Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
1.
Genet Test Mol Biomarkers ; 22(4): 266-269, 2018 Apr.
Article in English | MEDLINE | ID: mdl-29641284

ABSTRACT

AIMS: Dichloroacetate (DCA) represents the first targeted therapy for pyruvate dehydrogenase complex deficiency; it is metabolized by glutathione transferase zeta1 (GSTZ1). Variation in the GSTZ1 haplotype is the principal variable influencing DCA kinetics and dynamics in humans. We aimed to develop a sensitive and rapid clinical genetic screening test for determining GSTZ1 haplotype status in individuals who would be treated with DCA, and then apply the test for the investigation of the plasma pharmacokinetics (PK) of DCA as a function of GSTZ1 haplotype. MATERIALS AND METHODS: DNA samples from 45 healthy volunteer study participants were genotyped for three functional GSTZ1 single nucleotide polymorphisms (rs7975, rs7972, and rs1046428) by TaqMan®. Prior studies showed that subjects with at least one EGT haplotype (EGT carrier) metabolized DCA faster than EGT noncarriers. The clinical genetic test for GSTZ1 was developed and validated at our CLIA-certified Clinical Laboratory. Four fast metabolizer EGT carriers and four slow metabolizer EGT noncarriers were selected to complete a standard PK study. Each participant received a single oral dose of 25 mg/kg of DCA (IND 028625) for 5 days. RESULTS: The EGT haplotype carrier group demonstrated significantly faster metabolism of DCA and higher rates of plasma DCA clearance after 5 days of drug exposure compared with EGT noncarriers (p = 0.04). CONCLUSIONS: These preliminary data establish the validity and practicality of our rapid genotyping/haplotyping procedure for genetic-based DCA dosing to mitigate or prevent adverse effects in patients treated chronically with this drug.


Subject(s)
Dichloroacetic Acid/therapeutic use , Genotype , Glutathione Transferase/genetics , Precision Medicine , Adolescent , Adult , Aged , Dichloroacetic Acid/administration & dosage , Dichloroacetic Acid/pharmacokinetics , Female , Haplotypes , Humans , Male , Middle Aged , Reference Values , Reproducibility of Results , Young Adult
2.
Mitochondrion ; 42: 59-63, 2018 09.
Article in English | MEDLINE | ID: mdl-29129554

ABSTRACT

We developed an Observer-Reported Outcome (ObsRO) survey instrument to be applied in a multicenter, placebo-controlled, crossover randomized controlled trial of dichloroacetate in children with pyruvate dehydrogenase complex deficiency. The instrument quantifies a subject's at-home level of functionality, as reported by a parent/caregiver, who were instrumental in providing the clinical descriptors and domains that formed the instrument's content. Feasibility testing of the ObsRO tool showed it to be easy to use and comprehensive in capturing the major clinical functional limitations of affected children and requires less than 5min for a parent/caregiver to complete daily.


Subject(s)
Dichloroacetic Acid/administration & dosage , Pyruvate Dehydrogenase Complex Deficiency Disease/drug therapy , Rare Diseases/drug therapy , Symptom Assessment/methods , Treatment Outcome , Child , Child, Preschool , Cross-Over Studies , Female , Humans , Male , Placebos/administration & dosage
3.
Hum Gene Ther Clin Dev ; 28(4): 208-218, 2017 12.
Article in English | MEDLINE | ID: mdl-29160099

ABSTRACT

A first-in-human trial of diaphragmatic gene therapy (AAV1-CMV-GAA) to treat respiratory and neural dysfunction in early-onset Pompe disease was conducted. The primary objective of this study was to assess the safety of rAAV1-CMV-hGAA vector delivered to the diaphragm muscle of Pompe disease subjects with ventilatory insufficiency. Safety was assessed by measurement of change in serum chemistries and hematology, urinalysis, and immune response to GAA and AAV, as well as change in level of health. The data demonstrate that the AAV treatment was safe and there were no adverse events related to the study agent. Adverse events related to the study procedure were observed in subjects with lower baseline neuromuscular function. All adverse events were resolved before the end of the study, except for one severe adverse event determined not to be related to either the study agent or the study procedure. In addition, an anti-capsid and anti-transgene antibody response was observed in all subjects who received rAAV1-CMV-hGAA, except for subjects who received concomitant immunomodulation to manage reaction to enzyme replacement therapy, as per their standard of care. This observation is significant for future gene therapy studies and serves to establish a clinically relevant approach to blocking immune responses to both the AAV capsid protein and transgene product.


Subject(s)
Dependovirus/genetics , Genetic Therapy , Glycogen Storage Disease Type II/genetics , alpha-Glucosidases/administration & dosage , Animals , Child , Diaphragm/surgery , Female , Genetic Vectors/administration & dosage , Genetic Vectors/adverse effects , Glycogen Storage Disease Type II/blood , Glycogen Storage Disease Type II/pathology , Glycogen Storage Disease Type II/therapy , Humans , Immunomodulation , Male , Mice , Muscle, Skeletal , Thoracic Surgery, Video-Assisted , Transgenes/genetics , alpha-Glucosidases/adverse effects , alpha-Glucosidases/genetics
4.
Exp Neurol ; 287(Pt 2): 216-224, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27453480

ABSTRACT

Pompe disease is an inherited disorder due to a mutation in the gene that encodes acid α-glucosidase (GAA). Children with infantile-onset Pompe disease develop progressive hypotonic weakness and cardiopulmonary insufficiency that may eventually require mechanical ventilation (MV). Our team conducted a first in human trial of diaphragmatic gene therapy (AAV1-CMV-GAA) to treat respiratory neural dysfunction in infantile-onset Pompe. Subjects (aged 2-15years, full-time MV: n=5, partial/no MV: n=4) underwent a period of preoperative inspiratory muscle conditioning exercise. The change in respiratory function after exercise alone was compared to the change in function after intramuscular delivery of AAV1-CMV-GAA to the diaphragm with continued exercise. Since AAV-mediated gene therapy can reach phrenic motoneurons via retrograde transduction, we hypothesized that AAV1-CMV-GAA would improve dynamic respiratory motor function to a greater degree than exercise alone. Dependent measures were maximal inspiratory pressure (MIP), respiratory responses to inspiratory threshold loads (load compensation: LC), and physical evidence of diaphragm activity (descent on MRI, EMG activity). Exercise alone did not change function. After AAV1-CMV-GAA, MIP was unchanged. Flow and volume LC responses increased after dosing (p<0.05 to p<0.005), but only in the subjects with partial/no MV use. Changes in LC tended to occur on or after 180days. At Day 180, the four subjects with MRI evidence of diaphragm descent had greater maximal voluntary ventilation (p<0.05) and tended to be younger, stronger, and use fewer hours of daily MV. In conclusion, combined AAV1-CMV-GAA and exercise training conferred benefits to dynamic motor function of the diaphragm. Children with a higher baseline neuromuscular function may have greater potential for functional gains.


Subject(s)
Diaphragm/physiology , Exercise Therapy , Genetic Therapy , Glycogen Storage Disease Type II/complications , Respiratory Insufficiency/etiology , Respiratory Insufficiency/therapy , Adenoviridae/genetics , Adenoviridae/metabolism , Adolescent , Child , Child, Preschool , Electromyography , Female , Glycogen Storage Disease Type II/diagnostic imaging , Glycogen Storage Disease Type II/genetics , Glycogen Storage Disease Type II/therapy , Humans , Magnetic Resonance Imaging , Male , Muscle, Skeletal/physiopathology , Prospective Studies , Respiratory Insufficiency/diagnostic imaging , Treatment Outcome , alpha-Glucosidases/genetics , alpha-Glucosidases/therapeutic use
5.
Phys Ther ; 96(5): 696-703, 2016 May.
Article in English | MEDLINE | ID: mdl-26893511

ABSTRACT

BACKGROUND AND PURPOSE: Pompe disease is an inherited disorder notable for severe, progressive ventilatory compromise. Although ventilatory failure has been attributed to myofiber dysfunction secondary to diaphragmatic glycogen accumulation, neural involvement of the phrenic motor system is also a prominent feature. Direct diaphragm pacing supplements respiratory function in other disorders of the phrenic motor system. Accordingly, it is hypothesized that augmented neuromuscular activity via diaphragm pacing would promote weaning from mechanical ventilation in patients with Pompe disease who are unresponsive to conventional, muscle-directed treatments. CASE DESCRIPTION: Three patients with Pompe disease developed diaphragm paresis that resulted in chronic mechanical ventilation dependence. After preoperative inspiratory muscle strengthening exercises failed to improve function, fine-wire pacing electrodes were laparoscopically implanted into the diaphragm. Diaphragm conditioning was initiated the first postoperative week and consisted of gradual increases in stimulation parameters, lengthening of stimulation sessions, and ventilator weaning. Ventilation and intramuscular electromyographic activity were recorded periodically during conditioning to quantify diaphragm neuromuscular function. OUTCOMES: During paced breathing without mechanical ventilation, tidal volumes increased, and 2 patients were weaned from daytime ventilator dependence within the first 3 months of pacing, which has been sustained over the long-term. A third patient reduced reliance on daytime ventilation, but weaning was delayed by malacia of the large airways. In all patients, pacing appeared to facilitate spontaneous phrenic motor unit activity during independent breathing without ventilator or pacer support. DISCUSSION: The findings are consistent with the view that diaphragm pacing has potential rehabilitative value to reduce reliance on mechanical ventilation in people with Pompe disease, but further study is needed. Diaphragm pacing represents a paradigm shift in the management of respiratory insufficiency for Pompe disease that warrants further controlled examination.


Subject(s)
Diaphragm/physiopathology , Electric Stimulation Therapy , Glycogen Storage Disease Type II/physiopathology , Glycogen Storage Disease Type II/rehabilitation , Respiration , Respiratory Insufficiency/rehabilitation , Child, Preschool , Electrodes, Implanted , Electromyography , Glycogen Storage Disease Type II/complications , Humans , Male , Middle Aged , Positive-Pressure Respiration , Pulmonary Ventilation , Respiratory Insufficiency/etiology , Respiratory Insufficiency/physiopathology , Ventilator Weaning
6.
Muscle Nerve ; 53(2): 214-21, 2016 02.
Article in English | MEDLINE | ID: mdl-26351754

ABSTRACT

UNLABELLED: I NTRODUCTION: Individuals with X-linked myotubular myopathy (XLMTM) and other centronuclear myopathies (CNMs) frequently have profound respiratory insufficiency that requires support early in life. Still, few quantitative data exist to characterize respiratory motor function in CNM. METHODS: We evaluated the reliance upon mechanical ventilation (MV), ventilatory kinematics, unassisted tidal volumes, and maximal respiratory pressures in 14 individuals with CNMs, including 10 boys with XLMTM. RESULTS: Thirteen participants required full-time, invasive MV. Maximal inspiratory pressures were higher in subjects who breathed unsupported at least 1 hour/day as compared with 24-hour MV users [33.7 (11.9-42.3) vs. 8.4 (6.0-10.9) cm H(2)O, P < 0.05]. Years of MV dependence correlated significantly with MEP (r = -0.715, P < 0.01). CONCLUSIONS: Respiratory function in CNMs may be related to deconditioning from prolonged MV and/or differences in residual respiratory muscle strength. Results from this study may assist in evaluating severe respiratory insufficiency in neuromuscular clinical care and research.


Subject(s)
Myopathies, Structural, Congenital/complications , Respiration Disorders/diagnosis , Respiration Disorders/etiology , Respiratory Function Tests/methods , Adolescent , Adult , Aged , Child , Child, Preschool , Female , Humans , Infant , Male , Middle Aged , Myopathies, Structural, Congenital/therapy , Pressure , Respiration, Artificial/methods , Respiratory Muscles/physiopathology , Young Adult
7.
Muscle Nerve ; 51(6): 877-83, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25186912

ABSTRACT

INTRODUCTION: Pompe disease is a progressive disease that affects skeletal muscles and leads to loss of ambulation. We investigated the activation of the tibialis anterior (TA) in late-onset Pompe disease (LOPD) individuals during maximal voluntary contraction (MVC) and evoked involuntary responses. METHODS: Four LOPD patients and matched control subjects performed MVC of the TA using dorsiflexion and TA evoked responses. Activation of the TA was recorded with surface electromyography. RESULTS: The Pompe patients exhibited greater power at frequencies below 60 Hz and reduced power above 100 Hz. They also exhibited a reduced increase in M-wave and prolonged M-wave latency and duration in response to stimulation. CONCLUSIONS: These results provide evidence that LOPD individuals have an altered activation pattern of the TA during maximal contractions. The observed activation pattern may reflect impairments in voluntary command, neuromuscular junction pathology, or compensatory drive due to a reduced number of functional motoneurons.


Subject(s)
Glycogen Storage Disease Type II/complications , Isometric Contraction/physiology , Movement Disorders/etiology , Muscle, Skeletal/physiopathology , Adolescent , Adult , Electric Stimulation , Evoked Potentials, Motor/physiology , Female , Humans , Male , Young Adult
9.
Respir Physiol Neurobiol ; 189(2): 241-9, 2013 Nov 01.
Article in English | MEDLINE | ID: mdl-23797185

ABSTRACT

Pompe disease is due to mutations in the gene encoding the lysosomal enzyme acid α-glucosidase (GAA). Absence of functional GAA typically results in cardiorespiratory failure in the first year; reduced GAA activity is associated with progressive respiratory failure later in life. While skeletal muscle pathology contributes to respiratory insufficiency in Pompe disease, emerging evidence indicates that respiratory neuron dysfunction is also a significant part of dysfunction in motor units. Animal models show profound glycogen accumulation in spinal and medullary respiratory neurons and altered neural activity. Tissues from Pompe patients show central nervous system glycogen accumulation and motoneuron pathology. A neural mechanism raises considerations about the current clinical approach of enzyme replacement since the recombinant protein does not cross the blood-brain-barrier. Indeed, clinical data suggest that enzyme replacement therapy delays symptom progression, but many patients eventually require ventilatory assistance, especially during sleep. We propose that treatments which restore GAA activity to respiratory muscles, neurons and networks will be required to fully correct ventilatory insufficiency in Pompe disease.


Subject(s)
Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/pathology , Glycogen Storage Disease Type II/physiopathology , Motor Neurons/physiology , Respiratory Muscles/physiology , Animals , Humans , Neuromuscular Junction/physiology
10.
J Pediatr ; 163(3): 847-54.e1, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23601496

ABSTRACT

OBJECTIVE: To evaluate whether B-cell depletion before enzyme replacement therapy (ERT) initiation can block acid alpha-glucosidase (GAA) antibody responses and improve clinical outcomes. STUDY DESIGN: Six subjects with Pompe disease (including 4 cross-reacting immunologic material-negative infants) aged 2-8 months received rituximab and sirolimus or mycophenolate before ERT. Four subjects continued to receive sirolimus, rituximab every 12 weeks, and intravenous immunoglobulin monthly for the duration of ERT. Sirolimus trough levels, IgG, CD3, CD4, CD8, CD19, CD20, N-terminal pro-brain natriuretic peptide, creatine kinase, creatine kinase-MB, C-reactive protein, platelets, alkaline phosphatase, gamma-glutamyl transferase, aspartate aminotransferase, and alanine aminotransferase were measured regularly. RESULTS: Immunomodulation achieved B-cell depletion without adverse effects. After 17-36 months of rituximab, sirolimus and ERT, all subjects lacked antibodies against GAA, 4 continued to gain motor milestones, yet 2 progressed to require invasive ventilation. The absence of infusion-associated reactions allowed the use of accelerated infusion rates. CONCLUSION: B-cell depletion and T-cell immunomodulation in infants naïve to ERT was accomplished safely and eliminated immune responses against GAA, thereby optimizing clinical outcome; however, this approach did not necessarily influence sustained independent ventilation. Importantly, study outcomes support the initiation of immunomodulation before starting ERT, because the study regimen allowed for prompt initiation of treatment.


Subject(s)
Antibodies, Monoclonal, Murine-Derived/therapeutic use , Enzyme Replacement Therapy , Glycogen Storage Disease Type II/drug therapy , Immunosuppressive Agents/therapeutic use , Mycophenolic Acid/analogs & derivatives , Sirolimus/therapeutic use , alpha-Glucosidases/therapeutic use , Antigens, CD/blood , Autoantibodies/blood , B-Lymphocytes/metabolism , Biomarkers/blood , Drug Administration Schedule , Drug Therapy, Combination , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Glycogen Storage Disease Type II/enzymology , Glycogen Storage Disease Type II/immunology , Humans , Immunoglobulin G/blood , Immunoglobulins, Intravenous/therapeutic use , Infant , Male , Mycophenolic Acid/therapeutic use , Rituximab , Treatment Outcome , alpha-Glucosidases/immunology
11.
Hum Gene Ther ; 24(6): 630-40, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23570273

ABSTRACT

Pompe disease is an inherited neuromuscular disease caused by deficiency of lysosomal acid alpha-glucosidase (GAA) leading to glycogen accumulation in muscle and motoneurons. Cardiopulmonary failure in infancy leads to early mortality, and GAA enzyme replacement therapy (ERT) results in improved survival, reduction of cardiac hypertrophy, and developmental gains. However, many children have progressive ventilatory insufficiency and need additional support. Preclinical work shows that gene transfer restores phrenic neural activity and corrects ventilatory deficits. Here we present 180-day safety and ventilatory outcomes for five ventilator-dependent children in a phase I/II clinical trial of AAV-mediated GAA gene therapy (rAAV1-hGAA) following intradiaphragmatic delivery. We assessed whether rAAV1-hGAA results in acceptable safety outcomes and detectable functional changes, using general safety measures, immunological studies, and pulmonary functional testing. All subjects required chronic, full-time mechanical ventilation because of respiratory failure that was unresponsive to both ERT and preoperative muscle-conditioning exercises. After receiving a dose of either 1×10(12) vg (n=3) or 5×10(12) vg (n=2) of rAAV1-hGAA, the subjects' unassisted tidal volume was significantly larger (median [interquartile range] 28.8% increase [15.2-35.2], p<0.05). Further, most patients tolerated appreciably longer periods of unassisted breathing (425% increase [103-851], p=0.08). Gene transfer did not improve maximal inspiratory pressure. Expected levels of circulating antibodies and no T-cell-mediated immune responses to the vector (capsids) were observed. One subject demonstrated a slight increase in anti-GAA antibody that was not considered clinically significant. These results indicate that rAAV1-hGAA was safe and may lead to modest improvements in volitional ventilatory performance measures. Evaluation of the next five patients will determine whether earlier intervention can further enhance the functional benefit.


Subject(s)
Dependovirus/metabolism , Genetic Therapy/adverse effects , Glycogen Storage Disease Type II/therapy , Pulmonary Ventilation/physiology , Respiratory Insufficiency/therapy , alpha-Glucosidases/genetics , alpha-Glucosidases/therapeutic use , Adolescent , Antibodies/blood , Child, Preschool , Diaphragm/physiopathology , Female , Genetic Vectors , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/physiopathology , Glycogen Storage Disease Type II/surgery , Humans , Immunity, Cellular , Infant , Male , Postoperative Care , Preoperative Care , Resistance Training , Respiratory Insufficiency/blood , Respiratory Insufficiency/immunology , Respiratory Insufficiency/physiopathology , T-Lymphocytes/immunology , Treatment Outcome
12.
Hum Mol Genet ; 20(R1): R61-8, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21518733

ABSTRACT

Pompe disease is an autosomal recessive metabolic myopathy caused by the deficiency of the lysosomal enzyme acid alpha-glucosidase and results in cellular lysosomal and cytoplasmic glycogen accumulation. A wide spectrum of disease exists from hypotonia and severe cardiac hypertrophy in the first few months of life due to severe mutations to a milder form with the onset of symptoms in adulthood. In either condition, the involvement of several systems leads to progressive weakness and disability. In early-onset severe cases, the natural history is characteristically cardiorespiratory failure and death in the first year of life. Since the advent of enzyme replacement therapy (ERT), the clinical outcomes have improved. However, it has become apparent that a new natural history is being defined in which some patients have substantial improvement following ERT, while others develop chronic disability reminiscent of the late-onset disease. In order to improve on the current clinical outcomes in Pompe patients with diminished clinical response to ERT, we sought to address the cause and potential for the treatment of disease manifestations which are not amenable to ERT. In this review, we will focus on the preclinical studies that are relevant to the development of a gene therapy strategy for Pompe disease, and have led to the first clinical trial of recombinant adeno-associated virus-mediated gene-based therapy for Pompe disease. We will cover the preliminary laboratory studies and rationale for a clinical trial, which is based on the treatment of the high rate of respiratory failure in the early-onset patients receiving ERT.


Subject(s)
Dependovirus/genetics , Genetic Therapy/methods , Glycogen Storage Disease Type II/therapy , Clinical Trials as Topic , Enzyme Replacement Therapy , Genetic Vectors/administration & dosage , Glycogen/metabolism , Glycogen Storage Disease Type II/immunology , Glycogen Storage Disease Type II/pathology , Humans , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL
...