Therapeutic benefit after intracranial gene therapy delivered during the symptomatic stage in a feline model of Sandhoff disease

Victoria J McCurdy, Aime K Johnson, Heather L Gray-Edwards, Ashley N Randle, Allison M Bradbury, Nancy E Morrison, Misako Hwang, Henry J Baker, Nancy R Cox, Miguel Sena-Esteves, Douglas R Martin, Victoria J McCurdy, Aime K Johnson, Heather L Gray-Edwards, Ashley N Randle, Allison M Bradbury, Nancy E Morrison, Misako Hwang, Henry J Baker, Nancy R Cox, Miguel Sena-Esteves, Douglas R Martin

Abstract

Sandhoff disease (SD) is an autosomal recessive lysosomal storage disease caused by defects in the β-subunit of β-N-acetylhexosaminidase (Hex), the enzyme that catabolizes GM2 ganglioside. Hex deficiency causes neuronal storage of GM2 and related glycoconjugates, resulting in progressive neurodegeneration and death, typically in infancy. No effective treatment exists for human patients. Adeno-associated virus (AAV) gene therapy led to improved clinical outcome and survival of SD cats treated before the onset of disease symptoms. Most human patients are diagnosed after clinical disease onset, so it is imperative to test AAV-gene therapy in symptomatic SD cats to provide a realistic indication of therapeutic benefits that can be expected in humans. In this study, AAVrh8 vectors injected into the thalamus and deep cerebellar nuclei of symptomatic SD cats resulted in widespread central nervous system enzyme distribution, although a substantial burden of storage material remained. Cats treated in the early symptomatic phase showed delayed disease progression and a significant survival increase versus untreated cats. Treatment was less effective when administered later in the disease course, although therapeutic benefit was still possible. Results are encouraging for the treatment of human patients and provide support for the development AAV-gene therapy for human SD.

Trial registration: ClinicalTrials.gov NCT02716246 NCT03315182.

Conflict of interest statement

Conflict of Interest

The authors are beneficiaries of a licensing agreement with Axovant Gene Therapies (New York City) based partly on this technology. Drs. Sena-Esteves and Martin are shareholders in Lysogene (Neuilly-sur-Seine, France).

Figures

Figure 1.
Figure 1.
Survival and clinical progression of post-symptomatic AAV-treated SD cats and untreated SD cats. (a) Age of symptom onset is shown for untreated SD cats (mean ± s.d, n = 14). The scale is based on gait defects, which ultimately define humane endpoint, with initial deficits at 2.1 months old. However, disease onset begins at 1.3 months old with subtle tremors of the head/tail that progress to overt whole-body tremors at 2.4 months old. Neurologic humane endpoint is reached at a score of 3. (b) Kaplan-Meier survival curves for untreated SD cats (solid black line, n = 14) and SD cats treated post-symptomatically. Survival of the EPS cohort is significantly increased compared to untreated (P = 0.0010, dashed black line, n = 4) and compared to the LPS cohort (P = 0.048). Survival of the LPS cohort is not significantly increased compared to untreated (P < 0.28, solid grey line, n = 3). Normal controls (not shown) remained alive and healthy throughout the study. (c, d) Composite clinical scores are shown for untreated SD cats (n = 14) while EPS (c) and LPS (d) treated cats are depicted separately. Open symbols denote the presence of subtle tremors, and checkered symbols denote the presence of overt whole body tremors, which were absent in all EPS treated cats, but present in all LPS treated cats. Scores were assigned using two separate, independent readouts, as described in materials and methods. Normal controls (not shown) scored a 10 on the clinical rating scale and had no tremors for the duration of the study.
Figure 2.
Figure 2.
Neurological exam performance of post-symptomatic AAV-treated SD cats versus untreated SD cats. Cats were given a neurological examination twice monthly and deficits were recorded. * Onset of deficit was significantly delayed compared to untreated SD cats (P ≤ 0.011); ▼, onset of deficit was significantly earlier compared to the EPS cohort (P ≤ 0.026). In the LPS cohort, pelvic hopping deficits and pelvic wheelbarrowing deficits were already apparent at the time of treatment for cat 7-866 and all deficits except thoracic wheelbarrowing were apparent at the time of treatment for cat 7-801. The first deficit to appear in untreated SD cats is pelvic hopping at 2.2 ± 0.7 months, and the last deficit to appear is thoracic wheelbarrowing at 3.0 ± 0.7 months. Abbreviations: P = pelvic limb; T = thoracic limb; EPT = extensor postural thrust; LPS = late post-symptomatic; EPS = early post-symptomatic. Data is expressed as mean ± s.d. No deficits were recorded in normal control cats.
Figure 3.
Figure 3.
MRI evaluation of late post-symptomatic AAV-treated SD cats and untreated controls. T2-weighted MR images (3 Tesla) were taken at the level of the caudate nucleus (a, c and e) and DCN (b, d and f). Cortical white matter is hypointense to (darker than) gray matter in normal cats but hyperintense to (lighter than) gray matter in untreated SD cats. Also, the DCN area is hypointense to surrounding gray matter in normal cats (outlined black arrowhead in panel b) but becomes hyperintense with disease progression in untreated SD cats. In LPS cat 11-994 at humane endpoint, hypointensity of cortical white to gray matter is improved compared to untreated but is not normal. However, the DCN area has turned hyperintense to gray matter, similar to untreated SD cats. At the time of imaging cat 11-994 was on a clinical rating score of 3 (Figure 1a). There were no appreciable differences between a normal cat brain at 6 months old and at older ages. Brain atrophy, indicated by the amount of CSF (bright white area), in cat 11-994 was comparable to the untreated SD cat.
Figure 4.
Figure 4.
Therapeutic enzyme distribution in the CNS of post-symptomatic AAV-treated SD cats. Tissues from post-symptomatic AAV-treated SD cats were collected at humane endpoint. (a) Shown are injection sites (white circles) and 0.6 cm coronal blocks of the brain (A-H) and spinal cord (I-O) as depicted in a previous study. Brain blocks were halved, and the right half was assayed for enzyme activity. Lysosomal Hex activity (red) was visualized throughout the brain (b) and spinal cord (c) of early post-symptomatic AAV-treated SD cat 11-831 (SD + AAV) at 18.9 months old. Similarly, Hex activity was visualized throughout the brain (d) and spinal cord (e) of late post-symptomatic AAV-treated SD cat 11-994 (SD + AAV) at 11.5 months old. Representative control sections are shown from untreated normal cats along with untreated SD cats, which express ≤0.02 fold normal HexA activity in the brain and spinal cord. Corresponding HexA activity against MUGS substrate is shown below each block as fold normal level (fold N). Each block from treated and untreated SD cats is normalized to the corresponding block from age-matched normal controls (the mean value from 5 normal cats). HexA specific activity in normal control cats ranged from 28.1 ± 7.5 to 57.4 ± 5.7 nmol 4MU/mg protein/hr in the brain and from 6.7 ± 0.8 to 17.5 ± 3.2 nmol 4MU/mg/hr in the spinal cord.
Figure 5.
Figure 5.
Storage in the CNS of post-symptomatic AAV-treated SD cats. (a) Storage in untreated SD cats is visualized by dark PAS staining in gray matter. In treated brains, ganglioside storage was present in Hex deficient areas (see corresponding naphthol staining for the same cat, 11-831, in Figures 4b and c), such as the temporal lobe (blocks d and e) and cervical spinal cord (block k). (b) Sample sites for sialic acid quantitation (circles) in brain (A-H) and spinal cord (K and O; half of each block was used). (c) Sialic acid levels were measured in untreated SD cats (n = 4) and in the EPS (n = 4) and LPS (n = 3) cohorts for comparison to normal cats (n = 4 for each cohort). **, all samples from untreated cats were significantly higher than normal (P = 0.015 for each block) in the cerebrum (2.6 – 4.9 fold normal), brainstem and cerebellum (2.0 - 3.8 fold normal) and spinal cord (3.2 - 3.8 fold normal); *, samples from treated cats that were not significantly higher than normal. Samples without this symbol were significantly higher than normal (P ≤ 0.030 for each block); ▼, samples from treated cats that were significantly lower than untreated (P ≤ 0.030); ▲, sample from treated cats that was significantly higher than untreated (P = 0.015); ●, sample that was significantly lower in the LPS cohort versus the EPS cohort (P = 0.037). Abbreviations: EPS = early post-symptomatic; LPS = late post-symptomatic; tx = treatment. Data is expressed as mean ± s.d. Samples were analyzed in duplicate and the average value is reported.
Figure 6.
Figure 6.
Normalization of lysosomal mannosidase activity in the CNS and liver of post-symptomatic AAV-treated SD cats. Cats from the EPS (n = 4, black bars) and LPS (n = 3, striped bars) cohorts were euthanized at humane endpoint and lysosomal mannosidase activity was compared to untreated SD cats (n = 5, gray bars) and normal cats (n = 5) in the brain (A-H), spinal cord (I-O), and liver (Liv). CNS sample lettering corresponds to Figure 4a. *, all samples from untreated SD cats were significantly higher than normal (A-H, I-O, and liver,P = 0.0061 for each block); **, samples B-F, H, I-O, and liver from the LPS cohort remained significantly higher than normal (P ≤ 0.037 for each block); ***, all CNS samples from the EPS cohort remained significantly higher than normal (P ≤ 0.019 for each block), but liver was not significantly higher than normal; ▼, samples from AAV-treated SD cats that were significantly lower than untreated (P ≤ 0.037); ●, samples that were significantly higher in the LPS cohort versus the EPS cohort (P ≤ 0.040). Data is expressed as mean ± s.d. Samples were analyzed in duplicate and the average value is reported.

References

    1. Gravel RA, Kaback MM, Proia RL, Sandhoff K, Suzuki K, Suzuki K. The GM2 Gangliosidoses. In: Scriver CR, Beaudet AL, Sly WS, Valle D (eds). The Metabolic and Molecular Bases of Inherited Disease. 8th edn. (McGraw-Hill, New York, 2001) vol. 3, chapter 153.
    1. Conzelmann E, Kytzia H, Navon R, Sandhoff K. Ganglioside GM2 N-acetyl-beta-D-galactosaminidase activity in cultured fibroblasts of late-infantile and adult GM2 gangliosidosis patients and of healthy probands with low hexosaminidase level. Am J Hum Genet. 1983; 35: 900–914.
    1. Jacobs J, Willemsen M, Groot-Loonen J, Wevers R, Hoogerbrugge P. Allogeneic BMT followed by substrate reduction therapy in a child with subacute Tay-Sachs disease. Bone Marrow Transplant. 2005; 36: 925–926.
    1. Bembi B, Marchetti F, Guerci V, Ciana G, Addobbati R, Grasso D, et al. Substrate reduction therapy in the infantile form of Tay-Sachs disease. Neurology. 2006; 66: 278–280.
    1. Shapiro BE, Pastores GM, Gianutsos J, Luzy C, Kolodny EH. Miglustat in late-onset Tay-Sachs disease: a 12-month, randomized, controlled clinical study with 24 months of extended treatment. Genet Med. 2009; 11: 425–433.
    1. Maegawa GH, Banwell BL, Blaser S, Sorge G, Toplak M, Ackerley C, et al. Substrate reduction therapy in juvenile GM2 gangliosidosis. Mol Genet Metab. 2009; 98: 215–224.
    1. Tallaksen C, Berg J. Miglustat therapy in juvenile Sandhoff disease. J Inherit Metab Dis. 2009; 32: 289–293.
    1. Clarke JTR, Mahuran DJ, Sathe S, Kolodny EH, Rigat BA, Raiman JA, et al. An open-label Phase I/II clinical trial of pyrimethamine for the treatment of patients affected with chronic GM2 gangliosidosis (Tay–Sachs or Sandhoff variants). Mol Genet Metab. 2011; 102: 6–12.
    1. Osher E, Fattal-Valevski A, Sagie L, Urshanski N, Amir-Levi Y, Katzburg S, et al. Pyrimethamine increases β-hexosaminidase A activity in patients with Late Onset Tay Sachs. Mol Genet Metab. 2011; 102: 356–363.
    1. Neufeld EF, Fratantoni JC. Inborn errors of mucopolysaccharide metabolism. Science. 1970; 169: 141–146.
    1. Taylor RM, Wolfe JH. Decreased lysosomal storage in the adult MPS VII mouse brain in the vicinity of grafts of retroviral vector-corrected fibroblasts secreting high levels of beta-glucuronidase. Nat Med. 1997; 3: 771–774.
    1. Passini MA, Lee EB, Heuer GG, Wolfe JH. Distribution of a lysosomal enzyme in the adult brain by axonal transport and by cells of the rostral migratory stream. J Neurosci. 2002; 22: 6437–6446.
    1. Cearley CN, Wolfe JH. A single injection of an adeno-associated virus vector into nuclei with divergent connections results in widespread vector distribution in the brain and global correction of a neurogenetic disease. J Neurosci. 2007; 27: 9928–9940.
    1. Broekman M, Tierney L, Benn C, Chawla P, Cha J, Sena-Esteves M. Mechanisms of distribution of mouse β-galactosidase in the adult GM1-gangliosidosis brain. Gene Ther. 2008; 16: 303–308.
    1. Cachón-González MB, Wang SZ, McNair R, Bradley J, Lunn D, Ziegler R, et al. Gene Transfer Corrects Acute GM2 Gangliosidosis—Potential Therapeutic Contribution of Perivascular Enzyme Flow. Mol Ther. 2012; 20: 1489–1500.
    1. Haurigot V, Marco S, Ribera A, Garcia M, Ruzo A, Villacampa P, et al. Whole body correction of mucopolysaccharidosis IIIA by intracerebrospinal fluid gene therapy. J of Clin Invest. 2013; 123: 3254–3271.
    1. Cork L, Munnell JF, Lorenz MD, Murphy JV, Baker HJ, Rattazzi MC. GM2 ganglioside lysosomal storage disease in cats with beta-hexosaminidase deficiency. Science. 1977; 196: 1014–1017.
    1. Cork LC, Munnell JF, Lorenz MD. The pathology of feline GM2 gangliosidosis. Am J Pathol. 1978; 90: 723–734.
    1. Martin DR, Krum BK, Varadarajan G, Hathcock TL, Smith BF, Baker HJ. An inversion of 25 base pairs causes feline GM2 gangliosidosis variant 0. Exp Neurol. 2004; 187: 30–37.
    1. Cachón-González MB, Wang SZ, Lynch A, Ziegler R, Cheng SH, Cox TM. Effective gene therapy in an authentic model of Tay-Sachs-related diseases. PNAS. 2006; 103: 10373–10378.
    1. McCurdy VJ, Rockwell HE, Arthur JR, Bradbury AM, Johnson AK, Randle AN, et al. Widespread correction of central nervous system disease after intracranial gene therapy in a feline model of Sandhoff disease. Gene Ther. 2015; 22: 181–189.
    1. Rockwell HE, McCurdy VJ, Eaton SC, Wilson DU, Johnson AK, Randle AN, et al. AAV-mediated gene delivery in a feline model of Sandhoff disease corrects lysosomal storage in the central nervous system. ASN Neuro. 2015; 7(2): 1759091415569908. doi: 10.1177/1759091415569908.
    1. Bradbury AM, McCurdy VJ, Johnson AK, Gray-Edwards H, Brunson BL, Randle AN, et al. Intracranial AAV Gene Therapy Extends the Life Span of GM2 Gangliosidosis Cats >Four-Fold with No Clinical Evidence of Vector Toxicity. Mol Ther, 2013; 21; S226 (abstract 590).
    1. Bley AE, Giannikopoulos OA, Hayden D, Kubilus K, Tifft CJ, Eichler FS. Natural history of infantile GM2 gangliosidosis. Pediatrics. 2011: 128: e1233–e1241.
    1. Smith NJ, Winstone A, Stellitano L, Cox TM, Verity CM. GM2 gangliosidosis in a UK study of children with progressive neurodegeneration: 73 cases reviewed. Dev Med Child Neurol. 2012; 54: 176–182.
    1. Cachon-Gonzalez MB, Wang SZ, Ziegler R, Cheng SH, Cox TM. Reversibility of neuropathology in Tay-Sachs-related diseases. Hum Mol Genet. 2014; 23: 730–748.
    1. Bradbury AM, Cochran JN, McCurdy VJ, Johnson AK, Brunson BL, Gray-Edwards H, et al. Therapeutic Response in Feline Sandhoff Disease Despite Immunity to Intracranial Gene Therapy. Mol Ther. 2013; 21: 1306–1315.
    1. Broekman M, Comer L, Hyman B, Sena-Esteves M. Adeno-associated virus vectors serotyped with AAV8 capsid are more efficient than AAV-1 or-2 serotypes for widespread gene delivery to the neonatal mouse brain. Neuroscience. 2006; 138: 501–510.
    1. Bradbury AM, Morrison NE, Hwang M, Cox NR, Baker HJ, Martin DR. Neurodegenerative lysosomal storage disease in European Burmese cats with hexosaminidase beta-subunit deficiency. Mol Genet Metab. 2009; 97: 53–57.
    1. Lacorazza H, Jendoubi M. In situ assessment of beta-hexosaminidase activity. Biotechniques. 1995; 19: 434–440.
    1. McCurdy VJ, Johnson AK, Gray-Edwards HL, Randle AN, Brunson BL, Morrison NE. Sustained normalization of neurologic disease after intracranial gene therapy in a feline model. Science Translational Medicine. 2014; 6(231): 231ra48.
    1. Sondhi D, Hackett NR, Peterson DA, Stratton J, Baad M, Travis KM, et al. Enhanced survival of the LINCL mouse following CLN2 gene transfer using the rh. 10 rhesus macaque-derived adeno-associated virus vector. Mol Ther. 2006; 15: 481–491.
    1. Baek RC, Martin DR, Cox NR, Seyfried TN. Comparative analysis of brain lipids in mice, cats, and humans with Sandhoff disease. Lipids. 2009; 44: 197–205.
    1. Kolter T, Proia RL, Sandhoff K. Combinatorial ganglioside biosynthesis. J Biol Chem. 2002; 277: 25859–25862.
    1. Baker HJ, Reynolds GD, Walkley SU, Cox NR, Baker GH. The Gangliosidoses: Comparitive Features and Research Applications. Vet Pathol. 1979; 16: 635–649.
    1. Cressant A, Desmaris N, Verot L, Bréjot T, Froissart R, Vanier MT, et al. Improved behavior and neuropathology in the mouse model of Sanfilippo type IIIB disease after adeno-associated virus-mediated gene transfer in the striatum. Journal Neurosci. 2004; 24: 10229–10239.
    1. Lew RM, Proos AL, Burnett L, Delatycki M, Bankier A, Fietz MJ. Tay Sachs disease in Australia: reduced disease incidence despite stable carrier frequency in Australian Jews. Medical J Aust. 2012; 197: 652–654.
    1. Smith NJ, Winstone A, Stellitano L, Cox TM, Verity CM. GM2 gangliosidosis in a UK study of children with progressive neurodegeneration: 73 cases reviewed. Dev Med Child Neurol. 2012; 54: 176–182.
    1. Ahmed SS, Li H, Cao C, Sikoglu EM, Denninger AR, Su Q, et al. A single intravenous rAAV injection as late as P20 achieves efficacious and sustained CNS Gene therapy in canavan mice. Mol Ther. 2013; 21: 2136–2147.
    1. Cabrera-Salazar MA, Roskelley EM, Bu J, Hodges BL, Yew N, Dodge JC, et al. Timing of therapeutic intervention determines functional and survival outcomes in a mouse model of late infantile batten disease. Mol Ther. 2007; 15: 1782–1788.
    1. Ellinwood NM, Ausseil J, Desmaris N, Bigou S, Liu S, Jens JK, et al. Safe, efficient, and reproducible gene therapy of the brain in the dog models of Sanfilippo and Hurler syndromes. Mol Ther. 2011; 19: 251–259.
    1. Golebiowski D, van der Bom IMJ, Kwon C, Miller AD, Petrosky K, Bradbury AM, et al. Direct Intracranial Injection of AAVrh8 Encoding Monkey β-N-Acetylhexosaminidase Causes Neurotoxicity in the Primate Brain. Hum Gene Ther. 2017; 28: 510 – 522.
    1. Elliger S, Elliger C, Aguilar C, Raju N, Watson G. Elimination of lysosomal storage in brains of MPS VII mice treated by intrathecal administration of an adeno-associated virus vector. Gene Ther. 1999; 6: 1175–1178.
    1. Bradbury AM, Gray-Edwards H, Shirley JL, McCurdy VJ, Colaco AN, Randle AN, et al. Biomarkers for Disease Progression and AAV Therapeutic Efficacy in Feline Sandhoff Disease. Exp Neurol. 2015; 263: 102 – 112.
    1. Gray-Edwards HL, Brunson BL, Holland M, Hespel AM, Bradbury AM, McCurdy VJ. Mucopolysaccharidosis-like Phenotype in Feline Sandhoff Disease and Partial Correction after AAV Gene Therapy. Mol Genet Metab. 2015; 116: 80 – 87.
    1. Bradbury AM, Bagel JH, Brisson BK, Marshall MS, Pesayco Salvador J, 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. Gurda BL, De Guilhem De Lataillade A, Bell P, Zhu Y, Yu H, Wang P, et al. (2016) Evaluation of AAV-mediated Gene Therapy for Central Nervous System Disease in Canine Mucopolysaccharidosis VII. Mol Ther 24(2):206–216.
    1. Sun B, Zhang H, Bird A, Li S, Young SP, Koeberl DD. Impaired clearance of accumulated lysosomal glycogen in advanced Pompe disease despite high-level vector-mediated transgene expression. Journal Gene Med. 2009; 11: 913–920.
    1. Jeyakumar M, Thomas R, Elliot-Smith E, Smith DA, van der Spoel AC, d'Azzo A. Central nervous system inflammation is a hallmark of pathogenesis in mouse models of GM1 and GM2 gangliosidosis. Brain. 2003; 126: 974 – 87.
    1. Bradbury AM, Peterson TA, Gross AL, Wells SZ, McCurdy VJ, Wolfe KG. AAV Mediated Gene Delivery Attenuates Neuroinflammation in Feline Sandhoff Disease. Neuroscience. 2016; 340: 117 – 125.
    1. Walkley SU. Cellular pathology of lysosomal storage disorders. Brain Pathol. 1998; 8: 175–193.
    1. Walkley S, Wurzelmann S, Siegel D. Ectopic axon hillock-associated neurite growth is maintained in metabolically reversed swainsonine-induced neuronal storage disease. Brain Res. 1987; 410: 89–96.
    1. Vite CH, McGowan JC, Niogi SN, Passini MA, Drobatz KJ, Haskins ME, et al. Effective gene therapy for an inherited CNS disease in a large animal model. Ann Neurol. 2005; 57: 355–364.

Source: PubMed

3
Iratkozz fel