1 / 40

Risk Assessments in 510K Clinical Studies

Risk Assessments in 510K Clinical Studies. John McLane, Ph.D. COO & Vice President Clinical and Regulatory Affairs Clinquest, Inc. jmclane@clinquest.com. Clinical Trial Preparation. Do your homework: Start with the regs Literature Budget appropriately for R&D

issac
Download Presentation

Risk Assessments in 510K Clinical Studies

An Image/Link below is provided (as is) to download presentation Download Policy: Content on the Website is provided to you AS IS for your information and personal use and may not be sold / licensed / shared on other websites without getting consent from its author. Content is provided to you AS IS for your information and personal use only. Download presentation by click this link. While downloading, if for some reason you are not able to download a presentation, the publisher may have deleted the file from their server. During download, if you can't get a presentation, the file might be deleted by the publisher.

E N D

Presentation Transcript


  1. Risk Assessments in 510K Clinical Studies John McLane, Ph.D. COO & Vice President Clinical and Regulatory Affairs Clinquest, Inc. jmclane@clinquest.com

  2. Clinical Trial Preparation • Do your homework: • Start with the regs • Literature • Budget appropriately for R&D • Form a solid team of experts – • Scientific Advisory Board • Clinical Expertise • Careful Planning and Conduct are Key to Obtaining Useful Data and Assuring Human Subject Protections

  3. Medical Device Risk Regulations • Medical Devices Directive (MDD), 93/42/EC – covers all general medical devices except: • Active Implantable Medical Devices (AIMD), 90/385/EC –in humans • In Vitro Diagnostic Devices (IVDD), 98/79/EC • ISO regulations • ISO 14155 (clinical) • ISO 14971 • 21 CFR Parts 812, ICH GCP, • Japan GCPs, GHTF guidelines • EU MEDDEVs (2.7.1)

  4. ISO 14971 Risk Management • Risk Management standard • Must have that particular defined and documented RM Process that addresses • risk analysis • evaluation and control, • collection of production • post-production data to validate or change previous risk determinations

  5. ISO 14155:2011 Newly Released • Harmonize the ISO standard with the ICH-GCPs and MedDev guidances on clinical trials • Clarify the responsibilities of investigator, sponsor, monitor, and IRB/EC more thoroughly and consistently • Clarify risk and risk assessment with regard to the subject rather then device design • Update the literature review section to match MedDev and any developing GHTF documents: Investigator’s Brochure • Clarify the responsibilities of sponsors, monitors, investigators, institutions, IRBs/ECs, DSMB, auditors, and other parties • Clarify details of the Investigator ‘s Brochures, final report, adverse event handling, case report forms, and data mgmnt • Distinguish between adverse events, devise deficiencies, and deviations

  6. ISO 14155: 2011 key items • Definition of Clinical Data not included • Development of clinical data is in Annex A • Risk of Use Error (ref: ISO 14971) includes training • Use of selected literature allowed if meets similarity requirements • Intended use • Population • Conditions of use • Note: similarity differs from MDD “demonstration of equivalency” (Article 1) • Requires assessment of significance and weight of studies including published and unpublished studies • Note: FDA PMA require unpublished and published data • All AE (not just SAE) to be reported to all countries where trial conducted

  7. ISO 14155 Med Device Clinical • Step-by-step methodology, record-keeping, and reporting requirements • Ethical and legal approval requirements • Steps to construct a protocol • Risk assessment • Case report (data collection) forms • Instructions for preparing a final report

  8. Global Harmonization Task Force (GHTF) • Study Group 5 • Harmonize clinical definitions • Review applicable GHTF & ISO/ICH documents, to assure terminology is consistent and interfaces are clear • Develop guidance on how to conduct and document the clinical evaluation • Harmonise the content and format for clinical investigation reports.

  9. GHTF Essential Principles • Essential Principles of Safety and Performance of Medical Devices (Summary Technical Document (STED) • Classification and complexity of the device; • If it incorporates novel technology • If it is an already marketed device type that w/ intended use different from original use • If it is new to the manufacturer • Device type associated w/ significant number of adverse events/ user errors, novel or potentially hazardous materials, or raises specific public health concerns

  10. STED Premarket Use • General description and list specific features • Set of labels and language variants • Summary tech docs and design • Essential principles checklist • Risk analysis and control summary • Summary of V&V • Clinical Evaluation Report

  11. Are Essential Principles met? Principles of GHTF SG5 MORE NO YES REQUIRED - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - Outputs Clinical investigation Literature searching Clinical use CLINICAL EVALUATION Processes • CLINICAL INFO • - Literature • Clinical experience • Clinical Investigations • Other? DECLARATION OF CONFORMITY REPORT CLINICAL EVIDENCE ISO14155 S T E D NON CLINICAL EVIDENCE

  12. Clinical Design • Clear statement of objectives • Appropriate subject population(s) • Minimization of bias • randomization, blinding • Confounding factors • concurrent medications, co-morbidities • Appropriate controls • cohort, sham, historical • Design configuration • parallel, crossover, factorial • Type of comparison • superiority, non-inferiority, equivalence

  13. Conclusion • Consider the Clinical Trial as a comprehensive process • Get Experts (Reliance Medical Association) • Know your target product profile • Be prepared • Have the evidence • Preclinical • QSR • Work with the FDA and IRBs • Be realistic on potential risks

  14. GHTF Documents to Consider • SG1/N011:2008 Summary Technical Documentation for Demonstrating Conformity to the Essential Principles of Safety and Performance of Medical Devices (STED) • SG1/N029:2005 Information Document Concerning the Definition of the Term “Medical Device” • SG1/N041:2005 Essential Principles of Safety and Performance of Medical Devices • SG1/N040:2006 Principles of Conformity Assessment for Medical Devices • SG1/N43:2005 Labelling for Medical Devices • SG5/N1R8:2007 Clinical Evidence – Key definitions and Concepts • SG5/N2R8:2007 Clinical Evaluation

  15. Backup slides

  16. Classification Basis • Classification depends on intended use and indications for use, and level of risk • Intended use- What disease, symptom, or condition is the device intended to treat? How will the device be used? • Indications for use- What kinds of patients should this be used on? Can be based on age, disease state, medical history, allergies, etc. • Level of risk-Is the device life-saving? Is the device life-sustaining? Is there an unreasonable risk of illness or injury associated with use of the device?

  17. Device Classification Class I • Safety & effectiveness are well-established • Subject only to “General Controls” (registration, device listing, GMPs) Class II • Need “Special Controls” (guidances, postmarket surveillance, labeling, preclinical testing) Class III • General and special controls are insufficient to assure safety and effectiveness • Devices that are life-sustaining, life-supporting, or present unreasonable risk of illness or injury

  18. Protocol Deviations • CFR 812.150(a)(4) require prior approval from the sponsor of all planned deviations, including administrative and minor deviations.  • Planned deviations requested of a sponsor must be submitted for IRB review as a “Change in Research” prior to instituting any IDE research planned deviations • For device research, the PI must keep on file a copy of the written approval document from the sponsor and IRB when a deviation is granted.

  19. Medical Device Clinical Paths

  20. Core Principles • When is Clinical Data Needed To Assure That Essential Principles are Met? • Clinical Evaluation Process • Justification for Clinical Investigation • What Type / Design of Clinical Investigation is Appropriate?

  21. Studies Exempt from IDE Regulation • Legally marketed device when used in accordance with its labeling • Diagnostic device if it complies with the labeling requirements in §809.10(c) and if the testing: • Noninvasive • Does not require an invasive sampling procedure • Does not introduce energy into a subject • Has “back-up” approved confirmatory diagnostic tests • Consumer preference testing, testing of a modification, or testing of a combination of devices if the device(s) are legally marketed device(s) • Device intended solely for veterinary use or laboratory animal use

  22. Non-significant Risk Device IDE Applications • Abbreviated IDE application submitted to IRB: • Device Labeling : • CAUTION - Investigational Device. Limited by Federal (or United States) law to investigational use • Informed Consent –Investigators must obtain and document informed consent from each subject • Monitoring - All investigations must be properly monitored to protect the human subjects and assure compliance • Records and Reports - Sponsors and Investigators are required to maintain specific records and make certain reports as required by the IDE regulation • Prohibitions –Commercialization, promotion, test marketing, misrepresentation of an investigational device, and prolongation of the study are prohibited (§812.7)

  23. Test for Safety • Biocompatibilty • ISO 10993 • Rabbit epidural study • Implant – Tissue interface • Mechanical Performance • ASTM testing • Biomechanical Performance • Cadaveric, animal?? • Expulsion, subsidence, catastrophic failure

  24. Example: Implant Assessments • Static / Fatigue – endurance – 10M • Wear debris – amount & characterization • Long term creep • Quantity of Motion • Quality of Motion • How much work does the implant have to do – will affect lifespan of implant • Interface with tissue

  25. FDA Meeting Preparation • Prepare a target product profile • Key efficacy and safety objectives • Potential pt and user group description • Plan on submission questions • Keep questions focused • Don’t ask question of what you can easily find in the regulations • Can ask question to clarify approach to a regulation • Plan on providing support documentation • Evidence-based information most persuasive • Be prepared

  26. Reports of Prior Investigations • Provide all data that is relevant (whether adverse of supportive) • Including laboratory/animal data • Provide data on previous versions (models) of the device. • Explain what conclusions where reached from the clinical experience with previous device designs. • For each clinical investigation: • Rationale for subject selection • Statistical justification for N • Description of the study methods and endpoints • Efficacy and safety results (summary table AEs)

  27. Device Description • Description of each important component, property and principle of operation of the investigational device • Identify Human Factor tests • If applicable, state any anticipated change(s) in the investigational device during the course of the study • Identify potential device-related risks • Differentiate from clinical risks • Investigational use instructions

  28. Investigational Plan • Purpose • Protocol • Risk analysis • Description of device • Label to be on device • Monitoring Procedures • CRF • Patient information materials • Informed consent template

  29. Feasibility Clinical Study • Simple trial design to provide • Support for a future pivotal study • Answer basic research questions • Often not primary support for a marketing application • May be required by FDA prior to pivotal study to assess basic safety and potential for effectiveness • Endpoints and sample size generally not statistically driven • N=10-50 subjects

  30. FDA 1976 Medical Device Regulations • Prompted by Dalkon Shield IUD contraceptive device – caused injury, miscarriage, infertility • Established three classes of medical devices • Required safety and efficacy of all medical devices including diagnostic products • Required manufacturers to register with FDA and follow quality control procedures • Required pre-market approval for devices

  31. Pivotal study • Generally intended as the primary clinical support for a marketing application • Endpoints and sample size statistically driven • Assess both safety and effectiveness • Reasonable study conceptually? • Adequate preclinical validation of device? • Appropriate mitigation of potential risks? • Appropriate enrollment criteria? • Patients adequately informed? • Sample size appropriate?

  32. Key Components of Clinical Protocol • General study design • Proposed subject population • Anticipated number of subjects • Inclusion criteria • Exclusion criteria • Screening procedures • Study treatment (allocation, breaking the blind) • Follow-up assessment methods including the schedule of testing

  33. Biometrics Sections of Protocols • Identify primary effectiveness endpoint • Avoid composite or ambiguously defined terms • Describe how measured • How will safety be assessed and monitored (safety endpoint) • Not just well tolerated • Objective performance criteria • Sample size determination • Data and Safety Monitoring Committee

  34. Objective Performance Criteria • Type of comparison in medical device trials • Requires statistical pooling of prior investigations • Underlying disease and pt population well described and stable • Fixed Target(s) Positive Tx effect expected • Objective and Meaningful Standard • Provides Comparison in Evaluating Safety and Effectiveness • Usually a Rate • Surrogate for Control Group • Benchmark for Minimally Acceptable • Values • Not a Control Group

  35. Statistical Analysis Plan • Justification for sample size calculations • Type-1 error and multiplicity • Missing data handling • Assessment of critical endpoint covariates • Interim analyses and early stopping rules • Data handling • Contingency analysis • Provide enough detail to avoid ambiguity

  36. Anticipated and Unanticipated Safety Events • Use prior studies to clearly identify potential and anticipated risks • Similar devices • Engineering, animal, and human factor testing • Define how study design mitigates risk • Clinical training necessary? • Define how different safety events to be reported • Patients • Patient’s Investigator and all investigators • IRB • FDA

  37. Unanticipated Adverse Device Events (UADE) • UADEs must be reported by investigators to sponsors and reviewing IRBs as follows: • As soon as possible, but in no event later than 10 working days after the investigator first learns of the event • Sponsors must immediately conduct an evaluation of a UADE and must report the results of the evaluation to FDA, all reviewing IRBs, and participating investigators within 10 working days after the sponsor first receives notice of the effect • Guidance for Clinical Investigators, Sponsors, and IRBs. Adverse Event Reporting to IRBs—Improving Human Subject Protection Jan 2009

  38. Shared regulations with drugs • Part 50 – Protection of Human Subjects • Part 56 – Institutional Review Boards • Part 54 – Financial Disclosure by Clinical Investigators • Part 58 – Good Laboratory Practices for Nonclinical Laboratory Studies • Part 11 – Electronic Records; Electronic Signatures

  39. Adequate Monitoring • Trained monitors • Qualified investigator sites • Following the written procedures in the protocol • Collection of essential documents • Obtaining a signed investigator agreement from each participating investigator (can use FDA form 1572) • Provide investigators with the information they need to conduct the investigation properly • Documented training of all study personnel • Delegation log • Ensuring subjects sign informed consent form • Device quality check and accountability

  40. Questions for audience • Does “risk analysis”mean risks of the device or the risks of study participation? • •Is it always necessary to keep records of all the potential subjects screened? • •Must all data be copied into or collected in Case Report Forms vs. hospital records? • •Must marketed products that are studied for new uses have “Investigational”labeling? • How to integrate quality assurance for the study into the overall quality system of the Sponsor? Investigator or CRO?

More Related