Response to the consultation paper 'Delegated Act on Post-Authorisation Efficacy Studies' PCPAES/12/01 Public Consultation on PAES

Similar documents
EUROPEAN COMMISSION HEALTH AND CONSUMERS DIRECTORATE-GENERAL. Health systems and products Medicinal products authorisations, EMA

Secretary-General of the European Commission, signed by Mr Jordi AYET PUIGARNAU, Director

PGEU GPUE. Pharmaceutical Group of European Union Groupement Pharmaceutique de l Union Européenne

Accepted refereed manuscript of:

EUROPEAN COMMISSION HEALTH AND FOOD SAFETY DIRECTORATE-GENERAL. PHARMACEUTICAL COMMITTEE 21 October 2015

BACKGROUND + GENERAL COMMENTS

Guideline on good pharmacovigilance practices (GVP)

IAASB Exposure Draft, Proposed ISAE 3000 (Revised), Assurance Engagements Other Than Audits or Reviews of Historical Financial Information

2008 Public Status Report on the Implementation of the European Risk Management Strategy. Executive Summary

Vaccine shortages: Improving cooperation, communication and management Michael Sulzner

Re: Docket No. FDA D Presenting Risk Information in Prescription Drug and Medical Device Promotion

ICH Topic S1C(R2) Dose Selection for Carcinogenicity Studies of Pharmaceuticals. Step 5

C 178/2 Official Journal of the European Union

WORLD MEDICAL ASSOCIATION DECLARATION OF HELSINKI. Ethical Principles for Biomedical Research Involving Human Beings

Response to: Concept paper of 9 February 2011 submitted for public consultation by the European Commission on the

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

DOSE SELECTION FOR CARCINOGENICITY STUDIES OF PHARMACEUTICALS *)

Submission to the Senate Community Affairs Legislation Committee inquiry into the Therapeutic Goods Amendment (2016 Measures No.

Reflection paper on the use of extrapolation in the development of medicines for paediatrics

The Nutrition (Amendment) (EU Exit) Regulations 2018

Regulatory framework

EPF s response to the European Commission s public consultation on the "Summary of Clinical Trial Results for Laypersons"

15 May 2017 Exposure Draft. Response Due Date 23 June Exposure Draft

COUNCIL RECOMMENDATION of 2 December 2003 on cancer screening (2003/878/EC)

Basis for Conclusions: ISA 230 (Redrafted), Audit Documentation

(Legislative acts) REGULATIONS

July 7, Dockets Management Branch (HFA-305) Food and Drug Administration 5630 Fishers Lane, Rm Rockville, MD 20852

Access to newly licensed medicines. Scottish Medicines Consortium

Review of Medicines Act 1968: informal consultation on issues relating to the PLR regime and homeopathy

EMEA WORKING PARTY ON HERBAL MEDICINAL PRODUCTS

European Sponsorship Association

Reporting guidelines

ATMPs & EU GMP Update. Bryan J Wright July 2017

COMMISSION OF THE EUROPEAN COMMUNITIES. Proposal for a REGULATION OF THE EUROPEAN PARLIAMENT AND OF THE COUNCIL

ISA 540, Auditing Accounting Estimates, Including Fair Value Accounting Estimates, and Related Disclosures Issues and Task Force Recommendations

STANDING COMMITTEE ON THE FOOD CHAIN AND ANIMAL HEALTH SECTION ON GENERAL FOOD LAW. Summary Record of Meeting of 30 April 2012

REGULATION (EC) No.141/2000

Use of Standards in Substantial Equivalence Determinations

Additional monitoring of medicines and direct patient reporting impact on the package leaflet

GUIDELINES: PEER REVIEW TRAINING BOD G [Amended BOD ; BOD ; BOD ; Initial BOD ] [Guideline]

Overhauling The 510(k) Process

Collecting and Handling Health Data in a GDPR World

Clinical Evidence in the Daily Work of an Anthroposophic Hospital Dr. med. Harald Matthes, Medical Director, Havelhöhe Hospital in Berlin

The legal basis of this draft Regulation is Art. 11 (4) of Regulation (EC) No. 882/2204.

Dissemination experiences from CONSORT and other reporting guidelines

1. Introduction. 2. Role of PSI as Pharmacy Regulator

EFSA Statement regarding the EU assessment of glyphosate and the socalled

Ref: E 007. PGEU Response. Consultation on measures for improving the recognition of medical prescriptions issued in another Member State

COMMISSION DELEGATED REGULATION (EU).../... of XXX

RE: IESBA s Exposure Draft Proposed Revisions Pertaining to Safeguards in the Code Phase 1

Procedural advice on publication of information on withdrawals of applications related to the marketing authorisation of human medicinal products

EUCERD Recommendations on Rare Disease European Reference Networks (RD ERNs)

SUMMARY REPORT OF THE STANDING COMMITTEE ON THE FOOD CHAIN AND ANIMAL HEALTH HELD IN BRUSSELS ON 10 DECEMBER 2012 (Section General Food Law)

European Medicines Agency

Peer counselling A new element in the ET2020 toolbox

Single market, regulatory environment, industries under vertical legislation Pharmaceuticals : regulatory framework and market authorisations

1 March 2010 EMA/131300/2010 Human Medicines Development and Evaluation. 7 Westferry Circus Canary Wharf London E14 4HB United Kingdom

EUROPEAN COMMISSION DG Employment, Social Affairs and Inclusion. The Advisory Committee on Safety and Health at Work. Supplementary Opinion

Wyss Zürich Regulatory Affairs Seminar

IAASB Main Agenda (March 2005) Page Agenda Item. DRAFT EXPLANATORY MEMORANDUM ISA 701 and ISA 702 Exposure Drafts February 2005

Transparenzvorgaben in der Klinischen Prüfung

Meeting report, September 2005

EVIDENCE AND RECOMMENDATION GRADING IN GUIDELINES. A short history. Cluzeau Senior Advisor NICE International. G-I-N, Lisbon 2 November 2009

MINI SYMPOSIUM - EUMASS - UEMASS European Union of Medicine in Assurance and Social Security

on the advertising of medicinal products for human use

RECOMMENDATION ADOPTED BY THE INTERGOVERNMENTAL COMMITTEE FOR THE NAGOYA PROTOCOL AT ITS FIRST MEETING

GLP in the European Union Ecolabel detergents, GLP and accreditation

Roadmap to review the Nutrition and Health Claims legislation expression of interest to contribute to the upcoming external study

Re: JICPA comments on the IESBA Consultation Paper Improving the Structure of the Code of Ethics for Professional Accountants

COUNCIL OF THE EUROPEAN UNION. Brussels, 7 September 2009 (OR. en) 11261/09 Interinstitutional File: 2008/0002 (COD) DENLEG 51 CODEC 893

European Commission Initiative on Breast Cancer (ECIBC) MEETING OF MONITORING SUBGROUP Ispra, 14 September 2016 (10:00-17:00) Minutes

Other EU Activities Contributing to Harmonization of Labeling

The Joanna Briggs Institute Reviewers Manual 2014

Guideline on influenza vaccines submission and procedural requirements

Draft Agreed by Immunologicals Working Party January Adoption by CVMP for release for consultation 12 March 2009

Experimental Methods in Health Research A. Niroshan Siriwardena

Applicant (Invented) Name Strength Pharmaceutical Form. UMAN BIG 180 I.E./ml Injektionslösung. UMAN BIG 180 IU/ml Solution for injection

COMMITTEE FOR MEDICINAL PRODUCTS FOR HUMAN USE (CHMP)

COMMITTEE OF EXPERTS ON THE CLASSIFICATION OF MEDICINES AS REGARDS THEIR SUPPLY (CD-P-PH/PHO) PROGRAMME RESULTS

EER Assurance Criteria & Assertions IAASB Main Agenda (June 2018)

EUROPEAN COMMISSION SUMMARY REPORT OF THE STANDING COMMITTEE ON PLANTS, ANIMALS, FOOD AND FEED HELD IN BRUSSELS ON 09 JUNE 2015

The EU PIP - a step in Pediatric Drug Development. Thomas Severin Bonn,

Summary of responses: Consultation on determining the amount of a variable monetary penalty. April 2017

DRAFT (Final) Concept Paper On choosing appropriate estimands and defining sensitivity analyses in confirmatory clinical trials

Delegations will find attached document D042445/02.

Access to clinical trial information and the stockpiling of Tamiflu. Department of Health

EFSA PRE-SUBMISSION GUIDANCE FOR APPLICANTS INTENDING TO SUBMIT APPLICATIONS FOR AUTHORISATION OF HEALTH CLAIMS MADE ON FOODS

Survey results - Analysis of higher tier studies submitted without testing proposals

COMMISSION OF THE EUROPEAN COMMUNITIES. Proposal for a DIRECTIVE OF THE EUROPEAN PARLIAMENT AND OF THE COUNCIL

Holders of European Union marketing authorizations

Practical guidance for applicants on the submission of applications on food additives, food enzymes and food flavourings

STANDING COMMITTEE ON THE FOOD CHAIN AND ANIMAL HEALTH SECTION ON GENERAL FOOD LAW. Summary Record of Meeting of 15 July 2009

Dagmar Roth-Behrendt Vice-President of the European Parliament

Workshop for OIE national Focal Points for Veterinary Products November 2010, Johannesburg, South Africa. Presentation

Preliminary library of alcohol health information and warning labels

Problem solving therapy

COMMITTEE FOR PROPRIETARY MEDICINAL PRODUCTS (CPMP) POINTS TO CONSIDER ON MISSING DATA

Kurse Epidemiology & Public Health

EUROPEAN COMMISSION SUMMARY REPORT OF THE STANDING COMMITTEE ON THE FOOD CHAIN AND ANIMAL HEALTH HELD IN BRUSSELS ON 13 JUNE 2014

Transcription:

ESCAMP - Zechenweg 6 79111 Freiburg Germany Zechenweg 6 Tel +49 761-1560305 79111 Freiburg Fax +49 761-1560306 Germany info@escamp.org www.escamp.org Directorate-General for Health and Consumers Unit SANCO/D/5, BE-1049 Brussels Response to the consultation paper 'Delegated Act on Post-Authorisation Efficacy Studies' PCPAES/12/01 Public Consultation on PAES ESCAMP ID number in the Transparency Register: 930773810574-17 The European Scientific Cooperative on Anthroposophic Medicinal Products (ESCAMP, www.escamp.org) is an independent scientific cooperative. The aim of ESCAMP is to develop the scientific basis for a permanent regulatory framework for anthroposophic medicinal products in Europe. The issues raised in the consultation paper 'Delegated Act on Post-Authorisation Efficacy Studies [PAES] from the European Commission' (PCPAES/12/01, henceforth abbreviated PCPAES) concern ESCAMP. See also the Conflict of interest statement, below. Introduction The legal context for the proposed Delegated Act is the adoption of a new EU pharmacovigilance legislation in 2010, referring to the "possibility of requesting the marketing authorisation holder to conduct [PAES] complementing efficacy data that are available at the time of the initial authorization." (DA, p.3) This is further elaborated: "It is set out in the legislation that, in the case of an initial marketing authorisation, PAESs may be required where concerns relating to some aspects of the efficacy of the medicinal product are identified and can be resolved only after the medicinal product has been marketed. Following the granting of the marketing authorisation, they may be imposed when the understanding of the disease or the clinical methodology indicate that previous efficacy evaluations might have to be revised significantly'." (PCPAES, p.6) PAES are currently used "in the framework of conditional and exceptional marketing authorisations or as a follow-up to a serious pharmacovigilance signal or efficacy concern" (PCPAES, p.6). Thus, PAES "have a clear regulatory purpose". (PCPAES, p.6) Geschäftsnummer: VR Freiburg im Br. 700424 Vorsitzender / President: Dr. med. Harald J. Hamre Bankverbindung / Bank account: GLS Gemeinschaftsbank eg Kontonummer / Account No.: 7911875600 BLZ: 43060967 IBAN: DE 4306 0967 7911 8756 00 BIC: GENO DE M 1 GLS

Two possible changes to the present framework for PAES use are: Extension: "the new provisions in the 2010 legislation seem to imply that the boundaries for these studies have been expanded beyond their existing use" (PCPAES, p.5) Clarification: "determine the situations in which post-authorisation efficacy studies may be required" (PCPAES, p.3) and "a more global and systematic approach to post-authorisation efficacy studies Instead of referring to postmarketing studies in different pieces of legislation or hinting at their existence in several articles " (PCPAES, p.5) The PCPAES does not present any explicit scientific evidence or elaborated arguments for an extension of the present framework for PAES use but rather aims at a clarification of the framework, by describing seven "situations in which a post-authorisation efficacy study may be required" (PCPAES, Chapter 5. pp.8-11). These situations are defined by particular properties of the patient groups, the medicinal products in question, the standards of care for the disease, or the clinical studies submitted to regulatory authorities for initial market authorisation. Comments on the PCPAES in general and on Consultation item No 1 (Delegated act on the situations in which a PAES may be required) The PCPAES concerns the legal boundaries for a regulatory decision to require PAES as a result of new concerns about the efficacy of a medicinal product on the market. Such decisions will not only take the efficacy of the product into consideration, but also possible risks. Accordingly, the PCPAES issue can be seen in the context of benefit-risk assessments and their use to inform regulatory decision making, in this case the decision to act by requiring PAES or not to act. This is also clear from the context of the new pharmacovigilance as background for the PCPAES (see above). As a general principle, the criteria for such regulatory action should be appropriate to the concerns raised about the benefit-risk profile of the product; in other words: they should neither be too lax (risking regulatory inactivity where action would be necessary), nor too strict (leading to regulatory requirements of many unnecessary PAES, which would be unethical from a research ethics perspective, and also uneconomic). In addition, regulatory decision making should as far as possible be based on objective criteria, leading to a consistent, transparent practice, in order to prevent inequity and unpredictable situations for marketing authorisation holders, doctors, and patients. In this context, the contribution of the PCPAES document is to list and describe different situations where PAES may be required. Although of value, we do not think this alone is sufficient to assure appropriateness, objectivity, consistency, and transparency of regulatory practice towards PAES. As the PCPAES demonstrates (Chapter 5), the different situations for which PAES may be considered are very disparate in regard to patient characteristics, effects and risks of the products and in regard to several features of the clinical studies required for market authorisation (e.g. outcome measures, follow-up periods, or even study design). Accordingly, the nature and seriousness of concerns leading regulatory authorities to consider PAES, the impacts of such PAES, and consequently the threshold for the authorities to require PAES may vary substantially across different situations and medicinal products. Any delegated act on PAES would therefore have to include a framework in order to tackle this variability of settings for PAES. Since regulatory decisions an PAES will be based on benefit-risk assessments (see above), such a framework can and should be constructed with due consideration to the principles, methods and tools for benefit-risk assessment of medicinal products [1]. - 2 -

It is therefore highly surprising that the PCPAES document, although mentioning benefit-risk assessments repeatedly, does not comment on the available methodologies and tools for benefit-risk assessment and associated decision making especially since the European Medicines Agency recently carried out a large, comprehensive Benefit-risk Methodology Project with the main objective to "improve the current practice of benefit-risk assessment for medicinal products, with an aim to increase the consistency and transparency of the regulatory process" (EMA/213482/2010, Human Medicines Development and Evaluation, 30 March 2010). In order to construct a satisfactory legal framework for PAES, appropriate methodology and tools for risk-benefit assessment should be incorporated and adapted for the specific PAES situation (concerns vs. impact of PAES). The framework might for example include specific tools for the assessment of the concerns raised: seriousness of the concerns (e.g. severity of newly identified potential risk, quantification of potential reduction of efficacy, number of patients affected) quality of evidence for concerns (e.g. clinical studies, case reports and case series, extrapolation from animal or in-vitro experiments, pathophysiological speculation) admitted sources for concerns (e.g. peer-review publications, research or pharmacovigilance reports) Conclusions on the PCPAES in general A delegated act on PAES should not aim to extend of the present framework for PAES use, but should focus on the clarification of the present framework. Reason: No explicit scientific evidence or elaborated arguments for such an extension have been presented in the PCPAES. Conclusions on Consultation Item 1 We do not think that a delegated act which is based solely on a description of the situations in which a PAES may be required will be of added value. Before the Commission considers bringing forward a draft delegated act, the use of available benefit-risk assessment methodologies and tools in order to increase the consistency and transparency of regulatory practice towards PAES must be addressed. Reasons are stated above. Comments on Consultation items No 2 and 6 (Efficacy vs. effectiveness + Study designs) The PCPAES rightly refers to an ambiguity in current EU legislation, which refers to "[PAES] aimed at collecting data to enable the assessment of efficacy of medicinal products for human use in everyday medical practice" (PCPAES, p.8). The assessment of treatment effects "in everyday medical practice" would clearly be understood as 'effectiveness assessment' according to current terminology, while the concept of 'effectiveness' itself is not defined in the relevant EU legislation. Effectiveness in everyday medical practice is commonly evaluated in pragmatic trials, a pragmatic trial being defined as "a randomised controlled trial whose purpose is to inform decisions about practice" [2]. Accordingly, pragmatic trials, like their counterpart 'explanatory trials' (roughly equivalent to the term 'controlled clinical trial' used in medicinal product regulation and in the PCPAES) are both interventional studies, incorporating the key design feature of random assignment of patients to intervention or control groups [3]. The two designs differ in terms of research question, setting, patients, intervention, outcome measures, and relevance to practice (Table 1). - 3 -

Some descriptions of pragmatic trials in the PCPAES are unclear ("pragmatic trials outside the scope of a controlled clinical trial setting" (PCPAES, p.8)) or incorrect ("non-randomised design of the study or [pragmatic] trial" (PCPAES, p.8), "non-interventional studies, including pragmatic trials" (PCPAES, p.11)) and need to be amended. Table 1, copied from [2]: Key differences between trials with explanatory and pragmatic attitudes, adapted from a table presented at the 2008 Society for Clinical Trials meeting by Marion Campbell, University of Aberdeen Question Efficacy can the intervention work? Effectiveness does the intervention work when used in normal practice? Setting Well resourced, ideal setting Normal practice Participants Intervention Outcomes Relevance to practice Highly selected. Poorly adherent participants and those with conditions which might dilute the effect are often excluded Strictly enforced and adherence is monitored closely Often short term surrogates or process measures Indirect little effort made to match design of trial to decision making needs of those in usual setting in which intervention will be implemented Little or no selection beyond the clinical indication of interest Applied flexibly as it would be in normal practice Directly relevant to participants, funders, communities, and healthcare practitioners Direct trial is designed to meet needs of those making decisions about treatment options in setting in which intervention will be implemented Among observational study designs, "analysis of patient registries" are mentioned (PCPAES, p.7). Such registry or database analyses are often retrospective and rely on data collected for another purpose than scientific research (e.g. cost reimbursement or quality assurance). Prospective controlled observational studies (also called non-randomised controlled trials or cohort studies) on the other hand, share with randomised trials the prospective collection of data collected for study purposes. This observational study design is relevant (e.g. see below on vaccines) and should also be explicitly mentioned. To sum up, the designs available for PAES include one design tailored to evaluate efficacy (explanatory randomised trials / controlled clinical trials) and several designs that can be used to evaluate effectiveness (pragmatic randomised trials, observational designs such as cohort studies or database analyses). In this context, the PCPAES holds explanatory trials to be the generally preferred design: "In view of the clear regulatory purpose and the need for robust data as the outcome of a PAES, the large majority of studies will have a clinical trial design" and "generally speaking [PAES] should focus on generating efficacy data". However, according to the description in Table 1, pragmatic randomised trials would be better suited than exploratory randomised trials to assess - 4 -

effects of medicinal products in everyday medical practice which is one of the applications of PAES explicitly mentioned in the legislation (PCPAES, p.8). Furthermore, this and other specific properties of pragmatic trials (Table 1) are prominently featured in the PCPAES, Chapter 5: "Situations in which PAES may be required": need for more relevant, long-term outcomes (Chapters 5.1 and 5.6) flexible intervention [which may include co-medications], (Chapter 5.2) broader range of patient characteristics (Chapter 5.3) and studies in normal, everyday practice (Chapter 5.7) Accordingly, pragmatic randomised trials are at least highly relevant or perhaps even the preferred study design for many PAES situations. The use of observational designs for PAES are implied in the PCPAES (p. 11) with a reference to a Note for guidance on the clinical evaluation of vaccines, issued by the European Medicines Agency (EMEA/CHMP/VWP/164653/2005), according to which "pre-authorisation studies of protective efficacy are not always necessary or feasible" (EMEA Note, p.10), while PAES (the assessment of "vaccine effectiveness during the post-authorisation period", EMEA Note, p.14) can be conducted using randomised trials or "observational cohort studies" (EMEA Note, p.14). Thus, PAES are not a priori restricted to randomised trials. This is not against the principles of scientific or evidence based medicine: The relative merits of randomised trials and observational studies have been subject to a long [4-8] and still ongoing scientific debate [9;10]. Randomised trials have a number of limitations, including generalisability problems as discussed in the PCPAES, but also various threats to internal validity, against which randomisation does not protect (e.g. unmasking of blinding, attrition bias, non-compliance with prescribed medication intake in intervention group, use of study medication in control group) [5;11]. Accordingly, the GRADE system, the current state-of-the-art system for grading the quality of evidence from clinical studies [12], whilst starting with randomised trials placed 'above' observational studies, allows for 'upgrading' and 'downgrading' of both types of studies according to specific study features [13]. Conclusions on Consultation Items No 2 and 6 The descriptions of pragmatic trials and observational studies need some correction and expansion, as described above. We do not agree that "generally speaking [PAES] should focus on generating efficacy data". When planning a PAES, the study design should be suited to the main research question, with due consideration of study feasibility. If the research question is centred on efficacy in an ideal setting, explanatory randomised trials (roughly equivalent to 'controlled clinical trials') would be the preferred design. If effectiveness under everyday practice is more important, pragmatic randomised trials (see Table 1 above) and, for some situations, also well-designed observational studies or analyses, are the preferred candidates. Comments on Consultation item No 3 (Section 5.4: Studies in the context of the European standard of care) In this section different standards of care in two regions (EU vs. the rest of the world), and possible effects of such differences on the generalisability of study results are discussed. The general argumentation is clear, but the exact phrasing should be re-written to improve clarity, since both regions (EU and the rest of the world) include countries with very different standards of care. - 5 -

Freiburg, 13 February 2012 Harald J. Hamre Conflict of interest statement This response to the PCPAES was not sponsored by any third party. ESCAMP has previously received funding from Wala-Heilmittel GmbH and Weleda AG, manufacturers of anthroposophic medicinal products. These sponsors had no influence on the drafting, revision or finalization of the present document. The conflict of interest policy of ESCAMP is stated at: http://www.escamp.org/conflict-ofinterests.html About the author Dr. med. Harald Johan Hamre has previously worked as a general practitioner in Norway and Spain. He works as a senior research scientist at the Institute for Applied Epistemology and Medical Methodology at the University of Witten/Herdecke. He is president and scientific director of ESCAMP. E-Mail: harald.hamre@escamp.org References [1] Mt-Isa S, Tzoulaki I, Callum J, Micaleff A, Ashby D. Weighing benefit risk of medicines: concepts and approaches. Drug Discovery Today: Technologies 2011; 8(1): e29-e35. [2] Zwarenstein M, Treweek S, Gagnier JJ, Altman DG, Tunis S, Haynes B et al. Improving the reporting of pragmatic trials: an extension of the CONSORT statement. BMJ 2008; 337: a2390. [3] Schwartz D, Lellouch J. Explanatory and pragmatic attitudes in therapeutical trials. J Chronic Dis 1967; 20(8): 637-48. [4] Burkhardt R, Kienle G. Controlled clinical trials and medical ethics. Lancet 1978; 2(8104-8105): 1356-9. [5] Feinstein AR. An additional basic science for clinical medicine: II. The limitations of randomized trials. Ann Intern Med 1983; 99(4): 544-50. [6] Black N. Why we need observational studies to evaluate the effectiveness of health care. BMJ 1996; 312(7040): 1215-8. [7] Britton A, McPherson K, McKee M, Sanderson C, Black N, Bain C. Choosing between randomised and non-randomised studies: a systematic review. Health Technol Assess 1998; 2(13): 1-124. [8] Leichsenring F. Randomized controlled versus naturalistic studies: a new research agenda. Bull Menninger Clin 2004; 68(2): 137-51. [9] Jadad AR, Enkin MW. Randomized controlled trials: Questions, answers, and musings. Malden MA: Blackwell Publishing; 2007. [10] Rawlins M. De testimonio: on the evidence for decisions about the use of therapeutic interventions. Lancet 2008; 372(9656): 2152-61. [11] Kienle GS. Gibt es Gründe für pluralistische Evaluationsmodelle? Limitierungen der randomisierten klinischen Studie. Z aerztl Fortb Qual sich 2005; 99(4-5): 289-94. [12] Guyatt GH, Oxman AD, Schunemann HJ, Tugwell P, Knottnerus A. GRADE guidelines: a new series of articles in the Journal of Clinical Epidemiology. J Clin Epidemiol 2011; 64(4): 380-2. [13] Guyatt G, Oxman AD, Akl EA, Kunz R, Vist G, Brozek J et al. GRADE guidelines: 1. Introduction-GRADE evidence profiles and summary of findings tables. J Clin Epidemiol 2011; 64(4): 383-94. - 6 -