S. Garattini, J. Jakobsen, J. Wetterslev, V. Bertele, R. Banzi et al., Evidence-based clinical practice: Overview of threats to the validity of evidence and how to minimise them, European Journal of Internal Medicine, vol.32, issue.11, pp.13-21, 2016.
DOI : 10.1016/j.ejim.2016.03.020

P. Song, J. Gao, Y. Inagaki, N. Kokudo, and W. Tang, Rare diseases, orphan drugs, and their regulation in Asia: Current status and future perspectives, Intractable & Rare Diseases Research, vol.1, issue.1, pp.3-9, 2012.
DOI : 10.5582/irdr.2012.v1.1.3

A. Rath, N. Wakap, and S. , Orphanet Report Series -Prevalence of rare diseases: Bibliographic data, Number 1, 2017.

M. Schlander, S. Garattini, S. Holm, P. Kolominsky-rabas, E. Nord et al., Incremental cost per quality-adjusted life year gained? The need for alternative methods to evaluate medical interventions for ultra-rare disorders, Journal of Comparative Effectiveness Research, vol.194, issue.9, pp.399-422, 2014.
DOI : 10.1002/9781118644898

S. Bell, T. Smith, and C. , A comparison of interventional clinical trials in rare versus non-rare diseases: an analysis of ClinicalTrials.gov, Orphanet Journal of Rare Diseases, vol.71, issue.4, p.170, 2014.
DOI : 10.1111/j.1365-2125.2010.03877.x

E. Union, on an action in the field of rare diseases, pp.1510007-1510017, 2009.

S. Aymé and C. Rodwell, European Committee Expert Group on Rare Diseases Report on the State of the Art of Rare Disease Activities in Europe, p.15, 2014.

A. Rath and V. Salamon, Orphanet Report Series -Lists of medicinal products for rare diseases in Europe, 2017.

D. Food and . Administration, Rare Disease and Orphan Drug Designated Approvals, 2016.

R. Joppi, V. Bertele, and S. Garattini, Orphan drugs, orphan diseases. The first decade of orphan drug legislation in the EU, European Journal of Clinical Pharmacology, vol.68, issue.2, pp.1009-1033, 2012.
DOI : 10.1007/s00228-011-1115-3

K. Westermark, B. Holm, M. Soderholm, J. Llinares-garcia, F. Riviere et al., European regulation on orphan medicinal products: 10 years of experience and future perspectives, Nat Rev Drug Discov, vol.10, issue.5, pp.341-350, 2011.

D. Food and . Administration, Novel Drugs Summary Available from: http://www.fda.gov/Drugs, 2015.

A. European-medicines, Latest news, 2016.

C. Davis, J. Lexchin, T. Jefferson, P. Gotzsche, and M. Mckee, ???Adaptive pathways??? to drug authorisation: adapting to industry?, BMJ, vol.354, 2016.
DOI : 10.1136/bmj.i4437

H. Eichler, K. Oye, L. Baird, E. Abadie, J. Brown et al., Adaptive Licensing: Taking the Next Step in the Evolution of Drug Approval, Clinical Pharmacology & Therapeutics, vol.374, issue.3, pp.426-463, 2012.
DOI : 10.1016/S0140-6736(10)60141-9

J. Demotes-mainard and C. Kubiak, A European perspective - the European clinical research infrastructures network, Annals of Oncology, vol.22, issue.Suppl 7, pp.44-53, 2011.
DOI : 10.1093/annonc/mdr425

S. Djurisic, A. Rath, S. Gaber, S. Garattini, V. Bertele et al., Barriers to the conduct of randomised clinical trials within all disease areas, Trials, vol.497, issue.7451, p.360, 2017.
DOI : 10.1038/497557a

URL : https://hal.archives-ouvertes.fr/inserm-01572262

M. Laville, B. Segrestin, Y. Masson, C. Ruano-rodríguez, L. Serra-majem et al., Evidence-based practice within nutrition: what are the barriers for improving the evidence and how can they be dealt with?, Trials, vol.11, issue.10, p.425, 2017.
DOI : 10.1007/s11306-014-0750-y

URL : https://hal.archives-ouvertes.fr/inserm-01588782

E. Neugebauer, A. Rath, S. Antoine, M. Eikermann, D. Seidel et al., Specific barriers to the conduct of randomised clinical trials on medical devices, Trials, vol.526, issue.7572, p.427, 2017.
DOI : 10.1038/526187a

URL : https://hal.archives-ouvertes.fr/inserm-01588783

R. Valdez, L. Ouyang, and J. Bolen, Public Health and Rare Diseases: Oxymoron No More, Preventing Chronic Disease, vol.13, pp.5-15, 2016.
DOI : 10.5888/pcd13.150491

URL : https://www.cdc.gov/pcd/issues/2016/pdf/15_0491.pdf

E. Shash, A. Negrouk, S. Marreaud, V. Golfinopoulos, D. Lacombe et al., International clinical trials setting for rare cancers: organisational and regulatory constraints???the EORTC perspective, ecancermedicalscience, vol.7, p.321, 2013.
DOI : 10.3332/ecancer.2013.321

K. Boycott, M. Vanstone, D. Bulman, and A. Mackenzie, Rare-disease genetics in the era of next-generation sequencing: discovery to translation, Nature Reviews Genetics, vol.48, issue.10, pp.681-91, 2013.
DOI : 10.1172/JCI44929

A. Rath and S. Peixoto, Disease registries in Europe, 2016.

E. Augustine, H. Adams, and J. Mink, Clinical Trials in Rare Disease, Journal of Child Neurology, vol.79, issue.1, pp.1142-50, 2013.
DOI : 10.1212/WNL.0b013e31826c19b4

A. Rath, A. Olry, F. Dhombres, M. Brandt, B. Urbero et al., Representation of rare diseases in health information systems: The orphanet approach to serve a wide range of end users, Human Mutation, vol.32, issue.5, pp.803-811, 2012.
DOI : 10.1002/humu.21469

S. Rodger, H. Lochmuller, A. Tassoni, K. Gramsch, K. Konig et al., The TREAT-NMD care and trial site registry: an online registry to facilitate clinical research for neuromuscular diseases, Orphanet Journal of Rare Diseases, vol.8, issue.1, p.171, 2013.
DOI : 10.1212/WNL.0b013e318282334e

B. Potter, S. Khangura, K. Tingley, P. Chakraborty, and J. Little, Translating rare-disease therapies into improved care for patients and families: what are the right outcomes, designs, and engagement approaches in health-systems research?, Genetics in Medicine, vol.30, issue.2, pp.117-140, 2015.
DOI : 10.1186/1750-1172-7-37

T. Smith, C. Williamson, P. Beresford, and M. , Methodology of clinical trials for rare diseases, Best Practice & Research Clinical Rheumatology, vol.28, issue.2, pp.247-62, 2014.
DOI : 10.1016/j.berh.2014.03.004

S. Gupta, M. Faughnan, G. Tomlinson, and A. Bayoumi, A framework for applying unfamiliar trial designs in studies of rare diseases, Journal of Clinical Epidemiology, vol.64, issue.10, pp.1085-94, 2011.
DOI : 10.1016/j.jclinepi.2010.12.019

E. Korn, L. Mcshane, and B. Freidlin, Statistical Challenges in the Evaluation of Treatments for Small Patient Populations, Science Translational Medicine, vol.376, issue.9742, pp.178-181, 2013.
DOI : 10.1016/S0140-6736(10)61121-X

C. Cornu, B. Kassai, R. Fisch, C. Chiron, C. Alberti et al., Experimental designs for small randomised clinical trials: an algorithm for choice, Orphanet Journal of Rare Diseases, vol.8, issue.1, p.48, 2013.
DOI : 10.1007/s10985-007-9049-x

URL : https://hal.archives-ouvertes.fr/inserm-00818031

J. Gagne, L. Thompson, O. Keefe, K. Kesselheim, and A. , Innovative research methods for studying treatments for rare diseases: methodological review, BMJ, vol.349, issue.nov24 15, p.6802, 2014.
DOI : 10.1136/bmj.g6802

K. Button, J. Ioannidis, C. Mokrysz, B. Nosek, J. Flint et al., Power failure: why small sample size undermines the reliability of neuroscience, Nature Reviews Neuroscience, vol.80, issue.5, pp.365-76, 2013.
DOI : 10.1086/512821

M. Parmar, M. Sydes, and T. Morris, How do you design randomised trials for smaller populations? A framework, BMC Medicine, vol.28, issue.2, p.183, 2016.
DOI : 10.1016/j.berh.2014.03.004

R. Hilgers, K. Roes, and N. Stallard, Directions for new developments on statistical design and analysis of small population group trials, Orphanet Journal of Rare Diseases, vol.8, issue.10, pp.78-94, 2016.
DOI : 10.1186/1750-1172-8-48

K. Roes, A framework: make it useful to guide and improve practice of clinical trial design in smaller populations, BMC Medicine, vol.11, issue.8, p.195, 2016.
DOI : 10.1186/s13023-016-0464-5

S. Nikolakopoulos and K. Roes, van der Tweel I. Sequential designs with small samples: evaluation and recommendations for normal responses, Stat Methods Med Res, 2016.

J. Ioannidis, I. Hozo, and B. Djulbegovic, Optimal type I and type II error pairs when the available sample size is fixed, Journal of Clinical Epidemiology, vol.66, issue.8, pp.903-913, 2013.
DOI : 10.1016/j.jclinepi.2013.03.002

A. Vickers and P. Scardino, The clinically-integrated randomized trial: proposed novel method for conducting large trials at low cost, Trials, vol.59, issue.4, p.14, 2009.
DOI : 10.1016/S0025-7125(16)32001-6

A. European-medicines, Reflection paper on methodological issues in confirmatory clinical trials planned with an adaptive design Available from, 2007.

M. Mauer, L. Collette, and J. Bogaerts, 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, European Journal of Cancer, vol.48, issue.9, pp.1386-91, 2012.
DOI : 10.1016/j.ejca.2011.12.024

R. Tamura, J. Krischer, C. Pagnoux, R. Micheletti, P. Grayson et al., A small n sequential multiple assignment randomized trial design for use in rare disease research, Contemporary Clinical Trials, vol.46, pp.48-51, 2015.
DOI : 10.1016/j.cct.2015.11.010

G. Hlavin, F. Koenig, C. Male, M. Posch, and P. Bauer, Evidence, eminence and extrapolation, Statistics in Medicine, vol.91, issue.3, pp.2117-2149, 2016.
DOI : 10.1038/clpt.2011.345

URL : http://onlinelibrary.wiley.com/doi/10.1002/sim.6865/pdf

L. Billingham, K. Malottki, and N. Steven, Research methods to change clinical practice for patients with rare cancers, The Lancet Oncology, vol.17, issue.2, pp.70-80, 2016.
DOI : 10.1016/S1470-2045(15)00396-4

R. Joppi, V. Bertele, and S. Garattini, Orphan drug development is not taking off, British Journal of Clinical Pharmacology, vol.64, issue.5, pp.494-502, 2009.
DOI : 10.1111/j.1365-2125.2009.03369.x

URL : http://onlinelibrary.wiley.com/doi/10.1111/j.1365-2125.2009.03369.x/pdf

C. Gluud, J. Brok, Y. Gong, and R. Koretz, Hepatology may have problems with putative surrogate outcome measures, Journal of Hepatology, vol.46, issue.4, pp.734-776, 2007.
DOI : 10.1016/j.jhep.2007.01.003

G. Molenberghs, H. Geys, and M. Buyse, Evaluation of surrogate endpoints in randomized experiments with mixed discrete and continuous outcomes, Statistics in Medicine, vol.89, issue.20
DOI : 10.7326/0003-4819-118-9-199305010-00003

G. Molenberghs, T. Burzykowski, A. Alonso, P. Assam, A. Tilahun et al., A unified framework for the evaluation of surrogate endpoints in mental-health clinical trials, Statistical Methods in Medical Research, vol.5, issue.379, pp.205-241, 2009.
DOI : 10.1002/pst.207

H. Ensor, R. Lee, C. Sudlow, and C. Weir, Statistical approaches for evaluating surrogate outcomes in clinical trials: A systematic review, Journal of Biopharmaceutical Statistics, vol.27, issue.710, pp.1-21, 2015.
DOI : 10.1111/j.1541-0420.2011.01736.x

R. Joppi, C. Gerardi, V. Bertele, and S. Garattini, Letting post-marketing bridge the evidence gap: the case of orphan drugs: Table 1, BMJ, vol.353, 2016.
DOI : 10.1136/bmj.i2978

J. Jakobsen, C. Gluud, P. Winkel, T. Lange, and J. Wetterslev, The thresholds for statistical and clinical significance ??? a five-step procedure for evaluation of intervention effects in randomised clinical trials, BMC Medical Research Methodology, vol.346, issue.1, p.34, 2014.
DOI : 10.1136/bmj.f839

E. Perea-milla, L. Aycaguer, J. Cerda, F. Saiz, F. Rivas-ruiz 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, Trials, vol.3, issue.1, p.70, 2009.
DOI : 10.1186/1477-7517-3-16

M. Putzeist, A. Mantel-teeuwisse, C. Wied, A. Hoes, H. Leufkens et al., Drug development for exceptionally rare metabolic diseases: challenging but not impossible, Orphanet Journal of Rare Diseases, vol.8, issue.1, p.179, 2013.
DOI : 10.1186/1750-1172-6-59

L. Matza, D. Patrick, A. Riley, J. Alexander, L. Rajmil 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, Value in Health, vol.16, issue.4, pp.461-79, 2013.
DOI : 10.1016/j.jval.2013.04.004

G. Tafuri, M. Pagnini, J. Moseley, M. Massari, F. Petavy et al., How aligned are the perspectives of EU regulators and HTA bodies? A comparative analysis of regulatory-HTA parallel scientific advice, British Journal of Clinical Pharmacology, vol.97, issue.4, pp.965-73, 2016.
DOI : 10.1002/cpt.59

J. Forman, D. Taruscio, V. Llera, L. Barrera, T. Cote et al., The need for worldwide policy and action plans for rare diseases, Acta Paediatrica, vol.371, issue.8, pp.805-812, 2012.
DOI : 10.1016/S0140-6736(08)60875-2

M. Skoog, J. Saarimäki, C. Gluud, M. Sheinin, K. Erlendsson et al., Transparency and registration in clinical research in the Nordic countries (Report) Nordic Trial Alliance, NordForsk. Oslo. Norge, vol.2015, pp.1-108

A. Bajard, S. Chabaud, C. Cornu, A. Castellan, S. Malik et al., An in silico approach helped to identify the best experimental design, population, and outcome for future randomized clinical trials, Journal of Clinical Epidemiology, vol.69, pp.125-161, 2015.
DOI : 10.1016/j.jclinepi.2015.06.024

D. Bolignano and A. Pisano, Good-quality research in rare diseases: trials and tribulations, Pediatric Nephrology, vol.327, issue.11, pp.217-240, 2016.
DOI : 10.1136/bmj.327.7405.47

R. Ahmed, U. Duerr, K. Gavenis, R. Hilgers, and O. Gross, Challenges for Academic Investigator???Initiated Pediatric Trials for Rare Diseases, Clinical Therapeutics, vol.36, issue.2, pp.184-90, 2014.
DOI : 10.1016/j.clinthera.2014.01.013

T. Friede, C. Rover, S. Wandel, and B. Neuenschwander, Meta-analysis of few small studies in orphan diseases, Research Synthesis Methods, vol.36, issue.1, pp.79-91, 2016.
DOI : 10.18637/jss.v036.i03

J. Wetterslev, K. Thorlund, J. Brok, and C. Gluud, Trial sequential analysis may establish when firm evidence is reached in cumulative meta-analysis, Journal of Clinical Epidemiology, vol.61, issue.1, pp.64-75, 2007.
DOI : 10.1016/j.jclinepi.2007.03.013

G. Imberger, K. Thorlund, C. Gluud, and J. Wetterslev, False-positive findings in Cochrane meta-analyses with and without application of trial sequential analysis: an empirical review, BMJ Open, vol.6, issue.8, pp.11890-11906, 2016.
DOI : 10.1136/bmjopen-2016-011890

E. Kulinskaya and J. Wood, Trial sequential methods for meta-analysis, Research Synthesis Methods, vol.18, issue.7, pp.212-232, 2014.
DOI : 10.1002/(SICI)1097-0258(19990915/30)18:17/18<2271::AID-SIM254>3.0.CO;2-Z

J. Ioannidis, How to Make More Published Research True, PLoS Medicine, vol.51, issue.7145, p.1001747, 2014.
DOI : 10.1371/journal.pmed.1001747.t002

URL : https://doi.org/10.1371/journal.pmed.1001747

B. Alberts, M. Kirschner, S. Tilghman, and H. Varmus, Rescuing US biomedical research from its systemic flaws, Proceedings of the National Academy of Sciences, vol.10, issue.3, pp.5773-5780, 2014.
DOI : 10.1187/cbe.11-02-0013

URL : http://www.pnas.org/content/111/16/5773.full.pdf

J. Ioannidis, D. Fanelli, D. Dunne, and S. Goodman, Meta-research: Evaluation and Improvement of Research Methods and Practices, PLOS Biology, vol.505, issue.7485, pp.1002264-1002267, 2015.
DOI : 10.1371/journal.pbio.1002264.t002

P. Gotzsche, Blinding during data analysis and writing of manuscripts, Controlled Clinical Trials, vol.17, issue.4, pp.285-90, 1996.
DOI : 10.1016/0197-2456(95)00263-4

R. Maccoun and S. Perlmutter, Blind analysis: Hide results to seek the truth, Nature, vol.526, issue.7572, pp.187-196, 2015.
DOI : 10.1038/526187a

T. Jarvinen, R. Sihvonen, M. Bhandari, S. Sprague, A. Malmivaara et al., Blinded interpretation of study results can feasibly and effectively diminish interpretation bias, Journal of Clinical Epidemiology, vol.67, issue.7, pp.769-72, 2014.
DOI : 10.1016/j.jclinepi.2013.11.011