Skip to main content

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), and all other applicable regulations. Safety events must be collected and reported following the protocol and the guidelines presented in this SOP, as applicable.

Definitions and Acronyms

  • Adverse Event (AE): Any unfavorable medical occurrence or worsening of an existing condition in a clinical trial participant, which may or may not have a causal relationship with the study treatment.
  • Suspected Adverse Reaction: An adverse event for which there is a reasonable possibility that the drug caused the adverse event.
  • Serious Adverse Event (SAE): An adverse event or suspected adverse reaction is considered "serious" if it results in specific outcomes such as death, life-threatening conditions, hospitalization, significant incapacity, congenital anomalies, or other significant medical events.
  • Unanticipated Adverse Device Effect (UADE): Any serious adverse effect on the health or safety of a participant, including life-threatening issues or death, caused by or associated with a device not previously identified in the investigational plan or application, or any other unanticipated serious problem related to the device's impact on the rights, safety, or welfare of subjects.
  • Safety Event Evaluation Criteria: Safety events must be evaluated based on criteria relevant to the study protocol and applicable regulatory authorities. Common evaluation criteria include:
  • Severity: The intensity of the adverse event and its impact on the participant's health, categorized on a scale such as Mild, Moderate, Severe, Life-threatening, or Death.
  • Attribution or Relatedness: The determination of whether there is a causal relationship between an adverse event and the investigational product or intervention. This may be rated on a scale like Unrelated, Unlikely Related, Possibly Related, Probably Related, or Definitely Related.
  • Expectedness: Whether the adverse event and its characteristics are anticipated based on the current understanding of the research intervention or the expected progression of the participant's underlying condition.
  • Prompt Reporting: The urgency and speed required for reporting events that meet specific criteria, which may be defined by the IRB, Human Research Protection Program, study sponsor, or applicable oversight agencies.

Procedure

Collection of Safety Events

  • The PI and study staff should familiarize themselves with the safety profile of the investigational product by reviewing the study protocol, informed consent documents, Investigator's Brochure, package insert, safety reports related to the investigational product, and safety event evaluation criteria specified by the IRB, Human Research Protection Program, study sponsor, or oversight agencies.
  • Safety events should be collected according to the protocol-defined reporting window or, if not specified, until one of the following conditions is met: completion of the safety follow-up window, withdrawal of consent by the participant, or resolution or stabilization of safety events related to the investigational product.
  • A baseline assessment of the participant's medical history and comorbidities should be conducted before any clinical research intervention, including a review of the participant's systems and medical history.
  • Throughout the study, safety information should be collected from various sources, including participant reports, investigator observations, abnormal laboratory or imaging findings, and non-study-related medical encounters.

Evaluation of Safety Events

  • After identifying an adverse event, the PI or a qualified study team member should assess its severity, attribution, expectedness, seriousness, and, if applicable, whether it meets protocol-defined stopping rules such as Dose Limiting Toxicity (DLT).
  • Severity should be assessed based on the relevant scale specified in the protocol or by regulatory authorities.
  • Attribution or relatedness should be determined based on the policy of the reviewing IRB, the institution's Human Research Protection Program, the study sponsor, or oversight agencies. If the study involves multiple interventions, attribution should be assessed independently for each.
  • Expectedness should be determined by comparing the event to the information provided in the protocol, Investigator's Brochure, informed consent forms, product labeling, and the natural progression of the participant's underlying condition.

Documentation of Safety Events

  • Safety events for each participant should be recorded cumulatively and accurately, either in an adverse event log, source documents, or Case Report Forms (CRFs).
  • The assessment of each safety event, including severity, attribution, expectedness, seriousness, and whether it meets DLT criteria, should be documented, signed, and dated by the investigator or a designated team member.

Reporting Safety Events

  • Safety information must be reported to the relevant parties, including the Sponsor, Contract Research Organization (CRO) if applicable, IRB, Data Safety Monitoring Committee/Board, FDA, or other sponsoring agencies, as specified in the protocol.
  • Reporting methods and timelines may vary by study and should be outlined in the protocol or separate guidance documents. Common reporting methods include paper-based forms, Electronic Data Capture (EDC) systems, or other designated formats.
  • The reporting frequency and extent depend on the characteristics of the event, such as expectedness, relatedness, and seriousness. Expedited reporting is typically required for certain events, with initial notification within 24 business hours and follow-up reporting as necessary.

Institutional Review Board (IRB) Reporting

  • The IRB of record should be consulted for guidance on reporting safety information, including unanticipated problems or events.
  • The PI or a qualified team member should review and confirm safety events that meet IRB reporting requirements.
  • The IRB requires reporting of adverse events that are unexpected, definitely or probably related to the research, and place participants or others at greater risk of harm than previously known.
  • Reporting requirements and timeframes can be found on the IRB website.

Food and Drug Administration (FDA) Reporting

  • For studies falling under FDA regulations, adverse events meeting specific criteria must be promptly reported to the FDA. These criteria include events that are serious, unexpected, and related (definitely, probably, or possibly).
  • Reporting timeframes for FDA-regulated studies are provided in this SOP and may vary based on the characteristics of the event.
  • Responsibilities for reporting to the FDA depend on whether the study is industry-sponsored or investigator-initiated. The SOP provides guidance for both scenarios.

Reporting External Safety Events

  • External safety events, such as those from sponsors, coordinating centers, or IP manufacturers, should be reviewed and assessed for their impact on the study's conduct, informed consent document, or investigator brochure.
  • The sponsor is responsible for notifying investigators of important safety information and unanticipated problems that affect the research.
  • Investigators are not required to actively monitor external sources for safety information but should promptly report relevant safety events to the IRB.

Materials Required

  • Study protocol
  • IRB of record safety event reporting requirements
  • Template adverse event log (available at provided link)

References

  • 21 CFR Part 312.32 IND Safety Reporting
  • 21 CFR Part 812 Investigational Device Exemptions
  • FDA Guidance for Industry: Safety Reporting Requirements for INDs and BA/BE Studies
  • ICH Guidance for Industry E2: Clinical Safety Data Management
  • NCI Common Terminology Criteria for Adverse Events

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