109 results on '"Kevin M. Flanigan"'
Search Results
2. A first-in-human phase I/IIa gene transfer clinical trial for Duchenne muscular dystrophy using rAAVrh74.MCK.GALGT2
- Author
-
Kevin M. Flanigan, Tatyana A. Vetter, Tabatha R. Simmons, Megan Iammarino, Emma C. Frair, Federica Rinaldi, Louis G. Chicoine, Johan Harris, John P. Cheatham, Sharon L. Cheatham, Brian Boe, Megan A. Waldrop, Deborah A. Zygmunt, Davin Packer, and Paul T. Martin
- Subjects
Genetics ,Molecular Medicine ,Molecular Biology - Published
- 2022
3. Systemic PPMO-mediated dystrophin expression in the Dup2 mouse model of Duchenne muscular dystrophy
- Author
-
Liubov V. Gushchina, Tatyana A. Vetter, Emma C. Frair, Adrienne J. Bradley, Kelly M. Grounds, Jacob W. Lay, Nianyuan Huang, Aisha Suhaiba, Frederick J. Schnell, Gunnar Hanson, Tabatha R. Simmons, Nicolas Wein, and Kevin M. Flanigan
- Subjects
Drug Discovery ,Molecular Medicine - Abstract
Duchenne muscular dystrophy (DMD) is a devastating muscle-wasting disease that arises due to the loss of dystrophin expression, leading to progressive loss of motor and cardiorespiratory function. Four exon-skipping approaches using antisense phosphorodiamidate morpholino oligomers (PMOs) have been approved by the FDA to restore a
- Published
- 2022
4. Systemic delivery of an AAV9 exon-skipping vector significantly improves or prevents features of Duchenne muscular dystrophy in the Dup2 mouse
- Author
-
Nicolas Wein, Tatyana A. Vetter, Adeline Vulin, Tabatha R. Simmons, Emma C. Frair, Adrienne J. Bradley, Liubov V. Gushchina, Camila F. Almeida, Nianyuan Huang, Daniel Lesman, Dhanarajan Rajakumar, Robert B. Weiss, and Kevin M. Flanigan
- Subjects
Genetics ,Molecular Medicine ,Molecular Biology - Abstract
Duchenne muscular dystrophy (DMD) is typically caused by mutations that disrupt the
- Published
- 2022
5. Interim results of Transpher A, a multicenter, single-dose clinical trial of UX111 gene therapy for Sanfilippo syndrome type A (mucopolysaccharidosis IIIA)
- Author
-
Kevin M. Flanigan, Nicholas Smith, Maria Luz Couce, Deepa Rajan, Kristen Truxal, Kim L. McBride, Maria Jose de Castro Lopez, Maria Fuller, John Taylor, Ana B. Del Campo, Igor Grachev, and Heather Lau
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism ,Genetics ,Molecular Biology ,Biochemistry - Published
- 2023
6. Evaluation of biomarkers for Sanfilippo syndrome
- Author
-
Maria Fuller, Kim L. McBride, Kristen V. Truxal, Kevin M. Flanigan, and Jennifer T. Saville
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Oligosaccharides ,Mucopolysaccharidosis type III ,Neuropathology ,Urine ,030105 genetics & heredity ,Biochemistry ,Mucopolysaccharidosis III ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Gangliosides ,Internal medicine ,Genetics ,medicine ,Humans ,Molecular Biology ,Sanfilippo syndrome ,business.industry ,Metabolic disorder ,Infant ,medicine.disease ,Child, Preschool ,Pharmacodynamics ,Biomarker (medicine) ,Heparitin Sulfate ,Age of onset ,business ,Biomarkers ,030217 neurology & neurosurgery - Abstract
Sanfilippo syndrome or mucopolysaccharidosis type III (MPS III) is a childhood metabolic disorder marked by neuropathology arising due to impaired heparan sulphate (HS) catabolism. Consequently, partially degraded HS accumulates in the lysosomes of affected cells and is excreted in the urine. The measurement of HS in urine has long been considered a biomarker of Sanfilippo syndrome although it is largely non-specific. Using blood, urine and CSF collected from a cohort of Sanfilippo patients we investigated the utility of primary and secondary biomarkers to inform on disease activity. These included enzyme activity, specific oligosaccharides with non-reducing end residues reflective of the enzyme deficiency, and gangliosides. The diagnostic oligosaccharides - a HS disaccharide and tetrasaccharide - were elevated in the urine, plasma and CSF of all MPS IIIA and IIIB patients, respectively. There was no correlation between the concentrations in any of the matrices suggesting they reflect specific tissues and not overall disease burden. Enzyme activity did not inform on disease severity, with no measurable activity in CSF and activity approaching normal in MPS IIIA plasma. The concentration of gangliosides, GM2 and GM3, were significantly higher in the CSF of all MPS III subjects when compared to controls and correlated with the age of onset of first symptoms. Given that these gangliosides reflect delayed brain development they may be useful measures of disease burden, within the limitations of the clinical surrogates. Observation of these biochemical measurements in MPS III patients enrolled in clinical trials may determine whether they represent true pharmacodynamics biomarkers.
- Published
- 2019
7. Interim results of Transpher A, a multicentre, single-dose, phase 1/2 clinical trial of ABO-102 investigational gene therapy for Sanfilippo syndrome type A (mucopolysaccharidosis IIIA)
- Author
-
Kevin M. Flanigan, Nicholas J.N. Smith, Maria L. Couce, Maria Escolar, Kristen V. Truxal, Kim L. McBride, Maria J. de Castro, Maria Fuller, A. Pañeda, J. Ruiz, A.B. del Campo, and I. Grachev
- Subjects
Endocrinology ,Endocrinology, Diabetes and Metabolism ,Genetics ,Molecular Biology ,Biochemistry - Published
- 2022
8. Low-level dystrophin expression attenuating the dystrophinopathy phenotype
- Author
-
Diane E. Frank, Megan A. Waldrop, Felecia Gumienny, Robert B. Weiss, Kevin M. Flanigan, and Saleh El Husayni
- Subjects
Male ,musculoskeletal diseases ,0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Duchenne muscular dystrophy ,Nonsense mutation ,Gene Expression ,Biology ,Article ,Dystrophin ,03 medical and health sciences ,Exon ,0302 clinical medicine ,medicine ,Humans ,Muscular dystrophy ,Child ,Muscle, Skeletal ,Genetics (clinical) ,Genetics ,Muscle biopsy ,medicine.diagnostic_test ,Exons ,medicine.disease ,Exon skipping ,Muscular Dystrophy, Duchenne ,Phenotype ,030104 developmental biology ,Neurology ,Codon, Nonsense ,Pediatrics, Perinatology and Child Health ,RNA splicing ,biology.protein ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
The reading frame rule suggests that Duchenne muscular dystrophy (DMD) results from DMD mutations causing an out-of-frame transcript, whereas the milder Becker muscular dystrophy results from mutations causing an in-frame transcript. However, predicted nonsense mutations may instead result in altered splicing and an in-frame transcript. Here we report a 10-year-old boy with a predicted nonsense mutation in exon 42 who had a 6-minute walk time of 157% of that of age matched DMD controls, characterized as intermediate muscular dystrophy. RNA sequencing analysis from a muscle biopsy revealed only 6.0–9.8% of DMD transcripts were in-frame, excluding exon 42, and immunoblot demonstrated only 3.2% dystrophin protein expression. Another potential genetic modifier noted was homozygosity for the protective IAAM LTBP4 haplotype. This case suggests that very low levels of DMD exon skipping and dystrophin protein expression may result in amelioration of skeletal muscle weakness, a finding relevant to current dystrophin-restoring therapies.
- Published
- 2018
9. Updated results of Transpher A: multicenter, single-dose, phase 1/2 clinical trial of ABO-102 for Sanfilippo syndrome type A (mucopolysaccharidosis IIIA)
- Author
-
Luise Ammer, Claudia Ravelli, María L. Couce, Kim L. McBride, Kristen V. Truxal, Maria Fuller, Mona Lindschau, María J. De Castro, Juan Ruiz, Nicole Muschol, Bénédicte Héron, Kevin M. Flanigan, and Ana del Campo
- Subjects
Pediatrics ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Mucopolysaccharidosis ,medicine.disease ,Biochemistry ,Sanfilippo syndrome type a ,Clinical trial ,Endocrinology ,ABO blood group system ,Genetics ,Medicine ,business ,Molecular Biology - Published
- 2021
10. DMD – CLINICAL CARE
- Author
-
T. Vetter, Robert B. Weiss, S. Nicolau, Kevin M. Flanigan, Emma C Frair, and A. Bradley
- Subjects
medicine.medical_specialty ,Neurology ,business.industry ,Pediatrics, Perinatology and Child Health ,medicine ,Neurology (clinical) ,Clinical care ,Intensive care medicine ,business ,Genetics (clinical) - Published
- 2021
11. Clinicopathologic Conference: A Newborn With Hypotonia, Cleft Palate, Micrognathia, and Bilateral Club Feet
- Author
-
Daniel R. Boue, Megan A. Waldrop, Kevin M. Flanigan, Richard Shell, and Emily Sites
- Subjects
0301 basic medicine ,medicine.medical_specialty ,business.industry ,Congenital fiber type disproportion ,medicine.disease ,Native American myopathy ,Congenital myopathy ,Hypotonia ,Surgery ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Developmental Neuroscience ,Neurology ,Pediatrics, Perinatology and Child Health ,medicine ,Pierre Robin syndrome ,Neurology (clinical) ,medicine.symptom ,Malignant Hyperthermia Syndrome ,business ,030217 neurology & neurosurgery - Published
- 2017
12. LATE BREAKING NEWS ORAL PRESENTATION
- Author
-
Margaret Beatka, Johan Harris, Brenna R. Powers, Michael W. Lawlor, Tabatha R. Simmons, Tatyana Meyers Vetter, Maryann Kaler, Emma C Frair, Megan A. Waldrop, M. Iammarino, Nico Wein, Hui Meng, and Kevin M. Flanigan
- Subjects
Presentation ,medicine.medical_specialty ,Neurology ,business.industry ,media_common.quotation_subject ,General surgery ,Pediatrics, Perinatology and Child Health ,Medicine ,Neurology (clinical) ,business ,Genetics (clinical) ,media_common - Published
- 2020
13. Interim results of Transpher A, a multicenter, single-dose, phase 1/2 clinical trial of ABO-102 gene therapy for Sanfilippo syndrome type A (mucopolysaccharidosis type IIIA)
- Author
-
María José de Castro, Krista Cope, María L. Couce, MT Oreiro, Kevin M. Flanigan, Louise Jaensch, Maria Fuller, Tabatha R. Simmons, Nicholas J.C. Smith, Juan Ruiz, Kim L. McBride, and Kristen V. Truxal
- Subjects
medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Genetic enhancement ,Biochemistry ,Sanfilippo syndrome type a ,Gastroenterology ,Clinical trial ,Endocrinology ,ABO blood group system ,Internal medicine ,Interim ,Genetics ,medicine ,business ,Molecular Biology ,Mucopolysaccharidosis Type IIIA - Published
- 2020
14. Update in the Mucopolysaccharidoses
- Author
-
Kim L. McBride and Kevin M. Flanigan
- Subjects
Central Nervous System ,Newborn screening ,business.industry ,Early disease ,Infant, Newborn ,Genetic Therapy ,Mucopolysaccharidoses ,Bioinformatics ,Recombinant enzyme ,Multisystem disease ,03 medical and health sciences ,Neonatal Screening ,0302 clinical medicine ,030225 pediatrics ,Pediatrics, Perinatology and Child Health ,Humans ,Medicine ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Glycosaminoglycans - Abstract
The mucopolysaccharidoses (MPS) are a genetically heterogenous group of enzyme deficiencies marked by accumulation of glycosaminoglycans in lysosomes leading to multisystem disease. Although significant therapeutic advances have been made for the MPS disorders, including recombinant enzyme replacement approaches, the neuronopathic features of MPS lack adequate treatment. Gene therapies, including adeno-associated virus vectors targeting the central nervous system, hold significant promise for this group of disorders. Optimal outcomes of all therapies will require early disease identification and treatment, ideally by newborn screening.
- Published
- 2021
15. NEW GENES AND DISEASES / NGS & RELATED TECHNIQUES
- Author
-
C. Cottrell, A. Dave-Wala, M. Stein, R. Pfau, S. Hashimoto, S. Reshmi, M. Mathew, Kevin M. Flanigan, Megan A. Waldrop, D. Corsmeier, T. Matthews, K. Lee, V. Jayaraman, D. Mouhlas, and A. Meyer
- Subjects
Neurology ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) ,Computational biology ,Gene ,Genetics (clinical) - Published
- 2020
16. DMD – ANIMAL MODELS & PRECLINICAL TREATMENT
- Author
-
Nicolas Wein, D. Li, M. Spencer, C. Young, D. Lesman, C. Gaffney, Y. Rodriguez, D. Rajakumar, Kevin M. Flanigan, R. Rafferty, Nianyuan Huang, and Tabatha R. Simmons
- Subjects
Neurology ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) ,Genetics (clinical) - Published
- 2020
17. AUTOPHAGIC MYOPATHIES / MYOFIBRILLAR MYOPATHIES / DISTAL MYOPATHIES / POMPE DISEASE
- Author
-
M. Waldrop, W. Arnold, A. Meyer, and Kevin M. Flanigan
- Subjects
Pathology ,medicine.medical_specialty ,Neurology ,business.industry ,Pediatrics, Perinatology and Child Health ,Autophagy ,Distal Myopathies ,medicine ,Neurology (clinical) ,Disease ,Myofibril ,business ,Genetics (clinical) - Published
- 2020
18. Safety, tolerability and preliminary evidence of biopotency in Transpher B, a multicenter, single-dose, phase 1/2 clinical trial of ABO-101 gene therapy for Sanfilippo syndrome type B (mucopolysaccharidosis type IIIB)
- Author
-
Kim L. McBride, Kristen V. Truxal, Federica Rinaldi, MT Oreiro, María José de Castro, Tabatha R. Simmons, María L. Couce, Maria Fuller, Kevin M. Flanigan, and Juan Ruiz
- Subjects
medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Genetic enhancement ,Safety tolerability ,medicine.disease ,Biochemistry ,Gastroenterology ,Mucopolysaccharidosis type IIIB ,Clinical trial ,Endocrinology ,ABO blood group system ,Internal medicine ,Genetics ,medicine ,business ,Molecular Biology ,Sanfilippo syndrome - Published
- 2020
19. The first exon duplication mouse model of Duchenne muscular dystrophy: A tool for therapeutic development
- Author
-
Nicolas Wein, Andrew R. Findlay, Yuuki Kaminoh, Kevin M. Flanigan, Tabatha R. Simmons, Adeline Vulin, A. Rutherford, and Jacqueline A. Yurkoski
- Subjects
Male ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Duchenne muscular dystrophy ,Mice, Transgenic ,Biology ,Exon ,Gene Duplication ,Gene duplication ,medicine ,Animals ,Gene Knock-In Techniques ,RNA, Messenger ,Muscle, Skeletal ,Creatine Kinase ,Genetics (clinical) ,Body Weight ,Intron ,Skeletal muscle ,Exons ,medicine.disease ,Phenotype ,Molecular biology ,Exon skipping ,Muscular Dystrophy, Duchenne ,Disease Models, Animal ,medicine.anatomical_structure ,Neurology ,Pediatrics, Perinatology and Child Health ,Mice, Inbred mdx ,biology.protein ,Neurology (clinical) ,Dystrophin - Abstract
Exon duplication mutations account for up to 11% of all cases of Duchenne muscular dystrophy (DMD), and a duplication of exon 2 is the most common duplication in patients. For use as a platform for testing of duplication-specific therapies, we developed a mouse model that carries a Dmd exon 2 duplication. By using homologous recombination we duplicated exon 2 within intron 2 at a location consistent with a human duplication hotspot. mRNA analysis confirms the inclusion of a duplicated exon 2 in mouse muscle. Dystrophin expression is essentially absent by immunofluorescent and immunoblot analysis, although some muscle specimens show very low-level trace dystrophin expression. Phenotypically, the mouse shows similarities to mdx, the standard laboratory model of DMD. In skeletal muscle, areas of necrosis and phagocytosis are seen at 3 weeks, with central nucleation prominent by four weeks, recapitulating the "crisis" period in mdx. Marked diaphragm fibrosis is noted by 6 months, and remains unchanged at 12 months. Our results show that the Dup2 mouse is both pathologically (in degree and distribution) and physiologically similar to mdx. As it recapitulates the most common single exon duplication found in DMD patients, this new model will be a useful tool to assess the potential of duplicated exon skipping.
- Published
- 2015
20. Genetics and Emerging Treatments for Duchenne and Becker Muscular Dystrophy
- Author
-
Nicolas Wein, Lindsay N. Alfano, and Kevin M. Flanigan
- Subjects
Male ,musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Pediatrics ,medicine.medical_specialty ,Duchenne muscular dystrophy ,Dystrophin ,Adrenal Cortex Hormones ,Elevated serum transaminases ,medicine ,Humans ,Muscular dystrophy ,biology ,Gait Disturbance ,business.industry ,Infant ,medicine.disease ,Exon skipping ,Muscular Dystrophy, Duchenne ,Dmd gene ,Child, Preschool ,Mutation ,Pediatrics, Perinatology and Child Health ,biology.protein ,Physical therapy ,business - Abstract
Mutations in the DMD gene result in Duchenne or Becker muscular dystrophy due to absent or altered expression of the dystrophin protein. The more severe Duchenne muscular dystrophy typically presents around ages 2 to 5 with gait disturbance, and historically has led to the loss of ambulation by age 12. It is important for the practicing pediatrician, however, to be aware of other presenting signs, such as delayed motor or cognitive milestones, or elevated serum transaminases. Becker muscular dystrophy is milder, often presenting after age 5, with ambulation frequently preserved past 20 years and sometimes into late decades.
- Published
- 2015
21. Duchenne muscular dystrophy: meeting the therapeutic challenge
- Author
-
Kevin M. Flanigan
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,business.industry ,Duchenne muscular dystrophy ,medicine.disease ,Muscular Dystrophy, Duchenne ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine ,Humans ,Neurology (clinical) ,Muscular dystrophy ,business ,030217 neurology & neurosurgery - Published
- 2016
22. One Year Outcome of Boys With Duchenne Muscular Dystrophy Using the Bayley-III Scales of Infant and Toddler Development
- Author
-
Kate Bushby, Craig M. Zaidman, Elizabeth C. Malkus, Craig M. McDonald, Kevin M. Flanigan, J. Philip Miller, Anne M. Connolly, Mary Michaeleen Cradock, Michelle Eagle, Peter I. Karachunski, Paul T. Golumbek, Jerry R. Mendell, Julaine Florence, and Basil T. Darras
- Subjects
Male ,Pediatrics ,medicine.medical_specialty ,Duchenne muscular dystrophy ,Gross motor skill ,Motor Activity ,Article ,Child Development ,Cognition ,Developmental Neuroscience ,medicine ,Humans ,Longitudinal Studies ,Toddler ,Motor skill ,Language ,Language Tests ,Infant ,Repeated measures design ,medicine.disease ,Child development ,Muscular Dystrophy, Duchenne ,Clinical trial ,Bayley iii ,Neurology ,Motor Skills ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Physical therapy ,Neurology (clinical) ,Psychology ,Follow-Up Studies - Abstract
The pathogenesis of Duchenne muscular dystrophy starts before birth. Despite this, clinical trials exclude young boys because traditional outcome measures rely on cooperation. We recently used the Bayley-III Scales of Infant and Toddler Development to study 24 infants and boys with Duchenne muscular dystrophy. Clinical evaluators at six centers were trained and certified to perform the Bayley-III. Here, we report 6- and 12-month follow-up of two subsets of these boys.Nineteen boys (1.9 ± 0.8 years) were assessed at baseline and 6 months. Twelve boys (1.5 ± 0.8 years) were assessed at baseline, 6, and 12 months.Gross motor scores were lower at baseline compared with published controls (6.2 ± 1.7; normal 10 ± 3; P0.0001) and revealed a further declining trend to 5.7 ± 1.7 (P = 0.20) at 6 months. Repeated measures analysis of the 12 boys monitored for 12 months revealed that gross motor scores, again low at baseline (6.6 ± 1.7; P0.0001), declined at 6 months (5.9 ± 1.8) and further at 12 months (5.3 ± 2.0) (P = 0.11). Cognitive and language scores were lower at baseline compared with normal children (range, P = 0.002-0.0001) and did not change significantly at 6 or 12 months (range, P = 0.89-0.09). Fine motor skills, also low at baseline, improved1 year (P = 0.05).Development can reliably be measured in infants and young boys with Duchenne muscular dystrophy across time using the Bayley-III. Power calculations using these data reveal that motor development may be used as an outcome measure.
- Published
- 2014
23. Pharmacokinetics and safety of single doses of drisapersen in non-ambulant subjects with Duchenne muscular dystrophy: Results of a double-blind randomized clinical trial
- Author
-
Joanna Nakielny, A. Morgan, Naashika Quarcoo, Thomas Voit, C. Wardell, Xiomara Q. Rosales, Lia Liefaard, Kevin M. Flanigan, John E. Kraus, G. Campion, Laurent Servais, Tom Drury, Susie Dorricott, and Padraig Wright
- Subjects
Male ,Duchenne muscular dystrophy ,medicine.medical_specialty ,Adolescent ,Population ,Clinical Neurology ,Oligonucleotides ,Urology ,Non-ambulant ,Article ,law.invention ,Dystrophin ,Double-Blind Method ,Randomized controlled trial ,Pharmacokinetics ,law ,Oligonucleotide ,Exon 51 ,DMD ,Post-hoc analysis ,Humans ,Medicine ,Genetics(clinical) ,Pediatrics, Perinatology, and Child Health ,Child ,education ,Genetics (clinical) ,Drisapersen ,education.field_of_study ,biology ,business.industry ,medicine.disease ,Muscular Dystrophy, Duchenne ,Neurology ,Tolerability ,Pediatrics, Perinatology and Child Health ,biology.protein ,Physical therapy ,Neurology (clinical) ,Inflammation Mediators ,Safety ,business ,Biomarkers ,Oligonucleotide, Pharmacokinetics - Abstract
Duchenne muscular dystrophy (DMD) is a progressive, lethal neuromuscular disorder caused by the absence of dystrophin protein due to mutations of the dystrophin gene. Drisapersen is a 2′-O-methyl-phosphorothioate oligonucleotide designed to skip exon 51 in dystrophin pre-mRNA to restore the reading frame of the mRNA. This study assessed safety, tolerability, and pharmacokinetics of drisapersen after a single subcutaneous administration in non-ambulatory subjects. Eligible subjects were non-ambulant boys aged ≥9 years, in wheelchairs for ≥1 to ≤4 years, with a diagnosis of DMD resulting from a mutation correctable by drisapersen treatment. Four dose cohorts were planned (3, 6, 9 and 12 mg/kg), but study objectives were met with the 9 mg/kg dose. Less than proportional increase in exposure was demonstrated over the 3–9 mg/kg dose range, though post hoc analysis showed dose proportionality was more feasible over the 3–6 mg/kg range. Single doses of drisapersen at 3 and 6 mg/kg did not result in significant safety or tolerability concerns; however, at the 9 mg/kg dose, pyrexia and transient elevations in inflammatory parameters were seen. The maximum tolerated dose of 6 mg/kg drisapersen was identified for further characterization in multiple dose studies in the non-ambulant DMD population.
- Published
- 2014
24. DMD CLINICAL THERAPIES II
- Author
-
Lindsay N. Alfano, Tabatha R. Simmons, John P. Cheatham, M. Iammarino, Rui Xu, Sharon L. Cheatham, L. Chicoine, Federica Rinaldi, Paul T. Martin, Linda Lowes, Deborah A. Zygmunt, Kevin M. Flanigan, Megan A. Waldrop, and N. Miller
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Duchenne muscular dystrophy ,Gene transfer ,medicine.disease ,Clinical trial ,Phase i ii ,Neurology ,Internal medicine ,Pediatrics, Perinatology and Child Health ,Medicine ,Neurology (clinical) ,business ,Genetics (clinical) - Published
- 2018
25. P.140RNA-Seq shows an absence of off-target splicing effects in AAV9-U7snRNA mediated skipping of DMD exon 2
- Author
-
Megan A. Waldrop, Robert B. Weiss, Nicolas Wein, L. Gushchina, and Kevin M. Flanigan
- Subjects
Genetics ,Exon ,Neurology ,Pediatrics, Perinatology and Child Health ,RNA splicing ,Neurology (clinical) ,Biology ,Genetics (clinical) - Published
- 2019
26. P.141PPMO-mediated skipping therapy of duplicated exon 2 in the DMD gene
- Author
-
Nicolas Wein, H. Huang, Emma C Frair, K. Grounds, Kevin M. Flanigan, Tabatha R. Simmons, F. Schnell, L. Gushchina, and G. Hanson
- Subjects
Genetics ,Exon ,Neurology ,Dmd gene ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) ,Biology ,Genetics (clinical) - Published
- 2019
27. P.365Clinical outcomes in patients with spinal muscular atrophy type 1, 2 or 3 after 1 year of nusinersen therapy
- Author
-
Megan A. Waldrop, Richard Shell, Lindsay N. Alfano, N. Miller, Linda Lowes, G. Paul, Garey Noritz, C. Tsao, K. Kotha, J. Toops, Kevin M. Flanigan, M. Iammarino, and M. Moore-Clingenpeel
- Subjects
medicine.medical_specialty ,Neurology ,business.industry ,Pediatrics, Perinatology and Child Health ,medicine ,In patient ,Nusinersen ,Neurology (clinical) ,Spinal muscular atrophy ,medicine.disease ,business ,Genetics (clinical) ,Surgery - Published
- 2019
28. EP.89Clinical trials in young boys and infants with DMD: how do you handle maturation?
- Author
-
Linda Lowes, C. Tsao, N. Miller, Lindsay N. Alfano, J. Mendell, M. Iammarino, M. Moore-Clingenpeel, Kevin M. Flanigan, and Megan A. Waldrop
- Subjects
Neurology ,Pediatrics, Perinatology and Child Health ,Neurology (clinical) ,Genetics (clinical) - Published
- 2019
29. Phase 1/2 clinical trial of systemic gene transfer of scAAV9.U1a.hSGSH for MPS IIIA demonstrates 2 years of safety, tolerability, and biopotency
- Author
-
Kevin M. Flanigan, MT Oreiro, Juan Ruiz, Nicholas J.C. Smith, S. Alyward, Kim L. McBride, Krista L. Kunkler, Kristen V. Truxal, Tabatha R. Simmons, L Jaensch, Maria Fuller, and María L. Couce
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Safety tolerability ,Gene transfer ,Biochemistry ,Clinical trial ,Endocrinology ,Internal medicine ,Genetics ,medicine ,business ,Molecular Biology - Published
- 2019
30. RNA Interference Inhibits DUX4-induced Muscle Toxicity In Vivo: Implications for a Targeted FSHD Therapy
- Author
-
Jacqueline S. Domire, Scott Q. Harper, Kevin M. Flanigan, Jerry R. Mendell, Jian Liu, Sara E Garwick-Coppens, Susan M. Guckes, and Lindsay M. Wallace
- Subjects
musculoskeletal diseases ,congenital, hereditary, and neonatal diseases and abnormalities ,Genetic Vectors ,Biology ,Bioinformatics ,Mice ,03 medical and health sciences ,0302 clinical medicine ,DUX4 ,RNA interference ,Drug Discovery ,microRNA ,Genetics ,medicine ,Animals ,Gene silencing ,Facioscapulohumeral muscular dystrophy ,RNA, Small Interfering ,Muscular dystrophy ,Muscle, Skeletal ,Myopathy ,Molecular Biology ,Gene ,030304 developmental biology ,Homeodomain Proteins ,Pharmacology ,0303 health sciences ,Genetic Therapy ,Dependovirus ,medicine.disease ,Muscular Dystrophy, Facioscapulohumeral ,nervous system diseases ,3. Good health ,Mice, Inbred C57BL ,MicroRNAs ,Molecular Medicine ,Female ,RNA Interference ,Original Article ,medicine.symptom ,030217 neurology & neurosurgery - Abstract
No treatment exists for facioscapulohumeral muscular dystrophy (FSHD), one of the most common inherited muscle diseases. Although FSHD can be debilitating, little effort has been made to develop targeted therapies. This lack of focus on targeted FSHD therapy perpetuated because the genes and pathways involved in the disorder were not understood. Now, more than 2 decades after efforts to decipher the root cause of FSHD began, this barrier to translation is finally lowering. Specifically, several recent studies support an FSHD pathogenesis model involving overexpression of the myopathic DUX4 gene. DUX4 inhibition has therefore emerged as a promising therapeutic strategy for FSHD. In this study, we tested a preclinical RNA interference (RNAi)-based DUX4 gene silencing approach as a prospective treatment for FSHD. We found that adeno-associated viral (AAV) vector-delivered therapeutic microRNAs corrected DUX4-associated myopathy in mouse muscle. These results provide proof-of-principle for RNAi therapy of FSHD through DUX4 inhibition.
- Published
- 2012
31. Effects of Angiotensin-Converting Enzyme Inhibitors and/or Beta Blockers on the Cardiomyopathy in Duchenne Muscular Dystrophy
- Author
-
Philip T. Thrush, Laurence Viollet, Hugh D. Allen, Jerry R. Mendell, and Kevin M. Flanigan
- Subjects
Adult ,Male ,medicine.medical_specialty ,Time Factors ,Adolescent ,Duchenne muscular dystrophy ,Adrenergic beta-Antagonists ,Cardiomyopathy ,Angiotensin-Converting Enzyme Inhibitors ,Ventricular Function, Left ,Young Adult ,Pharmacotherapy ,Internal medicine ,medicine ,Humans ,Prospective Studies ,Muscular dystrophy ,Child ,Glucocorticoids ,Ejection fraction ,Dose-Response Relationship, Drug ,biology ,business.industry ,Angiotensin-converting enzyme ,medicine.disease ,Echocardiography, Doppler ,Muscular Dystrophy, Duchenne ,Dose–response relationship ,Treatment Outcome ,Endocrinology ,ACE inhibitor ,Disease Progression ,Electrocardiography, Ambulatory ,biology.protein ,Drug Therapy, Combination ,Female ,Cardiomyopathies ,Cardiology and Cardiovascular Medicine ,business ,Follow-Up Studies ,medicine.drug - Abstract
Cardiomyopathy is a consequence of Duchenne muscular dystrophy (DMD). Suggested treatments include angiotensin-converting enzyme (ACE) inhibitors and/or β blockers (BBs), but few large series have been reported. We present 42 patients with DMD and cardiomyopathy treated with an ACE inhibitor or an ACE inhibitor plus a BB. Serial echocardiograms were recorded. Adequate ejection fractions (EFs) were obtained at initiation of therapy (EF55%). ACE inhibitor dosage adjustments were made if a continued decrease in EF was noted. BB therapy was initiated when average heart rate on Holter monitoring exceeded 100 beats/min. Data were analyzed using paired t test and linear regression. Before ACE inhibition, patients (n = 22) demonstrated decreased EF over time (r(2) = 0.23). At ACE inhibitor therapy initiation, mean age was 14.1 ± 4.6 years and mean EF was 44.2 ± 6.8%. BB therapy was used in 24 of 42 patients. Mean age for the ACE inhibitor + BB group was 15.7 ± 3.9 years. The 2 groups showed significant improvement (p0.0001 for ACE inhibitor and ACE inhibitor plus BB) compared to the pretherapy group. No significant differences were noted between treatment groups. Patients with DMD demonstrated a gradual decrease in myocardial function. Treatment with ACE inhibitor or ACE inhibitor plus BB resulted in significant improvement compared to pretherapy. No significant difference occurred in EF improvement between treatment groups. In conclusion, treatment with ACE inhibitor or ACE inhibitor plus BB can delay progression of cardiomyopathy.
- Published
- 2012
32. DUCHENNE MUSCULAR DYSTROPHY - PHYSIOTHERAPY
- Author
-
M. Dugan, M. Iammarino, C. Tsao, K. Berry, N. Miller, Megan A. Waldrop, Linda Lowes, M. Moore-Clingenpeel, J. Mendell, Kevin M. Flanigan, Samiah Al-Zaidy, Lindsay N. Alfano, and Louise R. Rodino-Klapac
- Subjects
medicine.medical_specialty ,business.industry ,Duchenne muscular dystrophy ,medicine.disease ,03 medical and health sciences ,0302 clinical medicine ,Neurology ,030225 pediatrics ,Pediatrics, Perinatology and Child Health ,medicine ,Physical therapy ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,Genetics (clinical) - Published
- 2018
33. Clinical and genetic characterization of manifesting carriers of DMD mutations
- Author
-
Carsten G. Bönnemann, Andrew von Niederhausern, Sidney M. Gospe, Michael T. Howard, Julaine Florence, Olga L. Gurvich, Diane M. Dunn, Jerry R. Mendell, Eduard Gappmaier, Michael J. Friez, Kathryn J. Swoboda, Richard S. Finkel, Jacinda B. Sampson, Laura E. Taylor, Brenda Wong, Anne M. Connolly, Karen Kovak, Michael D. Sussman, Livija Medne, Katherine D. Mathews, Robert B. Weiss, Jonathan Zonana, Alan Pestronk, Kevin M. Flanigan, and Payam Soltanzadeh
- Subjects
Adult ,Cardiomyopathy, Dilated ,Male ,musculoskeletal diseases ,Heterozygote ,Adolescent ,Duchenne muscular dystrophy ,DNA Mutational Analysis ,Nonsense mutation ,Biology ,Compound heterozygosity ,medicine.disease_cause ,Article ,Dystrophin ,Young Adult ,X Chromosome Inactivation ,medicine ,Humans ,Family history ,Muscular dystrophy ,Child ,Muscle, Skeletal ,Genetics (clinical) ,Genetics ,Mutation ,Muscle Weakness ,Splice site mutation ,Point mutation ,Middle Aged ,medicine.disease ,Muscular Dystrophy, Duchenne ,Neurology ,Child, Preschool ,Heart Function Tests ,Pediatrics, Perinatology and Child Health ,Female ,Neurology (clinical) - Abstract
Manifesting carriers of DMD gene mutations may present diagnostic challenges, particularly in the absence of a family history of dystrophinopathy. We review the clinical and genetic features in fifteen manifesting carriers identified among 860 subjects within the United Dystrophinopathy Project, a large clinical dystrophinopathy cohort whose members undergo comprehensive DMD mutation analysis. We defined manifesting carriers as females with significant weakness, excluding those with only myalgias/cramps. DNA extracted from peripheral blood was used to study X chromosome inactivation patterns. Among these manifesting carriers, age at symptom onset ranged from 2 to 47 years. Seven had no family history and eight had male relatives with Duchene muscular dystrophy (DMD). Clinical severity among the manifesting carriers varied from a DMD-like progression to a very mild Becker muscular dystrophy-like phenotype. Eight had exonic deletions or duplications and six had point mutations. One patient had two mutations (an exonic deletion and a splice site mutation), consistent with a heterozygous compound state. The X chromosome inactivation pattern was skewed toward nonrandom in four out of seven informative deletions or duplications but was random in all cases with nonsense mutations. We present the results of DMD mutation analysis in this manifesting carrier cohort, including the first example of a presumably compound heterozygous DMD mutation. Our results demonstrate that improved molecular diagnostic methods facilitate the identification of DMD mutations in manifesting carriers, and confirm the heterogeneity of mutational mechanisms as well as the wide spectrum of phenotypes.
- Published
- 2010
34. A phase 1/2 clinical trial of systemic gene transfer of scAAV9.U1a.HSGSH for MPS IIIA: Safety, tolerability, and preliminary evidence of biopotency
- Author
-
Tabatha R. Simmons, Nicholas Zumberge, Krista L. Kunkler, Douglas M. McCarty, Kelly A. McNally, Christopher McKee, Shawn C. Aylward, Juan Ruiz, Marco Corridore, L. Martín, Kevin M. Flanigan, Kim L. McBride, and Kristen V. Truxal
- Subjects
0301 basic medicine ,Oncology ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Safety tolerability ,Gene transfer ,Biochemistry ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,Endocrinology ,Internal medicine ,Genetics ,Medicine ,business ,Molecular Biology - Published
- 2018
35. Spinocerebellar Ataxia Type 31 Is Associated with 'Inserted' Penta-Nucleotide Repeats Containing (TGGAA)n
- Author
-
Mari Yoshida, Kinya Ishikawa, Tohru Matsuura, Sawa Iwasaki, Shuichi Asakawa, Yuko Saito, Takeshi Amino, Nobuyoshi Shimizu, Shoji Tsuji, Fumitoshi Ishino, Yuji Takahashi, Hidehiro Mizusawa, Kevin M. Flanigan, Tatsushi Toda, Shigeo Murayama, Taro Ishiguro, Nozomu Sato, Yoshio Hashizume, Taiji Tsunemi, Kazuhiro Kobayashi, and Makoto Takahashi
- Subjects
Adult ,Heterochromatin ,Molecular Sequence Data ,Biology ,Thymidine Kinase ,Article ,Microsatellite Repeat ,Genetics ,medicine ,Humans ,Spinocerebellar Ataxias ,Genetics(clinical) ,Age of Onset ,Gene ,In Situ Hybridization ,Genetics (clinical) ,Genes, Dominant ,Southern blot ,Base Sequence ,Homozygote ,Intron ,Sequence Analysis, DNA ,Physical Chromosome Mapping ,medicine.disease ,Molecular biology ,Founder Effect ,Introns ,Mutagenesis, Insertional ,Haplotypes ,RNA splicing ,Anticipation (genetics) ,Spinocerebellar ataxia ,Chromosomes, Human, Pair 16 ,Microsatellite Repeats - Abstract
Spinocerebellar ataxia type 31 (SCA31) is an adult-onset autosomal-dominant neurodegenerative disorder showing progressive cerebellar ataxia mainly affecting Purkinje cells. The SCA31 critical region was tracked down to a 900 kb interval in chromosome 16q22.1, where the disease shows a strong founder effect. By performing comprehensive Southern blot analysis and BAC- and fosmid-based sequencing, we isolated two genetic changes segregating with SCA31. One was a single-nucleotide change in an intron of the thymidine kinase 2 gene (TK2). However, this did not appear to affect splicing or expression patterns. The other was an insertion, from 2.5-3.8 kb long, consisting of complex penta-nucleotide repeats including a long (TGGAA)n stretch. In controls, shorter (1.5-2.0 kb) insertions lacking (TGGAA)n were found only rarely. The SCA31 repeat insertion's length inversely correlated with patient age of onset, and an expansion was documented in a single family showing anticipation. The repeat insertion was located in introns of TK2 and BEAN (brain expressed, associated with Nedd4) expressed in the brain and formed RNA foci in the nuclei of patients' Purkinje cells. An electrophoretic mobility-shift assay showed that essential splicing factors, serine/arginine-rich splicing factors SFRS1 and SFRS9, bind to (UGGAA)n in vitro. Because (TGGAA)n is a characteristic sequence of paracentromeric heterochromatin, we speculate that the insertion might have originated from heterochromatin. SCA31 is important because it exemplifies human diseases associated with "inserted" microsatellite repeats that can expand through transmission. Our finding suggests that the ectopic microsatellite repeat, when transcribed, might cause a disease involving the essential splicing factors.
- Published
- 2009
36. A novel form of juvenile recessive ALS maps to loci on 6p25 and 21q22
- Author
-
Kevin M. Flanigan, Mark Leppert, Russell J. Butterfield, Brith Otterud, Deepa Ramachandran, Sandra J. Hasstedt, and Kathryn J. Swoboda
- Subjects
Male ,Candidate gene ,Adolescent ,Chromosomes, Human, Pair 21 ,Offspring ,DNA Mutational Analysis ,Chromosome Disorders ,Genes, Recessive ,Biology ,medicine ,Humans ,Genetic Predisposition to Disease ,Genetic Testing ,Age of Onset ,Motor Neuron Disease ,Child ,Gene ,Genetics (clinical) ,Linkage (software) ,Genetics ,Amyotrophic Lateral Sclerosis ,Brain ,Chromosome Mapping ,Syndrome ,Motor neuron ,Disease gene identification ,Amyotrophy ,medicine.disease ,medicine.anatomical_structure ,Neurology ,Mutation ,Pediatrics, Perinatology and Child Health ,Mutation testing ,Chromosomes, Human, Pair 6 ,Neurology (clinical) ,Brain Stem - Abstract
We describe a novel form of juvenile recessive ALS (JRALS) affecting four of six offspring from a consanguineous first cousin marriage. The syndrome is characterized by early and prominent upper motor neuron signs, along with striking amyotrophy of the upper and lower limbs and bulbar involvement. After excluding linkage to loci with known association to ALS and other motor neuron diseases, we used a homozygosity mapping approach to identify loci on chromosomes 6p25 and 21q22, each with an equal probability of linkage to the trait (with a LOD score = 3.1, the maximum possible given the family structure). Mutation analysis of seven candidate genes that are expressed in the CNS or have roles in neuronal function did not reveal any pathogenic mutations. Identification of additional families will help to distinguish between which of the two autosomal loci contains the disease-causing gene, or whether this is a digenic trait.
- Published
- 2009
37. Mitochondrial DNA depletion syndrome due to mutations in the RRM2B gene
- Author
-
Jaya Ganesh, Belén Bornstein, Sara Shanske, Jorida Coku, Kathryn J. Swoboda, Estela Area, Salvatore DiMauro, Kurenai Tanji, Ali Naini, Parul Jayakar, Kevin M. Flanigan, and Michio Hirano
- Subjects
Male ,Models, Molecular ,Mitochondrial DNA ,Mitochondrial Diseases ,SUCLA2 ,DNA Mutational Analysis ,Cell Cycle Proteins ,Biology ,medicine.disease_cause ,DGUOK ,DNA, Mitochondrial ,Article ,Mitochondrial myopathy ,Ribonucleotide Reductases ,medicine ,Animals ,Humans ,Child ,Muscle, Skeletal ,MPV17 ,Genetics (clinical) ,Genetics ,Mutation ,medicine.disease ,Neurology ,Child, Preschool ,Lactic acidosis ,Pediatrics, Perinatology and Child Health ,Mitochondrial DNA depletion syndrome ,Female ,Neurology (clinical) ,Gene Deletion - Abstract
Mitochondrial DNA depletion syndrome (MDS) is characterized by a reduction in mtDNA copy number and has been associated with mutations in eight nuclear genes, including enzymes involved in mitochondrial nucleotide metabolism (POLG, TK2, DGUOK, SUCLA2, SUCLG1, PEO1) and MPV17. Recently, mutations in the RRM2B gene, encoding the p53-controlled ribonucleotide reductase subunit, have been described in seven infants from four families, who presented with various combinations of hypotonia, tubulopathy, seizures, respiratory distress, diarrhea, and lactic acidosis. All children died before 4 months of age. We sequenced the RRM2B gene in three unrelated cases with unexplained severe mtDNA depletion. The first patient developed intractable diarrhea, profound weakness, respiratory distress, and died at 3 months. The other two unrelated patients had a much milder phenotype and are still alive at ages 27 and 36 months. All three patients had lactic acidosis and severe depletion of mtDNA in muscle. Muscle histochemistry showed RRF and COX deficiency. Sequencing the RRM2B gene revealed three missense mutations and two single nucleotide deletions in exons 6, 8, and 9, confirming that RRM2B mutations are important causes of MDS and that the clinical phenotype is heterogeneous and not invariably fatal in infancy.
- Published
- 2008
38. Abnormal expression of mu-crystallin in facioscapulohumeral muscular dystrophy
- Author
-
Robert J. Bloch, Neil C. Porter, Andrea M. Corse, Kevin M. Flanigan, and Patrick W. Reed
- Subjects
musculoskeletal diseases ,Silver Staining ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Biopsy ,CRYM ,Immunoblotting ,Biology ,Silver stain ,Pathogenesis ,Muscular Diseases ,Developmental Neuroscience ,Crystallin ,mu-Crystallins ,Internal medicine ,Rosaniline Dyes ,medicine ,Humans ,Facioscapulohumeral muscular dystrophy ,Electrophoresis, Gel, Two-Dimensional ,Muscular dystrophy ,Muscle, Skeletal ,Myopathy ,Thyroid ,medicine.disease ,Crystallins ,Muscular Dystrophy, Facioscapulohumeral ,Up-Regulation ,nervous system diseases ,Oxidative Stress ,medicine.anatomical_structure ,Endocrinology ,Neurology ,medicine.symptom - Abstract
To identify proteins expressed abnormally in facioscapulohumeral muscular dystrophy (FSHD), we extracted soluble proteins from deltoid muscle biopsies from unaffected control and FSHD patients and analyzed them using two-dimensional electrophoresis, mass spectrometry and immunoblotting. Muscles from patients with FSHD showed large increases over controls in a single soluble, 34 kDa protein (pI=5.08) identified by mass spectrometry and immunoblotting as mu-crystallin (CRYM). Soluble fractions of biopsies of several other myopathies and muscular dystrophies showed no appreciable increases in mu-crystallin. Mu-crystallin has thyroid hormone and NADPH binding activity and so may influence differentiation and oxidative stress responses, reported to be altered in FSHD. It is also linked to retinal and inner ear defects, common in FSHD, suggesting that its up-regulation may play a specific and important role in pathogenesis of FSHD.
- Published
- 2007
39. 78. An IND-Enabling GLP-Toxicology and Biodistribution Study Assessing Systemic rAAV9-hNAGLU Gene Delivery for Treating MPS IIIB: Genotype- and Sex-Specific Dose-Limiting Acute Liver Toxicity in Male Wild Type C57BL/6 Mice
- Author
-
Chrystal Mongomery, F. Jason Duncan, Brad Bolon, Christopher M. Walker, Marybeth Camboni, Kevin M. Flanigan, William G. Bremer, Haiyan Fu, Kathryn Waligura, Aaron S. Meadows, Douglas M. McCarty, and Kim L. McBride
- Subjects
C57BL/6 ,Pharmacology ,medicine.medical_specialty ,biology ,business.industry ,Mucopolysaccharidosis ,Cell ,Cardiomyopathy ,Wild type ,Gene delivery ,medicine.disease ,biology.organism_classification ,Endocrinology ,medicine.anatomical_structure ,Internal medicine ,Toxicity ,Immunology ,Drug Discovery ,Lysosomal storage disease ,medicine ,Genetics ,Molecular Medicine ,business ,Molecular Biology - Abstract
No treatment is currently available for mucopolysaccharidosis (MPS) IIIB, a neuropathic lysosomal storage disease due to autosomal recessive defect in α-N-acetylglucosaminidase (NAGLU). In preparation for a clinical trial, we performed an IND-enabling GLP-toxicology study to assess systemic rAAV9-CMV-hNAGLU administration in wt C57BL/6 mice (6-8wk-old), at 1e14 vg/kg or 2e14 vg/kg (n=30/group, M:F=1:1). These were 2 and 4-fold higher, respectively, than our proposed clinical high dose. No adverse clinical signs were observed during the entire 6mo study duration. NAGLU activity above wt levels and vector genomes were detected in all tested CNS and somatic tissues in rAAV-treated mice through 6mo. However, acute mortality was observed in 5 male mice at 2e14 vg/kg dose on Days 6 and 8 pi. All 5 animals exhibited liver discoloration correlating with severe hepatic necrosis and vacuolation, without detectable leukocyte infiltration. At 6, 12 and 24wk pi, minimal single cell hepatocyte necrosis and/or vacuolation, and minimal cardiomyopathy were noted in a dose-dependent manner, with slight transient elevation of ALT or AST at 6wk pi. T-cell responses to AAV9 capsid and rNAGLU were detected at 6wk pi, but partially and completely diminished at 12wk pi, respectively. ELISA showed antibody responses to both AAV9 and rNAGLU in treated animals. In response to the acute mortality in the GLP-tox test, we performed a 6-wk non-GLP study mimicking the GLP-tox study conditions in male MPS IIIB and wt littermates, to further assess the approach and obtain data prior to early mortality. We treated the mice with the same batch of rAAV9-CMV-hNAGLU at the high dose, 2e14 vg/kg, and performed necropsy at 5 days and 6wk pi (n=4/group). No mortality or clinical signs were observed in either rAAV9-treated MPS IIIB or wt mice. At Day 5 pi necropsy, of 4 rAAV9-treated male wt mice, 1 exhibited liver discoloration, correlating with multifocal, periportal and midzonal hepatocyte necrosis, and 2 showed minimal single cell hepatocyte necrosis. Minimal liver damage and elevation of ALT/AST in wt mice persisted to 6wk pi. However, no abnormal changes attributed to vector treatment were detected in the liver in MPS IIIB mice. Therefore, we have identified genotype- and sex-specific dose-limiting toxicity of systemic rAAV9-hNAGLU delivery, supporting a safe profile for our proposed approach for treating MPS IIIB in patients.
- Published
- 2015
- Full Text
- View/download PDF
40. Evaluating the effect of a monetary incentive on performance of the 100-meter timed test in Duchenne muscular dystrophy
- Author
-
Linda H. Cripe, N. Miller, Lindsay N. Alfano, Kevin M. Flanigan, Linda Lowes, K. Berry, and J. Mendell
- Subjects
medicine.medical_specialty ,business.industry ,Duchenne muscular dystrophy ,medicine.disease ,Test (assessment) ,Incentive ,Neurology ,Pediatrics, Perinatology and Child Health ,medicine ,Physical therapy ,Metre ,Neurology (clinical) ,business ,Genetics (clinical) - Published
- 2016
41. 497. Follistatin Gene Therapy Improves Six Minute Walk Distance in Sporadic Inclusion Body Myositis (sIBM)
- Author
-
Brian K. Kaspar, Jerry R. Mendell, Sarah Lewis, Zarife Sahenk, Markus McColly, Samiah Al-Zaidy, Linda Lowes, K. Berry, Igor Dvorchik, Mark Hogan, Kevin M. Flanigan, N. Miller, Louise R. Rodino-Klapac, Kathleen Church, Melissa Moore-Clingenpeel, and Lindsay N. Alfano
- Subjects
Pharmacology ,medicine.medical_specialty ,biology ,business.industry ,Urology ,Myostatin ,Gene delivery ,medicine.disease ,Inflammatory myopathy ,Fibrosis ,Interquartile range ,Drug Discovery ,Genetics ,medicine ,biology.protein ,Molecular Medicine ,Muscular dystrophy ,Intramuscular injection ,business ,Molecular Biology ,Follistatin - Abstract
Treatment of sIBM poses many challenges. The cause of this disease is enigmatic, and although considered to be an inflammatory myopathy, there is resistance to anti-inflammatory and immunosuppressive agents. sIBM muscle biopsies show vacuolated muscle fibers, widespread inflammation, and intracellular amyloid deposits. Follistatin is a potent inhibitor of the myostatin pathway and its potential as a therapeutic vehicle is enhanced by a pathway independent of the activin IIB receptor. We have demonstrated both safety and efficacy following direct intramuscular injection of follistatin in the quadriceps muscle in a previously reported gene therapy trial in Becker muscular dystrophy (Mendell JR, et al Mol Ther 2015). No off target effects were encountered attributed to the use of an alternatively spliced follistatin isoform, FS344, also used in the current sIBM gene therapy trial. Enrollment in the current gene therapy trial included 6 subjects with either definite or possible sIBM (Griggs RC, et al. Ann Neurol 1995). Pretreatment MRI's were obtained to determine areas of relative muscle sparing/lack of fibrosis. The intramuscular injections of AAV1.CMV.FS344 to 12 to 14 sites in the quadriceps muscle delivered 1.2X1012 vg/kg. Injections were performed with direct ultrasound guidance to target the most normal appearing muscle bundles, and intramuscular position was confirmed with simultaneous EMG. A three-patient, single limb, safety trial preceded the Phase I/IIA trial reported here. During the ongoing gene therapy trial, a control sIBM group (n=20) was prospectively studied by performance of the 6MWT with follow up from 9-28 months.The 6MWT was the primary functional outcome (See table below). sIBM patients treated with AAV1.CMV.FS344 increased the 6MWT distance by 46.5m (457 to 503.5, p =0.001). Untreated sIBM controls lost 38.5m over a similar time period resulting in net difference of 85.0m between groups (p=0.0007). To validate findings and confirm the lack of selection bias we compared a subgroup of untreated sIBM controls (n=8), matched for age, gender, and 6MWD at baseline. Matched controls lost 39m (p=0.0036) in the 6MWD, a virtually identical loss to the larger control group.The results of this study demonstrate that sIBM can benefit from follistatin gene therapy based on improvement in distance walked in the 6MWT. We did find a hierarchy of response based on muscle preservation and avoiding gene delivery to areas of fibrosis. In this study, gene delivery was limited to the quadriceps muscle, but in future trials more widespread delivery could potentially be more effective.Six Minute Walk Distance Pre- and Post-Treatment* [median values (interquartile ranges provided)]GroupBaseline (m)Final Compared to Baseline (m)Change from Baseline (m)Change per Month (m)sIBM Gene Therapy Pts (n =6)457 (431,475)Improved to 503.5 (443, 573)+46.5 (2,117)+3.09 (0.39, 8.9)Untreated sIBM Controls (n =20)393 (356.5,.451.5)Declined to 354.5 (303.5,410.5)−38.5 (−73,−22) p =0.0007−2.3 (−4,−1. 1) P =0.0032Matched sIBM Controls for age, gender, and 6MWD (n = 8)459 (439.5,469)Declined to 420 (388.5,447.5)−39.0 (−77,−8) p = 0.0036−2.2 (−4.8. −0.7) P=0.0118 View Table in HTML Data analysis used SAS 9.3 (SAS Institute, Cary NC) with two-sided p-values.
- Published
- 2016
42. 624. A Single Neonatal Delivery of an Exon 2 Directed AAV9.U7snRNA Vector Results in Long-Term Dystrophin Expression That Prevents Pathologic Features in the Dup2 Mouse
- Author
-
Nicolas Wein, Kevin M. Flanigan, Felecia Gumienny, Francesco Muntoni, Jacqueline A. Yurkoski, Nianyuan Huang, and Tabatha R. Simmons
- Subjects
Pharmacology ,Gene isoform ,biology ,Myogenesis ,Duchenne muscular dystrophy ,medicine.disease ,Molecular biology ,Exon ,Internal ribosome entry site ,Drug Discovery ,Gene duplication ,Genetics ,medicine ,biology.protein ,Molecular Medicine ,Dystrophin ,Molecular Biology ,Gene - Abstract
We recently identified an internal ribosome entry site (IRES) within exon 5 of the DMD gene. Mutations that truncate the reading frame 5’ of this exon can result in use of the IRES for alternate translational initiation beginning within exon 6 that results in expression of an N-truncated isoform. Despite lacking the calponin homology domain 1 (CH1) of the actin binding domain 1 (ABD1), this isoform is highly functional, as demonstrated by the minimal symptoms in patients who express it. Consistent with genotype-phenotype correlations in DMD patients, the IRES is not active in the presence of exon 2 duplication but is active when exon 2 is deleted. We developed an AAV9.U7snRNA vector to that truncates DMD reading frame by skipping of exon 2, and have shown that in a Duchenne muscular dystrophy (DMD) mouse model carrying a duplication of exon 2 (the Dup2 mouse), postnatal intramuscular (IM) or intravascular (IV) treatment results in functional and pathologic improvement in skeletal muscle. Relevant to efforts to identify and treat DMD patients at an earlier age, we sought here to determine whether earlier gene transfer might slow down or even prevent the development of pathology. Dup2 mice were injected via facial vein at postnatal day 1 (P1) with 1E12 total vector genomes of the AAV9.U7snRNA vector and sacrificed at either 1, 3, 6, or 12 months post-injection for evaluation of exon 2 skipping by RT-PCR, quantification of dystrophin expression, and characterization of histopathology. To model the applicability of this approach beyond exon 2 duplication patients, the same vector was used to treat 6 human patient fibroblast-derived transdifferentiated myoblasts (FibroMyoD cells) harboring various mutations within exons 1 to 4. In the Dup2 mouse, efficient skipping and abundant dystrophin expression were present up to one year following the single AAV injection. Dystrophic pathology was absent at all-time points; at one year, less than 1% of fibers showed central nucleation, in comparison to ~70% in untreated Dup2 mice. Two tests on the ex vivo diaphragm preparations: isometric force (providing assessment of strength), and eccentric contractions (evaluating sarcolemma stability) were performed at 3 and 6 months following P1 injection. Both tests demonstrated little to no difference between treated animals and wild type mice. In all FibroMyoD cultures, abundant exon 2 skipping and dystrophin expression were detected in myotubes at 14 days of culture after treatment. These results suggest that this exon-skipping vector offers a therapeutic approach not only to patients with exon 2 duplications but with all mutations within the first four DMD exons (~6% of patients), an area of the gene largely ignored by the current therapeutic approaches. This work strongly supports the idea that early treatment of these patients will have longstanding and significant benefit resulting in a better outcome.
- Published
- 2016
43. Design of a phase I/II gene transfer clinical trial of rAAV9.CMV.hNAGLU for mucopolysaccharidosis type IIIB
- Author
-
Tamar Sherrod-Canaan, Douglas M. McCarty, Kevin M. Flanigan, Kim L. McBride, Kelly A. McNally, Kristen V. Truxal, Shawn C. Aylward, Timothy M. Miller, Marco Corridore, Lisa Martin, Krista L. Kunkler, Nicholas Zumberge, Christopher McKee, and Haiyan Fu
- Subjects
business.industry ,Endocrinology, Diabetes and Metabolism ,Gene transfer ,02 engineering and technology ,021001 nanoscience & nanotechnology ,030226 pharmacology & pharmacy ,Biochemistry ,Virology ,Mucopolysaccharidosis type IIIB ,Clinical trial ,03 medical and health sciences ,0302 clinical medicine ,Endocrinology ,Phase i ii ,Genetics ,Medicine ,0210 nano-technology ,business ,Molecular Biology - Published
- 2016
44. Prospective natural history study of mucopolysaccharidosis types IIIA and IIIB (Sanfilippo syndrome)
- Author
-
Shawn C. Aylward, Linda Lowes, Rebecca Grimes, Kevin M. Flanigan, Tamar Sherrod-Canaan, Haiyan Fu, Douglas M. McCarty, Christopher McKee, K. Berry, Marco Corridore, Lisa Martin, Jill Twersky, Krista Kunkler, Nicholas Zumberge, Kelly A. McNally, Lindsay N. Alfano, Kim L. McBride, and Kristen V. Truxal
- Subjects
Pediatrics ,medicine.medical_specialty ,business.industry ,Endocrinology, Diabetes and Metabolism ,Mucopolysaccharidosis ,medicine.disease ,Biochemistry ,Endocrinology ,Genetics ,medicine ,business ,Molecular Biology ,Natural history study ,Sanfilippo syndrome - Published
- 2016
45. Cerebral proton magnetic resonance spectroscopy of a patient with giant axonal neuropathy
- Author
-
Jens Frahm, Folker Hanefeld, Knut Brockmann, Kevin M. Flanigan, Peter Dechent, and Petra J. W. Pouwels
- Subjects
Magnetic Resonance Spectroscopy ,Central nervous system ,White matter ,Leukoencephalopathy ,03 medical and health sciences ,0302 clinical medicine ,Nuclear magnetic resonance ,Developmental Neuroscience ,030225 pediatrics ,medicine ,Humans ,Child ,Spectroscopy ,Giant axonal neuropathy ,medicine.diagnostic_test ,Chemistry ,business.industry ,Brain ,Infant ,Magnetic resonance imaging ,General Medicine ,Nuclear magnetic resonance spectroscopy ,medicine.disease ,Axons ,Proton magnetic resonance ,3. Good health ,Cytoskeletal Proteins ,medicine.anatomical_structure ,Mutation ,Pediatrics, Perinatology and Child Health ,Disease Progression ,Female ,Neurology (clinical) ,Nuclear medicine ,business ,Metabolism, Inborn Errors ,030217 neurology & neurosurgery ,Demyelinating Diseases - Abstract
Magnetic resonance imaging of a girl with giant axonal neuropathy revealed a progressive white matter disease. In close agreement with histopathological features reported previously, localized proton magnetic resonance spectroscopy at 9 and 12 years of age indicated a specific damage or loss of axons (reduced N-acetylaspartate and N-acetylaspartylglutamate) accompanied by acute demyelination (elevated choline-containing compounds, myo-inositol, and lactate) in white matter as well as a generalized proliferation of glial cells (elevated choline-containing compounds and myo-inositol) in both gray and white matter. (C) 2002 Elsevier Science B.V. All rights reserved.
- Published
- 2003
46. The 100-meter timed test: ability to detect change over time in Duchenne muscular dystrophy
- Author
-
J. Mendell, Lindsay N. Alfano, Samiah Al-Zaidy, C. Tsao, N. Miller, Linda Lowes, and Kevin M. Flanigan
- Subjects
Change over time ,medicine.medical_specialty ,business.industry ,Duchenne muscular dystrophy ,medicine.disease ,Test (assessment) ,Physical medicine and rehabilitation ,Neurology ,Pediatrics, Perinatology and Child Health ,Medicine ,Metre ,Neurology (clinical) ,business ,Genetics (clinical) - Published
- 2017
47. A single neonatal injection of an AAV9.U7snRNA virus mediating skipping of dmd exon 2 allows dystrophin expression preventing apparition of pathologic features in the Dup2 mouse one year post injection
- Author
-
Nicolas Wein, Francesco Muntoni, Felecia Gumienny, Nianyuan Huang, Jacqueline A. Yurkoski, Tabatha R. Simmons, Louise R. Rodino-Klapac, K. Heller, and Kevin M. Flanigan
- Subjects
Pathology ,medicine.medical_specialty ,biology ,business.industry ,Post injection ,Virology ,Virus ,Clinical neurology ,Exon ,Neurology ,Pediatrics, Perinatology and Child Health ,biology.protein ,Medicine ,Neurology (clinical) ,business ,Dystrophin ,Genetics (clinical) - Published
- 2017
48. Daily versus weekend steroid use in DMD: age at loss of ambulation is equivalent in a retrospective patient cohort
- Author
-
Megan A. Waldrop, Kevin M. Flanigan, J. Kaminoh, and M. Moore-Clingenpeel
- Subjects
0301 basic medicine ,Pediatrics ,medicine.medical_specialty ,business.industry ,03 medical and health sciences ,030104 developmental biology ,Neurology ,Steroid use ,Pediatrics, Perinatology and Child Health ,Cohort ,Medicine ,Neurology (clinical) ,business ,Genetics (clinical) - Published
- 2017
49. Reduced Hedgehog signalling in Duchenne muscular dystrophy impairs muscle regeneration and function
- Author
-
H. Wang, N. Salgado, Christopher M. Penton, Paul M.L. Janssen, Kevin M. Flanigan, S. Devenport, and Federica Montanaro
- Subjects
Muscle regeneration ,Hedgehog signalling ,Neurology ,business.industry ,Duchenne muscular dystrophy ,Pediatrics, Perinatology and Child Health ,medicine ,Neurology (clinical) ,medicine.disease ,business ,Genetics (clinical) ,Function (biology) ,Cell biology - Published
- 2017
50. Systemic gene transfer of scAAV9.U1a.hSGSH for MPS IIIA: tolerability and preliminary evidence for a biochemical effect
- Author
-
Douglas M. McCarty, Kevin M. Flanigan, Kim L. McBride, Haiyan Fu, Kristen V. Truxal, Shawn C. Aylward, Marco Corridore, Lisa Martin, Christopher McKee, Nicholas Zumberge, Krista L. Kunkler, and Kelly A. McNally
- Subjects
Endocrinology ,Tolerability ,business.industry ,Endocrinology, Diabetes and Metabolism ,Genetics ,Medicine ,Gene transfer ,Pharmacology ,business ,Molecular Biology ,Biochemistry - Published
- 2017
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.