Skip to main content
Log in

Common data elements for clinical research in mitochondrial disease: a National Institute for Neurological Disorders and Stroke project

  • Original Article
  • Published:
Journal of Inherited Metabolic Disease

An Erratum to this article was published on 04 October 2017

This article has been updated

Abstract

Objectives

The common data elements (CDE) project was developed by the National Institute of Neurological Disorders and Stroke (NINDS) to provide clinical researchers with tools to improve data quality and allow for harmonization of data collected in different research studies. CDEs have been created for several neurological diseases; the aim of this project was to develop CDEs specifically curated for mitochondrial disease (Mito) to enhance clinical research.

Methods

Nine working groups (WGs), composed of international mitochondrial disease experts, provided recommendations for Mito clinical research. They initially reviewed existing NINDS CDEs and instruments, and developed new data elements or instruments when needed. Recommendations were organized, internally reviewed by the Mito WGs, and posted online for external public comment for a period of eight weeks. The final version was again reviewed by all WGs and the NINDS CDE team prior to posting for public use.

Results

The NINDS Mito CDEs and supporting documents are publicly available on the NINDS CDE website (https://commondataelements.ninds.nih.gov/), organized into domain categories such as Participant/Subject Characteristics, Assessments, and Examinations.

Conclusion

We developed a comprehensive set of CDE recommendations, data definitions, case report forms (CRFs), and guidelines for use in Mito clinical research. The widespread use of CDEs is intended to enhance Mito clinical research endeavors, including natural history studies, clinical trial design, and data sharing. Ongoing international collaboration will facilitate regular review, updates and online publication of Mito CDEs, and support improved consistency of data collection and reporting.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Change history

  • 04 October 2017

    An erratum to this article has been published.

References

  • Camp KM, Krotoski D, Parisi MA, Gwinn KA, Cohen BH, Cox CS, Enns GM, Falk MJ, Goldstein AC, Gopal-Srivastava R, Gorman GS, Hersh SP, Hirano M, Hoffman FA, Karaa A, MacLeod EL, McFarland R, Mohan C, Mulberg AE, Odenkirchen JC, Parikh S, Rutherford PJ, Suggs-Anderson SK, Tang WH, Vockley J, Wolfe LA, Yannicelli S, Yeske PE, Coates PM (2016) Nutritional interventions in primary mitochondrial disorders: developing an evidence base. Mol Genet Metab 119(3):187–206

    Article  CAS  PubMed  Google Scholar 

  • Chinnery PF (2014) Mitochondrial disorders overview. In: Pagon PA, Adam MP, Ardinger HH et al (eds) Gene reviews. University of Washington, Seattle

    Google Scholar 

  • DiMauro S, Schon E (2003) Mitochondrial respiratory-chain diseases. N Engl J Med 348:2656–2668

    Article  CAS  PubMed  Google Scholar 

  • Gorman GS, Schaefer AM, Ng Y et al (2015) Prevalence of nuclear and mitochondrial DNA mutations related to adult mitochondrial disease. Ann Neurol 77:753–759

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Gorman GS, Chinnery PF, DiMauro S, Hirano M, Koga Y, McFarland R, Suomalainen A, Thorburn DR, Zeviani M, Turnbull DM (2016) Mitochondrial diseases. Nat Rev Dis Primers. doi:10.1038/nrdp.2016.80

  • Grinnon ST, Miller K, Marler JR, Lu Y, Stout A, Odenkirchen J, Kunitz S (2012) National Institute of Neurological Disorders and Stroke Common Data Element Project - approach and methods. Clin Trials 9:322–329

    Article  PubMed  PubMed Central  Google Scholar 

  • Harrison SM, Riggs ER, Maglott DR, Lee JM, Azzariti DR, Niehaus A, Ramos EM, Martin CL, Landrum MJ, Rehm HL (2016) Using ClinVar as a resource to support variant interpretation. Curr Protoc Hum Genet 89:8.16.1–8.16.23

    Article  Google Scholar 

  • Lott MT, Leipzig JN, Derbeneva O, Xie HM, Chalkia D, Sarmady M, Procaccio V, Wallace DC (2013) mtDNA variation and analysis using mitomap and mitomaster. Curr Protoc Bioinformatics 44:1.23.1–26

    Article  Google Scholar 

  • MacArthur DG, Manolio TA, Dimmock DP (2014) Guidelines for investigating causality of sequence variants in human disease. Nature 24 508(7497):469–476

    Article  CAS  Google Scholar 

  • Nightingale H, Pfeffer G, Bargiela D, Horvath R, Chinnery PF (2016) Emerging therapies for mitochondrial disorders. Brain 139:1633–1648

    Article  PubMed  PubMed Central  Google Scholar 

  • Niyazov D, Kahler S, Frye R (2016) Primary mitochondrial disease and secondary mitochondrial dysfunction: importance of distinction for diagnosis and treatment. Mol Syndromol 7:122–137

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  • Parikh S, Goldstein A, Koenig MK et al (2015) Diagnosis and management of mitochondrial disease: a consensus statement from the Mitochondrial Medicine Society. Genet Med 17:689–701

    Article  CAS  PubMed  Google Scholar 

  • Rahman S (2015) Emerging aspects of treatment in mitochondrial disorders. J Inherit Metab Dis 38:641–653

    Article  CAS  PubMed  Google Scholar 

  • Richards S, Aziz N, Bale S et al (2015) Standards and guidelines for the interpretation of sequence variants: a joint consensus recommendation of the American College of Medical Genetics and Genomics and the Association for Molecular Pathology. Genet Med 17:405–424

    Article  PubMed  PubMed Central  Google Scholar 

  • Suomalainen A (2013) Fibroblast growth factor 21: a novel biomarker for human muscle-manifesting mitochondrial disorders. Expert Opin Med Diagn 7:313–317

    Article  CAS  PubMed  Google Scholar 

  • Yatsuga S, Fujita Y, Ishii A et al (2015) Growth differentiation factor 15 as a useful biomarker for mitochondrial disorders. Ann Neurol 7:814–823

    Article  Google Scholar 

Download references

Acknowledgements

The views expressed here are those of the authors and do not represent those of the NIH, NINDS or the US Government. Logistic support for this project was provided in part through NIH Contract HHSN271201200034C. The development of the NINDS Mito CDEs was made possible thanks to the great investment of time and effort of WG members and the members of the NINDS CDE Project team participating from 2013 to 2015.

Author information

Authors and Affiliations

Authors

Consortia

Corresponding author

Correspondence to Amy Goldstein.

Ethics declarations

Conflict of interest

John Shoffner has received research funds from the Department of Defense and owns stock in Medical Neurogenetics, LLC.

David Dimmock is a consultant to Audentes Therapeutics, Biomarin, Ilumina and Complete Genomics and involved in clinical trials with Genzyme/Sanofi, Biomarin, Shire, Cytonet.

Marni Falk has been a consultant to Mitokyne; received research funds from Cardero Therapeutics, Mitobridge, Raptor Pharmaceuticals, RiboNova, Stealth BioTherapeutics, and Vitaflo International; was a former scientific advisory board member for Perlstein Laboratory, Inc., and is involved in clinical trials with REATA pharmaceuticals.

Amel Karaa, Shamima Rahman, Anne Lombès, Patrick Yu-Wai-Man, Muniza K. Sheikh, Sherita Alai-Hansen, Bruce H. Cohen, Lisa Emrick, Shana McCormack, David Mirsky, Tony Moore, Sumit Parikh, Tanja Taivassalo, Mark Tarnopolsky, Ingrid Tein, Joanne C. Odenkirchen, and Amy Goldstein have no conflict of interest.

Studies with human or animal

This article does not contain any studies with human or animal subjects performed by the any of the authors.

Additional information

Communicated by: Eva Morava

MITO Working Group Members

Biomarkers WG

John Shoffner, MD (Chair), Atlanta, GA, USA

Mark Bamberger, PhD, Stealth Peptides, Inc, Newton Centre, MA, USA

Yasutoshi Koga, MD, PhD, Kurume University, Fukuoka-ken, Japan

Anne Lombès, MD, PhD, INSERM, Paris, France

Fernando Scaglia, MD, Baylor College of Medicine, Houston, TX, USA

Cognitive/Behavioral/Psychological Outcomes WG

Lisa Emrick, MD (Chair), Baylor College of Medicine/Texans Children’s Hospital, Houston, TX, USA

Rebecca Vaurio, PhD, Kennedy Krieger Institute, Baltimore, MD, USA

Endocrinology/Diabetes/GI/Nutrition WG

Shana McCormack, MD (Chair), The Children’s Hospital of Philadelphia, Philadelphia, PA, USA

J.E. Abdenur, MD, CHOC Children’s Hospital, Orange, CA, USA

Kristin Fiorino, MD, Children’s Hospital of Philadelphia, Philadelphia, PA, USA (ad hoc member)

Bret Goodpaster, PhD, Sanford, Burnham Medical Research Institute, Orlando, FL, USA

Robert O. Heuckeroth, MD PhD, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA

Kierstin Keller, CGC, Children’s Hospital of Philadelphia, Philadelphia, PA, USA (ad hoc member)

Robert McFarland, DoH, HEFCE, Newcastle University, UK

Philip Morgan, MD, Mitochondrial Research Guild, Seattle, WA, USA

Fernando Scaglia, MD, Baylor College of Medicine, Houston, TX, USA

Charles A. Stanley, MD, Children’s Hospital of Philadelphia, Philadelphia, PA, USA

Steven M. Willi, MD, The Children’s Hospital of Philadelphia, Philadelphia, PA, USA

Exercise Physiology WG

Tanja Taivassalo, PhD (Chair), McGill University, Montreal, QC, Canada

Amy Goldstein, MD, Children’s Hospital of Pittsburgh, Pittsburgh, PA, USA

Ronald Haller, MD, Southwestern Medical Center, Dallas, TX, USA

Mark Tarnopolsky, MD, PhD, McMaster University, Hamilton, Ontario, Canada

Karim Wahbi, MD, PhD, Institute of Myology, Paris, France

Genetics WG

David Dimmock, MD (Co-Chair), Medical College of Wisconsin, Milwaukee, Wisconsin, USA

Marn J. Falk, MD (Co-Chair), The Children’s Hospital of Philadelphia, Philadelphia, PA, USA

Xiaowu Gai, PhD, Ocular Genomics Institute

Michio Hirano, MD, Columbia University Medical Center

Anne Lombès, MD, PhD, INSERM, Paris, France

Shamima Rahman, FRCP, FRCPCH, PhD, UCL Great Ormond Street Institute of Child Health, London, UK

Fernando Scaglia, MD, Baylor College of Medicine, Houston, TX, USA

Curt Scharfe, MD, PhD, Stanford University School of Medicine, Stanford, CA, USA

David Thorburn, PhD, FHGSA, FFSC (RCPA), Murdoch Children’s Research Institute, Victoria, Australia

Stephan Züchner, MD, PhD, Dr. John T. Macdonald Foundation, Miami, FL, USA

Imaging WG

David M. Mirsky, MD (Chair), Children’s Hospital Colorado, Aurora, CO, USA

Abigail Collins, MD, Children’s Hospital Colorado, Aurora, CO, USA

Andrea L. Gropman, MD, Children’s National Medical Center, Washington, DC, USA

Edward Yang, MD, Boston Children’s Hospital, Boston, MA, USA

Neurological Assessments WG

Bruce Cohen, MD, FAAN (Co-Chair), Akron Children’s Hospital, Akron, OH, USA

Ingrid Tein, MD, FRCP (Co-Chair), University of Toronto, ON, Canada

Laurence Bindoff, MD, PhD, University of Bergen and Haukeland

Michio Hirano, MD, Columbia University Medical Center

Matthew Klein, MD, MS, FACS, Edison Pharmaceuticals

Thomas Klopstock, MD, Friedrich-Baur-Institute, Munich, Germany

Saskia Koene, MD, Radboud University Nijmegen Medical Centre, Netherlands

Pascal Laforêt, MD, PhD, Groupe Hospitalier Pitié-Salpêtrière Assistance Publique-Hôpitaux de Paris, Paris, France

Margherita Milone, MD, PhD, Mayo School of Graduate Medical Education, Rochester, MN, USA

Jan Smeitink, MD, PhD, Radboud University Nijmegen Medical Centre, Netherlands

Patient-Reported Outcomes QoL WG

Mary Kay Koenig, MD (Co-Chair), University of Texas Health Science Center, Houston, TX, USA

Sumit Parikh, MD (Co-Chair), Cleveland Clinic, Cleveland, OH, USA

Cristy Balcells, RN MSN, Mito Action, Boston, MA, USA

Michio Hirano, MD, Columbia University Medical Center

Amel Karaa, MD, Massachusetts General Hospital, Boston, MA, USA

Robert McFarland, DoH, HEFCE, Newcastle University, UK

Russel Saneto, DO, Seattle Children’s Hospital, Seattle, WA, USA

Peter W. Stacpoole, MD, PhD, University of Florida, Gainesville, FL, USA

Philip Yeske, PhD, United Mitochondrial Disease Foundation

Vision WG

Patrick Yu-Wai-Man, MD, PhD, FRCPath, FRCOphth (Chair), Newcastle University and Moorfields Eye Hospital, UK

Piero Barboni, MD, University of Bologna, Bologna, Italy

Valerio Carelli, MD, PhD, University of Bologna School of Medicine, Bologna, Italy

Patrick Chinnery, MD, PhD, FRCPath, FRCP, University of Cambridge, UK

Graham E. Holder, PhD, Moorfields Eye Hospital, London, UK

John L. Keltner, MD, UC Davis School of Medicine, Sacramento, CA, USA

Tony Moore, MA, FRCS, FCROphth, FMedSci, Ophthalmology UCSF School of Medicine, San Francisco, CA

Alfredo A. Sadun, MD, PhD, Doheny Eye Institute/UCLA, Los Angeles, CA, USA

Claire Sheldon, MD, PhD, University of British Columbia, Vancouver, Canada

Marcela Votruba, MD, PhD, FRCOphth, Cardiff University, UK

NINDS CDE Team

Joanne Odenkirchen, MPH, NINDS CDE Project Officer, National Institutes of Health/National Institute of Neurological Disorders and Stroke, (NIH/NINDS), Bethesda, MD, USA

Vernon Anderson, PhD

Kathryn Camp MS, RD, Consultant to the NIH/Office of Dietary Supplements (ODS), Bethesda, MD, USA

William C. Copeland, PhD, National Institute of Environmental Health Sciences (NIEHS), Research Triangle Park, NC, USA

Kerry Goetz, MS, National Eye Institute (NEI)/NIH, Bethesda, MD, USA

Katrina A Gwinn, MD, NINDS-NIH, Bethesda, MD, USA

Petra Kaufmann, MD, MSc, National Center for Advancing Translational Sciences (NCATS)/ NIH, Bethesda, MD, USA

Danuta Krotoski, PhD, Eunice Kennedy Shriver NICHD/NIH, Bethesda, MD, USA

Lynne A. Wolfe, MS, PNP, BC, Undiagnosed Disease Program, NHGRI/NIH, Bethesda, MD, USA

Steven Zullo, PhD, NIH/NIBIB, Bethesda, MD, USA

Sherita Ala’i-Hansen, MS, The Emmes Corporation, Rockville, MD, USA

Muniza K. Sheikh, MS, MBA, The Emmes Corporation, Rockville, MD, USA

An erratum to this article is available at https://doi.org/10.1007/s10545-017-0081-z.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Karaa, A., Rahman, S., Lombès, A. et al. Common data elements for clinical research in mitochondrial disease: a National Institute for Neurological Disorders and Stroke project. J Inherit Metab Dis 40, 403–414 (2017). https://doi.org/10.1007/s10545-017-0035-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10545-017-0035-5

Keywords

Navigation