Skip to main content

SOP on FDA-Regulated Investigational Device Exemption (IDE) Application

 Title: Standard Operating Procedure for FDA-Regulated Investigational Device Exemption (IDE) Application

Introduction and Purpose

Conducting a clinical investigation, often referred to as a clinical trial, under the umbrella of an FDA-Regulated Investigational Device Exemption (IDE) application entails navigating a complex set of FDA regulations, requirements, and obligations. The FDA places the responsibility for ensuring compliance with these regulations, requirements, and obligations squarely on the sponsor of the IDE application, as delineated in 21 CFR Part 812, Subpart C. While sponsors of IDE applications are typically pharmaceutical and device companies, the FDA's IDE regulations extend the sponsorship eligibility to individuals, governmental agencies, or academic institutions (21 CFR Part 812.3).

The primary purpose of this Standard Operating Procedure (SOP) is threefold:

  • To assist in determining when an IDE is required.
  • To provide guidance and resources to individuals conducting clinical investigations, including sponsor-investigators, on obtaining an IDE for the device under study.
  • To address the process for compassionate use, also known as expanded access, and emergency use of an investigational device.

IDE Definitions and Acronyms

  • 510(k) Pre-Marketing Notification: An FDA application type for high-risk Class III devices. In most cases, an IDE is required to clinically evaluate devices subject to PMA (Premarket Approval) requirements.
  • Classes of Devices: The FDA classifies devices into three categories based on safety and effectiveness levels.
  • Class I: Low-risk devices, generally exempt from FDA regulations. Exceptions exist if the device's use is of substantial importance in preventing health impairment or presents unreasonable risk.
  • Class II: Moderate-risk devices that often require 510(k) clearance. Clinical investigations may be necessary in specific cases to demonstrate substantial equivalence.
  • Class III: High-risk devices mandating Premarket Approval (PMA) due to a need for safety and efficacy establishment. Clinical investigations are usually essential.
  • Device: Any instrument, apparatus, implement, or other article intended for diagnosing, curing, mitigating, treating, or preventing disease, or affecting the body's structure or function, not relying primarily on chemical action and not needing metabolism to function. This also includes investigational software.
  • Emergency Use: An FDA Emergency Use exemption allowing the use of an investigational device in a life-threatening situation when no standard treatment is available. Emergency use is considered clinical care, not research.
  • Expanded Access: Mechanisms allowing the use of an investigational device outside of a formal clinical trial. Also known as compassionate use, it permits patients to access the product when they're ineligible for or no clinical trials are available.
  • HUD (Humanitarian Use Device): A device benefiting patients with a disease or condition affecting fewer than 4,000 individuals in the U.S. per year.
  • Investigational Device Exemption (IDE): Authorization from the FDA to use an investigational device in human subjects, typically when the device lacks FDA marketing approval.
  • IDE Application: Document submitted to the FDA seeking permission for clinical studies involving significant risk devices that are new or not approved for a specific use.
  • Nonsignificant Risk Device: An investigational device not classified as significant risk, usually exempt from IDE requirements.
  • PMA (Premarket Approval): FDA's review process to assess safety and effectiveness for Class III medical devices, mandatory before marketing.
  • Significant Risk Device: An investigational device meeting specific criteria indicating potential serious risks to subjects' health, safety, or welfare.
  • Sponsor-Investigator: An individual initiating and overseeing the investigation, responsible for both investigator and sponsor duties.
  • Clinical Research: Research involving human participants.
  • Clinical Trial: A specific type of clinical research designed to evaluate an intervention's effect on health-related outcomes.
  • CFR (Code of Federal Regulations): The codification of general and permanent rules and regulations published in the Federal Register by the executive departments and agencies of the federal government.
  • FDA (United States Food and Drug Administration): The federal agency responsible for regulating and supervising various aspects of food, drugs, medical devices, and more.
  • IRB (Institutional Review Board): A committee responsible for the ethical and regulatory oversight of research involving human subjects.
  • SOP (Standard Operating Procedure): A documented set of instructions and protocols to standardize and guide specific processes.

IDE Guidance

IDE Requirements: Clinical investigations involving devices vary in regulatory control based on risk levels. IDE regulations differentiate between significant and nonsignificant risk device studies, with different approval processes. Every clinical investigation involving devices must possess an approved IDE or be exempt from IDE regulations. Exemptions include the use of legally marketed devices, non-invasive diagnostic devices, veterinary devices, and devices for research with laboratory animals, among others.

IDE Pre-Submission Process: Sponsor-investigators are encouraged to engage with the FDA before submitting an IDE application. This is especially valuable for first-time sponsors or those exploring new technologies or uses. Early interaction helps sponsors understand FDA requirements, regulations, and guidance, expedites the regulatory process, and fosters mutual understanding. Pre-submissions can be used to discuss potential IDEs and to determine their risk classification.

Initial IDE Submission: Sponsor-investigators bear the responsibility of preparing and submitting the IDE application to the FDA. Each IDE application's content varies depending on the investigational device and the study's specifics, but it generally includes a complete protocol, informed consent, and information about the device. The FDA has 30 calendar days to review the IDE submission, during which they may request additional information. The investigator must not begin the clinical study during this review period. After 30 calendar days, assuming no clinical hold, the study can commence.

IRB Requirements: Sponsor-investigators must ensure that an Institutional Review Board (IRB) reviews and approves the study before initiation. The IRB may require proof of IDE submission or waivers with the initial submission. The Pre-IDE Audit must be completed before IRB approval. This audit process is initiated automatically upon IRB submission and includes a Pre-IDE Checklist.

Significant Risk Devices: These devices pose serious risks and require both FDA and IRB approval before initiating a clinical study. The FDA notifies the sponsor upon receiving an IDE application and may approve, approve with modifications, or disapprove it. The sponsor can provide requested information or request a hearing if dissatisfied. Clinical investigations can begin 30 days after FDA receipt of the IDE application if IRB approval has been obtained.

Non-Significant Risk Devices: These devices have lower risks and only necessitate IRB approval before starting a clinical study. Sponsor-investigators of nonsignificant risk device studies do not submit IDE applications to the FDA. Instead, submissions are made directly to the IRB. If the IRB disagrees with the nonsignificant risk determination, this should be reported to the FDA within five working days.

IDE Reporting Requirements: Sponsor-investigators must adhere to specific reporting obligations during the study, including IDE supplements, changes in the investigational plan, development changes, protocol deviations, and more. Annual reports are due within 60 days of the IDE anniversary date, even if the study hasn't begun. A final study report is submitted upon study completion for IDE closure.

IDE Submission Documentation: Comprehensive records of IDE submissions, FDA responses, and relevant correspondence must be maintained for the study's duration and afterward, following Clinical Research SOP, "Study Records Management." 

References

  • 21 CFR Part 812: Investigational Device Exemptions
  • 21 CFR Part 812, Subpart C: Responsibilities of Sponsors
  • 21 CFR Part 812.3: Definitions

Document Approval

Popular posts from this blog

Human Gene Therapy for Neurodegenerative Diseases: FDA Guidance Summary

  Neurodegenerative diseases are a diverse group of disorders characterized by the progressive degeneration of the central or peripheral nervous system, and they can have various causes and clinical characteristics. This guidance document is a resource for sponsors on different aspects of product development, preclinical testing, and clinical trial design. It acknowledges the unique challenges and considerations associated with developing GT products for such complex and varied diseases. Below are the key summaries from the guidance. CONSIDERATIONS FOR CHEMISTRY, MANUFACTURING AND CONTROLS (CMC) The considerations for Chemistry, Manufacturing, and Controls (CMC) when developing gene therapy (GT) products for the treatment of neurodegenerative diseases are crucial for ensuring the safety and efficacy of these advanced therapies. Here, we will elaborate on the specific CMC considerations outlined in your text: Route of Administration and Product Volume: Neurodegenerative diseases often r

Human Genome Editing: FDA Draft Guidance Summary

Consideration for Developing Gene Editing Product  1. Genome Editing Methods: Genome editing can be achieved through nuclease-dependent or nuclease-independent methods. Nuclease-dependent methods involve introducing site-specific breaks in DNA using technologies like zinc finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), modified-homing endonucleases, and CRISPR-associated (Cas) nucleases. These breaks can lead to modification of the DNA sequence at the cleavage site. Nuclease-independent methods can change DNA sequences without cleaving the DNA and include techniques like base editing and synthetic triplex-forming peptide nucleic acids. The choice of GE technology should consider factors such as the mechanism of action, the ability to target specific DNA sequences, and the potential to optimize components for efficiency, specificity, or stability. 2. Type and Degree of Genomic Modification: Different GE approaches rely on DNA repair pathways such as ho

SOP on Investigational New Drug (IND) Application and Clinical Investigation Policy

Title: Investigational New Drug (IND) Application and Clinical Investigation Policy Introduction and Purpose: The conduct of clinical investigations (clinical trials) under an FDA-Regulated Investigational New Drug (IND) application involves compliance with a complex set of FDA regulations, requirements, and responsibilities. The FDA holds the IND application sponsor accountable for ensuring compliance with these regulations (21 CFR Part 312, Subpart D). While pharmaceutical companies are typical IND sponsors, individuals, governmental agencies, and academic institutions may also serve as sponsors (21 CFR Part 312.3). This policy serves the following purposes: To assist in determining when an IND is necessary. To provide guidance and resources for individuals conducting clinical investigations, including sponsor-investigators, on obtaining an IND for the investigational drug. To outline the process for compassionate use (expanded access) and emergency use of investigational drugs. Defi

Engineering AAV vectors for enhanced safety profiles

 Engineering AAV vectors for enhanced safety profiles involves multiple strategies at both the vector genome and capsid levels. Here is a breakdown of these strategies: Vector Genome Level: Modifying Vector Genome Sequences: Scientists modify AAV vector genomes by adding, mutating, or deleting specific sequences. For example, self-complementary AAV (scAAV) vectors are designed by deleting key signals from the second inverted terminal repeat (ITR), allowing for more efficient genome replication. Codon Optimization: Optimizing the codon usage of the transgene can enhance its expression efficiency. Promoter and PolyA Sequence Selection: Careful selection and manipulation of promoter and polyadenylation (polyA) sequences can influence transgene expression and tissue specificity. Capsid Engineering: Capsid engineering strategies can be categorized into four main categories: Directed Evolution: This approach involves creating capsid mutant libraries using error-prone PCR or introducing pepti

SOP on Safety Events Reporting In Clinical Trials

Title: Standard Operating Procedure for Collection, Evaluation, Documentation, and Reporting of Safety Events in Clinical Trials Introduction and Purpose The assessment of safety events and the accurate reporting of these events are fundamental aspects of conducting clinical trials. These processes are crucial for ensuring the safety and well-being of research participants. This Standard Operating Procedure (SOP) outlines the procedures for collecting, evaluating, documenting, and reporting safety events, including Adverse Events (AE), Serious Adverse Events (SAE), Unanticipated Problems (UP), and other relevant safety events during the course of a clinical trial. The Principal Investigator (PI) holds the primary responsibility for the overall conduct of the trial, safeguarding the rights, safety, and welfare of study subjects, and ensuring that the investigation adheres to the protocol, Good Clinical Practice (GCP), Institutional Review Board (IRB), Food and Drug Administration (FDA),