Skip to main content

Standard Operating Procedure (SOP) on Study Records Collection and Maintenance for Clinical Trials

 Title: Study Records Collection and Maintenance

Introduction and Purpose:

The purpose of this SOP is to define the procedures for collecting and maintaining study records in the conduct of clinical research at the clinical site and relevant covered entities. In accordance with FDA regulations and Good Clinical Practice (GCP) guidelines, it is essential to maintain original records of clinical findings, observations, and activities for the reconstruction and evaluation of clinical research studies.

Subject-specific records, contained within study participant charts, serve as source documentation of research records. These records encompass various documents such as signed informed consent forms, medical histories, medication logs, treatment records, protocol assessment results, and records of adverse events. Additionally, general study records, including versions of the study protocol and informed consent form, Institutional Review Board (IRB) approvals, investigator's brochures, training records, delegation of authority logs, and monitoring reports, are preserved in the regulatory binder. While not all documents apply uniformly to every study, the fundamental principles outlined in this procedure remain consistent across all clinical research types.

Definitions and Acronyms:

  • Clinical Research: Encompasses all research involving human participants. It excludes secondary studies using existing biological specimens or publicly available data without identifiers.
  • Clinical Trial: Refers to clinical research studies involving human participants assigned to an intervention designed to assess its impact on health-related biomedical or behavioral outcomes.
  • Source Data: All information in original records of clinical findings, observations, or other activities in a clinical trial necessary for the reconstruction and evaluation of the study.
  • Source Documents: Original documents, data, and records (e.g., hospital records, clinical charts, laboratory reports, subjects’ diaries, pharmacy dispensing records, radiologic scans and imaging records, etc.) on which source data are recorded.
  • CRF: Case Report Form
  • CRO: Contract Research Organization
  • EHR: Electronic Health Record
  • FDA: United States Food and Drug Administration
  • GCP: Good Clinical Practice
  • ICH: International Council on Harmonization
  • IRB: Institutional Review Board
  • PHI: Protected Health Information
  • PI: Principal Investigator
  • SAE: Serious Adverse Event
  • SOP: Standard Operating Procedure

Procedure:

Standards and Responsibilities:

  • The documentation of source data is crucial for reconstructing, evaluating, and validating clinical research findings, observations, and activities.
  • The Principal Investigator (PI) bears ultimate responsibility for collecting and maintaining research records to ensure their accuracy, completeness, and accessibility throughout the study. However, the PI may delegate specific study record tasks to sub-investigators or coordinating staff, as outlined in the delegation of authority log or other delegation processes.

Clinical research data on source documentation should adhere to the following principles:

  • Attributable: The data's origin or source must be identified and recorded, including an audit trail for electronic and automated records, along with signatures/initials and dates for record entries. Additionally, source documents should include study, subject, and visit identifiers as needed.
  • Legible: Data and records should be clearly readable and interpretable.
  • Contemporaneous: Data must be recorded at the time of the event or observation.
  • Original: Data should represent the initial entry or recording.
  • Accurate: Data should reflect the true observation or results, with errors corrected appropriately.
  • Complete: Study records should encompass all information and data related to the study and subject's health (e.g., adverse events) throughout their participation.
  • The study protocol specifies the required screening (including medical history) and baseline assessments, study procedures, interventions, and follow-up assessments. Source documentation should be created and compiled (as originals or copies) in the study participant chart to demonstrate compliance with each eligibility criterion, protocol requirement, and to provide evidence for validating each case report form entry.
  • Source data may be directly entered into the Case Report Form (CRF) when stipulated in the study protocol or other study documents.
  • Electronic Health Record (EHR) systems, used by the clinical site and relevant covered entities, incorporate electronic signatures and audit trail functions. However, these systems are not validated or documented as compliant with 21 CFR Part 11, as the FDA does not intend to assess the compliance of electronic health records with Part 11.

Source Entry and Corrections:

  • Observations recorded as source data must be signed/initialized and dated by the PI or the study team member making the source entry.
  • Electronic records (e.g., EHR) are signed electronically. Printed copies of EHR source documents included in the study participant chart should feature the electronic signature and date stamp on the printed copy.
  • The practice of transcribing source data from the EHR to a "study visit form" or "source worksheet" before CRF entry should be minimized to reduce transcription errors.
  • When creating source data, blue or black ink should be used; pencil should not be used, original entries should not be overwritten, and correction fluid or markers should not be used to obliterate the original entry. Backdating entries is also discouraged.
  • All source documents should include clear identifiers associating the document with the trial (e.g., study title, study number), study participant (e.g., subject number, initials), and study visit (e.g., date, visit number, cycle).
  • In case an error is identified on a source document, it should be corrected correctly. Handwritten source data corrections involve drawing a single line through the incorrect information, adding the correction beside the original entry, and completing the entry with initials, date, and, if necessary, a reason for the change. The original entry must not be obscured.
  • Electronic source documents created and maintained using automated systems, including the EHR, should have an audit trail documenting any corrections made to the source.

Study Participant Charts:

  • Study participant charts should be consistently organized for all subjects.
  • Charts should be structured with documents arranged in reverse chronological order, with the latest documents on top. A uniform template for chart organization should be followed for all study participants enrolled in a specific study. A separate chart may be maintained to compile detailed records for screen failures (subjects who do not enroll).
  • For a comprehensive list of documents to include in study participant charts, refer to UUSOP-06 Supplement A.
  • Study participant charts must be regularly updated after each study visit or on a defined schedule (e.g., monthly) until the subject's study participation concludes.
  • In certain studies, both paper source documents and electronic source data captured directly in the electronic data capture system or stored electronically (e.g., EHR) may coexist. In such cases, providing an outline of where to find each source document helps avoid confusion during inspections. This outline should specify which data is directly entered into the CRF and therefore does not have a corresponding source document.

Regulatory Binder (Trial Master File):

  • Essential documents and general study records should be collected and maintained in the Regulatory Binder or Trial Master File, in either paper or electronic format.
  • Consistent organization templates for regulatory binders should be adhered to.
  • Certain essential documents, such as the Delegation of Authority log and Screening log, may receive updates with new entries throughout the study's conduct. In cases where new documents contain updated information (e.g., protocol amendment, renewed medical license, changes in financial disclosure), both the original and updated documents should be retained in the regulatory binder.
  • If essential documents are maintained electronically or in a central location, a note-to-file should be placed in the regulatory binder to indicate the location of these records.
  • Maintenance and Storage of Study Records:

  • Study participant charts and regulatory binders should be kept current and up-to-date throughout the clinical research.
  • All research records containing Protected Health Information (PHI) must be stored in an area with restricted access during working hours and in a locked room or cabinet when not in use or outside working hours.
  • Records created or maintained in the EHR are considered source documents. Copies of these records from the EHR included in the study participant charts are not regarded as source documents and do not require certification as such.

Monitoring and Auditing of Study Records:

  • All subject and regulatory records, including electronic records, should be available for review by sponsor representatives, the compliance office, IRB auditors, departmental monitors/auditors, regulatory inspectors, and other authorized individuals.
  • When utilizing an electronic data capture system for direct source entry or storing regulatory records electronically, consideration should be given to whether the system or program allows for external review by the listed entities.
  • At the clinical site, sponsor representatives and regulatory inspectors are permitted to review source documents in the EHR only under supervised log-in and in the presence of a study coordinator or authorized study team member. External visitors will not receive individual log-in credentials, and they will not have unrestricted access to the EHR.
  • Direct review of source documents in the EHR will be limited to study participants, and the duration will be reasonable (e.g., 1-2 hours per day).
  • Entire medical records should not be copied or transferred from the clinical site. A limited number of copies from the EHR may be made for external reviewers; however, personal information that could identify study subjects will be redacted from provided copies.

Archiving Study Records:

  • Following the completion of a clinical study, the study records may be examined and subject to review by internal departmental monitors/auditors, sponsors, or FDA inspectors.
  • After maintaining the study records on-site for a defined period, clinical research records may be moved to long-term archiving, such as clinical site Archives and Records Management, or another secure archive system.

Materials Required:

  • Study source documents and general study records.
  • Access to the EHR system.

References:

  • 21 CFR Part 312.62: Investigator Recordkeeping and Record Retention.
  • 21 CFR Part 812.140: Records and Reports.
  • 21 CFR Part 11: Electronic Records, Electronic Signatures.
  • ICH Guidance for Industry E6(R2): Good Clinical Practice.
  • FDA Guidance for Industry – Electronic Source Data in Clinical Investigations.

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),