Skip to main content

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.

Definitions and Acronyms:

  • Biologic: A product, including vaccines and gene therapies, applicable to the prevention, treatment, or cure of human diseases. Most biologics are considered drugs by the FDA.
  • Botanical: A finished product containing plant matter, which may include plant materials, algae, fungi, or combinations thereof.
  • DEA: Drug Enforcement Administration
  • Drug: A substance recognized by an official pharmacopeia, intended for use in diagnosis, cure, mitigation, treatment, prevention of disease, or affecting the structure/function of the body.
  • Schedule 1 drugs: Substances with no accepted medical use in the United States, often associated with high risk of substance use disorder.
  • Emergency Use: The use of an investigational product under strict criteria on a single subject in a life-threatening situation when no standard treatment is available. It is not considered research.
  • Expanded Access: Also known as compassionate use, it allows a patient to receive an investigational drug outside of a clinical trial when enrollment is not possible.
  • Investigational New Drug (IND): A request submitted to the FDA to administer an investigational drug to humans, necessary before clinical trials.
  • Sponsor-Investigator: An individual initiating and conducting an investigation and directly administering or dispensing the investigational drug.
  • Supplement: A product intended to supplement the diet, including vitamins, minerals, herbs, amino acids, dietary substances, or concentrates.
  • CFR: Code of Federal Regulations
  • CRSO: Contract Research Support Office
  • FDA: United States Food and Drug Administration
  • IRB: Institutional Review Board
  • SOP: Standard Operating Procedure

IND Guidance:

IND Requirements:

  • U.S. regulations mandate an IND before conducting clinical studies of an investigational drug.
  • Investigational use covers unapproved products or approved products used beyond labeled indications.
  • An IND may be required when the primary intent is to develop safety or efficacy information.
  • Schedule 1 drug investigations also require DEA approval.
  • Clinical investigations of marketed drugs may be exempt from IND submission under specific conditions.

Pre-IND Meeting:

  • Sponsor-investigators may request a pre-IND meeting with the FDA.
  • While not obligatory, pre-IND meetings can help confirm IND requirements, ensure study design suitability, minimize clinical hold risks, control costs, and facilitate early FDA interactions.
  • Meetings are typically scheduled within 60 days of request receipt.

Initial IND Submission:

  • The sponsor-investigator is responsible for preparing and submitting the IND application to the FDA.
  • IND content varies but must include a complete protocol, informed consent, and investigational product details.
  • The FDA reviews the submission within 30 days and may request additional information.
  • Clinical study may commence after 30 days, barring FDA notification of clinical hold.

IRB Requirements:

  • The sponsor-investigator ensures IRB review and approval before initiating the study.
  • Proof of IND submission or waivers may be required by the IRB.
  • The Pre-IND Audit must be completed before IRB approval.

IND Amendment Requirements:

  • Protocol, investigator, site changes, and other amendments necessitate IND updates.
  • Informational amendments report non-protocol, non-safety information.
  • IND Safety Reports: Sponsor notifies FDA and investigators of serious, related, unexpected adverse experiences promptly.
  • Follow-up reports may provide additional information.
  • Non-serious adverse events are summarized in Annual Reports.

IND Annual Reports:

  • Due within 60 days of the IND anniversary date.
  • Include study summary and adverse event summaries.
  • Required even if the study has not begun.

IND Final Study Report:

  • Submitted upon study completion for IND closure.

IND Submission Documentation:

  • Maintain complete submission records per Clinical Research SOP, "Study Records Management."
  • FDA submission documents, responses, and correspondence are retained by the CRSO IND Specialist for studies using their services.

References:

  • 21 CFR Part 312: Investigational New Drug Application
  • 21 CFR Part 312, Subpart D: Responsibilities of Sponsors and Investigators
  • 21 CFR Part 312.3: Definitions and Interpretations
  • SOP: Study Records Management

Document Approval

Popular posts from this blog

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

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

Cellular & Gene Therapy Guidances (Food and Drug Administration)

  Studying Multiple Versions of a Cellular or Gene Therapy Product in an Early-Phase Clinical Trial; Draft Guidance for Industry 11/2022 Human Gene Therapy for Neurodegenerative Diseases; Guidance for Industry 10/2022 Considerations for the Development of Chimeric Antigen Receptor (CAR) T Cell Products; Draft Guidance for Industry 3/2022 Human Gene Therapy Products Incorporating Human Genome Editing; Draft Guidance for Industry 3/2022 Interpreting Sameness of Gene Therapy Products Under the Orphan Drug Regulations; Guidance for Industry 9/2021 Manufacturing Considerations for Licensed and Investigational Cellular and Gene Therapy Products During COVID-19 Public Health Emergency; Guidance for Industry 1/2021 Chemistry, Manufacturing, and Control (CMC) Information for Human Gene Therapy Investigational New Drug Applications (INDs); Guidance for Industry 1/2020 Long Term Follow-up After Administration of Human Gene Therapy Products; Guidance for Industry 1/2020 Testing of Retroviral Vecto

Stem loop RT-PCR for Detection of siRNA in Animal Tissues

Step Loop RT-PCR for Detection of Small Interfering RNA (siRNA) The recent publications described a novel used the novel method for the detection of siRNAs using a TaqMan®-based approach. This approach utilizes similar strategy that has been used for microRNA detection. The approach is illustrated in below.  In brief, the RT step occurs in the presence of a stem-loop RT primer that is complementary to the last 6–10 bases of the 3′ end of the antisense strand of the target siRNA. The stem-loop primer contains an additional universal sequence at the 5′ end that facilitates a TaqMan-based detection strategy in the subsequent qPCR step. As in the case of microRNA, the forward primer for qPCR is sequence-specific for the target siRNA. For sequence compositions that yield a low predicted melting temperature (Tm), the forward primer is designed as a tailed primer to help increase Tm. Stem Loop PCR for SiRNA Detection Step 1: Preparation of liver and plasma samples for the quantification of si

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