Hostname: page-component-8448b6f56d-gtxcr Total loading time: 0 Render date: 2024-04-24T20:12:55.472Z Has data issue: false hasContentIssue false

The Evidence-based Medicine Paradigm: Where are We 20 Years Later? Part 1

Published online by Cambridge University Press:  23 September 2014

Shashi S. Seshia*
Affiliation:
Department of Pediatrics, Division of Pediatric Neurology, University of Saskatchewan, Royal University Hospital, Saskatoon, Saskatchewan
G. Bryan Young
Affiliation:
Department of Clinical Neurological Sciences, Western University, London, Ontario, Canada
*
Department of Pediatrics, Division of Pediatric Neurology, University of Saskatchewan, Royal University Hospital, 108 Hospital Drive, Saskatoon, Saskatchewan, S7N 0W8, Canada. Email: sseshia@yahoo.ca
Rights & Permissions [Opens in a new window]

Abstract:

Core share and HTML view are not available for this content. However, as you have access to this content, a full PDF is available via the ‘Save PDF’ action button.

The evidence-based medicine (EBM) paradigm, introduced in 1992, has had a major and positive impact on all aspects of health care. However, widespread use has also uncovered some limitations; these are discussed from the perspectives of two clinicians in this, the first of a two part narrative review. For example, there are credible reservations about the validity of hierarchical levels of evidence, a core element of the EBM paradigm. In addition, potential and actual methodological and statistical deficiencies have been identified, not only in many published randomized controlled trials but also in systematic reviews, both rated highly for evidence in EBM classifications. Ethical violations compromise reliability of some data. Clinicians need to be conscious of potential limitations in some of the cornerstones of the EBM paradigm, and to deficiencies in the literature.

Résumé:

Résumé:

Le paradigme de la médecine fondée sur des données probantes (MFDP) introduit en 1992 a eu un impact positif majeur sur tous les aspects des soins de santé. Cependant, son utilisation répandue a également mis au jour certaines limites. Nous discutons de ces limites du point de vue de deux cliniciens dans la première partie de cet examen narratif. Il existe, par exemple, des réserves crédibles concernant la validité des niveaux hiérarchiques de preuves, un élément clé du paradigme de la MFDP. De plus, des lacunes potentielles et réelles dans la méthodologie et l'analyse statistique ont été identifiées, non seulement dans plusieurs essais cliniques randomisés qui ont été publiés, mais également dans les revues systématiques, deux sources de données très prisées pour établir les classifications dans la MFDP. Les manquements à l'éthique compromettent la fiabilité de certaines données. Les cliniciens doivent être conscients des limites potentielles présentes dans certains principes de base du paradigme de la MFDP et des lacunes présentes dans la littérature.

Type
Review Article
Copyright
Copyright © The Canadian Journal of Neurological 2013

References

1. Evidence-Based Medicine Working Group. Evidence-based medicine. A new approach to teaching the practice of medicine. JAMA. 1992;268:2420–5.Google Scholar
2. Straus, SE, Glasziou, P, Richardson, WS, Haynes, RB. Evidence-based Medicine; 4th ed. Toronto: Churchill Livingstone Elsevier, 2011.Google Scholar
3. Goldenberg, MJ, Borgerson, K, Bluhm, R. The nature of evidence in evidence-based medicine: guest editors’ introduction. Perspect Biol Med. 2009;52:164–7.Google Scholar
4. Djulbegovic, B, Guyatt, GH, Ashcroft, RE. Epistemologic inquiries in evidence-based medicine. Cancer Control. 2009;16:158–68.CrossRefGoogle ScholarPubMed
5. Montori, VM, Guyatt, GH. Progress in evidence-based medicine. JAMA. 2008;300:1814–6.Google Scholar
6. Burneo, JG, Demaerschalk, BM, Jenkins, ME, editors. Neurology: An evidence-based approach. New York: Springer; 2012.Google Scholar
7. Wiebe, S, Demaerschalk, B. Evidence based care in the neurosciences. Can J Neurol Sci. 2002;29:115–9.Google Scholar
8. Gronseth, G, French, J. Practice parameters and technology assessments: what they are, what they are not, and why you should care. Neurology. 2008;71:1639–43.CrossRefGoogle Scholar
9. Cochrane Collaboration. [Cited 2012 Aug 27]. Available from: http://www.cochrane.org/cochrane-reviews/ Google Scholar
10. Centre for Evidence-Based Medicine, University of Oxford. Oxford, UK. [Cited 2012 Dec 12]. Available from: http://www.cebm.net/ Google Scholar
11. CONSORT group. [Cited 2012 Dec 11]. Avaiable from: http://www.consort-statement.org/ Google Scholar
12. Davidoff, F, Haynes, B, Sackett, D, Smith, R. Evidence based medicine. BMJ. 1995;310:1085–6.Google Scholar
13. Sackett, DL, Rosenberg, WM, Gray, JA, Haynes, RB, Richardson, WS. Evidence based medicine: what it is and what it isn’t. BMJ. 1996;312:71–2.Google Scholar
14. Straus, SE, McAlister, FA. Evidence-based medicine: a commentary on common criticisms. CMAJ. 2000;163:837–41.Google Scholar
15. Elamin, MB, Montori, VM. The hierarchy of evidence: From unsystematic clinical observations to systematic reviews. Chapter 2. In: Burneo, JG, Demaerschalk, BM, Jenkins, ME, editors. Neurology: An evidence-based approach. New York: Springer; 2012:p.1124.Google Scholar
16. Anonymous. The periodic health examination: 2. 1985 update. Canadian Task Force on the Periodic Examination. CMAJ. 1986;134:724–7.Google Scholar
17. French, J, Gronseth, G. Lost in a jungle of evidence: we need a compass. Neurology. 2008;71:1634–8.Google Scholar
18. Lee, SK, Singhal, N, Aziz, K, Cronin, CM. The EPIQ evidence reviews - practical tools for an integrated approach to knowledge translation. Paediatr Child Health. 2011;16:629–30.Google Scholar
19. Atkins, D, Eccles, M, Flottorp, S, et al. Systems for grading the quality of evidence and the strength of recommendations I: critical appraisal of existing approaches The GRADE Working Group. BMC Health Serv Res. 2004;4:38.Google Scholar
20. Feinstein, AR, Horwitz, RI. Problems in the “evidence” of “evidence-based medicine”. Am J Med. 1997;103:529–35.Google Scholar
21. Bluhm, R, Borgerson, K. Evidence-based Medicine. In: Gifford, F, editor. Handbook of the Philosophy of Science. Volume 16: Philosophy of Medicine. Amsterdam: Elsevier, BV; 2011: p. 203–38.Google Scholar
22. Cohen, AM, Stavri, PZ, Hersh, WR. A categorization and analysis of the criticisms of Evidence-Based Medicine. Int J Med Inform. 2004;73:3543.Google Scholar
23. Rawlins, M. De testimonio: on the evidence for decisions about the use of therapeutic interventions. Lancet. 2008;372:2152–61.Google Scholar
24. Rosner, AL. Evidence-based medicine: revisiting the pyramid of priorities. J Bodyw Mov Ther. 2012;16:42–9.Google Scholar
25. Tonelli, MR. The limits of evidence-based medicine. Respir Care. 2001;46:1435,40; discussion 1440-1.Google ScholarPubMed
26. Upshur, RE. Are all evidence-based practices alike? Problems in the ranking of evidence. CMAJ. 2003;169:672–3.Google Scholar
27. Upshur, R, Tracy, CS. Evidence-based medicine: perils and pitfalls. N Z Fam Physician. 2003;30(5):327–30.Google Scholar
28. Concato, J. Observational versus experimental studies: what’s the evidence for a hierarchy? NeuroRx. 2004;1:341–7.Google Scholar
29. Goodin, DS, Reder, AT. Evidence-based medicine: promise and pitfalls. Mult Scler. 2012;18:947–8.Google Scholar
30. Jadad, AR, Enkin, MW. Randomized controlled trials. Oxford, UK: Blackwell Publishing BMJI Books; 2007.Google Scholar
31. Rawlins, MD. De Testimonio: On the evidence for decisions about the use of therapeutic interventions. London, UK: Royal College of Physicians; 2008.Google Scholar
32. Atkins, D, Best, D, Briss, PA, et al. Grading quality of evidence and strength of recommendations. BMJ. 2004;328:1490.Google ScholarPubMed
33. Guyatt, GH, Oxman, AD, Vist, GE, et al. GRADE: an emerging consensus on rating quality of evidence and strength of recommendations. BMJ. 2008;336:924–6.CrossRefGoogle ScholarPubMed
34. Hill, AB. Reflections on controlled trial. Ann Rheum Dis. 1966;25:107–13.Google Scholar
35. Schwartz, D, Lellouch, J. Explanatory and pragmatic attitudes in therapeutical trials. J Chronic Dis. 1967;20:637–48.CrossRefGoogle ScholarPubMed
36. Cochrane, AL. Archie Cochrane in his own words. Selections arranged from his 1972 introduction to “Effectiveness and Efficiency: Random Reflections on the Health Services” 1972. Control Clin Trials. 1989;10:428–33.Google Scholar
37. Horwitz, RI. The dark side of evidence-based medicine. Cleve Clin J Med. 1996;63:320–3.CrossRefGoogle ScholarPubMed
38. Hartling, L, McAlister, FA, Rowe, BH, Ezekowitz, J, Friesen, C, Klassen, TP. Challenges in systematic reviews of therapeutic devices and procedures. Ann Intern Med. 2005;142:1100–11.Google Scholar
39. Patsopoulos, NA. A pragmatic view on pragmatic trials. Dialogues Clin Neurosci. 2011;13:217–24.Google Scholar
40. Buchanan, WW, Kean, WF. Evidence based medicine: the median is not the message. J Rheumatol. 2001;28:2371–2.Google Scholar
41. Brigo, F. New anti-epileptic drugs: overcoming the limits of randomised controlled trials. Int J Evid Based Healthc. 2011;9:440–3.Google Scholar
42. Glasziou, P, Chalmers, I, Rawlins, M, McCulloch, P. When are randomised trials unnecessary? Picking signal from noise. BMJ. 2007;334:349–51.Google Scholar
43. Wikipedia. The randomized controlled trial. [Cited 2012 Dec 12]. Available at: http://en.wikipedia.org/wiki/Randomized_controlled_trial Google Scholar
44. Schulz, KF, Altman, DG, Moher, D, CONSORT Group. CONSORT 2010 statement: updated guidelines for reporting parallel group randomized trials. Ann Intern Med. 2010;152:726–32.Google Scholar
45. Makhinson, M. Biases in the evaluation of psychiatric clinical evidence. J Nerv Ment Dis. 2012;200:7682.Google Scholar
46. Zwarenstein, M, Oxman, A. Pragmatic Trials in Health Care Systems (PRACTIHC). Why are so few randomized trials useful, and what can we do about it? J Clin Epidemiol. 2006;59:1125–6.Google Scholar
47. Weiss, NS, Koepsell, TD, Psaty, BM. Generalizability of the results of randomized trials. Arch Intern Med. 2008;168:133–5.CrossRefGoogle ScholarPubMed
48. Senn, S, Julious, S. Measurement in clinical trials: a neglected issue for statisticians? Stat Med. 2009;28:3189–209.Google Scholar
49. Gifford, F. Uncertainty about clinical equipoise. Clinical equipoise and the uncertainty principles both require further scrutiny. BMJ. 2001;322:795.Google Scholar
50. Oricha, BS, Yauri, MB. Uncertainty principle versus clincal equipoise in clinical trials in Sub-Saharan Africa: Are they really tenable? Ann African Med. 2003;2(2):99100.Google Scholar
51. Weijer, C, Shapiro, SH, Cranley Glass, K. For and against: clinical equipoise and not the uncertainty principle is the moral underpinning of the randomised controlled trial. BMJ. 2000;321:756–8.CrossRefGoogle Scholar
52. Djulbegovic, B, Lacevic, M, Cantor, A, et al. The uncertainty principle and industry-sponsored research. Lancet. 2000;356:635–8.Google Scholar
53. Rich, W, Finer, NN, Gantz, MG, et al. Enrollment of extremely low birth weight infants in a clinical research study may not be representative. Pediatrics. 2012;129:480–4.Google Scholar
54. Biau, DJ, Kerneis, S, Porcher, R. Statistics in brief: the importance of sample size in the planning and interpretation of medical research. Clin Orthop Relat Res. 2008;466:2282–8.Google Scholar
55. Heinzl, H, Benner, A, Ittrich, C, Mittlbock, M. Proposals for sample size calculation programs. Methods Inf Med. 2007;46:655–61.Google Scholar
56. van der Tweel, I, Askie, L, Vandermeer, B, et al. Standard 4: determining adequate sample sizes. Pediatrics. 2012; 129 Suppl 3:S13845.Google Scholar
57. Sakpal, TV. Sample size estimation in clinical trial. Perspect Clin Res. 2010;1:67–9.Google Scholar
58. Julious, SA, Campbell, MJ. Tutorial in biostatistics: sample sizes for parallel group clinical trials with binary data. Stat Med. 2012;31:2904–36.CrossRefGoogle ScholarPubMed
59. Charles, P, Giraudeau, B, Dechartres, A, Baron, G, Ravaud, P. Reporting of sample size calculation in randomised controlled trials: review. BMJ. 2009;338:b1732.Google Scholar
60. Bacchetti, P. Current sample size conventions: flaws, harms, and alternatives. BMC Med. 2010;8:17.Google Scholar
61. Bacchetti, P, Deeks, SG, McCune, JM. Breaking free of sample size dogma to perform innovative translational research. Sci Transl Med. 2011;3:87ps24.Google Scholar
62. Hill, AB. Medical ethics and controlled trials. BMJ. 1963;1:1043–9.Google Scholar
63. Hollis, S, Campbell, F. What is meant by intention to treat analysis? Survey of published randomised controlled trials. BMJ. 1999;319:670–4.Google Scholar
64. Gravel, J, Opatrny, L, Shapiro, S. The intention-to-treat approach in randomized controlled trials: are authors saying what they do and doing what they say? Clin Trials. 2007;4:350–6.Google Scholar
65. Abraha, I, Montedori, A. Modified intention to treat reporting in randomised controlled trials: systematic review. BMJ. 2010;340:c2697.Google Scholar
66. Kruse, RL, Alper, BS, Reust, C, Stevermer, JJ, Shannon, S, Williams, RH. Intention-to-treat analysis: Who is in? Who is out? J Fam Pract. 2002;51:969–71.Google Scholar
67. McAlister, FA. The “number needed to treat” turns 20-and continues to be used and misused. CMAJ. 2008;179:549–53.CrossRefGoogle Scholar
68. Smeeth, L, Haines, A, Ebrahim, S. Numbers needed to treat derived from meta-analyses-sometimes informative, usually misleading. BMJ. 1999;318:1548–51.Google Scholar
69. Hutton, JL. Number needed to treat and number needed to harm are not the best way to report and assess the results of randomised clinical trials. Br J Haematol. 2009;146:2730.Google Scholar
70. Julious, SA. Issues with number needed to treat. Stat Med. 2005;24:3233–5.Google Scholar
71. Greenhalgh, T. How to read a paper. Statistics for the non-statistician. II: “Significant” relations and their pitfalls. BMJ. 1997;315:422–5.Google Scholar
72. Goodman, S. A dirty dozen: twelve p-value misconceptions. Semin Hematol. 2008;45:135–40.Google Scholar
73. Bhandari, M, Montori, VM, Schemitsch, EH. The undue influence of significant p-values on the perceived importance of study results. Acta Orthop. 2005;76:291–5.Google Scholar
74. Guyatt, G, Jaeschke, R, Heddle, N, Cook, D, Shannon, H, Walter, S. Basic statistics for clinicians: 2. Interpreting study results: confidence intervals. CMAJ. 1995;152:169–73.Google Scholar
75. Guyatt, GH, Oxman, AD, Kunz, R, et al. GRADE guidelines 6. Rating the quality of evidence-imprecision. J Clin Epidemiol. 2011;64:1283–93.Google Scholar
76. Glasziou, P, Doll, H. Was the study big enough? Two cafe rules. Evid Based Med. 2006;11:6970.Google Scholar
77. Yudkin, JS, Lipska, KJ, Montori, VM. The idolatry of the surrogate. BMJ. 2011;343:d7995.CrossRefGoogle ScholarPubMed
78. Aronson, JK. Surrogate end points: studying their benefits, taxonomy, and semantics. BMJ. 2012;344:e750.Google Scholar
79. Colatsky, TJ. Reassessing the validity of surrogate markers of drug efficacy in the treatment of coronary artery disease. Curr Opin Investig Drugs. 2009;10:239–44.Google Scholar
80. Psaty, BM, Weiss, NS, Furberg, CD, et al. Surrogate end points, health outcomes, and the drug-approval process for the treatment of risk factors for cardiovascular disease. JAMA. 1999;282:786–90.Google Scholar
81. Fleming, TR, DeMets, DL. Surrogate end points in clinical trials: are we being misled? Ann Intern Med. 1996;125:605–13.Google Scholar
82. Montori, VM, Shah, ND. What have we learnt from the rosiglitazone saga? BMJ. 2011;342:d1354.Google Scholar
83. Cordoba, G, Schwartz, L, Woloshin, S, Bae, H, Gotzsche, PC. Definition, reporting, and interpretation of composite outcomes in clinical trials: systematic review. BMJ. 2010;341:c3920.Google Scholar
84. Ferreira-Gonzalez, I, Busse, JW, Heels-Ansdell, D, et al. Problems with use of composite end points in cardiovascular trials: systematic review of randomised controlled trials. BMJ. 2007;334:786.Google Scholar
85. Ferreira-Gonzalez, I, Permanyer-Miralda, G, Busse, JW, et al. Methodologic discussions for using and interpreting composite endpoints are limited, but still identify major concerns. J Clin Epidemiol. 2007;60:651,7; discussion 658-62.Google Scholar
86. Ferreira-Gonzalez, I, Permanyer-Miralda, G, Busse, JW, et al. Composite outcomes can distort the nature and magnitude of treatment benefits in clinical trials. Ann Intern Med. 2009;150:566–7.Google Scholar
87. Montori, VM, Busse, JW, Permanyer-Miralda, G, Ferreira, I, Guyatt, GH. How should clinicians interpret results reflecting the effect of an intervention on composite endpoints: should I dump this lump? ACP J Club. 2005;143:A8.Google Scholar
88. Montori, VM, Permanyer-Miralda, G, Ferreira-Gonzalez, I, et al. Validity of composite end points in clinical trials. BMJ. 2005;330:594–6.Google Scholar
89. Sun, X, Briel, M, Busse, JW, et al. Credibility of claims of subgroup effects in randomised controlled trials: systematic review. BMJ. 2012;344:e1553.Google Scholar
90. Wang, R, Lagakos, SW, Ware, JH, Hunter, DJ, Drazen, JM. Statistics in medicine-reporting of subgroup analyses in clinical trials. N Engl J Med. 2007;357:2189–94.CrossRefGoogle ScholarPubMed
91. Sacks, FM, Pfeffer, MA, Moye, LA, et al. The effect of pravastatin on coronary events after myocardial infarction in patients with average cholesterol levels. Cholesterol and Recurrent Events Trial investigators. N Engl J Med. 1996;335:1001–9.Google Scholar
92. Guyatt, GH, Briel, M, Glasziou, P, Bassler, D, Montori, VM. Problems of stopping trials early. BMJ. 2012;344:e3863.Google Scholar
93. Bassler, D, Ferreira-Gonzalez, I, Briel, M, et al. Systematic reviewers neglect bias that results from trials stopped early for benefit. J Clin Epidemiol. 2007;60:869–73.Google Scholar
94. Bassler, D, Montori, VM, Briel, M, et al. Reflections on meta-analyses involving trials stopped early for benefit: Is there a problem and if so, what is it? Stat Methods Med Res. 2013;22:159–68.Google Scholar
95. Cady, RK, Lipton, RB, Hall, C, Stewart, WF, O’Quinn, S, Gutterman, D. Treatment of mild headache in disabled migraine sufferers: results of the Spectrum Study. Headache. 2000;40:792–7.Google Scholar
96. Tellez-Zenteno, JF, Wiebe, S. Hippocampal stimulation in the treatment of epilepsy. Neurosurg Clin N Am. 2011;22:465,75, vi.CrossRefGoogle ScholarPubMed
97. Andrasik, F, Powers, SW, McGrath, PJ. Methodological considerations in research with special populations: children and adolescents. Headache. 2005;45:520–5.Google Scholar
98. Lewis, DW, Winner, P, Wasiewski, W. The placebo responder rate in children and adolescents. Headache. 2005;45:232–9.Google Scholar
99. Klassen, TP, Hartling, L, Craig, JC, Offringa, M. Children are not just small adults: the urgent need for high-quality trial evidence in children. PLoS Med. 2008;5:e172.Google Scholar
100. Klassen, TP, Hartling, L, Hamm, M, van der Lee, JH, Ursum, J, Offringa, M. StaR Child Health: an initiative for RCTs in children. Lancet. 2009;374:1310–2.Google Scholar
101. Hartling, L, Wittmeier, KD, Caldwell, PH, et al. StaR Child Health: developing evidence-based guidance for the design, conduct, and reporting of pediatric trials. Clin Pharmacol Ther. 2011;90:727–31.Google Scholar
102. Croskerry, P. Perspectives on diagnostic failure and patient safety. Healthc Q. 2012;15 Suppl:50-6.Google Scholar
103. Croskerry, P. A universal model of diagnostic reasoning. Acad Med. 2009;84:1022–8.Google Scholar
104. Croskerry, P. Context is everything or how could I have been that stupid? Healthc Q. 2009;12 Spec No Patient:e171-6.Google Scholar
105. Croskerry, P, Abbass, AA, Wu, AW. How doctors feel: affective issues in patients’ safety. Lancet. 2008;372:1205–6.Google Scholar
106. Heneghan, C, Glasziou, P, Thompson, M, et al. Diagnostic strategies used in primary care. BMJ. 2009;338:b946.Google Scholar
107. Cruz, MF, Edwards, J, Dinh, MM, Barnes, EH. The effect of clinical history on accuracy of electrocardiograph interpretation among doctors working in emergency departments. Med J Aust. 2012;197:161–5.Google Scholar
108. Katelaris, A. What influences clinical decision making? Med J Aust. 2012;197:129.Google Scholar
109. Perera, R, Heneghan, C, Badenoch, D. Statistics toolkit. Oxford, UK: Blackwell publishing; 2008.Google Scholar
110. Feinstein, AR. Misguided efforts and future challenges for research on “diagnostic tests”. J Epidemiol Community Health. 2002;56:330–2.Google Scholar
111. Stegenga, J. Is meta-analysis the platinum standard of evidence? Stud Hist Philos Biol Biomed Sci. 2011;42:497507.Google Scholar
112. Coyne, JC, Thombs, BD, Hagedoorn, M. Ain’t necessarily so: review and critique of recent meta-analyses of behavioral medicine interventions in health psychology. Health Psychol. 2010;29:107–16.Google Scholar
113. Clark, DA. The end of evidence-based medicine? Inflammopharmacology. 2012;20:187–93.Google Scholar
114. Rutjes, AW, Reitsma, JB, Di Nisio, M, Smidt, N, van Rijn, JC, Bossuyt, PM. Evidence of bias and variation in diagnostic accuracy studies. CMAJ. 2006;174:469–76.Google Scholar
115. Shojania, KG, Sampson, M, Ansari, MT, et al. Technical review number 16: Updating systematic reviews. AHRQ Publication No. 07-0087. Rockville (MD): U. S. Department of Health and Human Services; 2007.Google Scholar
116. Hartling, L, Bond, K, Santaguida, PL, Viswanathan, M, Dryden, DM. Testing a tool for the classification of study designs in systematic reviews of interventions and exposures showed moderate reliability and low accuracy. J Clin Epidemiol. 2011;64:861–71.Google Scholar
117. Mutasingwa, DR, Ge, H, Upshur, RE. How applicable are clinical practice guidelines to elderly patients with comorbidities? Can Fam Physician. 2011;57:e25362.Google Scholar
118. Lugtenberg, M, Burgers, JS, Clancy, C, Westert, GP, Schneider, EC. Current guidelines have limited applicability to patients with comorbid conditions: a systematic analysis of evidence-based guidelines. PLoS One. 2011;6:e25987.Google Scholar
119. Ballantyne, AJ, Rogers, WA. Sex bias in studies selected for clinical guidelines. J Womens Health (Larchmt). 2011;20:1297–306.Google Scholar
120. Fahy, K, Tracy, SK. Critique of Cochrane systematic review of home-like setting for birth. Int J Evid Based Healthc. 2007;5:360–4.Google Scholar
121. Cundiff, DK. A systematic review of Cochrane anticoagulation reviews. Medscape J Med. 2009;11:5.Google Scholar
122. Bow, S, Klassen, J, Chisholm, A, et al. A descriptive analysis of child-relevant systematic reviews in the Cochrane Database of Systematic Reviews. BMC Pediatr. 2010;10:34.Google Scholar
123. Roseman, M, Milette, K, Bero, LA, et al. Reporting of conflicts of interest in meta-analyses of trials of pharmacological treatments. JAMA. 2011;305:1008–17.Google Scholar
124. Roseman, M, Turner, EH, Lexchin, J, Coyne, JC, Bero, LA, Thombs, BD. Reporting of conflicts of interest from drug trials in Cochrane reviews: cross sectional study. BMJ. 2012;345:e5155.Google Scholar
125. Hartling, L, Hamm, M, Milne, A, et al. Validity and inter-rater reliability testing of quality assessment instruments. Rockville (MD): Agency for health care research and policy, US department of health and human services; 2012.Google Scholar
126. Tharyan, P. Evidence-based medicine: can the evidence be trusted? Indian J Med Ethics. 2011;8:201–7.Google Scholar
127. Tharyan, P. Criminals in the citadel and deceit all along the watchtower: Irresponsibility, fraud, and complicity in the search for scientific truth. Mens Sana Monogr. 2012;10:158–80.Google Scholar
128. Bonita, RE, Adams, S, Whellan, DJ. Reporting of clinical trials: publication, authorship, and trial registration. Heart Fail Clin. 2011;7:561–7.Google Scholar
129. Mowatt, G, Shirran, L, Grimshaw, JM, et al. Prevalence of honorary and ghost authorship in Cochrane reviews. JAMA. 2002;287:2769–71.Google Scholar
130. Wislar, JS, Flanagin, A, Fontanarosa, PB, Deangelis, CD. Honorary and ghost authorship in high impact biomedical journals: a cross sectional survey. BMJ. 2011;343:d6128.Google Scholar
131. Avraham, R. Clinical practice guidelines: the warped incentives in the U. S. healthcare system. Am J Law Med. 2011;37:740.Google Scholar
132. Guyatt, G, Akl, EA, Hirsh, J, et al. The vexing problem of guidelines and conflict of interest: a potential solution. Ann Intern Med. 2010;152:738–41.Google Scholar
133. Neuman, J, Korenstein, D, Ross, JS, Keyhani, S. Prevalence of financial conflicts of interest among panel members producing clinical practice guidelines in Canada and United States: cross sectional study. BMJ. 2011;343:d5621.Google Scholar
134. Scott, IA, Guyatt, GH. Clinical practice guidelines: the need for greater transparency in formulating recommendations. Med J Aust. 2011;195:2933.Google Scholar
135. Norris, SL, Holmer, HK, Burda, BU, Ogden, LA, Fu, R. Conflict of interest policies for organizations producing a large number of clinical practice guidelines. PLoS One. 2012;7:e37413.Google Scholar
136. Bailey, CS, Fehlings, MG, Rampersaud, YR, Hall, H, Wai, EK, Fisher, CG. Industry and evidence-based medicine: Believable or conflicted? A systematic review of the surgical literature. Can J Surg. 2011;54:321–6.Google Scholar
137. Lundh, A, Barbateskovic, M, Hrobjartsson, A, Gotzsche, PC. Conflicts of interest at medical journals: the influence of industry-supported randomised trials on journal impact factors and revenue - cohort study. PLoS Med. 2010;7:e1000354.Google Scholar
138. Upshur, R, Buetow, S, Loughlin, M, Miles, A. Can academic and clinical journals be in financial conflict of interest situations? The case of evidence-based incorporated. J Eval Clin Pract. 2006;12:405–9.Google Scholar
139. Doshi, P, Jefferson, T, Del Mar, C. The imperative to share clinical study reports: recommendations from the Tamiflu experience. PLoS Med. 2012;9:e1001201.Google Scholar
140. Moynihan, R. Rosiglitazone, marketing, and medical science. BMJ. 2010;340:c1848.Google Scholar
141. Wang, AT, McCoy, CP, Murad, MH, Montori, VM. Association between industry affiliation and position on cardiovascular risk with rosiglitazone: cross sectional systematic review. BMJ. 2010;340:c1344.Google Scholar
142. Zarin, DA, Tse, T. Medicine. Moving toward transparency of clinical trials. Science. 2008;319:1340–2.Google Scholar
143. Rhode, DL, Packel, AK. Ethics and Non Profits. [updated summer 2009; cited 2012 Aug 5] Available at: http://www.ssireview.org/articles/entry/ethics_and_nonprofits Google Scholar
144. Booth, A. Evidence-based practice: triumph of style over substance? Health Info Libr J. 2011;28:237–41.Google Scholar