonhuman primates. Mol Ther 2015;23:1298–1307.
    1. Taniike M, Suzuki K. Spacio-temporal progression of demyelination in twitcher mouse: with clinico-pathological correlation. Acta Neuropathol 1994;88:228–236.
    1. Workman AD, Charvet CJ, Clancy B, et al. . Modeling transformations of neurodevelopmental sequences across mammalian species. J Neurosci 2013;33:7368–7383.
    1. Arant BS Jr., Gooch WM, 3rd. Developmental changes in the mongrel canine brain during postnatal life. Early Hum Dev 1982;7:179–194.
    1. Bradbury AM, Bagel JH, Jiang X, et al. . Clinical, electrophysiological, and biochemical markers of peripheral and central nervous system disease in canine globoid cell leukodystrophy (Krabbe's disease). J Neurosci Res 2016;94:1007–1017.
    1. Hinderer C, Nosratbakhsh B, Katz N, et al. . A single injection of an optimized adeno-associated viral vector into cerebrospinal fluid corrects neurological disease in a murine model of GM1 gangliosidosis. Hum Gene Ther 2020;31:1169–1177.
    1. Marco S, Haurigot V, Jaen ML, et al. . Seven-year follow-up of durability and safety of AAV CNS gene therapy for a lysosomal storage disorder in a large animal. Mol Ther Methods Clin Dev 2021;23:370–389.
    1. Kwon JM, Matern D, Kurtzberg J, et al. . Consensus guidelines for newborn screening, diagnosis and treatment of infantile Krabbe disease. Orphanet J Rare Dis 2018;13:30.
    1. Mizisin AP, Weerasuriya A. Homeostatic regulation of the endoneurial microenvironment during development, aging and in response to trauma, disease and toxic insult. Acta Neuropathol 2011;121:291–312.
    1. Wolf NI, Breur M, Plug B, et al. . Metachromatic leukodystrophy and transplantation: remyelination, no cross-correction. Ann Clin Transl Neurol 2020;7:169–180.
    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. Rafi MA, Luzi P, Wenger DA. Conditions for combining gene therapy with bone marrow transplantation in murine Krabbe disease. Bioimpacts 2020;10:105–115.
    1. Kaminski D, Yaghootfam C, Matthes F, et al. . Brain cell type-specific endocytosis of arylsulfatase A identifies limitations of enzyme-based therapies for metachromatic leukodystrophy. Hum Mol Genet 2021;29:3807–3817.
    1. Bradbury A, Peterson D, Vite C, et al. . Diffusion tensor imaging analysis of the brain in the canine model of Krabbe disease. Neuroradiol J 2016;29:417–424.

Source: PubMed

3
Abonnieren