Safe and sustained expression of human iduronidase after intrathecal administration of adeno-associated virus serotype 9 in infant rhesus monkeys

J Hordeaux, C Hinderer, EL Buza… - Human Gene …, 2019 - liebertpub.com
J Hordeaux, C Hinderer, EL Buza, JP Louboutin, T Jahan, P Bell, JA Chichester, AF Tarantal…
Human Gene Therapy, 2019liebertpub.com
Many neuropathic diseases cause early, irreversible neurologic deterioration, which
warrants therapeutic intervention during the first months of life. In the case of
mucopolysaccharidosis type I, a recessive lysosomal storage disorder that results from a
deficiency of the lysosomal enzyme α-l-iduronidase (IDUA), one of the most promising
treatment approaches is to restore enzyme expression through gene therapy. Specifically,
administering pantropic adeno-associated virus (AAV) encoding IDUA into the cerebrospinal …
Many neuropathic diseases cause early, irreversible neurologic deterioration, which warrants therapeutic intervention during the first months of life. In the case of mucopolysaccharidosis type I, a recessive lysosomal storage disorder that results from a deficiency of the lysosomal enzyme α-l-iduronidase (IDUA), one of the most promising treatment approaches is to restore enzyme expression through gene therapy. Specifically, administering pantropic adeno-associated virus (AAV) encoding IDUA into the cerebrospinal fluid (CSF) via suboccipital administration has demonstrated remarkable efficacy in large animals. Preclinical safety studies conducted in adult nonhuman primates supported a positive risk–benefit profile of the procedure while highlighting potential subclinical toxicity to primary sensory neurons located in the dorsal root ganglia (DRG). This study investigated the long-term performance of intrathecal cervical AAV serotype 9 gene transfer of human IDUA administered to 1-month-old rhesus monkeys (N = 4) with half of the animals tolerized to the human transgene at birth via systemic administration of an AAV serotype 8 vector expressing human IDUA from the liver. Sustained expression of the transgene for almost 4 years is reported in all animals. Transduced cells were primarily pyramidal neurons in the cortex and hippocampus, Purkinje cells in the cerebellum, lower motor neurons, and DRG neurons. Both tolerized and non-tolerized animals were robust and maintained transgene expression as measured by immunohistochemical analysis of brain tissue. However, the presence of antibodies in the non-tolerized animals led to a loss of measurable levels of secreted enzyme in the CSF. These results support the safety and efficiency of treating neonatal rhesus monkeys with AAV serotype 9 gene therapy delivered into the CSF.
Mary Ann Liebert