Presentation is loading. Please wait.

Presentation is loading. Please wait.

Research Guidelines Research Governance

Similar presentations


Presentation on theme: "Research Guidelines Research Governance"— Presentation transcript:

1 Research Guidelines Research Governance
EU Directive on Clinical Trials ICH-GCP: Informed Consent Safety & Adverse Events Documentation - Audit

2 ICH definition - GCP "A standard for the design, conduct, performance, monitoring, auditing, recording, analyses, and reporting of clinical trials that provides assurance that the data and reported results are credible and accurate, and that the rights, integrity, and confidentiality of trial subjects are protected" ICH E6 1.24

3 ICH-GCP The International Conference on Harmonisation of Technical Requirements for Registration of Pharmaceuticals for Human Use ICH-GCP - Good Clinical Practice guidelines agreed at the conference This module will cover what is GCP; the major milestones in its development ; where we are to date and finally an overview of the responsibilities GCP outlines for those involved in clinical trials. These responsibilities will fall in to the 4 categories of consent, safety, data handling and study product. These 4 categories will be covered in more depth in the other 4 modules. Liz - I understand that you are producing the slides to introduce the other four modules.

4 The objectives of ICH GCP Guidelines
Developed with consideration of the current good clinical practices of the European Union, Japan & USA, plus those of Australia, Canada, the Nordic countries & World Health Organisation. Provide a unified standard for the European Union, Japan & USA to facilitate the mutual acceptance of clinical data by the regulatory authorities in these jurisdictions. This module will cover what is GCP; the major milestones in its development ; where we are to date and finally an overview of the responsibilities GCP outlines for those involved in clinical trials. These responsibilities will fall in to the 4 categories of consent, safety, data handling and study product. These 4 categories will be covered in more depth in the other 4 modules. Liz - I understand that you are producing the slides to introduce the other four modules.

5 Good Clinical Practice - GCP
What is GCP? “Ethical and scientific quality standards for designing, conducting, recording and reporting trials that involve participation of human subjects” Why is it needed? To ensure that the RIGHTS, SAFETY and WELLBEING of the trial subjects are protected Ensure the CREDIBILITY of clinical trial data Why has it developed into formal guidelines? Public disasters, serious fraud and abuse of human rights Liz - the bits in blue need to be ‘flashed’ in - I don’t know how to do it. Good clinical practice: “A standard for the design, conduct, performance, monitoring, auditing, recording, analyses, and reporting of clinical trials that provides assurance that the data and reported results are credible and accurate, and that the rights, integrity, and confidentiality of trial subjects are protected.” ref: ICH-GCP June 1996. In order to be able to unpack this slightly wordy explanation a little, we need to be aware of why GCP guidelines have come into existence and why these guidelines have become formal and soon be be integrated into legislation. ? Add here the latest EU directives?

6 ICH GCP Guidelines cover…
Ethics Committee /IRB Sponsor Investigator Investigator’s Brochure Essential Documents for conduct of a trial Clinical Trial Protocol & Amendments EU directive will ensure the same level of patient protection, scientific standards, it will rationalise administrative procedures across member states it is unlikely that the above timeframes will be upheld - more likely that it’s adopted 2001, implemented the following year.

7 Responsibilities of the Investigator
Agreements Compliance Ethics Informed Consent Investigational drug Qualifications Medical care of trial subjects Randomization Records Reports Resources Safety reporting Unblinding EU directive will ensure the same level of patient protection, scientific standards, it will rationalise administrative procedures across member states it is unlikely that the above timeframes will be upheld - more likely that it’s adopted 2001, implemented the following year.

8 World Medical Association Declaration of Helsinki
Adopted by 18th WMA Assembly, June 1964 (Helsinki, Finland) Covers all “medical research” Most recent amendment October 2000 changes include: Peer review of protocol,supervision of research, results to be made available, test against “gold standard”, access to best proven treatment at end of research.

9 Research Governance Framework

10 Scope of the Framework Research by staff with Trust and Honorary Trust Contracts Research involving patients, service users, care professionals, volunteers or their organs, tissues or data Research funded by the NHS Research using facilities funded by the NHS

11 Aims of the Framework set out in legislation and regulations
To promote a quality research culture To promote excellence To provide strong leadership for research To implement standards: set out in legislation and regulations required by the Department of Health established as good practice

12 Research Governance domains
Ethics Science Information Health, Safety and Employment Finance and Intellectual Property

13 Ethics - key points Independent review of research involving patients, service users, care professionals, volunteers, or their organs, tissues or data Consideration of the dignity, rights, safety and well being of participants Informed consent Data protection Consumer involvement Diversity of human culture Avoidance of animal experiments

14 Science - key points Peer review – commensurate with the scale of research Regulation by the MHRA and MDA Data retention to allow further analysis and support monitoring

15 Information - key points
Free access to information on research being conducted and research findings Publication of results in a format understandable to the public Making findings available to participants

16 Responsibilities of the Researcher
Developing proposals Seeking ethical committee approval Conducting research according to the agreed protocol Ensuring participant welfare Feeding back results to the participants

17 Responsibilities of the Sponsor
Assuring scientific quality (peer review) Ensuring research ethics committee approval Ensuring arrangements for the management and monitoring of research

18 Responsibilities of the Care Organisation
Ensuring that research using their patients, users, carers or staff meets the standards in the Research Governance Framework Ensuring ethics committee approval Retaining responsibility for research participants’ care

19 Sanctions “the assumption will be that there is full compliance (or else research is stopped)”

20 The Medicines for Human Use (Clinical Trials) Regulations 2004
The EU Clinical Trials Directive 2001/20/EC The Medicines for Human Use (Clinical Trials) Regulations 2004

21 Aims of the EU Directive
To simplify and harmonise the administrative provisions governing clinical trials in the EU To facilitate the internal market in medicinal products To maintain appropriate protection for public health

22 ICH-GCP is to be the GCP standard
Application of GCP ICH-GCP is to be the GCP standard (EU GCP Directive 2005) All drug trials will need: an ICH-GCP compliant protocol an investigator brochure case report forms a site file to report SAEs

23 The regulations cover:
All medicinal products – medicinal by function or presented as treating or preventing disease in human beings (including health products) All phases of trials - including Healthy Volunteer Trials Trials sponsored by industry, government, charities, universities, NHS organisations Trials recruiting participants after 1st May 2004 – the regulations are RETROSPECTIVE Devices are exempt if looking at mode of delivery (comparing devices) rather than dosage   Further guidance is still awaited

24 EU Directive & Informed Consent
Standards currently adopted in the UK comply with the Directive However, the Directive states that a “legal representative” may act for a trial subject that is not able to give informed consent A formal mechanism to appoint a legal representative for incapacitated adults will be introduced

25 Indemnity/Compensation
Provision must be made for compensation for non-negligent harm for all trials e.g The clinical negligence scheme for Trusts University Public Liability Insurance Specific Clinical Trials insurance ABPI model indemnity agreements to used for industry sponsored trials

26 Licensing Authority Approval
UK “competent authority” will be the Medicines and Health-care products Regularity Agency (MHRA) Prior authorisation is required for all trials and substantial amendments Consideration the MHRA within 30 days, extendable to 60 days SAE reporting protocols Report to the MHRA within 90 days of trials conclusion If trial is terminated or ended prematurely notification required within 15 days

27 EU Directive & Ethics Committees
60 day time limit for decisions A single request for additional information Approval or Rejection decisions Approval to take place in parallel with licensing authority approval Extended time limits for gene therapy, somatic cell therapy, medicinal products containing Genetically Modified Organisms and xenogenic cell therapy trials 35 day time limit to review amendments If trial is terminated or ended prematurely notification required within 15 days Substantial amendments to be notified

28 Substantial Amendments
Amendments are considered to be substantial if they are likely to impact on: Subject safety Scientific value of the trial Conduct or management of the trial Quality or safety of the IMP Duration of the trial - anything other than minor administrative amendment

29 EU Directive & Competent Authority - Information to Support Approval
Core information - Required by the Directive Covering letter EUDRACT form - register via MHRA Protocol Investigator Brochure Investigational medicinal products dossier - Ph1 trials List of CA to which an application has been made EC opinion if available Member State Specific Local information: consent forms, information sheets, recruitment, indemnity, manufacturing of IMPs

30 EU Directive & Good Manufacturing Practice and Investigational Medicinal Products
Manufacture or importation from countries outside the European Economic Area will require prior Licensing Authority authorisation All products to be approved by a “Qualified Person” (QP) as compliant with GMP Labelling requirements Unlikely that Manufacturer’s licence and QP will be required to cover reconstitution and packing of IMPs by hospital Pharmacists

31 GCP and GMP Inspections
Inspections to be undertaken (by MHRA) of trial sites, manufacturing sites, laboratories used for analysis and sponsors premises Charges to be levied, amount still to be determined Inspections will be of two types: Cyclical, systems based inspections of sponsors Triggered inspections as required

32 Safety Reporting PI to identify category
Complete Safety Report Form (s) Forward as appropriate to Trust and Sponsor within 24hrs If SUSAR Sponsor to forward to REC, MHRA and relevant CA outside UK if applicable (EUDRAVIGILANCE database). N.B. It is relevant to report events that may be related to placebo or reference drugs N.B. If SAE for non-CTIMP report within 15 days using non-CTIMP SAE form (COREC/UHL website)

33 Timelines for Sponsor for further reporting
Fatal /life threatening report within 7days to MHRA ‘Other’ report within 15 days to MHRA Further information to MHRA or/and REC within 8 days if requested Annual Safety Report listing all SARs and SUSARs for the study on anniversary of CTA approval (PI responsibility)

34 The Medicines for Human Use (Clinical Trials Regulations) 2004
Order came into force in the UK on 1 May 2004 following the EU Directive on Good Clinical Practice in clinical trials 2001/20/EC (The Clinical Trials Directive). Clinical Trials Authorisation has now replaced the DDX / CTX

35 The MHRA have produced a short description of the (the UK order which implements the EU Directive) which aims to help those involved in the conduct of clinical trials to follow and understand the Regulations

36 GOOD CLINICAL PRACTICE
Consent

37 What is Informed Consent?
"Informed consent is a process by which a subject voluntarily confirms his or her willingness to participate in a particular clinical trial, after having been informed of all aspects of the trial that are relevant to the subject's decision to participate” ICH 1.28

38 Who can consent a subject?
A medically qualified person (usually) - however, Declaration of Helsinki states “physician” UHL has a policy for Nurses & AHPs obtaining consenting for clinical trials – each study assessed and training package implemented as necessary. Not automatic right Consent may be delegated to a sub investigator (needs to be documented) – must have be approved by LREC The investigator retains overall responsibility Consent must be documented in the medical notes

39 When should a subject be consented?
Prior to participation in a trial Before ANY trial procedure - including the taking of blood to screen patients if it is not part of normal clinical practice or a questionnaire to access health etc

40 How should someone be consented?
The consent form must have been approved by the Ethics Committee There should be no coercion to enter the trial Non-technical language must be used The information must be presented to the subject in the most appropriate way The subject must have “ample” time to consider their decision

41 How should a consent form be completed?
Subject must sign & date the form (& preferably write their own name) Original PIL & consent form - site file Copies of PIL & consent form - Patient notes and to the patient The consent form & patient information leaflet should always be kept together

42 GOOD CLINICAL PRACTICE
Safety & Adverse Events

43 Safety Data collected in Clinical Trials
Adverse Events Serious Adverse Events Adverse Reactions Suspected Unexpected Serious Adverse Reaction Pregnancy Lab data Vital Signs Project specific data Explanatory - essentially, all data collected in a trial can be considered safety data. Ask audience – “Can anybody tell me what safety data is collected in clinical trials”

44 Adverse Event Any untoward medical occurrence
Not necessarily causal relationship with treatment Unfavourable /unintended sign ICH (1.1) ICH GCP definition of an adverse drug reaction - ADR's reported in clinical trials before AEs

45 Adverse Reaction Untoward or unintended response to IMP ICH (1.2)
ICHGCP definition of an adverse event - can be deemed positive response as well as negative e.g. increased libido. Ensure everyone understands the differences between AEs and ADRs

46 Suspected Unexpected Serious Adverse Reaction
Unexpected-not consistent with information already available in the protocol and the IB Imperative that ALL prescribers (doctors, dentists, coroners and pharmacists) report any suspected toxicity. “Does anyone know how we go about this in the UK? The yellow card system – yellow card (found at back of BNFs, compendium of data sheets, prescriptions….) allow all prescribers to report any suspected ADRs to the MCA (Medicines control agency)/CSM (Committee on safety of medicines) – essential for monitoring drug safety. The system is still being developed – nurses used it during the meningitis C campaign and in fact the MCA are looking to extend the use of nurse reporting.  Yellow card requires details of the reporting prescriber, patient, the suspected drug (dose, dates of use) and the suspected reactions. This card scheme is in the process of changing in that personal details (name and DOB) are no longer requested, instead patients age, sex and a reference number is asked for, which enables identification – this change is to ensure that the scheme is in line with current guidelines on confidentiality. Electronic yellow cards are being piloted at the moment to make it even easier to report suspected ADRs. To help prescribers a black triangle symbol (upside down triangle!) is placed onto the drug label of a medicine that is under intensive safety monitored by the MCA/CSM. These medicines can either have been recently introduced into the UK or can be a product containing previously licensed active substances. For these drugs all suspected ADRs should be recorded. It should be noted however, that it is recommended that all ADRs in children are reported to the MCA even if there is no black triangle (from 2001 BNF 41).  For drugs that don’t have a black triangle only serious suspected ADRs should be recorded to the MCA/CSM. The black triangle drugs are monitored closely for a minimum of two years and the black triangle symbol is not removed until the safety of the drug is well established. Post market surveillance - needs to be encouraged as drug are prescribed to huge numbers of patients when marketed in comparison to during the clinical trial phase.

47 SAE,SAR or SUSAR is SERIOUS and requires reporting if it:
Results in death Is life threatening Requires hospitalisation or prolongation of stay Results in persistent or significant disability/incapacity Consists of congenital anomaly or birth defect Other Exacerbation – Asthma attacks Increase in frequency and intensity – Migraines Diagnosis of condition after trial started that may have been present before the trial started – cancer/tumours Continuous disease worsens – symptoms of a condition that was not excluded by the study protocol that worsens.

48 Continued Results in death
- Record the event that lead to death as the SAE - “Death” is the outcome Life threatening - “The patient was, in the view of the investigator, at immediate risk of death from the event as it occurred. It does not include an event that, had it occurred in a more serious form, might have caused death”

49 Continued Prolonged hospitalisation - Record diagnosis NOT procedure
- Hospitalisation = in-patient admission - Not out-patient appointments or A & E visits Disabling or incapacitating - Event which is disabling or incapacitating or causes a disruption of one’s ability to carry out normal life functions or daily activities

50 Continued Congenital anomaly
- Diagnosed in the offspring of a subject who received study drug Other - Additional option given by some pharmaceutical companies - Event not covered by SAE categories but in the investigator’s opinion, should be considered serious

51 Pregnancy - what and when to report
Protocol specific - report on SAE or pregnancy reporting form All need reporting - don’t forget the female partners of men participating in the trial too!! All need to be followed up - Length of follow-up depends on drug Pregnancies can be reported as an SAE or on a pregnancy report – dictated by the Sponsor Company. Pregnancies are usually followed up throughout the pregnancy until approx. 3 months after birth (dictated by sponsors) – pharmaceutical companies tend to send out questionnaires at birth, 3 months and later if needed. Important to report and follow up if a partner of a subject participating in a clinical trial becomes pregnant. E.g. If partner becomes pregnant of someone participating in a HIV trial it is likely that the child will have HIV – need to know the combination of the drug that the child would have been exposed to be able to provide the new born child with the correct combination of medication.

52 GOOD CLINICAL PRACTICE
Documentation & Audit

53 Essential Documents “Are those documents, which permit evaluation of the conduct of the trial and the quality of data produced. These documents serve to demonstrate the compliance of the investigator, sponsor and monitor with the standards of GCP and with all regulatory requirements” ICH (8 .1) Slide 4 – Essential Documents This is ICH GCP definition (read).

54 Minimum list - see ICH section 8
Essential Documents Documents which… are audited by the regulatory authorities or sponsor company to confirm the validity of the data & integrity of the data collected are contained in the files established at the beginning of the trial at sponsors office and investigators site Minimum list - see ICH section 8

55 Essential Documents - source data
Records should be accurate, complete, legible & timely ICH (4.9.1) Data should be consistent with source documents (or discrepancies explained) ICH (4.9.2) Any changes should be initialled, dated & explained Document all deviations from protocol and explain ICH (4.5.3) Document all dose/therapy modifications, visits and tests not conducted ICH has a whole section dedicated to Essential Documents. Basically, the study file, as provided by the sponsor should contain all the essential documents and is your evidence of compliance with GCP, the protocol and requirements of the MCA or FDA. It will contain ALL the information required to verify the results of the study for the patients you have recruited. “Can anyone suggest the types of documents I mean by essential documents?”

56 What needs to be recorded in the patient notes?
Copy of signed and dated Consent Form and PIL Title of the trial including the drug to be received Study and patient number Visit dates Concomitant medicines taken Any adverse events A letter informing the GP that the patient has been enrolled in the clinical trial Meaning of source data is the ORIGINAL record, or the first place data is recorded, or written down. “Anyone give me some examples of source data?” (X-ray, lab report, writing BP in notes etc) Whatever form this takes, it MUST be kept. This is because every CRF entry should be verifiable against source data, known as Source Data Verification (SDV). This is mainly what your monitor will be doing. (Joanne’s example: Investigator who was called by a patient with an AE while he was at dinner, so wrote it all down on a paper napkin, which is still filed in the study file!) Explain bullets of slide e.g. timely is very important, as is initialling and dating changes. This will prevent a long list of data queries at the end of the study and ensures the data is robust.

57 Essential Documents continued
Essential documents should be retained for 2 yrs following last approval of marketing application in the ICH region (taken to be 15yrs) ICH (4.9.5) All records must be made available (direct access) for monitors, auditors & regulatory authorities ICH (4.9.7) Pharma companies should tell you this, some now archive for you. Is likely to be at least 5 years.

58 ICH definition - Audit "a systematic and independent examination of trial related activities and documents to determine whether the evaluated trial related activities were conducted, and the data were recorded, analysed and accurately reported according to the protocol, sponsor's standard operating procedures (SOPs), Good Clinical Practice (GCP), and the applicable regulatory requirement(s)" ICH E6 1.6)

59 Common audit findings Patients do not fulfil the entry criteria
Incomplete/incorrectly completed consent forms Drug accountability inadequate Hidden entry envelopes opened during study Many pharma companies now have audit departments, but you may also be audited by the MCA or FDA. “Has anyone been through an audit?” Very detailed and thorough process that will go back and check everything, every signature etc. (Joanne’s example: audit that picked up that the oxygen was out of date, many people don’t know oxygen has a shelf life.) MCA and FDA have very different approaches to auditing; FDA are distant, you won’t be allowed to give them coffee, they have a “20 minute rule” – if you cannot produce any document or information within 20 minutes of being asked, the FDA assumes it doesn’t exist, and won’t accept it after this time. This is why filing is so important. These are some common audit findings, all related to documentation, which have consistently been found for years, ie we’re not learning the lesson, and we need to….

60 Audit can be conducted to:
EU GCP Directive EU Clinical Trials Directive Protocol

61 Clinical Trial Audits Sponsor Audits - Independent & separate from routine monitoring or quality control functions and evaluate trial conduct & compliance with the protocol, SOPs, GCP & applicable regulatory requirements FDA Audits - In USA inspections occur without notice. In Europe, carried out with notice (about 14 days) MHRA Audits – increasingly common and at short notice

62 Audit of the SITE FILE will check ….
Approvals & correspondence – Ethics approval, all correspondence to & from Ethics /Trust , MHRA notification Laboratory – normal ranges, reports, procedures, etc Documentation – protocol & amendments (signed), Information Leaflet & Consent, signed consent forms, Investigator Brochure, Indemnity, all correspondence between sponsor & investigator

63 Audit of the SITE FILE will check ….
Personnel – CVs of those working on the study, training records (such as GCP) Drug – shipping records, drug receipt - but these may be in pharmacy Patient details – screening / enrolment / identity logs, SAE reports

64 Audit of the PATIENT NOTES will check ….
Data verification –Case report forms, looking at data queries, lab results, ECGs, x-rays etc Data verification patient notes – protocol details/number in the notes, date of birth, vital signs, all visit dates, concomitant medication & changes, medical examination, study specific procedures

65 An audit may also look at-
Drug storage – accountability, temperature logs, security, dispensing, labelling, Laboratories – procedures, handling samples, accreditation Other study specific procedures

66 The Inspection Report Contains details of all finding
Each finding labelled “Critical”, “Major”, “Minor” or “For Note” Each finding is referenced to the particular regulation / guideline to which it is attributable A reply to the report is required

67 Common Audit Findings Patient or Investigator signature on consent form not dated No version / date on consent form Staff CVs not on file Ethics Committee member list not up to date Ethics Committee letter did not list all docs reviewed Ethics Committee letter did not contain statement of GCP compliance Drug not temperature monitored (was required) Signatures not dated Investigator failed to report SAEs

68 Write it down– if it’s not written down it didn’t happen
In summary Legislation will affect us all - we all have to work to GCP and Research Governance guidelines The legislation aims to improve standards and build public confidence Ensure that: The site file and filing is up to date – have a complete “paper trail” The PIL & consent form in use are the most up to date and approved by LREC People obtaining informed consent have been “approved” Write it down– if it’s not written down it didn’t happen

69 Remember….. “ the assumption will be that there is full compliance (or else research is stopped)”

70 Useful Contacts in Research and Development at LGH
Office Manager - Nicky Turner 4109 Programme Board Co-ordinators - Jill Horsley 8239 Gemini Davda Marlene Chapman 8246 Clinical Trials Pharmacist – Claire Khalil 8241 Clinical Trainer - Sarah Nicholson 8180


Download ppt "Research Guidelines Research Governance"

Similar presentations


Ads by Google