A systematic literature review of evidence-based clinical practice for rare diseases: what are the perceived and real barriers for improving the evidence and how can they be overcome?

[1]  I. van der Tweel,et al.  Sequential designs with small samples: Evaluation and recommendations for normal responses , 2018, Statistical methods in medical research.

[2]  D. Pieper,et al.  Specific barriers to the conduct of randomised clinical trials on medical devices , 2017, Trials.

[3]  C. la Vecchia,et al.  Evidence-based practice within nutrition: what are the barriers for improving the evidence and how can they be dealt with? , 2017, Trials.

[4]  A. Rath,et al.  Barriers to the conduct of randomised clinical trials within all disease areas , 2017, Trials.

[5]  Tim Friede,et al.  Meta‐analysis of few small studies in orphan diseases , 2016, Research synthesis methods.

[6]  M. Parmar,et al.  How do you design randomised trials for smaller populations? A framework , 2016, BMC Medicine.

[7]  K. Roes A framework: make it useful to guide and improve practice of clinical trial design in smaller populations , 2016, BMC Medicine.

[8]  P. Gøtzsche,et al.  “Adaptive pathways” to drug authorisation: adapting to industry? , 2016, British Medical Journal.

[9]  K. Thorlund,et al.  False-positive findings in Cochrane meta-analyses with and without application of trial sequential analysis: an empirical review , 2016, BMJ Open.

[10]  Christian Gluud,et al.  Evidence-based clinical practice: Overview of threats to the validity of evidence and how to minimise them. , 2016, European journal of internal medicine.

[11]  G. Rasi,et al.  How aligned are the perspectives of EU regulators and HTA bodies? A comparative analysis of regulatory‐HTA parallel scientific advice , 2016, British journal of clinical pharmacology.

[12]  S. Garattini,et al.  Letting post-marketing bridge the evidence gap: the case of orphan drugs , 2016, British Medical Journal.

[13]  Ralf-Dieter Hilgers,et al.  Directions for new developments on statistical design and analysis of small population group trials , 2016, Orphanet Journal of Rare Diseases.

[14]  C. Sudlow,et al.  Statistical approaches for evaluating surrogate outcomes in clinical trials: A systematic review , 2016, Journal of biopharmaceutical statistics.

[15]  J. Little,et al.  Translating rare-disease therapies into improved care for patients and families: what are the right outcomes, designs, and engagement approaches in health-systems research? , 2015, Genetics in Medicine.

[16]  L. Billingham,et al.  Research methods to change clinical practice for patients with rare cancers. , 2016, The Lancet. Oncology.

[17]  D. Bolignano,et al.  Good-quality research in rare diseases: trials and tribulations , 2016, Pediatric Nephrology.

[18]  R. Valdez,et al.  Public Health and Rare Diseases: Oxymoron No More , 2016, Preventing chronic disease.

[19]  Martin Posch,et al.  Evidence, eminence and extrapolation , 2016, Statistics in medicine.

[20]  V. Volpert,et al.  An in silico approach helped to identify the best experimental design, population, and outcome for future randomized clinical trials. , 2016, Journal of Clinical Epidemiology.

[21]  Peter A Merkel,et al.  A small n sequential multiple assignment randomized trial design for use in rare disease research. , 2016, Contemporary clinical trials.

[22]  Saul Perlmutter,et al.  Blind analysis: Hide results to seek the truth , 2015, Nature.

[23]  S. Goodman,et al.  Meta-research: Evaluation and Improvement of Research Methods and Practices , 2015, PLoS biology.

[24]  C. Tudur Smith,et al.  A comparison of interventional clinical trials in rare versus non-rare diseases: an analysis of ClinicalTrials.gov , 2014, Orphanet Journal of Rare Diseases.

[25]  Joshua J Gagne,et al.  Innovative research methods for studying treatments for rare diseases: methodological review , 2014, BMJ : British Medical Journal.

[26]  John P. A. Ioannidis,et al.  How to Make More Published Research True , 2014, PLoS medicine.

[27]  Elena Kulinskaya,et al.  Trial sequential methods for meta‐analysis , 2014, Research synthesis methods.

[28]  S. Simoens,et al.  Incremental cost per quality-adjusted life year gained? The need for alternative methods to evaluate medical interventions for ultra-rare disorders. , 2014, Journal of comparative effectiveness research.

[29]  Mohit Bhandari,et al.  Blinded interpretation of study results can feasibly and effectively diminish interpretation bias. , 2014, Journal of clinical epidemiology.

[30]  Harold Varmus,et al.  Rescuing US biomedical research from its systemic flaws , 2014, Proceedings of the National Academy of Sciences.

[31]  P. Williamson,et al.  Methodology of clinical trials for rare diseases. , 2014, Best practice & research. Clinical rheumatology.

[32]  J. Jakobsen,et al.  The thresholds for statistical and clinical significance – a five-step procedure for evaluation of intervention effects in randomised clinical trials , 2014, BMC Medical Research Methodology.

[33]  O. Gross,et al.  Challenges for academic investigator-initiated pediatric trials for rare diseases. , 2014, Clinical Therapeutics.

[34]  A. Hoes,et al.  Drug development for exceptionally rare metabolic diseases: challenging but not impossible , 2013, Orphanet Journal of Rare Diseases.

[35]  K. Bushby,et al.  The TREAT-NMD care and trial site registry: an online registry to facilitate clinical research for neuromuscular diseases , 2013, Orphanet Journal of Rare Diseases.

[36]  K. Boycott,et al.  Rare-disease genetics in the era of next-generation sequencing: discovery to translation , 2013, Nature Reviews Genetics.

[37]  John P A Ioannidis,et al.  Optimal type I and type II error pairs when the available sample size is fixed. , 2013, Journal of clinical epidemiology.

[38]  Anne W. Riley,et al.  Pediatric patient-reported outcome instruments for research to support medical product labeling: report of the ISPOR PRO good research practices for the assessment of children and adolescents task force. , 2013, Value in health : the journal of the International Society for Pharmacoeconomics and Outcomes Research.

[39]  D. Lacombe,et al.  International clinical trials setting for rare cancers: organisational and regulatory constraints—the EORTC perspective , 2013, Ecancermedicalscience.

[40]  Brian A. Nosek,et al.  Power failure: why small sample size undermines the reliability of neuroscience , 2013, Nature Reviews Neuroscience.

[41]  Boris Freidlin,et al.  Statistical Challenges in the Evaluation of Treatments for Small Patient Populations , 2013, Science Translational Medicine.

[42]  Sylvie Chabaud,et al.  Experimental designs for small randomised clinical trials: an algorithm for choice , 2013, Orphanet Journal of Rare Diseases.

[43]  Roberta Joppi,et al.  Orphan drugs, orphan diseases. The first decade of orphan drug legislation in the EU , 2012, European Journal of Clinical Pharmacology.

[44]  Domenica Taruscio,et al.  The need for worldwide policy and action plans for rare diseases , 2012, Acta paediatrica.

[45]  M Mauer,et al.  Adaptive designs at European Organisation for Research and Treatment of Cancer (EORTC) with a focus on adaptive sample size re-estimation based on interim-effect size. , 2012, European journal of cancer.

[46]  F. Dhombres,et al.  Representation of rare diseases in health information systems: The orphanet approach to serve a wide range of end users , 2012, Human mutation.

[47]  H. Eichler,et al.  Adaptive Licensing: Taking the Next Step in the Evolution of Drug Approval , 2012, Clinical pharmacology and therapeutics.

[48]  Jianjun Gao,et al.  Rare diseases, orphan drugs, and their regulation in Asia: Current status and future perspectives. , 2012, Intractable & rare diseases research.

[49]  J. Demotes-Mainard,et al.  A European perspective--the European clinical research infrastructures network. , 2011, Annals of oncology : official journal of the European Society for Medical Oncology.

[50]  George A Tomlinson,et al.  A framework for applying unfamiliar trial designs in studies of rare diseases. , 2011, Journal of clinical epidemiology.

[51]  The Committee for Orphan Medicinal Products and the Eu Secretariat European regulation on orphan medicinal products: 10 years of experience and future perspectives , 2011, Nature Reviews Drug Discovery.

[52]  G. Molenberghs,et al.  A unified framework for the evaluation of surrogate endpoints in mental-health clinical trials , 2010, Statistical methods in medical research.

[53]  F. Rivas-Ruíz,et al.  Efficacy of prescribed injectable diacetylmorphine in the Andalusian trial: Bayesian analysis of responders and non-responders according to a multi domain outcome index , 2009, Trials.

[54]  S. Garattini,et al.  Orphan drug development is not taking off. , 2009, British journal of clinical pharmacology.

[55]  P. Scardino,et al.  The clinically-integrated randomized trial: proposed novel method for conducting large trials at low cost , 2009, Trials.

[56]  K. Thorlund,et al.  Trial sequential analysis may establish when firm evidence is reached in cumulative meta-analysis. , 2008, Journal of clinical epidemiology.

[57]  R. Koretz,et al.  Hepatology may have problems with putative surrogate outcome measures. , 2007, Journal of hepatology.

[58]  G Molenberghs,et al.  Evaluation of surrogate endpoints in randomized experiments with mixed discrete and continuous outcomes , 2001, Statistics in medicine.

[59]  P. Gøtzsche,et al.  Blinding during data analysis and writing of manuscripts. , 1996, Controlled clinical trials.