Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Wendy M. King is active.

Publication


Featured researches published by Wendy M. King.


The New England Journal of Medicine | 1989

Randomized, Double-Blind Six-Month Trial of Prednisone in Duchenne's Muscular Dystrophy

Richard T. Moxley; Robert C. Griggs; M.H. Brooke; G.M. Fenichel; J.P. Miller; Wendy M. King; Linda Signore; Shree Pandya; J. Florence; J. Schierbecker; J. Robison; K. Kaiser; Stephen Mandel; C. Arfken; B. Gilder

We performed a randomized, double-blind, controlled six-month trial of prednisone in 103 boys with Duchennes muscular dystrophy (age, 5 to 15 years). The patients were assigned to one of three regimens: prednisone, 0.75 mg per kilogram of body weight per day (n = 33); prednisone, 1.5 mg per kilogram per day (n = 34); or placebo (n = 36). The groups were initially comparable in all measures of muscle function. Both prednisone groups had significant improvement of similar degree in the summary scores of muscle strength and function. Improvement began as early as one month and peaked by three months. At six months the high-dose prednisone group, as compared with the placebo group, had improvement in the time needed to rise from a supine to a standing position (3.4 vs. 6.2 seconds), to walk 9 m (7.0 vs. 9.7 seconds), and to climb four stairs (4.0 vs. 7.1 seconds), in lifting a weight (2.1 vs. 1.2 kg), and in forced vital capacity (1.7 vs. 1.5 liters) (P less than 0.001 for all comparisons). There was an increase in urinary creatinine excretion (261 vs. 190 mg per 24 hours), which suggested an increase in total muscle mass. However, the prednisone-treated patients who had required long-leg braces (n = 5) or wheelchairs (n = 11) continued to require them. The most frequent side effects were weight gain, cushingoid appearance, and excessive hair growth. We conclude from this six-month study that prednisone improves the strength and function of patients with Duchennes muscular dystrophy. However, further research is required to identify the mechanisms responsible for these improvements and to determine whether prolonged treatment with corticosteroids may be warranted despite their side effects.


Annals of Neurology | 2008

A Phase I/II trial of MYO-029 in Adult Subjects with Muscular Dystrophy

Kathryn R. Wagner; James L. Fleckenstein; Anthony A. Amato; Richard J. Barohn; K. Bushby; Diana M. Escolar; Kevin M. Flanigan; Alan Pestronk; Rabi Tawil; Gil I. Wolfe; Michelle Eagle; Julaine Florence; Wendy M. King; Shree Pandya; Volker Straub; Paul Juneau; Kathleen Meyers; Cristina Csimma; Tracey Araujo; Robert Allen; Stephanie A. Parsons; John M. Wozney; Edward R. LaVallie

Myostatin is an endogenous negative regulator of muscle growth and a novel target for muscle diseases. We conducted a safety trial of a neutralizing antibody to myostatin, MYO‐029, in adult muscular dystrophies (Becker muscular dystrophy, facioscapulohumeral dystrophy, and limb‐girdle muscular dystrophy).


Neurology | 2001

Randomized controlled trial of IVIg in untreated chronic inflammatory demyelinating polyradiculoneuropathy

J. R. Mendell; Richard J. Barohn; Miriam Freimer; John T. Kissel; Wendy M. King; H. N. Nagaraja; R. Rice; W. W. Campbell; Peter D. Donofrio; Carlayne E. Jackson; Richard A. Lewis; Michael E. Shy; D. M. Simpson; Gareth Parry; Michael H. Rivner; Charles A. Thornton; Mark B. Bromberg; Rup Tandan; Yadollah Harati; M. J. Giuliani

Objective: To determine the efficacy of IV immunoglobulin (IVIg) given patients with untreated chronic inflammatory demyelinating polyradiculoneuropathy (CIDP). Methods: A randomized, double-blind, multicenter, investigator-initiated study compared IVIg (Aventis Behring LLC, King of Prussia, PA) with placebo (5% albumin). On days 1, 2, and 21, IVIg (1 g/kg) or placebo was given. The primary outcome measure was the change in muscle strength from baseline to day 42, using the average muscle score (AMS). Secondary outcome measures included change from baseline AMS at days 10 and 21, the Hughes’ functional disability scale, forced vital capacity (FVC), and nerve conduction studies (NCS) of four motor nerves (median, ulnar, peroneal, and tibial). Results: The patients (n = 33) were randomized. Of these, 30 (14 women, 16 men, aged 54 ± 20 years, range 13 to 82) received IVIg and 23 were given placebo (12 women, 11 men, aged 50 ± 18 years, range 23 to 73). Baseline AMS values of the groups were similar (IVIg 7.06 ± 1.31 versus placebo 7.28 ± 1.18, p = 0.53). There were two dropouts in placebo group and one in the IVIg group. Mean AMS improved at day 42 comparing IVIg with placebo (0.63 versus −0.1, p = 0.006). Improved strength was seen by day 10. The placebo group lost strength over this same interval. In the IVIg, 11 subjects improved by the functional disability scale; none worsened. This differed (p = 0.019) from those in the placebo-treated group (two improved, two got worse, remainder unchanged). Forced vital capacity did not improve with IVIg treatment. IVIg improved ulnar motor distal latency (p = 0.005), tibial distal compound muscle amplitude (p = 0.003), and peroneal nerve conduction velocity (p = 0.03). Conclusions: IVIg improves strength in patients with untreated CIDP by day 10 with continued benefit through day 42; more than one third improve by at least a functional grade on a disability scale. This study provides data supporting IVIg as the initial treatment for CIDP.


Neurology | 2009

Subcutaneous IGF-1 is not beneficial in 2-year ALS trial

Eric J. Sorenson; A. J. Windbank; Jayawant N. Mandrekar; William R. Bamlet; Stanley H. Appel; Carmel Armon; Paul E. Barkhaus; Peter E. Bosch; Kevin B. Boylan; William S. David; Eva L. Feldman; Jonathan D. Glass; Laurie Gutmann; J. I. Katz; Wendy M. King; Carlos A. Luciano; Leo McCluskey; Steven Nash; D. S. Newman; Robert M. Pascuzzi; Erik P. Pioro; L. J. Sams; Stephen N. Scelsa; Ericka Simpson; S. H. Subramony; Ezgi Tiryaki; Charles A. Thornton

Background: Previous human clinical trials of insulin-like growth factor type I (IGF-1) in amyotrophic lateral sclerosis (ALS) have been inconsistent. This phase III, randomized, double-blind, placebo-controlled study was undertaken to address whether IGF-1 benefited patients with ALS. Methods: A total of 330 patients from 20 medical centers were randomized to receive 0.05 mg/kg body weight of human recombinant IGF-1 given subcutaneously twice daily or placebo for 2 years. The primary outcome measure was change in their manual muscle testing score. Secondary outcome measures included tracheostomy-free survival and rate of change in the revised ALS functional rating scale. Intention to treat analysis was used. Results: There was no difference between treatment groups in the primary or secondary outcome measures after the 2-year treatment period. Conclusions: Insulin-like growth factor type I does not provide benefit for patients with amyotrophic lateral sclerosis. GLOSSARY: ALS = amyotrophic lateral sclerosis; ALSFRS-r = revised ALS functional rating scale; AUC = area under the curve; DVT = deep venous thromboses; IGF-1 = insulin-like growth factor type I; MMT = manual muscle testing; PE = pulmonary embolisms.


Annals of Neurology | 2010

Gentamicin-induced readthrough of stop codons in duchenne muscular dystrophy

Vinod Malik; L. Rodino-Klapac; Laurence Viollet; Cheryl Wall; Wendy M. King; Roula al-Dahhak; Sarah Lewis; C. Shilling; Janaiah Kota; Carmen Serrano-Munuera; John R. Hayes; John D. Mahan; Katherine J. Campbell; Brenda Banwell; Majed Dasouki; Victoria Watts; Kumaraswamy Sivakumar; Ricardo Bien-Willner; Kevin M. Flanigan; Zarife Sahenk; Richard J. Barohn; Christopher M. Walker

The objective of this study was to establish the feasibility of long‐term gentamicin dosing to achieve stop codon readthrough and produce full‐length dystrophin. Mutation suppression of stop codons, successfully achieved in the mdx mouse using gentamicin, represents an important evolving treatment strategy in Duchenne muscular dystrophy (DMD).


Human Mutation | 2009

Mutational spectrum of DMD mutations in dystrophinopathy patients: application of modern diagnostic techniques to a large cohort

Kevin M. Flanigan; Diane M. Dunn; Andrew von Niederhausern; Payam Soltanzadeh; Eduard Gappmaier; Michael T. Howard; Jacinda Sampson; Cheryl Wall; Wendy M. King; Alan Pestronk; Julaine Florence; Anne M. Connolly; Katherine D. Mathews; Carrie M. Stephan; Karla S. Laubenthal; Brenda Wong; P. Morehart; Amy Meyer; Richard S. Finkel; Carsten G. Bönnemann; Livija Medne; John W. Day; Joline Dalton; Marcia Margolis; Veronica J. Hinton; Robert B. Weiss

Mutations in the DMD gene, encoding the dystrophin protein, are responsible for the dystrophinopathies Duchenne Muscular Dystrophy (DMD), Becker Muscular Dystrophy (BMD), and X‐linked Dilated Cardiomyopathy (XLDC). Mutation analysis has traditionally been challenging, due to the large gene size (79 exons over 2.2 Mb of genomic DNA). We report a very large aggregate data set comprised of DMD mutations detected in samples from patients enrolled in the United Dystrophinopathy Project, a multicenter research consortium, and in referral samples submitted for mutation analysis with a diagnosis of dystrophinopathy. We report 1,111 mutations in the DMD gene, including 891 mutations with associated phenotypes. These results encompass 506 point mutations (including 294 nonsense mutations) and significantly expand the number of mutations associated with the dystrophinopathies, highlighting the utility of modern diagnostic techniques. Our data supports the uniform hypermutability of CGA>TGA mutations, establishes the frequency of polymorphic muscle (Dp427m) protein isoforms and reveals unique genomic haplotypes associated with “private” mutations. We note that 60% of these patients would be predicted to benefit from skipping of a single DMD exon using antisense oligonucleotide therapy, and 62% would be predicted to benefit from an inclusive multiexonskipping approach directed toward exons 45 through 55. Hum Mutat 30:1657–1666, 2009.


Journal of Neuropathology and Experimental Neurology | 2006

Limb-Girdle Muscular Dystrophy in the United States

Steven A. Moore; C. Shilling; Steven Westra; Cheryl Wall; Matthew Wicklund; Catherine A. Stolle; Charlotte A. Brown; Daniel E. Michele; F. Piccolo; Thomas L. Winder; Aaron Stence; Rita Barresi; Nick King; Wendy M. King; Julaine Florence; Kevin P. Campbell; Gerald M. Fenichel; Hansell H. Stedman; John T. Kissel; Robert C. Griggs; Shree Pandya; Katherine D. Mathews; Alan Pestronk; Carmen Serrano; Daniel Darvish

Limb-girdle muscular dystrophy (LGMD) has been linked to 15 chromosomal loci, 7 autosomal-dominant (LGMD1A to E) and 10 autosomal-recessive (LGMD2A to J). To determine the distribution of subtypes among patients in the United States, 6 medical centers evaluated patients with a referral diagnosis of LGMD. Muscle biopsies provided histopathology and immunodiagnostic testing, and their protein abnormalities along with clinical parameters directed mutation screening. The diagnosis in 23 patients was a disorder other than LGMD. Of the remaining 289 unrelated patients, 266 had muscle biopsies sufficient for complete microscopic evaluation; 121 also underwent Western blotting. From this combined evaluation, the distribution of immunophenotypes is 12% calpainopathy, 18% dysferlinopathy, 15% sarcoglycanopathy, 15% dystroglycanopathy, and 1.5% caveolinopathy. Genotypes distributed among 2 dominant and 7 recessive subtypes have been determined for 83 patients. This study of a large racially and ethnically diverse population of patients with LGMD indicates that establishing a putative subtype is possible more than half the time using available diagnostic testing. An efficient approach to genotypic diagnosis is muscle biopsy immunophenotyping followed by directed mutational analysis. The most common LGMDs in the United States are calpainopathies, dysferlinopathies, sarcoglycanopathies, and dystroglycanopathies.


Annals of Neurology | 2013

LTBP4 genotype predicts age of ambulatory loss in duchenne muscular dystrophy

Kevin M. Flanigan; Ermelinda Ceco; Kay Marie Lamar; Yuuki Kaminoh; Diane M. Dunn; Wendy M. King; Alan Pestronk; Julaine Florence; Katherine D. Mathews; Richard S. Finkel; Kathryn J. Swoboda; Eduard Gappmaier; Michael T. Howard; John W. Day; Craig M. McDonald; Elizabeth M. McNally; Robert B. Weiss

Duchenne muscular dystrophy (DMD) displays a clinical range that is not fully explained by the primary DMD mutations. Ltbp4, encoding latent transforming growth factor‐β binding protein 4, was previously discovered in a genome‐wide scan as a modifier of murine muscular dystrophy. We sought to determine whether LTBP4 genotype influenced DMD severity in a large patient cohort.


Human Molecular Genetics | 1993

Characterization of translational frame exception patients in Duchenne/Becker muscular dystrophy

Alissa V. Winnard; Christopher J. Klein; Daniel D. Coovert; Thomas W. Prior; Audrey C. Papp; Pamela J. Snyder; Dennis E. Bulman; Peter N. Ray; Patricia McAndrew; Wendy M. King; Richard T. Moxley; Arthur H.M. Burghes

The clinical progression of Duchenne muscular dystrophy (DMD) patients with deletions can be predicted in 93% of cases by whether the deletion maintains or disrupts the translational reading frame (frameshift hypothesis). We have identified and studied a number of patients who have deletions that do not conform to the translational frame hypothesis. The most common exception to the frameshift hypothesis is the deletion of exons 3 to 7 which disrupts the translational reading frame. We identified a Becker muscular dystrophy (BMD) patient, an intermediate, and a DMD patient with this deletion. In all three cases, dystrophin was detected and localized to the membrane. One DMD patient with an inframe deletion of exons 4-18 produced no dystrophin. One patient with a mild intermediate phenotype and a deletion of exon 45, which shifts the reading frame, produced no dystrophin. Two patients with large inframe deletions had discordant phenotypes (exons 3-41, DMD; exons 13-48, BMD), but both produced dystrophin that localized to the sarcolemma. The DMD patient, 113, indicates that dystrophin with an intact carboxy terminus can be produced in Duchenne patients at levels equivalent to some Beckers. The dystrophin analysis from these patients, together with patients reported in the literature, indicate that more than one domain can localize dystrophin to the sarcolemma. Lastly, the data shows that although most patients show correlation of clinical severity to molecular data, there are rare patients which do not conform.


Neurology | 1998

Pilot trial of albuterol in facioscapulohumeral muscular dystrophy

John T. Kissel; Michael P. McDermott; Rajeshwari Natarajan; Shree Pandya; Wendy M. King; Robert C. Griggs; Rabi Tawil

Background/Objective: Facioscapulohumeral muscular dystrophy (FSHD) is currently untreatable, and there have been few therapeutic trials of any agent in the disease. Animal studies have demonstrated that β2-adrenergic agonists induce muscle hypertrophy and prevent atrophy after a variety of physical and biochemical insults, and two human studies have shown that these agents increase certain measures of strength in healthy volunteers. We conducted an open-label pilot trial of a β2-agonist (albuterol) in patients with FSHD. Methods: Fifteen FSHD patients were given sustained-release albuterol (16.0 mg/day) for 3 months. The primary outcome measure was lean body mass, which was assessed through dual energy X-ray absorptiometry (DEXA). Strength was evaluated through maximal voluntary isometric contraction testing (MVICT) and manual muscle testing. Results: Albuterol significantly increased DEXA lean body mass (the skeletal muscle compartment) by 1.29 ± 1.18 kg (mean ± SD, p = 0.001). Strength assessed through composite MVICT scores also increased by an average of 0.33 ± 0.60 (p = 0.05), representing an overall 12% improvement in strength. Conclusions: These encouraging results suggest that β2-agonists may have a role in treating FSHD and possibly other neuromuscular diseases. The effects of albuterol in FSHD are currently being evaluated in a larger, randomized, double-blind, placebo-controlled trial lasting 1 year.

Collaboration


Dive into the Wendy M. King's collaboration.

Top Co-Authors

Avatar

Shree Pandya

University of Rochester

View shared research outputs
Top Co-Authors

Avatar

Julaine Florence

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kevin M. Flanigan

Nationwide Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Richard T. Moxley

University of Rochester Medical Center

View shared research outputs
Top Co-Authors

Avatar

Alan Pestronk

Washington University in St. Louis

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge