Network


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

Hotspot


Dive into the research topics where Paula R. Clemens is active.

Publication


Featured researches published by Paula R. Clemens.


Lancet Neurology | 2010

Diagnosis and management of Duchenne muscular dystrophy, part 1: diagnosis, and pharmacological and psychosocial management

K. Bushby; R. Finkel; David J. Birnkrant; Laura E. Case; Paula R. Clemens; Linda H. Cripe; Ajay Kaul; Kathi Kinnett; Craig M. McDonald; Shree Pandya; James Poysky; Frederic Shapiro; Jean Tomezsko; Carolyn Constantin

Duchenne muscular dystrophy (DMD) is a severe, progressive disease that affects 1 in 3600-6000 live male births. Although guidelines are available for various aspects of DMD, comprehensive clinical care recommendations do not exist. The US Centers for Disease Control and Prevention selected 84 clinicians to develop care recommendations using the RAND Corporation-University of California Los Angeles Appropriateness Method. The DMD Care Considerations Working Group evaluated assessments and interventions used in the management of diagnostics, gastroenterology and nutrition, rehabilitation, and neuromuscular, psychosocial, cardiovascular, respiratory, orthopaedic, and surgical aspects of DMD. These recommendations, presented in two parts, are intended for the wide range of practitioners who care for individuals with DMD. They provide a framework for recognising the multisystem primary manifestations and secondary complications of DMD and for providing coordinated multidisciplinary care. In part 1 of this Review, we describe the methods used to generate the recommendations, and the overall perspective on care, pharmacological treatment, and psychosocial management.


Lancet Neurology | 2010

Diagnosis and management of Duchenne muscular dystrophy, part 2: implementation of multidisciplinary care

K. Bushby; R. Finkel; David J. Birnkrant; Laura E. Case; Paula R. Clemens; Linda H. Cripe; Ajay Kaul; Kathi Kinnett; Craig M. McDonald; Shree Pandya; James Poysky; Frederic Shapiro; Jean Tomezsko; Carolyn Constantin

Optimum management of Duchenne muscular dystrophy (DMD) requires a multidisciplinary approach that focuses on anticipatory and preventive measures as well as active interventions to address the primary and secondary aspects of the disorder. Implementing comprehensive management strategies can favourably alter the natural history of the disease and improve function, quality of life, and longevity. Standardised care can also facilitate planning for multicentre trials and help with the identification of areas in which care can be improved. Here, we present a comprehensive set of DMD care recommendations for management of rehabilitation, orthopaedic, respiratory, cardiovascular, gastroenterology/nutrition, and pain issues, as well as general surgical and emergency-room precautions. Together with part 1 of this Review, which focuses on diagnosis, pharmacological treatment, and psychosocial care, these recommendations allow diagnosis and management to occur in a coordinated multidisciplinary fashion.


The New England Journal of Medicine | 2010

A Randomized Study of Alglucosidase Alfa in Late-Onset Pompe's Disease

Ans T. van der Ploeg; Paula R. Clemens; Deyanira Corzo; Diana M. Escolar; Julaine Florence; Geert Jan Groeneveld; Serge Herson; Priya S. Kishnani; P. Laforêt; Stephen Lake; Dale J. Lange; Robert Leshner; J. Mayhew; Claire Morgan; Kenkichi Nozaki; Dorothy J. Park; Alan Pestronk; Barry E. Rosenbloom; Alison Skrinar; Carine I. van Capelle; Nadine A. van der Beek; Melissa P. Wasserstein; Sasa Zivkovic

BACKGROUND Pompes disease is a metabolic myopathy caused by a deficiency of acid alpha glucosidase (GAA), an enzyme that degrades lysosomal glycogen. Late-onset Pompes disease is characterized by progressive muscle weakness and loss of respiratory function, leading to early death. We conducted a randomized, placebo-controlled trial of alglucosidase alfa, a recombinant human GAA, for the treatment of late-onset Pompes disease. METHODS Ninety patients who were 8 years of age or older, ambulatory, and free of invasive ventilation were randomly assigned to receive biweekly intravenous alglucosidase alfa (20 mg per kilogram of body weight) or placebo for 78 weeks at eight centers in the United States and Europe. The two primary end points were distance walked during a 6-minute walk test and percentage of predicted forced vital capacity (FVC). RESULTS At 78 weeks, the estimated mean changes from baseline in the primary end points favored alglucosidase alfa (an increase of 28.1+/-13.1 m on the 6-minute walk test and an absolute increase of 3.4+/-1.2 percentage points in FVC; P=0.03 and P=0.006, respectively). Similar proportions of patients in the two groups had adverse events, serious adverse events, and infusion-associated reactions; events that occurred only in patients who received the active study drug included anaphylactic reactions and infusion-associated reactions of urticaria, flushing, hyperhidrosis, chest discomfort, vomiting, and increased blood pressure (each of which occurred in 5 to 8% of the patients). CONCLUSIONS In this study population, treatment with alglucosidase alfa was associated with improved walking distance and stabilization of pulmonary function over an 18-month period. (ClinicalTrials.gov number, NCT00158600.)


Journal of Clinical Investigation | 2012

NF-κB inhibition delays DNA damage–induced senescence and aging in mice

Jeremy S. Tilstra; Andria Rasile Robinson; Jin Wang; Siobhán Q. Gregg; Cheryl L. Clauson; Daniel P. Reay; Luigi Aurelio Nasto; Claudette M. St. Croix; Arvydas Usas; Nam Vo; Johnny Huard; Paula R. Clemens; Donna B. Stolz; Denis C. Guttridge; Simon C. Watkins; George A. Garinis; Yinsheng Wang; Laura J. Niedernhofer; Paul D. Robbins

The accumulation of cellular damage, including DNA damage, is thought to contribute to aging-related degenerative changes, but how damage drives aging is unknown. XFE progeroid syndrome is a disease of accelerated aging caused by a defect in DNA repair. NF-κB, a transcription factor activated by cellular damage and stress, has increased activity with aging and aging-related chronic diseases. To determine whether NF-κB drives aging in response to the accumulation of spontaneous, endogenous DNA damage, we measured the activation of NF-κB in WT and progeroid model mice. As both WT and progeroid mice aged, NF-κB was activated stochastically in a variety of cell types. Genetic depletion of one allele of the p65 subunit of NF-κB or treatment with a pharmacological inhibitor of the NF-κB-activating kinase, IKK, delayed the age-related symptoms and pathologies of progeroid mice. Additionally, inhibition of NF-κB reduced oxidative DNA damage and stress and delayed cellular senescence. These results indicate that the mechanism by which DNA damage drives aging is due in part to NF-κB activation. IKK/NF-κB inhibitors are sufficient to attenuate this damage and could provide clinical benefit for degenerative changes associated with accelerated aging disorders and normal aging.


Muscle & Nerve | 2001

CLINICAL EVALUATOR RELIABILITY FOR QUANTITATIVE AND MANUAL MUSCLE TESTING MEASURES OF STRENGTH IN CHILDREN

Diana M. Escolar; Erik Henricson; J. Mayhew; Julaine Florence; Robert Leshner; K. M. Patel; Paula R. Clemens

Measurements of muscle strength in clinical trials of Duchenne muscular dystrophy have relied heavily on manual muscle testing (MMT). The high level of intra‐ and interrater variability of MMT compromises clinical study results. We compared the reliability of 12 clinical evaluators in performing MMT and quantitative muscle testing (QMT) on 12 children with muscular dystrophy. QMT was reliable, with an interclass correlation coefficient (ICC) of >0.9 for biceps and grip strength, and >0.8 for quadriceps strength. Training of both subjects and evaluators was easily accomplished. MMT was not as reliable, and required repeated training of evaluators to bring all groups to an ICC >0.75 for shoulder abduction, elbow and hip flexion, knee extension, and ankle dorsiflexion. We conclude that QMT shows greater reliability and is easier to implement than MMT. Consequently, QMT will be a superior measure of strength for use in pediatric, neuromuscular, multicenter clinical trials.


Molecular Therapy | 2011

Current Status of Pharmaceutical and Genetic Therapeutic Approaches to Treat DMD

Christophe Pichavant; Annemieke Aartsma-Rus; Paula R. Clemens; Kay E. Davies; George Dickson; Shin'ichi Takeda; S.D. Wilton; Jon A. Wolff; Christine I. Wooddell; Xiao Xiao; Jacques P. Tremblay

Duchenne muscular dystrophy (DMD) is a genetic disease affecting about one in every 3,500 boys. This X-linked pathology is due to the absence of dystrophin in muscle fibers. This lack of dystrophin leads to the progressive muscle degeneration that is often responsible for the death of the DMD patients during the third decade of their life. There are currently no curative treatments for this disease but different therapeutic approaches are being studied. Gene therapy consists of introducing a transgene coding for full-length or a truncated version of dystrophin complementary DNA (cDNA) in muscles, whereas pharmaceutical therapy includes the use of chemical/biochemical substances to restore dystrophin expression or alleviate the DMD phenotype. Over the past years, many potential drugs were explored. This led to several clinical trials for gentamicin and ataluren (PTC124) allowing stop codon read-through. An alternative approach is to induce the expression of an internally deleted, partially functional dystrophin protein through exon skipping. The vectors and the methods used in gene therapy have been continually improving in order to obtain greater encapsidation capacity and better transduction efficiency. The most promising experimental approaches using pharmaceutical and gene therapies are reviewed in this article.


Muscle & Nerve | 2013

THE COOPERATIVE INTERNATIONAL NEUROMUSCULAR RESEARCH GROUP DUCHENNE NATURAL HISTORY STUDY: GLUCOCORTICOID TREATMENT PRESERVES CLINICALLY MEANINGFUL FUNCTIONAL MILESTONES AND REDUCES RATE OF DISEASE PROGRESSION AS MEASURED BY MANUAL MUSCLE TESTING AND OTHER COMMONLY USED CLINICAL TRIAL OUTCOME MEASURES

Erik K. Henricson; R. Ted Abresch; Avital Cnaan; F. Hu; T. Duong; A. Arrieta; Jay Han; D. Escolar; Julaine Florence; Paula R. Clemens; Eric P. Hoffman; Craig M. McDonald

Introduction: Glucocorticoid (GC) therapy in Duchenne muscular dystrophy (DMD) has altered disease progression, necessitating contemporary natural history studies. Methods: The Cooperative Neuromuscular Research Group (CINRG) DMD Natural History Study (DMD‐NHS) enrolled 340 DMD males, ages 2–28 years. A comprehensive battery of measures was obtained. Results: A novel composite functional “milestone” scale scale showed clinically meaningful mobility and upper limb abilities were significantly preserved in GC‐treated adolescents/young adults. Manual muscle test (MMT)‐based calculations of global strength showed that those patients <10 years of age treated with steroids declined by 0.4±0.39 MMT unit/year, compared with −0.4±0.39 MMT unit/year in historical steroid‐naive subjects. Pulmonary function tests (PFTs) were relatively preserved in steroid‐treated adolescents. The linearity and magnitude of decline in measures were affected by maturational changes and functional status. Conclusions: In DMD, long‐term use of GCs showed reduced strength loss and preserved functional capabilities and PFTs compared with previous natural history studies performed prior to the widespread use of GC therapy. Muscle Nerve, 2013


Gene Therapy | 1998

Ex vivo gene transfer using adenovirus-mediated full-length dystrophin delivery to dystrophic muscles

S. S. Floyd; Paula R. Clemens; M. R. Ontell; Stefan Kochanek; C. S. Day; Yang Jp; S. D. Hauschka; L. Balkir; Je Morgan; M. S. Moreland; G. W. Feero; Michael W. Epperly; Johnny Huard

Duchenne muscular dystrophy (DMD) is an X-linked recessive muscle disease characterized by a lack of dystrophin expression. Myoblast transplantation and gene therapy have the potential of restoring dystrophin, thus decreasing the muscle weakness associated with this disease. In this study we present data on the myoblast mediated ex vivo gene transfer of full-length dystrophin to mdx (dystrophin deficient) mouse muscle as a model for autologous myoblast transfer. Both isogenic primary mdx myoblasts and an immortalized mdx cell line were transduced with an adenoviral vector that has all viral coding sequences deleted and encodes β-galactosidase and full-length dystrophin. Subsequently, these transduced myoblasts were injected into dystrophic mdx muscle, where the injected cells restored dystrophin, as well as dystrophin- associated proteins. A greater amount of dystrophin replacement occurred in mdx muscle following transplantation of mdx myoblasts isolated from a transgenic mouse overexpressing dystrophin suggesting that engineering autologous myoblasts to express high amounts of dystrophin might be beneficial. The ex vivo approach possesses attributes that make it useful for gene transfer to skeletal muscle including: (1) creating a reservoir of myoblasts capable of regenerating and restoring dystrophin to dystrophic muscle; and (2) achieving a higher level of gene transfer to dystrophic muscle compared with adenovirus-mediated direct gene delivery. However, as observed in direct gene transfer studies, the ex vivo approach also triggers a cellular immune response which limits the duration of transgene expression.


Human Gene Therapy | 1999

Polylysine Modification of Adenoviral Fiber Protein Enhances Muscle Cell Transduction

Khaled Bouri; W. Greg Feero; Mike M. Myerburg; Thomas J. Wickham; Imre Kovesdi; Eric P. Hoffman; Paula R. Clemens

Adenoviral vectors (ADVs) are used widely for gene delivery to different tissues including muscle. One particularly promising use for ADVs is in the transfer of the dystrophin gene to the muscle of patients with Duchenne muscular dystrophy (DMD). However, studies in different animal models of DMD suggest that ADVs inefficiently transduce mature skeletal muscle. In this article we test whether AdZ.F(pK7), a genetically modified ADV that expresses a polylysine moiety on the end of the fiber protein, could enhance transduction of muscle cells and circumvent the maturation-dependent loss of muscle infectivity by ADVs. The efficiency of transduction was tested at different levels of muscle maturation. In vitro, AdZ.F(pK7) showed a higher level of transduction at all stages of differentiation including myoblasts, myotubes, and single muscle fibers. In vivo, mature skeletal muscle was transduced fourfold better by AdZ.F(pK7) than by the unmodifled vector (AdZ.F). Together, these observations demonstrate improved ADV transduction of skeletal muscle by modifying ADV tropism, and provide a proof-of-principle that modification of ADVs to target muscle-specific molecules could result in tissue-specific transfer of skeletal muscle tissue as well.


Annals of Neurology | 2005

CINRG randomized controlled trial of creatine and glutamine in Duchenne muscular dystrophy

Diana M. Escolar; Gunnar Buyse; Erik Henricson; Robert Leshner; Julaine Florence; J. Mayhew; Carolina Tesi-Rocha; Ksenija Gorni; Livia Pasquali; Kantilal M. Patel; Robert McCarter; Jennifer Huang; Thomas P. Mayhew; Tulio E. Bertorini; Jose Carlo; Anne M. Connolly; Paula R. Clemens; Nathalie Goemans; Susan T. Iannaccone; Masanori Igarashi; Yoram Nevo; Alan Pestronk; S. H. Subramony; V. V. Vedanarayanan; Henry B. Wessel

We tested the efficacy and safety of glutamine (0.6gm/kg/day) and creatine (5gm/day) in 50 ambulant boys with Duchenne muscular dystrophy in a 6‐month, double‐blind, placebo‐controlled clinical trial. Drug efficacy was tested by measuring muscle strength manually (34 muscle groups) and quantitatively (10 muscle groups). Timed functional tests, functional parameters, and pulmonary function tests were secondary outcome measures. Although there was no statistically significant effect of either therapy based on manual and quantitative measurements of muscle strength, a disease‐modifying effect of creatine in older Duchenne muscular dystrophy and creatine and glutamine in younger Duchenne muscular dystrophy cannot be excluded. Creatine and glutamine were well tolerated. Ann Neurol 2005;58:151–155

Collaboration


Dive into the Paula R. Clemens's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel P. Reay

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Lauren P. Morgenroth

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Eric P. Hoffman

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Avital Cnaan

Children's National Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erik Henricson

University of California

View shared research outputs
Top Co-Authors

Avatar

Heather Gordish-Dressman

Children's National 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

Anne M. Connolly

Washington University in St. Louis

View shared research outputs
Researchain Logo
Decentralizing Knowledge