August 3, 2011

Study Explains Why Muscles Weaken with Age and Points to Possible Therapy

 
Released: 8/1/2011 8:00 AM EDT 
Embargo expired: 8/2/2011 12:00 PM EDT
Source: Columbia University Medical Center

 

EMBARGOED UNTIL: 12 NOON (ET), TUESDAY, AUGUST 2, 2011

Newswise — (NEW YORK, NY, (August 2, 2011) – Researchers at Columbia University Medical Center have discovered the biological mechanism behind age-related loss of muscle strength and identified a drug that may help reverse this process. Their findings were published in the August 2 online edition of Cell Metabolism.

As we grow older, our skeletal muscles tend to wither and weaken, a phenomenon known as sarcopenia. Sarcopenia, which begins to appear at around age 40 and accelerates after 75, is a major cause of disability in the elderly. Exercise can help counter the effects of age-related muscle loss. Otherwise, there are no established treatments.

According to the new study, conducted in mice, sarcopenia occurs when calcium leaks from a group of proteins in muscle cells called the ryanodine receptor channel complex. These leaks then trigger a chain of events that ultimately limits the ability of muscle fibers to contract, reports study leader Andrew R. Marks, M.D., chairman and professor of physiology and cellular biophysics, the Clyde and Helen Wu Professor of Medicine, and director of the Wu Center for Molecular Cardiology at Columbia University Medical Center (CUMC).

Ryanodine receptors, which are calcium channels found in most body tissues, have been the focus of Dr. Marks’ research since 1989. After cloning the ryanodine receptor gene, he later discovered, in studies of mice, that leaky ryanodine receptors are involved in the development of heart failure and arrhythmias. In 2009, he showed that leaks in these channels also contribute to Duchenne muscular dystrophy, a genetic disorder characterized by rapidly progressing muscle weakness and early death.

Since muscular dystrophy and sarcopenia have some commonalities, Dr. Marks suspected that ryanodine receptor leakage may also be involved in age-related muscle loss, which the present study shows is the case.

“This is a completely new concept — that the damage that occurs in aging is very similar to what happens in muscular dystrophy,” says Dr. Marks, “thus as we age we essentially develop an acquired form of muscular dystrophy.”

Both the aging process and the genetic defect responsible for muscular dystrophy cause an increase in the production of oxygen free radicals, highly reactive and harmful molecules. “Our data suggest that this sets up a vicious cycle, in which the free radicals cause ryanodine receptors to leak calcium into the cell. The calcium poisons mitochondria — organelles that power the cell — leading to the release of even more free radicals. This, in turn, causes more calcium leakage. With less calcium available for contraction, the muscles get weaker,” says first author Daniel C. Andersson, M.D., Ph.D., a postdoctoral fellow in physiology and cellular biophysics at CUMC.

The study also points to a possible therapy for sarcopenia: an experimental drug called S107, developed by Dr. Marks and his colleagues. The drug acts by stabilizing calstabin1, a protein that binds to ryanodine receptors and prevents calcium leakage.

In the study, 24-month-old mice (roughly the equivalent of 70-year-old humans) were given S107 for four weeks. The mice showed significant improvements in both muscle force and exercise capacity, compared with untreated controls. “The mice ran farther and faster during voluntary exercise,” says Dr. Andersson. “When we tested their muscles, they were about 50 percent stronger.” The drug had no effect on younger mice with normal ryanodine receptors.

A similar drug is now in phase II clinical trials for the treatment of heart failure.

“Most investigators in the field of aging have been saying that the way to improve muscle strength is to build muscle mass, using such therapies as testosterone, growth hormone, and insulin-like growth factor-1,” says Dr. Marks. “But an increase in muscle mass is not necessarily accompanied by an increase in muscle function. Our results suggest that you can improve muscle function by fixing leaky calcium channels. And in fact, treating aged mice with S107 enhanced muscle strength without increasing muscle size, at least during the four-week treatment period.”

Dr. Marks’ paper is titled, “Ryanodine Receptor Oxidation Causes Intracellular Calcium Leak and Muscle Weakness in Aging.” In addition to Dr. Andersson, his coauthors include Mathew J. Betzenhauser, Steven Reiken, Albano C. Meli, Alisa Umanskaya, Wenjun Xie, Takayuki Shiomi, and Ran Zalk at CUMC, and Alain Lacampagne at Universités Montpellier, Montpellier, France.

A.R. Marks is a consultant for a start-up company, ARMGO Pharma, Inc., which is targeting ryanodine receptors to improve exercise capacity in muscle diseases.

This research was supported by grants from the National Heart, Lung, and Blood Institute and the Swedish Research Council.
###

Columbia University Medical Center provides international leadership in basic, pre-clinical and clinical research, in medical and health sciences education, and in patient care. The medical center trains future leaders and includes the dedicated work of many physicians, scientists, public health professionals, dentists, and nurses at the College of Physicians and Surgeons, the Mailman School of Public Health, the College of Dental Medicine, the School of Nursing, the biomedical departments of the Graduate School of Arts and Sciences, and allied research centers and institutions. Established in 1767, Columbia's College of Physicians and Surgeons was the first institution in the country to grant the M.D. degree and is among the most selective medical schools in the country. Columbia University Medical Center is home to the largest medical research enterprise in New York City and State and one of the largest in the United States.

-####-


April 21, 2011

GSK Receives FDA Approval to Move Forward with Exon Skipping Trial in the US

The US Food and Drug Administration has lifted the partial clinical hold on the Investigational New Drug Application for GSK2402968.  This means they can now proceed with longer term clinical studies of GSK2402968 in DMD patients in the US.  As a result, plans for a randomised placebo controlled study in the US are advancing.  Details regarding the study, including inclusion/exclusion criteria and investigator sites, will be posted to www.clinicaltrials.gov as soon as they are finalized. 

 


 Apr 05, 2011 08:25 ET

SOURCE: Phrixus Pharmaceuticals, Inc.

Phrixus Pharmaceuticals, Inc. Announces $890,000 in NIH Funding for Its Programs in Duchenne Muscular Dystrophy and Heart Failure

Awards to Fund Respiratory Studies for CarmesealTM in Duchenne Muscular Dystrophy and Mechanism of Action Studies in Heart Failure

ANN ARBOR, MI--(Marketwire - April 5, 2011) - Phrixus Pharmaceuticals, Inc., a clinical-stage,specialty pharmaceutical company focused on innovative therapies for Duchenne muscular dystrophy (DMD) and heart failure, today announced that it has received a total of $890,000 in awards from the National Institutes of Health (NIH). Funding is in the form of one SBIR Phase 1 award titled "Effects of P-188 on Respiratory Function and Diaphragm Degeneration in the mdx Mouse" and one STTR Phase 1 award titled "Poloxamer 188 Mechanism of Action in Ischemic Heart Failure." The latter award is in collaboration with Dr. Joseph M. Metzger, Chair of Integrative Biology and Physiology at the University of Minnesota.

"This funding constitutes validation for the potential utility of Carmeseal not just in cardiomyopathy but also in respiratory disease in patients with DMD, for whom it is the leading killer," said Thomas A. Collet, president and CEO. "Completion of the work funded by these grants will significantly advance our knowledge of how Carmeseal achieves its dramatic effects," adds Dr. Bruce Markham, Vice President of Research and Chief Scientific Officer.

DMD is the most devastating of the muscular dystrophies. No drug is approved for its treatment. It is a genetic disease that affects about one out of every 3,500 boys. Approximately 20,000 boys and young men live with this disease in the United States. The hallmarks of DMD are skeletal muscle weakness, respiratory distress, and cardiomyopathy. It is a degenerative disease that leads to premature death.

Heart failure occurs when the heart is unable to pump enough blood around the body. It affects five million Americans and costs the health care system $37 billion annually according to the American Heart Association. Acute Decompensated Heart Failure (ADHF) is the most severe form of heart failure. It causes one million hospitalizations each year.

About Carmeseal™
Carmeseal, generically known as poloxamer 188 (P-188), has been shown to boost the blood pumping capacity of damaged hearts. When Carmeseal, which appears to act as a molecular bandaid, is infused into the bloodstream, it encounters and binds to microscopic tears in the heart muscle. This may prevent the pathological leak of calcium into the heart cells, which could cause calcium overload and keep the heart from delivering sufficient oxygenation to the vital organs. Carmeseal, which has been shown to be effective in four animal models of DMD and heart failure, is expected to have its effect in patients with DMD irrespective of the genetic defect that causes the disease.

About Phrixus Pharmaceuticals, Inc.
Phrixus Pharmaceuticals is developing Carmeseal for DMD and for acute decompensated heart failure. For more information about Phrixus Pharmaceuticals, please visitwww.phrixuspharmaceuticals.com.

Phrixus Pharmaceuticals, Inc. Forward-Looking Statement Disclaimer
This announcement may contain, in addition to historical information, certain forward-looking statements that involve risks and uncertainties. Such statements reflect management's current views and are based on certain assumptions. Actual results could differ materially from those currently anticipated as a result of a number of factors. The company is developing several products for potential future marketing. There can be no assurance that such development efforts will succeed, that such products will receive required regulatory clearance or that, even if such regulatory clearance were received, such products would ultimately achieve commercial success.

http://www.marketwire.com/press-release/Phrixus-Pharmaceuticals-Inc-Announces-890000-NIH-Funding-Its-Programs-Duchenne-Muscular-1422585.htm

Thomas A. Collet
President and CEO
Phrixus Pharmaceuticals, Inc.
+1 (734) 926-0966 ext. 12


STUDY BLURB

March 22, 2011

The first few young men with DMD have enrolled in the clinical trial REVERSE-DMD at The Kennedy Krieger Institute and Johns Hopkins School of Medicine.  This trial, being conducted by neurologist Kathryn Wagner and cardiologist Daniel Judge  will examine whether Revatio (Sildenafil) improves cardiac function in DMD.  Sildenafil has been shown to improve cardiac function in multiple different animal models including the mouse model of DMD.   Thirty males, aged 15 and older who have been diagnosed with DMD and cardiac dysfunction are being recruited to participate in this randomized, double-blind, placebo-controlled study for six months.  An open-label period during which all participants will receive Revatio (Sildenafil) will follow for an additional six months.   For more information regarding the trial, visit: http://clinicaltrials.gov/ct2/show/NCT01168908?term=Duchenne.


Phase III Study of Idebenone in Duchenne Muscular Dystrophy (DMD) (DELOS)
This study is currently recruiting participants.
Verified by Santhera Pharmaceuticals, November 2010
First Received: December 8, 2009   Last Updated: November 26, 2010   History of Changes
Sponsor: Santhera Pharmaceuticals
Information provided by: Santhera Pharmaceuticals
ClinicalTrials.gov Identifier: NCT01027884
  Purpose

The aim of this Phase III study is to assess the efficacy of idebenone on pulmonary function, motor function, muscle strength and quality of life in patients with DMD. Furthermore, the safety and tolerability of idebenone will be assessed. The DELOS study follows a group sequential design. In the first enrolment period patients never used glucocorticoids and/or patients that did stop using glucocorticoids at least 12 months prior to enrolment (i.e. "glucocorticoid non-users") are enrolled. The enrolment of patients using glucocorticoids is foreseen in a second phase of the study.


Condition Intervention Phase
Muscular Dystrophy, Duchenne
Ambulatory Care
Drug: Idebenone (CATENA®)
Phase III

Study Type: Interventional
Study Design: Allocation: Randomized
Control: Placebo Control
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Parallel Assignment
Masking: Double Blind (Subject, Caregiver, Investigator, Outcomes Assessor)
Primary Purpose: Treatment
Official Title: A Phase III Double-Blind, Randomised, Placebo-Controlled Study of the Efficacy, Safety and Tolerability of Idebenone in 10-18 Year Old Patients With Duchenne Muscular Dystrophy

Resource links provided by NLM:


Further study details as provided by Santhera Pharmaceuticals:

Primary Outcome Measures:
  • Primary Objective: to assess the efficacy of idebenone, compared to placebo, in improving or delaying the loss of respiratory function in patients with DMD. Primary endpoint: percent predicted peak expiratory flow (PEF) [ Time Frame: Efficacy assessments: Baseline, Week 13, 26 , 39, 52 ] [ Designated as safety issue: No ]

Secondary Outcome Measures:
  • Pulmonary function tests [ Time Frame: 12 months ] [ Designated as safety issue: No ]
  • Muscle strength and motor function [ Time Frame: 12 months ] [ Designated as safety issue: No ]
  • Quality of life [ Time Frame: 12 months ] [ Designated as safety issue: No ]
  • Safety and tolerability [ Time Frame: 12 months ] [ Designated as safety issue: Yes ]

Estimated Enrollment: 240
Study Start Date: July 2009
Estimated Study Completion Date: December 2012
Estimated Primary Completion Date: December 2011 (Final data collection date for primary outcome measure)
Arms Assigned Interventions
Placebo: Placebo Comparator
Intervention: Drug: Idebenone (CATENA®)
Drug: Idebenone (CATENA®)
Idebenone (900 mg idebenone/day) 2 tabl (150 mg each) x 3 times orally with meals
Idebenone: Experimental
Idebenone 900 mg/day
Intervention: Drug: Idebenone (CATENA®)
Drug: Idebenone (CATENA®)
Idebenone (900 mg idebenone/day) 2 tabl (150 mg each) x 3 times orally with meals

Detailed Description:

This study is a Phase III, multicenter, randomized, double-blind, placebo-controlled efficacy and safety study. DMD patients (ambulatory and non-ambulatory) at age 10-18 years are eligible for enrolment at sites in Europe and North America. Study subjects will be randomized in a 1:1 ratio to receive either idebenone (900 mg/day) or placebo 3 times a day with meals for 52 weeks. Enrollment of patients occurs in phases, depending on the systemic use of glucocorticoids; patients not using glucocorticoids are enrolled first. From randomization efficacy and safety assessments are planned during 6 study visits within 52 weeks.

  Eligibility

Ages Eligible for Study:   10 Years to 18 Years
Genders Eligible for Study:   Male
Accepts Healthy Volunteers:   No
Criteria

Inclusion Criteria:

  1. Patients 10 - 18 years of age at Baseline.
  2. Signed and dated informed consent.
  3. Documented diagnosis of DMD or severe dystrophinopathy and clinical features consistent of typical DMD at diagnosis (i.e. documented delayed motor skills and muscle weakness by age 5 years). DMD should be confirmed by mutation analysis in the dystrophin gene or by substantially reduced levels of dystrophin protein (i.e. absent or <5% of normal) on Western blot or immunostain.
  4. Ability to provide reliable and reproducible repeat PEF within 15% of the first assessment (i.e. Baseline vs. Screening).
  5. Patients assessed by the investigator as willing and able to comply with the requirements of the study, possess the required cognitive abilities and are able to swallow study medication.

Exclusion Criteria:

  1. Patients dependent on assisted ventilation at Screening and/or Baseline (defined as non-invasive nocturnal ventilation, daytime non-invasive ventilation or continuous invasive ventilation).
  2. Patients with documented DMD-related hypoventilation for which assisted ventilation is needed according to current standard of care guidelines (e.g. FVC< 30%) or is required in the opinion of the Investigator.
  3. Patients with a percent predicted PEF > 80% at Baseline.
  4. Patients unable to form a mouth seal to allow precise respiratory flow measurements and mouth pressures.
  5. Symptomatic heart failure (high probability of death within one year of Baseline) and/or symptomatic ventricular arrhythmias.
  6. Participation in the previous Phase II or Phase II Extension study (SNT-II-001 or SNT-II-001-E) for idebenone.
  7. Participation in any other therapeutic trial and/or intake of any investigational drug within 90 days prior to Baseline.
  8. Use of carnitine, creatine, glutamine, oxatomide, or any herbal medicines within 30 days prior to Baseline.
  9. Use of coenzyme Q10 or vitamin E (if taken at a dose of 5 times above the daily physiological requirement) within 30 days prior to Baseline.
  10. Any previous use of idebenone.
  11. Any concomitant medication with a depressive or stimulating effect on respiration or the respiratory tract.
  12. Planned or expected spinal fixation surgery during the study period (as judged by the investigator).
  13. Asthma, bronchitis/COPD, bronchiectasis, emphysema, pneumonia or the presence of any other non-DMD respiratory illness that affects PEF.
  14. Use of bronchodilating medication (i.e. inhaled steroids, sympathomimetics, anti-cholinergics).
  15. Moderate or severe hepatic impairment or severe renal impairment.
  16. Prior or ongoing medical condition or laboratory abnormality that in the Investigator's opinion could adversely affect the safety of the subject
  17. Relevant history of or current drug or alcohol abuse or use of any tobacco/marijuana products/smoking
  18. Known individual hypersensitivity to idebenone or to any of the ingredients/excipients of the study medication
  19. For "glucocorticoid non-users" only

    1. Chronic use of systemic glucocorticoid therapy for DMD related conditions within 12 months of Baseline (the "12 month non-use period")
    2. More than 2 rounds of acute systemic glucocorticoid burst therapy (of ≤2 week duration) for non-DMD related conditions within the 12 month non-use period
    3. Use of any round of systemic glucocorticoid burst therapy of longer than 2 weeks duration within the 12 month non-use period
    4. Use of systemic glucocorticoid burst therapy less than 8 weeks prior to baseline
  20. For "glucocorticoid users" only

    1. Prior to Interim Analysis 1: All "glucocorticoid users"
    2. After the Interim Analysis 1: Initiation, cessation or any relevant change (i.e. dose change of >15% above any dose adaptation for body weight increase/decrease) in systemic glucocorticoid therapy within 6 months prior to Baseline
  Contacts and Locations
Please refer to this study by its ClinicalTrials.gov identifier: NCT01027884

Contacts
Contact: Thomas Meier, Dr, PhD +41(0)61 906 89 64 thomas.meier@santhera.com
Contact: Laurence Lemoal +41(0)61 906 89 33 laurence.lemoal@santhera.com

Locations
United States, Pennsylvania
Prof. Richard S. Finkel, M.D., PhD Recruiting
Philadelphia, Pennsylvania, United States, 19104-1771
Contact: Prof. Richard S. Finkel, MD, PhD         finkel@email.chop.edu    
Contact: Nancy Videon         VIDEON@email.chop.edu    
Principal Investigator: Prof.Richard S. Finkel, MD, PHD            
Austria
Dr. Günther Bernert, Prim. Univ. Doz. Recruiting
Wien, Austria, 1100
Contact: Dr. Günther Bernert, PhD         guenther.bernert@wienkav.at    
Contact: Herrmanns Verena, verena.herrmanns@wienkav.at            
Principal Investigator: Dr. Günther Bernert, MD, PhD            
Belgium
University Hospitals Leuven- Children Hospital Recruiting
Leuven, Belgium, B - 3000
Contact: Gunnar Buyse, MD, PhD         gunnar.buyse@uz.kuleuven.ac.be    
Contact: Kim Rowan, Ms         kim.rowan@uzleuven.be    
Principal Investigator: Gunnar Buyse, MD, PhD            
France
Prof. Thomas Voit , MD, PhD Recruiting
Paris Cedex 13, France, 75651
Contact: Prof. Thomas Voit, MD, PhD         t.voit@institut-myologie.org    
Contact: Dr Valérie Doppler         v.doppler@institut-myologie.org    
Principal Investigator: Prof. Thomas Voit, MD, PhD            
Germany
Universitätsklinikum Essen, Zentrum für Kinderheikunde Recruiting
Essen, Germany, D-45122
Contact: Ulrike Schara, MD, Phd         ulrike.schara@uk-essen.de    
Contact: Bärbel Leiendecker, Ms         baerbel.leiendecker@uk-essen.de    
Principal Investigator: Ulrike Schara, MD, PhD            
Netherlands
Ass. Prof. Jan Verschuuren , MD, PhD Recruiting
Leiden, P.O. Box 9600, Netherlands, 2300 RC
Contact: Ass.Prof. Jan Verschuuren, MD, PhD         J.J.G.M.Verschuuren@lumc.nl    
Contact: Dr Chiara Straathof         C.S.M.Straathof@lumc.nl    
Principal Investigator: Ass. Prof. Jan Verschuuren, MD, PhD            
Sweden
Prof. Thomas Sejersen, MD, PhD Recruiting
Stockholm, Sweden, 17176
Contact: Prof. Thomas Sejersen, MD, PhD         thomas.sejersen@ki.se    
Contact: Marie von Platen         marie.vonplaten@ki.se    
Principal Investigator: Prof. Thomas Sejersen, MD, PhD            
Switzerland
Dr. Pierre-Yves Jeannet, Médecin Associé, MER Recruiting
Lausanne, Switzerland, 1011
Contact: Dr. Pierre-Yves Jeannet Jeannet, Med. Ass., MER         Pierre-yves.Jeannet@chuv.ch    
Contact: Dr Clemens Blötzer         Clemens.Bloetzer@chuv.ch    
Principal Investigator: Dr Pierre-Yves Jeannet, MED.Ass., MER            
Sponsors and Collaborators
Santhera Pharmaceuticals
Investigators
Principal Investigator: Prof. Gunnar Buyse, MD, PhD. University Hospitals Leuven, B-3000, Belgium
Principal Investigator: Dr. Ulrike Schara, MD, PhD Universitätsklinikum Essen, D-45122 Essen, Germany
Principal Investigator: Ass. Prof. Jan Verschuuren, MD, PhD Leiden University Medical Center (LUMC), 2300 RC Leiden, the Netherlands
Principal Investigator: Dr. Pierre-Yves Jeannet, Médecin Associé, MER Unité de Neuropédiatrie, CHUV - BH11, 1011 Lausanne-CH, Switzerland
Principal Investigator: Pr of. Thomas Voit, MD, PhD Université Pierre et Marie curie VI - Institut de Myologie - groupe hospitalier Pitié Salpétričre - 47/83 boulevard de l'hôpital, 75651 Paris Cedex 13, France
Principal Investigator: Prof. Thomas Sejersen, MD, PhD Astrid Lindgrens Barnsjukhus- Karolinska Universitetssjukhuset, SE-17176 Stockholm, Sweden
Principal Investigator: Dr. Günther Bernert, Prim. Univ. Doz. Vorstand der Abteilung für Kinder- und Jugendheilkunde, Gottfried v. Preyer'sches Kinderspital, 1100 Wien, Austria
Principal Investigator: Prof. Richard S. Finkel, M.D., PhD Division of Neurology - The Children's Hospital of Philadelphia - 34th Street and Civic Center Blvd, Philadelphia, PA 19104-1771, USA
  More Information
Additional Information:
No publications provided

Responsible Party: University Hospitals Leuven & University of Leuven School of Medicine ( Gunnar BUYSE, MD, PhD. Professor of Child Neurology & Pediatrics )
ClinicalTrials.gov Identifier: NCT01027884     History of Changes
Other Study ID Numbers: SNT-III-003
Study First Received: December 8, 2009
Last Updated: November 26, 2010
Health Authority: Belgium: Federal Agency for Medicinal Products and Health Products;   Germany: Federal Institute for Drugs and Medical Devices

Keywords provided by Santhera Pharmaceuticals:
Idebenone
Duchenne Muscular Dystrophy (DMD)
Respiratory function
Ambulatory and non-ambulatory patients
Subjects not using glucocorticoids (first enrollment period)
Subjects using glucocorticoids (second enrollment period)

Additional relevant MeSH terms:
Muscular Dystrophy, Duchenne
Muscular Dystrophies
Muscular Disorders, Atrophic
Muscular Diseases
Musculoskeletal Diseases
Neuromuscular Diseases
Nervous System Diseases
Genetic Diseases, X-Linked
Genetic Diseases, Inborn
Idebenone
Antioxidants
Molecular Mechanisms of Pharmacological Action
Pharmacologic Actions
Protective Agents
Physiological Effects of Drugs

ClinicalTrials.gov processed this record on December 19, 2010

Duchenne Muscular Dystrophy Is Ultimately a Stem Cell Disease, Researchers Find

 

ScienceDaily (Dec. 10, 2010) — For years, scientists have tried to understand why children with Duchenne muscular dystrophy experience severe muscle wasting and eventual death. After all, laboratory mice with the same mutation that causes the disease in humans display only a slight weakness. Now research by scientists at the Stanford University School of Medicine, and a new animal model of the disease they developed, points a finger squarely at the inability of human muscle stem cells to keep up with the ongoing damage caused by the disorder.

"Patients with muscular dystrophy experience chronic muscle damage, which initiates a never-ending cycle of repair and wasting," said Helen Blau, PhD, the Donald E. and Delia B. Baxter Professor and a member of Stanford's Institute for Stem Cell Biology and Regenerative Medicine. "We found that in mice the muscle stem cells can keep up with the demands on them to cycle."

The difference is caused, the researchers found, by the fact that mice have significantly longer protective caps on the ends of their chromosomes. The caps, called telomeres, allow the cells to continue to divide and replenish the damaged muscle long after the human cells have reached their capacity for division.

The research marks the first time that muscular dystrophy has been shown definitively to be a stem-cell-based disorder, according to the scientists, who also generated the first-ever mouse model of Duchenne muscular dystrophy that closely mimics the human disease. Similar to human patients, the animals exhibit severe muscle weakness and shortened life span. The mouse model will allow clinicians and researchers to better study the disease and test new therapies.

"The results suggest that treatments directed solely at the muscle fiber will not suffice and could even exacerbate the disease. The muscle stem cells must be taken into consideration," said Blau. Former postdoctoral fellow Jason Pomerantz, MD, co-corresponding author and now an assistant professor at the University of California-San Francisco, said, "if a treatment does not replenish the stem cell compartment, it will likely fail; it would be like pushing the gas pedal to the floor when there is no reserve."

Blau is the senior author of the research, which will be published online Dec. 9 in Cell. Postdoctoral scholars Alessandra Sacco, PhD, and Foteini Mourkioti, PhD, are co-first authors of the work. Sacco is now an assistant professor at the Sanford-Burnham Medical Research Institute.

Duchenne muscular dystrophy is the most prevalent form of the muscular dystrophies. It is caused by a mutation in the dystrophin gene, which connects the interior cytoskeleton of the muscle fiber to the extracellular matrix. Its absence leads to death of the muscle tissue and progressive weakness, which eventually affects a patient's ability to breathe; 10-year-olds are often wheelchair-bound. Death usually occurs by the second or third decade as a result of respiratory and heart problems. The disorder affects about one of every 3,500 boys in the United States, whereas girls are generally spared because the gene lies on the X-chromosome.

Unfortunately, for decades the trusty laboratory mouse failed scientists trying to study the disease in animals. Mice with the same mutation showed only minimal muscle weakness. This left researchers without an easy way to test drugs and therapies. It also gave them a puzzle: Why were the mice so resistant to the muscle damaged caused by the dystrophin mutation?

Blau, Pomerantz, Sacco and Mourkioti, thought the answer might lie in the muscle stem cells. Like other types of stem cells, the muscle stem cells can divide to both replenish themselves and to make new muscle cell precursors. These precursor cells can replace damaged or dead muscle cells that make up the muscle fiber. But even muscle stem cells have their limits, and in this case, the mouse cells outperform their human counterparts.

The reason, the Stanford researchers found, is in the length of the telomeres on the DNA of the two species. The average length of telomeres in laboratory mice is greater than 40 kilobases; in humans it's about 5 to 15 kilobases. Telomeres serve as protective caps on the ends of chromosomes, buffering them from the gradual shortening that occurs during each round of replication. When the telomeres become too short, the cells are no longer able to divide.

To test their theory, the researchers blocked the expression of a component of the telomerase enzyme, which maintains telomeric DNA. Mice with both the dystrophin mutation and the faulty telomerase expression experienced progressive, debilitating muscle degeneration with age -- as exhibited by treadmill stamina tests and muscle damage assays -- and had shorter-than-normal life spans. Muscle stem cells from the mice also had a reduced ability to proliferate, both in the animals and in culture, and were less able to engraft and begin growing when transplanted into wild-type animals.

"What we're seeing is that muscular dystrophy is a multi-factorial disease," said Blau. "The lack of dystrophin causes muscle damage. These damaged muscles are replaced by dividing muscle stem cells, but the repeated rounds of division cause the telomeres to shorten until the stem cells can't fix the damage anymore. This is what happens in humans, and in our new mouse model."

The idea that the symptoms of muscular dystrophy reflect an inability of stem cells to repair ongoing damage has some interesting implications. It implies that any successful treatment should begin early, before the stem cell pool is depleted. It also indicates that researchers and clinicians should investigate stem-cell-based therapies as well as those aimed at protecting the muscle fibers themselves. Finally, it suggests that a highly targeted approach to increase telomerase activity in the muscle stem cells could be useful.

"Finding out that this is a stem cell defect is really exciting," said Blau. "In the early 1980s we reported that muscle cells from DMD patients had less capacity to divide but we did not have the tools to figure out why, since muscle stem cells, the dystrophin gene and telomere function had yet to be identified. Finally, now we can get a handle on what is going on, and learn how best to target future therapies. Having a mouse model that mimics the human disease will benefit all in the field and is very exciting for patients."

Other Stanford researchers involved in the work include Rose Tran, now a graduate student; Peggy Kraft, research assistant and Blau lab manager; postdoctoral scholars Jinkuk Choi, PhD, and Marina Shkreli, PhD; research fellow Michael Llewellyn, PhD; Steve Artandi, MD, PhD, associate professor of medicine; and Scott Delp PhD, the James H. Clark Professor of Bioengineering, Mechanical Engineering and Orthopaedic Surgery.

The research was funded by the American Heart Association, the National Institutes of Health, the Muscular Dystrophy Association and the Baxter Foundation.

Disclaimer: This article is not intended to provide medical advice, diagnosis or treatment. Views expressed here do not necessarily reflect those of ScienceDaily or its staff.

 

New compound brings hope of muscular dystrophy remedy

 

The Mainichi Daily News (Dec. 11, 2010) — Researchers have developed a chemical compound that can curb the progress of muscular dystrophy in dogs, paving the way for a remedy for the genetic disease in humans.

A joint research team at the Osaka Bioscience Institute and the National Center of Neurology and Psychiatry (NCNP) has developed a chemical compound that can restrain the progression of Duchenne muscular dystrophy while at the same time promoting muscle repair.

In their experiment, researchers continued to administer the compound to dogs and found that the subjects were able to keep up their physical strength. The finding was announced at a meeting of the Japanese Biochemical Society in Kobe on Dec. 10.

Duchenne muscular dystrophy is a genetic disease only seen in males, afflicting one in 3,500 boys. Because the disease progresses quickly, many patients are forced to start using wheelchairs at age 5 or 6. No basic remedy for the disease has yet been discovered.

Yoshihiro Urade, a researcher with the Osaka Bioscience Institute, and his fellow researchers synthesized a chemical compound that can curb the activities of the enzyme that generates Prostaglandin D2 (PGD2), an inflammatory substance that is seen in abundance around damaged muscles.

When researchers administered a mouse suffering from Duchenne muscular dystrophy with the compound for five days, the volume of dead muscle tissue in the animal halved. The researchers then turned to two male canine siblings suffering from the same disease, giving one of them the new treatment and using the other as the control. The one administered with the compound was able to run as fast as healthy dogs after a year-long course of the medication.

"Patients may become able to move on their own for lengthy periods of time by taking the medication while effectively using their muscles. We'd like to start clinical tests on humans in a couple of years," Urade said.

 

A Clinical Study to Assess the Efficacy and Safety of GSK2402968 in Subjects With Duchenne Muscular Dystrophy (DMD114044)

 

ClinicalTrials.gov processed this record on December 12, 2010

This study is not yet open for participant recruitment.
Verified by GlaxoSmithKline, December 2010
First Received: October 21, 2010   Last Updated: December 2, 2010   History of Changes
Sponsor: GlaxoSmithKline
Information provided by: GlaxoSmithKline
ClinicalTrials.gov Identifier: NCT01254019
  Purpose

The purpose of this study is to determine whether GSK2402968 is effective in the treatment of ambulant boys with Duchenne muscular dystrophy resulting from a mutation thought to be corrected by exon 51 skipping.


Condition Intervention Phase
Muscular Dystrophies
Duchenne Muscular Dystrophy
Drug: 6mg/kg/week
Phase III

Study Type: Interventional
Study Design: Allocation: Randomized
Endpoint Classification: Safety/Efficacy Study
Intervention Model: Single Group Assignment
Masking: Double Blind (Subject, Caregiver, Investigator, Outcomes Assessor)
Primary Purpose: Treatment
Official Title: A Phase III, Randomized, Double Blind, Placebo-controlled Clinical Study to Assess the Efficacy and Safety of GSK2402968 in Subjects With Duchenne Muscular Dystrophy

Resource links provided by NLM:


Further study details as provided by GlaxoSmithKline:

Primary Outcome Measures:
  • To assess the efficacy of subcutaneous 6 mg/kg GSK2402968 versus placebo; specifically to have 90% power to detect a difference in 6MWD between GSK2402968 and placebo of 30 meters, assuming a common standard deviation of 55meters [ Time Frame: one year ] [ Designated as safety issue: No ]

Secondary Outcome Measures:
  • To evaluate the safety and tolerability of subcutaneous 6 mg/kg GSK2402968 with respect to AEs, ECG results, vital signs and laboratory tests [ Time Frame: one year ] [ Designated as safety issue: Yes ]
  • Plasma concerntration of subcutaneous 6 mg/kg GSK2402968 calculated at each time point for each subject (sample size (n), mean, standard deviation (SD), percentage of coefficient of variation (%CV), geometric mean, median, minimum, and maximum [ Time Frame: one year ] [ Designated as safety issue: No ]
  • To evaluate the diffrence on quality of life of between GSK2402968 and placebo using PedsQOL, CGI-I and HUI [ Time Frame: one year ] [ Designated as safety issue: No ]

Estimated Enrollment: 180
Study Start Date: November 2010
Estimated Study Completion Date: March 2011
Estimated Primary Completion Date: March 2011 (Final data collection date for primary outcome measure)
Arms Assigned Interventions
GSK2402968: Experimental
6mg/kg
Intervention: Drug: 6mg/kg/week
Drug: 6mg/kg/week
Placebo: Experimental
dose-matched
Intervention: Drug: 6mg/kg/week
Drug: 6mg/kg/week

  Eligibility

Ages Eligible for Study:   5 Years and older
Genders Eligible for Study:   Male
Accepts Healthy Volunteers:   No
Criteria

Inclusion Criteria:

  • Ambulant subjects with Duchenne muscular dystrophy resulting from a mutation/deletion within the DMD gene, confirmed by a state-of-the-art DNA diagnostic technique covering all DMD gene exons, including but not limited to MLPA (Multiplex Ligation-dependent Probe Amplification), CGH (Comparative Genomic Hybridisation), SCAIP (Single Condition Amplification/Internal Primer) or H-RMCA (High-Resolution Melting Curve Analysis), and correctable by GSK2402968-induced DMD exon 51 skipping.
  • Males, aged at least 5 years, and with life expectancy of at least 1 year
  • Able to complete 6MWD test with minimal distance of at least 75m at each predrug visit. In addition, results of 6MWD must be within 20% of each other at each pre-drug visit
  • Receiving glucocorticoids for a minimum of 6 months immediately prior to screening, with no significant change in total daily dosage or dosing regimen for a minimum of 3 months immediately prior to screening and a reasonable expectation that total daily dosage and dosing regimen will not change significantly for the duration of the study
  • QTc <450msec (based on single or average QTc value of triplicate ECGs obtained over a brief recording period), or <480 msec for subjects with Bundle Branch Block. Note: QTc may be either QTcB or QTcF, and machine read or manual overread.
  • Subjects, where appropriate, must be willing to use adequate contraception (condoms or abstinence) for the duration of the study and for at least 5 months after the last dose of study drug.
  • Willing and able to comply with all protocol requirements and procedures,
  • Able to give informed assent and/or consent in writing signed by the subject and/or parent(s)/legal guardian (according to local regulations).
  • French subjects: In France, a subject will be eligible for inclusion in this study only if either affiliated to or a beneficiary of a social security category.

Exclusion Criteria:

  • Any additional missing exon for DMD that cannot be treated with GSK2402968
  • Current or history of liver or renal disease or impairment
  • Acute illness within 4 weeks of the first anticipated administration of study medication which may interfere with study assessments
  • Use of anticoagulants, antithrombotics or antiplatelet agents, previous treatment with investigational drugs, within 6 months of the first administration of study medication; and idebenone or other forms of Coenzyme Q10 within 1 month of the first administration of study medication.
  • Current or anticipated participation in any investigational clinical studies
  • Positive hepatitis B surface antigen, hepatitis C antibody test (if verified via RIBA or PCA testing), or human immunodeficiency virus (HIV) test at screening,
  • Symptomatic cardiomyopathy. If subject has a left ventricular ejection fraction <45% at Screening, the investigator should discuss inclusion of subject in the study with the medical monitor,
  • Children in Care. The definition of a Child in Care is a child who has been placed under the control or protection of an agency, organisation, institution or entity by the courts, the government or a government body, acting in accordance with powers conferred on them by law or regulation. The definition of a child in care can include a child cared for by foster parents or living in a care home or institution, provided that the arrangement falls within the definition above. The definition of a child in care does not include a child who is adopted or has an appointed legal guardian.
  Contacts and Locations
Please refer to this study by its ClinicalTrials.gov identifier: NCT01254019

Contacts
Contact: US GSK Clinical Trials Call Center 877-379-3718 info@clinicaltrialsforgsk.com

Locations
France
GSK Investigational Site
Nantes cedex 01, France, 44093
GSK Investigational Site
Toulouse cedex 9, France, 31059
Germany
GSK Investigational Site
Freiburg, Baden-Wuerttemberg, Germany, 79106
GSK Investigational Site
Muenchen, Bayern, Germany, 80337
GSK Investigational Site
Goettingen, Niedersachsen, Germany, 37075
Korea, Republic of
GSK Investigational Site
Seoul, Korea, Republic of, 110-744
Sponsors and Collaborators
GlaxoSmithKline
Investigators
Study Director: GSK Clinical Trials GlaxoSmithKline
  More Information
No publications provided

Responsible Party: GSK Clinical Disclosure ( E.D. Derilus; Clinical Disclosure Advisor )
ClinicalTrials.gov Identifier: NCT01254019     History of Changes
Other Study ID Numbers: 114044
Study First Received: October 21, 2010
Last Updated: December 2, 2010
Health Authority: France: Afssaps - French Health Products Safety Agency;   Canada: Health Canada;   Germany: Bundesinstitut für Arzneimittel und Medizinprodukte;   Europe: European Medicines Agency

Keywords provided by GlaxoSmithKline:
Duchenne Muscular Dystrophy
DMD
968
GSK
Duchenne

Additional relevant MeSH terms:
Muscular Dystrophy, Duchenne
Muscular Dystrophies
Muscular Disorders, Atrophic
Muscular Diseases
Musculoskeletal Diseases