Which Clinical Study Report section would be most useful for a Data Manager to review?
Clinical narratives of adverse events
Enumeration and explanation of data errors
Description of statistical analysis methods
Rationale for the study design
The section of theClinical Study Report (CSR)that is most useful for a Data Manager is the one that includes theenumeration and explanation of data errors. This section provides a summary of thedata quality control findings, including error rates, missing data summaries, and any issues identified during data review, validation, or database lock.
According to theGCDMP (Chapter: Data Quality Assurance and Control), post-study reviews of data errors and quality findings are essential for evaluating process performance, identifying recurring issues, and informing continuous improvement in future studies.
Other sections, such as clinical narratives (A) or statistical methods (C), are outside the core scope of data management responsibilities. Thedata error enumeration sectiondirectly reflects the quality and integrity of the data management process and is therefore the most relevant for review.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Data Quality Assurance and Control, Section 6.4 – Quality Reporting and Error Analysis
ICH E3 – Structure and Content of Clinical Study Reports, Section 14.3 – Data Quality Evaluation
The result set from the query below would be which of the following?
SELECT Pt_ID, MRN, SSN FROM patient
Wider than the patient table
Shorter than the patient table
Longer than the patient table
Narrower than the patient table
In aSQL (Structured Query Language)database, theSELECTstatement specifies which columns to display from a table. In this query, only three columns —Pt_ID,MRN, andSSN— are being selected from thepatienttable.
This means the resulting dataset will contain:
The same number ofrows (records)as the original table (assuming noWHEREfilter), and
Fewer columnsthan the full table.
In database terminology:
“Wider” refers to more columns (fields).
“Narrower” refers to fewer columns (fields).
Since this query retrieves only 3 columns (out of potentially many in the original table), the result set isnarrower than the patient table, makingoption Dcorrect.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Database Design and Build, Section 5.1 – Relational Databases and Query Logic
ICH E6(R2) GCP, Section 5.5.3 – Data Retrieval and Integrity Principles
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations, Section 6.4 – Database Query Controls
Which of the following ensures that the trials are conducted and the data are generated, documented (recorded), and reported in compliance with the protocol, GCP, and the applicable regulatory requirement(s)?
Standard Operating Procedures (SOP)
Statistical Analysis Plan (SAP)
Data Management Plan (DMP)
CRFs
Standard Operating Procedures (SOPs)are formal, controlled documents that definestandardized processesto ensure clinical trials are conducted in compliance withGood Clinical Practice (GCP), the study protocol, and regulatory requirements (such as ICH and FDA).
According toGood Clinical Data Management Practices (GCDMP)andICH E6(R2) GCP, SOPs are fundamental to quality management systems. They describehowtasks are performed, ensuring consistency, accountability, and traceability across all studies and team members. Proper adherence to SOPs guarantees that data areaccurately generated, documented, and reportedin compliance with ethical and regulatory standards.
Other options serve different purposes:
SAP (B)defines statistical methodology, not compliance control.
DMP (C)focuses on study-specific data handling, not organizational compliance.
CRFs (D)are tools for data collection but do not enforce compliance by themselves.
Therefore,option A (SOP)is correct.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Quality Management and Compliance, Section 5.1 – Role of SOPs in Regulatory Compliance
ICH E6(R2) GCP, Section 2.13 and 5.1.1 – Quality Management Systems and SOP Requirements
FDA 21 CFR Part 312.50 – Sponsor Responsibilities and Compliance Systems
For ease of data processing, the study team would like the database codes for a copyrighted rating scale preprinted on the CRF. What is the most critical task that the CRF designer must do to ensure the data collected on the CRF for the scale are reliable and will support the results of the final analysis?
Consult the independent source and determine database codes will not influence subject responses.
Consult the study statistician regarding the change and determine that database codes will not influence the analysis.
Consult the independent source of the rating scale for approval and document that continued validity of the tool is not compromised.
Complete the requested changes to the instrument and ensure the correct database codes are associated with the appropriate responses.
When using acopyrighted or validated rating scale(e.g., Hamilton Depression Scale, Visual Analog Pain Scale), anymodification to the original instrument, including preprinting database codes on the CRF, must beapproved by the instrument’s owner or licensing authorityto ensure thevalidity and reliabilityof the instrument are not compromised.
According to theGCDMP (Chapter: CRF Design and Data Collection), validated rating scales are psychometrically tested tools. Any visual or structural modification (such as adding codes, changing layout, or rewording questions) can invalidate prior validation results. Therefore, the CRF designer mustconsult the independent source (copyright holder)for approval anddocument that the validity of the tool remains intact.
Merely consulting statisticians (option B) or verifying database alignment (option D) does not ensure compliance. Thus,Option Censures scientific and regulatory integrity.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: CRF Design and Data Collection, Section 6.1 – Use of Validated Instruments and Rating Scales
ICH E6 (R2) GCP, Section 5.5.3 – Validation of Instruments and Data Capture Tools
FDA Guidance for Industry: Patient-Reported Outcome Measures – Use in Medical Product Development to Support Labeling Claims, Section 4 – Instrument Modification and Validation
Which metric reveals the timeliness of the site-work dimension of site performance?
Time from Last Patient Last Visit to database lock
Time from final protocol to first patient enrolled
Time from site contract execution to first patient enrolled
Median and range of time from query generation to resolution
Thesite-work dimension of site performanceevaluates how efficiently sites manage and resolve data-related tasks — particularly query resolution, data entry, and correction timelines. Among the given metrics, themedian and range of time from query generation to resolution (D)directly measures the site’s responsiveness and data management efficiency.
According to theGCDMP (Chapter on Metrics and Performance Measurement), this indicator helps identify sites that delay query resolution, which can impact overall study timelines and data quality. Tracking this metric allows the data management team to proactively provide additional training or communication to underperforming sites.
Other options measure different aspects of project progress:
Areflects overall database closure speed.
BandCrelate to study startup and enrollment readiness, not ongoing data work.
Thus,option Daccurately represents asite performance timeliness metric, aligning with CCDM principles for operational performance measurement.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Metrics and Performance Management, Section 5.4 – Site Query Resolution Metrics
ICH E6(R2) Good Clinical Practice, Section 5.18 – Monitoring and Site Performance Oversight
A study has an expected enrollment period of one year but has subject recruitment issues. Twelve new sites are added toward the end of the expected enrollment period to help boost enrollment. What is the most likely impact on data flow?
The database set-up will need to be changed to allow for additional sites as they are added to the study.
The distribution of subjects selected for quality control will need to be stratified to allow for the twelve new sites.
A bolus of CRFs at the end of the study will result in the need to increase data entry and cleaning rates to meet existing timelines.
Additional sites will likely have increased query rates since site training is occurring closer to study close.
Adding multiple new sites late in the enrollment period creates aconcentrated influx of new datanear the end of the study. These sites typically start enrolling patients later, resulting in a“bolus” of Case Report Forms (CRFs)that must be entered, validated, and cleaned within a shorter timeframe to meet database lock deadlines.
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Project Management and Data Flow), late site activation compresses the timeline for data management tasks, necessitating increased resources fordata entry, query management, and cleaning. Data management teams must anticipate this surge and plan accordingly—either by increasing staffing or revising timelines to prevent bottlenecks and maintain quality.
Whileoption D(increased query rates) can occur, it is a secondary effect. Themost direct and consistent impactis the surge in data volume requiring expedited processing near study end.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Project Management, Section 5.3 – Managing Changes in Site Activation and Data Flow
ICH E6(R2) GCP, Section 5.1 – Quality Management and Oversight
Which information is required by most systems to specify data entry screens?
User role, access level, and permissions
Data type, prompt, and response format
Page number and total number of pages
Help text, review parameters, and answers
When designing or configuringdata entry screenswithin an Electronic Data Capture (EDC) system, three critical components are required for each field:
Data Type– Defines the nature of the data (e.g., text, numeric, date).
Prompt– The label or question displayed to the user.
Response Format– Specifies how the user enters or selects data (e.g., free text, drop-down, checkbox).
According to theGCDMP (Chapter: EDC Systems and Database Design), these three attributes form thelogical data structurerequired to build and validate data entry interfaces. They ensure consistency in how information is captured, displayed, and validated during data entry.
Whileuser roles (A)andhelp text (D)are system-level configurations, not field-level specifications,page numbers (C)relate to printed CRFs rather than digital data screens.
Therefore,option B (Data type, prompt, and response format)correctly identifies the essential information needed to define data entry screens.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: EDC Systems and Database Design, Section 4.3 – Screen Design Specifications
CDISC CDASH Implementation Guide, Section 3.2 – Data Field Attributes
ICH E6(R2) GCP, Section 5.5.3 – Data Capture and Input Standards
According to theFDA Guidance for Industry, Providing Regulatory Submissions in Electronic Format (April 2006)andGood Clinical Data Management Practices (GCDMP, May 2007), which of the following is the most acceptable for aderived field?
Providing CRF annotation "not entered in the database" next to the average score
Providing the algorithm for calculating the average score on the CRF
Providing the algorithm for calculating the average score in the dataset definition file
Providing CRF annotation AVE next to the average score
In clinical data management, aderived fieldrefers to any variable that is not directly collected from the Case Report Form (CRF) but is instead calculated or inferred from one or more collected variables (for example, calculating an average blood pressure from multiple readings). Proper documentation of derived fields is essential for ensuringdata traceability, transparency, and compliancewith both FDA and SCDM guidelines.
According to theGood Clinical Data Management Practices (GCDMP, May 2007), all derivations and transformations applied to clinical data must beclearly defined and documentedin metadata such as thedataset definition file (also referred to as data specifications, variable definition tables, or Define.xml files). The derivation algorithm should be explicitly stated in this documentation to allow independent verification, regulatory review, and reproducibility of results.
TheFDA Guidance for Industry (April 2006)on electronic submissions further emphasizes thatderived fields must be supported by comprehensive metadatathat defines the computational method used. This documentation enables the FDA or any regulatory body to audit andreproduce analytical results without ambiguity. Annotating or describing derivations directly on the CRF (as in options A, B, or D) isnot sufficient, as CRFs represent data collection instruments—not analytical documentation.
Therefore, the correct and regulatory-compliant practice isto provide the derivation algorithm for a calculated field within the dataset definition file, aligning with bothFDAandGCDMPexpectations for data integrity and auditability.
Reference (CCDM-Verified Sources):
Society for Clinical Data Management (SCDM), Good Clinical Data Management Practices (GCDMP), Chapter: Data Handling and Processing – Derived and Calculated Data Fields, Section 5.3.3
FDA Guidance for Industry: Providing Regulatory Submissions in Electronic Format, April 2006, Section 3.2 on Dataset Documentation Requirements
CDISC Define.xml Implementation Guide – Metadata and Algorithm Documentation for Derived Variables
In a physical therapy study, range of motion is assessed by a physical therapist at each site using a study-provided goniometer. Which is the most appropriate quality control method for the range of motion measurement?
Comparison to the measurement from the previous visit
Programmed edit checks to detect out-of-range values upon data entry
Reviewing data listings for illogical changes in range of motion between visits
Independent assessment by a second physical therapist during the visit
In this scenario, the variable of interest—range of motion (ROM)—is aclinically measured, observer-dependent variable. The accuracy and reliability of such data depend primarily on theprecision and consistency of the measurement technique, not merely on data entry validation. Therefore, the most appropriatequality control (QC) methodisindependent verification of the measurement by a second qualified assessor during the visit(Option D).
According to theGood Clinical Data Management Practices (GCDMP, Chapter on Data Quality Assurance and Control), quality control procedures must be tailored to the nature of the data. Forclinically assessed variables, especially those involving human judgment (e.g., physical measurements, imaging assessments, or subjective scoring),real-time verification by an independent qualified assessorensures that data are valid and reproducible at the point of collection. This approach directly addressesmeasurement bias,observer variability, andinstrument misuse, which are primary sources of data error in clinical outcome assessments.
Other options, while valuable, address onlydata consistency or plausibilityafter collection:
Option A (comparison to previous visit)andOption C (reviewing data listings)are retrospective data reviews, suitable for identifying trends but not preventing measurement error.
Option B (programmed edit checks)detects only extreme or impossible values, not measurement inaccuracies due to technique or observer inconsistency.
The GCDMP andICH E6 (R2) Good Clinical Practiceguidelines emphasize that data quality assurance should beginat the source, through standardized procedures, instrument calibration, and dual assessments for observer-dependent measures. Having anindependent second assessorensures inter-rater reliability and provides direct confirmation that the recorded value reflects an accurate and valid measurement.
Reference (CCDM-Verified Sources):
Society for Clinical Data Management (SCDM), Good Clinical Data Management Practices (GCDMP), Chapter: Data Quality Assurance and Control, Section 7.4 – Measurement Quality and Verification
ICH E6 (R2) Good Clinical Practice, Section 2.13 – Quality Systems and Data Integrity
FDA Guidance for Industry: Patient-Reported Outcome Measures and Clinical Outcome Assessment Data, Section 5.3 – Quality Control of Clinician-Assessed Data
SCDM GCDMP Chapter: Source Data Verification and Quality Oversight Procedures
QA is conducting an audit on a study for ophthalmology which is ready for lock. Inconsistencies are found between the database and the source. Of the identified fields containing potential data errors, which fields are considered critical for this particular study?
Subject Identifier
Concomitant Medications
Weight
Medical History
In anophthalmology clinical study, data criticality is determined by how directly a data element affectssafety evaluation,efficacy assessment, andregulatory decision-making. According to theGood Clinical Data Management Practices (GCDMP, Chapter on Data Validation and Cleaning), critical data fields are those that:
Have a direct impact on theprimary and secondary endpoints, or
Are essential forsafety interpretation and adverse event causality assessment.
Among the listed options,Concomitant Medications (Option B)are consideredcritical datafor ophthalmology studies. This is because many ocular treatments and investigational products can interact with systemic or topical medications, potentially affectingocular response,intraocular pressure,corneal healing, orvisual function outcomes. Any inconsistency in concomitant medication data could directly influencesafety conclusionsorefficacy interpretations.
Other options, while important, are less critical for this study type:
Subject Identifier (A)is essential for data traceability and audit purposes but is not directly related to safety or efficacy outcomes.
Weight (C)may be relevant in dose-dependent drug trials but is rarely a pivotal variable in ophthalmology, where local administration (eye drops, intraocular injections) is common.
Medical History (D)provides contextual background but does not have the same immediate impact on endpoint analysis as current concomitant treatments that can confound the therapeutic effect or cause ocular adverse events.
PerGCDMPandICH E6 (R2) GCPguidelines, data validation plans must definecritical data fieldsduring study setup, reflecting therapeutic area–specific priorities. For ophthalmology,concomitant medications, ocular assessments (visual acuity, intraocular pressure, retinal thickness, etc.), and adverse eventsare typically designated as critical fields requiring heightened validation, source verification, and reconciliation accuracy before database lock.
Thus, when QA identifies discrepancies between the CRF and source, theConcomitant Medications field (Option B)is the most critical to address immediately to ensure clinical and regulatory data integrity.
Reference (CCDM-Verified Sources):
Society for Clinical Data Management (SCDM), Good Clinical Data Management Practices (GCDMP), Chapter: Data Validation and Cleaning, Section 6.4 – Critical Data Fields and Data Validation Prioritization
ICH E6 (R2) Good Clinical Practice, Section 5.18 – Monitoring and Source Data Verification
FDA Guidance for Industry: Oversight of Clinical Investigations — A Risk-Based Approach to Monitoring, Section 5.3 – Identification of Critical Data and Processes
SCDM GCDMP Chapter: Data Quality Assurance and Control – Therapeutic Area–Specific Data Criticality Examples (Ophthalmology Studies)
A Clinical Data Manager is drafting data element definitions for a new study. One of the definitions provided is:
"Baby's crown to heel length measured lying on back, measured physical quantity, precision of 0.1."
Which of the following is missing from the definition?
Discrete values for a drop-down list
Enumeration
Data type of the data element
Unit or dimensionality of measure
A completedata element definitionin clinical data management should include:
Nameandclear descriptionof the data element,
Data type(e.g., numeric, text, date),
Precisionor scale (if numeric), and
Unit or dimensionalityof measure (e.g., centimeters, inches).
In this example, while the data type (“measured physical quantity”) and precision (0.1) are defined, theunit of measurement(e.g., centimeters or inches) is missing. This omission leads to ambiguity and could cause serious discrepancies when comparing or analyzing measurements.
TheGCDMP (Chapter: Database Design and Build)emphasizes that units and dimensionality must be explicitly defined and consistently applied in all CRFs, metadata dictionaries, and data transformations.
Thus,option D (Unit or dimensionality of measure)is correct.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Database Design and Build, Section 5.2 – Metadata and Data Element Definitions
CDISC CDASH Implementation Guide, Section 3.3 – Data Element Metadata Requirements
ICH E6(R2) GCP, Section 5.5.3 – Data Accuracy and Standardized Definitions
In an EDC study, an example of an edit check that would be inefficient to run at data entry is a check:
Against a valid list of values.
Across visits for consistency.
Against a valid numeric range.
On the format of a date.
InElectronic Data Capture (EDC)systems, edit checks are categorized based on when and how they are executed — typicallyimmediate (at data entry)orbatch (post-entry). Checks that require data frommultiple visits or formsare generallyinefficient to run at data entrybecause they depend on information that may not yet exist in the system.
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Data Validation and Cleaning),cross-visit consistency checks— such as comparing baseline and follow-up blood pressure or verifying date order between screening and dosing — should be executed asbatch or scheduled validations, not at the point of data entry. Running these complex checks in real time can slow system performance, increase query load unnecessarily, and confuse site users if related data are not yet entered.
Conversely, edit checks against valid ranges, formats, or predefined value lists (options A, C, and D) are simple, local validations ideally performed immediately at data entry to prevent basic errors.
Therefore,cross-visit consistency checks(Option B) are best executed later, making theminefficient for real-time data entry validation.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Validation and Cleaning, Section 6.4 – Real-Time vs. Batch Edit Checks
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations – Section on Edit Checks and Data Validation Logic
CDISC SDTM Implementation Guide – Section on Temporal Data Consistency Validation
If a data manager generated no additional manual queries on data in an EDC system and the data were deemed clean, why could the data appear to be not clean during the next review?
The study coordinator can change the data due to re-review of the source.
The CRA can change the data during a quality review of source to database.
The medical monitor can override safety information entered in the system.
The data manager may have accidentally changed the data.
In anElectronic Data Capture (EDC)system, even after a data manager completes all manual queries and marks data as "clean," the data may later appearuncleanifthe site (study coordinator)makes subsequent updates in the system after re-reviewing thesource documents.
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Electronic Data Capture Systems), site users maintain the authority to modify data entries as long as the system remains open for data entry. TheEDC system audit trailcaptures such changes, which can automatically invalidate prior data reviews, triggering new discrepancies or changing system edit-check statuses.
This situation commonly occurs when the site identifies corrections in the source (e.g., wrong date or lab result) and updates the EDC form accordingly. These post-cleaning changes require additional review cycles to ensure the database reflects accurate and verified information before final lock.
Options B, C, and D are incorrect — CRAs and medical monitors cannot directly change EDC data; they can only raise queries or request updates.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Electronic Data Capture Systems, Section 6.3 – Post-Cleaning Data Changes and Audit Trails
ICH E6 (R2) GCP, Section 5.5.3 – Data Integrity and Change Control
FDA 21 CFR Part 11 – Electronic Records: Change Documentation Requirements
A study is collecting ePRO assessments as well as activity-monitoring data from a wearable device. Which data should be collected from the ePRO and activity-monitoring devices to synchronize the device data with the visit data entered by the site?
Study subject identifier
Study subject identifier and date/time
Geo-spatial location
Geo-spatial location and study subject identifier
To synchronize data fromelectronic patient-reported outcomes (ePRO)andwearable activity-monitoring deviceswith site-entered visit data, both thestudy subject identifieranddate/timeare essential.
According to theGCDMP (Chapter: Data Management Planning and Study Start-up), each dataset must containkey identifiersthat allow for accuratedata integration and temporal alignment. In studies involving multiple digital data sources, time-stamped subject identifiers are necessary to ensure that the device-generated data correspond to the correct subject and study visit.
Thesubject identifierensuresdata traceability and linkageto the appropriate participant, whiledate/timeallows synchronization of device data (e.g., activity or physiological measurements) with the corresponding site-reported visit or event. Geo-spatial data (options C and D) are typically not relevant to study endpoints and pose unnecessary privacy risks underHIPAAandGDPRguidelines.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Integration and eSource Data, Section 5.2 – Data Alignment and Synchronization Principles
FDA Guidance for Industry: Use of Electronic Health Record Data in Clinical Investigations, Section 4.2 – Data Linking and Synchronization
ICH E6 (R2) GCP, Section 5.5.3 – Data Traceability and Integrity
Which Clinical Study Report section would be most useful for a Data Manager to review?
Description of statistical analysis methods
Rationale for the study design
Description of how data were processed
Clinical narratives of adverse events
The section of theClinical Study Report (CSR)most useful for aData Manageris thedescription of how data were processed.
According to theGCDMP (Chapter: Data Quality Assurance and Control), this section details thedata handling methodology— includingdata cleaning, coding, transformation, and derivation procedures— all of which are core responsibilities of data management. Reviewing this section ensures that the data processing methods documented in the CSR align with theData Management Plan (DMP),Data Validation Plan (DVP), anddatabase specifications.
Thestatistical methods section (option A)is primarily for biostatistics, and therationale for study design (option B)pertains to clinical and regulatory affairs.Clinical narratives (option D)are used by medical reviewers, not data managers.
By reviewing how data were processed, the Data Manager verifies that the study data lifecycle—from collection to analysis—was conducted in compliance with regulatory and GCDMP standards.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Quality Assurance and Control, Section 6.3 – Documentation of Data Processing in Clinical Study Reports
ICH E3 – Structure and Content of Clinical Study Reports, Section 11.3 – Data Handling and Processing
FDA Guidance for Industry: Clinical Study Reports and Data Submission – Data Traceability and Handling Documentation
A study team member suggests that data for a small, 50-patient, 2-year study can be entered and cleaned in two weeks before lock. Which are important other considerations?
Processing the data in two weeks after the study is over would save money because the data manager would not be involved until the end
Without the ability to capture the data electronically, the data cannot be checked or used to monitor and manage the study
Processing the data in two weeks after the study is over would save money because the EDC system would only be needed for a month
It would take more than two weeks to get second iteration queries generated and resolved
The most critical consideration is thatdata cleaning is an iterative process, and completing all necessary steps — includingquery generation, site resolution, and second-pass validation— cannot realistically be accomplished within two weeks after study close.
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Data Validation and Cleaning), data cleaning must occurcontinuously throughout the study, not only at the end. Post-database lock activities typically include running final validation checks, resolving outstanding queries, performing reconciliation (e.g., SAEs, labs, coding), and conducting final quality review.
Even in small studies,query turnaround and response cyclesfrom sites take time — typically2–4 weeks per iteration— making a two-week total cleaning period unrealistic.
Therefore,Option Dis correct: it would take more than two weeks to handle second-round (follow-up) queries and confirm final resolutions prior to database lock.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Validation and Cleaning, Section 5.4 – Ongoing vs. End-of-Study Data Cleaning
ICH E6 (R2) Good Clinical Practice, Section 5.5.3 – Data Quality and Timeliness
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations – Data Management and Cleaning
An organization conducts over fifty studies per year. Currently each study is specified and set-up from scratch. Which of the following organizational infrastructure options would streamline database set-up and study-to-study consistency?
Adopting an ODM compliant database system
Maintaining a library of form or screen modules
Improving the form or screen design process
Implementing controlled terminology for adverse events
To improve efficiency and ensure consistency across multiple studies, the most effective infrastructure solution is tomaintain a centralized library of standardized forms or screen modules(e.g., CRF/eCRF templates).
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Database Design and Build), using aform libraryallows reuse of validated data collection modules for commonly collected domains such as demographics, adverse events, and vital signs. This reduces database setup time, enhances uniformity in data definitions, and ensures alignment with standards such asCDISC CDASH and SDTM.
While adoptingODM (A)provides standardized data exchange and interoperability, it does not inherently reduce setup workload.Improving design processes (C)enhances efficiency but doesn’t guarantee consistency, andimplementing controlled terminology (D)helps with coding standardization, not database structure.
Therefore,option B—maintaining a library of form or screen modules— provides the most direct and sustainable improvement for scalability and quality.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Database Design and Build, Section 5.3 – Use of Standard Libraries and Templates
CDISC CDASH Implementation Guide, Section 3.2 – Reusable CRF Modules and Standardization
ICH E6(R2) GCP, Section 5.5.3 – Standardization and Reuse in Data Collection Systems
Which database table structure is most appropriate for vital signs data collected at every-other visit for each patient in a study?
One record per visit
One record per patient per study
One record per patient per visit
One record per patient
In a relational clinical database, themost efficient and normalized structurefor data collected repeatedly over time—such asvital signs—isone record per patient per visit.
Each patient will have multiple records, one for each visit when vital signs are assessed. This structure supports:
Time-based analysis (e.g., trends across visits),
Accurate data linkage with visit-level metadata, and
Efficient querying for longitudinal data.
According to theGCDMP (Chapter: Database Design and Build), the relational design principle dictates that data should be stored at thelowest unique level of observation. Since vital signs vary by both patient and visit, the combination ofpatient ID + visit IDforms a unique key for each record.
Option A (per visit) lacks patient identification, while options B and D aggregate data too broadly, losing temporal detail.
Thus,option C (One record per patient per visit)correctly represents the normalized design structure.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Database Design and Build, Section 4.2 – Normalization and Table Structure
CDISC SDTM Implementation Guide, Section 5.3 – Visit-Level and Observation-Level Data Structures
ICH E6(R2) GCP, Section 5.5.3 – Data Handling Principles
An international study collects lab values. Sites use different units in the source documents. Which of the following data collection strategies will have fewer transcription errors?
Allow values to be entered as they are in the source document and derive the units based on the magnitude of the value
Allow values to be entered as they are in the source and the selection of units on the data collection form
Use a structured field and print standard units on the data collection form
Have all sites convert the values to the same unit system on the data collection form
In international or multicenter clinical studies,laboratory dataoften originate from different laboratories that use varying measurement units (e.g., mg/dL vs. mmol/L). TheGood Clinical Data Management Practices (GCDMP, Chapter on CRF Design and Data Collection)provides clear guidance on managing this variability to ensuredata consistency,traceability, andminimized transcription errors.
The approach that results infewer transcription errorsis toallow sites to enter lab values exactly as recorded in the source document (original lab report)and to requireexplicit selection of the corresponding unitfrom a predefined list on the data collection form or within the electronic data capture (EDC) system. This method (Option B) preserves the original source data integrity while enabling centralized or automated unit conversion later during data cleaning or statistical processing.
Option B also supports compliance withICH E6 (R2) Good Clinical Practice (GCP), which mandates that transcribed data must remain consistent with the source documents. Attempting to derive units automatically (Option A) can lead to logical errors, while forcing sites to manually convert units (Option D) introduces unnecessary complexity and increases the risk of miscalculation or inconsistent conversions. Printing only standard units on the CRF (Option C) ignores local lab practices and can lead to discrepancies between CRF entries and source records, triggering numerous data queries.
TheGCDMPemphasizes that CRF design must account for local variations in measurement systems and ensure thatunit selection is structured (dropdowns, controlled lists)rather than free-text to prevent typographical errors and facilitate standardization during data transformation.
Therefore, OptionB—“Allow values to be entered as they are in the source and the selection of units on the data collection form”—is the most compliant, accurate, and efficient strategy for minimizing transcription errors in international lab data collection.
Reference (CCDM-Verified Sources):
Society for Clinical Data Management (SCDM), Good Clinical Data Management Practices (GCDMP), Chapter: CRF Design and Data Collection, Section 5.4 – Laboratory Data Management and Unit Handling
ICH E6 (R2) Good Clinical Practice, Section 5.18 – Data Handling and Record Retention
CDISC SDTM Implementation Guide, Section 6.3 – Handling of Laboratory Data and Standardized Units
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations, Section 6 – Source Data and Accuracy of Data Entry
A Data Manager is designing a report to facilitate discussions with sites regarding late data. Which is the most important information to display on the report to encourage sites to provide data?
Number of forms entered in the last week
Expected versus actual forms entered
List of outstanding forms
Total number of forms entered to date
In managingsite data timeliness, the most actionable and effective tool is areport listing all outstanding (missing or incomplete) CRFs.
According toGCDMP (Chapter: Communication and Study Reporting), Data Managers must providesite-level performance reportshighlighting:
Outstanding CRFs not yet entered,
Unresolved queries, and
Pending data corrections.
Such reports help sites prioritize and address data gaps efficiently.
Option AandDare historical metrics without actionable context.
Option Bgives a general overview but lacks specific site-level actionability.
Hence,option C (List of outstanding forms)provides the clearest and most motivating feedback to sites for timely data entry and query resolution.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Communication and Study Reporting, Section 5.3 – Data Timeliness and Reporting Metrics
ICH E6(R2) GCP, Section 5.1.1 – Sponsor Oversight and Data Communication Requirements
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations, Section 6.5 – Site-Level Data Timeliness Reporting
When reviewing local lab data from a paper study, a Data Manager notices there are lab values not entered. What should the Data Manager request data-entry personnel do?
Flag the module for review
Call the patient to verify the information
Issue a query
Nothing
Whenlaboratory dataare missing from a paper-based clinical study, theData Managershould directdata-entry personnel to issue a queryto the investigative site for clarification or correction.
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Data Validation and Cleaning), every missing, inconsistent, or out-of-range data point must be reviewed and, if necessary, resolved through the formalquery management process. This ensures that all discrepancies between the source documents and database entries are properly documented, traceable, and auditable.
Data-entry staff arenot authorizedto infer or fill in missing information. They must escalate such discrepancies to the site via query, preservingdata integrityandregulatory compliancewithICH E6 (R2)andFDA 21 CFR Part 11. Calling the patient directly (option B) would violate confidentiality and site communication protocol, while simply flagging or ignoring the issue (options A and D) would not meet GCDMP query resolution standards.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Validation and Cleaning, Section 5.2 – Query Management and Resolution
ICH E6 (R2) Good Clinical Practice, Section 5.18.4 – Communication of Data Discrepancies
FDA 21 CFR Part 11 – Electronic Records; Query Audit Trails Requirements
An astute monitor discovers that a site is using nebulized albuterol rather than the inhaler provided in the study screening kit for the albuterol challenge. Which is the best response from the Data Manager?
No response is needed, the problem does not impact data
Contact the Ethics Committee
Update the CRF Completion Guidelines and notify all sites of the update
Query the site to enter a Protocol Violation
In this scenario, the site hasdeviated from the approved study protocolby using adifferent formulation (nebulized albuterol instead of inhaler). This is considered aprotocol deviation or violation, depending on study definitions.
PerGCDMP (Chapter: Data Validation and Cleaning)andICH E6(R2), Data Managers are responsible for ensuring that all protocol deviations affecting data integrity or subject safety areaccurately captured and documentedwithin the clinical database. The appropriate action is toissue a data queryprompting the site to record the deviation in the designated section (e.g., “Protocol Deviations” CRF).
Option A:Incorrect — it affects data comparability.
Option B:Escalation to the Ethics Committee is handled by the sponsor, not the Data Manager.
Option C:Updating the CRF guidelines is premature; first, the deviation must be logged and assessed.
Therefore,option D (Query the site to enter a Protocol Violation)is the correct and compliant action.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Data Validation and Cleaning, Section 6.2 – Query Management and Protocol Deviations
ICH E6(R2) GCP, Section 4.5 – Compliance with Protocol
FDA Guidance for Industry: Oversight of Clinical Investigations — Compliance and Protocol Deviation Reporting
Which mode of data entry is most commonly used in EDC systems?
Double entry
Blind verification
Single entry
Third party compare
Themost common mode of data entryinElectronic Data Capture (EDC)systems issingle data entry.
According to theGCDMP (Chapter: Electronic Data Capture Systems), EDC systems have built-inedit checks, validation rules, and audit trailsthat ensure data accuracy and integrity at the point of entry. These real-time validation capabilities makedouble data entry(a legacy practice from paper studies) unnecessary.
EDC systems automatically verify data as they are entered by site staff, generating queries for inconsistencies or out-of-range values immediately.Blind verification (option B)andthird-party comparisons (option D)are not standard data entry modes but may be used for specialized reconciliation or external data imports.
Thus,single data entry (Option C)is the industry standard approach, ensuring both efficiency and compliance withFDA 21 CFR Part 11andICH E6 (R2)data integrity requirements.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Electronic Data Capture (EDC) Systems, Section 5.4 – Data Entry and Verification Processes
ICH E6 (R2) Good Clinical Practice, Section 5.5.3 – Computerized Systems and Data Validation
FDA 21 CFR Part 11 – Electronic Records and Electronic Signatures: Validation and Data Entry Requirements
What are the key deliverables for User Acceptance Testing?
Project Plan
Training
Test Plan/Script/Results
eCRF Completion Guidelines
Thekey deliverables for User Acceptance Testing (UAT)are theTest Plan, Test Scripts, and Test Results.
According to theGCDMP (Chapter: Database Design and Validation), UAT is the final validation step before a clinical database is released for production. It confirms that the system performs according to user requirements and protocol specifications.
The deliverables include:
UAT Test Plan:Defines testing objectives, scope, acceptance criteria, and responsibilities.
UAT Test Scripts:Provide step-by-step instructions for testing database functionality, edit checks, and workflows.
UAT Test Results:Document actual test outcomes versus expected outcomes, including any deviations and their resolutions.
These deliverables form part of the system validation documentation required underFDA 21 CFR Part 11andICH E6 (R2)to demonstrate that the database has been properly validated.
Project Plans (option A) and Training (option B) occur in earlier phases, while eCRF Completion Guidelines (option D) support site data entry, not system validation.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Database Design and Validation, Section 5.3 – User Acceptance Testing Deliverables
FDA 21 CFR Part 11 – Validation Documentation Requirements
ICH E6 (R2) Good Clinical Practice, Section 5.5.3 – System Validation Records
In a study conducted using paper CRFs, a discrepancy is discovered in a CRF to database QC audit. What is the reason why this discrepancy would be considered an audit finding?
Discrepancy not explained by the protocol
Discrepancy not explained by the CRF completion guidelines
Discrepancy not explained by the data handling conventions
Discrepancy not explained by the data quality control audit plan
In aCRF-to-database quality control (QC) audit, auditors compare data recorded on the paper Case Report Form (CRF) with data entered in the electronic database. If discrepancies exist thatcannot be explained by documented data handling conventions, they are classified asaudit findings.
PerGCDMP (Chapter: Data Quality Assurance and Control),data handling conventionsdefine acceptable data entry practices, transcription rules, and allowable transformations. These conventions ensure that CRF data are consistently interpreted and entered.
If a discrepancy deviates from these established rules, it indicates a process gap or error in data entry, validation, or training. Discrepancies justified by protocol design or CRF guidelines would not constitute findings.
Therefore,option C (Discrepancy not explained by the data handling conventions)correctly identifies the criterion for a true QC audit finding.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Data Quality Assurance and Control, Section 6.1 – Data Handling Conventions and QC Auditing
ICH E6(R2) GCP, Section 5.1 – Quality Management and Documentation of Deviations
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations, Section 6.5 – Data Verification and Audit Findings
A Data Manager receives an audit finding of missing or undocumented training for two database developers according to the organization's training SOP and matrix. Which is the best response to the audit finding?
Identify the root cause and improve the process to prevent it
Remove the training items from the training matrix
Reprimand the person responsible for maintaining training documentation
Send the two developers to the required training
When an audit identifiesmissing or undocumented training, the most appropriate and compliant response is toidentify the root causeof the issue andimplement corrective and preventive actions (CAPA)to ensure that similar findings do not recur.
According toGood Clinical Data Management Practices (GCDMP, Chapter: Quality Management and Auditing), effective quality systems require root cause analysis (RCA) for all audit findings. The process involves:
Investigating why the documentation gap occurred (e.g., poor tracking, outdated SOP, or lack of oversight).
Correcting the immediate issue (e.g., ensuring the developers complete or document training).
Updating processes, training systems, or oversight mechanisms to prevent recurrence.
While sending the two developers to training (D) addresses thesymptom, it does not resolve thesystemic issueidentified by the audit. Options B and C are non-compliant and do not address quality system improvement.
Therefore,option A (Identify the root cause and improve the process)is the best and CCDM-compliant response.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Quality Management and Auditing, Section 6.2 – Corrective and Preventive Actions (CAPA)
ICH E6(R2) GCP, Section 5.1.1 – Quality Management and Continuous Process Improvement
FDA 21 CFR Part 820.100 – Corrective and Preventive Action (CAPA) Requirements
Which is a minimum prerequisite that should be in place before choosing an EDC system?
Knowledge of functional requirements
Completed installation qualification
Updated governance documentation
Draft validation plan
Before selecting anElectronic Data Capture (EDC)system for a clinical trial, it is essential to have a clear understanding of thefunctional requirements. This serves as theminimum prerequisiteto guide system selection, ensuring that the EDC solution aligns with the protocol needs, data workflow, security requirements, and regulatory compliance.
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Computerized Systems and Compliance), functional requirements describe what the system must do—such as data entry capabilities, edit checks, query management, user roles, audit trails, and integration with external systems (e.g., labs, ePRO). This understanding allows sponsors and CROs to evaluate vendor systems effectively during the selection and qualification phase.
Other options:
B. Installation qualificationandD. Validation planoccuraftersystem selection.
C. Governance documentationsupports operations but is not required before choosing the system.
Hence,option Ais correct — the first and most essential prerequisite before EDC selection is a solid understanding of thefunctional requirements.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Computerized Systems and Compliance, Section 4.2 – Requirements Gathering and System Selection
FDA 21 CFR Part 11 – System Validation and Intended Use Requirements
ICH E6(R2) GCP, Section 5.5.3 – Computerized System Selection and Qualification
All range and logic checks have been resolved in a study. An auditor found discrepancies between the database and the source. Which reason is most likely?
The auditor made an error
The discrepant data values were logical and in range
Data were changed after the checks were run
Data were not abstracted correctly from the source
Even when allrangeandlogic checksare successfully resolved, discrepancies may still exist between theclinical databaseand thesource documents. This typically indicates anerror in data abstraction or transcription, meaning that data were incorrectly entered or extracted from the source records during the data entry or verification process.
According to theGood Clinical Data Management Practices (GCDMP, Chapter on Data Validation and Cleaning),data validation rulessuch as range and logic checks are designed to identify inconsistencies, missing data, or out-of-range valueswithin the databaseitself. However, they donot verify the accuracy of data entry against the original source documents— that responsibility falls undersource data verification (SDV), typically conducted by clinical monitors or auditors.
When an auditor detects discrepancies between source and database values after all edit checks have passed, the most probable explanation is thatdata were not transcribed correctly from the source, rather than a failure in programmed edit checks. This could occur due to human error during manual data entry, misinterpretation of the source document, or oversight during SDV.
OptionC (Data were changed after checks were run)might occur in rare cases but would normally be documented in an audit trail per21 CFR Part 11andICH E6 (R2)standards. OptionBmisinterprets the issue, since “logical and in range” values can still be incorrect relative to the source. OptionA (Auditor error)is possible but statistically less likely, as source data verification follows strict, documented audit procedures.
Therefore, themost likely reasonfor such discrepancies isOption D: Data were not abstracted correctly from the source, emphasizing the importance of robust data entry training, dual data entry, and verification procedures.
Reference (CCDM-Verified Sources):
Society for Clinical Data Management (SCDM), Good Clinical Data Management Practices (GCDMP), Chapter: Data Validation and Cleaning, Section 6.5 – Source Data Verification and Reconciliation
ICH E6 (R2) Good Clinical Practice, Section 5.18 – Monitoring and Source Data Verification
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations, Section 6 – Source Data Accuracy and Audit Trails
21 CFR Part 11 – Electronic Records and Electronic Signatures, Subpart B: Audit Trails and Record Accuracy
An external organization has been hired to manage SAE follow-up for a large study. Which of the following would be used as guidance for exchange of the SAE data between the EDC system and the vendor's safety management system?
Medical Document for Regulatory Activities
Biomedical Research Domain Model
Individual Case Safety Report
Submission Data Tabulation Model
TheIndividual Case Safety Report (ICSR)is the standard format used globally for the exchange ofSerious Adverse Event (SAE)data between clinical data management systems (EDC) and safety management systems.
According toICH E2B(R3)andGood Clinical Data Management Practices (GCDMP, Chapter: Safety Data Management and SAE Reconciliation), the ICSR provides thedata structure and content standardsfor electronic transmission of safety data, including patient demographics, event details, outcomes, and product information. It ensures interoperability between systems by defining standardized message elements and controlled terminologies.
Other options are not applicable:
A. Medical Document for Regulatory Activities (MDRA)is not a recognized standard.
B. Biomedical Research Domain Model (BRIDG)provides conceptual modeling but not data exchange guidance.
D. SDTMis used for regulatory submission datasets, not real-time SAE exchange.
Thus,option C (Individual Case Safety Report)is correct, as it defines the internationally accepted electronic format for SAE data exchange between safety and clinical databases.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Safety Data Management and SAE Reconciliation, Section 4.3 – SAE Data Exchange and Standards
ICH E2B(R3): Electronic Transmission of Individual Case Safety Reports
FDA Guidance for Industry: Providing Regulatory Submissions in Electronic Format — Postmarketing ICSRs (2014)
To ensure data quality and efficient integration of data, which of the following best describes the main topic that should be covered in initial discussions with a vendor providing the external data?
Metrics that will be used to measure data quality
Criteria to trigger audits based on performance-monitoring reports
Acceptable record, field, and file formats
Standard dictionary versioning and maintenance
In initial vendor discussions forexternal data integration(e.g., central lab, ECG, imaging vendors), themost critical and foundational topicis defining theacceptable record, field, and file formats.
According to theGCDMP (Chapter: External Data Transfers and Integration), establishing theData Transfer Specifications (DTS)early in the process ensures consistent structure, proper mapping, and compatibility between the vendor’s system and the sponsor’s database. These specifications define:
Data structure (variable names, formats, delimiters)
File naming conventions
Frequency of transfers
Methods of secure data transmission
Discussing formats first allows later alignment on data validation, quality metrics, and dictionary standards (which occur in subsequent stages). Without format agreement, all downstream processes risk misalignment, resulting in data incompatibility and rework.
Thus,option C (Acceptable record, field, and file formats)correctly represents the foundational focus of initial vendor discussions for ensuring data quality and integration efficiency.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: External Data Transfers and Integration, Section 4.1 – Data Transfer Planning and Specification Development
ICH E6(R2) GCP, Section 5.5.3 – Data Handling and System Validation
FDA Guidance: Computerized Systems Used in Clinical Investigations, Section 6.3 – Data Import and Format Control
A site study coordinator attempts to make an update in a study database in an EDC system after lock. What occurs?
The old value is replaced in all locations by the new value
The change is approved by the Data Manager before it is applied
The site study coordinator is not able to make the change
The change is logged as occurring after lock
Once a clinical database islocked, it becomesread-only— no further data modifications can be made by any users, including site personnel. This ensures that the data arefinalized, consistent, and auditablefor statistical analysis and regulatory submission.
According to theGCDMP (Chapter: Database Lock and Archiving), the lock process involves freezing the database to prevent accidental or unauthorized changes. After lock, access permissions are restricted, and all edit and update functions are disabled. If any corrections are required post-lock, the database must beunlocked under controlled procedures(with full audit trail documentation).
Thus,option C—The site study coordinator is not able to make the change— correctly reflects standard EDC functionality and regulatory compliance.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Database Lock and Archiving, Section 5.2 – Database Lock Procedures and Controls
ICH E6(R2) GCP, Section 5.5.3 – Data Integrity and Audit Trail Requirements
FDA 21 CFR Part 11 – Controls for Electronic Records and System Lock Functions
Which information should an auditee expect prior to an audit?
Auditor's credentials and certification number
Corrective action requests
Standard operating procedures
Audit plan or agenda
Prior to an audit, theauditeeshould expect to receive anaudit plan or agenda, which outlines thescope, objectives, schedule, and logisticsof the audit.
According to theGCDMP (Chapter: Quality Assurance and Audits), anaudit planensures transparency, preparation, and efficient execution. It typically includes details such as:
The audit scope and objectives,
The audit team members,
Documents or processes to be reviewed, and
The audit schedule and timeframe.
This allows the auditee to prepare the necessary records, staff, and facilities. While the auditor’s credentials (option A) may be shared informally, they are not a regulatory requirement.Corrective actions (option B)are outcomes of the audit, not pre-audit materials.Standard Operating Procedures (option C)may be requested during the audit but are not provided in advance.
Thus,Option D – Audit Plan or Agenda– is the correct and compliant answer.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Quality Assurance and Audits, Section 6.1 – Pre-Audit Planning and Communication
ICH E6 (R2) Good Clinical Practice, Section 5.19.3 – Audit Procedures and Responsibilities
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations – Section 8.1 – Audit Preparation and Planning
When a data manager runs a report on resolution types of discrepancy status, which of the following wouldNOTbe a part of resolution types?
Cannot be resolved (but data incorrect)
Received from site and not yet reviewed
Resolved with data/confirmed as is (non problematic)
Data management – self evident corrections
In a discrepancy management workflow,“Received from site and not yet reviewed”isnot a resolution type— it represents astatus, not a final resolution outcome.
According to theGCDMP (Chapter: Data Validation and Cleaning), resolution types describe how a data discrepancy was finalized or addressed, such as:
Resolved with data correction,
Confirmed as correct (no change required),
Self-evident correction applied by data management, or
Unresolvable discrepancies documented.
In contrast,statusesdescribe the stage of the query (e.g., open, sent, answered, pending review, closed). “Received from site and not yet reviewed” indicates anintermediate workflow statewhere the response awaits validation by data management.
Proper classification of resolution types is essential for performance reporting, audit readiness, and ensuring the traceability of query management actions underICH E6 (R2)andFDA 21 CFR Part 11.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Validation and Cleaning, Section 5.3 – Discrepancy Resolution Lifecycle
ICH E6 (R2) Good Clinical Practice, Section 5.5.3 – Data Handling and Record Management
FDA 21 CFR Part 11 – Electronic Records; Audit Trails and Discrepancy Tracking Requirements
Which is the MOST appropriate flow for EDC set-up and implementation?
CRF “wire-frames” created, CRFs reviewed, CRFs printed, CRFs distributed to sites
Protocol finalized, Database created, Edit Checks created, Database tested, Sites trained
Database created, Subjects enrolled, Database tested, Sites trained, Database released
Database created, Database tested, Sites trained, Protocol finalized, Database released
The correct and compliant sequence forEDC system setup and implementationbegins onlyafter the study protocol is finalized, as all case report form (CRF) designs, database structures, and validation rules derive directly from the finalized protocol.
According toGCDMP (Chapter: EDC Systems Implementation), the proper order is:
Protocol finalized– defines endpoints and data requirements.
Database created– built according to the protocol and CRFs.
Edit checks created– programmed to validate data entry accuracy.
Database tested (UAT)– ensures functionality, integrity, and compliance.
Sites trained and system released– only then can data entry begin.
Option B follows this logical and regulatory-compliant sequence. Other options (A, C, D) are eitherpaper-based workflowsor violateGCP-compliant timelines(e.g., enrolling subjects before database validation).
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Electronic Data Capture (EDC) Systems, Section 5.2 – System Setup and Implementation Flow
ICH E6(R2) GCP, Section 5.5.3 – Computerized Systems Validation and User Training Before Use
FDA 21 CFR Part 11 – Validation and System Release Requirements
A study is collecting pain levels three times a day. Which is the best way to collect the data?
Using paper pain diary cards completed by study subjects
Sites calling patients daily and administering a pain questionnaire
Study subjects calling into an IVRS three times a day to enter pain levels
Using ePRO with reminders for data collection at each time point
The optimal method for collectingfrequent patient-reported pain datais throughelectronic Patient-Reported Outcomes (ePRO)with built-inreminder functionality.
According to theGCDMP (Chapter: Electronic Data Capture Systems), ePRO systems provide avalidated, real-time, and user-friendly interfacefor subjects to record time-sensitive data accurately. The use ofautomated remindersensures compliance with protocol-specified data collection times, improving data completeness and accuracy.
Paper diaries (option A) are prone torecall bias and backfilling, while daily site calls (option B) areresource-intensiveand introduce human error. IVRS systems (option C) are acceptable but less efficient and user-friendly than modern ePRO applications, which can integrate timestamp validation, compliance monitoring, and real-time alerts.
ePRO systems also comply withFDA 21 CFR Part 11andICH E6 (R2)for audit trails, authentication, and validation, making them the preferred solution for repeated PRO data collection.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Electronic Data Capture (EDC) Systems, Section 6.1 – Use of ePRO for Repeated Measures
FDA Guidance for Industry: Electronic Source Data in Clinical Investigations, Section 5 – ePRO Compliance and Validation
ICH E6 (R2) GCP, Section 5.5.3 – Electronic Data Systems and Recordkeeping
During a database audit, it was determined that there were more errors than expected. Who is responsible for assessing the overall impact on the analysis of the data?
Data Manager
Statistician
Quality Auditor
Investigator
TheStatisticianis responsible for assessing theoverall impact of data errors on the analysis and study results.
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Data Quality Assurance and Control)andICH E9 (Statistical Principles for Clinical Trials), while theData Managerensures data accuracy and completeness through cleaning and validation, theStatisticiandetermines whether the observed data discrepancies are statistically significant or if they may affect thevalidity, power, or interpretabilityof the study’s outcomes.
TheQuality Auditor (C)identifies and reports issues but does not quantify analytical impact. TheInvestigator (D)is responsible for clinical oversight, not statistical assessment. Thus, after a database audit, theStatistician (B)performs a formal evaluation to determine whether the magnitude and nature of the errors could bias results or require reanalysis.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Quality Assurance and Control, Section 7.3 – Data Audit and Impact Assessment
ICH E9 – Statistical Principles for Clinical Trials, Section 3.2 – Data Quality and Analysis Impact Assessment
FDA Guidance for Industry: Computerized Systems Used in Clinical Investigations – Data Validation and Analysis Review
Based on the project Gantt chart as of 01 Nov 2019, an interim analysis is scheduled to occur early Q2 of 2020. All of the following are valid for initially assessing the status of data cleanliness EXCEPT:
Determining CRF data entry status of received pages
Identifying missing pages where visits have been completed to date
Identifying the number of discrepancies resolved to date
Identifying all outstanding discrepancies to date and aging
When initially assessingdata cleanlinessin preparation for aninterim analysis, the focus should be onoutstanding issuesthat could affect data completeness and reliability.
According to theGCDMP (Chapter: Data Quality Assurance and Control), key indicators of readiness include:
TheCRF data entry statusof received pages (option A) to confirm completeness.
Identification ofmissing pages or visits(option B) to verify subject-level completeness.
A listing ofoutstanding discrepancies and their aging(option D) to assess unresolved data issues.
Counting the number ofdiscrepancies resolved to date (option C), however, does not reflect data quality or current data readiness—it indicates past actions rather than current unresolved risks. Therefore, it isnot a valid measurefor assessing interim data cleanliness.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Quality Assurance and Control, Section 6.1 – Data Readiness Assessments for Analysis
ICH E6 (R2) GCP, Section 5.18.4 – Ongoing Data Quality Review
FDA Guidance for Industry: Oversight of Clinical Investigations – Risk-Based Monitoring, Section 7 – Data Quality Indicators
Which of the following actions is particularly important in merging data from different trials?
Use of a common software platform
Enrollment of investigative sites with similar patient populations
Exclusion of studies that use a cross-over design
Use of a common adverse event dictionary
Whenmerging data from different clinical trials, theuse of a common adverse event (AE) dictionary(such asMedDRAorWHO Drug) is essential to ensure consistency and comparability across datasets.
According to theGCDMP (Chapter: Standards and Data Mapping)andCDISC SDTM Implementation Guide, data integration across studies requires standardized terminology for adverse events, medications, and clinical outcomes. Using the same AE dictionary ensures that similar terms are coded consistently, allowing accurate cross-study analysis, pooled summaries, and safety reporting.
A sharedsoftware platform (option A)is not necessary if data are mapped to standard formats (e.g., CDISC SDTM). Patient population similarity (option B) affects interpretation but not technical data merging. Study design differences (option C) may influence statistical analysis but not data integration mechanics.
Therefore,Option D – Use of a common adverse event dictionary– is the correct and most critical action for consistent multi-study data integration.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Standards and Data Mapping, Section 5.1 – Use of Standardized Coding Dictionaries
CDISC SDTM Implementation Guide, Section 4.3 – Controlled Terminology and Cross-Study Integration
ICH E3 and E2B – Clinical Data Standards and Safety Coding Requirements
An asthma study is taking into account local air quality and receives that data from the national weather bureau. Which information is needed to link research subject data to the air-quality readings?
Location identifier
Location and time identifiers
Location, time and subject identifiers
Location, time, subject and site identifiers
When integratingexternal environmental datasuch asair quality readingswith clinical study data, it is essential to uselocation and time identifiersto properly align the environmental data with subject-level data.
According to theGood Clinical Data Management Practices (GCDMP, Chapter: Data Management Planning and Study Start-up), external data sources (like national weather or pollution databases) must be merged usingcommon linkage variablesthat allow synchronization without breaching subject confidentiality. In this case:
Location identifiers(e.g., city, postal code, or region) align the subject’s study site or residential area with the environmental dataset.
Time identifiers(e.g., date and time of data collection) ensure that the environmental readings correspond to the same period as the subject’s clinical observations.
Including subject identifiers (option C or D) is unnecessary and would poseprivacy and data protection risks. Instead, linkage is typically done at theaggregate (site or regional) level, maintaining compliance withHIPAAandGDPR.
Reference (CCDM-Verified Sources):
SCDM Good Clinical Data Management Practices (GCDMP), Chapter: Data Integration and External Data Handling, Section 4.3 – Linking External Data Sources
ICH E6 (R2) GCP, Section 5.5.3 – Data Traceability and External Data Management
FDA Guidance for Industry: Use of Electronic Health Record Data in Clinical Investigations, Section 5.2 – Linking and Integration Principles
A relational database has tables for PATIENT_DEMOGRAPHY and VITAL_SIGNS data collected during a visit. The primary key for the VITAL_SIGNS table is a composite key that includes the unique patient identifier, visit number, and vital signs parameter name. The two tables are joined on the patient identifier. What will be the number of records in the result set?
One record per patient
One record per visit
One record per patient per visit per vital sign parameter
One record per patient per visit
In arelational database structure, each record in a table is uniquely identified by aprimary key. In this case, theVITAL_SIGNStable uses acomposite primary keyconsisting of:
Patient Identifier,
Visit Number, and
Vital Signs Parameter Name.
This means each record represents aunique measurement of a specific parameter (e.g., blood pressure, pulse)for a patient at a specific visit.
When joiningPATIENT_DEMOGRAPHYandVITAL_SIGNStables on thepatient identifier, the result set will includeone record for every combination of patient, visit, and parameter— i.e.,one record per patient per visit per vital sign parameter.
Therefore,option Ccorrectly describes the expected number of records.
Reference (CCDM-Verified Sources):
SCDM GCDMP, Chapter: Database Design and Build, Section 5.2 – Primary and Foreign Key Relationships in Relational Models
CDISC SDTM Implementation Guide, Section 5.3 – Observation-Level Data Structures
ICH E6(R2) GCP, Section 5.5.3 – Data Organization and Integration Principles
TESTED 09 Oct 2025
Copyright © 2014-2025 DumpsTool. All Rights Reserved