Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 14 de 14
Filter
Add more filters










Publication year range
1.
EBioMedicine ; 99: 104894, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38086156

ABSTRACT

BACKGROUND: X-linked myotubular myopathy (XLMTM) is a rare, life-threatening congenital muscle disease caused by mutations in the MTM1 gene that result in profound muscle weakness, significant respiratory insufficiency, and high infant mortality. There is no approved disease-modifying therapy for XLMTM. Resamirigene bilparvovec (AT132; rAAV8-Des-hMTM1) is an investigational adeno-associated virus (AAV8)-mediated gene replacement therapy designed to deliver MTM1 to skeletal muscle cells and achieve long-term correction of XLMTM-related muscle pathology. The clinical trial ASPIRO (NCT03199469) investigating resamirigene bilparvovec in XLMTM is currently paused while the risk:benefit balance associated with this gene therapy is further investigated. METHODS: Muscle biopsies were taken before treatment and 24 and 48 weeks after treatment from ten boys with XLMTM in a clinical trial of resamirigene bilparvovec (ASPIRO; NCT03199469). Comprehensive histopathological analysis was performed. FINDINGS: Baseline biopsies uniformly showed findings characteristic of XLMTM, including small myofibres, increased internal or central nucleation, and central aggregates of organelles. Biopsies taken at 24 weeks post-treatment showed marked improvement of organelle localisation, without apparent increases in myofibre size in most participants. Biopsies taken at 48 weeks, however, did show statistically significant increases in myofibre size in all nine biopsies evaluated at this timepoint. Histopathological endpoints that did not demonstrate statistically significant changes with treatment included the degree of internal/central nucleation, numbers of triad structures, fibre type distributions, and numbers of satellite cells. Limited (predominantly mild) treatment-associated inflammatory changes were seen in biopsy specimens from five participants. INTERPRETATION: Muscle biopsies from individuals with XLMTM treated with resamirigene bilparvovec display statistically significant improvement in organelle localisation and myofibre size during a period of substantial improvements in muscle strength and respiratory function. This study identifies valuable histological endpoints for tracking treatment-related gains with resamirigene bilparvovec, as well as endpoints that did not show strong correlation with clinical improvement in this human study. FUNDING: Astellas Gene Therapies (formerly Audentes Therapeutics, Inc.).


Subject(s)
Muscle, Skeletal , Myopathies, Structural, Congenital , Male , Infant , Humans , Muscle, Skeletal/pathology , Genetic Therapy/adverse effects , Genetic Therapy/methods , Muscle Weakness , Muscle Strength , Myopathies, Structural, Congenital/genetics , Myopathies, Structural, Congenital/therapy , Myopathies, Structural, Congenital/pathology
2.
JCI Insight ; 8(11)2023 06 08.
Article in English | MEDLINE | ID: mdl-37288657

ABSTRACT

Fragile X syndrome is a neurodevelopmental disorder caused by the absence of the mRNA-binding protein fragile X messenger ribonucleoprotein (FMRP). Because FMRP is a highly pleiotropic protein controlling the expression of hundreds of genes, viral vector-mediated gene replacement therapy is viewed as a potential viable treatment to correct the fundamental underlying molecular pathology inherent in the disorder. Here, we studied the safety profile and therapeutic effects of a clinically relevant dose of a self-complementary adeno-associated viral (AAV) vector containing a major human brain isoform of FMRP after intrathecal injection into wild-type and fragile X-KO mice. Analysis of the cellular transduction in the brain indicated primarily neuronal transduction with relatively sparse glial expression, similar to endogenous FMRP expression in untreated wild-type mice. AAV vector-treated KO mice showed recovery from epileptic seizures, normalization of fear conditioning, reversal of slow-wave deficits as measured via electroencephalographic recordings, and restoration of abnormal circadian motor activity and sleep. Further assessment of vector efficacy by tracking and analyzing individual responses demonstrated correlations between the level and distribution of brain transduction and drug response. These preclinical findings further demonstrate the validity of AAV vector-mediated gene therapy for treating the most common genetic cause of cognitive impairment and autism in children.


Subject(s)
Fear , Fragile X Mental Retardation Protein , Animals , Humans , Mice , Fragile X Mental Retardation Protein/genetics , Mice, Knockout , Protein Isoforms/genetics , Protein Isoforms/metabolism , Seizures/genetics , Seizures/therapy
3.
Hum Gene Ther ; 34(3-4): 94-111, 2023 02.
Article in English | MEDLINE | ID: mdl-36606687

ABSTRACT

Adeno-associated virus (AAV)-based gene therapies, exemplified by the approved therapy for spinal muscular atrophy, have the potential to deliver disease-course-altering treatments for central nervous system (CNS) indications. However, several clinical trials have reported severe adverse events, including patient deaths following high-dose systemic administration for muscle-directed gene transfer, highlighting the need to explore approaches utilizing lower doses when targeting the CNS. Animal models of disease provide insight into the response to new AAV therapies. However, translation from small to larger animals and eventually to humans is hampered by anatomical and biological differences across the species and their impact on AAV delivery. We performed a literature review of preclinical studies of AAV gene therapy biodistribution following cerebrospinal fluid (CSF) delivery (intracerebroventricular, intra-cisterna magna, and intrathecal lumbar). The reviewed literature varies greatly in the reported biodistribution of AAV following administration into the CSF. Differences between studies, including animal model, vector serotype used, method used to assess biodistribution, and route of administration, among other variables, contribute to differing outcomes and difficulties in translating these preclinical results. For example, only half of the published AAV-based gene therapy studies report vector copy number, the most direct readout following administration of a vector; none of these studies reported details such as the empty:full capsid ratio and quality of encapsidated genome. Analysis of the last decade's literature focusing on AAV-based gene therapies targeting the CNS underscores limitations of the body of knowledge and room for continued research. In particular, there is a need to understand the biodistribution achieved by different CSF-directed routes of administration and determining if specific cell types/structures of interest will be transduced. Our findings point to a clear need for a more systematic approach across the field to align the assessments and elements reported in preclinical research to enable more reliable translation across animal models and into human studies.


Subject(s)
Dependovirus , Genetic Therapy , Animals , Humans , Dependovirus/genetics , Tissue Distribution , Genetic Therapy/methods , Central Nervous System , Models, Animal , Genetic Vectors/genetics , Gene Transfer Techniques
4.
Genes (Basel) ; 15(1)2023 Dec 24.
Article in English | MEDLINE | ID: mdl-38254921

ABSTRACT

Inactivating mutations and the duplication of methyl-CpG binding protein 2 (MeCP2), respectively, mediate Rett syndrome (RTT) and MECP2 duplication syndrome. These disorders underscore the conceptual dose-dependent risk posed by MECP2 gene therapy for mosaic RTT patients. Recently, a miRNA-Responsive Autoregulatory Element (miRARE) mitigated the dose-dependent toxicity posed by self-complementary adeno-associated viral vector serotype 9 (AAV9) miniMECP2 gene therapy (scAAV9/miniMECP2-myc) in mice. Here, we report an efficacy assessment for the human-ready version of this regulated gene therapy (TSHA-102) in male Mecp2-/y knockout (KO) mice after intracerebroventricular (ICV) administration at postnatal day 2 (P2) and after intrathecal (IT) administration at P7, P14 (±immunosuppression), and P28 (±immunosuppression). We also report qPCR studies on KO mice treated at P7-P35; protein analyses in KO mice treated at P38; and a survival safety study in female adult Mecp2-/+ mice. In KO mice, TSHA-102 improved respiration, weight, and survival across multiple doses and treatment ages. TSHA-102 significantly improved the front average stance and swing times relative to the front average stride time after P14 administration of the highest dose for that treatment age. Viral genomic DNA and miniMECP2 mRNA were present in the CNS. MiniMeCP2 protein expression was higher in the KO spinal cord compared to the brain. In female mice, TSHA-102 permitted survivals that were similar to those of vehicle-treated controls. In all, these pivotal data helped to support the regulatory approval to initiate a clinical trial for TSHA-102 in RTT patients (clinical trial identifier number NCT05606614).


Subject(s)
Mental Retardation, X-Linked , MicroRNAs , Rett Syndrome , Adult , Humans , Female , Male , Animals , Mice , Rett Syndrome/genetics , Rett Syndrome/therapy , Brain , DNA, Viral , Genetic Therapy , Mice, Knockout
5.
Genes (Basel) ; 13(9)2022 09 15.
Article in English | MEDLINE | ID: mdl-36140822

ABSTRACT

Epileptic encephalopathies may arise from single gene variants. In recent years, next-generation sequencing technologies have enabled an explosion of gene identification in monogenic epilepsies. One such example is the epileptic encephalopathy SLC13A5 deficiency disorder, which is caused by loss of function pathogenic variants to the gene SLC13A5 that results in deficiency of the sodium/citrate cotransporter. Patients typically experience seizure onset within the first week of life and have developmental delay and intellectual disability. Current antiseizure medications may reduce seizure frequency, yet more targeted treatments are needed to address the epileptic and non-epileptic features of SLC13A5 deficiency disorder. Gene therapy may offer hope to these patients and better clinical outcomes than current available treatments. Here, we discuss SLC13A5 genetics, natural history, available treatments, potential outcomes and assessments, and considerations for translational medical research for an AAV9-based gene replacement therapy.


Subject(s)
Epilepsy , Symporters , Citrates , Epilepsy/genetics , Epilepsy/therapy , Genetic Therapy , Humans , Mutation , Seizures/genetics , Seizures/therapy , Sodium , Spasms, Infantile , Symporters/genetics
6.
Hum Gene Ther ; 33(23-24): 1228-1245, 2022 12.
Article in English | MEDLINE | ID: mdl-35994385

ABSTRACT

Adeno-associated viruses (AAVs) are being increasingly used as gene therapy vectors in clinical studies especially targeting central nervous system (CNS) disorders. Correspondingly, host immune responses to the AAV capsid or the transgene-encoded protein have been observed in various clinical and preclinical studies. Such immune responses may adversely impact patients' health, prevent viral transduction, prevent repeated dosing strategies, eliminate transduced cells, and pose a significant barrier to the potential effectiveness of AAV gene therapy. Consequently, multiple immunomodulatory strategies have been used in attempts to limit immune-mediated responses to the vector, enable readministration of AAV gene therapy, prevent end-organ toxicity, and increase the duration of transgene-encoded protein expression. Herein we review the innate and adaptive immune responses that may occur during CNS-targeted AAV gene therapy as well as host- and treatment-specific factors that could impact the immune response. We also summarize the available preclinical and clinical data on immune responses specifically to CNS-targeted AAV gene therapy and discuss potential strategies for incorporating prophylactic immunosuppression regimens to circumvent adverse immune responses.


Subject(s)
Central Nervous System Diseases , Dependovirus , Humans , Dependovirus/genetics , Genetic Vectors/genetics , Genetic Vectors/therapeutic use , Genetic Therapy , Immunity, Humoral , Immunosuppressive Agents , Central Nervous System Diseases/genetics , Central Nervous System Diseases/therapy
7.
Front Neurol ; 13: 805007, 2022.
Article in English | MEDLINE | ID: mdl-35847198

ABSTRACT

Genetic epilepsies are a spectrum of disorders characterized by spontaneous and recurrent seizures that can arise from an array of inherited or de novo genetic variants and disrupt normal brain development or neuronal connectivity and function. Genetically determined epilepsies, many of which are due to monogenic pathogenic variants, can result in early mortality and may present in isolation or be accompanied by neurodevelopmental disability. Despite the availability of more than 20 antiseizure medications, many patients with epilepsy fail to achieve seizure control with current therapies. Patients with refractory epilepsy-particularly of childhood onset-experience increased risk for severe disability and premature death. Further, available medications inadequately address the comorbid developmental disability. The advent of next-generation gene sequencing has uncovered genetic etiologies and revolutionized diagnostic practices for many epilepsies. Advances in the field of gene therapy also present the opportunity to address the underlying mechanism of monogenic epilepsies, many of which have only recently been described due to advances in precision medicine and biology. To bring precision medicine and genetic therapies closer to clinical applications, experimental animal models are needed that replicate human disease and reflect the complexities of these disorders. Additionally, identifying and characterizing clinical phenotypes, natural disease course, and meaningful outcome measures from epileptic and neurodevelopmental perspectives are necessary to evaluate therapies in clinical studies. Here, we discuss the range of genetically determined epilepsies, the existing challenges to effective clinical management, and the potential role gene therapy may play in transforming treatment options available for these conditions.

8.
EMBO Mol Med ; 14(1): e13968, 2022 01 11.
Article in English | MEDLINE | ID: mdl-34850579

ABSTRACT

Pompe disease is a severe disorder caused by loss of acid α-glucosidase (GAA), leading to glycogen accumulation in tissues and neuromuscular and cardiac dysfunction. Enzyme replacement therapy is the only available treatment. AT845 is an adeno-associated viral vector designed to express human GAA specifically in skeletal muscle and heart. Systemic administration of AT845 in Gaa-/- mice led to a dose-dependent increase in GAA activity, glycogen clearance in muscles and heart, and functional improvement. AT845 was tolerated in cynomolgus macaques at low doses, while high doses caused anti-GAA immune response, inflammation, and cardiac abnormalities resulting in unscheduled euthanasia of two animals. Conversely, a vector expressing the macaque GAA caused no detectable pathology, indicating that the toxicity observed with AT845 was an anti-GAA xenogeneic immune response. Western blot analysis showed abnormal processing of human GAA in cynomolgus muscle, adding to the species-specific effects of enzyme expression. Overall, these studies show that AAV-mediated GAA delivery to muscle is efficacious in Gaa-/- mice and highlight limitations in predicting the toxicity of AAV vectors encoding human proteins in non-human species.


Subject(s)
Glycogen Storage Disease Type II , Animals , Dependovirus/genetics , Genetic Therapy/methods , Genetic Vectors , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Mice , Mice, Knockout , Muscle, Skeletal/metabolism , alpha-Glucosidases/genetics , alpha-Glucosidases/metabolism
9.
Bioanalysis ; 7(3): 373-81, 2015.
Article in English | MEDLINE | ID: mdl-25697194

ABSTRACT

An appropriate assessment strategy with validated anti-drug antibody (ADA) assays is critical for comparative evaluation of immunogenicity between a proposed biosimilar and its reference product. The strategy should aim to identify potential differences in immune responses between these products. While an ADA assay employing the proposed biosimilar product as the detecting reagent has been generally recommended for such evaluation, a product-specific assay using the product of interest may be of use as it offers a capability of detecting antibodies against specific epitopes from the respective product. Regardless of assay strategy, the performance of the assay must be fully assessed and method needs to be validated to meet the comparative purpose of immunogenicity assessment.


Subject(s)
Biosimilar Pharmaceuticals/pharmacology , Drug Discovery/methods , Immunity/drug effects , Animals , Humans
10.
J Immunoassay Immunochem ; 29(1): 42-57, 2008.
Article in English | MEDLINE | ID: mdl-18080879

ABSTRACT

Functions and binding properties of four CD11c-specific mAbs are described here. The mAb 496B stimulated, while 496K inhibited ligand binding of CD11c. The stimulatory mAb, 496B, as well as the inhibitory mAbs BU15 and 496 K appear to act allosterically, as they do not bind the CD11c I domain. The mAb 3.9 bound preferentially to activated forms of CD11c and the binding was divalent cation dependent. CD11c binding to 3.9 recapitulates many of the integrin-ligand interactions. Our data suggest that 3.9 is a competitive antagonist, BU15 and 496K are allosteric antagonists, and 496B is an allosteric agonist of CD11c. These mAbs provide a set of tools to study the functions of the dendritic cell marker, CD11c.


Subject(s)
Antibodies, Monoclonal/immunology , CD11c Antigen/analysis , CD11c Antigen/immunology , Antibodies, Blocking/immunology , Cations, Divalent/chemistry , Humans
11.
Biochem Biophys Res Commun ; 358(3): 938-41, 2007 Jul 06.
Article in English | MEDLINE | ID: mdl-17512497

ABSTRACT

Natural Killer (NK) cells kill certain tumor cells and virus infected cells in an antigen-independent manner. Members of CD18 integrins such as CD11a, CD11b, and CD11c are expressed in all NK cells. CD18-blocking mAbs inhibit the killing activity of NK cells implying an essential role of these integrins in NK cell cytotoxicity. In this report we show that the pan CD18-activating mAb, 240Q, augments cytotoxicity of resting NK cells. Since activation of either CD11a or CD11c alone fails to augment the NK cell activity, we postulate that a functional synergy of the individual CD18 integrins is responsible for the observed stimulatory effect of pan CD18 activation on NK cell cytotoxicity.


Subject(s)
Antibodies/chemistry , CD18 Antigens/biosynthesis , Integrins/metabolism , Killer Cells, Natural/cytology , Antibodies, Monoclonal/chemistry , CD11a Antigen/metabolism , CD11b Antigen/metabolism , CD11c Antigen/metabolism , Cell Adhesion , Flow Cytometry , Humans , Hybridomas/metabolism , K562 Cells , Killer Cells, Natural/metabolism
12.
J Leukoc Biol ; 81(6): 1395-403, 2007 Jun.
Article in English | MEDLINE | ID: mdl-17389580

ABSTRACT

CD11c, a member of the leukointegrin family, is expressed prominently on tissue macrophages and dendritic cells and binds to complement fragment (iC3b), provisional matrix molecules (fibrinogen), and the Ig superfamily cell adhesion molecule, ICAM-1. CD11c has been proposed to function in phagocytosis, cell migration, and cytokine production by monocytes/macrophages as well as induction of T cell proliferation by Langerhans cells. Using assays to quantify CD11c-mediated cell adhesion, we demonstrate that CD11c recognizes ICAM-2 and VCAM-1. The CD11c-binding site on VCAM-1 appears to be different from that used by the integrin alpha4. CD11c and alpha4beta1 contributed to monocyte capture and transmigration on inflamed human aortic endothelial cells. We discovered that the anti-mouse CD11c mAb N418 blocks CD11c binding to iC3b, ICAM-1, and VCAM-1. Treatment of mice with N418 reduced SRBC-induced delayed-type hypersensitivity significantly. CD11c appeared to contribute predominantly to the sensitization phase and somewhat less to the response to SRBC challenge. This suggests a novel role for CD11c during leukocyte recruitment, antigen uptake, and the survival of APC.


Subject(s)
CD11c Antigen/immunology , CD18 Antigens/immunology , Hypersensitivity, Delayed/immunology , Animals , Antibodies, Monoclonal/pharmacology , Antigens, CD/metabolism , Aorta/cytology , Binding Sites , CD11c Antigen/metabolism , Cell Adhesion/immunology , Cell Adhesion Molecules/metabolism , Cells, Cultured , Complement C3b/immunology , Endothelial Cells/immunology , Endothelial Cells/physiology , Erythrocytes/immunology , Humans , Integrin alpha4/metabolism , Ligands , Mice , Monocytes/immunology , Monocytes/physiology , Protein Binding , Sheep , Vascular Cell Adhesion Molecule-1/metabolism
13.
Biochem Biophys Res Commun ; 308(4): 764-9, 2003 Sep 05.
Article in English | MEDLINE | ID: mdl-12927784

ABSTRACT

Although members of the class I phosphoinositide 3-kinases (PI3Ks) have been implicated in neutrophil inflammatory responses, the contribution of the individual PI3K isoforms in neutrophil activation has not been tractable with the non-selective inhibitors, LY294002 and wortmannin. We have developed a novel series of PI3K inhibitors that is selective for PI3K delta, an isoform expressed predominantly in hematopoietic cells. In addition to being selective between members of class I PI3Ks, representatives of these inhibitors such as IC980033 and IC87114 did not inhibit any protein kinases tested. Utilizing these inhibitors we report here a novel role for PI3K delta in neutrophil activation. Inhibition of PI3K delta with IC980033 and IC87114 blocked both fMLP- and TNF1 alpha-induced neutrophil superoxide generation and elastase exocytosis. The PI3K delta inhibitor IC87114 also blocked TNF1 alpha-stimulated elastase exocytosis from neutrophils in a mouse model of inflammation. To our knowledge, this is the first in vivo efficacy demonstration of a PI3K delta inhibitor in an animal model. Inhibition of PI3K delta, however, had no effect on in vitro neutrophil bactericidal activity and Fc gamma R-stimulated superoxide generation. Thus, PI3K delta plays an essential role in certain signaling pathways of neutrophil activation and appears to be an attractive target for the development of an anti-inflammatory therapeutic.


Subject(s)
Neutrophils/enzymology , Neutrophils/immunology , Phosphatidylinositol 3-Kinases/physiology , Adenine/analogs & derivatives , Adenine/pharmacology , Androstadienes/pharmacology , Animals , Anti-Inflammatory Agents/pharmacology , Chromones/pharmacology , Class I Phosphatidylinositol 3-Kinases , Disease Models, Animal , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Exocytosis , Inflammation , Leukocyte Elastase/chemistry , Mice , Mice, Inbred BALB C , Morpholines/pharmacology , Neutrophils/metabolism , Phosphatidylinositol 3-Kinases/chemistry , Protein Isoforms , Quinazolines/pharmacology , Signal Transduction , Superoxides , Time Factors , Wortmannin
14.
J Immunol ; 170(5): 2647-54, 2003 Mar 01.
Article in English | MEDLINE | ID: mdl-12594293

ABSTRACT

Neutrophil chemotaxis is a critical component of the innate immune response. Neutrophils can sense an extremely shallow gradient of chemoattractants and produce relatively robust chemotactic behavior. This directional migration requires cell polarization with actin polymerization occurring predominantly in the leading edge. Synthesis of phosphatidylinositol (3,4,5) trisphosphate (PIP3) by phosphoinositide 3-kinase (PI3K) contributes to asymmetric F-actin synthesis and cell polarization during neutrophil chemotaxis. To determine the contribution of the hemopoietic cell-restricted PI3K delta in neutrophil chemotaxis, we have developed a potent and selective PI3K delta inhibitor, IC87114. IC87114 inhibited polarized morphology of neutrophils, fMLP-stimulated PIP3 production and chemotaxis. Tracking analysis of IC87114-treated neutrophils indicated that PI3K delta activity was required for the directional component of chemotaxis, but not for random movement. Inhibition of PI3K delta, however, did not block F-actin synthesis or neutrophil adhesion. These results demonstrate that PI3K delta can play a selective role in the amplification of PIP3 levels that lead to neutrophil polarization and directional migration.


Subject(s)
Chemotaxis, Leukocyte/physiology , Neutrophils/cytology , Neutrophils/enzymology , Phosphatidylinositol 3-Kinases/physiology , Cell Movement/drug effects , Cell Movement/physiology , Cell Polarity/drug effects , Cell Polarity/physiology , Chemotaxis, Leukocyte/drug effects , Chromones/pharmacology , Class I Phosphatidylinositol 3-Kinases , Enzyme Inhibitors/pharmacology , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/physiology , Morpholines/pharmacology , N-Formylmethionine Leucyl-Phenylalanine/pharmacology , Neutrophil Activation/drug effects , Neutrophil Activation/physiology , Neutrophils/drug effects , Neutrophils/metabolism , Phosphatidylinositol Phosphates/biosynthesis , Phosphoinositide-3 Kinase Inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...