Vincent Bontrop
banner
bontrop.com
Vincent Bontrop
@bontrop.com
Faciliteren, accelereren & excelleren! | πάντα ῥεῖ | Psychologist | (coi) Ambtelijk secretaris METC NedMec | Views expressed are my own

www.bontrop.com

#GCP #CTR #MDR #IVDR #GDPR #ClinicalTrials #ClinicalResearch
EU Nieuws: The EUDAMED four first modules will be mandatory to use as from 28 May 2026
The EUDAMED four first modules will be mandatory to use as from 28 May 2026
The EUDAMED four first modules will be mandatory to use as from 28 May 2026
health.ec.europa.eu
November 27, 2025 at 10:10 AM
EU Nieuws: Commission authorises first treatment for serious chronic lung disease
Commission authorises first treatment for serious chronic lung disease
Commission authorises first treatment for serious chronic lung disease
health.ec.europa.eu
November 18, 2025 at 1:25 PM
Nieuws van ACT EU: Improved scientific advice for medicines for public health threats including antimicrobial resistance
Improved scientific advice for medicines for public health threats including antimicrobial resistance
New approach brings together clinical trial and ethics expertise to accelerate development of medicines for use ahead of or during emergencies
www.ema.europa.eu
November 18, 2025 at 10:26 AM
EU Nieuws: Agenda - Biosimilar medicines: Multistakeholder Event (4 December 2025)
Biosimilar medicines - Multistakeholder Event
Biosimilar medicines - Multistakeholder Event
health.ec.europa.eu
November 17, 2025 at 4:10 PM
Nieuws van ACT EU: Improved scientific advice for medicines for public health threats including antimicrobial resistance
Improved scientific advice for medicines for public health threats including antimicrobial resistance
New approach brings together clinical trial and ethics expertise to accelerate development of medicines for use ahead of or during emergencies
accelerating-clinical-trials.europa.eu
November 17, 2025 at 2:31 PM
Nieuws van ACT EU: Annex I - Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2.1
Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS)
Official address Domenico Scarlattilaan 6 ● 1083 HS Amsterdam ● The Netherlands Address for visits and deliveries Refer to www.ema.europa.eu/how-to-find-us Send us a question Go to www.ema.europa.eu/contact Telephone +31 (0)88 781 6000 An agency of the European Union © European Medicines Agency, 2024. Reproduction is authorised provided the source is acknowledged. Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Table I – Published CTIS Structured Data and relevant disclosure timelines ..........................................................2 Table II – CTIS Documents ‘for publication’ and relevant disclosure timelines ......................................................3 Table III – Documents ‘for publication’: templates and personal data usually included .........................................4 Table IV – Publication rules of historical trials and of non-authorised applications ...............................................5 Table V – Trial categories and definition of trial phases subject to each category ..................................................6 Table VI – CTIS Structured Data and documents that are not subject to publication .............................................8 Table VII – Acronyms ........................................................................................................................................... 14 Table VIII - Definitions ........................................................................................................................................ 16 Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 2/20 Table I – Published CTIS Structured Data and relevant disclosure timelines The detailed list of structured data that are, or are not subject to publication is specified in documents on CTIS application fields and Notifications and Results. Disclosure timelines are provided in this table, and modalities in table IV. To know the type of trials belonging to each Category, refer to table V. An indicative list of structured data that is not subject to publication is provided in table VI. 1 Any contact detail provided (e.g., scientific and public sponsor contact point, PI) is expected to be a functional or professional contact detail (not personal). 2 The following fields are disclosed at time of decision for Category 1 trials conducted solely on adult population: public title (= title in lay terms), trial identifiers in registers, protocol code, phase, medical condition, rare disease, therap. Area, Population age, gender, Sponsor details, Details of clinical investigator sites in MSC(s) Structured data fields Category 1 Category 2 integrated ph1&2 Category 2 & 3 (excl. integr. ph1&2) Paediatrics and/or PIP Adults CTIS Application fields1, excluding the ones specified in the row below and in table III First MSC decision First MSC decision 2 First MSC decision First MSC decision 30 months after EU/EEA End of Trial CTIS Application fields: maximum duration of treatment, maximum daily dose allowed, daily dose unit of measure, maximum total dose allowed, Total dose unit of measure 30 months after EU/EEA End of Trial MSC(s) conclusions and decision outcomes That MSC decision Notifications on trial status and recruitment As soon as submitted by sponsor Notifications on serious breaches, urgent safety measures, unexpected events After MSC assessment 30 months after EU/EEA End of Trial & MSC assessment After MSC assessment Corrective measures (suspension, revocation, modification request) When applied by MSC(s) Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 3/20 Table II – CTIS Documents ‘for publication’ and relevant disclosure timelines The detailed list of documents that are, or are not subject to publication is specified in documents on CTIS application fields and Notification and Results. Disclosure timelines are provided in this table, and modalities in table IV (including exceptions applicable to ‘historical trials’). Caution: any document inadvertently uploaded ‘for publication’ into the below document upload sections will be published. Example: if an IB is uploaded into the SmPC section of a Cat 2 or 3 trial, the IB will be made public if not corrected by the sponsor before the decision on the application. Documents3 to be submitted in two versions ‘for publication’ and ‘not for publication Publication timelines4 Category 1 Category 2 and 3 including integrated ph1&2 Paediatrics and/or PIP Adults Protocol, including patients facing documents5 Upon results’ submission 30 months after EU/EEA EoT First MSC decision Protocol synopsis SmPC, if available Never Recruitment arrangements, including procedures for inclusion and copy of advertising material6 That MSC decision Subject information and informed consent form Lay person summary of results As soon as submitted 30 months after EU/EEA EoT As soon as submitted Final summary of results7 Clinical study report, if available8 As soon as submitted 3 Table III lists the type of personal data generally contained in documents ‘for publication’, while for an indicative list of documents that are not published: see table VI 4 To know the type of trials belonging to each Category, refer to table V 5 Protocol: this is referred to any kind of protocol (including master protocol, sub-protocol, etc.); patients facing documents: see definition in Clinical Trial Regulation 536/2014 Q&A 6 Recruitment arrangements: provide a clear indication of what the first act of recruitment is; advertising material: this includes any printed materials and audio or visual recordings 7 Interim results are not made publicly available, only final summary of results are: those documents are distinguished through a dropdown menu of the relevant placeholder 8 It is the Marketing Authorisation Applicants/Holders that provide Clinical Study Reports (CSRs), within 30 days from the issuing of marketing authorisation, or variation/line extension Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 4/20 Table III – Documents ‘for publication’: templates and personal data usually included In this table there is a suggested list of personal data that may be contained in the document version ‘not for publication’ and shall be removed the relevant ‘for publication’ version (with exception of PI details). For each document, any available template as per current guidelines is also linked, so that users can know which information is required to be included in the documents, avoiding unnecessary inclusion of CCI. Documents to be submitted in two versions ‘for publication’ and ‘not for publication Personal data9 to be anonymised in the doc version ‘for publication’ Websites on the standard templates Protocol, including patients facing documents Personal details of sponsor staff, including signatures https://www.ema.europa.eu/en/ich-m11-guideline-clinical-study- protocol-template-and-technical-specifications-scientific-guideline Protocol synopsis SmPC, if available Not expected https://www.ema.europa.eu/en/human-regulatory- overview/marketing-authorisation/product-information- requirements/product-information-templates-human and, for Nationally Authorised Products: https://www.hma.eu/human-medicines/cmdh/templates/qrd.html Recruitment arrangements, including procedures for inclusion and copy of advertising material Name, surname or identifying element of PI (to be disclosed10) or of other individual(s) including trial site personnel https://health.ec.europa.eu/medicinal-products/eudralex/eudralex- volume-10_en#set-of-documents-applicable-to-clinical-trials- authorised-under-regulation-eu-no-5362014 Subject information and informed consent form Lay person summary of results Not expected https://health.ec.europa.eu/document/download/8a42b8f5-4ec3- 4667-969c-3dd89ea8b270_en?filename=glsp_en.pdf Final summary of results Personal details of sponsor staff, including signatures Pseudonymised data of trial participants Clinical study report, if available https://database.ich.org/sites/default/files/E3_Guideline.pdf 9 Personal data should be provided in the document version ‘not for publication’ if reduced to a minimum: only when required and necessary to facilitate collaboration within the parties [Article 81(6) referring to 81(2) of the CTR]. Personal data of the author of a document appearing in the file properties should be removed from any file before being uploaded on CTIS. 10 Name, surname and professional contact details of the Principal Investigator should be included in version ‘not for publication’ as well as in version ‘for publication’ (see revised rules) Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 5/20 Table IV – Publication rules of historical trials and of non-authorised applications This table provides an overview of the modality of disclosure for non-authorised applications, for historical trials applications as well as for all applications submitted to CTIS as of 18 June 2024 (non-historical trials). The ‘historical’ trials are trials submitted to CTIS before the 18 June 2024, date of applicability of the revised transparency rules. Note that only the most recent authorised11 application of any trial, as well as any ‘not authorised’ initial application, is made publicly available. Application type Authorisation outcome Structured data (most recent version) Documents (most recent version) submitted as part of the application Applications published as of 18 June 2024, of trials submitted before 18 June 2024 (historical trials): IN, SM, NSM, AMSC Authorised11 Published as per Table I Not published SM, AMSC Not authorised Not published Not published Applications submitted as of 18 June, on trials submitted before 18 June 2024 (historical trials): SM, NSM12 Authorised11 Published as per Table I Published as per Table II13 AMSC Authorised Published as per Table I Part I documents: not published14 Part II documents: published as per Table II SM, AMSC Not authorised Not published Not published Trials submitted as of 18 June 2024 (non-historical trials): IN, SM, NSM, AMSC Authorised11 Published as per Table I12 Published as per Table II IN Not authorised Published as per Table I Published as per Table II SM, AMSC Not authorised11 Not published Not published 11 In case of NSM: ‘N/A’ 12 Note that between 18 June 2024 and 6 November 2025, the submissions of NSMs on historical trials did not trigger the publication of part I documents. 13 Due to technical reasons, structured data fields are updated depending on which part of the application the specific value is publicly available: for example, fields updated through SM/NSM part I, that affect fields disclosed as part II (e.g. number of subjects), will not be updated, and can only be updated if a part II update of those fields occurs 14 No part I documents are published in case of AMSC occurring on a historical trial. If the AMSC follows the SM, documents which were published with the SM will remain as they are Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 6/20 Table V – Trial categories and definition of trial phases subject to each category CTIS disclosure timelines depend mainly on the trial category. In case of category 1 trials, trial population age is also affecting those timelines. In addition, in case of integrated phase 1 and 2 trials, exceptions apply: see table I. Category Definition of category Acceptable values in application dossier data field “Trial phase” Comment Category 1 Pharmaceutical development clinical trials: • Phase I clinical trial in healthy volunteers or patients • Phase 0 trial - in healthy volunteers or patients, without therapeutic or prophylactic intent • Bioequivalence and bioavailability trials • Similarity trials for biosimilar product including those conducted in patients where efficacy endpoints are used to determine biosimilarity, where pharmacokinetic and or pharmacodynamic studies are not possible • Equivalence trial for combination products or topical products where a pharmacodynamic or efficacy endpoint is used to determine equivalence, and where pharmacokinetic and or pharmacodynamic studies are not possible. Human Pharmacology (Phase I) - First administration to humans Human Pharmacology (Phase I) - Bioequivalence Study Human Pharmacology (Phase I) - Other Category not permitted for clinical trial in emergency situations acc. to article 35 of EU CTR Category not permitted for integrated phase I and phase II trials Only for similarity trials and equivalence trials: the phase selected in the CTIS application could be different than phase I Category 2 Therapeutic exploratory and confirmatory clinical trials • Phase I and phase II integrated clinical trial • Phase II clinical trial • Phase III clinical trial Phase I and Phase II (Integrated)- First administration to humans Phase I and Phase II (Integrated)- Bioequivalence Study Phase I and Phase II (Integrated)- Other Category includes safety and efficacy trials in patients, or target populations for prophylaxis, i.e. carried out for treatment, diagnosis or prevention in the subjects included in the clinical trial during clinical development of a new product or during exploration of new indications, pharmaceutical forms, Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 7/20 Category Definition of category Acceptable values in application dossier data field “Trial phase” Comment strengths, and routes of administration for an existing product that already has a marketing authorisation When a protocol sets out a multiphase or adaptive design that falls in both category 1 and 2, the trial should be treated according to category 2. Category not permitted for integrated phase III and Phase IV trials. Category 3 Therapeutic use clinical trials • Phase III and phase IV integrated clinical trial • Phase IV clinical trial • Low intervention clinical trial Therapeutic use (Phase IV) Phase III and Phase IV (Integrated) Category for clinical trial carried out for treatment, diagnosis or prevention in the subjects included in the clinical trial, using an authorised IMP, used in accordance with the terms of the marketing authorisation, or the use of the IMPs is evidence-based and supported by published scientific evidence on the safety and efficacy of those IMPs in any of the Member States concerned Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 8/20 Table VI – CTIS Structured Data and documents that are not subject to publication The detailed list of structured data and documents that are, or are not subject to publication is specified in documents on CTIS application fields and Notifications and Results. The below is an indicative list that allows to identify easily CTIS data and documents submitted by Sponsors and Authority(ies) (including MSC - National Competent Authorities & Ethics Committees - and European Commission) that are not subject to publication and that might contain commercially confidential information and personal data. Personal data should be provided in CTIS only when required and necessary to facilitate collaboration within the parties [Article 81(6) referring to 81(2) of the Clinical Trials Regulation], further details are provided in the Q&A. Personal data of the author of a document appearing in the file properties should be removed from any file before being uploaded in CTIS. Location Data/document type Categories of personal data captured in CTIS15 Structured data submitted by the sponsor CTIS Application Sponsor Contact point for Union Sponsor Legal representative, if applicable Names and surnames of individual(s) Justification for no IMPD/AMPD upload, if applicable Non substantial modification description, if applicable Substantial modification details (information, reason, scope), if applicable Justification of application withdrawal, if applicable Not expected , Assessment and other sections Request for information (RFI) responses structured data Sponsor opinion requested as part of intended corrective measure, if applicable Sponsor response(s) to request of ad hoc assessment(s), if applicable Third country inspection details, if applicable Not expected Documents submitted by the sponsor CTIS Application Form Cover letter Not expected Proof of payment (per MSC) Not expected. However, where required by specific 15 Personal data should be provided in the document version ‘not for publication’ if reduced to a minimum: only when required and necessary to facilitate collaboration within the parties [Article 81(6) referring to 81(2) of the CTR]. Personal data of the author of a document appearing in the file properties should be removed from any file before being uploaded on CTIS Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 9/20 Location Data/document type Categories of personal data captured in CTIS15 Member States (see Q&A): name, surname and signature of individual(s) Statement of compliance with GDPR EU Regulation 2016/679 (see template) Name and surname of individual(s) issuing the statement Documents provided only in case of Substantial Modification (modification description, supporting information documentation), if applicable Document with supporting information on withdrawal, if applicable Not expected CTIS Application Part I Justification for low interventional trial, if applicable Not expected Data Safety Monitoring Committee Charter Name and surname of members of the Data Safety Monitoring Committee, see Q&A Study design Not expected Summary of scientific advice Summary of scientific advice – quality Not expected Paediatric Investigational Plan (PIP) opinion/decision Not expected. It may exceptionally include pseudonymised data of trial participants Written agreement from the sponsor - of any previous submitted applications that are associated with this clinical trial Name and surname of individual(s) Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 10/20 Location Data/document type Categories of personal data captured in CTIS15 Medicinal product documents for test/comparator/auxiliary/placebo, as applicable: • Investigator brochure (IB) • (GMP) Authorisation of manufacturing and import Not expected. The IB may exceptionally include pseudonymised data of trial participants • Certification by the qualified person (QP) In the Union that the manufacturing complies with Good Manufacturing Practice (GMP) Name, surname and signature of the qualified person (QP) • Quality (IMPD-Q) - Full and simplified Not expected • Safety and Efficacy (IMPD-S&E) - Full and simplified Not expected. It may exceptionally include pseudonymised data of trial participants • Auxiliary medicinal product dossier (AMPD) • Placebo medicinal product dossier - quality (IMPD-Q) Not expected Content of the labelling of the investigational medicinal products Not expected CTIS Application Part II Suitability of the investigator: Principal Investigator (PI) Curriculum Vitae (CV) Suitability of the investigator other than the investigator's CV Suitability of the facilities: suitability of the clinical trial site facilities Name and surname of the principal investigator and of any other personnel. Where required by specific Member States only, other signatures are to be provided: see Q&A. Name and surname of the head of the clinic/institution or of another responsible person issuing the document on site suitability, including relevant signature(s). Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 11/20 Location Data/document type Categories of personal data captured in CTIS15 Proof of insurance cover or indemnification Name, surname and potential signatures already present on document, if required in specific member state(s): see Q&A Financial and other arrangements Compliance with national requirements on data protection Compliance with use of biological sample Not expected Assessment and other sections Documents submitted with responses to the RFI (for validation, part I/part II, as applicable): list of changes to the application, General documentation, Quality related documentation, Documents related to the response, as applicable Sponsor opinion requested document(s) as part of intended corrective measure, if applicable Documents to support response(s) to request of ad hoc assessment(s), if applicable Name and surname of relevant individual(s) (e.g. principal investigator, head of the clinic/institution or other responsible person issuing the site suitability declaration, sponsor legal representative, Qualified Person), as applicable depending on the document type. Notifications documents submitted during trial duration Documents submitted as part of notifications uploaded for trial start, end of trial, early termination, temporary halt, restart trial, start recruitment, end recruitment, restart recruitment, as applicable Notification supporting documentation, supporting information uploaded for unexpected event(s), serious breach(es), urgent safety measure(s), as applicable Third country inspectorate inspection documents (inspection report and related document(s)), if applicable Not expected. They may exceptionally include pseudonymised data of trial participants, as applicable. Names, surnames, signatures of third countries inspectors, personal data of sponsor staff, trial site personnel or pseudonymised data of trial participants. Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 12/20 Location Data/document type Categories of personal data captured in CTIS15 Trial results Intermediate data analysis summary of results document(s), if applicable Personal data of sponsor staff, signatures and pseudonymised data of trial participants. Structured data submitted by the Member State(s) Concerned Assessment and other sections RFIs Validation conclusion details Assessment decision conditions (if any) Ad hoc assessment details, if applicable Corrective measure consultation with MSCs, sponsor opinion request, as applicable Annual Safety Report RFI, if applicable Not expected Documents submitted by the Member State(s) Concerned Assessment and other sections Documents to support requests for information (RFI) to sponsor during evaluation of an application (validation, part I/part II assessment), on any element of the dossier, including quality, as applicable Draft assessment reports (part I and/or part II as applicable) Final assessment reports (part I and/or part II as applicable) Authorisation supporting documentation (decision document), if applicable Revert decision supporting documentation, if applicable Part I disagreement justification Corrective measure supporting documents: justification document(s), sponsor opinion, investigator opinion, consultation with MSCs related document(s), sponsor opinion request(s) related document(s), as applicable Ad hoc assessment related documents: supporting document(s), document(s) added to RFI, summary of assessment and outcome documentation, as applicable Inspection report, if applicable Assessment reports for serious breaches, urgent safety measures, unexpected events, if applicable Union control plans/programmes/reports, as applicable Not expected. However, they could include name and surname of relevant individual(s) (e.g. principal investigator, head of the clinic/institution or other responsible person issuing the site suitability declaration, Qualified Person), as applicable depending on the document type. The inspection report may include names, surnames, signatures of EU/EEA inspectors, personal data of PI, head of the institution, sponsor staff/site personnel Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 13/20 Location Data/document type Categories of personal data captured in CTIS15 or pseudonymised data of trial participants. In the assessment report(s) for serious breaches, urgent safety measures and unexpected events, personal data are not expected, however they may exceptionally include pseudonymised data of trial participants Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 14/20 Table VII – Acronyms In this Guidance document and Annex the following acronyms apply: Acronym Description AMSC Additional Member State Concerned Application ASR Annual Safety Report CCI Commercially Confidential Information CTIS Clinical Trials Information System CTR Clinical Trials Regulation or Regulation (EU) No 536/2014 of the European Parliament and of The Council of 16 April 2014 on clinical trials on medicinal products for human use, and repealing Directive 2001/20/EC. EC European Commission EEA European Economic Area EMA European Medicines Agency, also referred to hereafter as the Agency EoT End of Trial date EU European Union EUDPR Regulation (EU) 2018/1725 of the European Parliament and of the Council of 23 October 2018 on the protection of natural persons with regard to the processing of personal data by the Union institutions, bodies, offices, and agencies and on the free movement of such data, and repealing Regulation (EC) No 45/2001 and Decision No 1247/2002/EC (European Data Protection Regulation) EUPD European Union Portal and Database GCP Good Clinical Practice GDPR Regulation (EU) 2016/679 of the European Parliament and of the Council of 27 April 2016 on the protection of natural persons with regard to the processing of personal data and on the free movement of such data, and repealing Directive 95/46/EC (General Data Protection Regulation) Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 15/20 Acronym Description IAM Identity Access Management IMPD Investigational Medicinal Product Dossier MAA Marketing Authorisation Applicant MAH Marketing Authorisation Holder MSs Member States MSC Member State Concerned NCA National Competent Authority NSM Non-Substantial Modification Application OMS Organisation Management Service PI Principal Investigator RFI Request for information SM Substantial Modification Application XEVMPD Extended EudraVigilance Medicinal Product Dictionary Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 16/20 Table VIII - Definitions In this Guidance document and Annex the following definitions apply: Definition Description Aggregated data Data about several individuals that have been combined/grouped to present general trends or values without identifying (either directly or indirectly) individuals within the data generated for statistical or research purposes. Anonymisation The process of rendering personal data anonymous as described in recital 16 of the EUDPR and recital 26 of the GDPR i.e., namely information which does not relate to an identified or identifiable natural person or to personal data rendered anonymous in such a manner that the data subject is not or no longer identifiable. Anonymous data (also called as anonymised) Information which does not relate to an identified or identifiable natural person or personal data rendered anonymous in such a manner that the data subject is not, or no longer, identifiable. Article 29 Data Protection Working Party (Art. 29 WP) The ‘Article 29 Working Party’ is the short name of the Article 29 Data Protection Working Party established by Article 29 of Directive 95/46/EC. It provided the European Commission with independent advice on data protection matters and helped in the development of a harmonised implementation of data protection rules in the EU Member States. As of 25 May 2018, the Article 29 Working Party ceased to exist, and has been replaced by the European Data Protection Board (EDPB). Clinical trial information submitted to CTIS Data (captured in structured data fields) and documents submitted to CTIS in the context of a clinical trial application, during the evaluation of an application and during the clinical trial life cycle including the supervision of the clinical trial and the clinical trials results. Commercially Confidential Information (CCI) Any information submitted to CTIS which is not in the public domain, or publicly available, and where disclosure may undermine the legitimate economic interest or competitive position of the concerned entities, e.g. clinical trial sponsors, marketing authorisation applicants/holders or service providers.16 Data ‘Data’ means any digital representation of acts, facts or information and any compilation of such acts, facts, or information, including in the form of sound, visual or audio-visual recording (Article 2 of Data Act). 16 HMA/EMA recommendations on transparency approved in November 2010 - Recommendations on release of information with regard to new applications for medicinal products before and after opinion or decision on granting of a marketing authorisation (EMA/484118/2010). CCI potentially claimed by service providers is unlikely to appear in clinical trial information submitted in CTIS and will be subject to a particular degree of rigor when scrutinizing its existence. Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 17/20 Definition Description Database An organized collection of data stored as multiple datasets. Dataset A dataset is a structured collection of data. A table where each column represents a particular variable, and each row corresponds to a different record is an example of a dataset17. Data controller (or controller) ‘‘Controller’ means the natural or legal person, public authority, agency, or other body which, alone or jointly with others, determines the purposes and means of the processing of personal data; where the purposes and means of such processing are determined by Union or Member State law, the controller or the specific criteria for its nomination may be provided for by Union or Member State law.’ (Article 4(7) of Regulation (EU) 2016/679). or, as applicable to the entity in question ‘‘Controller’ means the Union institution or body or the directorate-general or any other organisational entity which, alone or jointly with others, determines the purposes and means of the processing of personal data; where the purposes and means of such processing are determined by a specific Union act, the controller or the specific criteria for its nomination can be provided for by Union law.’ (Article 3(8) of Regulation (EU) 2018/1725). Data minimisation principle ‘Personal data shall be adequate, relevant and limited to what is necessary in relation to the purposes for which they are processed.’ (Article 5(1)(c) of Regulation (EU) 2016/679 and Article 4(1)(c) of Regulation (EU) 2018/1725). Data processor (or processor) ‘‘Data processor’ means a natural or legal person, public authority, agency, or other body which processes personal data on behalf of the controller.’ (Article 4(8) of Regulation (EU) 2016/679 and Article 3(12) of Regulation (EU) 2018/1725). Data protection principles Regulation (EU) 2016/679 and Regulation (EU) 2018/1725 prescribe adherence to 7 data protection principles, i.e.: • Lawfulness, fairness, and transparency • Purpose limitation • Data minimisation • Accuracy • Storage limitation • Integrity and confidentiality (security) 17 See AEPD-EDPS joint paper on 10 misunderstandings related to anonymisation, https://edps.europa.eu/data-protection/our-work/publications/papers/aepd-edps-joint-paper-10- misunderstandings-related_en Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 18/20 Definition Description • Accountability Data subject ‘An identified or identifiable natural person to whom personal data relates. An identifiable natural person is one who can be identified, directly or indirectly, in particular by reference to an identifier such as a name, an identification number, location data, an online identifier or to one or more factors specific to the physical, physiological, genetic, mental, economic, cultural, or social identity of that natural person.’ (Article 4(1) of Regulation (EU) 2016/679 and Article 3(1) of Regulation (EU) 2018/1725). Data subjects applicable to CTR and CTIS are trial participants, CTIS users, principal investigators, sponsor staff, etc. Disclosure The act of making data available to one or more third parties. EU Clinical Trials Information System (CTIS) The IT platform, including the EU portal and EU database, that allows the exchange of clinical trials information in the European Union. CTIS interacts with other databases such as IAM (Identity Access Management), XEVMPD (Extended EudraVigilance Medicinal Product Dictionary) and OMS (Organisation Management Service) which are also managed by the Agency. EU Clinical Trials Information System (CTIS) user The natural person(s) being granted access to the secure domains of CTIS, that submitted the clinical trial information to CTIS in the context of a clinical trial application, or that has access to the system during the evaluation of an application, or during the clinical trial life cycle including supervision of the clinical trial. EU Portal and Database (EUPD) In accordance with Articles 80 and 81, and Recitals 66 and 67 of the Clinical Trials Regulation, the Agency has the obligation, in collaboration with the Member States and the Commission, to set up and maintain both a Clinical Trials Portal, as a single-entry point for the submission of data and information relating to clinical trials, and a Clinical Trials Database containing data and information submitted in accordance with that Regulation. Joint Controller ‘Where two or more controllers jointly determine the purposes and means of processing, they shall be joint controllers. They shall in a transparent manner determine their respective responsibilities for compliance with the obligations under this Regulation, in particular as regards the exercising of the rights of the data subject and their respective duties to provide the information referred to in Articles 13 and 14, by means of an arrangement between them unless, and in so far as, the respective responsibilities of the controllers are determined by Union or Member State law to which the controllers are subject. The arrangement may designate a contact point for data subjects.’ (Article 26(1) of Regulation (EU) 2016/679) Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 19/20 Definition Description or, as applicable to the entity in question ‘Where two or more controllers or one or more controllers together with one or more controllers other than Union institutions and bodies jointly determine the purposes and means of processing, they shall be joint controllers. They shall in a transparent manner determine their respective responsibilities for compliance with their data protection obligations, in particular as regards the exercising of the rights of the data subject and their respective duties to provide the information referred to in Articles 15 and 16, by means of an arrangement between them unless, and in so far as, the respective responsibilities of the joint controllers are determined by Union or Member State law to which the joint controllers are subject. The arrangement may designate a contact point for data subjects.’ (Article 28(1) of Regulation (EU) 2018/1725). Personal data ‘‘Personal data’ means any information relating to an identified or identifiable natural person (‘data subject’); an identifiable natural person is one who can be identified, directly or indirectly, in particular by reference to an identifier such as a name, an identification number, location data, an online identifier or to one or more factors specific to the physical, physiological, genetic, mental, economic, cultural or social identity of that natural person’. (Article 4(1) of Regulation (EU) 2016/679 and Article 3(1) of Regulation (EU) 2018/1725). Special categories of personal data Personal data revealing racial or ethnic origin, political opinions, religious or philosophical beliefs, or trade union membership, and the processing of genetic data, biometric data for the purpose of uniquely identifying a natural person, data concerning health or data concerning a natural person's sex life or sexual orientation. (Article 9(1) of Regulation (EU) 2016/679 and Article 10(1) of Regulation (EU) 2018/1725). Personal data breach ‘'Personal data breach’ means a breach of security leading to the accidental or unlawful destruction, loss, alteration, unauthorised disclosure of, or access to, personal data transmitted, stored, or otherwise processed’. (Article 4(12) of Regulation (EU) 2016/679 and Article 3(16) of Regulation (EU) 2018/1725). Process, processes, processing ‘'Processing’ means any operation or set of operations which is performed on personal data or on sets of personal data, whether or not by automated means, such as collection, recording, organisation, structuring, storage, adaptation or alteration, retrieval, consultation, use, disclosure by transmission, dissemination or otherwise making available, alignment or combination, restriction, erasure, or destruction’. (Article 4(2) of Regulation (EU) 2016/679 and Article 3(3) of Regulation (EU) 2018/1725). Pseudonymised, pseudonymisation ‘'Pseudonymisation’ means the processing of personal data in such a manner that the personal data can no longer be attributed to a specific data subject without the use of additional information, provided that such additional Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) EMA/194159/2023 Page 20/20 Definition Description information is kept separately and is subject to technical and organisational measures to ensure that the personal data are not attributed to an identified or identifiable natural person’. (Article 4(5) of Regulation (EU) 2016/679 and Article 3(6) of Regulation (EU) 2018/1725). Publishing The act of making data publicly available. Redaction Masking of text in a document by applying a permanent and unremovable overlay, rendering the text unreadable. Re-identification The process of analysing data, or combining it with other data, with the result that individuals become identifiable. Re-identification risk (or re- identification likelihood, risk of re-identification) The re-identification risk (or likelihood) is the probability in a given dataset of re-identifying an individual, by turning anonymised data back into personal data through the use of data matching or similar techniques.18 Sponsor The term ‘Clinical Trial Sponsors’ in the tables of the present Annex applies to sponsors or entities working on behalf of the sponsors, like Clinical Research Organisations (CROs). Study subject, trial participant ‘An individual who participates in a clinical trial, either as a recipient of an investigational medicinal product or as a control’. Article 2(17) of Regulation (EU) No 536/2014. Use is made in the guidance of the term ‘trial participant’ as an equivalent to ‘trial subject/study subject’. Version of the document ‘for publication’ The version of the document provided in CTIS by the users which should not contain commercial confidential information (CCI) and personal data19. It is the responsibility of the user to ensure that this version does not contain such information. Version of the document ‘not for publication’ The version of the document provided in CTIS by the users which may contain personal data insofar that this is necessary for the purposes listed in Article 81(2) of the Clinical Trials Regulation and/or commercial confidential information (CCI). 18 See AEPD-EDPS joint paper on 10 misunderstandings related to anonymisation, https://edps.europa.eu/data-protection/our-work/publications/papers/aepd-edps-joint-paper-10- misunderstandings-related_en. 19 With the exceptions defined by the present guidance
accelerating-clinical-trials.europa.eu
November 7, 2025 at 8:54 AM
Nieuws van ACT EU: Revised CTIS transparency rules & historical trials: quick guide for users
Revised CTIS transparency rules and historical trials: quick guide for users
An agency of the European Union CTIS team, Data Analytics and Methods Taskforce (TDA) Classified as public by the European Medicines Agency Table of contents 1. Implementation of revised CTIS transparency rules 2. Revised rules: modality of disclosure of CTIS trials’ information 3. Publication of structured data 4. Publication of documents 5. Historical trials, submitted before 18 June 2024 6. Reference documentation Article 81(4) of Regulation (EU) No. 536/2014: legal basis for the establishment a publicly accessible EU clinical trials database, while protecting commercially confidential information (CCI), personal data (PD) and confidential information on the assessment conducted by MSs Classified as public by the European Medicines Agency Implementation of revised CTIS transparency rules Classified as public by the European Medicines Agency Implementation of revised CTIS transparency rules As of 18 June 2024, the Revised CTIS transparency rules are the publication rules of the Clinical Trial Information System: • All trials submitted on or after 18 June follow the principles and timelines defined in the revised rules • Trials submitted to CTIS before 18 June are considered ‘historical’ and have only their structured data published, see ‘historical trials’ section Rules are summarised in the Annex 1 to the Guidance document on how to approach the protection of personal data and CCI while using the CTIS More info is in the Q&A on the protection of CCI and Personal Data, in a dedicated CTIS bitesize talk and in the List of CTIS application fields and documents + notifications fields Classified as public by the European Medicines Agency Revised rules: modality of disclosure of CTIS trials’ information Chapter 2 of Guidance document on how to approach the protection of personal data and commercially confidential information while using the CTIS Classified as public by the European Medicines Agency Revised rules: modality of disclosure of CTIS trials’ information Category Trial type Category 1 Pharmaceutical development clinical trials Phase I clinical trials in healthy volunteers or patients Phase 0 trial in healthy volunteers or patients Bioequivalence and bioavailability trials Similarity trials for biosimilars Equivalence trials for combination or topical products Category 2 Therapeutic exploratory & confirmatory clinical trials Phase I and phase II integrated clinical trials Phase II clinical trials Phase II and phase III integrated clinical trials Phase III clinical trials Category 3 Therapeutic use clinical trials Phase III and phase IV integrated clinical trials Phase IV clinical trial and low interventional trials The most recent authorised application of any trial, as well as any ‘not authorised’ initial application, is made publicly available as per timelines based on: • trial category, selected in the ‘Form’ section as per below table • population age • trial phase (in case of category 2 trials that are integrated phase 1&2) Classified as public by the European Medicines Agency Publication of structured data Table I of Annex 1 to Guidance document, List of CTIS application and notifications fields and documents Classified as public by the European Medicines Agency Structured data – what will be published & when Structured data Category 1 Category 2 integrated p h1&2 Category 2 & 3 (excl. integr. ph1&2) Paediatrics and/or PIP Adults CTIS application fields First MSC decision First MSC decision First MSC decision First MSC decision 30 months after EU/EEA End of Trial CTIS application fields on dose and treatment duration 30 months after EU/EEA End of Trial MSC(s) conclusions and decision outcomes That MSC decision Notifications on trial status and recruitment As soon as submitted by sponsor Notific. on serious breaches, urgent safety measures, unexpected events After MSC assessment 30 months after EU/EEA EoT & MSC assessment After MSC assessment Corrective measures (suspension, revocation, modification request) When applied by MSC(s) Classified as public by the European Medicines Agency Structured data – what will be published & when Structured data Category 1 Category 2 integrated ph1&2 Paediatrics and/or PIP Adults CTIS application fields populated by the sponsor, including: •Public title (= title in lay terms) •Trial identifiers in registers, protocol code •Phase, medical cond., rare disease, therap. area •Population age, gender •Sponsor details •Details of clinical investigator sites in MSC(s) First MSC decision First MSC decision First MSC decision Remaining CTA fields populated by the sponsor 30 months after EU/EEA End of Trial CTIS application fields on Maximum duration of treatment, Maximum daily dose allowed, Daily dose unit of measure, Maximum total dose allowed, Total dose unit of measure 30 months after EU/EEA End of Trial Classified as public by the European Medicines Agency Structured data – what will be published & when Structured data All categories Sponsor legal representative details Never Any request for information (RFI) and RFI responses Validation conclusion details, assessment decision conditions (if any) MSC(s) assessment(s) on notifications 3rd country inspection details Classified as public by the European Medicines Agency Publication of documents Table II of Annex 1 to Guidance document, List of CTIS application and notifications fields and documents Classified as public by the European Medicines Agency Documents – what will be published & when Category 1 Category 2 and 3 including integrated ph1&2Documents type Paediatrics and/or PIP Adults Protocol, synopsis, patients facing documents Upon results’ submission 30 months after EU/EEA End of Trial First MSC decision SmPC, if available Never Subject information and informed consent form That MSC decisionRecruitment arrangements, including procedures for inclusion and copy of advertising material Final summary of results, Lay person summary of results As soon as submitted 30 months after EU/EEA End of Trial As soon as submitted Clinical study report, if available As soon as submitted (requirement: 30 days from MA) All other documents, including any MS document Never 11 Classified as public by the European Medicines Agency For all those documents that are no longer published (e.g. IB), the system had to remove the former version ‘for publication’, unless no ‘not for publication’ version was linked to it As of 18 June 2024, for any application created before this date before 18/6/24 as of 18/6/24 workspace view Examples are in the next slide Note question 1.9 of Q&A: when submitting a modification application of those trials, sponsors are not requested to re-upload the clean versions of those documents ‘not for publication’ that are no longer available. Only the clean version and corresponding track-changes version of those documents that have been modified as part of the application should be uploaded, unless MSCs requests further uploads. 12 Classified as public by the European Medicines Agency Examples Before 18 June, sponsor had uploaded: As of 18 June, Sponsor/MS sees, in the same placeholder: In slot ‘for publication’ and linked to ‘not for publication’ Doc 1 ‘for publication’ - Doc 1 (formerly ‘for publication’) Doc 1 ‘for publication’ Doc 1 not for publication Doc 1 (formerly ‘not for publication’) Doc 1 ‘for publication’ 2 or more docs ‘not for publication’, linked to the same unique document for pub All ‘not for publication’ docs (doc ‘for publication’ removed) Doc 1 For publication Doc 2 For publication (e.g. translation) Doc 1 ‘not for publication’ Doc 2 ‘not for publication’ (e.g. translation) Doc 1 (formerly ‘not for publication’) Doc 2 (formerly ‘not for publication’ ,e.g. translation) Doc 1 For publication Doc 2 For publication Doc 1 ‘not for publication’ linked to doc 1 - Doc 1 (formerly ‘not for publication’) Doc 2 (formerly ‘for publication’) Doc 1 ‘for publication’ Doc 2 ‘for publication’ Doc 3 ‘for publication’ Doc 4 ‘for publication’ Doc 1 ‘not for publication’ Doc 2 ‘not for publication’ - - Doc 1 (formerly ‘not for publication’) Doc 2 (formerly ‘not for publication’) Doc 3 (formerly ‘for publication’) Doc 4 (formerly ‘for publication’) Doc 1 ‘for publication’ English Doc 2 ‘for publication’ German Doc 3 ‘not for publication’ Spanish Doc 4 ‘not for publication’ Italian Doc 1 (formerly ‘for publication’ English) Doc 2 (formerly ‘for publication’ German) Doc 3 (formerly ‘not for publication’ Spanish) Doc 4 (formerly ‘not for publication’ Italian) In some cases, users could still see both ‘for publication’ and ‘not for publication’ versions. Note that also in these cases the document is not published. 13 Classified as public by the European Medicines Agency Historical trials, submitted before 18 June 2024 Section 2.3 of Guidance document on how to approach the protection of personal data and CCI while using the CTIS + specific behaviour of SM change of sponsor Classified as public by the European Medicines Agency Historical trials: what is published and when For all those CTIS applications submitted* before 18 June 2024: • the structured data are published for all trials’ categories as per revised rules • documents are not published (this applies to all historical trials, regardless of the previous use of deferrals or publication status) The following kinds of CTIS applications submitted* on or after 18 June 2024 trigger publication of those documents that are in scope of the application and of the revised rules: • Substantial Modifications (part I and/or part II) • Non-Substantial Modification (part I and/or part II) • Additional Member State (triggering publication of part II docs only) Warning: ‘SM part I change of sponsor’ triggers publication of part I documents: see slide 17 *the date determining whether a trial is ‘historical’ or not, is the submission date, not the date of creation of your draft documents in scope of publication should be redacted accordingly Classified as public by the European Medicines Agency Historical trials: what is published and when AM applications done on ‘historical trials’ do not trigger the publication of part I documents. Apart from AM applications, for all the other kinds of applications the following applies: Example: an initial application submitted before 18 June 2024 has the latest version of its structured data published on 18 June 2024 while its documents were not published; if, on this trial: • A SM part 1 or a NSM part I is submitted, with the purpose of updating the IB: protocol and synopsis will be published as per revised rules and should be redacted accordingly. Note: part II documents will not be published • A SM part I change of sponsor is submitted: this will trigger publication of part I documents, see next slide • An SM Part II or a NSM part II is submitted: this triggers publication of part II documents only & update of all structured data fields. • Submission of trial notifications or results do not trigger publication of application’s documents of the same trial. Note: in order to remove ‘for publication’ versions to replace them with redacted versions, it is necessary to remove the relevant ‘non for publication’ versions, and upload them again once the redacted ‘for publication’ versions are uploaded  for an application that is submitted to CTIS on any ‘historical’ trial, the documents in scope of publication are going to be published and should be redacted accordingly Classified as public by the European Medicines Agency SM part I change of sponsor* triggers publication of part I docs For a trial submitted to CTIS before 18 June 2024: For questions: contact the Reporting Member State (RMS) 17 Do part I docs ‘for publication’ contain personal data/CCI? 1. Create NSM part I where the part I docs ‘for publication’ are without CCI, keeping the previous versions in the slot ‘not for publication’ 2. Submit NSM part I 1. Create SM part I change of sponsor & specify in the cover letter that the part I docs ‘for publication’ do not contain personal data/CCI 2. Submit SM part I change of sponsor Yes No * Specific SM that allows the change of the Sponsor’s ORG ID Classified as public by the European Medicines Agency Reference documentation ACT EU – Implementation of clinical trial regulation website Classified as public by the European Medicines Agency Reference documentation • Revised transparency rules • Quick guide for users • Guidance document on how to approach the protection of personal data and commercially confidential information (CCI) while using CTIS and its Annex I • Q&A on the protection of CCI and Personal Data while using CTIS • List of CTIS application fields and documents (with publication details) • List of CTIS notifications fields and documents (with publication details) • CTIS Bitesize talk on the transparency rules • Sponsor handbook 19
accelerating-clinical-trials.europa.eu
November 7, 2025 at 8:45 AM
Nieuws van ACT EU: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2.1
2. Rules of clinical trial information in CTIS pertaining to submission and publication .................................................................................................. 9
Official address Domenico Scarlattilaan 6 ● 1083 HS Amsterdam ● The Netherlands Address for visits and deliveries Refer to www.ema.europa.eu/how-to-find-us Send us a question Go to www.ema.europa.eu/contact Telephone +31 (0)88 781 6000 An agency of the European Union © European Medicines Agency, 2024. Reproduction is authorised provided the source is acknowledged. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2.1 This document provides guidance to users on the revised Clinical Trials Information System (CTIS) transparency rules and on the protection of personal data and commercially confidential information (CCI) submitted to CTIS, the EU database established in accordance with the requirements of Regulation (EU) No 536/2014 (CTR). It should be read in conjunction with its Annex I. A Questions and Answers (Q&A) document is also available to users, see Q&A on the protection of Commercially Confidential Information and Personal Data while using CTIS. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 2/28 Document version Publication date Changes introduced in the text Version 1.0 3 May 2023 N/A Version 1.1 10 July 2023 - new chapter 4 on management of commercially confidential information (CCI) in clinical trial information submitted to CTIS - new chapter 5 on GCP inspection reports Version 2 18 June 2024 - alignment with revised CTIS transparency rules, including removal of chapter 5 (no longer applicable) - new sections on the ‘historical trials’ publication principles and on transition trials Principles of protection of personal data and CCI remained unchanged compared to the former versions Version 2.1 7 November 2025 - chapter 2: updated rules for NSM submitted on historical trials and publication details of SM to change the sponsor Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 3/28 Table of contents 1. General information ................................................................................ 4 1.1. Introduction......................................................................................................... 4 1.2. Scope ................................................................................................................. 5 1.3. Legal framework .................................................................................................. 5 2. Rules of clinical trial information in CTIS pertaining to submission and publication .................................................................................................. 9 2.1. Introduction......................................................................................................... 9 2.2. Clinical trial information submitted to CTIS and disclosure rules ............................... 10 2.2.1. Submission of structured data ........................................................................... 11 2.2.2. Submission of documents for ‘for publication’ and ‘not for publication’ .................... 11 2.3. Publication rules of the so called ‘historical trials’ .................................................... 11 2.4. Transparency aspects of transition trials ................................................................ 13 3. Management of personal data in structured data fields and in documents submitted to CTIS...................................................................................... 14 3.1. Introduction....................................................................................................... 14 3.2. The principles of anonymisation ........................................................................... 15 3.3. Anonymisation of personal data in the document version ‘for publication’ .................. 16 3.3.1. Anonymisation of personal data other than those of trial participants in the document version ‘for publication’ .............................................................................................. 17 3.3.2. Anonymisation of personal data of trial participants in the document version ‘for publication’ .............................................................................................................. 18 3.4. Documents not subject to publication: the principles of minimisation and pseudonymisation of personal data ............................................................................. 19 4. Management of commercially confidential information (CCI) in clinical trial information submitted to CTIS ........................................................... 21 4.1. Introduction....................................................................................................... 21 4.2. Mechanisms available in CTIS to protect CCI.......................................................... 21 4.3. Redaction of CCI in the document version ‘for publication’ ....................................... 22 4.4. Relevant expertise and consistent decision-making process on the identification and redaction of CCI........................................................................................................ 23 4.5. Information that may be considered CCI ............................................................... 24 4.6. Information that may not be considered CCI .......................................................... 25 4.6.1. Information that is already in the public domain or publicly available ..................... 25 4.6.2. Information that does not bear any innovative elements ...................................... 26 4.6.3. Information that would not qualify as commercially confidential ............................ 26 Annex I: Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS)……………………………………………………………..………………..…….see Annex I Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 4/28 1. General information 1.1. Introduction The European Clinical Trials Regulation (EU) No 536/20141 (hereinafter ‘the Clinical Trials Regulation’ or ‘CTR’) repeals Directive 2001/20/EC on clinical trials2 (CTs) and establishes a harmonised approach to the submission, assessment, supervision, and reporting of clinical trials information with the implementation of consistent rules throughout the European Union (EU)/European Economic Area (EEA) Member States (MSs). The Clinical Trials Regulation aims to foster innovation through harmonised content of clinical trial applications submitted to Member States for assessment, to increase collaboration between the Member States on the assessment of clinical trial applications, and to increase transparency and availability of information on clinical trials and their results. Publicly available information foreseen by the CTR should contribute to protecting public health and fostering the innovation capacity of European medical research, while recognising the legitimate economic interests of sponsors and protecting personal data. In accordance with Recitals 66 and 67 and Articles 80 and 81 of the Clinical Trials Regulation, the Agency, in collaboration with the Member States and the European Commission (EC), has the obligation to set up and maintain an EU Portal as a single-entry point for the submission of data and documents relating to clinical trials, and an EU Database containing the data and documents submitted via the EU Portal. The EU Clinical Trials Portal and Database are jointly referred to as the EU Portal and Database (EUPD). To ensure transparency of clinical trials, the EU Database should be publicly accessible, and data should be presented in an easily searchable format. The EU database is a key instrument to ensure transparency of clinical trial information. The database serves as the source of public information on assessed clinical trial applications, from the time of decision until the submission of summary results. Access to this information is fundamental to enable trust in the clinical research conducted in the European Union. The EUPD and associated workspaces provide MSs, the European Commission, the Agency, sponsors, and applicants3 of a marketing authorisation with an effective network to streamline and facilitate the preparation of the flow of information for the authorisation and supervision of clinical trials in the EU/EEA. The EUPD enables the submission and storing of clinical trial information and is one of the two components of the Clinical Trials Information System (CTIS), also including the module for submission of the Annual Safety Reports (ASRs). Throughout the document overall reference is made to the use of CTIS. To streamline the use of the already available information stored in other databases managed by the Agency and to promote consistency and standardisation, CTIS consumes data from the following data sources: • Extended EudraVigilance Medicinal Product Dictionary (XEVMPD) • Organisation Management Service (OMS) 1 Regulation (EU) No 536/2014 of the European Parliament and of The Council of 16 April 2014 on clinical trials on medicinal products for human use, and repealing Directive 2001/20/EC. 2 Directive 2001/20/EC 3 Note that where this document refers to ‘sponsor users’ or ‘sponsor domain’: this may refer to, respectively as applicable, users acting on behalf of marketing authorisation applicants/holders and related user domain areas in the system. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 5/28 • Identity Access Management (IAM) The interface of CTIS with other EMA data sources is shown in the figure below: 1.2. Scope The CTR brings an unprecedented level of transparency in terms of publication of clinical trials information for trials conducted in the EU/EEA. Access to this information, including trial results, is important to allow prompt recruitment of patients at the site, avoid duplication of efforts and ultimately foster innovation and promote clinical research in the European Union and the European Economic Area. This guidance document aims to help CTIS users to navigate through the system functionalities and understand the main principles to be followed to enable protection of personal data and commercially confidential information while using CTIS and publishing clinical trials data and documents. The former version of this document described those system functionalities that were implemented under the previous transparency rules. Those functionalities implied the possibility for CTIS users to defer the publication of certain structured data and/or documents, for a specific timeframe depending on the trial’s development phase. The principles of protection of personal data and CCI that were described in the former versions of this document were not affected by this revision. The following chapters provide information on: • Description of CTIS structure and components, including a description of the functionalities and publication rules for clinical trials information submitted to CTIS (chapter 2). • Principles to be followed enabling protection of personal data as part of the clinical trial information submitted to CTIS (chapter 3). • Principles to be followed enabling protection of commercially confidential information (CCI) as part of the clinical trial information submitted to CTIS (chapter 4). 1.3. Legal framework The CTR sets out requirements for the protection of personal data, CCI and increased transparency of clinical trials in the EU/EEA. These requirements apply to information contained in the EU Database. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 6/28 Data and documents defined in the CTR are submitted via the EU Portal, stored in the EU Database and subject to the disclosure rules. Article 81(4) of the CTR states that the EU Database shall be publicly accessible unless, for all or parts of the data and information contained therein, confidentiality is justified on any of the following grounds: a) protecting personal data in accordance with Regulation (EU) 2018/17254; b) protecting commercially confidential information, in particular through taking into account the status of the marketing authorisation for the medicinal product, unless there is an overriding public interest in disclosure. c) protecting confidential communication between Member States in relation to the preparation of the assessment report. d) ensuring effective supervision of the conduct of a clinical trial by Member States. Recital 68 of the CTR states that: in general, the data included in a clinical study report should not be considered commercially confidential once a marketing authorisation has been granted, the procedure for granting the marketing authorisation has been completed, the application for marketing authorisation has been withdrawn. In addition, the main characteristics of a clinical trial, the conclusion on Part I of the assessment report for the authorisation of a clinical trial, the decision on the authorisation of a clinical trial, the substantial modification of a clinical trial, and the clinical trial results including reasons for temporary halt and early termination, in general, should not be considered confidential. Structured data fields and documents from the clinical trial application dossier can be made public after the decision on the clinical trial has been taken (Article 81(5) of the CTR), unless there is an overriding public interest for a particular clinical trial to do so earlier. This applies only in exceptional circumstances where the general public interest in having information made publicly available may outweigh considerations for the information to remain confidential (e.g. in case of declared pandemic, public health emergency). Thus, only applications on which a decision has been issued by a Member State concerned (MSC) will be made public. This applies to any decision outcome, i.e., authorisation, authorisation with condition(s) or whether the authorisation is refused. Information on initial applications which are only for assessment of Part I of the dossier (Article 11 applications) will not be made public until a Part II has been submitted to the MSC and a decision has been issued by at least one of the MSC(s). Applications which are not validated, that are lapsed, or those withdrawn by the sponsors before a decision is issued will not be made public. In addition, the following provisions related to the protection of personal data and CCI should be also considered as part of the guidance provided in this document. Data protection related provisions: Article 93 of the CTR expressly makes reference to EU data protection legislation i.e., to the now applicable General Data Protection Regulation (GDPR) with reference to the processing of personal data carried out in MSs (including processing by regulatory authorities and ethics committees) as well as sponsors, marketing authorisation applicants or holders and the European Data Protection Regulation 4 Article 81(4) of Regulation EU (No) 536/2014 refers to Regulation (EU) No 45/2001 replaced by Regulation 2018/1725, the EUDPR Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 7/28 (EUDPR), which applies to the processing of personal data by the European Commission and the Agency. CTR details the need for the protection of personal data as follows: • Recital 67: No personal data of data subjects participating in a clinical trial should be recorded in the EU database. The information in the EU database should be public, unless specific reasons require that a piece of information should not be published, in order to protect the right of the individual to private life and the right to the protection of personal data, recognised by Articles 7 and 8 of the Charter (…). • Article 56(1): All clinical trial information shall be recorded, processed, handled, and stored by the sponsor or investigator, as applicable, in such a way that it can be accurately reported, interpreted, and verified while the confidentiality of records and the personal data of the subjects remain protected in accordance with the applicable law on personal data protection. • Article 56(2): Appropriate technical and organisational measures shall be implemented to protect information and personal data processed against unauthorised or unlawful access, disclosure, dissemination, alteration, or destruction or accidental loss, in particular where the processing involves the transmission over a network. • Article 81(2): The EU database shall be established to enable cooperation between the competent authorities of the Member States concerned to the extent that it is necessary for the application of this Regulation and to search for specific clinical trials. It shall also facilitate the communication between sponsors and Member States concerned and enable sponsors to refer to previous submissions of an application for authorisation of a clinical trial or a substantial modification (…). • Article 81(4): The EU database shall be publicly accessible unless, for all or part of the data and information contained therein, confidentiality is justified on any of the following grounds: (a) protecting personal data in accordance with Regulation (EC) No 45/2001. • Article 81(6): The EU database shall contain personal data only insofar as this is necessary for the purposes of paragraph 2. • Article 81(7): No personal data of subjects shall be publicly accessible. • Article 93(1): Member States shall apply Directive 95/46/EC5 to the processing of personal data carried out in the Member States pursuant to this Regulation. • Article 93(2): Regulation (EC) No 45/20016 shall apply to the processing of personal data carried out by the Commission and the Agency pursuant to this Regulation. Commercially Confidential Information (CCI) related provisions: • Recital 68 clarifies that, for the purposes of the CTR, in general the data included in a clinical study report should not be considered commercially confidential once the procedure is finalised. • For clinical trials intended to be used in a marketing authorisation application in the EU/EEA, Article 37(4) of the CTR requires that the applicant for a marketing authorisation submits the clinical study report to the EU database within 30 days after the day the marketing authorisation has been granted, the procedure for granting marketing authorisation has been completed, or the applicant has withdrawn the application. 5 Replaced by Regulation (EU) 2016/679 (GDPR). 6 Replaced by Regulation (EU) 2018/1725 (EUDPR). Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 8/28 • Article 81(4) of the CTR states that: The EU database shall be publicly accessible unless, for all or part of the data and information contained therein, confidentiality is justified on any of the following grounds: ………. (b) protecting commercially confidential information, in particular through taking into account the status of the marketing authorisation for the medicinal product, unless there is an overriding public interest in disclosure. Overriding public interest anticipating the publication of clinical trials information means that the general public interest in having information made publicly available may outweigh considerations that the same information should remain confidential. It applies in exceptional circumstances only (e.g. in case of declared pandemic, public health emergency). In the context of inspection reports, the CTR sets out the following: • Article 53(2): The sponsor shall submit to the Member States concerned, through the EU portal, all inspection reports of third country authorities concerning the clinical trial. When requested by a Member State concerned, the sponsor shall submit a translation of the report or of its summary in an official language of the Union indicated in the request. • Article 78(6): Following an inspection, the Member State under whose responsibility the inspection has been conducted shall draw up an inspection report. That Member State shall make the inspection report available to the inspected entity and the sponsor of the relevant clinical trial and shall submit the inspection report through the EU portal. • Furthermore, Article 13 of the Commission Implementing Regulation (EU) 2017/556 of 24 March 20177 states (…) The inspection reports submitted through the EU portal shall not contain personal data of clinical trials' subjects. • The implementation of the disclosure rules of the Clinical Trials Regulation is without prejudice to the application of Regulation (EC) No 1049/2001 and citizens’ right to request documents under that Regulation. 7 COMMISSION IMPLEMENTING REGULATION (EU) 2017/ 556 - of 24 March 2017 - on the detailed arrangements for the good clinical practice inspection procedures pursuant to Regulation (EU) No 536 / 2014 of the European Parliament and of the Council (europa.eu). Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 9/28 2. Rules of clinical trial information in CTIS pertaining to submission and publication 2.1. Introduction This chapter describes the type of clinical trial information, including data and documents, submitted to CTIS and how this information is managed to protect personal data and commercially confidential information (CCI), while ensuring publication principles are met as per revised CTIS transparency rules. Principles of protection of personal data and CCI should be followed while using CTIS, as required in the CTR. The clinical trial information flow starts in the CTIS secure domain with an initial clinical trial application submitted by the sponsor, or delegated entities, to ask for authorisation of a clinical trial in the EU/EEA and the corresponding evaluation performed by the Member States concerned (MSC). Following the evaluation of the application, a decision is issued by each MSC for the application, on whether the trial is authorised, authorised with conditions, or not authorised. After a decision has been issued by the MSC, those data and documents submitted to CTIS for the trial will be made available to the public, depending to the trial’s development phase and population age, in line with the revised CTIS transparency rules. After the authorisation is obtained, the trial may start, and the MSC(s) will supervise the trial running in their territory. After the initial application, other application types may be submitted by the sponsor for the same trial such as, substantial modifications to the initial application or the addition of new MSC which are also subject to the assessment and decision by the relevant MSC. In addition to the above, non-substantial modifications to the content of the application dossier can be applied by the sponsor during the trial life cycle up to its completion, as well as notifications to the MSC(s) by the trial, of events of relevance, such as the occurrence of a serious breach or an urgent safety measure. The MSC(s) supervise the conduct of the trial in their territory with different means, including monitoring and assessing safety reports such as Annual Safety Report (ASRs), performing ad hoc assessments including for safety related matters, performing Good Clinical Practice (GCP) inspections, and having the possibility to apply corrective measures to request modifications, suspend the trial or revoke the trial authorisation, for example. The sequence of events occurring during the trial life cycle might require the collection and processing of personal data for the purposes set out in Article 81(2) of the Clinical Trials Regulation. Data and documents provided by the users in CTIS may also contain information that is considered commercially confidential. As defined in Article 81(4) of the CTR, personal data of trial participants, as well as other types of personal data, and commercial confidential information should be exempted from publication. Within CTIS secure domains for sponsors and Member States, users can have access to clinical trial data and documents for the trials of their concern. These users are clinical trial sponsors or delegated parties, marketing authorisation applicants/holders, EU/EEA Member States (encompassing responsible national competent authorities and Ethics Committees), the European Commission and the Agency. Access to data and documents in CTIS secure domain is managed through the user’s profile. The image below represents the different domains in CTIS, including sponsors’ and authorities’ domains with secure access and a public domain. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 10/28 CTIS therefore allows for the submission of required information in the secure domain and provides users access depending on their user profile, thus protecting personal data and the legitimate economic interest of sponsors for what concerns CCI. The following sections of this chapter describe the principles and timelines of public disclosure of those submitted data and documents. 2.2. Clinical trial information submitted to CTIS and disclosure rules The detailed list of structured data and documents that are, or are not subject to publication is specified in the CTIS application fields and Notifications, ASR and Results documents. Table I and table II of the Annex I to the present guidance document provide a list of the types of data and documents submitted to CTIS that are subject to publication, in line with the revised CTIS transparency rules. Their disclosure timelines are also mentioned, which depend on the trial category, on the population age (in case of category 1 trials) and on the trial phase (in case of category 2 trials that are integrated phase 1 and 2). The trial category is chosen by the sponsor when filling in the ‘form’ section of the application, based on definitions provided in table V of Annex I. Exceptions to those disclosure rules apply to all trials submitted before 18 June 2024 (so called ‘historical trials’), which have only their structured data published; moreover, for those trials documents submitted through additional member state applications are also not published: see section 2.3. of the present document and table IV of Annex I. A list of structured data fields and documents placeholders that are not subject to publication is provided in Table VI of Annex I: those type of submitted information will never be made publicly available (e.g. details of the legal representative, Investigator’s Brochure, IMPD documents, assessment reports). The partial initial applications are not made public until a decision is issued by at least one of the MSC. Applications which are not valid, that are lapsed, or those withdrawn by the sponsors before a decision is issued will not be made public. Note that only the most recent authorised application of any trial (or submitted, in case of Non- Substantial Modifications), as well as any ‘not authorised’ initial application, is made publicly available. Data and documents of authorised applications that are then superseded by subsequent authorised modifications are no longer disclosed, to increase the public’s readability of data; their conclusion and decision dates that are however subject to publication, for information purposes. Non authorised Substantial Modifications and Additional member state concerned applications, are not made publicly available: refer to Table IV of Annex I. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 11/28 2.2.1. Submission of structured data There is no equivalent of having a version ‘for publication’ and ‘not for publication’ in the structured data fields populated by CTIS users, as these fields cannot be redacted. Therefore, sponsors and Member States users should be mindful of this aspect when populating structured data fields that are subject to publication (as per Table I of Annex I), to protect personal data and commercially confidential information. Of note, the main characteristics of a clinical trial, the conclusions, and the decision on the authorisation of a clinical trial, the substantial modification of a clinical trial, and the clinical trial results including reasons for temporary halt and early termination, in general, should not be considered confidential8. 2.2.2. Submission of documents for ‘for publication’ and ‘not for publication’ Sponsors should submit high quality documentation to CTIS to enable a proper assessment by the MSC(s). For all those documents that are in scope of publication as per revised CTIS transparency rules (see Table II of Annex I), users need to provide a document version ‘for publication’, where Personal data and Commercially Confidential Information should be properly redacted or should not appear. This version will be published in line with principles and timelines foreseen in the revised transparency rules. For more information on what can be considered personal data and CCI, as well as on how to remove them, see chapters 3. and 4. Please note: any document inadvertently uploaded ‘for publication’ into the relevant CTIS document upload sections will be published; for example, if an IB is uploaded into the SmPC section of a Category 2 or 3 trial, this IB will be made public if not corrected by the sponsor before the decision on the application. A corresponding version ‘not for publication’ is also to be provided for MSC(s) assessment in case a redaction of personal data and CCI occurred in the version ‘for publication.’ Submission of both versions depend on the document type and content, and it may not be required in every instance (for example: in case of documents where no redaction is needed because there are no personal data or CCI, e.g. SmPC, providing only the version ‘for publication’ is sufficient). When both versions are required, these documents should be provided at the same time. In principle, no other alteration of documents’ content should occur between the document version ‘for publication’ and ‘not for publication,’ where the difference should be only personal data and commercially confidential information being redacted. With respect to those documents that are not subject to publication, note that quality and non-quality documents need to be submitted as separate documents. This includes the IMPD-Q, Scientific Advice - Quality and Quality RFI response documents. CTIS user roles depend on maintaining separate quality documents to ensure only authorised users can view quality information. 2.3. Publication rules of the so called ‘historical trials’ The ‘historical’ trials are trials submitted to CTIS before the 18 June 2024 whether they were previously fully publicly available or not, and with/without deferrals (i.e. whether the sponsor asked to defer the disclosure of certain documents uploaded in the placeholders ‘for publication’, see section 1.2. ). Those trials, including subsequent applications submitted on them, are subject to specific publication rules, designed to avoid unintended disclosure of confidential information contained in 8 Recital 68 of the Clinical Trials Regulation. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 12/28 documents which could have been subject to deferrals under the previous transparency rules. This section and table IV of Annex I summarise those rules, which for technical reasons are applicable to all historical trials, regardless of whether a deferral was applied in the past. As of 18 June 2024, for those trials: • Structured data fields: published in line with the timelines and principles of the revised CTIS transparency rules (see Annex I, table I). • Documents: all documents contained in applications submitted to CTIS before 18 June 2024 are not published9. This applies to all historical trials in the system, and regardless of the previous use of deferrals or publication status. If submitted on historical trials, the following kinds of applications trigger the publication of those documents that are in scope of the application and of the revised rules: • Substantial Modifications (part I and/or part II), including the Substantial Modification Part I ‘change of sponsor’ • Non-Substantial Modifications (part I10 and/or part II) • Additional Member State (triggering publication of part II docs only) It is therefore expected that the sponsor protects CCI and personal data on those documents in scope of publication, as per table II of Annex I. Note that CTIS considers all documents in scope of that application, regardless of the fact that their update was in scope of the modification or not. Note that the Substantial Modification Part I ‘change of sponsor’ does not allow the change of documents that are ‘for publication’ as part of the application (protocol, synopsis, SmPC): for those historical trials for which no part I documents were published yet, the sponsor needs to review the Part I ‘for publication’ documents to see if they contain CCI or personal data. If they do, the sponsor needs to submit first a Non-Substantial Modification Part I to update those documents so that they can be published and then submit the Substantial Modification Part I ‘change of sponsor’. As of 18 June 2024, if submitted on historical trials, the Additional Member State (AMSC) application does not trigger publication of part I documents. This type of application does not foresee the possibility of updating (and therefore redacting) protocol and protocol synopsis, that are in scope of the application and that would be published under the revised transparency rules. For this reason, for historical trials, all part I documents belonging to AMSC application will never be published (this includes protocol translations for AMSC applications, for technical reasons). Note that the above is only applicable for historical trials. AMSC applications of non-historical trials are published according to timelines and content defined in the revised transparency rules. A full overview can be found in Table IV of Annex I. Example: a multinational trial’s application submitted before 18 June 2024 displays the latest version of its structured data in line with revised transparency rules, while its documents are not published; as of 18 June 2024, for this trial: • a SM Part I is submitted with the purpose of updating the IB: documents uploaded in the protocol, synopsis and SmPC placeholders will be subject to publication as per revised rules  the sponsor needs to make sure that the ‘for publication’ versions of all part I documents are properly redacted 9 Including documents updated as part of an RFI response that is submitted after 18 June 2024, on applications submitted before this date. 10 Note that between 18 June 2024 and 6 Novemberr 2025, submissions of NSMs part I on historical trials did not trigger the publication of part I documents Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 13/28 • a SM Part I ‘change of sponsor’ is submitted with the purpose of updating the sponsor: documents uploaded in the protocol, synopsis and SmPC placeholders will be subject to publication as per revised rules  the sponsor needs to make sure that the ‘for publication’ versions of all part I documents are properly redacted: if not, a NSM part I needs to be submitted before the SM Part I ‘change of sponsor’ • a NSM Part I is submitted after 6 November 2025 with the purpose of updating the SmPC in line with Art 81(9) of the CTR: protocol and synopsis will be subject to publication as per revised rules  the sponsor needs to make sure that the ‘for publication’ versions of all part I documents are properly redacted • a SM Part II or a NSM Part II is submitted, concerning only one member state: part II documents are subject to disclosure rules only for that member state (part II documents concerning other member states will continue not to be disclosed)  the sponsor needs to make sure that the ‘for publication’ versions of all documents for the relevant member state’s part II are properly redacted. 2.4. Transparency aspects of transition trials For all those trials that had to be transitioned to the CTR as of 30 January 2025, Sponsors were encouraged to transition clinical trials from CTD to CTR based on available existing guidance documents. In particular, the Guidance for the Transition of clinical trials from the Clinical Trials Directive to the Clinical Trials Regulation11 clarifies that the redacted documents to be published with the administrative transitioning applications are the protocol, subject information sheet and informed consent form, in addition to submission of the non-redacted documents already approved by the MSCs. This is valid for all trials’ categories, except for category 1 trials, where it is sufficient to provide a redacted version of the protocol only, and a redacted version of the subject information sheet and informed consent form should not be provided, in line with the revised transparency rules (see Annex I, table I). In place of redacted versions for other parts of the application dossier, a document referring to the National Competent Authority (NCA) and/or ethics committee who assessed and gave a positive opinion on the clinical trial under the CTD could be uploaded in CTIS12. At the time of submission of subsequent modifications, the dossier can then be updated with the documents for publication falling within the scope of the application and of the revised transparency rules, e.g., including patient facing documents, synopsis and recruitment arrangements. For these documents, the same redaction principles as described in the present document apply. 11 Guidance for the Transition of clinical trials from the Clinical Trials Directive to the Clinical Trials Regulation 12 CTCG Best Practice Guide, see CLINICAL TRIALS COORDINATION GROUP (CTCG) ‘key documents list’ Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 14/28 3. Management of personal data in structured data fields and in documents submitted to CTIS 3.1. Introduction Personal data is any information that relates to an identified or identifiable living individual. Different pieces of information, which collected together can lead to the identification of a particular person, also constitute personal data13. The protection of personal data processed in CTIS is a joint responsibility of the EMA, the European Commission, the Member States (including National Competent Authorities and Ethics Committees) and commercial, non-commercial organisations including academia acting as sponsors of clinical trials and marketing authorisation applicants/holders. This joint responsibility is documented in the Joint Controllership Arrangement (JCA) for CTIS14, which includes in its Annex II the EMA Data Protection Notice regarding personal data processing in the Clinical Trials Information System (CTIS), addressed to data subjects namely CTIS users, sponsors, principal investigators, trial participants and that explains the purpose of the processing of personal data, the way CTIS collects, handles and protects personal data, how the information is evaluated and the rights of data subjects in relation to their personal data. The processing of personal data in CTIS, entailing the collection, publication and archiving of clinical trial information in documents and structured data fields is necessary for the management and functioning of the Agency and the performance of its tasks carried out in the public interest mandated by Union law, as joint controller of the CTIS, which includes the EU Portal and Database, for the effective materialisation of the objectives of the Clinical Trials Regulation. Therefore, this data processing by the Agency is lawful under Article 5(1)(a) of the EUDPR and justified on the grounds of public interest. In addition, the Member States, the European Commission, the commercial, non-commercial organisation including academia acting as sponsors of clinical trials and marketing authorisation applicants/holders, are also joint controllers in the CTIS. They are legally obliged to collect and upload relevant documents in the CTIS. Therefore, the data processing by the Member States and the European Commission also relies on the lawful ground of public interest under Article 6(1)(e) of the GDPR and Article 5(1)(a) of the EUDPR, respectively. In the case of sponsors and marketing authorisation applicants/holders their activities in CTIS and the related personal data processing is necessary for compliance with their legal obligations under the Clinical Trials Regulation in accordance with Article 6(1)(c) of the GDPR. In the context of transparency of clinical trials in CTIS, to protect the rights of trial participants to private life and the right to the protection of personal data, Article 81(7) of the CTR sets out that no personal data of trial participants shall be publicly accessible, which is further reinforced by Article 81(4) of the CTR that states that the CTIS shall be publicly accessible except where justified to protect the confidentiality of personal data. Personal data, including special categories of personal data of trial participants, should only be provided in CTIS as strictly necessary to allow for the scientific and regulatory assessment of the documents submitted to Member States. 13 https://commission.europa.eu/law/law-topic/data-protection/reform/what-personal-data_en 14 https://www.ema.europa.eu/en/documents/other/joint-controllership-arrangement-regard-clinical-trials-information- system-ctis_en.pdf Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 15/28 Chapter 2.1 of the EMA Privacy Statement (Annex II of the CTIS JCA 15 ), referring to the personal data in documents provided by the joint controllers in CTIS, states the following: ‘Should any of these documents contain personal data, as applicable and as required in light of Article 81(2) of Regulation (EU) No 536/2014, this can be provided in the version of the documents ‘not for publication’. The version of the documents ‘for publication’ should not contain personal data. To ensure that no personal data are made public these data should be anonymised, in the versions of documents ‘for publication’ with a few exceptions: see section 3.3. 3.2. The principles of anonymisation Anonymisation refers to information which does not relate to an identified or identifiable natural person or to personal data rendered anonymous in such a manner that the data subject is not or no longer identifiable (Recital 26 of GDPR and Recital 16 of EUDPR). The processing of such anonymous information is not subject to the provisions of the GDPR/EUDPR. To determine whether a natural person is identifiable, account should be taken of all the means likely to be used, such as singling out, either by the controller or by another person to identify the natural person directly or indirectly. To ascertain whether means are reasonably likely to be used to identify the natural person, account should be taken of all objective factors, such as the costs of and the amount of time required for identification, taking into consideration the available technology at the time of the processing and technological developments (Recital 26 of GDPR and Recital 16 of EUDPR). The Article 29 Working Party has issued an Opinion on Anonymisation Techniques16. The Opinion discusses that the effectiveness of anonymisation techniques should be checked against three criteria: i. is it still possible to single out an individual, ii. is it still possible to link records relating to an individual, and iii. can information be inferred concerning an individual?17 The Opinion also recognises that the use of one individual anonymisation technique alone may not meet with certainty, in every instance, the criteria of effective anonymisation. However, some of the criteria may be met in whole or in part by a given anonymisation technique, therefore a combination of techniques should be carefully applied together to enhance the robustness of the outcome.18 Combination of anonymisation techniques could be used, for example, in clinical study reports. For documents part of the CTIS application, which are expected to contain mainly direct identifiers, redaction would be most likely the anonymisation technique of choice. An anonymisation report describing the anonymisation techniques used is not expected to be provided in CTIS, unless specifically requested. When establishing a process for ensuring an adequate level of anonymisation, the following factors may be considered: • the likelihood of re-identification being attempted. • the likelihood the reidentification would be successful. • the anonymisation techniques which are available to use 15 https://www.ema.europa.eu/en/documents/other/joint-controllership-arrangement-regard-clinical-trials-information- system-ctis_en.pdf 16 Opinion 05/2014 on Anonymisation Techniques, 0829/14/EN WP216, available : https://ec.europa.eu/justice/article- 29/documentation/opinion-recommendation/files/2014/wp216_en.pdf 17 Ibid, Executive Summary. 18 Ibid, Section 5.2. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 16/28 • the quality of the data after anonymisation has taken place and whether this will meet the needs of the organisation (and the public) using the anonymised information. For example, once the anonymisation has been completed, an analysis of the interpretability of the anonymised data and information could be conducted to ensure they still remain meaningful and having utility for the public. 3.3. Anonymisation of personal data in the document version ‘for publication’ Personal data of individuals including names and surnames are captured, as applicable, in CTIS documents in the version ‘not for publication’ and should be anonymised in the document version ‘for publication’ (see table III of Annex I). However, exceptions applied to this rule, see 3.3.1. Regarding anonymisation of personal data in documents submitted to CTIS, the following principles should be taken into account: • Anonymisation of personal data in the documents submitted to CTIS ‘for publication’ should occur outside of CTIS and be applied consistently across all documents. • The publication of documents in CTIS occurs at the time of decision on an application, or later depending on the trial category (or population age, in case of Category 1 trials), see Table II of Annex I, in line with the principles defined in the revised CTIS transparency rules. • Where only one version of a document is provided in CTIS secure domain, namely the version ‘for publication’ this version will be subject to publication and used for review by the MSC(s), in the absence of a version ‘not for publication.’ • It is the sole responsibility of CTIS users to ensure the quality, accuracy, and adequacy of anonymisation applied and that the document versions ‘for publication’ are anonymised in accordance with the applicable process agreed within their organisation. • CTIS does not automatically verify if anonymisation has been applied in the version of documents intended for publication. • When progressing with the submission of the documents via CTIS, the authorised user confirms that the recording, storage, and publication of the documents in question are in accordance with Union data protection legislation. A dedicated template is available for use19. • The Agency, as the system administrator, is entitled to delete corrupted, incorrect, or unlawfully processed data, including removing information from CTIS public domain. This refers to requests for removal raised by the parties20 that uploaded the document in CTIS. Such requests can be raised by contacting the dedicated EMA service desk21. • In addition, EMA can delete incorrect information identified in the public domain, in which case EMA will inform the party that has provided the document, that an amendment to the published document is needed. The Agency, or other joint controllers in accordance with the joint controllership arrangement, can also edit the inaccurate or outdated information contained in the CTIS secure domain to comply with Union data protection legislation. • The Agency, the European Commission, the Member States, commercial and non-commercial organisations, including academia acting as sponsors and/or marketing authorisation applicants/holders, have joint responsibilities in submitting clinical trial data and documents in 19 https://health.ec.europa.eu/system/files/2022-09/compliance_reg2016_679_template_en.pdf 20 Deletion of incorrect/corrupted documentation should not occur on routine basis but rather on justified grounds to remove corrupted/unlawful information. This should not be seen as an instrument for modification / protection of personal data or commercial confidential information provided by CTIS users that retain the ultimate responsibility. 21 https://support.ema.europa.eu/esc?id=emp_taxonomy_topic&topic_id=2111dcb6c39d9d10e68bf1f4e40131ee Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 17/28 accordance with the Clinical Trials Regulation and Union data protection legislation. They also have joint responsibilities towards the data subjects and should have clear, defined processes in place to deal with any personal data breaches. • Other shared aspects of CTIS falling under the joint controllership scheme, such as the handling of data subjects’ rights, is addressed in a published joint controllership arrangement (JCA) for CTIS.22 In addition to the EMA data protection notice (Annex II of JCA31), Table II in the Annex to this document should be consulted for a more detailed description of the documents submitted via CTIS that are subject to publication and Table III of the same Annex for the type of personal data that they might typically contain. In the context of anonymising personal data within the CTIS documents ‘for publication,’ it is paramount to differentiate between: • Personal data, other than those of trial participants, such as of staff of the sponsor and of the marketing authorisation applicant/holder, author of a document (even if included as metadata), principal investigators, etc. • Personal data of clinical trial participants The following sections define the two different approaches to be followed for those data, in the documents that are and that are not subject to publication. 3.3.1. Anonymisation of personal data other than those of trial participants in the document version ‘for publication’ The anonymisation of personal data of individuals other than those of trial participants in the document version ‘for publication’ can be achieved by applying redaction as the sole anonymisation technique. Redactions can be performed by using any available tool which ensures that the redacted information is irreversibly blacked out by applying a permanent and unremovable overlay and, at the same time, making the redacted text unreadable and unsearchable in the document. Redaction of pre-specified identifiers, e.g., names, surnames, telephone numbers, can be done manually and/or automatically with software functionalities which enable the user to identify the pre- specified identifiers intended for redaction. Signatures should never be disclosed in the document version ‘for publication.’ Personal data of the author of a document, included as part of the metadata of a file, should equally be removed prior to uploading the document in CTIS secure domain and subsequent publication of the document. Instructions are available in dedicated CTIS training material, Module 02 – Guide on CTIS common features23. The following exceptions apply to names and surnames that should be disclosed in the document version ‘for publication’: • Names and surnames of principal investigators, head of the clinic/institution or other responsible person of the trial site, which are subject to publication as explained in the revised CTIS transparency rules. 22 https://www.ema.europa.eu/en/documents/other/joint-controllership-arrangement-regard-clinical-trials-information- system-ctis_en.pdf 23 Section 3 in training module 02: clinical-trials-information-system-ctis-common-features-ctis-training-programme- module-02_en.pdf (europa.eu) Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 18/28 • On the clinical study report: the full name of the sponsor and coordinating investigator signatories of the clinical study report and the identities of the principal investigator(s) who conducted the trial, which are also subject to publication as explained in the revised CTIS transparency rules. Note: the relevant signatures should, instead, be anonymised. All contact details (i.e. e-mail addresses and telephone numbers) of the above-mentioned individuals should be their professional contact details or functional contact details. If applicable (i.e. in the case of the principal investigator) those details are also published through the relevant structured data fields. These contact details should, therefore, not be redacted, or otherwise anonymised, in the documents uploaded in CTIS. Private contact details should not be provided in structured data fields in CTIS and if included in the documents, they should be redacted in the published documents. Note that the scientific and public sponsor contact details are also expected to be functional and not containing personal data (for example: [email protected]). 3.3.2. Anonymisation of personal data of trial participants in the document version ‘for publication’ Personal data of trial participants may only appear, as applicable, in CTIS documents that are not subject to publication and encompass personal data in a pseudonymised format (e.g., clinical trial subject ID number) as well as indirect identifiers such as weight, height, age, gender, etc. These personal data are to be anonymised in any document version ‘for publication’ (see table III of Annex I). Protection of personal data of trial participants in the document version ‘for publication’ should be achieved by using the entire range of available anonymization techniques that might require modification of the text, as redaction might not be the most suitable anonymisation technique to retain a meaningful level of data utility in all cases. The following elements should be considered when applying anonymisation in the documents to be published: • The choice of anonymisation techniques36 In the context of CTIS, no specific anonymisation methodology to protect personal data of clinical trial participants is prescribed, acknowledging that each anonymisation technique has its own strengths and weaknesses. The robustness of each anonymisation technique is based upon the anonymisation criteria and will help in identifying the most suitable technique (or combination of different techniques) to establish an adequate anonymisation process for a given document or set of documents. • Data utility Personal data of trial participants could be present in documents that are not subject to publication (e.g., notification of serious breaches, unexpected events or urgent safety measures, clinical study reports ‘not for publication’ version). It should be noted that it is equally important to preserve data utility in the public version of the documents, as much as possible, whilst ensuring adequate anonymisation. Besides, a quantitative approach to the measurement of the risk of re-identification could be favoured. This is particularly important in documents such as the final summary of results. • The sensitivity of the data The specificities of the relevant data should be taken into consideration when selecting the most appropriate anonymisation technique(s). For example, clinical trials conducted on rare diseases and/or on small populations may carry a high risk of re-identification of trial participants. A thorough risk assessment should be performed for such scenarios and the anonymisation of personal data should be adapted to the identified risk. Moreover, such an approach is also applicable to genetic information and low frequency events (e.g., rare events, extreme values, unusual treatments, pregnancy outcomes). Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 19/28 For a more detailed description of the available anonymisation techniques and their strengths and weaknesses refer to Article 29 Working Party Opinion on Anonymisation Techniques24. The same principles will apply to the protection of personal data of trial participants in the documents submitted to CTIS. 3.4. Documents not subject to publication: the principles of minimisation and pseudonymisation of personal data Personal data, if needed during the scientific and regulatory review carried out by the MSC, should be included in the document version ‘not for publication.’ Examples can be found in section 2 of the Q&A. This will enable the MSC to have all the necessary information for evaluation. The following principles should however be followed when including personal data in any document that is uploaded to CTIS. • Data minimisation Principles of minimisation should be followed when providing personal data, only as needed in light of Articles 81(6) referring to 81(2) of the CTR. This also applies also to personal data of the author of a document, included as part of the metadata of a file, which should be removed prior to uploading the document in CTIS secure domain. Signatures may also be reduced to a minimum, unless required by the MSC. Further information is provided in in section 2 of the Q&A. With regards to personal data of trial participants, the principle of data minimisation should also be followed, even if those data are pseudonymised in the documents that are not subject to publication in CTIS secure domain (see below). The use of personal data of trial participants should be proportionate. The clinical trial documents should include sufficient level of details to permit for the scientific evaluation and include sufficient data to evaluate the benefit/risk profile of the investigational medicinal product(s) used. • Pseudonymisation of data of trial participants The documents uploaded in CTIS may contain personal data belonging to trial participants, in a pseudonymised format. Most frequently included one is the clinical trial subject ID number. For the reasons presented above the clinical trial subject ID number should not be disclosed in the document version ‘for publication.’ It should be adequately anonymised by employing appropriate anonymisation techniques. The pseudonymisation of personal data can reduce the risks to the data subjects concerned (e.g., trial participants). Pseudonymisation refers to processing of personal data in such a manner that the personal data can no longer be attributed to a specific data subject without the use of additional information, provided that such additional information is kept separately and is subject to technical and organisational measures to ensure that the personal data are not attributed to an identified or identifiable natural person (Article 4(5) of GDPR and Article 3(6) of the EUDPR). Practically, pseudonymisation consists of replacing one attribute (typically a unique attribute) in a record by another. When pseudonymisation is used, the natural person could still be identified indirectly. Therefore, pseudonymisation reduces the probability to link the data variables belonging to a single data subject within a pseudonymised dataset with the identity of the data subject contained within the source records, and when used will not result in an anonymised dataset. Thereby, pseudonymisation is not an anonymisation technique but a useful security measure. 24 Opinion 05/2014 on Anonymisation Techniques, 0829/14/EN WP216 Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 20/28 Personal data which have undergone pseudonymisation and which could be attributed to a natural person by the use of additional information, is considered to be information on an identifiable natural person, therefore data protection rules still apply. Personal data of trial participants in a pseudonymised format (e.g., clinical trial subject ID number) and relevant indirect identifiers such as weight, height, age, gender, etc. may be contained in CTIS documents not subject to publication. A non-exhaustive list of documents that may contain them is provided below: • Investigator Brochure • Paediatric Investigational Plan • IMPD sections on Safety and Efficacy • Unexpected event reports and supporting information • Urgent safety measure reports and supporting information • Serious Breach Reports and supporting information • Clinical study reports (version ‘not for publication’) • Assessment reports • Inspection reports Note that the principle of data minimisation should also be followed, as mentioned above. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 21/28 4. Management of commercially confidential information (CCI) in clinical trial information submitted to CTIS 4.1. Introduction This chapter concerns the identification of CCI in the clinical trials information submitted to CTIS and present the system functionalities that enable its protection. CCI could be contained in a clinical trial application dossier or provided during the trial life cycle including in the CSR. For the purpose of this guidance document commercial confidential information (CCI) means any information which is not in the public domain or publicly available and when its disclosure may undermine the legitimate economic interest or competitive position of the concerned entities, e.g., clinical trial sponsors, marketing authorisation applicants/holders or service providers.25 Sponsors should use redaction as the method to protect CCI in the document version ‘for publication.’ When identifying potential CCI, sponsors are strongly encouraged to consider whether the information is already published, for example via the structured data fields in CTIS, or via other publication sources. In addition, sponsors should consider whether the documents submitted as part of a clinical trial application, are already in the public domain in connection to other trials registered in CTIS public website, or via other public sources, and if any redactions had been applied in these published documents. Consistency should be maintained, and the extent of the redactions should be similar across published documents. Nonetheless CCI should be available in the document version ‘not for publication’ as needed for Member State evaluation, and therefore it should not be redacted. It is not necessary for sponsors to mark/highlight the text that they consider CCI in documents ‘not for publication’ for awareness of the Regulatory Authorities (see relevant question of the Q&A). The version of a document ‘not for publication’ should be considered as the original, integral version of the document containing all information required for the assessment by the MSC. The identification of the commercially confidential information available in the structured data fields and in the documents submitted to CTIS is time dependent and should be approached as such by the CTIS users: more details are provided in section 4.3. 4.2. Mechanisms available in CTIS to protect CCI In line with the Revised CTIS transparency rules, in order to allow sponsors to protect their commercially confidential information, CTIS foresees the following means: • the public disclosure of a limited number of documents that are of key interest to the public, all listed in Table II of Annex I. All other documents including those that often contain CCI are not made public, such as the Investigator’s Brochure, IMPD documents, assessment reports, request for information (RFI) and corresponding responses, financial arrangements and more. An indicative list of those documents is available in Annex I, Table VI. • the possibility for sponsors to provide a version ‘for publication’ of those key documents of interest that would contain CCI redactions: further details are provided in sections to 4.3. to 4.6. • different timelines for disclosure of structured data and documents depending on the trial category, on the population age and on the trial category (see table I and II of Annex I) Trial categories are established in the context of CTIS, to distinguish the system behaviour on publication of information, essentially depending on the trial phase: table V of Annex I provides a 25 EMA’s definition of CCI in Policy 0043 has been endorsed by the Court of Justice in its case-law on access to documents. The definition of CCI in this document is an extrapolation and adaptation of the definition appearing in Policy 0043. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 22/28 description of those categories. For category one trials certain documents are never subject to publication (i.e. SmPC, Recruitment arrangements and Subject information and informed consent form), while others (protocol, relevant patient facing documents and synopsis) are only subject to publication at the time of results disclosure (for those trials conducted in paediatric population) or 30 months after the end of trial in the EU/EEA (for trials conducted solely on adults). With regards to structured data, most of data fields of category one trials conducted solely on adults are made publicly available only 30 months after the end of trial date in EU/EEA, limiting the publication of data for those trials to only fields of essential public interest. In addition, a specific distinction is made for the disclosure of certain Investigational Medicinal product (IMP) details of category two trials that are integrated phase one and two, for which the system foresees the same timeline for disclosure of all Category one trials (30 months after the end of trial in EU/EEA). Any document that is not listed in table II of Annex I is not subject to publication (see table VI of the same Annex for an indicative list). Sections 4.5. and 4.6. of this document aim to ensure a common understanding of what may be, or may not be, considered CCI within clinical trial structured data fields and documents provided in a clinical trial application and throughout the trial life cycle. 4.3. Redaction of CCI in the document version ‘for publication’ The sponsor is responsible to redact any CCI that may be present in the documents ‘for publication’ at the time of their submission to CTIS. The assessment of what needs to be redacted on each document is to be performed based on: • the definition of CCI, as provided in section 4.1. , complemented with information contained in section 4.5. • the timeline for disclosure of each document, which vary depending on the trial category and population age, see section 2.2. For Category 1 trials’ documents are made publicly available only several26 months after the end of the trial in EU/EEA: any redaction performed at time of submission should concern only those pieces of information that would still be considered CCI at the time of documents’ disclosure (e.g., of quality data in the trial protocol). The application of redactions to protect CCI should be limited to information that meets the provided definition and should carefully be weighed against the principles of transparency and ease of access to clinical trial information. It is expected that as the development plans advance, information on clinical trials which initially was considered CCI may no longer be considered as such due to technical and scientific advancements in that research field. This should, therefore, translate into a decreased level of CCI redactions applied over time in the new and modified documents submitted to CTIS during the trial life cycle, while the development plan for the medicinal product progresses. Retrospective removal of redactions from the documents already published is not expected. The latest version of each document type should, with time, be less and less redacted, as applicable, during the trial life cycle. 26 Protocol and synopsis documents for category 1 trials are published 30 months after the end of trial in EU/EEA if the trial is conducted only on adult subjects, while together with results submission if the trial includes paediatric subjects, refer to table II of Annex I Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 23/28 See an example below for illustrative purposes only: Redacted documents submitted to CTIS for publication have to remain meaningful to the public, including potential trial participants and health care professionals27. Sponsors are responsible for the redactions applied and for maintaining the clinical utility of the relevant redacted documents. In addition to the scientific and regulatory review of the documentation provided during the lifecycle of a clinical trial, RMS/MSC might reserve the right to comment, via an RFI, on the extent of the redactions applied by the sponsor to ensure that the principles of transparency are followed28. When requested by the RMS/MSC, sponsors should be able to demonstrate why the redaction in the documents is needed, (as their disclosure might, otherwise, impact their legitimate economic interest or competitive position). In the clinical study report (CSR) only minimum amount of redaction to protect CCI is expected: in line with recital 68 of the CTR29, CSR content should in principle not be considered CCI at the end of the marketing authorisation process30. Further details on redaction of CSR are provided in section 4.6.3. 4.4. Relevant expertise and consistent decision-making process on the identification and redaction of CCI The following elements should be considered when identifying CCI in the clinical trial information submitted to CTIS: • involve in the CCI identification process experts with relevant scientific and technical skills, and • to follow a consistent decision-making process. It is envisaged that incorporating these two elements into the CCI identification strategy would not only significantly reduce the need for applying redactions in the CTIS documents but would also increase the efficiency during the process of reviewing the documents to identify those pieces of information which may be considered CCI. According to the definition provided in section 4.1 a piece of information can be considered CCI if it meets simultaneously two criteria: (1) not being in the public domain or publicly available and (2) its disclosure would undermine the legitimate economic interests or competitive position of the concerned entities, e.g., sponsor, marketing authorisation applicants/holders or service providers. Based on this, in order to facilitate the identification of CCI a 2-step approach is suggested below: 27 Clinical Trials Regulation (EU) No 536/2014, Questions & Answers, 28 Article 94 (2)(a) of the Regulation (EU) No 536/2014 refers to application of penalties including non-compliance with the provisions laid down in the Regulation on submission of information intended to be made publicly available to the EU database. This is also specified in the ACT EU Questions and answers on the protection of Commercially Confidential Information and Personal Data while using CTIS 29 For the purposes of this Regulation, in general the data included in a clinical study report should not be considered commercially confidential once a marketing authorisation has been granted, the procedure for granting the marketing authorisation has been completed, the application for marketing authorisation has been withdrawn. 30 https://www.ema.europa.eu/en/documents/regulatory-procedural-guideline/external-guidance-implementation- european-medicines-agency-policy-publication-clinical-data_en-1.pdf Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 24/28 First step: rule out the possibility that the particular piece of information is available in the public domain (for further guidance please see section 4.5.1), or otherwise made publicly available. In case the information is already available in the public domain, it cannot constitute CCI, therefore no redaction should be implemented and the second step below is no longer applicable. and Second step: in case the information is not available in the public domain, it can be determined, in collaboration with experienced professionals having a relevant expertise in the clinical research area, whether the disclosure of the piece of information may undermine the legitimate economic interest or competitive position of the sponsor, marketing authorisation applicant/holder or service providers. If step 2 is also confirmed, the relevant portion of information can be considered CCI and be redacted from the documents. Medical writing can also play an important role in reducing the need for redactions. It is expected that embedding a CCI identification and tracking strategy during the drafting of the CTIS related documentation would limit the unnecessary dissemination of commercially confidential information in documents where these pieces of information are not essential, required, or relevant. This strategy can be further complemented by using document templates which specifically indicate which information is required to be included in the documents according to the legislation, scientific guidelines and regulatory guidance for clinical trials: Table III of Annex I provides an overview of the available templates. As a complementary approach, tagging those pieces of information which are considered CCI at the time the clinical trial documents are written would facilitate the preparation of the redacted document versions meant to be published. 4.5. Information that may be considered CCI It is recommended that where sponsors or marketing authorisation applicants/holders, identify a portion of information such as a word or figure, part of a sentence, part of a paragraph that they wish to include amongst the redactions to protect CCI, they should consider whether that portion of information meets the definition of CCI. If the information is CCI, the extent of the redactions should be limited to the word(s), figure(s), and specific sentences/elements in the text that, in the CTIS user’s view, can be considered CCI. In case only some sentences within the text or some specific figures Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 25/28 within the tables are claimed to be CCI, the users should not redact entire pages, sub-sections of a document or full tables, which for instance is accepted instead in respect of quality-related information. In order to facilitate the identification of CCI, a short list of specific types of pieces of information that may carry commercially confidential value is presented below. CTIS users should not redact these types of information automatically in the documentation submitted for publication in CTIS, yet they should assess whether the information is CCI on a case-by-case basis. The list of elements that may be considered CCI at the time of submission of a CTA and during the trial life cycle including submission of results/CSRs, includes as indicative examples: • The names, address and contact details of manufacturers or suppliers of the active substance or the excipients and finished product as well as of investigational medical devices, unless disclosure is required as per current pharmaceutical legislation (e.g., for some biological products). • The excipients' quantitative composition of the investigational/authorised product. • Detailed information on the synthesis or manufacture of the active substance. • Detailed descriptions of the manufacturing and control processes for the investigational/authorised final product. • Information related to future development plans for indications other than the one under investigation and not yet disclosed in the public domain. • New biomarkers or novel methodologies not yet qualified (to the extent that the information is not yet disclosed in the public domain), including new methodologies to support future developments as regards secondary or exploratory endpoints. • Detailed information concerning innovative analytical methods. • Detailed information on the facilities and equipment available at the sponsors and clinical sites. • Only in clinical trial application dossiers: the details of the daily dose allowed and maximum dose allowed for the medicinal product under investigation on justified grounds, i.e. when the sponsor proves that the specific information on the posology is not in the public domain and constitutes patentable matter, the disclosure of which before a patent application is filed (typically, after the completion of the trial and during the trial readout) would jeopardise its protection. This might be applicable, for example, to integrated phase I/phase II trials that are to be marked in CTIS as category 2 trials. The grounds for considering dose details as CCI should be clearly documented in the cover letter of the application31. 4.6. Information that may not be considered CCI In order to achieve a high level of consistency in the identification of CCI across the clinical trial documents, the sections presented below list some additional examples of types of information which may not be considered CCI32. 4.6.1. Information that is already in the public domain or publicly available It is recommended that the clinical trial sponsor and marketing authorisation applicants/holders compile a list of the most common websites/locations where information regarding their own medicinal 31 ACT EU questions and answers pm protection of personal data and commercially confidential information while using CTIS 32 These examples reflect the most common redactions proposed by applicants/MAHs which are usually rejected by EMA in the framework of Access to Documents in accordance with Regulation (EC) No 1049/2001. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 26/28 product is usually made available. They may consider creating and maintaining their own specific lists detailing the level of public information concerning their product(s). The following sources of information be included in the list (as a minimum): • Sponsors, Applicants’/MAHs’ own website(s). • EMA web-site (e.g. scientific guidelines, and for, centrally authorised products, the product EPAR,); • Clinical trials registries (such as CTIS, EU Clinical Trials Register, ClinicalTrials.gov); • Web-sites of other regulatory authorities within the EU and outside the EU (such as FDA, PMDA, TGA, Health Canada) especially when the product (or another product containing the same active substance) is approved in those specific jurisdictions; • Scientific literature and articles (such as Textbooks, PubMed, Medline). The information sources suggested above are not intended to constitute an exhaustive list, but rather to serve as a starting point for the creation of their own (more exhaustive, customized) lists. In this case, the above-mentioned examples should be considered as the minimum number of information sources to be scrutinised in order to reach a basic level of awareness on publicly available information related to the product concerned. 4.6.2. Information that does not bear any innovative elements Information which has already been revealed to certain extent, which can be inferred from information available in the public domain or has the content of textbooks or scientific guidelines as basis, should not be withheld from the public versions of the clinical trial documents. The fact that certain pieces of information are not in the public domain as such does not necessarily mean that they should be considered by default to constitute CCI. In many instances, particular pieces of text contained in clinical trial documents describe how the sponsors and marketing authorisation applicants/holders complied with regulatory and scientific guidelines and how they applied the scientific knowledge available at that time to their own development programme. In essence, these pieces of text do not reveal any innovative elements (of any regulatory or scientific nature) as the approaches described in the text are built upon logic and common sense in line with the content of publicly available documents such as: • Scientific literature and articles (Textbooks, PubMed, Medline). • Scientific and regulatory guidelines and guidance documents (ICH). • Treatment/clinical practice/disease management guidelines (Learn societies, HTAs). 4.6.3. Information that would not qualify as commercially confidential When considering commercially confidential information while using CTIS it is important to stress once again that the concept of CCI is time dependent, with a particular focus on the development phase of the medicinal product used in a clinical trial. It is important, therefore, to differentiate between CCI applicable in an earlier development phase at the time of submission of a clinical trial application and during the trial life cycle, and CCI at the end of the development cycle when trial results are provided in the clinical study report as part of a marketing authorisation procedure. Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 27/28 As mentioned in the introductory section 4.1 of this chapter it is expected that the redaction of CCI in the documents uploaded and submitted in CTIS will decrease overtime in line with the evolution of the development plan. Redaction of information that is already present as structured data field in CTIS should be done in line with the applicable publication timelines of those fields (see table I of Annex I). For early phase trials (e.g. category 1 trials), details on medicinal product, as well as on other characteristics such as the third parties and service providers are disclosed 30 months after the end of trial date in EU/EEA and therefore may be equally protected in any document that could be published earlier in time (e.g. protocol of a paediatric trial, summary results), if the disclosure would undermine the legitimate economic interest or competitive position of the concerned parties. Some data elements should not be redacted from CTIS documentation since they are unlikely to constitute commercially confidential information at any point in time, in an application nor in a CSR. Some of these data elements are presented below. The list is not intended to be exhaustive, rather as indicative examples about details of the data elements generally not considered to be CCI: • Unit measurements, in such cases only the actual value may be considered CCI. [e.g.] 2.5mL/kg  xx mL/kg. • Study identification number(s) (e.g., EudraCT, ClinicalTrials.gov Identifier (NCT…), sponsor’s internal study number). • Names and addresses of investigator sites and the names of the principal investigators at each trial site. • Names of the countries where the clinical study is/was conducted. • Number (how many) of study sites/research facilities were involved in the research. • Name of the applicant’s/sponsor’s own research facility(ies) where clinical studies were conducted (e.g., phase I studies). • Name of the trial sponsor or the legal entity (CRO) that acted on behalf of the sponsor for clinical trial application submission. • Names/identifying elements of all CROs, vendors and service providers involved in trial-related duties and functions (e.g., central laboratories, IVRS provider, image reading centres), unless referred to Category 1 trials. • Standard Operating Procedure (SOP) numbers and titles. • Information on worldwide approval status, Marketing Authorisation dates and launch status. In the clinical trial application(s), and during the whole trial life cycle, sponsors should only redact in the document version ‘for publication’ the CCI identified based on the principles described in section 4.3 above. In case the sponsors wish to flag what they consider CCI in the document version ‘not for publication’ uploaded in CTIS, they can mark the text with red border boxes. (see description in the Q&A). For clinical study reports to be submitted to CTIS, marketing authorisation applicants/holders should follow the same principles for the protection of CCI that are described in chapter 4 of the document on Policy 007033 on the publication of clinical study reports submitted to EMA as part of the centralised procedures for marketing authorisation, or variation or line extension of these. Where applicable, duplication of efforts should be avoided when preparing for publication those CSRs supporting 33 EMA guidance on policy 0070 Guidance document on how to approach the protection of personal data and commercially confidential information while using the Clinical Trials Information System (CTIS) Version 2 Page 28/28 centralised applications for marketing authorisation or variations thereof. As the submission and corresponding publication of CSRs via CTIS and publication via Policy 0070 initiative are triggered by the same regulatory milestone (i.e., the completion of the marketing authorisation procedure), the same level of CCI redaction applied in the CSR published on Clinical Data Publication portal (under Policy 0070 initiative) should be applied in the CSR provided in CTIS.
accelerating-clinical-trials.europa.eu
November 7, 2025 at 8:36 AM
Nieuws van ACT EU: Recommendations for clinical trial sponsors
Recommendations for clinical trial sponsors
Following consultation with the Multi-stakeholder platform Advisory Group, new documents have been published to support sponsors in the implementation of the Clinical Trials Regulation (CTR).
accelerating-clinical-trials.europa.eu
October 31, 2025 at 9:58 AM
Nieuws van ACT EU: Helpdesk for non-commercial sponsors: 6-month report
4. Analysis of the first 6-months of activity of the helpdesk
EMA/283849/2025 Page 1/14 EMA/283849/2025 European Medicines Agency Helpdesk for non-commercial sponsors: 6-month report 1. Background The Accelerating Clinical Trials in the EU (ACT EU) priority action on the implementation of the Clinical Trials Regulation (EU) No 536/2014 (CTR), aims to promote clinical trial research by supporting non- commercial sponsors (NCSs) in using the Clinical Trial Information System (CTIS) and navigating the clinical trials landscape. An important part of this activity has been the establishment of a dedicated helpdesk with the intention to facilitate dialogue and reduce the gap between NCSs and regulators. The NCS helpdesk offers dedicated and expedited support from the moment a request is submitted; the whole process is managed through ServiceNow: a centralized support system that provides assistance to users requiring technical support and service requests while using CTIS. It offers tailored guidance and training to help NCSs strengthen their capabilities for successfully submitting clinical trial applications, while also allowing EMA to better understand their challenges and adapt its support accordingly. 2. Participants: A dedicated team responsible for managing enquiries submitted and following a defined process flow was established within EMA; National Competent Authorities (NCAs) play an active role in the helpdesk by collaborating in responding to queries that require joint solutions or are complex in nature, particularly those not directly related to CTIS functionalities. In addition, NCAs may provide direct support to NCS users, following consultation with the helpdesk, when queries concern specific national requirements. 3. Process for CTR/CTIS helpdesk: Entry points are identified for requests of support on CTR and CTIS use via ServiceNow where the user needs to select the request among three possible options (Fig. 1): - “Report an Issue with CTIS” for technical issues; the so-called “incidents”, Helpdesk for non-commercial sponsors: 6-month report Page 2/14 - “Request a CTIS Service” for assistance (e.g. for support regarding the system functionalities), - “Request for information CTIS” for the use of CTIS and regulatory questions. Fig. 1: The image shows the three options available in ServiceNow to request support on CTIS. After the request of interest is selected, the user needs to fill in the following fields and the affiliation where it is possible to specify that the request is raised by a NCS: Once the form is completed and submitted, the request is received and validated by the ServiceNow dedicated team. Validation process confirms the CTR/CTIS scope and affiliation of the submitter. If the question is within the scope, the team provides a reply in writing within the timelines assigned based on the priority of the topic; these timelines are defined as SLAs (Service Level Agreements). The user may require additional clarifications; in which case further support is provided accordingly. In case the issue persists, a virtual meeting can be organised with NCS representatives and CTIS experts, technical team, NCA members, as applicable. After fulfilling the request, a satisfaction survey is submitted, and the process is closed. Helpdesk for non-commercial sponsors: 6-month report Page 3/14 4. Analysis of the first 6-months of activity of the helpdesk 4.1. Issues reported / Incidents in the period October 2024 – March 2025 and time for resolution The issues reported in ServiceNow related to technical problem with CTIS are also called “incidents”. The graph below (Fig. 2) shows the total number of incidents reported by NCSs and the number of resolved incidents for the 6-month period Oct 2024-March 2025 divided per month. All tickets raised under this category (tot=578) in the timeframe are considered closed. Fig. 2: Number of tickets raised on incidents in the timeframe considered Oct 2024- March 2025. The graph below (Fig. 3) shows the percentage of incidents resolution based on SLAs timelines above mentioned, including percentage of tickets closed within and above the expected timeline. An average of 87% of incidents has been resolved within the expected timelines based on the period Oct 2024–March 2025. Helpdesk for non-commercial sponsors: 6-month report Page 4/14 Fig. 3 Resolution timeline related to the incidents raised by NCSs in ServiceNow in the timeframe considered Oct 2024- March 2025. 4.2. Request for service and for information / Questions in the period October 2024 – March 2025 and time for resolution For the purpose of this report, requests for service and requests for information have been grouped together under the term “questions”. The graph below (Fig. 4) shows the total number of questions submitted by NCSs and the number of resolved questions for the 6-month period Oct 2024-Mar 2025 divided per month. All tickets raised under this category (tot=596) in the timeframe are considered closed. Helpdesk for non-commercial sponsors: 6-month report Page 5/14 Fig. 4 Number of tickets raised on questions in the timeframe considered Oct 2024- March 2025. The graph below (Fig. 5) shows the percentage of questions resolution based on the SLAs timeline, including percentage of tickets closed within and above the expected timeline. An average of 90% of questions has been resolved within the expected timelines based on the period Oct 2024–March 2025. Helpdesk for non-commercial sponsors: 6-month report Page 6/14 Fig. 5 Resolution timeline related to the questions raised by NCS in ServiceNow in the timeframe considered Oct 2024- March 2025. Helpdesk for non-commercial sponsors: 6-month report Page 7/14 5. Main topics of the enquiries raised by NCSs The following table (Table 1) shows a list of tags used to categorise the enquiries raised by NCSs. For each incoming enquiry one or more tags were assigned to accurately capture and categorise the nature of the request, enabling a structured and systematic classification of all requests. This categorisation will enable targeted support and clarification, as well as help gain a better understanding of the challenges faced by NCSs. After tagging all requests, data was compiled and displayed in two sub-sections: one for incidents and one for questions. These sub-sections show the most common types of requests, offering insights into recurring issues and key areas of interest for NCSs. Table 1. Tags used to categorise the enquiries (both incidents and questions) raised by NCSs. Tag When to be used Change_sponsor_data When a request is submitted for support to update sponsor data or the issue is related to a Sponsor (ORG ID) change CTIS_improvement For suggestions to improve CTIS functionality CTIS_SM incident For tickets related to incidents of SMs for trials (not blocking) CTIS_SM incident blocked For tickets related to SMs of trials whereby submission or response to RFI is blocked CTIS_training For tickets related to users needing advice on functionality of CTIS CTIS_training_submission For tickets related to user needing advice on how to proceed with the submission of the applications CTIS_known issue For tickets related to known issues and when tickets are associated to created Problems (issue reproducible) CTIS_publication process For tickets related to transparency CTIS_RFI For tickets related to RFI issues CTIS_CTR interpretation For tickets requesting general advice of interpretation of EU CTR and related to regulatory topics CTIS_General error For tickets related to general errors in CTIS functionality not associated to a Problem because not reproducible (e.g. temporary glitch) CTIS_Transition For tickets related to transition applications CTIS_Lapsed CTA For tickets related to lapsed trials CTIS_Notification date For tickets related to issues with notification dates (e.g. start of recruitment, temporary halt, global/local EOT) Helpdesk for non-commercial sponsors: 6-month report Page 8/14 CTIS_User access_roles For tickets related to users not being able to access a trial or user is able to access part of a trial that they do not have access to or related to training about roles in general CTIS_XEVMPD For tickets related to integration between CTIS and XEVMPD CTIS_OMS For tickets related to integration between CTIS and OMS CTIS_Notices_alerts For tickets related to notices and alerts CTIS_other For tickets that cannot be classified otherwise (e.g related to MFA, login issues) CTIS_ASR For tickets related to ASRs NCS_NCA For tickets submitted by Non-commercial Sponsors in which NCA will be involved or where advice to refer to NCA is provided 5.1. Main topics of the incidents raised by NCSs in the period Oct 2024 – Mar 2025 The graph below (Fig. 6) shows the most frequent categories of incidents raised by NCSs in the reporting period, considering a total number of 578 incidents. The two most frequent categories identified are “training” and “known issues” which are broader categories that include additional subcategories categories. The “training” category covers tickets from users seeking guidance on CTIS functionalities. While the information shared by the helpdesk is generally available in the official user manuals and training materials, the assistance serves to help navigating these materials to the users who demonstrate a lack of familiarity with the system. Notably, 45 tickets under this category relate specifically to RFI (Request for Information), highlighting that sponsors often face difficulties managing the RFI response process. The “known issues” category (108 tickets) includes cases associated with previously identified technical problems, which are recurrent under specific conditions and reproducible by the technical team. The most common issues reported in this category concern incorrect notices and alerts being triggered (44 tickets) and problems with submitting notification dates (12 tickets). A significant number of incidents also concern other systems essential for correct sponsor registration in CTIS, such as OMS and XEVMPD. Helpdesk for non-commercial sponsors: 6-month report Page 9/14 Fig. 6 Main topics of the incidents in the timeframe considered. 5.1.1. Incidents raised by NCSs with implication of NCAs Among the incidents reported by the NCSs, 20 required interventions at the level of the NCAs involved in the Clinical Trial Assessment. The figures below illustrate the primary topics associated with these incidents and identify the NCAs involved (Fig.7 and Fig. 8). Helpdesk for non-commercial sponsors: 6-month report Page 10/14 Fig. 7 Topics where NCAs has been involved. Fig. 8 NCAs involved in the incidents. Helpdesk for non-commercial sponsors: 6-month report Page 11/14 5.2. Main topics of the questions raised by NCSs in the period Oct 2024 - Dec 2025 The graph below (Fig. 9) shows the most frequent categories of questions raised by NCSs in the reporting period considering a total number of questions of 596. The most frequent category is “training”. As this is used to identify tickets related to users needing advice on functionality of CTIS not surprisingly it is the most selected; the second most selected category is “CTR interpretation” that is clearly related to the interpretation of the Regulation and other regulatory questions. This is followed by “roles” related to users not being able to access a trial, or simply not able to proceed with the assignment of roles to perform certain actions. Many enquiries are again related to RFI and to transition of clinical trial applications from the Clinical Trials Directive to the Clinical Trials Regulation during the transition period that ended in January 2025. 51 questions (out of 596) required the intervention or are referred somehow to one NCA to facilitate the resolution (Fig. 10 and Fig.11). Fig. 9 Main topics of the questions in the timeframe considered. Helpdesk for non-commercial sponsors: 6-month report Page 12/14 5.2.1. Questions raised by NCSs requiring involvement of NCAs Among the questions raised by NCSs, 51 required interventions at the level of the NCAs involved in the Clinical Trial Assessment. The figures below illustrate the primary topics associated with these questions and identify the NCAs involved (Fig.10 and Fig. 11). Fig. 10 Main topics of the questions referred to NCAs. Helpdesk for non-commercial sponsors: 6-month report Page 13/14 Fig. 11 NCAs to whom the questions were referred to. 6. Conclusion The analysis of the tickets submitted by NCSs to the helpdesk between October 2024 and March 2025 provides valuable insights into the nature and frequency of the issues encountered with CTIS. A total of 578 incidents and 596 questions were reported during this six-month period. All tickets raised in this timeframe have been resolved. Most tickets were resolved in a timely manner, as matter of fact 87% of incidents and 90% of questions were addresses within the expected timeline demonstrating effective handling and closure by the support team. The categorisation of incidents revealed that the most frequently selected tags were “training” and “known issues”. These broad categories reflect a recurring need for user guidance on CTIS functionalities, and the presence of system-related problems already identified and tracked. Beyond these, a significant number of incidents were related to RFI, Notices and Alerts, Annual Safety Reports (ASRs), and (CTD to CTR) transition activities, as well as integration issues with external systems such as OMS and XEVMPD. Notably, 20 incidents required further actions or involvement of the NCA for resolution. Similarly, the analysis of questions showed that “training” was again the most common category, followed by “CTR interpretation”, which highlights the need for regulatory clarification. Other frequent Helpdesk for non-commercial sponsors: 6-month report Page 14/14 topics included user roles, RFI, and transition. Among the questions raised by NCS, 51 required NCA input. Overall, the data indicates that while many requests are related to user support and known system issues, a substantial portion also involves regulatory interpretation and cross-system dependencies. These findings will be useful to support future improvements in user training, system documentation, and inter-agency coordination.
accelerating-clinical-trials.europa.eu
October 30, 2025 at 3:57 PM
Nieuws van ACT EU: ACT EU multi-stakeholder workshop on methodology guidance report
ACT EU multi-stakeholder workshop: A patient-centered approach to methodologies
Report of the methodology guidance workshop ACT EU multi-stakeholder workshop on methodology guidance – workshop report 2 Contents Executive Summary ______________________________________________________ 3 Introduction ____________________________________________________________ 5 Complex trials I _________________________________________________________ 6 Paediatric trials __________________________________________________________ 8 Pragmatic trials _________________________________________________________ 10 Digital Endpoints ________________________________________________________ 11 Beyond RCTs ___________________________________________________________ 13 Patient-centricity, diversity and representativeness in clinical trials ________________ 14 Decentralised elements in clinical trials ______________________________________ 15 Complex trials II ________________________________________________________ 16 Closing remarks ________________________________________________________ 18Glossary ______________________________________________________________ 19 More information _______________________________________________________ 20 ACT EU multi-stakeholder workshop on methodology guidance – workshop report 3 Executive Summary The ACT EU multi-stakeholder workshop on methodology guidance facilitated a discussion between relevant stakeholders on key topics of clinical trial methodology. The objective of the workshop was to identify the challenges stakeholders face, propose ways for improving patient centricity and suggest possible solutions. In break-out sessions, participants from relevant stakeholder groups discussed complex clinical trials, paediatric trials, pragmatic trials, digital endpoints, evidence generated in non-randomised designs, patient centricity, decentralised trials, and platform trials. The following key themes emerged across the different break-out sessions. Challenges • The multitude of guidance documents and requirements from a variety of stakeholders makes it difficult to plan a trial that is acceptable to all decision-makers. • With the In Vitro Diagnostic Regulation, Medical Device Regulation and Pharma Legislation, multiple legislative frameworks apply within the EU that are difficult to navigate. • Requirements of the Clinical Trial Regulation are new and implementation may differ across EU member states. • Evidentiary requirements for decision-making are not always clear. • Patient communities are often not aware of relevant planned and ongoing clinical trials. Improving patient-centricity • Consult patients and practitioners consistently and early on during the design and conduct of clinical trials and the development of guidance on clinical trial methodology. • Provide easily accessible and understandable information about the clinical trial for participants. • Ensure adequate compensation for patient input into clinical trial design. • Improve the visibility of clinical trials. Solutions • Clarify regulatory requirements by developing new guidance or updating existing guidance. • Improve the alignment between Regulatory Agencies, Health Technology Assessment bodies and Notified bodies within Europe and aim to harmonise regulatory requirements internationally. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 4 • Clarify requirements of the Clinical Trial Regulation and strive for alignment between EU Member States. • Reduce the bureaucratic burden for trial conduct and improve processes and IT systems for clinical trial registration. • Increase clarity and consistent use of existing regulatory pathways. • Improve pre-competitive collaboration of stakeholders and data sharing. The discussions also highlighted the topics that would benefit from further clarification, by developing new guidance or updating existing guidance: • The definition of “normal clinical practice” to facilitate treatment optimisation trials with already approved medicinal products. • The definition of “unmet medical need”. • Age-inclusive research and including paediatric patients into adult clinical trials. • The recommendation paper on Ethical considerations for clinical trials conducted with minors. • Drug development for neonates. • Conducting clinical trials in small populations with a specific focus on N of 1 clinical trials. • Evidentiary requirements for the qualification of digital endpoints for use in clinical trials. • Systematically guiding the dialogue between sponsors, regulatory bodies (National Competent Authorities and Ethic committees) and Health Technology Assessment bodies on the acceptability of non-RCT designs. • The operationalisation of the Clinical Trial Regulation requirement describing patient involvement in clinical trial designs. • The regulatory acceptability of methodological aspects of platform trials. As next steps, focussed follow-up discussions and further workshops will be needed to consider the outcomes from the break-out sessions. The workshop report will be disseminated publicly and presented to relevant expert groups in the European Medicines Regulatory Network to acknowledge the challenges identified by all stakeholders and to consider the proposed ways for improving patient centricity and addressing the stakeholder challenges. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 5 Introduction Opening remarks by Emer Cooke (EMA), Karl Broich (HMA), Isabelle Clamou (European Commission) and Workshop Chairs Monique Al (CTCG) & Kit Roes (MWP) Excellent clinical trials are core to the generation of high-quality clinical evidence benefitting patients and healthcare in Europe. Accelerating Clinical Trials in the EU (ACT EU) is a joint initiative of the European Commission, Heads of Medicines Agencies (HMA) and the European Medicines Agency (EMA) that supports clinical trials conducted in Europe through regulatory, technological and process innovation. The evolving clinical trials landscape is an opportunity to improve coordination between stakeholders, regulators and ethics committees, aiming for clinical trials that meet the needs of all involved stakeholders, and particularly benefit patients and healthcare in the EU. To ensure that clinical trials generate fit-for-purpose evidence, the European Medicines Regulatory Network provides guidance on clinical trial methodology, supporting trial sponsors in the implementation of new and innovative approaches for the design and conduct of clinical trials. Therefore, the development of guidance on clinical trial methodologies is among the ACT EU priority areas. As part of this work, ACT EU has already supported the publication of a Q&A document on complex clinical trials and a recommendation paper on decentralised clinical trials. The ACT EU multi-stakeholder workshop on methodology guidance invited early input from all relevant stakeholders on key methodology topics, selected based on a public call for topics. A Break-out session was dedicated to each selected topic. Participating experts discussed challenges and needs around clinical trial methodology guidance, while keeping patients’ needs at the centre of discussions. Participants also discussed possible ways for the European Medicines Regulatory Network to address the identified challenges and for improving patient centricity and involvement. These discussions will inform the future development of guidance by the European Medicines Regulatory Network. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 6 Session A Complex trials I Moderators: Olga Kholmanskikh (FAMHP), Frank Petavy (EMA) Presenters: Olga Kholmanskikh (FAMHP), Laura Arenare (Istituto Nazionale Tumori IRCCS Fondazione Pascale), Sahar Barjesteh van Waalwijk van Doorn-Khosrovani (CZ Health Insurance, Leiden University Medical Centre), Nicky Best (GSK) and Kaspar Rufibach (Roche) The session opened with an introduction to the evolving clinical trials ecosystem and to the Q&A document on complex clinical trials followed by a presentation on the concept of Patients’ Journey Studies (PJS), where each node of clinical decision-making can be a randomisation if there is equipoise between different options. This type of trial addresses questions regarding subsequent lines of treatment and overall strategies that are generally left unanswered at the time of marketing authorisation. The Drug Rediscovery Protocol (DRUP) and the DRUP-like trials (the PRIME-ROSE consortium) were presented as examples of multi-drug, pan-cancer precision oncology trials, where the provision of treatments to patients belonging to rare and under-studied subpopulations is combined with data- collection and analysis, from preliminary stages to expansion cohorts. In these trials, successful cohorts will undergo central assessment and groundwork will be established to facilitate Health Technology Assessment (HTA) analysis and reimbursement decisions to ensure equitable access. The session closed with a presentation on the different estimands of interest for different stakeholders and of the technical and operational problems in pursuing estimation of all those in a development plan, highlighting the need for guidance for the role of augmented control. Key challenges identified by stakeholders • There is a trade-off between trying to answer many relevant questions in one trial (which would favour the option of designing complex trials) and interpretability and robustness of trial results (which would favour simpler studies). • The multitude of guidance documents and requirements from a variety of stakeholders makes it difficult to plan a trial that is acceptable to all decision-makers. • Assessing the feasibility of trials can also be challenging, given the imperfect knowledge of patients’ and prescribers’ preferences and the different treatment opportunities outside of trials. How to improve patient-centricity • The value of answering all the questions that inform joint decision-making of doctors and patients (including with post-authorisation trials) has to be recognised by all stakeholders, and trials that are suited for this purpose have to be prioritised. • Patients and practitioners should be increasingly consulted in designing clinical trials; the acceptability of certain designs and the desirability of collecting specific outcome measures should not be assumed, but proactively discussed and supported with data. Suggested ways forward to address the challenges • Methodological research on the risk of false positive conclusions (especially at a regulatory decision level) and the potential of bias in different types of complex trials, as well as on possible mitigation strategies, has to be prioritised. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 7 • Guidance should be developed with increasing degrees of harmonisation between different decision-makers (clinical trial approval bodies, marketing authorisation bodies, HTA bodies, prescribers), so that trials that satisfy the requirements of all are increasingly enabled. • Post-authorisation clinical trials (of the type of DRUG-Access Protocol and PJS) – including by academic sponsors - that aim at addressing questions that go beyond the requirements of marketing authorisation have to be promoted. As these trials often require complex designs, provision of robust guidance is important. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 8 Session B Paediatric trials Moderators: Anette Solli Karlsen (Norwegian Medicines Agency) and Dina Apele-Freimane (State agency of Medicines of Latvia) Presenters: Dominik Karres (EMA), Monique Al (CTCG), Wolfgang Berdel (Association of Medical Ethics Committees in Germany), Solange Corriol Rohou (AstraZeneca), Begonya Nafria Escalera (Sant Joan de Déu Research Foundation) The session focused on paediatric trials as part of the drug development and related methodology guidance needs. The changes in requirements for paediatric clinical trials brought about with the implementation of the Clinical Trial Regulation (CTR) and the need to revise the recommendations on ethical considerations for clinical trials with minors were presented. Academic studies investigating treatment optimisation in paediatric oncology were discussed, providing the view of clinicians on the interrelation between the Clinical Trial Directive (CTD) and CTR and these academic studies. The industry perspective on the current situation for paediatric clinical trials was also presented, focusing on opportunities for optimisation and highlighting the value of public-private partnerships. The session highlighted the importance of patient involvement from the conception and design phase to address the challenges in paediatric clinical trials. Key challenges identified by stakeholders • Paediatric drug development is mostly conducted in global drug development programs with diverse regional guidelines which need to be adhered to. • Terminology lacks a clear definition and is used heterogeneously, for example, the terms “normal clinical practice” and “unmet medical need”. • Paediatric clinical trials are often conducted for overlapping patient populations, also in relation to adult trials, which creates a barrier to the conduct based on small populations. • Paediatric patients and/or their parents/caregivers are rarely included in the planning, conduct and reporting of paediatric clinical trials. • Age-inclusive research is important for faster knowledge on the best treatment as well as timely access for the paediatric population, but is challenging with no guidance on when it is acceptable. • The CTR requirement for paediatric clinical trials to have direct benefit for the individual taking part in the trial has not been implemented in a harmonised manner at national level. • In small populations randomised clinical trials (RCTs) are most often not feasible; in such cases, data collection from N of 1 trials may be considered as a solution. There is lack of guidance and understanding, however, on how such trials should be conducted and fit within the regulatory system. • Some diseases occur in neonates-only, but dedicated guidance on trials in this special population is lacking. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 9 How to improve patient-centricity Paediatric patients and/or their parents/caregivers should be involved in the conception and design of clinical trials to ensure the relevance of the design elements for paediatric patients and to increase feasibility from a recruitment and adherence standpoint. • Diversity needs to be considered when designing patient and public involvement in clinical trials aiming to represent all relevant groups affected by the disease studied in these trials. • Documentation of patient involvement during the drug development process, including the design of clinical trials, should be part of a clinical trial application. Regulators should request information about patient involvement. • Patient and public involvement activities need to be led by paediatric experts to ensure the quality of the methods and outcomes, the protection of children’s rights and ethical principles, and to avoid any kind of conflict of interest or bias in the process. • In paediatric clinical trials, patient involvement should be interpreted to include family members and other caretakers. Suggested ways forward to address the challenges Key general points to consider when writing guidance for paediatric clinical trials are: • Guidance should not be developed in isolation in one regulatory region but rather be cross- regional, if possible, to ensure equal opportunities for patients around the world. • When methodology guidance is developed, special attention should be given to aspects which might be different for a paediatric population. • Multi-stakeholder collaboration is necessary when developing guidance. Suggestions for development/updating of guidance: • Clarify the definitions of “normal clinical practice” to facilitate treatment optimisation trials with already approved medicinal products. It is recommended that clinical medical societies should drive the definition of this term. • Clarify the definition of “unmet medical need”. • Develop guidance on when and how age inclusive research, including paediatric patients into adult clinical trials, is acceptable. • Update the recommendation paper on Ethical considerations for clinical trials on medicinal products conducted with minors from the Commission as published on Eudralex volume 10. • Clarify the CTR requirement for paediatric clinical trials to have direct benefit for the individual taking part in the trial, and the expectation for medical conditions that occur in minors and adults, but which manifest themselves in a different way at a young age. Harmonisation is needed between EU members state views, including that of the ethics committees. • Develop guidance on drug development for neonates. • Update the EMA guidance on how to conduct clinical trials in small populations with a specific focus on N of 1 clinical trials. • Ensure consistent use of terminology on extrapolation in the context of paediatric drug development when methodology guidance is updated. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 10 Session C Pragmatic trials Moderators: Frederik Grell Noergaard (DKMA), Claire Bahans (Research Ethics committees, FR) Presenters: Beate Wiesler (IQWiQ), Rudolf Huber (Ludwig-Maximilians-Universität Munich), Denis Lacombe (EORTC), Nafsika Kronidou Horst (Roche). The benefits of pragmatic trials to provide data on comparative effectiveness in routine care were presented, noting that randomisation is a key element for HTA. The session also explored the role of pragmatic trials in generating relevant data, optimising treatment and informing routine clinical practice. The value of pragmatic trials for patients, clinicians and payers was underlined, as these trials combine the methodological strengths of RCTs with the inclusiveness of a real-world setting. Pragmatic trials were highlighted as key to optimising the appropriate use of medicinal products and, for example, adjusting dose and schedule notably in de-escalation trials. The EFPIA position paper on randomised pragmatic trials to inform regulatory decisions was presented, focusing on the key considerations, learning opportunities and possible regulatory settings for the use of such trials. Key challenges identified by stakeholders • Randomised pragmatic trials are often conducted by non-commercial sponsors, where the burden to comply with the CTR is too resource-consuming rendering the trials unfeasible. The main challenges are the workload for reporting of safety events with lack of clarity on the low interventional clinical trial borderline and the requirements for reimbursement of investigational medicinal products. • The relevance of research questions and outcomes can differ according to stakeholders’ points of view (e.g. patients, HTA bodies, National Competent Authorities (NCAs), Ethics Committees) and across Member States, e.g. in regard to the choice for “standard of care” and “clinical practice’’. • The definition of pragmatic trials is not clear with partial overlap with other study types such as trials with decentralised elements. How to improve patient-centricity • Patients should be involved from the beginning of research conception and design. Treatment optimisation and quality of life are key points for patients, together with the reduction of operational complexities e.g. lengthy and complex informed consent. • The trial summary for patients in a pragmatic trial should be simplified. Suggested ways forward to address the challenges • Reduce the bureaucratic burden for pragmatic trials e.g. with a risk-based approach regarding safety registration and reporting, in line with ICH E19 and the CTR. Initiatives should include sponsors, patients and practitioners and focus on simplifying trial protocols. • Ensure that the design of clinical trials can provide data suitable for marketing authorisation and reimbursement decisions. To this point, joined scientific advice including HTA and ethics could be explored. • Designing clinical trials with more pragmatic elements during medicines development could reduce the need for additional trials/evidence after drug approval. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 11 Session D Digital Endpoints Moderators: Jesper Kjaer (DKMA), Thorsten Vetter (EMA) Presenters: Martin Daumer (School of Computation, Information and Technology), Lada Leyens (Takeda), Laurent Servais (Oxford University) The session opened with presentations on the development and validation of digital endpoints from an academic perspective, followed by the EFPIA perspective on opportunities and challenges in validating digital endpoints. Lastly, experience with the EMA qualification procedure for the “Stride velocity 95th centile” (SV95c) as a qualified primary endpoint for studies in Duchenne Muscular Dystrophy was presented. Key challenges identified by stakeholders • It is difficult for a single developer to generate sufficient high-quality data needed for the development, validation and qualification of digital endpoints. • With the (IVDR), Medical Device Regulation (MDR) and Pharma Legislation, multiple regulatory frameworks apply within the EU that are difficult to navigate and are not always aligned. Furthermore, there is a lack of international harmonisation. • Evidentiary requirements for validating and qualifying digital endpoints are not clear. • Including (too) many measures to generate evidence for the development and validation of new (digital) endpoints in a clinical trial puts burden on patients, challenging patient retention and trial conduct. How to improve patient-centricity • Involvement of patients from the earliest stages in the development of new endpoints and digital measures is key to ensure relevance and practicality. • Ensure proportionality in generating data for the development and validation of a new endpoint in a clinical trial, balancing the burden of the patients participating in the trial with the benefit of developing a new relevant endpoint. Suggested ways forward to address the challenges • Build on the experience of the CHMP Qualification Opinion for a digital endpoint in Duchenne Muscular Dystrophy (SV95c), as well as other examples, regarding evidentiary requirements and processes as well as scientific and technical learnings in the development of digital endpoints in related diseases. • Stakeholders should engage in early regulatory dialogue using the available support platforms: EMA support to SMEs, EMA Innovation Task Force briefing meetings, EMA Scientific Advice for product specific methodology development, EMA Qualification of Novel Methodologies. • To accelerate the development and validation of digital endpoints in a learning eco-system, the pre-competitive collaboration of stakeholders needs to be increased, including data sharing by clinical trial sponsors and secondary use of clinical trial data. Therefore, it is important to make optimal use of public-private partnerships and support the development of novel pre- competitive collaboration frameworks for the development, validation and qualification of ACT EU multi-stakeholder workshop on methodology guidance – workshop report 12 digital endpoints that encourage the translation of new technology into clinical research in practice. • Improve the alignment between Regulatory Agencies, HTA bodies and Notified bodies within Europe and harmonise regulatory requirements internationally. • Increase the clarity of existing regulatory pathways for digital endpoint qualification and consider process improvements, in particular on the need for Digital Health Technologies to be certified as medical devices by Notified Bodies based on the MDR/IVDR and documents needed for clinical trial applications. • Provide guidance on the evidentiary requirements for the qualification of digital endpoints for use in clinical trials. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 13 Session E Beyond RCTs Moderators: Elke Stahl (BfArM), Antoine Vanier (HTA, HAS France); Frank Petavy (EMA) Presenters: Mouna Akacha (Novartis), Denis Lacombe (EORTC), Pierre Henri Bertoye and Claire Bahans (EC France) and Antoine Vanier (HTA, HAS France) The session opened with the industry perspective on clinical trial methodology beyond RCTs, followed by an ethics view and an academic perspective on synthetic and external controls for clinical trials. The introduction concluded with an HTA perspective on non-randomised studies as source of clinical evidence. Key challenges identified by stakeholders • Non-RCT designs are generally associated with an increased risk of bias and reduced confidence in the reliability of results. • Non-RCT designs are generally associated with higher methodological complexities, such that a strengthened dialogue between decision-making entities could be beneficial. • A critical consideration for the acceptability of non-RCT designs is the need for strong knowledge about the counterfactual (i.e., what the clinical outcome would have been, had a patient not taken the treatment), which is a challenge in almost all therapeutic indications. • Experience and knowledge are constantly evolving, which complicates the specification acceptance criteria for non-randomised designs. • Optimised use of the accumulated knowledge is impeded by the limited sharing of data across stakeholders. • Stakeholders expressed uncertainty regarding the required level of evidence across decision- making entities, and in which situations non-RCT designs could be considered acceptable. A challenge which was acknowledged in this regard was that the amount of information which can be presented to decision-makers will evolve over time. How to improve patient-centricity • When developing a methodological framework to guide the dialogue on the acceptability of non-RCT designs, include considerations related to the specific target indication (such as medical knowledge about the counterfactual). • Foster and facilitate the conduct of multi-national trials for overcoming perceived obstacles of trials conducted in small patient populations. Suggested ways forward to address the challenges • Develop a framework to systematically guide the dialogue between sponsors, regulatory bodies (NCAs and Ethics) and HTAs on the acceptability of non-RCT designs. • Foster enhanced collaboration and data sharing across stakeholders for optimised use of accumulated medical knowledge. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 14 Session F Patient-centricity, diversity and representativeness in clinical trials Moderators: Anneliene Jonker (World Duchenne Organization / UPPMD) and Mårten Wendt (CTCG) Presenters: Louise Veltrop-Duits (Central Committee on Research involving Human Subject (CCMO)), Tarec Christoffer El-Galaly (Aalborg University Hospital and Danish Medical Research Ethics Committees), Mireille Muller (Novartis) and Michal Rataj (European Patients Forum). The session opened with an introduction to patient engagement and its importance. The second presentation on Diversity and inclusion in clinical trials focused on scientific needs and ethical obligations. This was followed by a presentation of an industry perspective on methodological priorities on patient centricity, inclusion and representativeness and, lastly, some proposals for making research more patient-driven. Key challenges identified by stakeholders • Unclear expectations about clinical trial participation can lead to dropout during trial conduct and follow-up. • From a patient perspective, potential clinical trials are difficult to identify, reducing the accessibility of trials. • Trial results are rarely communicated in a timely manner to participants, or it is difficult for patients to locate them. How to improve patient-centricity • Patient-centricity should be a general guiding principle in the design and conduct of clinical trials. • Patients should be involved early on in the design of clinical trials. Suggested ways forward to address the challenges • Develop guidance on the operationalisation of the CTR requirement describing patient involvement in clinical trial designs. • Use Real-World Evidence to complement insights from clinical trials. • Ensure that patients are adequately compensated for their time and input into clinical trial design and methodology. • Clarify inclusion and exclusion criteria and their rationale in clinical trials. • Align on terminologies, especially on diversity and representativeness in clinical trials. • Increase clarity on expectations of trial participation by providing adequate information to patients. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 15 Session G Decentralised elements in clinical trials Moderators: Monique Al (CTCG), Wolfgang Berdel (Association of Medical Ethics Committees, DE) Presenters: Christine Dehn (German Heart Foundation), Mira Zuidgeest (University Medical Center Utrecht), Alison Bond (Amgen), David Wright (AstraZeneca) The session opened with a patient perspective on use of decentralised elements in clinical trials (DCTs), emphasising that the main barrier to successful trial conduct is awareness of the trials with decentralised elements. An academic perspective on DCTs was then presented, promoting a change in perspective from the site-based approach as the ‘gold standard’ to the use of decentralised elements as a normal approach in clinical trials. The perspective from EFPIA emphasised the lack of harmonisation across Member States and the lack of clarity on acceptability and validation of digital/remote endpoints as the key challenges. Key challenges identified by stakeholders • Despite the enthusiasm, clinical trial applications with decentralised elements are still sparse. There is uncertainty on the regulatory acceptability of data generated by DCTs for marketing authorisation. • Awareness of planned and ongoing DCTs among patients is lacking. It is a major challenge to inform patients about the possibilities to take part in a DCT, which is not being conducted by the already treating physician. How to improve patient-centricity • Introduce flexibility for patients to participate onsite or remotely may improve the possibilities for patients to participate in trials but may also pose operational and statistical challenges. Clear guidance on how to mitigate the additional challenges can help to realise the benefits of decentralised trial elements for patients. • Ensure reimbursement for trial related tests and/or services not performed onsite but with local health care providers. • Implement adequate procedures to ensure equal importance of patients using decentralised and centralised elements. Suggested ways forward to address the challenges • More clarity on and faster validation of digital/remote endpoints. • Increase awareness among investigators and patients on the concept of decentralised elements in clinical trials and explore better use of the local health care practitioners in the conduct of clinical trials. • More case studies/examples are needed to develop best practices and facilitate harmonisation of requirements across Member States. Ongoing data and knowledge exchange between Member States from applications on DCT elements is important. • Increase awareness of the existing recommendations published by the EMRN: “Recommendation paper on Decentralised elements in Clinical Trials”. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 16 Session H Complex trials II Moderators: Elina Asikanius (FIMEA), Olga Kholmanskikh (FAMHP), Benjamin Hofner (PEI) Presenters: Saskia Litiere (EORTC), Tobias Mielke (Johnson & Johnson), Beate Wieseler (IQWiG) The session opened with an introduction to the regulatory activities related to platform trials and summary of the status of the Reflection Paper on platform trials. Thereafter, an academic organisation’s perspective on complex clinical trials was presented, covering operational and methodological challenges. The EFPIA perspective on platform trials with a focus on opportunities and challenges, particularly in relation to the EMA concept paper on platform trials followed. Lastly, an HTA perspective was presented on how pre- and post-approval platform trials across different new interventions can facilitate the assessment of relative effectiveness. Key challenges identified by stakeholders • Conceptualising and operationalising collaboration and sponsorship in multi-stakeholder platform trials investigating products from different competing developers, who may have differing standard operating procedures and legal requirements. • Lack of sponsor experience, resulting in perceived higher risk for delays and (operational) study failure for investigational interventions. • Confidence in regulatory acceptability of various design elements, including: o Definition and use of type-I-error in platform trials across study arms. o Choice of comparator and use of non-concurrent controls; need for guidance on conditions for acceptability of using non-concurrent controls. o Partial unblinding of ongoing trial arms after analysing and publishing information on earlier closed trial arms, which makes information on the shared control arm available while other arms are still ongoing. o Definition of the population in a platform trial and impact on the randomisation, e.g. in biomarker-driven designs. o Response-adaptive randomisation in platform trials with registrational intent. • Challenges in initiating and conducting platform trials under the CTR and registration via CTIS: o Deciding on the optimal submission strategy, either with a single trial application approach (with the platform trial as a single trial) or a multiple trial application approach (with each sub-protocol as individual trials) and anticipating their consequences. o Complexity of using CTIS, especially with a lack of functionalities specific to platform trials. o The sequential handling of trial amendments via CTIS and related waiting times conflict with the dynamic addition or modification of sub-studies in platform trials. o Transition of previously initiated complex clinical trials to the CTR. How to improve patient centricity • Accurate and well-described staged process for informed consent. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 17 • Broad exploration of optimal indication in a targeted development (e.g. exploring various biomarkers or combinations thereof, including biomarker positive and negative patients). • Early ideation of platform trials within indication(s) to facilitate comparisons for downstream decision-making for HTAs, caregivers and patients, by inclusion of as many of the relevant products developed in the respective disease as possible. Suggested ways forward to address the challenges • Collaborative and iterative multi-stakeholder dialogue exploring and defining the requirements for the design of a platform trial and facilitating early information sharing on lessons learned and acceptable design elements between various developers. • Guidance on the regulatory acceptability of methodological aspects of platform trials, to be read in conjunction with the Q&A on complex clinical trials. • Sponsors collaborate as co-sponsors or third-party organisations act as sponsors in multi- developer platform trials. Important aspects to consider in this setting are the governance structure of the platform trial, experience of the organisation with conducting complex trials, data sharing, data quality and accountability of the applicant in regulatory submissions. • Clarifications on clinical trial application, approval and conduct processes and procedures for clinical trials that are considered or labelled ‘complex’, including functionalities in CTIS. • Identification of novel pilot cases for platform trials in situations with medical need, limited competitive nature and existent regulatory requirement for the study conduct (e.g. paediatric studies) to generate and share experience in design and conduct of cross-company platform trials. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 18 Closing remarks Chairs: Monique Al (CTCG), Kit Roes (MWP), Peter Arlett (EMA), Florian Lasch (EMA) To facilitate better, faster and optimised clinical trials in the EU, clinical trial methodology is essential. Therefore, guidance on clinical trial methodology provided by the European Medicines Regulatory Network facilitates the implementation of innovative approaches. Patients are at the core of clinical development and successfully incorporating patient’s needs and perspectives is key for feasible clinical trials that generate fit-for-purpose evidence. The ACT EU multi-stakeholder workshop on methodology guidance has brought together experts representing all relevant stakeholders of clinical trials in the EU. In line with ACT EU’s vision to increase collaboration and coordination across the EU, key topics related to clinical trial methodologies were discussed from a multi-stakeholder perspective. The discussions identified challenges and needs related to guidance development, as well as proposals for how these can be addressed. As a next step, focussed follow-up discussions and further workshops will be needed to consider the outcomes from the break-out sessions. The workshop report will be disseminated publicly and presented to relevant expert groups in the European Medicines Regulatory Network to acknowledge the challenges identified by all stakeholders and to consider the proposed ways for improving patient centricity and addressing the stakeholder challenges for inclusion in their workplans. ACT EU multi-stakeholder workshop on methodology guidance – workshop report 19 Glossary ACT EU Accelerating Clinical Trials in the EU CHMP Committee for Medicinal Products for Human Use CTCG Clinical Trials Coordination Group CTIS Clinical Trials Information System CTR Clinical Trials Regulation DCT Decentralised Clinical Trial DKMA Danish Medicines Agency EMA European Medicines Agency EMRN European Medicines Regulatory Network EORCT European Organisation for Research and Treatment of Cancer EFPIA European Federation of Pharmaceutical Industries and Associations EFSPI European Federation of Statisticians in the Pharmaceutical Industry FAMHP Federal Agency for Medicines and Health Products FIMEA Finnish Medicines Agency HMA Heads of Medicines Agencies HTA Health Technology Assessment IQWiG Institut für Qualität und Wirtschaftlichkeit im Gesundheitswesen IVDR In Vitro Diagnostic Regulation MDR Medical Device Regulation MS Member State MWP Methodology Working Party NCA National Competent Authority PEI Paul-Ehrlich-Institut PJS Patients’ Journey Studies RCT Randomised Controlled Trial SME Small and Medium-sized Enterprise ACT EU multi-stakeholder workshop on methodology guidance – workshop report 20 More information The Accelerating Clinical Trials in the EU (ACT EU) initiative aims to develop the European Union further as a competitive centre for innovative clinical research. ACT EU seeks to deliver on the clinical trial innovation recommendations of the European medicines agencies network strategy and the European Commission’s Pharmaceutical strategy for Europe. ACT EU builds on the Clinical Trials Regulation and Clinical Trials Information System launched on 31 January 2022. The European Commission, EMA and Heads of Medicines Agencies launched ACT EU in January 2022 and run the initiative together, establishing a steering group in March 2022. The programme's strategy paper features ten priority action (PA) areas that are the basis for the ACT EU workplan; an eleventh PA on clinical trials in public health emergencies has been created since the launch of the programme. The ACT EU workplan was published on 10 November 2023 and sets out deliverables and timelines for the programme for 2023-26. The deliverables for 2024 include: • The implementation of the Clinical Trials Regulation, including support for the transition of clinical trials to the CTR and the CTR Collaborate project, which aims to optimise collaboration between national health authorities and national ethics bodies. • The creation of a regulatory helpdesk for non-commercial sponsors conducting multi- national clinical trials. • Creation of the Multi-Stakeholder Platform Advisory Group of stakeholder representatives. • A scientific advice pilot to provide consolidated advice for clinical trial and marketing authorisation applications. • Regulatory support to clinical trials in public health emergencies. European Medicines Agency Domenico Scarlattilaan 6 1083 HS Amsterdam The Netherlands Telephone +31 (0)88 781 6000 Send a question www.ema.europa.eu/contact www.ema.europa.eu ACT EU multi-stakeholder workshop on methodology guidance – workshop report EMA/105992/2024 © European Medicines Agency, 2024. Reproduction is authorised provided the source is acknowledged.
accelerating-clinical-trials.europa.eu
October 30, 2025 at 12:39 PM
Nieuws van ACT EU: Recommendation paper on decentralised elements in clinical trials
RECOMMENDATION PAPER ON DECENTRALISED ELEMENTS IN CLINICAL TRIALS
Recommendation paper DCT, V02, 1 October 2025 1 Version 02, 1 October 2025 Document history and effective date: Date of endorsement by CTAG 15 October 2025 Supersedes: Version December 2022 Changes compared to superseded version: - Front page includes a statement about cross- border trials with decentralised elements - The national provision overview has been updated Date come into effect Date of publication 29 October 2025 Important notice: The views expressed in this recommendation paper on decentralised elements in clinical trials in the European Union/European Economic Area are not legally binding. Ultimately, only the European Court of Justice can give an authoritative interpretation of Community law. This recommendation paper is restricted to decentralised elements in general. For cross-border trials with decentralised elements, it is the responsibility of the sponsor to check the national requirements for cross-border trials and contact the national contact point of the Member State if authorisation of the clinical trial is needed in home country of trial participant. Recommendation paper DCT, V02, 1 October 2025 2 Recommendation paper on decentralised elements in clinical trials TABLE OF CONTENT ABBREVATION LIST ............................................................................................................. 3 1. INTRODUCTION, SCOPE AND GENERAL CONSIDERATIONS .............................................. 4 General considerations ................................................................................................ 5 2. CLINICAL TRIAL OVERSIGHT: ROLES AND RESPONSIBILITIES ......................................... 8 Considerations on responsibilities ................................................................................. 8 Considerations on keeping oversight on incoming data .................................................... 9 3. INFORMED CONSENT PROCESS ................................................................................. 11 Informed consent interview ....................................................................................... 11 Digital information leaflet .......................................................................................... 12 Informed consent signature ....................................................................................... 12 4. DELIVERY OF INVESTIGATIONAL MEDICINAL PRODUCTS AND ADMINISTRATION AT HOME14 Considerations on IMP delivery direct to trial participants .............................................. 14 Considerations on IMP storage and administration at the trial participant’s home ............. 15 5. TRIAL RELATED PROCEDURES AT HOME ..................................................................... 17 6. DATA COLLECTION AND MANAGEMENT INCL. DEFINING AND HANDLING SOURCE DATA .. 18 7. TRIAL MONITORING ................................................................................................. 19 APPENDIX: NATIONAL PROVISIONS OVERVIEW .................................................................... 20 Recommendation paper DCT, V02, 1 October 2025 3 ABBREVATION LIST AE Adverse Event CRF Case Report Form CTCG Clinical Trial Coordination Group CTEG Clinical Trial Expert Group CTR Clinical Trial Regulation EC European Commission EEA European Economic Area EMA European Medicines Agency EMRN European Medicines Regulatory Network ePRO Electronic Patient Reported Outcome EU European Union GCP Good Clinical Practice GDP Good Distribution Practice GMP Good Manufacturing Practice GDPR General Data Protection Regulation HMA Heads of Medicine Agencies ICH International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use IMP Investigational Medicinal Product IT Information Technology IVD In Vitro Diagnostic IVDR In Vitro Diagnostics Regulation IWG Inspector Working Group MDR Medical Device Regulation MS Member State SAE Serious Adverse Event SAWP Scientific Advice Working Party SNSA Simultaneous National Scientific Advice Recommendation paper DCT, V02, 1 October 2025 4 1. INTRODUCTION, SCOPE AND GENERAL CONSIDERATIONS Clinical trials on Investigational Medicinal Products (IMPs) are increasingly using procedures conducted outside the traditional ‘clinical trial site’, a concept usually referred to as decentralisation. In addition, there is increasing use of digital tools within clinical trials. The COVID-19 pandemic highlighted the importance and usefulness of digital tools and decentralised procedures in a healthcare setting and in clinical trials. The guidance on the management of clinical trials during COVID-19 pandemic provided a set of recommendations that included adjustments to the informed consent process, the distribution of IMPs and in monitoring under specific circumstances. This guidance is specific to the COVID-19 health crisis in the European Union (EU)/European Economic Area (EEA) and is intended to be revoked when there is a consensus that the period of the COVID-19 outbreak in the EU/EEA has passed. The above context and trend highlight the need to provide further recommendations on the introduction of decentralised elements in the conduct of clinical trials in the EU/EEA, regardless of any health crisis, and in consideration of the currently limited national guidances. The aim of this recommendation paper is to address this requirement. The intention is to facilitate the use of decentralised elements in clinical trials in the EU/EEA. However, the necessary level of trial participant’s safety, protection of their rights and dignity should be ensured. In addition, the reliability of data for publication and submission for regulatory decision-making should be guaranteed. It is acknowledged that certain decentralised elements in clinical trials have been adopted for some time and that not all of these elements are likely to have a significant impact on scientific validity, data integrity, benefit-risk ratio or the protection of trial participants’ rights. If a decentralised element has been identified as a critical-to-quality factor as defined in International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) E8, a risk-proportionate approach should be followed and adapted to the risk of trial participants, trial integrity of the research carried out and to the risk related to reliability of trial results. This is in line with the Recommendations on risk proportionate approaches in clinical trials from the expert group on clinical trials for the implementation of Regulation (EU) No 536/2014. The recommendation paper will address the roles and responsibilities of the sponsor and investigator, electronic informed consent, IMP delivery, trial related procedures at home, data management and monitoring in a decentralised clinical trial setting. An overview of the current national provisions applicable in each Member State in relation to these topics is outlined in the appendix. It should be noted that the national provision appendix is for guidance purposes only as it is not feasible to give a complete overview of all scenarios for implementing decentralised elements in a clinical trial. It is at the discretion of the Member State involved in the assessment of a clinical trial whether the use of certain decentralised elements is acceptable in a specific clinical trial. Sponsors are encouraged to seek scientific advice via the European Medicine Agency [EMA, scientific advice working party (SAWP)], or via national competent authorities [national or simultaneous national scientific advice (SNSA)] regarding the use of specific decentralised elements, especially on decentralised elements where experience and the evidence of their impact may be limited. Sponsors may also request a consolidated opinion via the Clinical Trial Coordination Group (CTCG) for regulatory issues of general impact not related to a specific trial. This recommendation paper was created as part of the priority action 8 ‘Methodology guidances’ of the ACT EU initiative of the European Commission (EC), the Heads of Medicines Agencies (HMA) and the EMA. It was drafted in a collaboration between the HMA Clinical Trial Coordination Group (CTCG), EC Clinical Trial Expert Group (CTEG) and the EMA GCP Inspectors Working Group (GCP- IWG). It includes broad perspectives from the European medicines regulatory network (EMRN) as well as perspectives by patient and health care professional representatives. Given the rapid Recommendation paper DCT, V02, 1 October 2025 5 advances in the field of decentralised clinical trials, the paper is expected to evolve when new insights and experiences are gained. General considerations Clinical trials with medicinal products have already adopted many decentralised elements such as electronic diaries, wearables, phone calls and online appointments. How decentralised elements are used in clinical trials depends on many factors including the type of clinical trial, the trial population, the disease being treated, the condition of the trial participant, the type of medicinal product, its characteristics and development stage. These elements should be considered individually and in combination when planning for and implementing the use of decentralised elements. In addition, the following general considerations should be taken into account: • The rights, safety, dignity and well-being of the trial participants1 should be protected and prevail over all other interests. The implementation of decentralised elements in the conduct of a clinical trial should not result in increased risks to the safety, rights, and well-being of trial participants. The appropriateness of decentralised elements depends in particular on (but not limited to) the specific trial population, its disease, the type of assessment, the characteristics of the investigational medicinal product(s), including its/their stage of development and thus the current knowledge about its/their efficacy and safety profile. ▪ Adherence to EU and national applicable laws, regulations and established standards and guidances for clinical trials (e.g. Clinical Trial Regulation: CTR EU no 536/2014, ICH E6, ICH E8, applicable Good Manufacturing Practice (GMP) provisions, applicable Good Distribution Practice (GDP) principles) and international ethical and scientific principles of medical research (e.g. Declaration of Helsinki) is required for all clinical trials regardless the use of decentralised elements. Particular emphasis should be placed on compliance with the General Data Protection Regulation (GDPR EU no 2016/679). • Sponsors and investigators should engage potential trial participants, patients or patient organisations in a meaningful participatory process that involves them in an early and sustained manner in the design, development and implementation of the clinical trial. Early participant involvement in the design of the clinical trial is likely to increase scientific value. It may help develop trust in the trial, facilitate recruitment, and promote adherence. Patients also provide their perspective of living with a condition, which may contribute to the choice of decentralised elements, for example, the feasibility of appointments by videoconference instead of a physical visit, the use of digital tools, or the measurements of endpoints that are meaningful to patients and selection of the appropriate population. • When developing a clinical trial with decentralised elements, investigators/healthcare professionals should be involved in the design, development, and implementation of the clinical trial. The expertise of the investigators/health care professionals may contribute to ensure clinically relevant objectives and endpoints, efficient safety monitoring and adequate medical care. They can also contribute to identify the consequences of having less personal contact or how to manage data collection and the quality and integrity of the (source) data. • Any transfer of burden of trial related procedures to trial participants and/or investigators should be weighed against the potential benefits of using decentralised elements in the clinical trial. The sponsor may provide adequate support to trial participants and/or investigators to facilitate the appropriate conduct of their tasks. 1 Where patient/trial participant is mentioned in the paper, relatives and/or legal representatives of patient/trial participant are meant as well, whatever is applicable. Recommendation paper DCT, V02, 1 October 2025 6 • For transparency reasons, and to facilitate the assessment of the clinical trial by authorities and ethics committees, a summary of the decentralised elements planned in the clinical trial should be provided in the cover letter of the clinical trial application. • If it is determined that decentralised elements are likely to have a significant impact on scientific validity, data integrity, benefit-risk ratio or impact on the protection of trial participants’ rights, these should be considered in a specific and documented risk benefit assessment. This risk benefit assessment as well as any risk mitigation action taken should be clearly described in the clinical trial protocol or other protocol related document as part of the clinical trial application to the Member State. This is required for any element impacting the risk benefit assessment. • In clinical trials with decentralised elements, parts of the clinical trial may be conducted outside the traditional patient care centers, with the involvement of service providers. General medical rules to protect patient’s/trial participant’s safety should be upheld in trials with decentralised elements especially when patients/trial participants are separated from traditional patient care centers. Among those is the assessment of individual patient’s risk profile, including appropriate anamnestic information, physical examination and laboratory or imaging data by a responsible investigator with the required trial population specific medical background. Exceptions should be justified in the clinical trial application to ensure appropriate case-by-case review. • The sponsor should provide in the clinical trial application a description of the funding of the clinical trial and any other (financial) arrangements between funder, investigator and service providers involved in the conduct of the clinical trial. Any conditions, such as economic interests and institutional affiliations, that might influence the impartiality of the investigator should be provided as well, as would be expected for any trial. • Trials with decentralised elements should be designed to generate reliable and robust data. Regarding regulatory decisions supporting marketing authorisation, the data is required to meet the same expectations as those from trials with on-site procedures. Sponsors should carefully discuss expected challenges prospectively and clarify how they plan to address potential limitations introduced by decentralised elements in advance to ensure scientific quality of the clinical trial. The following are examples: o potential differences between the study population and target population which may trigger discussion on the generalisability of the results (e.g. due to potential exclusion of digitally illiterate persons or people who live in areas with limited internet connection). o imposed modifications in outcome assessments which may trigger a discussion on their validity (e.g, due to heterogeneous implementation of decentralised procedures across clinical trials sites or among trial participants). o the potential increase in missing data, overall or for specific endpoints. See also chapter 6 on data management. These considerations are of utmost importance especially in trials identified as pivotal in marketing authorisation applications. Sponsors are strongly encouraged to seek scientific advice for these trials. In addition, qualification advice is encouraged when new methods or endpoints are planned to be used. • IT devices / technologies which are developed and utilised should be fit for the purpose of reliable data collection and handling in accordance with the protocol. The use of computerised systems and/or the creation/capture of electronic clinical data, should be compliant with the ‘Guideline on computerised systems and electronic data in clinical trials’2. 2 Guideline on computerised systems and electronic data in clinical trials, EMA, GCP IWG, 2023 Recommendation paper DCT, V02, 1 October 2025 7 • A contingency plan should be in place to minimise the impact of any risk, for example malfunction of a digital tool or disruption of a planned decentralised visit, for identified critical- to-quality decentralised elements. • When medical devices, including in-vitro diagnostics (IVDs), are used in the clinical trial, their use should be compliant with the applicable medical device legislation, such as the Medical Device Regulation (MDR) EU no 2017/745, the In Vitro Diagnostic Directive 98/79/EC and/or the In Vitro Diagnostic Regulation (IVDR) EU no 2017/746. The following chapters outline more specific considerations regarding the decentralisation of certain clinical trial aspects. Recommendation paper DCT, V02, 1 October 2025 8 2. CLINICAL TRIAL OVERSIGHT: ROLES AND RESPONSIBILITIES When parts of the clinical trial are conducted off-site, and when additional service providers such as home nurses or providers of technology become involved, it is essential that the specific roles and responsibilities of the sponsor, investigator, and any additional parties are clearly defined and understood prior to the start of the trial. In addition, when trial participants are visiting the clinical trial site less frequently, alternate methods of clinical monitoring of the trial participants’ current health status and related data collection may need to be utilised. Data may be received from different routes, for example collected at home by the participants themselves, by visiting (external) healthcare professionals, or by digital tools. This poses a challenge to the oversight on the rights, safety, dignity and well-being of the trial participants as well as the reliability of trial results. As a general concept, introducing decentralised elements should be considered as an extension of the clinical trial site with the inclusion of the trial participants’ home, resulting in an additional obligation of oversight for investigators and sponsors. It is therefore important that, when decentralised elements are implemented, it is ensured that the investigator and sponsor still can fulfil their legal obligations as laid down in the CTR and ICH E6. In addition, with a potential increase in the number of parties involved in the clinical trial, adherence to the GDPR needs to be safeguarded. The protocol should reflect that the sponsor and the investigator are in full control of their respective areas of responsibilities at all times, e.g. with respect to the data processing, the communication flow, and ultimately the rights, safety, dignity and well-being of the trial participants and reliability of the trial data. In this section considerations in relation to investigator and sponsor oversight are outlined. Considerations on responsibilities • Notwithstanding the potential involvement of additional service providers, the clinical trial specific tasks as described in the protocol are ultimately the responsibility of either the investigator or the sponsor, in accordance with ICH E6. Great care should be taken that the delegation of tasks to the different parties is well defined. The introduction of decentralised elements in a clinical trial may have a relevant impact in the trial conduct, therefore, it should be clearly documented which tasks are conducted when, by whom, and in which setting (e.g. at the clinical site, at the trial participant’s home, etc.), and how the required oversight by the sponsor and/or supervision by the investigator is achieved. The general overview of the workflow of these different tasks and actions to be taken within the trial should be described in the protocol, and in more detail in a protocol related document. • In case service providers have been delegated trial specific tasks, a corresponding rationale and the extent of their involvement should be described in a high level in the protocol, and in detail in a protocol related document. The investigator retains the ultimate responsibility for tasks involving trial related medical decisions (i.e. trial participant eligibility and enrolment, protocol specified medical procedures, changes in medication, etc.) and for the rights, safety, dignity and well-being of the trial participants. See also the appendix for current national provisions regarding the involvement of external health care providers. • Any trial specific task that is delegated to a service provider should be specified in a written agreement between the responsible party for the task (according to ICH E6) and the service provider (see also the EMA GCP IWG Q&A B.2, and B.8). When the sponsor selects a service provider and the investigator is not involved in the contractual arrangement with this service provider, the contract between the sponsor and the investigator should clearly document the contractual arrangements with the service provider if it concerns tasks under investigator’s Recommendation paper DCT, V02, 1 October 2025 9 responsibility. This allows the investigator to agree or not to the deployment of service providers for certain trial specific tasks related to the medical care of trial participants. As stated previously, in the general considerations of this paper, it is recommended that the investigator should be involved in an early stage when designing the decentralised elements in the clinical trial. In this way it can be assessed early on what are the needs of the investigator with regard to the use of service providers for trial specific tasks that fall under the responsibility of the investigator. • The sponsor should ensure that the contracted service provider is qualified and experienced in the tasks they conduct for the trial. This should be reflected in the contract between the sponsor and the investigator, in order that the investigator is aware of, and can agree or not with the qualification of the service provider when the delegated tasks lie within the investigator’s responsibility. The investigator should have the possibility to ask for any additional information in order to perform due diligence and to require any change to the agreement or to the service when considered necessary, including the possibility to reject a certain service provider. It is the investigator’s responsibility to ensure that the service provider is properly trained on the trial specific tasks they have to conduct, when these tasks concern the medical care of the trial participants or lie within the investigator’s responsibility. • To maintain the investigator’s responsibility regarding the medical care and safety of the trial participant and to ensure that the sponsor has adequate oversight over the conduct of the clinical trial effective lines of communication should be established, documented and shared with all relevant parties, including trial participants, investigators, sponsor and any service providers. All parties involved should have access to the information required to fulfil their roles and responsibilities related to the conduct of the clinical trial at any time. In case of an emergency, an effective communication plan needs to be in place, so that all relevant parties can act without undue delay. The trial participant should be well informed and receive contact details for all necessary situations including who to contact for acute cases, but also for device failures, questions on home visits, etc. Considerations on keeping oversight on incoming data • Trial participants, investigators and service providers involved in the trial should receive training on how to use the digital tools employed in the trial, to ensure proper data collection, review, and transmission. In addition, the trial participants and service providers should receive training on what is considered an (serious) adverse event ((S)AE), who they should report this to, in what timeframe, and how to manage the (S)AE. • When AEs are reported via several routes (digital tool, external healthcare professional, or trial participant) it is important that procedures are in place to identify potential duplicates. • The use of digital tools (such as wearables) result in an increase in the amount of incoming data. This may challenge the capacity of the investigators to fulfil their responsibilities. Emerging data could be continuously at hand, and a clear procedure should be in place to determine how to handle this constant flow of information. The review frequency of the incoming data by the investigator should be based on the relevance of the data to the safety and well-being of the trial participant, and the relevance of the data for the efficacy. The review of safety data should be planned with a risk-based perspective, which may include the IMP safety profile, the indication, known potential risks, the use of notifications and alerts. Recommendation paper DCT, V02, 1 October 2025 10 The priority is to capture and assess SAEs in a timely manner, without creating an unacceptable burden for the investigator and/or the trial participant. The use of notifications and alerts is recommended to ensure timely assessment of SAE related data. In designing a trial with digital tools, the sponsor and investigator should anticipate what kind of safety alerts may occur and specify in the protocol how these alerts will be handled. If it is foreseen that a digital tool may generate critical safety data that needs immediate medical attention, a plan should be in place describing this. It should be outlined in the protocol how the investigator and/or the service provider should manage these situations, what actions should be taken and by whom. A schematic overview of parties involved, information flow and respective duties is recommended. The trial participant should be informed what to expect and what actions they may need to follow in these situations. In addition, a participant targeted scheme of the duties and information flow with the parties involved might enhance understanding. • The sponsor should ensure that digital tools are transmitting the required alerts as planned. The tool that generates alerts to the investigator should be validated. A risk mitigation plan should be in place for times that the tool may not work as intended. • The trial participant should be fully informed in advance on how the information transmitted via digital tools, for example electronic Patient Reported Outcomes (ePROs), will be acted upon. It should be made clear to the trial participant that the investigator may not review such data in real time, and that if the trial participant experiences any specific safety concern they need to directly contact the investigator to report such an issue. Recommendation paper DCT, V02, 1 October 2025 11 3. INFORMED CONSENT PROCESS An important aspect of a clinical trial is that the potential trial participants give their voluntarily informed consent to participate. To give consent, the potential participant needs adequate information. Informed consent is not only of ethical and legal importance: good communication between the investigator and the trial participant is beneficial for mutual trust and may promote trial compliance. Therefore, when considering the appropriateness of conducting the informed consent process in a remote manner, to use digital information leaflets, and/or to use electronic methods for the signature of the informed consent form, several aspects have to be thoroughly assessed. These include the design of the clinical trial, the characteristics of the trial population, and the risks, burdens and potential benefit related to participating in the clinical trial. The entire procedure for obtaining informed consent, i.e. the selection, the evaluation of the eligibility, and the actual informed consent process, should be described step-by-step in the clinical trial application to ensure appropriate ethical review. The rationale for not having a physical examination as part of this procedure should be given in the protocol or other protocol related document. The sponsor should also describe in the protocol the chosen method for obtaining informed consent. Regardless if only a part of or the whole informed consent process is conducted remotely, the process should still be carried out in compliance with the principles laid down in the CTR, ICH E6, the GDPR and national legislation. The informed consent process should be documented in a manner that allows verification of the receipt of information by the trial participant, the discussion between the person qualified to obtain the consent and the trial participant, as well as giving of the consent. Informed consent interview ICH E6 requires that all potential trial participants are fully informed on the clinical trial and are given the opportunity to ask questions. In general, this should be a physical meeting between the investigator and the potential trial participant. However, in some cases it can be justified that this is done remotely. The more vulnerable the trial population, the more limited the current knowledge of the efficacy and safety profile of the IMP(s), the more complex the trial concept and the higher the risks associated with the trial-specific interventions, the more necessary is a physical meeting between trial participant and investigator for the purpose of informed consent. In case the potential trial participant is not visiting the clinical trial site, the following aspects should be considered and addressed in the clinical trial application: • As part of the process of obtaining informed consent, it is considered essential that face-to- face communication takes place between the potential trial participant and the investigator, or a qualified person designated by the investigator. If this discussion is done in a digital/virtual meeting, it is recommended that this takes place in real time where the parties can both see and communicate with each other via audio and video. The remote face-to-face contact should allow for asking questions and the investigator should make every effort to check the identity of the participant if they are not already known by them, and conversely, the participant should have the right to ask for proof of the investigator’s identity if they have not been in contact before. Deviation from (remote) face-to-face communication should be justified in the clinical trial application, together with a description of how the verification of the identity of the investigator and the trial participant will be performed in such cases, and how it will be determined that the trial participant has understood the information. See also the appendix for the current national provisions. • The sponsor should ensure that trial participants and investigators are given the option to Recommendation paper DCT, V02, 1 October 2025 12 have the informed consent interview on site if this is preferred by the trial participant or the investigator. However, in duly justified cases only the remote option may be offered. • Individual participant related factors affecting the use of decentralised elements of the clinical trial should be evaluated by the investigator during the informed consent interview. • The reliability and confidentiality of the method used should be ensured. As a general principle, the communication channel used for the informed consent interview should be encrypted to protect the confidential information that will be discussed. Digital information leaflet • The use of different kinds of media may enhance the trial participant’s comprehension of the trial. However, when considering the use of electronic methods, the sponsor should also be aware that its use may unintentionally discriminate against participants who cannot or prefer not to use such technology. Alternative methods for the electronic provision of information should be available. There may be exceptions where the sponsor only provides a digital information leaflet. In such circumstances, this should be described in the protocol and justified in the clinical trial application. • The sponsor is responsible to verify whether a clinical trial site and/or the data protection officer of that site agrees to the use and storage of electronic methods for the consent process. • It should be ensured that the information provided to the trial participant is in a form that can be stored and retrieved by the trial participant. Informed consent signature • There are various ways of obtaining a signed informed consent form by remote means. This includes for example a paper consent form sent to the participant signed with a ‘wet ink signature’ and sent back by post, or a digital consent form signed with an electronic signature, i.e. completely digital. Regardless of the format of the informed consent, the method should allow reconstruction of the process, including the validity of the signatures. The sponsor should ensure that the systems used have proportionate security levels and that safeguards regarding confidentiality are in place. In general, the electronic signature functionality should be in accordance with the requirements described in the ‘Guideline on computerised systems and electronic data in clinical trials’ 3. In addition, the method used to record informed consent should follow national requirements with regards to acceptability of electronic signatures (see appendix for current national provisions). • When using electronic methods, the trial participants should be able to download an electronic copy of the signed and dated informed consent form, or to receive a print-out of the electronic copy. If an electronic copy, it should be protected against modification; any modification should invalidate the signatures. • Existing procedures related to re-consent should be adapted to the use of electronically signed • Procedures should be in place to handle follow-up steps after the consent has been withdrawn 3 Guideline on computerised systems and electronic data in clinical trials, EMA, GCP IWG, 2023 Recommendation paper DCT, V02, 1 October 2025 13 electronically, including partial withdrawal and complete withdrawal, due to the impact on patient participation and data collection. These procedures should include timely notification to the investigator and a communication plan with all other stakeholders. By any means, withdrawals should also be possible outside of the system, and this should be recorded by the investigator. Recommendation paper DCT, V02, 1 October 2025 14 4. DELIVERY OF INVESTIGATIONAL MEDICINAL PRODUCTS AND ADMINISTRATION AT HOME Where it is intended for the IMP4 to be delivered and/or administered at the trial participant’s home, a risk assessment should be completed to determine if such an approach is appropriate. The risk assessment should at a minimum take into account the following aspects: the knowledge and uncertainty of the IMP and its safety profile, the route of administration, the trial population, whether an observation period is required, the need for emergency plans, the preparation of the final IMP for administration, its stability, the storage conditions, and the robustness of IMP delivery logistics (the risk of an inadvertently IMP delivery to a non-intended recipient). The CTR aims to harmonise the rules of the conduct of clinical trials in the member states, while setting high standards of quality and safety of IMPs to ensure the protection of public health. Therefore, the import of IMPs into the EU requires an authorisation (CTR article 61), and the applicable principles of GDP should be considered in the logistics of IMPs. Shipping and the contractual agreements regarding IMP shipment between sponsor and investigator site or pharmacy are covered by the ‘Guideline on the responsibilities of the sponsor with regard to handling and shipping of investigational medicinal products for human use in accordance with Good Clinical Practice and Good Manufacturing Practice’. IMP delivery to the trial participant is not, however, within the scope of that guideline. In this section considerations are given concerning the delivery of the IMP and the administration at the trial participant’s home. Considerations on IMP delivery direct to trial participants • If the IMP is not dispensed to the trial participant by the investigator or delegated healthcare professional at the site, it is recommended that the vendors responsible for delivery to the participant are authorised to distribute or dispense medicinal products as much as possible. Any non-authorised vendor used in the logistics should be qualified and supervised by the authorisation holder, in accordance with the principles of GDP. There must be a written contract which clearly establishes the duties of each party. It is recommended that the number of separate transportation steps are minimised. • The investigator remains responsible for the decision of treatment which should be documented (for example prescription or Interactive Response Technology system) prior to any delivery of IMP to the trial participant’s home. Delivery to the participant’s home could mean another suitable address the participant prefers to receive the IMP at, provided that: o regulatory requirements are complied with; o risks of exposure to conditions that could impact quality and integrity of the product are minimised; o the applicable principles of the guidelines on GDP of medicinal products for human use are taken into consideration. When the given address is abroad, it should be verified whether the national legislation of that country allows the IMP to be delivered there (see appendix on national provisions). The given address should also be the place where the IMP is stored and administered, to avoid additional transport by the trial participant themselves. • There are several options for delivery of the IMP to the trial participant’s home, depending on what is permitted by national requirements. This can include delivery from the pharmacy of the investigator site, from a delegated pharmacy, or from a depot. The sponsor has the overall 4 The recommendations for IMP delivery and administration at home also apply to auxiliary medicinal products (AxMP). Recommendation paper DCT, V02, 1 October 2025 15 responsibility for the process and the contracts or agreements, which should reflect the principal investigator’s responsibilities pursuant to ICH E6. Please refer to the appendix for the acceptable options in the member states regarding the delivery of the IMP to the trial participant’s home. The arrangements for delivery of the IMP to the trial participant should be described in the clinical trial protocol or the Investigational Medicinal Product Dossier. • The sponsor should ensure that the personal data of the trial participants required for the delivery of the IMP is used in accordance with the GDPR on a need-to-know basis. For example, it should be ensured that personal data is solely accessible to those involved in the delivery of the IMP and will not be stored for other purposes than the delivery of the IMP. Access to the personal data should be restricted as soon as the final delivery is completed. Information should be made available only for the purpose of monitoring, auditing, inspections, and to trial participants for the exercise of their GDPR rights. • Trial participants should be made aware during the informed consent process that their contact details will be used for delivery purposes if the IMP is to be delivered to the trial participant’s home. Details regarding the use of contact information should be outlined in the participant • When delivering the IMP, it should only be handed over to the trial participant (or a representative, if applicable), or the present health care professional involved in the clinical trial. Sponsor procedures should be in place covering delivery and receipt of the IMP. With regard to receipt of IMP, the procedure should detail the steps and responsibilities in relation to confirmation of IMP identity (e.g. batch number) to ensure that what has been dispatched has actually been delivered. In some cases, the trial participant (or a representative) may not be available to accept and sign the receipt of the IMP. In this case, the IMP should be brought back by the service provider to the original location (investigator’s site, (central) pharmacy, or depot). • As an alternative to delivering the IMP to the trial participant’s home, the IMP could be dispensed by local pharmacies (based on a prescription issued by the investigator), provided that the labelling requirements in the CTR are fulfilled, and if national requirements allow (see appendix). In particular, sponsors are reminded to consult Article 61(5) of CTR regarding labelling requirements. The local pharmacy should be aware that the prescription of the IMP is in the context of a clinical trial, and if necessary, be trained to dispense the IMP. Considerations on IMP storage and administration at the trial participant’s home • The sponsor and the investigator should consider during the planning stage of the clinical trial how the appropriate storage conditions of the IMP can be met, and whether the IMP is suitable for administration at home. The inclusion/exclusion criteria should include provisions related to the adequacy of the trial participant’s home for storage of the IMP, such as temperature control and restricted access where necessary. Sponsors may consider providing trial participants additional equipment necessary for IMP storage. This should be described in the protocol or other protocol related document (e.g. pharmacy manual), including the documentation provided to participants. The investigator should give instructions to the trial participants on the use and storage of the IMP. The instructions should be realistic, feasible and the additional burden for the trial participant should be part of the aforementioned risk assessment. • The investigator and the sponsor should consider whether administration at home can be done by the trial participants themselves or if a trained, experienced and qualified healthcare professional is required for administration. In the case of complex administrations, special preparation or handling requirements, or when required by the safety profile of the IMP (e.g. unknown or potential serious adverse events in connection to the administration), health care professionals should always be involved. Recommendation paper DCT, V02, 1 October 2025 16 • Generally, if an IMP is required to be administered by a healthcare professional, shipping directly to the trial participant may not be appropriate. In the event it is required for IMP to be shipped directly to the trial participant separately, clear instructions for storage of the IMP prior to healthcare professional visit should be given, as well as a clear explanation that the IMP is not to be administrated before the visit of the healthcare professional nor before the investigator’s decision. • If it is anticipated that the trial participants will prepare and administer the IMP as outlined in the protocol, they should be instructed in advance about these aspects. Where appropriate, there should be instructions provided regarding these steps as well as dosing, in addition to what is already present on the IMP label or package leaflet. These instructions should be adapted to the needs of the individual trial participants. The use of electronic step-by-step instructions which are easily and promptly accessible (such as QR code scanning), could be considered. Depending on the safety profile of the IMP, the investigator should contact the trial participants after the first delivery of the IMP to ensure proper handling of the IMP. The sponsor may consider providing additional equipment necessary for the safe administration, use and destruction of the IMP to the trial participants, in which case this should be described in the protocol or other protocol related document (e.g. pharmacy manual), including the documentation provided to the participants. • The investigator should follow-up, at regular intervals, with participants to ensure the IMP is taken appropriately and according to the IMP instructions. • Procedures should be in place for IMP accountability and treatment compliance of trial participants. These tasks fall under the investigator’s responsibility according to ICH E6. • Procedures should be in place for IMP return from the trial participant’s home, and destruction of the unused IMPs, in compliance with the protocol and local safety requirements. The procedure should also cover recalls during the conduct of the trial, and the steps taken to avoid that the IMP remains at the trial participant’s home beyond the envisaged treatment period. Recommendation paper DCT, V02, 1 October 2025 17 5. TRIAL RELATED PROCEDURES AT HOME In a clinical trial with decentralised elements, trial related procedures may take place outside of the clinical trial site, such as in the trial participant’s home5. These procedures could be performed by the trial participant, the investigator staff visiting the trial participant at home, or a person contracted for the trial and delegated to perform them. For these procedures or examinations performed at home, considerations include, but may not be limited to the following: • The investigator should ascertain whether the trial participant's home situation and premises are suitable to have trial related procedures performed at home. It should be considered that there may be personal/social circumstances which could exclude home visits. • Inclusion/exclusion criteria should include provisions related to the adequacy of the trial participant’s home for critical trial related procedures at home. The trial participant should be informed during informed consent process about trial procedures planned to take place at home. • Performing trial-related procedures at home should only be done if the procedures do not cause additional risk to trial participant or reliability of the data and the person performing the task is qualified and/or trained to perform the task. For example, if biological samples are collected at home, it should be considered whether persons taking the sample are qualified and allowed by legislation to take the sample. In addition, adequate handling and storage conditions for the samples throughout the entire process should be assured. • In the event of trial participants performing trial related tasks it should be ensured that appropriate training is provided to them, and any additional trial participant burden duly considered, including tasks related to digital data collection. • The sponsor and/or investigator should ensure that appropriate guidance and training is provided to the delegated person(s) to conduct the tasks at home correctly. • The insurance or indemnity or a guarantee or a similar arrangement foreseen by CTR should be in place to cover any damage resulting from trial related procedures performed at home. • The investigator should monitor compliance of the trial participant considering the lack of/decrease in the number of face-to-face visits/meetings between the trial participant and the investigator and/or delegated staff. • The trial participants should be given the opportunity to visit the investigator in person if needed/preferred and they should be able to have a direct contact line if further support to perform a trial related task/collect data is needed. • There should be procedures in place for reporting and management of adverse events noticed by the trial participant or by any delegated person during home visits (see also chapter 2 about considerations on maintaining oversight on incoming safety data). • The sponsor should provide alternatives if a trial participant is unable or not willing to use her/his/their own private device (mobile phone, tablet, etc.) to capture trial data. 5 A trial participant’s home can be more than one home, e.g. children with parents that are separated. Recommendation paper DCT, V02, 1 October 2025 18 6. DATA COLLECTION AND MANAGEMENT INCLUDING DEFINING AND HANDLING SOURCE DATA Decentralised clinical trials are characterized by an extensive shift of data collection from the investigator/investigator site to the trial participants and/or their caregiver and/or service providers (e.g. home nurses). Direct data capture by electronic systems (e.g. electronic Case Report Forms (CRFs), ePROs, wearables etc.) may occur, for instance, at the clinical trial site or off-site locations. According to ICH E6, the data recorded during the clinical trial should be credible, reliable and verifiable. In addition, the data protection requirements according to the GDPR should be adhered to (see also chapter 1, general considerations). Utilising multiple systems and parties adds complexity and requires an adequate oversight and implementation of adequate measures by the sponsor. To this end, the sponsor should: • Ensure that all parties involved in the clinical trial have an overview of the data flow; a data flow diagram with additional explanations in the protocol is highly recommended. • Ensure that the used data acquisition tools are configured and validated in accordance with their intended use. • Determine the type and scope of the trial participants’ personal data to be collected and ensure adequate protection in compliance with the GDPR of such personal data in any step of the process. • Ensure that when source data captured by a data acquisition tool is transferred to another location and subsequently irreversibly deleted from the data acquisition tool, both the data and the metadata are transferred (see ICH E6 1.63 Certified Copy). • Implement measures such as encryption to minimise the risk of unauthorised access, when transferring the data from a data acquisition tool to a server. • Ensure access to trial data is controlled by defined user rights and methods of access for all relevant parties involved. Unauthorised access should be prevented using appropriate security measurers e.g. firewalls. • Ensure control of and continuous and complete access by the investigator to both source data generated either on-site or off-site as well as source data reported to the sponsor (e.g. central lab data). The risk of erroneous data entry for data measured and entered directly by trial participants, especially on primary, key-secondary or safety endpoints should be minimised by appropriate Additional advice on elements specific to digital data capture systems can be found in the ‘Qualification opinion on eSource Direct Data Capture (DDC) (EMA/CHMP/SAWP/483349/2019)’ from the EMA Scientific Advice Working Party (SAWP) and the ‘Notice to sponsors on validation and qualification of computerised systems used in clinical trials (EMA/INS/GCP/467532/2019)’. In addition, reference is made to the EMA GCP matters Q&A B3 ‘How and where should source data be defined’ as well as to the EMA GCP matters Q&A B5 ’What are the expectations of the investigator’s copy of the CRF when using a web based application’. Recommendation paper DCT, V02, 1 October 2025 19 7. TRIAL MONITORING Trial monitoring is part of the quality control processes in clinical trials. • As detailed in ICH E6, the monitoring strategy should be based on the specifics of a clinical trial. These specifics may include, as applicable, decentralised processes and tools described in the previous sections. For example, if according to the trial protocol, safety and/or efficacy data are collected via ePRO or wearables, or if key processes (e.g. those related to primary endpoints) are performed outside the investigator site (e.g. at central reading facilities, central laboratories), the specific risks associated with these decentralised processes, tools, locations, and individuals involved should be taken into account in the monitoring strategy. • Monitoring procedures can be divided into centralised and site monitoring, and generally a combination of them both is appropriate. Site monitoring is usually performed on-site. Depending on its purpose and suitability it may be performed off-site (remotely). • When establishing remote access for the purpose of monitoring, the principles of necessity and proportionality should always be adhered to. The monitoring strategy chosen should not unduly burden the site. • If remote access to source data and documents is foreseen, additional measures with respect to confidentiality of data access and security of the systems should be in place. Further guidance on this is being drafted by the GCP IWG. See appendix with current national provision overview per Member State whether remote access to medical records by the monitor or auditor is allowed. Recommendation paper DCT, V02, 1 October 2025 20 APPENDIX: NATIONAL PROVISIONS OVERVIEW This overview of national provisions does not purport to be an interpretation of law and/or regulations and is for guidance purposes only. The answers to the questions stated in the national provision overview are giving by the individual Member States and relates to the context and general recommendation as provided in the recommendation paper. References for the relevant sections within the recommendation paper are given in the question header. Please note that footnotes from each Member State are given in the tables following the national provision overview. The footnotes provide legislation reference, background or conditions for a ‘No’ or a ‘Yes’. Recommendation paper DCT, V02, 1 October 2025 21 Please see relevant footnotes for responses marked with an asterisk. A footnote may be raised even though no response is given. AT BE BG CY CZ DE DK EE EL ES FI FR HR HU IE IS IT LI LT LU LV MT NL NO PL PT RO SE SI SK The delivery of IMPs from sponsor/site, in relation to RP section 4. Q1: Is it possible to deliver IMPs directly to trial participants from their associated trial site? No * No * Yes * No * Yes * Yes* Yes Yes * Yes * Yes * No * Yes Yes * Yes Yes * Yes * Yes * Yes * Yes * Yes Yes Yes * Yes Yes * No * Yes Q2: Is it possible to deliver IMPs directly to trial participants from the pharmacy associated with the trial site? No * No * Yes * No * Yes * Yes* Yes No * * Yes * Yes * * No * Yes Yes * Yes No * No * Yes * Yes * Yes * Yes Yes Yes * No * Yes * * Yes Q3: Is it possible to deliver IMPs directly to trial participants from any delegated pharmacy? No * No * No No Yes * Yes Yes * No * No * No * No * Yes No * Yes Yes * Yes * No * No * Yes * No Yes * Yes No * Yes * No * Yes * No * Yes Q4: Is it possible to deliver IMPs directly to trial participants from the IMP manufacturer with a MIA license? No No * No No No * No * No * No * No * No * No * No * Yes No * Yes * No * No Yes * No No No * No * Yes * No No No * No Q5: Is it possible to deliver IMPs directly to trial participants from the trial sponsor (sponsors intermediaries/depots)? If yes, footnote states if a licence is required for the depot to carry out this task and how to obtain this licence. No No * No No No * No * No * No * No * No * No No * No Yes * Yes * No No Yes * No No No * No * No * Yes * No * No The shipment of IMPs from sponsor/site across boarders within the EU, in relation to RP section 4. Q6: Is it possible to deliver IMPs directly to trial participants from e.g. distribution/manufacturing/pharmacy licence holders located in other EU MSs if legally allowed to carry out this task in the country of origin? No * No * No No No * No* Yes No * No * No * No * No * No * Yes Yes * Yes * No * No Yes No No * No * No * No * Yes No * No * No Q7: Is it possible to deliver IMPs directly to investigators from e.g. distribution/manufacturing/pharmacy licence holders located in other EU MSs if legally allowed to carry out this task in the country of origin? Yes * Yes * No * Yes Yes* Yes Yes Yes * No * No * Yes * Yes * Yes Yes * * Yes No Yes Yes No * No * No * Yes * Yes No * No * Yes Recommendation paper DCT, V02, 1 October 2025 22 Labelling of IMP, in relation to RP section 4. Q8: Is it possible for any delegated pharmacy to label IMP or is this restricted to the pharmacy associated with the trial site? No No * No * Yes * * Yes * Yes * No * No * No * Yes Yes * Yes Yes * No No * No * No * No No Yes * * Yes Recommendation paper DCT, V02, 1 October 2025 23 Please see relevant footnotes for responses marked with an asterisk. A footnote may be raised even though no response is given. AT BE BG CY CZ DE DK EE EL ES FI FR HR HU IE IS IT LI LT LU LV MT NL NO PL PT RO SE SI SK The shipment and hand-out of IMPs from pharmacies. This is currently not included in the recommendation paper but may be relevant in next version of the RP. Q9: Is it possible to deliver or dispense authorised IMPs directly to trial participants from pharmacies not associated with the clinical trial sites? This include authorised investigational medicinal products not used according to their SmPC. Yes * No * No No No * Yes Yes * No * No * No * No * No * No * Yes Yes * Yes * No * No Yes No Yes * Yes * No * No * No * Yes * * Yes * Q10: Is it possible to deliver or dispense non-authorised IMPs directly to trial participants from pharmacies not associated with the clinical trial sites? No No * No No No * Yes No* No * No * No * No * No * No * Yes No Yes * No * No No Yes * Yes * No * No * No * Yes * * Yes * The eConsent process, in relation to RP section 3. Q11: Is a physical face to face meeting between the trial participant and the PI or a member of the research team always mandatory during the consent procedure (even if the rest is conducted remotely)? No No Yes * Yes No * Yes * No* * * No * No No * Yes * Yes * No No * No Yes No No No * No No * Yes * No No * No Q12: Is it possible to use electronic signatures instead of wet ink? If yes, please specify in the footnotes which eIDAS category is expected for the electronic signature. Yes * Yes * Yes * Yes Yes * Yes * Yes * Yes * * Yes * Yes * Yes * Yes * Yes * Yes Yes * Yes * No Yes * Yes Yes * Yes * Yes * Yes * Yes * Yes * * Yes * Trial participant oversight and home visits, in relation to RP section 2 and 5. Q13: Is it possible for the PI to delegate tasks under their responsibility to a qualified (for the delegated task) external healthcare provider? Yes Yes * Yes * * Yes * Yes * Yes Yes * Yes * Yes * Yes Yes * Yes Yes * Yes Yes Yes * No Yes Yes Yes * Yes * Yes * Yes * Yes * Yes * Yes Yes Q14: Certain tasks/procedures carried out at home may require supervision of the investigator (a physician). Is it allowed for the physician to supervise remotely? Yes Yes * Yes * * No * Yes * Yes * * * Yes * Yes * Yes * Yes * Yes Yes Yes * * Yes Yes * Yes Yes * Yes * No * Yes * * No Recommendation paper DCT, V02, 1 October 2025 24 Trial Monitoring using remote access to source data, in relation to RP paper section 7 Q15: Is remote access to the medical records allowed by the monitor or auditor? Yes * No * Yes * * Yes * Yes * Yes * * No * Yes Yes * No * Yes * Yes * Yes * Yes * No * Yes Yes * * * Yes * No No No * No Recommendation paper DCT, V02, 1 October 2025 25 Footnotes to the DCT Provision Overview by Member States BE Q1 Not allowed currently, unless specified in the CTA why a waiver should be authorized, referring to the Q&A n°10: https://www.ema.europa.eu/en/human-regulatory/research-development/compliance/good-clinical-practice/qa-good-clinical- practice-gcp [Change in national legislation ongoing] Q2 Same approach as in Q1 and according to the RD of 21 January 2009, a pharmacy in Belgium must deliver each medication in person to a patient, with the exception in art. 29 of medication free of prescription. [Change in national legislation ongoing] Q3 According to the RD of 21 January 2009, a pharmacy in Belgium must deliver each medication in person to a patient, with the exception in art. 29 of medication free of prescription. [Change in national legislation ongoing] Q4 According to the RD of 21 January 2009, a pharmacy in Belgium must deliver each medication in person to a patient, with the exception in art. 29 of medication free of prescription. An investigator can also provide the trial participant in person with an amount of medication (IMP). [Change in national legislation ongoing] Q5 Same as Q4 Q6 Same as Q4 Q7 According to art. 43 of the RD 9/10/2017 Q8 Labelling is a manufacturing operation. Labelling is only possible if the site/pharmacy has a GMP licence. Q9 Only a delegated pharmacy (delegated by the PI) can deliver or dispense IMPs. Q10 Only a delegated pharmacy (delegated by the PI) can deliver or dispense IMPs. Q11 - Q12 According to the national guidance on e-ICF, for remote signing, only an advanced or a qualified electronic signature as defined in the eIDAS regulation (Ref. 3) should be used as they uniquely identify the individual signing. Only a qualified electronic signature has the equivalent legal effect of a handwritten signature (eIDAS, art 25. §2.). Signatures via e-ID (Ref. 4) or itsme® (Ref. 5) are qualified electronic signatures. The advanced signature should comply with the defined requirements as described in the article 26 of the eIDAS Regulation that give guarantees of the identification of the individual signing. More information is available on the website of the FPS Economy (Ref. 6). References are available in https://consultativebodies.health.belgium.be/en/e- ICF%20guidance%20Belgium_30-09-2020 Q13 Provided that the delegated tasks fall under the qualification of the study personnel according to the Belgian legislation. Q14 Provided that the delegated tasks fall under the qualification of the study personnel according to the Belgian legislation. Q15 Remote source data verification is as such not allowed. This is only possible in specific cases, approved during the CTA process under the following conditions: - An agreement has been setup describing rSDV which is approved by all parties (institution, principal investigator and the sponsor or the CRO assigned). - The rSDV can be organized by the investigator’s site and is therefore technically feasible without compromising the confidentiality of the Electronic Medical Records data. AT Q1 §57, §59(1), (9) AMG (Austrian Medicines Act) Change in national legislation/guidelines is ongoing Q2 §57 AMG (Austrian Medicines Act), exception: Authorised or registered, non-prescription medicinal products could be delivered directly to patients as per §59(10) AMG (Austrian Medicines Act). Change in national legislation/guidelines is ongoing Q3 §57 AMG (Austrian Medicines Act), exception: Authorised or registered, non-prescription medicinal products could be delivered directly to patients as per §59(10) AMG (Austrian Medicines Act). Q4 - Q5 - Q6 §57 AMG (Austrian Medicines Act), exception: Authorised or registered, non-prescription medicinal products could be delivered directly to patients as per §59(10) AMG (Austrian Medicines Act). Q7 §57, §59(1), (9) AMG (Austrian Medicines Act) Q8 - Q9 (For non-prescription medicinal products only), packaging and labelling must not be changed, IMP must be from trial stock Q10 - Q11 - Q12 The use of advanced and qualified electronic signatures is accepted. Integrity and authenticity of the signature must be undisputable. Q13 - Q14 - Q15 Allowed for original electronic medical records only. The electronic medical record system must be validated for that purpose. Recommendation paper DCT, V02, 1 October 2025 26 BG Q1 It is possible, but it will be assessed on a case by case basis with respect to the character of the particular IMP and the medical condition. Q2 It is possible, but the pharmacy must be part of the approved trial site. It will be assessed on a case by case basis with respect to the character of the particular IMP and the medical condition. Q3 - Q4 - Q5 - Q6 - Q7 - Q8 - Q9 - Q10 - Q11 Face to face meeting between the trial participant and the PI or a member of the research team is always mandatory during the consent procedure. Q12 It is possible to electronically sign the Informed consent form (ICF), as the only permissible method is using qualified electronic signature (оnly qualified electronic signature issued by a licensed provider), replacing the handwritten signature but without fully digitalizing the entire informed consent process. Q13 It is possible, but it will be assessed on a case by case basis. In case that clinical trial activities will be carried out outside the trial site, the location outside the site shall be indicated with the name of the medical institution, the structure therein and the exact address of the activity, as well as the names and qualifications of the relevant person carrying out the activity, where applicable. The information should be present in the Site Suitability Form and in the ICF. Home visits are generally not allowed, except in limited cases after assessment. Q14 - Q15 in certain cases- Information should be provided and assessed. CY Q1 Any exemption is subject to a justified request and a case-by-case assessment and approval. Q2 Any exemption is subject to a justified request and a case-by-case assessment and approval. Q3 - Q4 - Q5 - Q6 - Q7 IMP delivery to investigational sites is possible upon a case-by-case assessment and approval. Q8 Not addressed in the national pharmaceutical legislation. Case by case assessment and approval. Q9 - Q10 - Q11 - Q12 - Q13 Case-by-case assessment. Comprehensive documentation and adherence to GCP guidelines is expected. Q14 Case-by-case assessment. Comprehensive documentation and adherence to GCP guidelines is expected. Q15 Case by case assessment. Comprehensive documentation and adherence to GDPR rules and GCP guidelines is expected. Recommendation paper DCT, V02, 1 October 2025 27 CZ Q1 It is possible, but it will be assessed on a case by case basis with respect to the character of the particular IMP and its pharmaceutical form (tablets, infusion etc.) It would not be possible in case of IMPs that need to be diluted or reconstituted before administration, because these operations have to be carried out by healthcare professionals appointed by the healthcare service provider (Section 79 (10) of Act no. 378/2007 Coll., on Medicinal Products). The sponsor must then proceed in accordance with national guideline VYR-44, available on SUKL’s website. Q2 It is possible if the pharmacy is closely connected with the trial site. It is possible then to deliver IMPs directly to trial participants from the pharmacy based on investigator´s request. But it will be assessed on a case by case basis with respect to the character of the particular IMP and its pharmaceutical form (tablets, infusion etc.) It would not be possible in case of IMPs that need to be diluted or reconstituted before administration, because these operations have to be carried out by healthcare professionals appointed by the healthcare service provider (Section 79 (10) of Act no. 378/2007 Coll., on Medicinal Products). The sponsor must then proceed in accordance with national guideline VYR-44, available on SUKLS’s website. Q3 It is possible if the pharmacy is connected with the trial site per contract. It is possible then to deliver IMPs directly to trial participants from the pharmacy based on investigator´s request. But it will be assessed on a case by case basis with respect to the character of the particular IMP and its pharmaceutical form (tablets, infusion etc.) It would not be possible in case of IMPs that need to be diluted or reconstituted before administration, because these operations have to be carried out by healthcare professionals appointed by the healthcare service provider (Section 79 (10) of Act no. 378/2007 Coll., on Medicinal Products). The sponsor must then proceed in accordance with national guideline VYR-44, available on SUKLS’s website. Q4 In general, the trial site pharmacy or PI is responsible for IMP handling. (Section 75 (5) and section 77 Act no. 378/2007 Coll., on Medicinal Products). Also, in line with the GCP the sponsor must not know trial participant's identification. Q5 In general, the trial site pharmacy or PI is responsible for IMP handling. (Section 75 (5) and section 77 Act no. 378/2007 Coll., on Medicinal Products) Also, in line with the GCP the sponsor must not know trial participant's identification. Q6 In general, the trial site pharmacy or PI is responsible for IMP handling. (Section 75 (5) and section 77 Act no. 378/2007 Coll., on Medicinal Products) Q7 - Q8 Delegated pharmacy can label IMP, but it must be treated contractually in case that delegated pharmacy and trial site are not the same legal entities. Q9 In general, the trial site pharmacy or PI is responsible for IMP handling. (Section 75 (5) and section 77 Act no. 378/2007 Coll., on Medicinal Products) Q10 In general, the trial site pharmacy or PI is responsible for IMP handling. (Section 75 (5) and section 77 Act no. 378/2007 Coll., on Medicinal Products) Q11 But in accordance with Act no. 378/2007 Coll., on Medicinal Products, an investigator must always be a physician and only the investigator is responsible for dialogue with the participant during the consent procedure. Q12 In our view, it is possible to use a qualified electronic signature in accordance with Regulation (EU) no. 910/2014 of the European Parliament and of the Council of 23 July 2014 on Electronic Identification and Trust Services for Electronic Transactions in the Internal Market and Repealing Directive 1999/93/EC (the „eIDAS regulation“). However, given that the acceptability of electronic signatures concerns all EU member states and the approach should be harmonised, we recommend to confirm with the European Commission should there be any differing views. Q13 Home care is possible in the CZ, but with certain limits – please see more information on the website here https://sukl.gov.cz/en/en-reg-kh-doplnujici-informace/home-care-3/ Q14 Due to safety of patients. Supervision of a physician is necessary due to situations where potential action from a physician would be required in case of any urgent emergency (risk procedure etc.) In such case remote supervision can put the participant in danger (connection loss, misunderstanding in communications etc.) Q15 It is only possible in trial sites, where all documentation is in electronical form in validated IT systems. But as the medical records are currently available in the paper form at the trial sites, the remote access is not allowed. With the ongoing digitization of healthcare systems at the trial sites, the remote access to the medical data will be reviewed case by case. Recommendation paper DCT, V02, 1 October 2025 28 DE Q1 Not subject to CT legislation Q2 Certain restrictions for e.g. hospital pharmacies may apply. Q3 - Q4 In principle: Not allowed. According to Section 47 (1) sentence 1 number 2 letter g German Medicinal Product Act pharmaceutical entrepreneurs and wholesalers may only supply pharmacy only medicinal products only then directly and only to hospitals and doctors if the medicinal products are labelled "Intended for clinical trials", provided they are supplied free of charge. Exceptions: According to an exemption valid until 31. December 2023, the NCA may, by way of derogation from Section 47 (1) sentence 1 number 2 letter g AMG, permit pharmaceutical entrepreneurs (also: sponsors) and wholesalers to make medicinal products labelled for clinical trials available free of charge to participants in a clinical trial if, after an assessment to be carried out by the sponsor on a case-by-case basis, the safety of the persons participating in the clinical trial and the validity of the data collected in the clinical trial are guaranteed and the pseudonymisation of the data is ensured by appropriate measures that the trial participants have the right to contact the sponsor. This should be reflected in the protocol Q5 Q6 See explanation to Q4; only for pharmacy licence holders Q7 Section 47 (1) sentence 1 number 2 letter g and section 73 (2) number 2 German Medicinal Product Act. Q8 It is crucial, that the manufacturer or the pharmacy has a manufacturing license Q9 Q10 Q11 Face to face meeting not subject to German Medicinal Product Act. According to Model Professional Code for Physicians of the German Medical Association exclusive counselling or treatment via communication media is permitted in individual cases if this is justifiable from a medical point of view and the required medical care is observed, in particular by the way in which the findings are ascertained, counselling, treatment and documentation are carried out and the patient is also informed about the special features of exclusive counselling and treatment via communication media. Q12 Only possible, when qualified electronical signature (eIDAS) Q13 Possible in principle, but medical activities must be carried out by a physician Q14 „In principle, yes, subject to the qualification requirements under national law for the medical personnel to whom a task is assigned; a number of medical tasks may according to national law only be performed by physicians (with a medical license to „In principle, yes, subject to the qualification requirements under national law for the medical personnel to whom a task is assigned; a number of medical tasks may according to national law only be performed by physicians (with a medical license to practice medicine Q15 In principle, yes, provided that the investigators can and does comply with their obligation to maintain the confidentiality of his patients' health records; therefore, the responsibility for answering this question is with the respective investigator DK Q1 - Q2 - Q3 Yes, for hospital pharmacies. Q4 A framework is being developed and will be provided in national guidance. Q5 A framework is being developed and will be provided in national guidance. Q6 - Q7 - Q8 - Q9 If dispensed and then delivered according to normal prescription practice and taken by pharmacy standard stock of medicinal products. Q10 - Q11 The physical face-to-face meeting is the primary expectation, but video-based communication can be accepted in certain situations, as per decision of the ethical committee. Q12 Currently both NemID (OCES standard) and MitID (eIDAS-compliant) is accepted. Q13 - Q14 - Q15 Please consult the DKMA DCT guidance for specific requirements if utilised for rSDV. Recommendation paper DCT, V02, 1 October 2025 29 EE Q1 - Q2 Not allowed for hospital pharmacies Q3 The conditions and procedure for the issue of prescriptions for medicinal products and for the dispensation of medicinal products by pharmacies and the format of the prescription Q4 Medicinal Products Act Q5 Medicinal Products Act Q6 Medicinal Products Act Q7 - Q8 Re-labelling is restricted to associated pharmacy Q9 The conditions and procedure for the issue of prescriptions for medicinal products and for the dispensation of medicinal products by pharmacies and the format of the prescription Q10 The conditions and procedure for the issue of prescriptions for medicinal products and for the dispensation of medicinal products by pharmacies and the format of the prescription Q11 If justified. Remote authentication and facial recognition. Q12 Qualified electronic signature (Smart ID, Mobile ID and ID card) Q13 - Q14 Case by case Q15 Remote SDV not allowed EL Q1 Direct shipment to the patient is not addressed in CT legislation. In general, the trial site Head of hospital pharmacy or PI is responsible for IMP handling. Ministerial Decree O.J. 4131/2016, Art 6. Q2 Direct shipment to the patient is not addressed in CT legislation. In general, the trial site Head of hospital pharmacy or PI is responsible for IMP handling. Ministerial Decree O.J. 4131/2016, Art 6. Q3 In general, the trial site Head of hospital pharmacy or PI is responsible for IMP handling. Ministerial Decree O.J. 4131/2016, Art 6. Q4 In general, the trial site Head of hospital pharmacy or PI is responsible for IMP handling. Ministerial Decree O.J. 4131/2016, Art 6. Q5 In general, the trial site Head of hospital pharmacy or PI is responsible for IMP handling. Ministerial Decree O.J. 4131/2016, Art 6. Q6 In general, the trial site Head of hospital pharmacy or PI is responsible for IMP handling. Ministerial Decree O.J. 4131/2016, Art 6. Q7 Yes, however IMPs are shipped to the trial site (not to specific investigators). Q8 - Q9 In general, the trial site Head of hospital pharmacy or PI is responsible for IMP handling. Ministerial Decree O.J. 4131/2016, Art 6. Q10 In general, the trial site Head of hospital pharmacy or PI is responsible for IMP handling. Ministerial Decree O.J. 4131/2016, Art 6. Q11 Although not specifically addressed in CT legislation, a face-to-face meeting is implied. Ministerial Decree O.J. 4131/2016, Art. 9. In addition, the National Ethics Committee interpretation is that a face-to-face meeting is required for the provision of Informed Consent. Q12 In general, electronic signatures are acceptable in Greece. However, CT specific legislation does not address the issue. The National Ethics Committee requires wet ink signatures. Q13 Ministerial Decree O.J. 4131/2016, Art. 3 Q14 Not specifically addressed in current CT legislation Q15 - Recommendation paper DCT, V02, 1 October 2025 30 ES Q1 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ) (however this activity should be managed by the Pharmacy Department [unless the site has no Pharmacy]). Q2 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ) Q3 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ). It is covered by Autonomous Communities legislation (e.g. Law 19/1998 the organization of pharmaceutical services in Madrid) Q4 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ) Q5 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ) Q6 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ). It is covered by Autonomous Communities legislation (e.g. Law 19/1998 the organization of pharmaceutical services in Madrid) Q7 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ). Except if the trial site has no Pharmacy Department. Q8 Regulation 536/2014 article 61.5 a). If the delegated pharmacy is taking part in the same clinical trial in the same Member State. Q9 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ) Q10 Real Legislative Decree 1/2015 article 3.6 c) and Royal Decree 1090/2015 article 39.3 ñ) Q11 Physical face to face is not specifically established by any national provisions. Q12 If high level of eIDAS and if the confidentiality of the personal data, data security and secure access to the data is ensured. See national Guidelines for undertaking decentralised items in clinical trials Q13 If an adequate investigator oversight and proper contractual arrangements between sponsor, trial site and investigator is ensured. Data protection aspects should also be considered. Q14 Certain tasks/procedures should be defined in order to provide proper assessment. General Data Protection Regulation (GDPR) should be addressed in this regard. Q15 There are no specific national provisions; GDPR covers this aspect in the entire EU and should be considered. FI Q1 - Q2 In special and justified circumstances only Q3 Medicinal Act §15 b. Fimea Administrative Regulation 2/2016 Q4 Medicinal Act §15 b. Medicinal Act §31 Q5 Medicinal Act §15 b Q6 Medicinal Act §17. Medicinal Act §15a Q7 Medicinal Act §17 Q8 Medicinal Act §15a Q9 Fimea Administrative Regulation 2/2016 Q10 Fimea Administrative Regulation 2/2016 Q11 - Q12 The national Client Data Act 703/2023 8 and 22§: the participant should be identified reliably (authentication. for example suomi.fi or respective) and the signature method must be advanced or qualified. Q13 - Q14 - Q15 - Recommendation paper DCT, V02, 1 October 2025 31 FR Q1 The shipment to patient home by PUI (hospital pharmacy) are allowed by 'retrocession' (article L. 5126-6 CSP). For experimental drugs in the context of a CT and in absence of any legal specification, ANSM and Ethics Committees may accept it. Q2 The shipment to patient home by PUI is allowed by 'retrocession' (article L. 5126-6 CSP). For experimental drugs in the context of a CT and in absence of any legal specification, ANSM and Ethics Committees may accept it. Q3 - Q4 In accordance with art R 5124-2 and -3 of CSP Q5 - Q6 As per French legislation. Q7 Yes, but under conditions for the shipping to the investigators as per article L 5126-7 of CSP. PUIs are allowed to provide investigators of the same CT or professionals with similar activities outside of France (article R 5124-4 of CSP), with experimental drugs. In the same way dispensers of experimental drugs who are based outside of France, may be allowed to provide French investigators of the same CT with experimental drugs. However, these deliveries should be in conformity with articles L5121-108 and L5124-13 of CSP and customs code (importation authorization if clinical trial is not already authorized). Under these conditions the ANSM may agree. The labelling should be in French language. Q8 The community pharmacy is not allowed to label experimental drugs. The labelling of experimental drugs should be performed either by a pharmaceutical entity with a GMP manufacturing authorization, or by a PUI pharmacy with authorization of preparing products needed for clinical trials (article R.5126-9 CSP). Q9 Not possible for a PUI pharmacy in a hospital that is not involved in the given clinical trial (article L. 5126-1 CSP). Possible for a community pharmacy under conditions of article D. 5125-45-1 of CSP - if specified in the protocol, the ANSM may agree. Q10 Not possible (article L. 5126-1 and D. 5125-45-1 of CSP) Q11 Not mandatory Q12 Based on the following legal references: - Article L1122-1-1 alinéa 1st of CSP & article 1367 of Civil code. - The European regulation (UE) number 910/2014 of European Parlement and of Counsil of 23 juillet 2014 concerning the electronic identification does distinguish three types of electronic signatures: the simple electronic signature, the advanced signature (article 26 of regulation) and qualified signature. The e-consent is legal (via these 3 categories of signature) but not yet used in France under this scope. Q13 Provided that the delegation is appropriately planned in the study protocol and validated by competent authorities. This possibility allows to involve the private physicians who work outside the hospitals in clinical studies. Q14 As of today, it is not forbidden in the regulation. As a consequence, it is allowed for a given trial provided that there is no opposition from ANSM or from the Ethics Committees. Q15 Fulfilling the recommendations of the CNIL (French DPA) about regulation and sensitive data protection. cf. provisional recommendations during the Covid crisis (https://www.cnil.fr/fr/recommandations-provisoires-controle-qualite- essais-cliniques-crise-sanitaire)” Recommendation paper DCT, V02, 1 October 2025 32 HR Q1 The IMP must be managed by trial site pharmacy or PI. Q2 According to the Medicinal Product Act only wholesale distributors can distribute medicinal products and the IMP is dispensed to the participant by the trial site pharmacy or PI. Q3 According to the Medicinal Product Act only wholesale distributors can distribute medicinal products and the IMP is dispensed to the participant by the trial site pharmacy or PI. Q4 According to the Medicinal Product Act only wholesale distributors can distribute medicinal products and the IMP is dispensed to the participant by the trial site pharmacy or PI. Q5 According to the Medicinal Product Act only wholesale distributors can distribute medicinal products and the IMP is dispensed to the participant by the trial site pharmacy or PI. Q6 According to the Medicinal Product Act only wholesale distributors can distribute medicinal products and the IMP is dispensed to the participant by the trial site pharmacy or PI. Q7 IMP can be delivered to principal investigator from wholesale licence holders located in other EU MSs. Q8 According to the Medicinal Product Act only wholesale distributors with a manufacturing authorisation are allowed to perform labelling of IMPs. Q9 According to the Medicinal Product Act only wholesale distributors can distribute medicinal products and the IMP is dispensed to the participant by the trial site pharmacy or PI. Q10 According to the Medicinal Product Act only wholesale distributors can distribute medicinal products and the IMP is dispensed to the participant by the trial site pharmacy or PI. Q11 Physical face to face meeting between the trial participant and the PI or a member of the research team is yet mandatory in Croatia. Q12 Although not specifically addressed in CT legislation, the use of qualified electronic signatures is accepted. Q13 - Q14 It depends on the task/procedure. Q15 If on-site SDV is not possible, remote SDV is allowed if EU (EC, EMA, HMA) GUIDANCE ON THE MANAGEMENT OF CLINICAL TRIALS DURING THE COVID-19 (CORONAVIRUS) PANDEMIC is respected. Remote SDV must be submitted as substantial protocol amendment to CEC/RA and must be precisely specified in the protocol as to how it will be carried out. Permission to perform remote monitoring should be previously carefully considered with clinical trial site and prior permission and agreement must be reached between the site and the sponsor/CRO. HU Q1 - Q2 - Q3 - Q4 - Q5 - Q6 - Q7 - Q8 - Q9 - Q10 - Q11 35/2005. (VIII. 26.) Health Ministry Decree Q12 e-IDAS Art. 6., 7. , 26. , 27. , 36. , 37. Q13 Consent of the trial participant to the home visits does not reduce in any aspect the responsibilities of the PI. Q14 Consent of the trial participant to the home visits does not reduce in any aspect the responsibilities of the PI. Q15 In case of emergency remote SDV should be allowed if EMA „GUIDANCE ON THE MANAGEMENT OF CLINICAL TRIALS DURING THE COVID-19 (CORONAVIRUS) PANDEMIC (most recent version)” is respected with special attention to paragraph 11. d) and Annex 1. In general, the prerequisite of RSDV may not be less stringent. Recommendation paper DCT, V02, 1 October 2025 33 IE Q1 Could be allowed on case-by-case basis, when justified and clearly described in the clinical trial application. Generally recommended to speak with the HPRA. Q2 Could be allowed on case by case basis, when justified and clearly described in the clinical trial application. Generally recommended to speak with the HPRA Q3 Could be allowed on case by case basis, when justified and clearly described in the clinical trial application. Generally recommended to speak with the HPRA Q4 An MIA in itself does not provide for direct distribution to clinical trial participants Q5 Could be allowed on case by case basis, when justified and clearly described in the clinical trial application. Generally recommended to speak with the HPRA Q6 Could be allowed on case by case basis, when justified and clearly described in the clinical trial application. Generally recommended to speak with the HPRA. The IMP should comply in all respects with details which have been approved as part of the clinical trial application in Ireland e.g. labelling. Q7 The IMP should comply in all respects with details which have been approved as part of the clinical trial application in Ireland e.g. labelling. Q8 In accordance with the terms of CTR Art 61.5, S.I. No. 99/2022 and the HPRA Guide to Registration of Processes Exempted under Article 61(5) of the CTR. Generally recommended to speak with the HPRA. Q9 When justified and clearly described in the clinical trial application. Generally recommended to speak with the HPRA Q10 - Q11 - Q12 - Q13 - Q14 Dependant on task Q15 There are no provisions in national legislation which prohibit remote access to medical records. However, such access can only be permitted by the institution / persons responsible for control of the data, in consideration of application data protection requirements. No footnotes for: IS IT Q1 - Q2 - Q3 National guideline published on Aug 2024 allowing direct delivery Q4 National guideline published on Aug 2024 allowing direct delivery Q5 National guideline published on Aug 2024 allowing direct delivery Q6 National guideline published on Aug 2024 allowing direct delivery Q7 Not according to the current provisions for CT according to Directive; national provisions for CT according to CTR are to be made available shortly Q8 - Q9 National guideline published on Aug 2024 allowing direct delivery Q10 National guideline published on Aug 2024 allowing direct delivery Q11 (CTR art.29 does not foresee a face to face interview, no additional national provision exists on this aspect) Q12 PADES electronic signature on pdf files is surely accepted; other formats (e.g. CADES) to be confirmed Q13 - Q14 - Q15 In compliance with the GDPR No footnotes for: LI Recommendation paper DCT, V02, 1 October 2025 34 LT Q1 National legislation does not foresee the delivery of IMP directly to trial participants. Nevertheless, the IMP delivery from trial site to trial participant could be allowed on case by case basis, when justified and clearly described in the clinical trial application. Q2 Law of pharmacy https://www.e-tar.lt/portal/lt/legalActEditions/TAR.FF33B3BF23DD Q3 Law of pharmacy https://www.e-tar.lt/portal/lt/legalActEditions/TAR.FF33B3BF23DD Q4 Law of pharmacy https://www.e-tar.lt/portal/lt/legalActEditions/TAR.FF33B3BF23DD Q5 - Q6 Law of pharmacy https://www.e-tar.lt/portal/lt/legalActEditions/TAR.FF33B3BF23DD Q7 - Q8 Re-packing and re-labelling can be performed in the hospital pharmacy, associated with the clinical trial site: The Order of the Minister of Health of the Republic of Lithuania No V-571 regarding the procedure of re-packing and re-labelling of IMPs at the clinical trial sites Q9 Law of pharmacy https://www.e-tar.lt/portal/lt/legalActEditions/TAR.FF33B3BF23DD Q10 Law of pharmacy https://www.e-tar.lt/portal/lt/legalActEditions/TAR.FF33B3BF23DD Q11 - Q12 Qualified electronic signature should be used. Q13 Delegation of specific functions is possible, contract needed with the relevant and licensed Health care institutions. Q14 - Q15 Yes, if electronic medical records are used and access to medical records of particular patient is feasible. LU Q1 It is delivered by the hospital pharmacist to the investigator who then delivers it to the patient. Q2 The delivery is coordinated by the hospital pharmacy. The pharmacist provides the investigator with the IMP. Q3 Clinical trials are only performed in hospitals in Luxembourg, and there are no delegated pharmacies for IMPs. IMPs can only be managed by hospital pharmacies. Q4 - Q5 - Q6 - Q7 - Q8 - Q9 - Q10 - Q11 - Q12 Not applicable as not yet done in practice. Q13 Delegated tasks are performed by qualified research team members. Q14 It depends on the clinical trial and task/procedure modalities Q15 - LV Q1 No restrictions in national legislation Q2 No restrictions in national legislation Q3 No restrictions in national legislation Q4 Delivery to patients is acceptable if patient’s confidentiality is ensured. Q5 Licensed depot is deemed acceptable for IMP delivery directly to the trial participants. Not specifically mentioned in national legislation. Q6 - Q7 - Q8 - Q9 - Q10 - Q11 The physical face-to-face meeting is the primary expectation, but video-based communication can be accepted in certain situations, as per decision of the ethical committee. Q12 - Recommendation paper DCT, V02, 1 October 2025 35 Q13 - Q14 Case-by-case evaluation Q15 As the medical records are currently mainly available in the paper form at the trial sites, the remote access is possible only if justified. For electronic records access is possible if justified. On site SDV is preferred option. MT Q1 Activity has to be approved so a request should be made to the Medicines Authority Q2 Activity has to be approved so a request should be made to the Medicines Authority Q3 - Q4 - Q5 - Q6 - Q7 - Q8 - Q9 - Q10 - Q11 - Q12 - Q13 - Q14 - Q15 - NL Q1 Only in specific circumstances, see Medicines Act (Geneesmiddelenwet), 61.2. Q2 Medicines Act (Geneesmiddelenwet), article 61.2. Q3 Medicines Act (Geneesmiddelenwet), article 61.2. Q4 Medicines Act (Geneesmiddelenwet), article 34.2 and 61.2. Q5 - Q6 Medicines Act (Geneesmiddelenwet), article 61.2, 61.5 and 61.6. Q7 Medicines Act (Geneesmiddelenwet), article 34.2 and 61.2. Q8 Medicines Act (Geneesmiddelenwet) article 18.1 and 18.7. Q9 Under the conditions mentioned in Medicines Act (Geneesmiddelenwet) articles 18.1, 34.2 and 61.2. Q10 - Q11 However, see requirements in article 6 of the Medical Research Involving Human Subjects Act (Wet medisch-wetenschappelijk onderzoek met mensen), especially 6.5, 6.6 and 6.7. Q12 Article 6.2 of the Medical Research Involving Human Subjects Act (Wet medisch-wetenschappelijk onderzoek met mensen). Guidance on electronic signature (in Dutch): https://www.ccmo.nl/onderzoekers/publicaties/publicaties/2022/08/31/handreiking-elektronische-toestemmingsverlening Q13 Taking into account the requirements mentioned in the Healthcare Professionals Act (Wet op de beroepen in de individuele gezondheidszorg). Q14 Taking into account the requirements mentioned in the Healthcare Professionals Act (Wet op de beroepen in de individuele gezondheidszorg). Q15 Taking into account the requirements mentioned in the General Data Protection Regulation (GDPR). NO Q1 NoMA defines "deliver" as physical delivery to patient, and not shipment. Shipment to patient must be applied for and approved by NoMA. Q2 - Q3 - Q4 Forskrift om tilvirkning og import av legemidler § 3-2/Forskrift om grossistvirksomhet § 13 Q5 Forskrift om tillvirkning og import av legemidler § 3-2 /Forskrift om grossistvirksomhet § 13 Q6 Forskrift om tillvirkning og import av legemidler § 3-2/forskrift om grossistvirksomhet § 13 Recommendation paper DCT, V02, 1 October 2025 36 Q7 Forskrift om tillvirkning og import av legemidler § 3-2/forskrift om grossistvirksomhet § 13 Q8 Pharmacies with a “pharmacy manufacturing license” are allowed to re-label IMP. Only pharmacies with an ordinary MIA can label IMP. Q9 Provided agreement with the sponsor (Apotekforskriften § 27 g) Q10 Provided agreement with the sponsor (Apotekforskriften § 27 g) Q11 - Q12 Qualified electronic like for instance BankID is acceptable. Q13 Provided contractual arrangement Q14 - Q15 No national legislations regulating such activities. However, compliance with GDPR is a prerequisite as well as adherence to local procedures. PL Q1 Poland indicated acceptance of this solution in 2022 but it is not specifically addressed in current PL legislation Q2 - Q3 Public pharmacies do not IMPs. 68.1 pharmaceutical law (u.p.f). Retail trade in medicinal products is carried out in generally accessible pharmacies, subject to the provisions of para. 2, art. 70 sec. 1 I art. 71 sec. 1. When Art. 86 par. 2a upf pharmaceutical services referred to in art. 4 sec. 3 points 5 and 7 of the Act of 10 December 2020 on the pharmaceutical profession and professional tasks referred to in art. 4 sec. 4 points 1, 2, 5-7, 15 and 16 of this Act, may be provided only in a hospital pharmacy, company pharmacy or hospital pharmacy separated from these pharmacies" (Article 4 paragraph 4 point 2 of the Act - "Examination in auxiliary tests including research conducted in the hospital as a member of the research team.” Thus, community pharmacies cannot provide IMP for research. Q4 No. Annex 13 of the Regulation GMP defines the obligations of the manufacturer of investigational medicinal products, including the points below are as follows: point 44. The distribution of investigational medicinal products is carried out in accordance with the instructions given by or on behalf of the sponsor in the distribution order. 46. Detailed inventory records of shipments of investigational product sent by the manufacturer or supplier shall be maintained. In particular, it includes data identifying recipients. Currently, the PF Act, art. 42 par. 2 point 2 defines to which entities the manufacturer/importer may distribute medicinal products, there is no direct recipient - study participant listed there. Q5 - Q6 No, in relation to generally available pharmacies - they cannot participate in clinical trials, they can only trade medicinal products - Art. 68 sec. 1 u.p.f. Q7 No, in relation to generally available pharmacies - they cannot participate in clinical trials, they can only trade medicinal products - Art. 68 sec. 1 u.p.f. Q8 Not. Any delegated pharmacy may not label the IMP, this is limited to the pharmacy associated with the study site. Article 38b upf Q9 Pharmaceutical services referred to in Art. 4 sec. 3 points 5 and 7 of the Act of 10 December 2020 on the profession of a pharmacist, and the professional tasks referred to in art. 4 sec. 4 points 1, 2, 5-7, 15 and 16 of this Act, may be provided only in a hospital pharmacy, company pharmacy or a hospital pharmacy department established instead of these pharmacies" (Article 4 paragraph 4 point 2 of the u.o.z.f - "participation in research clinical trials, including trials conducted in a hospital as a member of the research team". Thus, generally accessible pharmacies cannot provide IMPs to trial participants. This is the task of a hospital pharmacy, an in-house pharmacy, a medical entity where a clinical trial is conducted Q10 as above Q11 In accordance with the Act on the professions of physician and dentist Art. 42. 1. A physician shall rule on the health status of a given person after previously examining him personally or examining him via teleinformatic systems or communication systems (...) Q12 Currently qualified electronic signature is required. Q13 Poland indicated acceptance of this solution in 2022 but it is not specifically addressed in current PL legislation Q14 Poland indicated acceptance of this solution in 2022 but it is not specifically addressed in current PL legislation Q15 Not forbidden Recommendation paper DCT, V02, 1 October 2025 37 PT Q1 As long as shipping conditions are kept under control. Q2 As long as shipping conditions are kept under control. Q3 To be assessed on a case-by-case basis; IMP circuit should be clearly and in detail described in the CT protocol. Q4 To be assessed on a case-by-case basis; IMP circuit should be clearly and in detail described in the CT protocol. Q5 Article 32nd, Law 21/2014, from the 16th of April, current version. Q6 Article 32nd, Law 21/2014, from the 16th of April, current version. Q7 To be assessed on a case-by-case basis; IMP circuit should be clearly and in detail described in the CT protocol. - Pharmacies with a “pharmacy manufacturing license” are allowed to re-label IMP. Only pharmacies with an ordinary MIA can label IMP. Q9 Article 32nd, Law 21/2014, from the 16th of April, current version; the pharmacies must be included on the IMP circuit described in detail in the CT protocol Q10 Article 32nd, Law 21/2014, from the 16th of April, current version Q11 Dialogue is mandatory Q12 On a case-by-case basis; wet ink use should also be possible, along with e-signatures; according to the EC website, reuse of CEF eID sample implementation software is described as being implemented in Portugal; please refer to Autenticacao.gov.pt Q13 Healthcare provider must be in direct dependency of the IP Q14 Should be clearly specified in the CT protocol Q15 This should be approached under the EU GDPR. Remote monitoring may be performed using video or by sending the de-identified data via email using secure means. Direct remote access to the site´s patient records computer systems is not allowed RO Q1 - Q2 In Romania, authorizes activities for pharmacies are explicitly mentioned in the Pharmacy Law nr 266/2008 which permit shipping activities only for OTC medicines Q3 In Romania, authorizes activities for pharmacies are explicitly mentioned in the Pharmacy Law nr 266/2008 which permit shipping activities only for OTC medicines Q4 - Q5 Based on authorisation issued by ANMDMR Q6 - Q7 - Q8 - Q9 In Romania, authorizes activities for pharmacies are explicitly mentioned in the Pharmacy Law nr 266/2008 which permit shipping activities only for OTC medicines Recommendation paper DCT, V02, 1 October 2025 38 Q10 In Romania, authorizes activities for pharmacies are explicitly mentioned in the Pharmacy Law nr 266/2008 which permit shipping activities only for OTC medicines Q11 - Q12 Advanced electronic signature/ Qualified electronic signatures Q13 Only if it is an accredited medical service provider Q14 Not yet. National provisions are under development Q15 - SE Q1 If relevant national legislation (for example "Lag (2009:366) om handel med läkemedel") is followed. Q2 If relevant national legislation (for example "Lag (2009:366) om handel med läkemedel") is followed. Any pharmacy that is handling IMPs for a clinical trial site should have a CT specific delegation (either from site or sponsor/CRO) and established routines for handling IMP. Q3 If relevant national legislation (for example "Lag (2009:366) om handel med läkemedel") is followed. Any pharmacy that is handling IMPs for a clinical trial site should have a CT specific delegation (either from site or sponsor/CRO) and established routines for handling IMP. Q4 - Q5 - Q6 Current interpretation of "Lag (2009:366) om handel med läkemedel" Q7 Current interpretation of "Lag (2009:366) om handel med läkemedel" Q8 Labelling of IMPs in pharmacies is restricted to e.g. auxiliary labelling of authorised IMPs, if performed in accordance with relevant national pharmacy legislation. Please refer also to national legislation HSLF-FS 2021:109. Any pharmacy that is handling IMPs for a clinical trial site should have a CT specific delegation (either from site or sponsor/CRO) and established routines for handling IMP. Q9 If relevant national legislation (for example "Lag (2009:366) om handel med läkemedel") is followed. Any pharmacy that is handling IMPs for a clinical trial site should have a CT specific delegation (either from site or sponsor/CRO) and established routines for handling IMP. Q10 If relevant national legislation (for example "Lag (2009:366) om handel med läkemedel") is followed. Any pharmacy that is handling IMPs for a clinical trial site should have a CT specific delegation (either from site or sponsor/CRO) and established routines for handling IMP. Q11 - Q12 The system used (level of category) is the responsibility of the sponsor. Q13 If relevant national healthcare legislation and hospital practices allows for it. Q14 If relevant national healthcare legislation and hospital practices allows for it. Q15 - Recommendation paper DCT, V02, 1 October 2025 39 SI Q1 In case the trial site is a Hospital, the IMP must be managed by Hospital pharmacy Art 62, Par 3 and Par 7 of Pharmacy Practice Act Q2 Always under supervision of the investigator of the trial site Q3 In case the trial site is a Hospital, the IMP must be managed by Hospital pharmacy article 67 point 7 (1) Q4 In case the trial site is a Hospital, the IMP must be managed by Hospital pharmacy article 67 point 7 (1) Q5 Only in exceptional situation (IMP shortage due to for example COVID-19 lock-down and should be on the basis of a risk assessment with patient safety as utmost priority and only after agreement with the investigator and on the basis of the investigator’s prescription. Q6 In case the trial site is a Hospital, the IMP must be managed by Hospital pharmacy article 67 point 7 (1) Only after agreement with the investigator and on the basis of the investigator’s prescription. Q7 In case the trial site is a Hospital, the IMP must be managed by Hospital pharmacy article 67 point 7 (1) Only after agreement with the investigator and on the basis of the investigator’s prescription. Q8 Delegated pharmacy must comply with special requirements in accordance with Article 13 Par 2. Q9 It is not appropriate if IMP is blinded. Under oversight of investigator of trial site. The trial drug must be marketed and used within the approved indication (according to the SmPC). Q10 It is not appropriate if IMP is blinded. Under oversight of investigator of trial site. The trial drug must be marketed and used within the approved indication (according to the SmPC). Q11 In accordance to ICH GCP The investigator, or a person designated by the investigator, should fully inform the participant or the participant’s legally acceptable representative, of all pertinent aspects of the trial & and should answer all participant questions. The communication of this information should be documented. Q12 Currently, there is no established practice. Q13 - Q14 depending on the procedure/task Q15 If on-site SDV is not possible for a longer period of time due to for example lock-down due to pandemic, remote rSDV must be submitted as substantial protocol amendment to EC/RA and must be precisely specified in the protocol as to how it will be carried out, so that the rights of the participants will be protected and will not unnecessarily burden the staff at the trial site who must agree to such a method of data verification. Monitors should sign a written confidentiality agreement committing to securely destroy any copy of redacted documents, whether paper or electronic, as soon as they have been used for source data verification and committing not to make any copy (or recording in the case of video access) of any non-pseudonymised document. References from SI: (1) Pharmacy Practice Act (Official Gazette of the RS, no. 85/16, 77/17, 73/19 and 186/21) (2) Regulation on the implementation of the Regulation (EU) on clinical trials of medicinal products for human use (Official Gazette of the Republic of Slovenia, No. 132/22) SK Q1 Q2 Q3 Q4 Q5 Q6 Q7 Q8 Q9 Pharmacist must be delegated by PI. Q10 Pharmacist must be delegated by PI. Q11 Q12 Qualified electronic signature for investigator and participant is required according to the Act No. 272/2016 on credible services for electronic transaction for domestic trade. According to the EU regulation, the new forms of eIDAS are AsiC-E, AsiC-S and are acceptable. Q13 Q14 According to § 2(46) of Act No. 576/2004 Coll. on Health Care, Services Related to the Provision of Health Care, and on the Amendment and Supplementation of Certain Acts Q15
accelerating-clinical-trials.europa.eu
October 30, 2025 at 12:31 PM
Nieuws van ACT EU: Meeting highlights MSP Advisory Group 18 September 2025
Advisory Group 18 September 2025, 09:30-13:30 (CEST), Webex
Agenda – ACT EU Multi-stakeholder Platform Advisory Group Meeting highlights – ACT EU Multi-stakeholder Platform Co-Chairs: Maria Jesús Lamas (Regulatory co-chair), Denis Lacombe (Stakeholder co-chair) 1. Opening of the meeting The co-chairs welcomed participants to the Multi-stakeholder Platform Advisory Group (MSP AG) meeting and briefly outlined the agenda, which focused on key policy initiatives, industry proposals, and regulatory updates relevant to the EU clinical trials landscape, including the core dossier proposal, the reduction of clinical trial review timelines, cross-border clinical trials and EU clinical trial metrics. Further, the membership updates were briefly presented. Links to presentations and meeting agenda. 2. Cross-border Clinical Trials 2.1 Cross-border clinical trials and arrangements to take part in a trial abroad The presentation highlighted three main challenges facing cross-border clinical trials in Europe: liability insurance conditions, import and delivery of investigational and authorised medicinal products (IMP/AMP), and patient recruitment. Current insurance coverage is limited to the host country, leaving gaps when patients return home, while a lack of harmonisation on IMP/AMP delivery creates operational barriers. A proposal was made for the ACT EU to organise a multi-stakeholder workshop focused on improving recruitment, including through matchmaking and reducing barriers to cross-border participation. Enhancing the CTIS clinical trial map and database by including a “cross-border readiness” feature was also suggested to increase transparency on trials open to cross-border patients. Q&A session The discussion focused on challenges and opportunities for cross-border clinical trial participation. While some suggested prioritising rare and life-threatening diseases, others stressed access should not be restricted, as neighbouring countries may be more accessible than domestic sites. The patient community voiced strong support for addressing cross-border access more broadly, underlining that access should not depend on where patients live. Key barriers identified included gaps in liability insurance, with current policies limited to the trial country and the European Health Insurance Card not applying to clinical trials. In addition, some countries require direct negotiations with insurers in their own jurisdiction, adding complexity and Meeting highlights – ACT EU Multi-stakeholder Platform Advisory Group Page 2/7 delays. The need for EU-level harmonisation to ensure clarity and consistent patient protection were underscored. Participants also highlighted the lack of harmonisation across Member States in IMP/AMP import and delivery, stressing the need for EU guidance to provide more consistent rules for sponsors/sites and avoid delays and inequities. Recruitment was highlighted as a central challenge, with barriers such as matchmaking and cross-border cooperation. An ACT EU-organised workshop to address these issues was well received, and EU-level support was considered valuable. Further concerns were raised about the ability of very ill patients and those with disabilities to travel for trials, highlighting the need to consider accessibility and patient well-being. At the same time, it was noted that some patients may be willing to travel despite health challenges, emphasising the importance of providing flexible options rather than one-size-fits-all solutions. During the discussion, it was noted that a form of classification of clinical trials could be useful, distinguishing between those that naturally call for cross-border participation, such as trials requiring access to sites with specialised technologies, and more conventional clinical trials that should ideally be available to patients within their own countries. The discussion also underlined that clinical trials should be integrated within broader cross-border healthcare systems, requiring stronger collaboration with healthcare providers to ensure continuity of care and patient well-being. Enhancing the Clinical Trials Information System (CTIS) trial map to indicate sites ready for cross-border patients was seen as a promising and transparent tool to support trial planning. Next Steps / Action points • The ACT EU programme will consider the proposal for a potential multi-stakeholder workshop to discuss options for improving recruitment, including through matchmaking and reducing barriers to cross-border participation in Europe. • The potential addition of a “cross-border readiness” field in the CTIS trial map will be further explored. 3. CTR/CTA optimisation 3.1 Core Dossier: opportunities for increasing efficiency and future proofing the CT ecosystem in Europe The presentation introduced the concept of a core dossier model as a shift from a trial-by-trial approach to a product-based approach. The proposed core dossier would consolidate key documents, such as the Investigator’s Brochure (IB) and the Investigational Medicinal Product Dossier (IMPD), into a continuously updated single reference to support multiple trials. This would reduce duplication, improve efficiency, and embed reliance on prior assessments throughout a product’s lifecycle. It was emphasised that the focus of this presentation was on the conceptual framework rather than on technical solutions, with a request for stakeholder feedback on potential enablers, barriers, and benefits. Initial analysis indicated that the CTR does not prevent the implementation of this model, though practical implementation challenges remain. Q&A Session Participants generally supported the concept of a core dossier model as a way to streamline submissions, reduce duplication, and improve efficiency, but raised concerns on practical implementation aspects, particularly the administrative burden of keeping trial sites information up to date. Additional considerations on eventual responsibilities for the core dossiers were flagged, especially when trial-specific information should supplement the dossier and how deviations should be handled. International experience, including the FDA’s use of a similar model, demonstrated feasibility, but protocol-specific requirements would still need to be addressed alongside the core dossier. Safeguards were deemed essential to ensure accuracy and reliability. Meeting highlights – ACT EU Multi-stakeholder Platform Advisory Group Page 3/7 The discussion stressed the importance of distinguishing what is possible under the current Clinical Trials Regulation (CTR) and CTIS framework from what may need longer-term legislative changes. No major legal barriers were identified, but CTIS functionality was seen as a limitation. A pilot within CTIS was proposed to test feasibility and gather evidence. The European Commission noted that the CTR and CTIS already support cross-referencing (e.g., IMPD-Q), with current policy developments like the Biotech Act aligned with this approach. Furthermore, stakeholders were strongly encouraged to share their views through the ongoing impact assessment surveys that have been launched by the European Commission in the context of the upcoming Biotech Act. Participants confirmed that a modification to a single core quality dossier could apply across linked trials, though practical and legal details would need clarification under the Biotech Act or CTR annexes. Overall, strong support was expressed for the model’s alignment with EU priorities and its potential efficiency gains, but expectations must be managed. The Commission confirmed its role at this stage is primarily to listen and gather input while the regulatory and operational analysis is completed. Next Steps / Action points Stakeholders were invited to share via the ongoing European Commission impact assessment survey their perspective and suggestions on the above topic. 3.2 Reducing review timelines to 60 days: enhancing competitiveness through faster CTA approval The presentation introduced industry’s proposal to reduce clinical trial review timelines from the current 106 days stipulated in the CTR to 60 days to boost Europe’s competitiveness. National pilots show feasibility of this proposal, with no legislative change needed. Proposed measures to reduce assessment timelines include removing holiday pauses, strengthening reliance on the Reporting Member State (RMS), and harmonising Member State requirements, with key actions focusing on streamlining procedures, addressing operational inefficiencies, and adopting best practices already applied in some Member States. Q&A session Participants noted that Europe’s timelines often cause delays in trial initiation and recruitment. A differentiated approach between simple and complex trials was suggested, while emphasising that faster timelines must not compromise patient safety and data quality. Stakeholders discussed scaling up good national practices, noting that some Member States already managed to implement shorter timelines through efficient coordination and better resource management. Industry stressed that faster, more predictable reviews would boost Europe’s competitiveness for multinational trials. Recruitment competition was flagged as a key challenge, with delays in authorisation reducing time for patient enrolment and limiting access. Inefficiencies in authorising substantial modifications were also seen as barriers to timely trial adjustments, patient safety and the expansion of recruitment. The proposal was recognised as timely in the context of the Biotech Act, with stakeholders encouraged to provide concrete evidence and proposals through the ongoing European Commission impact assessment surveys. Regulators underlined the importance of strengthening the role of the RMS while noting that any acceleration must be carefully planned to support multinational collaboration. The Commission acknowledged that long timelines place Europe at a competitive disadvantage for multinational trials and confirmed ongoing work through the CTIS Simplification Taskforce to identify where efficiencies could be introduced. At the same time, it was noted that structural and resource constraints may limit how far timelines can realistically be shortened. Stakeholders welcomed the support shown and called for piloting the 60-day model, though concerns were raised about Member States’ capacity to manage increased trial volumes. It was Meeting highlights – ACT EU Multi-stakeholder Platform Advisory Group Page 4/7 agreed that Advisory Group feedback will inform the Commission’s Biotech Act policy work, with piloting and targeted efficiency measures to be further explored. 4. ACT EU training curriculum 5.1 Proposals on content for ACT EU clinical trials curriculum The presentation highlighted the need for a sustainable, multi-level training curriculum to support academic clinical trials, extending beyond investigators to include all staff involved in a clinical trial. Existing national and EU initiatives were noted, though many face sustainability issues after funding ends. ACT EU was proposed to integrate these resources into an agile curriculum covering regulatory, scientific, operational, and technical aspects. The new ACT EU focus group on clinical trials training was suggested as the forum to align priorities, avoid duplication, and incorporate stakeholder feedback to ensure the curriculum meets practical needs. Q&A session Participants welcomed the initiative, emphasising sustainability and better integration of existing training resources. Fragmentation and the short-lived nature of project-based programmes were highlighted as key challenges, with mapping and signposting seen as a valuable first step. Concerns were raised about the burden of training for each trial, leading to calls for a structured curriculum that distinguishes between generic and trial-specific content. Stronger generic training was suggested to streamline protocol-specific training and reduce duplication while ensuring quality and consistency. Training was seen as essential for all trial team members, including study nurses, pharmacists, clinical research associates and other healthcare professionals, to build a competent workforce. Participants highlighted the challenge of high staff turnover in public research organisations, which often leads to repeated retraining. High-quality training opportunities were also considered important for attracting and retaining staff. It was underlined that training should equip both assessors and investigators for future innovations, with adaptable courses addressing new scientific and digital skill needs. Participants supported integrating risk-based and pragmatic approaches, as well as multinational trial and portfolio management, to build capacity and career pathways. Alignment with international initiatives like WHO’s work to accelerate access to training for the research workforce was encouraged to avoid duplication. In addition, the proposed agile, multi-level structure was welcomed as a way to tailor training to staff roles. Coordination with regulatory training and inclusion of operational and technical aspects, such as quality management and project oversight, were also seen as priorities. The absence of SME representation in the ACT EU clinical trials training focus group was noted, with their input considered essential. The focus group will lead the mapping and integration of existing initiatives and gather further stakeholder input to refine the curriculum, ensuring it remains responsive to evolving needs such as innovation, digitalisation, and proportionality. Next steps/ Action points: Launch a call for SME representatives (e.g., EUCOPE or EuropaBio) and healthcare professional representatives to join the ACT EU focus group on clinical trials, with deadline of 6 October to express interest. 5. Ensuring Regulatory Agility for Clinical Trials During Public Health Crises 5.1 PA11 Regulatory flexibility for CTs during Public health emergency The presentation addressed regulatory flexibility for clinical trials during public health emergencies, presenting the new guidance, which is being drafted, drawing on lessons from the COVID-19 pandemic as well as existing regulations. The guidance will expand beyond pandemic situations to cover other crisis scenarios, such as natural disasters, and aims to provide a framework for adapting Meeting highlights – ACT EU Multi-stakeholder Platform Advisory Group Page 5/7 clinical trials in emergencies. Key reference points include the CTR and ICH guidelines E6(R3) and E8(R1), which support risk-proportionate approaches and a focus on critical-to-quality factors. Q&A session Participants reflected on lessons from COVID-19, stressing the need for preparedness in future crises. A key priority was clarifying when and how regulatory flexibilities can be applied, supported by practical examples for sponsors and investigators. While crisis-specific flexibilities are essential, some MSP AG members suggested that certain principles could also be applied in routine settings, fostering a preparedness mindset across the system. Safeguards must remain in place to protect patients and ensure data integrity. Operational challenges were noted, including harmonisation across Member States, logistical barriers, and oversight when conventional monitoring is disrupted. The importance of consultation with stakeholders, particularly sponsors, ethics committees, and regulators, was emphasised to ensure the guidance is both practical and implementable. Ongoing EU and international initiatives were highlighted as important for global coordination, with investigator groups and academia leading efforts, and industry engagement considered vital to avoid misalignment. It was confirmed that stakeholders will be consulted once the draft guidance is ready, while sponsors were encouraged to share if they already include crisis preparedness in their trial design and to identify the main challenges, they face in applying regulatory flexibility. The guidance will be refined based on this input, aiming to balance flexibility with quality and patient safety. Post-meeting note: A dedicated email was circulated to the MSP AG representatives on 29 September, with a deadline for responses set for 13 October. 6. Clinical trials related legislative developments 6.1 Update on policy development at the EU level The update on policy and legislative developments highlighted the Pharmaceutical Legislation reform, the Biotech Act, and ACT EU’s role in modernising the CTR. Alignment with ICH guidelines, work on decentralised trials, crisis preparedness, and patient access were noted. The importance of synergies with EU digitalisation, data governance, and the European Health Data Space was underlined, as these will directly shape how clinical trial data are generated, shared, and assessed, supporting Europe’s competitiveness in clinical research. Q&A session Participants welcomed the policy update and underlined the need for transparency and predictability in ongoing reforms. The potential impact of the Biotech Act on clinical trial submissions and timelines was discussed, with emphasis on the importance of continuous dialogue. ACT EU was recognised as central in aligning implementation with legislative changes and ensuring CTR/CTIS readiness. Stakeholders stressed the value of integrating ICH guidance on risk-proportionate approaches, pragmatic designs, and digital elements, while avoiding duplication with international efforts. Concerns were raised about ensuring that legislative changes do not overburden sponsors, investigators, or Member States with additional administrative requirements. Instead, participants stressed that reforms should deliver simplification, harmonisation, and improved efficiency, while maintaining patient safety and scientific integrity. Next steps include continued stakeholder engagement in legislative processes, close alignment of ACT EU activities with evolving frameworks, monitoring ICH updates, and continuous update on cross-cutting digital initiatives such as the European Health Data Space. 7. Clinical trials metrics 7.1 Update on EU clinical trials metrics Meeting highlights – ACT EU Multi-stakeholder Platform Advisory Group Page 6/7 An update was provided on clinical trial metrics and key performance indicators (KPIs), aimed at monitoring the attractiveness, efficiency, and impact of the EU clinical trial ecosystem. Two core KPIs were outlined: the number of authorised multinational trials and the number of clinical trials with first patient recruited within 200 calendar days of CTA submission. The overall impact of clinical trials on public health and research innovation will also be monitored although no dedicated KPI has been set for this parameter due to its multidimensional nature. These will serve as the reference for tracking Europe’s performance, supported by sub-metrics under development and a forthcoming three-year analysis report. Q&A session Participants welcomed the introduction of KPIs and encouraged expanding them to capture innovation and decentralised trial elements, ensuring Europe’s leadership in modern trial design is reflected. It was clarified that the initial focus will remain on agreed KPIs to establish a baseline, with additional measures to be integrated over time and targets reviewed regularly to adjust accordingly and remain ambitious. It is believed that Europe is the first region to adopt such metrics, and stakeholders valued the transparency, stressing their importance for future dialogue. Continued feedback and engagement in upcoming communication activities, including the live LinkedIn update on 24 September, were encouraged. Further input was invited on possible additional parameters to include in CTIS, as adjustments can still be made while the system is being updated to reflect the agreed KPIs. CTIS was praised as a valuable information source and a strong foundation for evidence-based discussions on EU clinical trial performance. Next steps/Actions: MSP AG members are invited to provide proposals for additional fields to be included in CTIS to facilitate the search for specific types of clinical trials, such as complex and/or decentralised trials. Please submit any suggestions to the MSP AG Secretariat ([email protected]) by close of business on 31 October 2025. Post meeting note: More information on the clinical trial metrics can be found at the three-year report, the news announcement, and the LinkedIn Live session. 8. Pre-read No formal presentation was provided. Participants were reminded that all supporting documents and updates, including focus group work, ongoing actions, workshop outcomes, patient involvement, and overall ACT EU progress, were shared in advance and were available on the ACT EU website. Q&A session Participants requested further insight into the timeline of ongoing initiatives. Updates were provided, noting that the AxMP document was in its final stages and expected to be endorsed and published soon. Work on RFI Part I and II was close to completion, with both sets of questions to be consolidated into a single document. The role of the RMS continued to be discussed in CTCG, with efforts focused on strengthening trust and ensuring more consistent practices. Stakeholders also asked about harmonisation of requirements, and it was explained that assessors are working towards identifying critical considerations only, to avoid unnecessary requests or deletions. An update was also provided on the risk-based approaches drafting group, which had launched in the summer, and which will work in collaboration with the MSP AG focus group. The focus group will meet on a monthly basis and provide further detailed examples and input to inform the revision of the 2017 Recommendation paper on risk proportionate approaches. Meeting highlights – ACT EU Multi-stakeholder Platform Advisory Group Page 7/7 A.O.B. Participants were informed that, from 2026 both permanent and alternate representatives of the MSP AG will be invited to attend all MSP AG meetings. The revised 2026 meeting dates were announced as 20 March and 18 June and have been published online. Further, participants were encouraged to register for the upcoming ACT EU workshop on external controls, scheduled for 3 November, with details available on the EMA website and included in the chat and post-meeting package. Closing remarks The co-chairs closed the meeting by thanking participants for their contributions and engagement. Actions arising will be followed up as needed. Next meeting is on 20 March 2026 (virtual) and deadline for topic submissions is 18 January 2026.
accelerating-clinical-trials.europa.eu
October 30, 2025 at 9:38 AM
EU Nieuws: New and updated documents - EudraLex Volume 10: Clinical trials guidelines
New and updated documents - EudraLex Volume 10: Clinical trials guidelines
New and updated documents - EudraLex Volume 10: Clinical trials guidelines
health.ec.europa.eu
October 30, 2025 at 9:20 AM
Nieuws van ACT EU: Slides from ACT EU Workshop for assessors of paediatric clinical trials
accelerating-clinical-trials.europa.eu
October 27, 2025 at 5:33 PM
Nieuws van ACT EU: ACT EU multi-annual workplan 2025-2026
ACT EU workplan
Contents ACT EU workplan 2025-2026 2 Introduction Workplan 3 Introduction 3 The Accelerating Clinical Trials in the European Union (ACT EU) initiative supports smarter clinical trials through regulatory, technological and process innovation. The vision is to have better, faster and optimised clinical trials, benefitting patients and healthcare in Europe. Seamless coordination between stakeholders, regulators and ethics committees strengthens cross-border collaboration. The ACT EU multi-stakeholder platform is central to this. The 3rd HMA-EC-EMA ACT EU workplan was adopted in December 2024 and covers activities for 2025 and 2026, together with an overview of the achievements from the previous year. Workshops reflected in the workplan will be organised in collaboration with the European Medicines Regulatory Network (EMRN). The document was informed by stakeholder and expert consultations and reflects the revised structure of the ACT EU programme based on the evolution of activities over time. ACT EU structure ACT EU workplan 2025-2026 4 Overarching activities: • ACT EU governance • Multi-stakeholder Platform Underpinning activities: • Communication • Clinical trials analytics • Clinical trials training Operation of the Clinical Trials Regulation: • Implementation of the Clinical Trials Regulation • Support for non-commercial sponsors • Clinical trials safety Maximising impact of clinical trials – design and conduct of excellent clinical trials: • Good clinical practice modernisation • Consolidated advice on clinical trials • Clinical trials methodologies Multi-stakeholder platform Operation of the CTR Maximising impact of CTs Underpinning activities Clinical trials in public health emergencies Overarching activities Workplan Overarching activities 6 FUNCTIONAL AREA OVERVIEW ACT EU workplan update 2025 ACT EU Governance Multi-stakeholder Platform (MSP) Q1 - 2025 Q2 - 2025 Q3 - 2025 Q4 - 2025 Q1 - 2026 Q2 - 2026 Q3 - 2026 Q4 - 2026 Annual event of the Multi-stakeholder Platform Monthly meetings of the ACT EU Steering Group 2026 Event TimeframeDeliverable MSP Advisory Group quarterly meetings Annual event of the Multi-stakeholder Platform ACT EU workplan update 7 ACT EU Governance and Multi-stakeholder Platform The ACT EU Governance includes the ACT EU Steering Group that meets on monthly basis underpinned by programme management delivered by representatives of the Commission, HMA and EMA. The ACT EU Multi-stakeholder Platform (MSP) and its Advisory Group have been created. The Multi-stakeholder Platform meets yearly with a public meeting, while the advisory group meets on quarterly basis. Via this forum, stakeholders can discuss their needs and priorities and communicate these to the ACT EU regulatory partners. Delivered in 2024: ACT EU workplan 2025-2026 Q2-Q3 2024 Mapping of Governance for clinical trials Closure of the priority action 1 on mapping and governance Q2 2024 Creation of MSP and its Advisory Group (MSP AG) The objective of priority action 3 to establish the MSP has been achieved, and the priority action can be considered closed Operation of the Clinical Trials Regulation 8 FUNCTIONAL AREA OVERVIEW Complete definition on non-commercial sponsors 2025 Implementation of the Clinical Trials Regulation (CTR) Support for non-commercial sponsors (NCS) Clinical trials safety Q1 - 2025 Q2 - 2025 Q3 - 2025 Q4 - 2025 Q1 - 2026 Q2 - 2026 Q3 - 2026 Q4 - 2026 Workshop identified priorities on CTR implementation Review CTR/CTIS training material 2026 Event TimeframeDeliverable Maintaining a pharmacovigilance network for clinical trials Cooperation between CTs and post-marketing pharmacovigilance Network of Regulatory helpdesks Quarterly reporting on CTR implementation Working with regulatory partners to address stakeholders’ needs (see separate table p.9) 3-year KPI report Workshop on contractual agreements Workshop on risk-based approaches Workshop on fostering clinical research by NCS Workshop on fostering clinical research by NCS SAFE CT assessors meeting SAFE CT assessors meeting Review IT functionalities for safety Review IT functionalities for safety Network initiatives and activities To address the most important issues reported by stakeholders on the CTR implementation ACT EU workplan 2025-2026 9 Issue reported Responsible bodies Preparation of requests for information (RFI) CTCG (CTR Collaborate), MedEthicsEU (part II of CTA), CTAG Strengthening the role of the reporting Member States (RMS) CTAG, CTCG, NCA and Ethics Committees Harmonisation on CTA part II requirements CTAG, MedEthicsEU, CTCG Use of common templates in CTA MedEthicsEU, CTAG Translation aspects (CTA documents/RFI) MedEthicsEU, CTAG, CTCG Risk based approach and Low Interventional Clinical Trials CTAG, CTCG, MedEthicsEU Patients’ involvement in clinical trials CTCG Interplay CTR/IVDR/MDR COMBINE Programme - Combined studies - European Commission CTIS functionalities CTIS Programme - Clinical trials in human medicines | European Medicines Agency (EMA) Additional relevant initiatives Strengthening funding mechanisms European Commission ACT EU workplan 2025-2026 10 Implementation of the Clinical Trials Regulation The priority action is focused on the implementation of the Clinical Trials Regulation (CTR). This includes aspects such as: • Tracking the performance of the European clinical trials environment though regular reporting. • Re-organisation of existing training material and guidance documents on CTR/CTIS, to facilitate access to users and content update to align with latest developments. • Working with the ACT EU regulatory partners, as defined in the overview table, to jointly address main issues on CTR implementation. • Organisation of dedicated workshops, in liaison with the MSP AG to address issues raised by stakeholders. Delivered in 2024: Q2 2024 Implementation of CTIS revised transparency rules Q1-Q4 2024 Increasing transition of clinical trials to CTR, via CTIS Q3 2024 Yearly survey to sponsors with corresponding published report Q1-Q4 2024 Publication of monthly KPI reports ACT EU workplan 2025-2026 11 Support for non-commercial sponsors The priority action is focused on understanding the bottlenecks that prevent non-commercial sponsors (NCS) from planning and initiating clinical trials. This is particularly relevant for multinational clinical trials, and subsequently, the aim is to establish an action plan with clear measures in place, which consists of: • Creating and maintaining a network of regulatory helpdesks, building on national activities, including extra support for questions related to CTIS/CTR. • Finalising a definition of non-commercial sponsors. • Organise dedicated workshops, in liaison with the MSP AG, to address prioritised topics. Delivered in 2024: Q2-Q3 2024 Publication of national initiatives at MS level to support NCS Q2-Q3 2024 Publication of network initiatives (i.e. ECRIN, Enpr-EMA) to support NCS Q3 2024 Provision of extra support for CTIS related questions ACT EU workplan 2025-2026 12 Clinical trials safety The priority action is focused on strengthening clinical trials safety monitoring in the EU, by maintaining a clinical trials pharmacovigilance network. This includes regular dialogue via the safety assessors’ roundtables and enabling collaboration between clinical trial and post-marketing pharmacovigilance. This activity enables Member States to work together to improve trial safety through coordinated work-sharing assessment. The EU4Health Joint Action 12 (SAFE CT), which is focused on capacity building and training, facilitates this activity. In addition, a training curriculum for safety assessors has been developed. Delivered in 2024: Q1 2024 Organisation of SAFE CT workshop Q1-Q4 2024 Establishment of roundtable for safety assessors, maintaining a clinical trials pharmacovigilance network Q4 2024 Delivery of the training curriculum for safety assessors Design and conduct of excellent clinical trials 13 FUNCTIONAL AREA OVERVIEW Workshop on ICH E6(R3) (Annex 1/principles) 2025 Good clinical practice modernisation Consolidated advice on clinical trials Clinical trials methodologies Q1 - 2025 Q2 - 2025 Q3 - 2025 Q4 - 2025 Q1 - 2026 Q2 - 2026 Q3 - 2026 Q4 - 2026PRIORITY ACTIONS Workshop on external controls 2026 Event TimeframeDeliverable Change management and training activities for ICH E6(R3) Implement changes in EU guidance documents Enhancement of intra-network information exchange Roadmap of guidance documents Sustainable coordination of guidance development Best practice on guidance development Explore advanced search for guidance documents Running expanded pilot phaseOperation of pilots Evaluation of pilots Workshop on ICH E6(R3) implementation Publish learnings from initial pilots Signpost of existing guidance Overview on CTs guidance development Workshop on platform trialsWorkshop with Enpr-EMA Workshop on Bayesian Statistics Assessors workshop on paediatric CTs ACT EU workplan 2025-2026 14 Good clinical practice modernisation The renovation of the International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use (ICH) E6 guideline on “Good Clinical Practice” (GCP), aims to address the application of GCP principles to the increasingly diverse range of clinical trial types and data sources. Additionally, it provides flexibility, when appropriate, to facilitate the use of technological innovations in clinical trials. The priority action is focused on supporting the implementation of ICH E6(R3), also via dedicated workshops in liaison with the MSP and its advisory group. The activities of the priority action include developing a communication and change management strategy to support smooth adoption and implementation of the revised guideline, in addition to updating the relevant EU guidelines impacted by the change. Delivered in 2024: Q1-4 2024 Activities at ICH level on E6 (R3) revision ACT EU workplan 2025-2026 15 Consolidated advice on clinical trials The priority action is focused on bringing together key actors in clinical trials in the EU. Pilot initiatives are in place to facilitate the dialogue between regulators and sponsors/applicants prior to submission of applications. One pilot aims at reinforcing the scientific advice coordination between clinical trial approval and clinical trial design in a collaboration between SAWP/CTCG. A second initiative, coordinated by CTCG, covers pre-CTA advice on regulatory aspects. An analysis of the pilot initiatives, followed by a publication of the learnings will take place in 2025. Subsequent proposals for definitive processes will be considered, to facilitate the development of safe and effective medicines for the benefit of patients. Delivered in 2024: Q2 2024 Launch of consolidated pilots on clinical trials: SWAP/CTCG and pre-CTA (CTCG) Q2 2024 Publication of guidance documents for sponsors/applicants on the pilot initiatives Q3 2024 Evaluation of the first two iterations of pre- CTA consolidated advice (10 pre-CTA procedures) ACT EU workplan 2025-2026 16 Clinical trials methodologies The priority action is focused on facilitating activities on methodological aspects for clinical trials. To reach this goal, the objectives of the work are: • Ensure aligned clinical trial guidance development across the EU network resulting in high impact guidance documents implemented in practice. • Bring together key decision makers during the clinical trial life cycle (including MWP, CTCG, HTA) via sustainable coordination of guidance development. • Help stakeholders navigate the EU clinical trial guidance landscape, by developing a signposting of existing MWP/CTCG/HTA guidance documents and an advanced search tool. In addition, an overview of future guidance documents in line with corresponding workplans of the relevant groups will be generated. • Organise dedicated workshops on topics of interest, also in liaison with the MSP AG. Q2-Q4 2024 Coordination of guidance between MWP, CTCG and HTA coordination group established Q2-Q4 2024 Ongoing activities on best practice on guidance development Delivered in 2024: Clinical trials in public health emergencies 17 FUNCTIONAL AREA OVERVIEW Training on the guidance document 2025 Clinical trials in public health emergencies (PHEs) Q1 - 2025 Q2 - 2025 Q3 - 2025 Q4 - 2025 Q1 - 2026 Q2 - 2026 Q3 - 2026 Q4 - 2026 Workshop on activities of the priority action Simplified CTA package 2026 Event TimeframeDeliverable Strengthening involvement of ethics committees Develop and implement fit-for-purpose regulatory flexibility in clinical trials Agree process for expedited assessment of CTs by MSs (NCA and ethics) Document on strengthening engagement of PHE ethics advisory group Publish simplified CTA package Publish recommendations on expedited assessment Publish guidance document on regulatory flexibility Training on the expedited assessment Training on simplified package ACT EU workplan 2025-2026 18 Clinical trials in public health emergencies The priority action is focused on enabling multinational clinical trials during public health emergencies (PHEs). Different aspects of the process of clinical trial approval are tackled, including: • Strengthen collaboration across National Competent Authorities and Medical Research Ethics Committees in the assessment of clinical trials in PHE and liaise with the EMA Emergency Task Force (ETF), to foster alignment and discussion across Member States. This includes also an analysis on possible benefits of having a central ethics committee during PHE. • Define a simplified package for submission of applications for clinical trials in PHE and expedited assessments. • Enable regulatory flexibilities in the assessment and conduct of clinical trials, based on the experience gained during COVID-19 pandemic. Delivered in 2024: Q2 2023 Work structure defined with 3 dedicated tracks Q3 2024 Establishment of PHE ethics advisory group Q4 2024 Workshop with Emergency Task Force Underpinning activities 19 FUNCTIONAL AREA OVERVIEW 2025 Communication Clinical trials analytics Clinical trials training Q1 - 2025 Q2 - 2025 Q3 - 2025 Q4 - 2025 Q1 - 2026 Q2 - 2026 Q3 - 2026 Q4 - 2026 Training needs academia/SME 2026 Event TimeframeDeliverable Continuous communication support activities (events, newsletters, website updates) Support access and analysis of clinical trials data Continuous dialogue to address needs to analyse clinical trials data Stakeholder engagement to strengthen education ecosystem Deliver training on CTIS/GCP/Methodologies/PHEs Training needs of other relevant groups Campaign on clinical trials Campaign on clinical trials Publication of research priorities Launch of web-based trial map to enable patient recruitment Publish summary of training needs Publish summary of training needs ACT EU workplan 2025-2026 20 Communication The priority action is focused on the communication aspects related to clinical trials. In addition, it supports regular dissemination of newsletters and information about events organised under ACT EU. Annual awareness campaigns are planned, with the intention to highlight the importance of clinical trials and their benefits for patients to increase public understanding and promote trust in the research process. Delivered in 2024: Q1-4 2024 Training, events, newsletters, communication campaign, regular updates of the ACT EU website ACT EU workplan 2025-2026 21 Clinical trials analytics The priority action is focused on engagement with the community of stakeholders to understand their needs and how data about clinical trials could support them. The stakeholders’ use cases are being collected to inform research priorities which may influence future public funding calls. A trial map for patients and health care professionals is being developed to facilitate access to clinical trials information in a user-friendly manner. The map will make it easier for patients to identify trials relevant to them. By highlighting research needs and facilitating access and analysis of clinical trial data, this priority action will inform better research and development of medicines and policy making in the EU. Delivered in 2024: Q1 2024 Clinical trials analytics workshop ACT EU workplan 2025-2026 22 Clinical trials training To support high quality clinical trials and enable better knowledge sharing, a training curriculum informed by regulatory experience is being developed. An overarching strategy and collection of training needs for different stakeholder groups will serve as the basis for the development of curricula. Engaging with stakeholders, identified via the dedicated training strategy, the curriculum will feed into an educational ‘ecosystem’ which will benefit from bidirectional exchanges to enable targeted training on clinical trials. Delivered in 2024: Q2-Q4 2023 Q1 2024 Training gap analysis for regulators Publication and dissemination of gap analysis summary/report Q2-Q4 2024 Ongoing analysis of training needs for academia and SME European Medicines Agency Official address Domenico Scarlattilaan 6 ● 1083 HS Amsterdam ● The Netherlands Telephone +31 (0)88 781 6000 Send us a question Go to www.ema.europa.eu/contact
accelerating-clinical-trials.europa.eu
October 27, 2025 at 4:21 PM
Nieuws van ACT EU: ACT EU workshop on paediatric clinical trials report
Meeting report
Workshop for assessors from NCAs and MRECs on paediatric clinical trials Meeting report Contents Introduction .......................................................................................................... 3 Setting the scene ................................................................................................... 3 Panel discussion .................................................................................................... 4 Breakout sessions .................................................................................................. 5 Conclusions .......................................................................................................... 5 Next steps ............................................................................................................ 6 Closing remarks from CTCG, PDCO and MWP ............................................................. 7 More information ................................................................................................... 8 Glossary .............................................................................................................. 9 3 Report: Workshop for assessors of paediatric clinical trials Introduction On 14 and 15 July 2025, the Clinical Trial Coordination Group (CTCG), Paediatric Committee (PDCO) and Methodology Working Party (MWP) organized a workshop for assessors from NCAs and ethics committees to discuss real cases of paediatric clinical trial applications which show different assessment approaches across EU/EEA. This workshop is a direct follow up to the ACT EU multi-stakeholder methodology workshop, which identified harmonization in the interpretation of CTR Article 32 (inclusion of minors in clinical trials) in the assessment of paediatric clinical trials as a priority. The objective of the workshop was to strengthen collaboration between national competent authorities, CTCG, ethics committees and the PDCO by bringing together all regulatory decision makers in the ecosystem of paediatric drug development. Through discussion and exchange on real use cases, the workshop provided an opportunity to address the challenges of assessing paediatric clinical trials under the Clinical Trials Regulation (CTR) as well as to develop together with the participants potential solutions for addressing inconsistencies in the interpretation of Article 32 of the CTR, in context of the revised Declaration of Helsinki, while ensuring ethical and scientific rigor. Setting the scene Moderators: Marianne Lunzer (CTCG chair, Austrian Agency for Health and Food Safety (AGES), Austria) and Monique Al (Central Committee on Research Involving Human Subjects (CCMO), Netherlands)/Vice- chair CTCG/Co-chair MedEdthicsEU). Presenters: Laura Fankhauser (Hopp Children’s Cancer Center Heidelberg (KiTZ) Germany), Dominik Karres (Paediatric Medicines Office EMA) and Sabine Scherer (PDCO member, Federal Institute for Drugs and Medical Devices (BFARM) in Germany), Martine de Vries (Head of the Department of Medical Ethics and Health Law Paediatrician Leiden University Medical Center, Department of Medical Ethics & Health Law, The Netherlands). Results from a descriptive analysis of paediatric clinical trial assessments revealed that there is significant variability in how Member States assess trials. Particularly, differences were noted in the interpretation of Article 32(1)(e), (f), (g) of the CTR (especially around “direct benefit” and the required evidence, e.g. Key messages • Despite regulatory progress, children remain underrepresented in clinical development, delaying access to innovative treatments and increases off-label use. • There are differences in interpretation of Article 32 and variability in how Member States assess paediatric clinical trials. • The revised Declaration of Helsinki emphasises that excluding vulnerable groups (like children) from research can potentially perpetuate or exacerbate their disparities. • Patients and caregivers are often willing to accept higher risks and burden, especially in life- threatening conditions. • Greater involvement of patient representatives in trial planning, regulatory and ethical discussions is essential. 4 Report: Workshop for assessors of paediatric clinical trials proof of concept; need for adult evidence). With regards to the inclusion of adolescents in adult trials, Member States differ in their acceptance, often requiring additional adult data to be generated first. It also showed that there is inconsistent use of paediatric investigation plans (PIPs) in assessments, with some Member States being more conservative, leading to none, delayed authorisation or authorisation only with certain conditions of trials despite the clinical trial application (CTA) being in line with an agreed PIP; appreciating that of course evidence may have evolved after the initial PIP agreement over time This first description of this as-is analyses provided the foundation for the subsequent discussions. It underpinned the need for more harmonised interpretation of Article 32, but also value submission of the full PIP into a CTA, to allow scientific considerations reflected upon during the clinical trial assessment process. It was emphasised that there are common challenges within the ecosystem of drug development in general. Those hold true across diseases and populations. But it remains key to acknowledge paediatric specificities, as represented by the EU Paediatric Regulation. Paediatric specific clinical development challenges, mainly related to small patient numbers due to disease prevalence, reflected upon in context of current methodological guidelines, such as ICH E11A on the use of extrapolation were discussed. It was emphasised that the key principle behind the use of extrapolation is to avoid unnecessary clinical trials in children whenever possible. And to using existing evidence from across populations and/or same in class products, as appropriate, in support of a paediatric clinical trial design, which addresses clear scientific questions in order to move forward in addressing identified unmet medical needs. This was complemented by the experience from the PDCO and how these considerations are reflected in the PIP assessment. From the regulation, the discussion moved to the principle of equitable access to health care through research and how to view paediatric clinical trials in context of the revised Declaration of Helsinki and CRT Article 32. It was highlighted that the revised Declaration of Helsinki emphasises that excluding vulnerable populations (like children) from research can potentially perpetuate or exacerbate their disparities. Therefore, the harms of exclusion must be considered and weighed against the harm of inclusion. This paradigm shifts from a “no unless” to a “yes, if justified” mindset should be taken into account when designing and reviewing paediatric clinical trials; as in line with relevant guidelines, such as E11A. It was concluded that justice and equitable access to research must be central to a scientific and ethical review. Panel discussion Moderator: Anette Solli Karlsen (PDCO member, Norwegian Medical Products Agency (NOMA), Norway) • Industry perspective • Academic perspective • Patient perspective Panellists: Scott Diede from Industry (MSD), Michel Zwaan from Academia [Prinses Maxima Centre Utrecht, Netherlands] and Tomasz Grybek from Patient perspective [Eurordis Rare Diseases Europe] Laura Fankhauser (Hopp Children’s Cancer Center Heidelberg (KiTZ) Germany), Dominik Karres (Pediatrics Medicines Office EMA) and Sabine Scherer (PDCO member, BFARM Germany), Martine de Vries (Head of the Department of Medical Ethics and Health Law Paediatrician Leiden University Medical Center, Department of Medical Ethics & Health Law, The Netherlands) 5 Report: Workshop for assessors of paediatric clinical trials The panellists provided additional perspectives, with the industry representative highlighting challenges with inconsistent assessments and subsequent delays in trial approvals. The academic perspective stressed the need for early engagement with regulators and patients/ caregivers at stage of trial design and pointing to the need for paediatric clinical trials which are acceptable to all regions globally. The patient/ parent representative stressed that patients and caregivers are often willing to accept higher risks, especially in life-threatening conditions and raised the concern that their voices remain underrepresented in trial design discussions and in clinical trial assessments. The panel discussed the value of potential alignment in the interpretation of Article 32 across NCAs and ethics committees. Reflecting on ways to improving exchange and learnings across regulatory decision makers (such as PDCO, CTCG/ NCAs and ethics committees). It was discussed how established platforms, like the CTCG assessor’s roundtable can be used more targeted and efficient to enable discussions and alignment. The challenges in applying the revised Declaration of Helsinki and CIOMS guidelines consistently were touched upon, pointing to the need for ways to further enhancing ethical review practices. Breakout sessions Moderators: Marianne Lunzer (CTCG chair, AGES Austria) and Monique Al (CCMO Netherlands /Vice-chair CTCG/Co-chair MedEdthicsEU) Breakout group topics The breakout session groups were introduced. This were closed meetings for NCA and Ethic Committees (EC members only). Real use cases of clinical trial applications were discussed. The use cases were chosen to represent of the following key issues: Dosing; clinical trials without prospect of direct benefit; what is sufficient proof of concept and how much adult data is needed; methodological challenges in relation to extrapolation; inclusion of adolescents in adult trials; situation of prospect of direct benefit/benefit to the group; minimum burden and risk; challenges seen from a patient perspective; how to support a global development. Five groups discussed in individual breakout sessions these use cases with a focus on potential solutions. Feedback from the breakout groups and Conclusions Moderators: Marianne Lunzer (CTCG chair, AGES Austria) and Monique Al (CCMO Netherlands /Vice-chair CTCG/Co-chair MedEdthicsEU) A recurring theme coming out of all five groups was the need for better alignment: • Between (NCAs) and Ethics in context of clinical trial approvals. • Inconsistent interpretations of Article 32 of the CTR and the revised Declaration of Helsinki remains a barrier in the assessment of paediatric clinical trials. • Between regulatory decision makers such as PDCO and NCAs and Ethics in context of a PIP agreement leading to a clinical trial application 6 Report: Workshop for assessors of paediatric clinical trials The groups discussed the cases in light of the interpretation of Article 32 (CTR), with a particular focus on: • Article 32.1(e) on the amount of adult data needed before including minors • Article 32.1(f): relation to the medical condition of the minor • Article 32(g): direct benefit vs group benefit with minimal risk/burden. Here the value to develop a common EU-wide interpretation were emphasized, especially regarding: • What constitutes “minimal additional risk.” • When group benefit justifies inclusion without direct benefit. The value of inclusion of adolescents in adult trials was raised, with a strong support for including adolescents when scientifically justified. Here it was proposed for sponsors to justify exclusion, not just inclusion. It was however flagged that some Member States may have national laws or practices that hinder such inclusion, pointing to the need for a better understanding of national legal frameworks. All groups agreed that there remains a need to further develop and/or update practical tools and guidance, such as: • Calling for a decision-making framework supporting the assessment of paediatric trials. • Consider updating the recommendation paper on ethical considerations in clinical trials with minors. • Considering developing a sponsor guidance on what to include in a paediatric CTA, including full PIP Decision, including its Annexes if available). • Need for further regulatory clarification on how to apply e.g. use of extrapolation. Next steps Moderators: Marianne Lunzer (CTCG chair, AGES Austria) and Monique Al (CCMO Netherlands /Vice-chair CTCG/Co-chair MedEdthicsEU) Analyses to better understand hurdles in MSs: Survey on national laws Current ‘guidance landscape’ fit for purpose? Analyse the document Ethical considerations for clinical trials with minors to see if an update is needed in the light of the workshop discussions and conclusions Need to develop an assessor’s best practice document on considerations specific for paediatric trials Guidance for sponsors directing towards good justification of inclusion of minors and provision of full PIP as part of the submission dossier, early engagement of investigators and patients, guidance should aim to explain paradigm shift (DoH, CTR) Enable information sharing, discussions and learnings across decision makers (from PIP to CTA): Organise meeting on interpretation of CTR art 32 e, f, g (PDCO, NCA, Ethics) Implement process for sharing and discussing critical cases between CTCG and PDCO 7 Report: Workshop for assessors of paediatric clinical trials Involvement of stakeholders, but also regulatory decision makers, such as ethics committees in guidance/Q&A updates (such as Q&A on paediatric dosing) created by PDCO, CTCG and MWP as co- author or in commenting phase Closing remarks from CTCG, PDCO and MWP Moderators: Marianne Lunzer (CTCG chair, AGES Austria) and Monique Al (CCMO Netherlands /Vice-chair CTCG/Co-chair MedEdthicsEU), Brian Aylward (PDCO chair), Kit Roes (MWP chair) Key messages • Timely workshop to address relevant issues related to assessment of paediatric clinical trials in EU/EEA • Clear message to continue this dialogue between regulatory decision makers, Ethics, investigators, sponsors and patient representatives • Concrete follow up actions, including formal implementation of a collaboration between PDCO and CTCG • Acknowledgement about the importance to extend methodology implementation activities, such as related to ICH E11A Guideline to include also, e.g. clinical trial assessors and ethics committees • Acknowledgement to everyone who has contributed to this workshop. • Acknowledgement to everyone who has contributed to this workshop.assessors, ethics committees, HTA bodies and payers (for downstream alignment) 8 Report: Workshop for assessors of paediatric clinical trials More information The Accelerating Clinical Trials in the EU (ACT EU) initiative aims to develop the European Union further as a competitive centre for innovative clinical research. ACT EU seeks to deliver on the clinical trial innovation recommendations of the European medicines agencies network strategy and the European Commission’s Pharmaceutical strategy for Europe. ACT EU builds on the Clinical Trials Regulation (CTR) and Clinical Trials Information System (CTIS) launched on 31 January 2022. The European Commission, EMA and Heads of Medicines Agencies launched ACT EU in January 2022 and run the initiative together, establishing a steering group in March 2022. The programme's strategy paper features ten priority action (PA) areas that are the basis for the ACT EU workplan. The third version of the ACT EU workplan was published on December 2024 and sets out deliverables and timelines for the programme for 2025-2026. 9 Report: Workshop for assessors of paediatric clinical trials Glossary ACT EU Accelerating Clinical Trials in the European Union CTA Clinical Trial Application CTCG Clinical Trials Coordination Group CTR Clinical Trial Regulation (Regulation (EU) No 536/2014 of the European Parliament and of the Council) EC Ethic Committee EU/EEA European Union/ European Economic Area ICH E11 International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use, Guideline on Clinical Investigation of Medicinal Products in the Paediatric Population MS Member States MWP Methodology Working Party NCA National Competent Authority NCA National Competent Authority PDCO Paediatric Committee PIP Paediatric Investigation Plan 10 Report: Workshop for assessors of paediatric clinical trials European Medicines Agency Domenico Scarlattilaan 6 1083 HS Amsterdam The Netherlands Telephone +31 (0)88 781 6000 Send a question www.ema.europa.eu/contact www.ema.europa.eu © European Medicines Agency, 2023. Reproduction is authorised provided the source is acknowledged.
accelerating-clinical-trials.europa.eu
October 27, 2025 at 4:12 PM
Nieuws van ACT EU: ACT EU workshop on paediatric clinical trials report
Meeting report
Workshop for assessors from NCAs and MRECs on paediatric clinical trials Meeting report Contents Introduction .......................................................................................................... 3 Setting the scene ................................................................................................... 3 Panel discussion .................................................................................................... 4 Breakout sessions .................................................................................................. 5 Conclusions .......................................................................................................... 5 Next steps ............................................................................................................ 6 Closing remarks from CTCG, PDCO and MWP ............................................................. 7 More information ................................................................................................... 8 Glossary .............................................................................................................. 9 3 Report: Workshop for assessors of paediatric clinical trials Introduction On 14 and 15 July 2025, the Clinical Trial Coordination Group (CTCG), Paediatric Committee (PDCO) and Methodology Working Party (MWP) organized a workshop for assessors from NCAs and ethics committees to discuss real cases of paediatric clinical trial applications which show different assessment approaches across EU/EEA. This workshop is a direct follow up to the ACT EU multi-stakeholder methodology workshop, which identified harmonization in the interpretation of CTR Article 32 (inclusion of minors in clinical trials) in the assessment of paediatric clinical trials as a priority. The objective of the workshop was to strengthen collaboration between national competent authorities, CTCG, ethics committees and the PDCO by bringing together all regulatory decision makers in the ecosystem of paediatric drug development. Through discussion and exchange on real use cases, the workshop provided an opportunity to address the challenges of assessing paediatric clinical trials under the Clinical Trials Regulation (CTR) as well as to develop together with the participants potential solutions for addressing inconsistencies in the interpretation of Article 32 of the CTR, in context of the revised Declaration of Helsinki, while ensuring ethical and scientific rigor. Setting the scene Moderators: Marianne Lunzer (CTCG chair, Austrian Agency for Health and Food Safety (AGES), Austria) and Monique Al (Central Committee on Research Involving Human Subjects (CCMO), Netherlands)/Vice- chair CTCG/Co-chair MedEdthicsEU). Presenters: Laura Fankhauser (Hopp Children’s Cancer Center Heidelberg (KiTZ) Germany), Dominik Karres (Paediatric Medicines Office EMA) and Sabine Scherer (PDCO member, Federal Institute for Drugs and Medical Devices (BFARM) in Germany), Martine de Vries (Head of the Department of Medical Ethics and Health Law Paediatrician Leiden University Medical Center, Department of Medical Ethics & Health Law, The Netherlands). Results from a descriptive analysis of paediatric clinical trial assessments revealed that there is significant variability in how Member States assess trials. Particularly, differences were noted in the interpretation of Article 32(1)(e), (f), (g) of the CTR (especially around “direct benefit” and the required evidence, e.g. Key messages • Despite regulatory progress, children remain underrepresented in clinical development, delaying access to innovative treatments and increases off-label use. • There are differences in interpretation of Article 32 and variability in how Member States assess paediatric clinical trials. • The revised Declaration of Helsinki emphasises that excluding vulnerable groups (like children) from research can potentially perpetuate or exacerbate their disparities. • Patients and caregivers are often willing to accept higher risks and burden, especially in life- threatening conditions. • Greater involvement of patient representatives in trial planning, regulatory and ethical discussions is essential. 4 Report: Workshop for assessors of paediatric clinical trials proof of concept; need for adult evidence). With regards to the inclusion of adolescents in adult trials, Member States differ in their acceptance, often requiring additional adult data to be generated first. It also showed that there is inconsistent use of paediatric investigation plans (PIPs) in assessments, with some Member States being more conservative, leading to none, delayed authorisation or authorisation only with certain conditions of trials despite the clinical trial application (CTA) being in line with an agreed PIP; appreciating that of course evidence may have evolved after the initial PIP agreement over time This first description of this as-is analyses provided the foundation for the subsequent discussions. It underpinned the need for more harmonised interpretation of Article 32, but also value submission of the full PIP into a CTA, to allow scientific considerations reflected upon during the clinical trial assessment process. It was emphasised that there are common challenges within the ecosystem of drug development in general. Those hold true across diseases and populations. But it remains key to acknowledge paediatric specificities, as represented by the EU Paediatric Regulation. Paediatric specific clinical development challenges, mainly related to small patient numbers due to disease prevalence, reflected upon in context of current methodological guidelines, such as ICH E11A on the use of extrapolation were discussed. It was emphasised that the key principle behind the use of extrapolation is to avoid unnecessary clinical trials in children whenever possible. And to using existing evidence from across populations and/or same in class products, as appropriate, in support of a paediatric clinical trial design, which addresses clear scientific questions in order to move forward in addressing identified unmet medical needs. This was complemented by the experience from the PDCO and how these considerations are reflected in the PIP assessment. From the regulation, the discussion moved to the principle of equitable access to health care through research and how to view paediatric clinical trials in context of the revised Declaration of Helsinki and CRT Article 32. It was highlighted that the revised Declaration of Helsinki emphasises that excluding vulnerable populations (like children) from research can potentially perpetuate or exacerbate their disparities. Therefore, the harms of exclusion must be considered and weighed against the harm of inclusion. This paradigm shifts from a “no unless” to a “yes, if justified” mindset should be taken into account when designing and reviewing paediatric clinical trials; as in line with relevant guidelines, such as E11A. It was concluded that justice and equitable access to research must be central to a scientific and ethical review. Panel discussion Moderator: Anette Solli Karlsen (PDCO member, Norwegian Medical Products Agency (NOMA), Norway) • Industry perspective • Academic perspective • Patient perspective Panellists: Scott Diede from Industry (MSD), Michel Zwaan from Academia [Prinses Maxima Centre Utrecht, Netherlands] and Tomasz Grybek from Patient perspective [Eurordis Rare Diseases Europe] Laura Fankhauser (Hopp Children’s Cancer Center Heidelberg (KiTZ) Germany), Dominik Karres (Pediatrics Medicines Office EMA) and Sabine Scherer (PDCO member, BFARM Germany), Martine de Vries (Head of the Department of Medical Ethics and Health Law Paediatrician Leiden University Medical Center, Department of Medical Ethics & Health Law, The Netherlands) 5 Report: Workshop for assessors of paediatric clinical trials The panellists provided additional perspectives, with the industry representative highlighting challenges with inconsistent assessments and subsequent delays in trial approvals. The academic perspective stressed the need for early engagement with regulators and patients/ caregivers at stage of trial design and pointing to the need for paediatric clinical trials which are acceptable to all regions globally. The patient/ parent representative stressed that patients and caregivers are often willing to accept higher risks, especially in life-threatening conditions and raised the concern that their voices remain underrepresented in trial design discussions and in clinical trial assessments. The panel discussed the value of potential alignment in the interpretation of Article 32 across NCAs and ethics committees. Reflecting on ways to improving exchange and learnings across regulatory decision makers (such as PDCO, CTCG/ NCAs and ethics committees). It was discussed how established platforms, like the CTCG assessor’s roundtable can be used more targeted and efficient to enable discussions and alignment. The challenges in applying the revised Declaration of Helsinki and CIOMS guidelines consistently were touched upon, pointing to the need for ways to further enhancing ethical review practices. Breakout sessions Moderators: Marianne Lunzer (CTCG chair, AGES Austria) and Monique Al (CCMO Netherlands /Vice-chair CTCG/Co-chair MedEdthicsEU) Breakout group topics The breakout session groups were introduced. This were closed meetings for NCA and Ethic Committees (EC members only). Real use cases of clinical trial applications were discussed. The use cases were chosen to represent of the following key issues: Dosing; clinical trials without prospect of direct benefit; what is sufficient proof of concept and how much adult data is needed; methodological challenges in relation to extrapolation; inclusion of adolescents in adult trials; situation of prospect of direct benefit/benefit to the group; minimum burden and risk; challenges seen from a patient perspective; how to support a global development. Five groups discussed in individual breakout sessions these use cases with a focus on potential solutions. Feedback from the breakout groups and Conclusions Moderators: Marianne Lunzer (CTCG chair, AGES Austria) and Monique Al (CCMO Netherlands /Vice-chair CTCG/Co-chair MedEdthicsEU) A recurring theme coming out of all five groups was the need for better alignment: • Between (NCAs) and Ethics in context of clinical trial approvals. • Inconsistent interpretations of Article 32 of the CTR and the revised Declaration of Helsinki remains a barrier in the assessment of paediatric clinical trials. • Between regulatory decision makers such as PDCO and NCAs and Ethics in context of a PIP agreement leading to a clinical trial application 6 Report: Workshop for assessors of paediatric clinical trials The groups discussed the cases in light of the interpretation of Article 32 (CTR), with a particular focus on: • Article 32.1(e) on the amount of adult data needed before including minors • Article 32.1(f): relation to the medical condition of the minor • Article 32(g): direct benefit vs group benefit with minimal risk/burden. Here the value to develop a common EU-wide interpretation were emphasized, especially regarding: • What constitutes “minimal additional risk.” • When group benefit justifies inclusion without direct benefit. The value of inclusion of adolescents in adult trials was raised, with a strong support for including adolescents when scientifically justified. Here it was proposed for sponsors to justify exclusion, not just inclusion. It was however flagged that some Member States may have national laws or practices that hinder such inclusion, pointing to the need for a better understanding of national legal frameworks. All groups agreed that there remains a need to further develop and/or update practical tools and guidance, such as: • Calling for a decision-making framework supporting the assessment of paediatric trials. • Consider updating the recommendation paper on ethical considerations in clinical trials with minors. • Considering developing a sponsor guidance on what to include in a paediatric CTA, including full PIP Decision, including its Annexes if available). • Need for further regulatory clarification on how to apply e.g. use of extrapolation. Next steps Moderators: Marianne Lunzer (CTCG chair, AGES Austria) and Monique Al (CCMO Netherlands /Vice-chair CTCG/Co-chair MedEdthicsEU) Analyses to better understand hurdles in MSs: Survey on national laws Current ‘guidance landscape’ fit for purpose? Analyse the document Ethical considerations for clinical trials with minors to see if an update is needed in the light of the workshop discussions and conclusions Need to develop an assessor’s best practice document on considerations specific for paediatric trials Guidance for sponsors directing towards good justification of inclusion of minors and provision of full PIP as part of the submission dossier, early engagement of investigators and patients, guidance should aim to explain paradigm shift (DoH, CTR) Enable information sharing, discussions and learnings across decision makers (from PIP to CTA): Organise meeting on interpretation of CTR art 32 e, f, g (PDCO, NCA, Ethics) Implement process for sharing and discussing critical cases between CTCG and PDCO 7 Report: Workshop for assessors of paediatric clinical trials Involvement of stakeholders, but also regulatory decision makers, such as ethics committees in guidance/Q&A updates (such as Q&A on paediatric dosing) created by PDCO, CTCG and MWP as co- author or in commenting phase Closing remarks from CTCG, PDCO and MWP Moderators: Marianne Lunzer (CTCG chair, AGES Austria) and Monique Al (CCMO Netherlands /Vice-chair CTCG/Co-chair MedEdthicsEU), Brian Aylward (PDCO chair), Kit Roes (MWP chair) Key messages • Timely workshop to address relevant issues related to assessment of paediatric clinical trials in EU/EEA • Clear message to continue this dialogue between regulatory decision makers, Ethics, investigators, sponsors and patient representatives • Concrete follow up actions, including formal implementation of a collaboration between PDCO and CTCG • Acknowledgement about the importance to extend methodology implementation activities, such as related to ICH E11A Guideline to include also, e.g. clinical trial assessors and ethics committees • Acknowledgement to everyone who has contributed to this workshop. • Acknowledgement to everyone who has contributed to this workshop.assessors, ethics committees, HTA bodies and payers (for downstream alignment) 8 Report: Workshop for assessors of paediatric clinical trials More information The Accelerating Clinical Trials in the EU (ACT EU) initiative aims to develop the European Union further as a competitive centre for innovative clinical research. ACT EU seeks to deliver on the clinical trial innovation recommendations of the European medicines agencies network strategy and the European Commission’s Pharmaceutical strategy for Europe. ACT EU builds on the Clinical Trials Regulation (CTR) and Clinical Trials Information System (CTIS) launched on 31 January 2022. The European Commission, EMA and Heads of Medicines Agencies launched ACT EU in January 2022 and run the initiative together, establishing a steering group in March 2022. The programme's strategy paper features ten priority action (PA) areas that are the basis for the ACT EU workplan. The third version of the ACT EU workplan was published on December 2024 and sets out deliverables and timelines for the programme for 2025-2026. 9 Report: Workshop for assessors of paediatric clinical trials Glossary ACT EU Accelerating Clinical Trials in the European Union CTA Clinical Trial Application CTCG Clinical Trials Coordination Group CTR Clinical Trial Regulation (Regulation (EU) No 536/2014 of the European Parliament and of the Council) EC Ethic Committee EU/EEA European Union/ European Economic Area ICH E11 International Council for Harmonisation of Technical Requirements for Pharmaceuticals for Human Use, Guideline on Clinical Investigation of Medicinal Products in the Paediatric Population MS Member States MWP Methodology Working Party NCA National Competent Authority NCA National Competent Authority PDCO Paediatric Committee PIP Paediatric Investigation Plan 10 Report: Workshop for assessors of paediatric clinical trials European Medicines Agency Domenico Scarlattilaan 6 1083 HS Amsterdam The Netherlands Telephone +31 (0)88 781 6000 Send a question www.ema.europa.eu/contact www.ema.europa.eu © European Medicines Agency, 2023. Reproduction is authorised provided the source is acknowledged.
accelerating-clinical-trials.europa.eu
October 27, 2025 at 3:36 PM
Nieuws van BAREC: Save the date: Symposium 2026
Save the date: Symposium 2026
We are delighted to invite you to our next joint CT-College – BAREC symposium on 28 March 2026.
barec.be
October 20, 2025 at 4:10 PM