Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
Add more filters










Publication year range
1.
Mol Ther ; 2024 Mar 27.
Article in English | MEDLINE | ID: mdl-38549375

ABSTRACT

Leukoencephalopathy with vanishing white matter (VWM) is a progressive incurable white matter disease that most commonly occurs in childhood and presents with ataxia, spasticity, neurological degeneration, seizures, and premature death. A distinctive feature is episodes of rapid neurological deterioration provoked by stressors such as infection, seizures, or trauma. VWM is caused by autosomal recessive mutations in one of five genes that encode the eukaryotic initiation factor 2B complex, which is necessary for protein translation and regulation of the integrated stress response. The majority of mutations are in EIF2B5. Astrocytic dysfunction is central to pathophysiology, thereby constituting a potential therapeutic target. Herein we characterize two VWM murine models and investigate astrocyte-targeted adeno-associated virus serotype 9 (AAV9)-mediated EIF2B5 gene supplementation therapy as a therapeutic option for VWM. Our results demonstrate significant rescue in body weight, motor function, gait normalization, life extension, and finally, evidence that gene supplementation attenuates demyelination. Last, the greatest rescue results from a vector using a modified glial fibrillary acidic protein (GFAP) promoter-AAV9-gfaABC(1)D-EIF2B5-thereby supporting that astrocytic targeting is critical for disease correction. In conclusion, we demonstrate safety and early efficacy through treatment with a translatable astrocyte-targeted gene supplementation therapy for a disease that has no cure.

2.
Mol Ther ; 32(1): 44-58, 2024 Jan 03.
Article in English | MEDLINE | ID: mdl-37952085

ABSTRACT

Hematopoietic stem cell transplantation (HSCT) is the only approved treatment for presymptomatic infantile globoid cell leukodystrophy (GLD [Krabbe disease]). However, correction of disease is not complete, and outcomes remain poor. Herein we evaluated HSCT, intravenous (IV) adeno-associated virus rh10 vector (AAVrh10) gene therapy, and combination HSCT + IV AAVrh10 in the canine model of GLD. While HSCT alone resulted in no increase in survival as compared with untreated GLD dogs (∼16 weeks of age), combination HSCT + IV AAVrh10 at a dose of 4E13 genome copies (gc)/kg resulted in delayed disease progression and increased survival beyond 1 year of age. A 5-fold increase in AAVrh10 dose to 2E14 gc/kg, in combination with HSCT, normalized neurological dysfunction up to 2 years of age. IV AAVrh10 alone resulted in an average survival to 41.2 weeks of age. In the peripheral nervous system, IV AAVrh10 alone or in addition to HSCT normalized nerve conduction velocity, improved ultrastructure, and normalized GALC enzyme activity and psychosine concentration. In the central nervous system, only combination therapy at the highest dose was able to restore galactosylceramidase activity and psychosine concentrations to within the normal range. These data have now guided clinical translation of systemic AAV gene therapy as an addition to HSCT (NCT04693598, NCT05739643).


Subject(s)
Hematopoietic Stem Cell Transplantation , Leukodystrophy, Globoid Cell , Dogs , Animals , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/therapy , Galactosylceramidase/genetics , Psychosine , Hematopoietic Stem Cell Transplantation/methods , Genetic Therapy/methods , Disease Models, Animal
4.
Ann Neurol ; 94(5): 969-986, 2023 11.
Article in English | MEDLINE | ID: mdl-37526361

ABSTRACT

OBJECTIVE: GM2 gangliosidosis is usually fatal by 5 years of age in its 2 major subtypes, Tay-Sachs and Sandhoff disease. First reported in 1881, GM2 gangliosidosis has no effective treatment today, and children succumb to the disease after a protracted neurodegenerative course and semi-vegetative state. This study seeks to further develop adeno-associated virus (AAV) gene therapy for human translation. METHODS: Cats with Sandhoff disease were treated by intracranial injection of vectors expressing feline ß-N-acetylhexosaminidase, the enzyme deficient in GM2 gangliosidosis. RESULTS: Hexosaminidase activity throughout the brain and spinal cord was above normal after treatment, with highest activities at the injection sites (thalamus and deep cerebellar nuclei). Ganglioside storage was reduced throughout the brain and spinal cord, with near complete clearance in many regions. While untreated cats with Sandhoff disease lived for 4.4 ± 0.6 months, AAV-treated cats lived to 19.1 ± 8.6 months, and 3 of 9 cats lived >21 months. Correction of the central nervous system was so effective that significant increases in lifespan led to the emergence of otherwise subclinical peripheral disease, including megacolon, enlarged stomach and urinary bladder, soft tissue spinal cord compression, and patellar luxation. Throughout the gastrointestinal tract, neurons of the myenteric and submucosal plexuses developed profound pathology, demonstrating that the enteric nervous system was inadequately treated. INTERPRETATION: The vector formulation in the current study effectively treats neuropathology in feline Sandhoff disease, but whole-body targeting will be an important consideration in next-generation approaches. ANN NEUROL 2023;94:969-986.


Subject(s)
Gangliosidoses, GM2 , Sandhoff Disease , Child , Animals , Cats , Humans , Sandhoff Disease/genetics , Sandhoff Disease/therapy , Sandhoff Disease/veterinary , Multiple Organ Failure/therapy , Genetic Vectors , Central Nervous System/pathology , Genetic Therapy
5.
J Exp Med ; 220(9)2023 09 04.
Article in English | MEDLINE | ID: mdl-37310382

ABSTRACT

Globoid cell leukodystrophy (GLD) or Krabbe's disease is a fatal genetic demyelinating disease of the central nervous system caused by loss-of-function mutations in the galactosylceramidase (galc) gene. While the metabolic basis for disease is known, the understanding of how this results in neuropathology is not well understood. Herein, we report that the rapid and protracted elevation of CD8+ cytotoxic T lymphocytes occurs coincident with clinical disease in a mouse model of GLD. Administration of a function-blocking antibody against CD8α effectively prevented disease onset, reduced morbidity and mortality, and prevented CNS demyelination in mice. These data indicate that subsequent to the genetic cause of disease, neuropathology is driven by pathogenic CD8+ T cells, thus offering novel therapeutic potential for treatment of GLD.


Subject(s)
Leukodystrophy, Globoid Cell , Animals , Mice , Leukodystrophy, Globoid Cell/genetics , Central Nervous System , Disease Models, Animal , Antibodies, Blocking , CD8-Positive T-Lymphocytes
6.
Mol Ther ; 31(1): 7-23, 2023 01 04.
Article in English | MEDLINE | ID: mdl-36196048

ABSTRACT

Krabbe disease (KD) is a lysosomal storage disease (LSD) caused by mutations in the galc gene. There are over 50 monogenetic LSDs, which largely impede the normal development of children and often lead to premature death. At present, there are no cures for LSDs and the available treatments are generally insufficient, short acting, and not without co-morbidities or long-term side effects. The last 30 years have seen significant advances in our understanding of LSD pathology as well as treatment options. Two gene therapy-based clinical trials, NCT04693598 and NCT04771416, for KD were recently started based on those advances. This review will discuss how our knowledge of KD got to where it is today, focusing on preclinical investigations, and how what was discovered may prove beneficial for the treatment of other LSDs.


Subject(s)
Leukodystrophy, Globoid Cell , Lysosomal Storage Diseases , Child , Humans , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/therapy , Leukodystrophy, Globoid Cell/pathology , Combined Modality Therapy , Mutation , Lysosomal Storage Diseases/genetics , Lysosomal Storage Diseases/therapy
7.
Mol Ther ; 30(7): 2416-2428, 2022 07 06.
Article in English | MEDLINE | ID: mdl-35585789

ABSTRACT

We are in an emerging era of gene-based therapeutics with significant promise for rare genetic disorders. The potential is particularly significant for genetic central nervous system disorders that have begun to achieve Food and Drug Administration approval for select patient populations. This review summarizes the discussions and presentations of the National Institute of Mental Health-sponsored workshop "Gene-Based Therapeutics for Rare Genetic Neurodevelopmental Psychiatric Disorders," which was held in January 2021. Here, we distill the points raised regarding various precision medicine approaches related to neurodevelopmental and psychiatric disorders that may be amenable to gene-based therapies.


Subject(s)
Mental Disorders , Precision Medicine , Humans , Mental Disorders/genetics , Mental Disorders/psychology , Mental Disorders/therapy , Rare Diseases , United States , United States Food and Drug Administration
8.
Hum Gene Ther ; 33(9-10): 499-517, 2022 05.
Article in English | MEDLINE | ID: mdl-35333110

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.


Subject(s)
Leukodystrophy, Globoid Cell , Animals , Child, Preschool , Dependovirus/genetics , Disease Models, Animal , Dogs , Genetic Therapy , Humans , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/therapy , Macaca mulatta/genetics , Mice , Psychosine
9.
Semin Pediatr Neurol ; 37: 100876, 2021 04.
Article in English | MEDLINE | ID: mdl-33892849

ABSTRACT

Leukodystrophies and genetic leukoencephalopathies comprise a growing group of inherited white matter disorders. Diagnostic rates have improved with increased utilization of next generation sequencing. As treatment options continue to advance for leukodystrophies, so will candidacy for inclusion in the United States' newborn Recommended Universal Screening Panel as was achieved for X-linked adrenoleukodystrophy. Stem cell therapies have become standard of care for selected leukodystrophies. However, transplantation-related risks remain high and outcomes are not fully satisfactory. Transduction of autologous hematopoietic stem cells with lentiviral vectors, referred to as ex vivo gene therapy, circumvents some, but not all, of the risks of traditional transplantation and has recently been demonstrated to be safe and efficective in clinical studies of X-linked adrenoleukodystrophy and metachromatic leukodystrophy. Gene therapy, through direct infusion of adeno-associated virus vectors, has emerged as a safer alternative for many monogenetic pediatric neurological disorders. Numerous preclinical studies have shown safety and efficacy of adeno-associated virus gene therapy in leukodystrophies allowing expanded access treatment for Canavan disease prior to initiation of a clinical trial. For inherited white matter disorders resulting from overexpression of a protein, such as Pelizaeus-Merzbacher disease, emerging RNA therapies have shown success in preclinical studies and promise for rapid translation to the clinic. Lastly, small molecule and protein therapies remain a long-term treatment option for a number of leukodystrophies, including intrathecal enzyme replacement therapy for metachromatic leukodystrophy. Herein we review recent advances in diagnosis and treatment of inherited white matter disorders.


Subject(s)
Demyelinating Diseases , Leukodystrophy, Metachromatic , Leukoencephalopathies , Neurodegenerative Diseases , Child , Genetic Therapy , Humans , Leukodystrophy, Metachromatic/diagnosis , Leukodystrophy, Metachromatic/genetics , Leukodystrophy, Metachromatic/therapy , Leukoencephalopathies/therapy
10.
Neurosci Lett ; 752: 135841, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33766733

ABSTRACT

Krabbe disease (globoid cell leukodystrophy) is a lysosomal storage disease (LSD) characterized by progressive and profound demyelination. Infantile, juvenile and adult-onset forms of Krabbe disease have been described, with infantile being the most common. Children with an infantile-onset generally appear normal at birth but begin to miss developmental milestones by six months of age and die by two to four years of age. Krabbe disease is caused by a deficiency of the acid hydrolase galactosylceramidase (GALC) which is responsible for the degradation of galactosylceramides and sphingolipids, which are abundant in myelin membranes. The absence of GALC leads to the toxic accumulation of galactosylsphingosine (psychosine), a lysoderivative of galactosylceramides, in oligodendrocytes and Schwann cells resulting in demyelination of the central and peripheral nervous systems, respectively. Treatment strategies such as enzyme replacement, substrate reduction, enzyme chaperones, and gene therapy have shown promise in LSDs. Unfortunately, Krabbe disease has been relatively refractory to most single-therapy interventions. Although hematopoietic stem cell transplantation can alter the course of Krabbe disease and is the current standard-of-care, it simply slows the progression, even when initiated in pre-symptomatic children. However, the recent success of combinatorial therapeutic approaches in small animal models of Krabbe disease and the identification of new pathogenic mechanisms provide hope for the development of effective treatments for this devastating disease. This review provides a brief history of Krabbe disease and the evolution of single and combination therapeutic approaches and discusses new pathogenic mechanisms and how they might impact the development of more effective treatment strategies.


Subject(s)
Galactosylceramidase/deficiency , Leukodystrophy, Globoid Cell/therapy , Animals , Combined Modality Therapy/methods , Disease Models, Animal , Enzyme Replacement Therapy/methods , Galactosylceramidase/genetics , Galactosylceramides/metabolism , Genetic Therapy/methods , Hematopoietic Stem Cell Transplantation/methods , Humans , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/pathology , Myelin Sheath/metabolism , Myelin Sheath/pathology , Sphingolipids/metabolism
11.
Gene Ther ; 28(3-4): 142-154, 2021 04.
Article in English | MEDLINE | ID: mdl-32884151

ABSTRACT

Sandhoff disease (SD) is an autosomal recessive lysosomal storage disease caused by defects in the ß-subunit of ß-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.


Subject(s)
Sandhoff Disease , Animals , Cats , Dependovirus/genetics , Disease Models, Animal , Genetic Therapy , Genetic Vectors/genetics , Humans , Sandhoff Disease/genetics , Sandhoff Disease/therapy , beta-N-Acetylhexosaminidases/genetics
12.
J Clin Invest ; 130(9): 4906-4920, 2020 09 01.
Article in English | MEDLINE | ID: mdl-32773406

ABSTRACT

Globoid cell leukodystrophy (GLD; Krabbe disease) is a progressive, incurable neurodegenerative disease caused by deficient activity of the hydrolytic enzyme galactosylceramidase (GALC). The ensuing cytotoxic accumulation of psychosine results in diffuse central and peripheral nervous system (CNS, PNS) demyelination. Presymptomatic hematopoietic stem cell transplantation (HSCT) is the only treatment for infantile-onset GLD; however, clinical outcomes of HSCT recipients often remain poor, and procedure-related morbidity is high. There are no effective therapies for symptomatic patients. Herein, we demonstrate in the naturally occurring canine model of GLD that presymptomatic monotherapy with intrathecal AAV9 encoding canine GALC administered into the cisterna magna increased GALC enzyme activity, normalized psychosine concentration, improved myelination, and attenuated inflammation in both the CNS and PNS. Moreover, AAV-mediated therapy successfully prevented clinical neurological dysfunction, allowing treated dogs to live beyond 2.5 years of age, more than 7 times longer than untreated dogs. Furthermore, we found that a 5-fold lower dose resulted in an attenuated form of disease, indicating that sufficient dosing is critical. Finally, postsymptomatic therapy with high-dose AAV9 also significantly extended lifespan, signifying a treatment option for patients for whom HSCT is not applicable. If translatable to patients, these findings would improve the outcomes of patients treated either pre- or postsymptomatically.


Subject(s)
Dependovirus , Galactosylceramidase , Genetic Therapy , Leukodystrophy, Globoid Cell , Animals , Disease Models, Animal , Dogs , Galactosylceramidase/biosynthesis , Galactosylceramidase/genetics , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/metabolism , Leukodystrophy, Globoid Cell/pathology , Leukodystrophy, Globoid Cell/therapy
13.
Front Vet Sci ; 7: 80, 2020.
Article in English | MEDLINE | ID: mdl-32219101

ABSTRACT

Mouse models of human disease remain the bread and butter of modern biology and therapeutic discovery. Nonetheless, more often than not mouse models do not reproduce the pathophysiology of the human conditions they are designed to mimic. Naturally occurring large animal models have predominantly been found in companion animals or livestock because of their emotional or economic value to modern society and, unlike mice, often recapitulate the human disease state. In particular, numerous models have been discovered in dogs and have a fundamental role in bridging proof of concept studies in mice to human clinical trials. The present article is a review that highlights current canine models of human diseases, including Alzheimer's disease, degenerative myelopathy, neuronal ceroid lipofuscinosis, globoid cell leukodystrophy, Duchenne muscular dystrophy, mucopolysaccharidosis, and fucosidosis. The goal of the review is to discuss canine and human neurodegenerative pathophysiologic similarities, introduce the animal models, and shed light on the ability of canine models to facilitate current and future treatment trials.

14.
Mol Cell Neurosci ; 102: 103451, 2020 01.
Article in English | MEDLINE | ID: mdl-31794880

ABSTRACT

Globoid cell leukodystrophy (GLD, Krabbe disease, Krabbe's disease) is caused by genetic mutations in the gene encoding, galactosylceramidase (GALC). Deficiency of this enzyme results in central and peripheral nervous system pathology, and is characterized by loss of myelin and an infiltration of globoid cells. The canine model of GLD provides a translational model which faithfully recapitulates much of the human disease pathology. Targeted lipidomic analysis was conducted in serum and cerebrospinal fluid (CSF) over the lifetime of GLD affected and normal canines, and in brain tissue at humane endpoint to better understand disease progression and identify potential biomarkers of disease. Psychosine, a substrate of GALC and primary contributor to the pathology in GLD, was observed to be significantly elevated in the serum and CSF by 2 or 4 weeks of age, respectively, and steadily increased over the lifetime of affected animals. Importantly, psychosine concentration strongly correlated with disease severity. Galactosylceramide, glucosylceramide, and lactosylceramide were also found to be elevated in the CSF of affected animals and increased with age. Psychosine and galactosylceramide were found to be significantly increased in brain tissue at humane endpoint. This study identified several biomarkers which may be useful in the development of therapeutics for GLD.


Subject(s)
Dog Diseases/cerebrospinal fluid , Galactosylceramides/blood , Galactosylceramides/cerebrospinal fluid , Leukodystrophy, Globoid Cell/veterinary , Psychosine/cerebrospinal fluid , Animals , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Dog Diseases/blood , Dog Diseases/pathology , Dogs , Female , Leukodystrophy, Globoid Cell/blood , Leukodystrophy, Globoid Cell/cerebrospinal fluid , Leukodystrophy, Globoid Cell/pathology , Male , Psychosine/blood
15.
Hum Gene Ther ; 29(7): 785-801, 2018 07.
Article in English | MEDLINE | ID: mdl-29316812

ABSTRACT

Globoid cell leukodystrophy (GLD), or Krabbe disease, is an inherited, neurologic disorder that results from deficiency of a lysosomal enzyme, galactosylceramidase. Most commonly, deficits of galactosylceramidase result in widespread central and peripheral nervous system demyelination and death in affected infants typically by 2 years of age. Hematopoietic stem-cell transplantation is the current standard of care in children diagnosed prior to symptom onset. However, disease correction is incomplete. Herein, the first adeno-associated virus (AAV) gene therapy experiments are presented in a naturally occurring canine model of GLD that closely recapitulates the clinical disease progression, neuropathological alterations, and biochemical abnormalities observed in human patients. Adapted from studies in twitcher mice, GLD dogs were treated by combination intravenous and intracerebroventricular injections of AAVrh10 to target both the peripheral and central nervous systems. Combination of intravenous and intracerebroventricular AAV gene therapy had a clear dose response and resulted in delayed onset of clinical signs, extended life-span, correction of biochemical defects, and attenuation of neuropathology. For the first time, therapeutic effect has been established in the canine model of GLD by targeting both peripheral and central nervous system impairments with potential clinical implications for GLD patients.


Subject(s)
Galactosylceramidase/administration & dosage , Genetic Therapy , Leukodystrophy, Globoid Cell/therapy , Peripheral Nervous System Diseases/therapy , Animals , Brain/drug effects , Central Nervous System/metabolism , Central Nervous System/pathology , Dependovirus/genetics , Disease Models, Animal , Dogs , Galactosylceramidase/genetics , Genetic Vectors/administration & dosage , Humans , Infant , Leukodystrophy, Globoid Cell/genetics , Leukodystrophy, Globoid Cell/pathology , Peripheral Nervous System Diseases/genetics , Peripheral Nervous System Diseases/pathology
16.
Hum Gene Ther ; 29(3): 312-326, 2018 03.
Article in English | MEDLINE | ID: mdl-28922945

ABSTRACT

Tay-Sachs disease (TSD) is a fatal neurodegenerative disorder caused by a deficiency of the enzyme hexosaminidase A (HexA). TSD also occurs in sheep, the only experimental model of TSD that has clinical signs of disease. The natural history of sheep TSD was characterized using serial neurological evaluations, 7 Tesla magnetic resonance imaging, echocardiograms, electrodiagnostics, and cerebrospinal fluid biomarkers. Intracranial gene therapy was also tested using AAVrh8 monocistronic vectors encoding the α-subunit of Hex (TSD α) or a mixture of two vectors encoding both the α and ß subunits separately (TSD α + ß) injected at high (1.3 × 1013 vector genomes) or low (4.2 × 1012 vector genomes) dose. Delay of symptom onset and/or reduction of acquired symptoms were noted in all adeno-associated virus-treated sheep. Postmortem evaluation showed superior HexA and vector genome distribution in the brain of TSD α + ß sheep compared to TSD α sheep, but spinal cord distribution was low in all groups. Isozyme analysis showed superior HexA formation after treatment with both vectors (TSD α + ß), and ganglioside clearance was most widespread in the TSD α + ß high-dose sheep. Microglial activation and proliferation in TSD sheep-most prominent in the cerebrum-were attenuated after gene therapy. This report demonstrates therapeutic efficacy for TSD in the sheep brain, which is on the same order of magnitude as a child's brain.


Subject(s)
Dependovirus , Genetic Therapy , Tay-Sachs Disease/therapy , beta-Hexosaminidase alpha Chain/biosynthesis , beta-Hexosaminidase beta Chain/biosynthesis , Animals , Brain/diagnostic imaging , Brain/enzymology , Disease Models, Animal , Echocardiography , Humans , Magnetic Resonance Imaging , Microglia/enzymology , Sheep , Tay-Sachs Disease/diagnostic imaging , Tay-Sachs Disease/enzymology , Tay-Sachs Disease/genetics , beta-Hexosaminidase alpha Chain/genetics , beta-Hexosaminidase beta Chain/genetics
17.
Yale J Biol Med ; 90(3): 417-431, 2017 09.
Article in English | MEDLINE | ID: mdl-28955181

ABSTRACT

For many lethal or debilitating genetic disorders in patients there are no satisfactory therapies. Several barriers exist that hinder the developments of effective therapies including the limited availability of clinically relevant animal models that faithfully recapitulate human genetic disease. In 1974, the Referral Center for Animal Models of Human Genetic Disease (RCAM) was established by Dr. Donald F. Patterson and continued by Dr. Mark E. Haskins at the University of Pennsylvania with the mission to discover, understand, treat, and maintain breeding colonies of naturally occurring hereditary disorders in dogs and cats that are orthologous to those found in human patients. Although non-human primates, sheep, and pig models are also available within the medical community, naturally occurring diseases are rarely identified in non-human primates, and the vast behavioral, clinicopathological, physiological, and anatomical knowledge available regarding dogs and cats far surpasses what is available in ovine and porcine species. The canine and feline models that are maintained at RCAM are presented here with a focus on preclinical therapy data. Clinical studies that have been generated from preclinical work in these models are also presented.


Subject(s)
Genetic Diseases, Inborn , Rare Diseases , Animals , Cats , Disease Models, Animal , Dogs , Humans , Sheep , Swine
18.
Mol Ther ; 25(4): 892-903, 2017 04 05.
Article in English | MEDLINE | ID: mdl-28236574

ABSTRACT

GM1 gangliosidosis is a fatal neurodegenerative disease that affects individuals of all ages. Favorable outcomes using adeno-associated viral (AAV) gene therapy in GM1 mice and cats have prompted consideration of human clinical trials, yet there remains a paucity of objective biomarkers to track disease status. We developed a panel of biomarkers using blood, urine, cerebrospinal fluid (CSF), electrodiagnostics, 7 T MRI, and magnetic resonance spectroscopy in GM1 cats-either untreated or AAV treated for more than 5 years-and compared them to markers in human GM1 patients where possible. Significant alterations were noted in CSF and blood of GM1 humans and cats, with partial or full normalization after gene therapy in cats. Gene therapy improved the rhythmic slowing of electroencephalograms (EEGs) in GM1 cats, a phenomenon present also in GM1 patients, but nonetheless the epileptiform activity persisted. After gene therapy, MR-based analyses revealed remarkable preservation of brain architecture and correction of brain metabolites associated with microgliosis, neuroaxonal loss, and demyelination. Therapeutic benefit of AAV gene therapy in GM1 cats, many of which maintain near-normal function >5 years post-treatment, supports the strong consideration of human clinical trials, for which the biomarkers described herein will be essential for outcome assessment.


Subject(s)
Biomarkers , Gangliosidosis, GM1/genetics , Gangliosidosis, GM1/metabolism , Genetic Therapy , Animals , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Biomarkers/urine , Cats , Dependovirus/classification , Dependovirus/genetics , Disease Models, Animal , Electroencephalography , Gangliosidosis, GM1/mortality , Gangliosidosis, GM1/therapy , Genetic Therapy/methods , Genetic Vectors/administration & dosage , Genetic Vectors/genetics , Humans , Hypocalcemia/metabolism , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy , Treatment Outcome
19.
Hum Gene Ther ; 28(6): 510-522, 2017 06.
Article in English | MEDLINE | ID: mdl-28132521

ABSTRACT

GM2 gangliosidoses, including Tay-Sachs disease and Sandhoff disease, are lysosomal storage disorders caused by deficiencies in ß-N-acetylhexosaminidase (Hex). Patients are afflicted primarily with progressive central nervous system (CNS) dysfunction. Studies in mice, cats, and sheep have indicated safety and widespread distribution of Hex in the CNS after intracranial vector infusion of AAVrh8 vectors encoding species-specific Hex α- or ß-subunits at a 1:1 ratio. Here, a safety study was conducted in cynomolgus macaques (cm), modeling previous animal studies, with bilateral infusion in the thalamus as well as in left lateral ventricle of AAVrh8 vectors encoding cm Hex α- and ß-subunits. Three doses (3.2 × 1012 vg [n = 3]; 3.2 × 1011 vg [n = 2]; or 1.1 × 1011 vg [n = 2]) were tested, with controls infused with vehicle (n = 1) or transgene empty AAVrh8 vector at the highest dose (n = 2). Most monkeys receiving AAVrh8-cmHexα/ß developed dyskinesias, ataxia, and loss of dexterity, with higher dose animals eventually becoming apathetic. Time to onset of symptoms was dose dependent, with the highest-dose cohort producing symptoms within a month of infusion. One monkey in the lowest-dose cohort was behaviorally asymptomatic but had magnetic resonance imaging abnormalities in the thalami. Histopathology was similar in all monkeys injected with AAVrh8-cmHexα/ß, showing severe white and gray matter necrosis along the injection track, reactive vasculature, and the presence of neurons with granular eosinophilic material. Lesions were minimal to absent in both control cohorts. Despite cellular loss, a dramatic increase in Hex activity was measured in the thalamus, and none of the animals presented with antibody titers against Hex. The high overexpression of Hex protein is likely to blame for this negative outcome, and this study demonstrates the variations in safety profiles of AAVrh8-Hexα/ß intracranial injection among different species, despite encoding for self-proteins.


Subject(s)
Dependovirus/genetics , Dyskinesias/etiology , Gangliosidoses, GM2/therapy , Genetic Vectors/adverse effects , Necrosis/etiology , Neurons/metabolism , beta-N-Acetylhexosaminidases/genetics , Animals , Apathy , Dependovirus/metabolism , Disease Models, Animal , Dyskinesias/genetics , Dyskinesias/metabolism , Dyskinesias/pathology , Female , Gangliosidoses, GM2/genetics , Gangliosidoses, GM2/metabolism , Gangliosidoses, GM2/pathology , Gene Expression , Genetic Therapy/methods , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Gray Matter/metabolism , Gray Matter/pathology , Injections, Intraventricular , Macaca fascicularis , Male , Necrosis/genetics , Necrosis/metabolism , Necrosis/pathology , Neurons/pathology , Protein Subunits/adverse effects , Protein Subunits/genetics , Protein Subunits/metabolism , Thalamus/metabolism , Thalamus/pathology , Transgenes , White Matter/metabolism , White Matter/pathology , beta-N-Acetylhexosaminidases/adverse effects , beta-N-Acetylhexosaminidases/metabolism
20.
Neuroscience ; 340: 117-125, 2017 01 06.
Article in English | MEDLINE | ID: mdl-27793778

ABSTRACT

Sandhoff disease (SD) is a lysosomal storage disorder characterized by the absence of hydrolytic enzyme ß-N-acetylhexosaminidase (Hex), which results in storage of GM2 ganglioside in neurons and unremitting neurodegeneration. Neuron loss initially affects fine motor skills, but rapidly progresses to loss of all body faculties, a vegetative state, and death by five years of age in humans. A well-established feline model of SD allows characterization of the disease in a large animal model and provides a means to test the safety and efficacy of therapeutic interventions before initiating clinical trials. In this study, we demonstrate a robust central nervous system (CNS) inflammatory response in feline SD, primarily marked by expansion and activation of the microglial cell population. Quantification of major histocompatibility complex II (MHC-II) labeling revealed significant up-regulation throughout the CNS with areas rich in white matter most severely affected. Expression of the leukocyte chemokine macrophage inflammatory protein-1 alpha (MIP-1α) was also up-regulated in the brain. SD cats were treated with intracranial delivery of adeno-associated viral (AAV) vectors expressing feline Hex, with a study endpoint 16weeks post treatment. AAV-mediated gene delivery repressed the expansion and activation of microglia and normalized MHC-II and MIP-1α levels. These data reiterate the profound inflammatory response in SD and show that neuroinflammation is abrogated after AAV-mediated restoration of enzymatic activity.


Subject(s)
Brain/immunology , Genetic Therapy , Sandhoff Disease/immunology , Sandhoff Disease/therapy , Adaptor Proteins, Signal Transducing/metabolism , Animals , Astrocytes/immunology , Astrocytes/pathology , Brain/pathology , Cats , Dependovirus/genetics , Disease Models, Animal , Genes, MHC Class II/physiology , Genetic Vectors , Gliosis/immunology , Gliosis/pathology , Gliosis/therapy , Immunohistochemistry , Microglia/immunology , Microglia/pathology , Neurons/immunology , Neurons/pathology , Polymerase Chain Reaction , Sandhoff Disease/pathology , Transcription Factors/genetics , Transcription Factors/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...