Week 8: Protocol Development and Study Management

TABLE OF CONTENTS

  1. Learning Objectives & Exam Context
  2. ICH E6(R2) Section 6: Protocol Requirements (14 Sections)
  3. Inclusion/Exclusion Criteria Development & Application
  4. Protocol Amendments vs. Administrative Changes
  5. Protocol Deviations vs. Violations (CRITICAL CONTENT)
  6. IRT/IWRS/RTSM Systems Overview
  7. Drug Accountability & Chain of Custody
  8. Interactive Case Studies
  9. Glossary & Key Terminology
  10. Exam Preparation Strategy

SECTION 1: LEARNING OBJECTIVES & EXAM CONTEXT

Week 8 Learning Outcomes

By the end of this week, you will be able to:

  1. Identify all 14 required sections of ICH E6(R2) Section 6 protocol content
  2. Apply inclusion/exclusion criteria to real subject enrollment scenarios
  3. Distinguish protocol amendments from administrative changes with regulatory reasoning
  4. Classify protocol deviations as minor/major and violations with supporting rationale
  5. Explain how IRT/IWRS systems prevent deviations through automated controls
  6. Calculate drug accountability reconciliation with complex scenarios
  7. Bridge implementation science background to IND/IDE regulatory frameworks

Why Week 8 Matters for Your Exam Score

SOCRA Exam Strategy for Week 8


SECTION 2: ICH E6(R2) SECTION 6 - PROTOCOL REQUIREMENTS

Overview: The "Gold Standard" Framework

ICH E6(R2) Section 6 provides the internationally recognized template for clinical trial protocol content. FDA expects IND protocols to follow this structure. SOCRA exam heavily tests protocol section identification and content requirements.

Critical fact: Protocols don't MUST include every section word-for-word, but missing content can trigger IND Response Letters. For FDA submissions, the expectation is comprehensive coverage of all topics in Section 6.

The 14 Major Sections at a Glance

SECTION 6.1: General Information

Who is responsible? Administrative identification of key parties

Required Elements:

Why It Matters: Identifies the chain of responsibility. In regulatory audits, inspectors verify these contacts. If sponsor moves and contact isn't updated → potential deviation identification.

Exam Question Type: "Who signs amendments?" Answer: Person authorized by sponsor (from 6.1)


SECTION 6.2: Background Information

What are we studying and why is it important?

Required Elements:

Why It Matters: Provides scientific rationale. Supports the "why we're doing this" foundation. Informs subject selection (population description) and risk/benefit disclosure requirements.

Implementation Science Bridge: Your implementation science protocols may discuss feasibility, stakeholder engagement, organizational readiness. IND protocols focus more narrowly on medical/pharmacological rationale and patient safety.

Exam Question Type: "The protocol must discuss known and potential risks. Why?" Answer: To inform informed consent and establish that benefits justify risks for the population.


SECTION 6.3: Trial Objectives & Purpose

What is the study question? How will we know if it worked?

Required Elements:

CRITICAL DISTINCTION:

Why It Matters: Primary endpoint determines statistical power. Changing it mid-study = protocol amendment. SOCRA exams frequently test recognition of endpoints vs. objectives.

Exam Question Type: "A protocol states 'To improve quality of life.' Is this sufficient?" Answer: No—needs measurable endpoint (e.g., QOL score improvement ≥10 points).


SECTION 6.4: Trial Design

How will the study be conducted? (The largest, most detailed section)

Subsection Overview:

6.4.1: Type of Trial

6.4.2: Trial Treatment Assignment and Blinding

6.4.3: Inclusion and Exclusion Criteria

6.4.4: Withdrawal Criteria

6.4.5: Investigational Product Accountability

6.4.6: Dosing and Treatment Modifications

6.4.7: Efficacy Assessments and Procedures

6.4.8: Safety Assessments and Adverse Event Reporting

Why 6.4 Matters: This section defines the CONDUCT of the study. Protocol deviations frequently occur when procedures in 6.4 aren't followed. Understanding 6.4 is essential for identifying deviations.

Exam Question Type: "A subject's visit occurred 4 days outside the protocol window. Where in 6.4 would this violation be identified?" Answer: Section 6.4.7 (efficacy assessment procedures define visit windows)


SECTIONS 6.5-6.14: Conduct & Documentation

Brief overview of sections 6.5 through 6.14

6.5 - Trial Procedures and Study Conduct

6.6 - Statistical Considerations

6.7 - Safety and Efficacy Evaluation Criteria

6.8 - Records and Reports

6.9 - Financing and Insurance

6.10 - Quality Overall Summary

6.11 - Protocol Signature Block and Version History

6.12-6.14 - Appendices


ICH E6 Section 6: Quick Reference Checklist

Use this to identify missing protocol sections on exam:

Section Content Key Elements
6.1 Identification Title, contacts, signatures
6.2 Background Rationale, risks, population
6.3 Objectives Primary & secondary endpoints
6.4 Design Methodology (8 subsections)
6.5 Procedures Visit schedule, consent process
6.6 Statistics Sample size, analysis plan
6.7 Evaluation Safety/efficacy criteria
6.8 Records CRF, lab specs, bioanalytical
6.9 Financing Funding, compensation, insurance
6.10 Quality Manufacturing, nonclinical data
6.11 Signature Authors, version history
6.12-6.14 Appendices IB, ICF, CRF, other

SECTION 3: INCLUSION/EXCLUSION CRITERIA

Definition & Purpose

Inclusion Criteria: Characteristics subjects MUST have to qualify for enrollment

Exclusion Criteria: Characteristics that prevent enrollment

Why I/E Criteria Matter (Regulatory + Scientific)

Regulatory perspective:

Scientific perspective:

Implementation Science Bridge: In implementation science, I/E criteria might be organizational (e.g., "Clinic must have ≥500 ED visits/year"). In IND trials, they're individual subject characteristics. The SOCRA exam focuses on individual-level criteria.


Common Inclusion Criteria Elements

Demographic Criteria

Example: "Age 18-75 years at time of enrollment"
- Clearly defined age range
- Tied to enrollment (not enrollment visit - week 1)

Disease Criteria

Example: "Confirmed diagnosis of Type 2 diabetes mellitus
  • Confirmed by HbA1c ≥7.0% within 3 months of enrollment
  • Duration ≥1 year at time of enrollment"
- Specific diagnosis with measurement
- Timeframe for confirmation
- Disease stability requirement (≥1 year)

Health Status Criteria

Example: "Estimated glomerular filtration rate (eGFR) ≥60 mL/min/1.73m²
  at screening (within 30 days of enrollment)"
- Numeric threshold (measurable)
- Specific assessment window
- Tied to enrollment timing

Medication Criteria

Example: "Stable dose on current antidiabetic medication for ≥30 days
  prior to enrollment"
- Medication stability (prevents confounding)
- Specific duration (measurable)
- Prevents medication changes affecting data

Common Exclusion Criteria Elements

Safety-Based Exclusions

Medical History:
- Recent myocardial infarction (within 6 months)
  Rationale: Cardiac stability needed for study drug safety

Medication:
- No concurrent ACE inhibitors (within 2 weeks of enrollment)
  Rationale: Drug-drug interaction with study medication

Physiologic:
- Pregnancy or breastfeeding
  Rationale: Teratogenic risk to fetus/infant

Organ Function:
- eGFR <45 mL/min/1.73m²
  Rationale: Reduced renal function affects drug clearance

Scientific-Based Exclusions

- Concurrent participation in other trials (washout period)
  Rationale: Drug-drug interactions, confounding

- Known drug allergy to study medication class
  Rationale: Safety and data validity

- Inability to provide informed consent
  Rationale: Ethical requirement for voluntary participation

Screening Failures vs. Subject Refusal

IMPORTANT DISTINCTION:

Screening Failure:

Subject Refusal:

Exam Context: FDA wants to know enrollment rate = Enrolled ÷ Screened


Washout Periods & Pharmacokinetic Principles

Definition: Time required for drug to be substantially eliminated from the body before enrollment

Pharmacokinetic Principle:

Calculation Example:

Drug half-life: 6 hours
5 half-lives = 5 × 6 hours = 30 hours = 1.25 days

Protocol requirement: "No drug X within 30 hours of enrollment"
Subject's last dose: Monday 2:00 PM
Acceptable enrollment: Tuesday 8:00 PM or later

Protocol Language Example:

"No ACE inhibitors within 2 weeks (14 days) of enrollment"

Subject's last dose: May 10
Earliest enrollment: May 10 + 14 days = May 24 or later
Subject enrolls: May 23 = DOES NOT MEET CRITERION = screening failure

Screening Window Timing (Frequently Confused):

Criterion: "Baseline labs within 30 days of enrollment"
Does this mean:
a) 30 days before enrollment? NO
b) 30 days before baseline visit? NO
c) Labs obtained within 30 days of the enrollment date? YES

Subject enrolls: June 15
Labs must be dated June 15 ± 30 days
Acceptable lab dates: May 16 - July 15

I/E Application: Practical Scenarios

Scenario 1: eGFR Threshold

Protocol: Inclusion—"eGFR ≥60 mL/min/1.73m²"
Subject's screening eGFR: 62
Decision: ELIGIBLE ✓

Same protocol
Subject's screening eGFR: 45
Decision: INELIGIBLE ✗
  (Might feel "close to 60," but mathematically 45 < 60)
  (The "eGFR <60 excluded" is a real deviation risk)

Scenario 2: Disease Duration

Protocol: "Type 2 diabetes diagnosis ≥2 years at enrollment"
Subject diagnosed: May 2022
Enrollment date: April 2024
Duration: ~2 years (23 months - NOT yet 2 full years)
Decision: Does NOT meet criterion until May 2024 ✗

Enrollment date: May 2024
Duration: Exactly 2 years
Decision: ELIGIBLE ✓

Scenario 3: Medication Stability

Protocol: "Stable dose on current antidiabetic for ≥30 days prior"
Subject's medication record:
- Started metformin 1000mg: March 1
- Enrollment: April 15
- Days on stable dose: 45 days
Decision: ELIGIBLE ✓

vs.

- Started metformin 1000mg: April 1
- Enrollment: April 15
- Days on stable dose: 14 days
Decision: INELIGIBLE (needs 30 days) ✗

SECTION 4: PROTOCOL AMENDMENTS VS. ADMINISTRATIVE CHANGES

The Critical Distinction

PROTOCOL AMENDMENT:

ADMINISTRATIVE CHANGE:

Why This Distinction Matters


Clear Amendment Examples (Must Have Approval)

Change Classification Why
Change primary endpoint AMENDMENT Changes main study objective
Modify inclusion criteria AMENDMENT Changes eligible population
Increase sample size AMENDMENT Changes statistical power
Add new efficacy measurement AMENDMENT Adds data collection burden
Extend study duration AMENDMENT Extends subject follow-up
Reduce dose range AMENDMENT Changes treatment intensity
Add safety monitoring requirement AMENDMENT Affects subject safety oversight

Key Teaching Point: If change affects WHO can enroll, WHAT is measured, or HOW it's measured → likely an amendment.


Clear Administrative Examples (Minimal Approval)

Change Classification Why
Sponsor phone number update ADMINISTRATIVE Notification routing only
Investigator office relocation (same institution) ADMINISTRATIVE Location update, no conduct change
Protocol formatting/layout ADMINISTRATIVE No content change
Correcting typo in protocol ADMINISTRATIVE Clerical error correction
Update laboratory contact ADMINISTRATIVE Notification routing
Correct investigator name spelling ADMINISTRATIVE Clerical correction

Key Teaching Point: If change affects ONLY how protocol is documented (not conducted) → administrative.


Gray Areas & Decision-Making Framework

Some changes could be EITHER amendment or administrative depending on context:

Example 1: Change Clinical Laboratory

Scenario A: Same test, different facility
Action: Might be ADMINISTRATIVE (if assay unchanged)

Scenario B: Different assay methodology
Action: AMENDMENT (affects data validity)

Scenario C: Different turnaround time (1 day vs. 7 days)
Action: Depends on whether timing matters for protocol
        If timing affects safety → AMENDMENT
        If timing doesn't affect protocol window → ADMINISTRATIVE

Example 2: Modify Case Report Form

Scenario A: Add new field requiring data collection
Action: AMENDMENT (adds burden)

Scenario B: Remove unused field
Action: ADMINISTRATIVE (doesn't affect collection)

Scenario C: Change field label for clarity (same data)
Action: ADMINISTRATIVE (same data, clearer label)

Rule of Thumb: When unsure, classify as AMENDMENT (conservative approach protects study integrity)


Amendment Approval Process & Timeline

Standard Amendment Process:

Step 1: Identify Need (sponsor, investigator, FDA communication)
        ↓
Step 2: Sponsor Drafts Amendment
        - Describes change
        - Provides rationale
        - References revised protocol sections
        ↓
Step 3: Sponsor Submits to IRB
        - Electronic submission
        - Convened review typical
        ↓
Step 4: IRB Review (typically 30 days)
        - Approval
        - Conditional approval (requests clarification)
        - Request for revisions
        ↓
Step 5: Sponsor Issues Final Amendment
        - Sends to all sites simultaneously (CRITICAL)
        - Includes IRB approval documentation
        ↓
Step 6: Sites Implement & Document
        - Acknowledge receipt
        - Train staff on changes
        - Implement per amendment effective date
        ↓
Step 7: Ongoing Compliance Tracking
        - Monitor sites for amendment compliance
        - Verify all new enrollments follow amended criteria

Timeline Considerations:

Planned Amendments:

Urgent Amendments (Safety-Driven):


Amendment Example: Adding Safety Monitoring

Scenario:

Week 3 of study: FDA publishes safety communication
  "Drug X may cause sudden cardiac arrhythmia (QTc prolongation)"

Protocol currently: No specific QTc monitoring required

Sponsor decision: Add "Baseline ECG screening—exclude if QTc >450ms"

Classification: AMENDMENT (changes inclusion criteria)

Process:
1. Sponsor drafts amendment: "Add 6.4.3 inclusion criterion: QTc ≤450ms"
2. Submits to IRB (expedited review possible—safety issue)
3. IRB approves within 5 business days
4. Sponsor notifies all sites immediately
5. Implementation effective date: Next Monday
6. New subjects screened with ECG before enrollment
7. Already-enrolled subjects: Safety assessment (test existing subjects for QTc, monitor if >450ms)

SECTION 5: PROTOCOL DEVIATIONS VS. VIOLATIONS (CRITICAL CONTENT)

The Most Important Week 8 Topic

Why? This distinction is tested 6-8 times on SOCRA exam AND is the most commonly misunderstood concept.

Foundational Definitions

PROTOCOL DEVIATION: Any noncompliance with the protocol (ICH E6)

PROTOCOL VIOLATION: Deviation that seriously compromises:

Critical Relationship:

ALL VIOLATIONS ⊂ DEVIATIONS
ALL VIOLATIONS ARE DEVIATIONS
SOME DEVIATIONS ARE VIOLATIONS
SOME DEVIATIONS ARE NOT VIOLATIONS

Types of Deviations

PLANNED DEVIATIONS (Medical Necessity)

Definition: Intentional deviation from protocol authorized in advance

Example 1: Medical Emergency Dosing

Protocol: "No antihypertensive medications during study"
Subject develops: Hypertensive crisis (BP 210/110) with chest pain
Action: Administer IV labetalol per emergency protocol
Documentation: "Planned deviation—medical emergency. Subject BP 210/110, risk of stroke, antihypertensive administered per critical care guidelines."
Status: NOT a violation (documented, medically justified, subject safety prioritized)

Example 2: Temporary Medication Use

Protocol: "No NSAIDs during trial"
Subject develops: Severe migraines during study
Action: Short-course NSAID prescribed (5 days) for medical necessity
Documentation: "Planned deviation—medical necessity. Subject experiencing severe migraines unresponsive to alternatives. 5-day NSAID course approved by investigator and sponsor."
Status: NOT a violation (if documented and medically justified)

Why Planned Deviations Aren't Always Violations:

UNPLANNED DEVIATIONS (Error/Oversight)

Definition: Unintentional protocol noncompliance

Example 1: Visit Timing

Protocol: "Week 4 visit ±3 days from Week 0"
Subject scheduled: Day 28
Subject actually visits: Day 31 (3 days late)
Status: UNPLANNED DEVIATION (oversight in scheduling)
Classification: Could be MINOR or MAJOR depending on:
  - If timing affects efficacy measurement: MAJOR
  - If timing doesn't affect data validity: MINOR

Example 2: Missed Assessment

Protocol: Requires 6 secondary endpoint assessments
Subject completes: 5 of 6 assessments (staff forgot one)
Status: UNPLANNED DEVIATION (staff oversight)
Classification: Likely MINOR (subject has 5 of 6, secondary endpoint, data partially available)

Example 3: Medication Taken by Subject

Protocol: "No NSAIDs during trial"
Subject takes: Ibuprofen for headache (didn't understand exclusion)
Duration: 2 days, then stops
Status: UNPLANNED DEVIATION (subject didn't follow protocol)
Classification: Could be MAJOR (drug-drug interaction possible) depending on study drug

Deviation Severity: Minor vs. Major

MINOR DEVIATION: Unlikely to affect subject safety or data validity

Examples:

Characteristics of Minor Deviations:

MAJOR DEVIATION: Could affect subject safety OR data validity

Examples:

Characteristics of Major Deviations:


Protocol Violations: Serious Breaches

DEFINITION: Deviation affecting SAFETY, DATA INTEGRITY, or INFORMED CONSENT

Type 1: Safety Violation

Scenario 1: Unevaluated Subject at Risk

Protocol: "eGFR ≥60 mL/min/1.73m² required"
Subject enrolled: eGFR 42 (below threshold)
Study drug: Renally metabolized
Safety issue: Reduced kidney function → increased drug levels → toxicity risk
Safety monitoring: Standard monitoring (insufficient for reduced renal clearance)
Status: VIOLATION
  Why? Subject at unknown risk; safety monitoring didn't account for renal impairment

Scenario 2: Unmonitored Safety Risk

Protocol: "Subjects with cardiac arrhythmia history excluded"
Subject enrolled: History of atrial fibrillation (not disclosed)
Protocol-required monitoring: Standard EKG at baseline and week 4
What happened: No EKG performed at baseline
Status: VIOLATION
  Why? Subject at higher cardiac risk; monitoring omitted; subject's risk undetected

Type 2: Data Integrity Violation

Scenario: Drug-Drug Interaction

Protocol: "No ACE inhibitors during study"
Study drug: Antiarrhythmic (interacts with ACE inhibitors)
Subject took: ACE inhibitor for 3 days (undetected)
Efficacy data: Subject shows unexpectedly poor response
Status: VIOLATION
  Why? Data validity compromised; can't attribute response to study drug (might be drug interaction)

Scenario: Undisclosed Risk

Protocol: Includes unknown risk of sudden cardiac arrhythmia
ICF: Does not mention arrhythmia risk
Subject enrolled: Given no specific cardiac monitoring (protocol doesn't require it)
Subject develops: Serious arrhythmia during study
Status: VIOLATION
  Why? Subject didn't consent to known/potential cardiac risk; monitoring inadequate for disclosed risk

Decision Framework: Classify as Deviation or Violation

Use this step-by-step approach:

STEP 1: Did protocol compliance fail?
        ├─ NO → Not a deviation
        └─ YES → Continue to Step 2

STEP 2: Could this affect subject SAFETY?
        ├─ NO → Continue to Step 3
        └─ YES (and unmonitored/unreported) → VIOLATION
                OR YES (but monitored) → MAJOR DEVIATION
        
STEP 3: Could this affect DATA INTEGRITY?
        ├─ NO → Continue to Step 4
        └─ YES (and compromises validity) → VIOLATION
                OR YES (but assessable) → MAJOR DEVIATION

STEP 4: Could this affect INFORMED CONSENT?
        ├─ NO → Continue to Step 5
        └─ YES → VIOLATION

STEP 5: Is this minor, documented, non-significant?
        ├─ YES → MINOR DEVIATION
        └─ NO (and affects something) → MAJOR DEVIATION

Reporting Requirements by Severity

MINOR DEVIATIONS

Documentation:

Reporting to Sponsor:

Reporting to IRB:

Retention:


MAJOR DEVIATIONS

Documentation:

Reporting to Sponsor:

Reporting to IRB:

Follow-up:


VIOLATIONS

Documentation:

Reporting to Sponsor:

Reporting to IRB:

Reporting to FDA:

Subject Management:


Corrective Action Plans (CAPA) for Violations

When a violation occurs, develop systematic CAPA addressing:

ROOT CAUSE ANALYSIS

Example Violation: Subject enrolled with eGFR 42 (protocol required ≥60)

Possible Root Causes:
1. Staff didn't understand the criterion (education gap)
2. Staff understood but made arithmetic error (QA gap)
3. Lab result misinterpreted (training gap)
4. Inclusion checklist missing (process gap)
5. Investigator didn't review labs (oversight gap)

Most Likely: Staff trained on I/E criteria but didn't understand numeric thresholds
  (thought 42 "was close enough to 60")

CORRECTIVE ACTIONS (Fix the Problem)

For above root cause:

1. Immediate action:
   - Retraining session on inclusion criteria
   - Specifically focus on numeric thresholds
   - Use examples: "42 is less than 60. Not eligible."

2. Verification:
   - Quiz staff on I/E criteria after training
   - Review next 5 subjects' screening documentation
   - Confirm all thresholds checked correctly

PREVENTIVE MEASURES (Prevent Recurrence)

1. Implement inclusion criteria checklist
   [ ] Age ≥40 and <75? YES / NO
   [ ] eGFR ≥60? YES / NO (not "close to 60")
   [ ] BP 140-180? YES / NO
   
2. Pharmacist review before enrollment
   - Independent verification of all labs against I/E
   - Double-check numeric thresholds
   
3. Weekly audit
   - Site coordinator audits first screening and enrollment of week
   - Confirms all I/E met
   - Reports to investigator
   
4. Monthly QA meeting
   - Track any I/E deviations
   - Discuss during team meeting
   - Reinforce importance

DOCUMENTATION & TIMELINE

CAPA documentation includes:
- Written by: Site Coordinator
- Approved by: Principal Investigator
- Reviewed by: Sponsor Clinical Monitor
- Completion date: All actions by [specific date, e.g., 2/28/2024]

Follow-up verification:
- Site provides evidence of completion (training records, audit results)
- Sponsor verifies measures taken
- Next monitoring visit confirms implementation

Subject Continuation After Deviations/Violations

When a deviation or violation occurs with an enrolled subject, make a determination:

STEP 1: SAFETY ASSESSMENT

Question: Is subject at risk if continuation?

Example 1: Visit 3 days late
- Impact: Minor. Subject still monitored.
- Decision: Can continue

Example 2: Enrolled with low eGFR, renally-metabolized drug
- Impact: High. Drug clearance reduced. Toxicity risk.
- Decision: Assess continuation. Enhanced monitoring OR withdrawal

Example 3: Subject had prohibited medication for 2 days
- Impact: Depends. Drug-drug interaction possible?
- If yes: Monitor closely OR discontinue
- If no: Can continue with enhanced monitoring

STEP 2: DATA INTEGRITY ASSESSMENT

Question: Is subject's data valid?

Example 1: Missed one of six secondary assessments
- Impact: 5 of 6 data points. Partial data.
- Decision: Data partially usable. Note limitation in analysis.

Example 2: Primary endpoint measurement outside timeframe
- Impact: Measurement timing critical. Data suspect.
- Decision: Data invalid for primary analysis (exclude from analysis)

Example 3: Subject received prohibited drug affecting pharmacokinetics
- Impact: Confounds efficacy assessment.
- Decision: Data unreliable. Exclude from analysis.

STEP 3: INFORMED CONSENT ASSESSMENT

Question: Did subject have adequate information to consent?

Example 1: Protocol excluded subjects with condition X, subject has condition X
- Impact: Subject didn't know their condition was a risk.
- Decision: Offer re-consent or withdrawal

Example 2: New safety information emerges mid-study
- Impact: Subjects consented without current risk information.
- Decision: May require re-consent form distribution

Example 3: Subject doesn't understand monitoring requirement
- Impact: Informed consent compromised.
- Decision: Offer additional counseling or withdrawal

FINAL DECISION: CONTINUE VS. WITHDRAW

Decision Matrix:

Safety Risk | Data Valid | IC Adequate | Decision
Minimal     | Yes        | Yes         | CONTINUE
Minimal     | Partial    | Yes         | CONTINUE (note limitation)
Minimal     | No         | Yes         | CONTINUE (exclude from analysis)
High        | Yes        | Yes         | ASSESS (may discontinue)
High        | No         | No          | DISCONTINUE (multiple issues)
Any         | Any        | No          | OFFER WITHDRAWAL (IC issue)

Documentation: All continuation/withdrawal decisions documented in subject's study record with:


SECTION 6: IRT/IWRS/RTSM SYSTEMS

System Overview

IRT = Interactive Response Technology (comprehensive randomization + drug management system)

IWRS = Interactive Voice Response System (phone-based version of IRT)

RTSM = Randomization and Trial Supply Management (broader term for system)

Key Purpose: Automate critical functions to PREVENT deviations while maintaining blinding and ensuring drug accountability


IRT Functions: How Systems Prevent Deviations

FUNCTION 1: RANDOMIZATION & ALLOCATION CONCEALMENT

What it does:

How it prevents deviation:

Example:

Without IRT:
Investigator knows from experience: "Subjects on Drug A get better outcomes"
When screening eligible subjects, investigator unconsciously enrolls those they "think" will benefit
Result: Allocation bias (systematically enrolling particular type)

With IRT:
Investigator can't predict next assignment
Enrollment follows truly random sequence
Result: Unbiased allocation (protective of study validity)

FUNCTION 2: ENROLLMENT VERIFICATION (I/E Screening)

What it does:

How it prevents deviation:

Example:

Protocol: eGFR ≥60 required
Subject's eGFR: 42

Without IRT (paper form):
Staff fills out screening form, might miss the low eGFR
Subject enrolled despite exclusion → DEVIATION

With IRT:
System asks: "Is eGFR ≥60?" 
Correct answer based on subject's lab: "NO"
IRT blocks enrollment → Can't randomize
Prevention: No enrollment of ineligible subject

What Happens if I/E Not Met:

FUNCTION 3: DRUG SUPPLY MANAGEMENT

What it does:

How it prevents deviation:

Example:

Manual drug accountability:
- Pharmacy staff maintain hand-written log
- Monthly count done by one person
- Easy to miss entries or transposition errors
- Discrepancies found months later (hard to investigate)

IRT drug tracking:
- Each drug unit scanned on receipt
- Dispensing entered with subject ID
- Returns scanned on collection
- System automatically reconciles daily
- Discrepancies identified immediately (easier to investigate)

FUNCTION 4: EMERGENCY UNBLINDING

What it does:

How it prevents deviation:

Emergency Unblinding Criteria:

ACCEPTABLE reasons for emergency unblinding:
✓ Severe anaphylaxis (treatment affects allergy management)
✓ Severe bleeding requiring antidote (need to know if on anticoagulant)
✓ Cardiac emergency where treatment matters (arrhythmia, need ECG interpretation)
✓ Pregnancy (need to know teratogenic risk)

NOT acceptable reasons:
✗ Subject curiosity
✗ Staff wanting to confirm "which treatment" a subject got
✗ Subject's family asking
✗ Administrative convenience
✗ Suspected benefit (want to confirm they got active)

Blinding: Levels & IRT Protection

SINGLE-BLIND TRIALS

Definition: Subject doesn't know treatment assignment; investigator/staff knows

Why Used:

IRT Role:

DOUBLE-BLIND TRIALS (Most Common)

Definition: Neither subject nor investigator knows treatment assignment

Why Used:

IRT Role:

Example:

IRT randomizes subject: "Assigned to Arm 1"

If Investigator could see: "Arm 1 = Active Drug (50mg daily)"
Then investigator is UNBLINDED (they know what drug subject got)

Proper IRT setup:
Investigator sees: "Assigned to Arm 1"
Staff dispensing knows: "Arm 1 corresponds to Drug Code H5P2"
Mapping stays hidden until database lock
Result: True double-blind maintained

TRIPLE-BLIND TRIALS (Rare)

Definition: Subject, investigator, AND sponsor don't know assignments until analysis

Why Used:

IRT Role:


IRT Audit Trail & Regulatory Compliance

What IRT Records (Permanent Documentation)

RANDOMIZATION EVENTS:
- Date and time of randomization
- Subject ID
- Treatment assignment code
- User ID who accessed system
- IP address / system access log

ENROLLMENT SCREENING:
- Subject demographic data entered
- Each I/E criterion answer (YES/NO)
- Date and time of each I/E check
- Which criteria failed (if applicable)
- Whether enrollment proceeds

DRUG DISPENSING:
- Date and time dispensing recorded
- Subject ID receiving drug
- Quantity dispensed
- Lot number and expiration
- User ID entering dispensing record
- Investigator/pharmacy signature timestamp

UNBLINDING EVENTS (if any):
- Date and time of unblinding request
- Subject ID
- Reason for unblinding
- Who approved unblinding
- Timestamp of approval
- Treatment code provided

EXCEPTIONS & OVERRIDES:
- Any manual changes to automated system
- Reason for override
- Who approved
- Timestamp of approval

Why This Audit Trail Matters

FDA Regulatory Requirement:

IRT provides all of this automatically.

Comparison:

Manual Record-Keeping (Paper):
"Subject 101 dispensed 30 capsules"
Inspector asks: "When exactly?" → "Sometime in February, I think"
"Who recorded this?" → "Our old coordinator, she left last year"
"Can you verify?" → "The log is in a box somewhere..."
Result: Incomplete audit trail, regulatory risk

IRT Record-Keeping:
"2024-02-15 10:23:47 - User ID: CC_JSmith - Subject 101 dispensed 30 capsules Lot XYZ, Exp 06/2024"
Inspector asks: "When?" → "February 15, 2024 at 10:23:47"
"Who recorded?" → "Jane Smith, Site Coordinator"
"Can you verify?" → "Here's the system export with timestamps"
Result: Complete audit trail, regulatory confidence

IWRS vs. Web-Based IRT Systems

IWRS (Interactive Voice Response System)

Technology:

Advantages:

Process Example:

Investigator calls IWRS number
"Welcome to Study X IRT. Please enter site number..."
Investigator: "0152"
"Thank you. Confirming subject screening data. Is subject age 40-75?"
Investigator: "Yes, press 1"
"Subject inclusion confirmed. Randomization complete. Your assignment is Arm 2. Record code H7K9."
Investigator records code, subject dispensed coded study drug

Limitations:

WEB-BASED IRT (Modern Standard)

Technology:

Advantages:

Process Example:

Investigator logs into IRT portal
Enters Subject ID: 101
Clicks "Screen Subject"
System displays screening questions
- Age: "Yes, 52"
- eGFR: "Yes, 68"
- Hypertension: "Yes, diagnosed 2018"
System displays: "Subject meets inclusion criteria"
Clicks "Randomize"
System shows: "Subject 101 randomized to Arm 1, Drug Code HX9L"
Dashboard shows: Randomized subjects: 34/100, Drug lot inventory: 67/100
Sponsor portal shows: Real-time enrollment across all sites

Regulatory Advantage: Most modern trials use web-based for superior audit trail and real-time monitoring


IRT System Failures & Contingencies

What if IRT System Fails?

EXAMPLE: Server down, can't access randomization system

Protocol typically includes:
"In case of system failure, randomization codes may be obtained from sponsor 
emergency contact by phone. System access restored within 24 hours."

Contingency process:
- Investigator calls sponsor emergency line
- Provides subject screening data by phone
- Sponsor manually verifies I/E from protocol
- Provides randomization code by phone
- Recorded as "Emergency randomization"
- IRT record created when system comes back online
- Audit trail shows deviation (emergency randomization) and reason

SECTION 7: DRUG ACCOUNTABILITY & CHAIN OF CUSTODY

Regulatory Framework

Legal Basis:

Fundamental Principle: Every dose of investigational product must be accounted for from manufacture to destruction.


Complete Drug Accountability Cycle

┌─────────────────────────────────┐
│ MANUFACTURE & QUALITY CONTROL   │
│ Drug made, tested, approved     │
└──────────────┬──────────────────┘
               │
               ↓
┌──────────────────────────────────┐
│ SHIPMENT TO SITE                 │
│ Drug sent with tracking          │
└──────────────┬───────────────────┘
               │
               ↓
┌──────────────────────────────────┐
│ RECEIPT AT SITE                  │
│ Inventory recorded, condition    │
└──────────────┬───────────────────┘
               │
               ↓
┌──────────────────────────────────┐
│ STORAGE & TEMPERATURE MONITORING │
│ Maintained at protocol conditions│
└──────────────┬───────────────────┘
               │
               ↓
┌──────────────────────────────────┐
│ DISPENSING TO SUBJECTS           │
│ Subject ID, dose, lot recorded   │
└──────────────┬───────────────────┘
               │
               ↓
┌──────────────────────────────────┐
│ SUBJECT USE OR RETURN            │
│ Subject takes home OR returns    │
└──────────────┬───────────────────┘
               │
               ↓
       ┌───────┴────────┐
       ↓                ↓
┌─────────────┐  ┌──────────────┐
│ DESTRUCTION │  │ RETURN TO    │
│ (expired)   │  │ MANUFACTURER │
└─────────────┘  └──────────────┘
       │                │
       └────────┬───────┘
                ↓
        FINAL ACCOUNTING

Drug Accountability Reconciliation Formula

Basic Formula:

Beginning Inventory + Receipts - Dispensed - Returned - Destroyed = Ending Inventory

Must Match Physical Count:

Expected Ending Inventory (calculated) = Physical Count (actual)
If NOT equal → Discrepancy requires investigation

Reconciliation Examples

EXAMPLE 1: SIMPLE SINGLE SHIPMENT

Received: 100 capsules on Day 1
Dispensed: 70 capsules (14 subjects × 5 capsules each)
Returned by subjects: 12 capsules (unused)
Destroyed/expired: 0 capsules

Calculation:
100 + 0 - 70 - 12 - 0 = 18 capsules expected

Physical count: 18 capsules
Status: BALANCED ✓

All drug accounted for:
- 70 capsules taken by subjects (in use)
- 12 capsules returned (available for reuse or destruction)
- 18 capsules in stock
- Total: 70 + 12 + 18 = 100 ✓

EXAMPLE 2: MULTIPLE SHIPMENTS

Beginning inventory: 50 capsules

Shipment 1 (Jan 15): 100 capsules received ✓
Shipment 2 (Feb 10): 75 capsules received ✓

Dispensed: 140 capsules
Returned: 20 capsules
Destroyed (expired): 8 capsules

Calculation:
50 + 100 + 75 - 140 - 20 - 8 = 57 capsules expected

Physical count: 57 capsules
Status: BALANCED ✓

Interpretation:
- 140 capsules given to subjects (in use/consumed)
- 20 capsules returned (available)
- 8 capsules destroyed (expired, disposed)
- 57 capsules in stock
- Total: 140 + 20 + 8 + 57 = 225
- Receipts: 50 + 100 + 75 = 225 ✓

EXAMPLE 3: DISCREPANCY INVESTIGATION

Beginning: 50 capsules
Received: 100 capsules
Dispensed: 60 capsules
Returned: 15 capsules
Destroyed: 5 capsules

Expected: 50 + 100 - 60 - 15 - 5 = 70 capsules
Physical count: 67 capsules

DISCREPANCY: Missing 3 capsules

Investigation Steps:
1. Recount inventory → Still 67 capsules ✓
2. Review dispensing log:
   - Subject 101: 5 capsules ✓
   - Subject 102: 5 capsules ✓
   - Subject 103: 5 capsules (only 4 recorded!) ← Found 1
3. Review return log:
   - Subject 104: 8 returned ✓
   - Subject 105: 7 returned ✓
   - Total: 15 returned ✓
4. Review destruction records:
   - Expired: 5 capsules ✓
5. Interview staff:
   - "Two capsules spilled when opening bottle during inventory"
   - Spillage: 2 capsules

Final accounting:
- Lost dispensing record: +1 capsule
- Spillage (unwitnessed): 2 capsules
- Account for: 1 + 2 = 3 capsules ✓

Status: DISCREPANCY RESOLVED
Documentation: 
"Week 4 inventory count revealed 3-capsule discrepancy. Investigation identified 
1 capsule unrecorded dispensing (added to Subject 103 record) and 2 capsules 
spillage (documented in physical inventory form). All drug now accounted for."

Temperature Excursions & Drug Stability

Definition: Drug stored outside protocol temperature specifications

Protocol Example:

"Investigational Product Storage: 15-25°C (59-77°F), protect from light"

Excursion Example:

Temperature monitoring log shows:
- Normal range: 15-25°C maintained
- Day 7: Freezer malfunction
- Temperature: Dropped to -5°C for 8 hours
- Recovery: Technician corrected, returned to 18°C

Status: TEMPERATURE EXCURSION

Management Process:

STEP 1: DISCOVERY & DOCUMENTATION
- Identify excursion from temperature logs
- Document: Date, duration, temperatures, affected product lots

STEP 2: NOTIFICATION
- Immediately notify sponsor
- Provide: Temperature readings, lot numbers, quantity affected, timeline

STEP 3: SPONSOR ASSESSMENT
- Sponsor reviews drug stability data
- Questions: Can drug be exposed to -5°C and remain stable?
- If answer is NO → destroy product
- If answer is YES with monitoring → allow use with caution

STEP 4: DECISION & ACTION
Case A: Drug stable at -5°C
- Allow continued use with enhanced monitoring
- Subjects receiving affected lots monitored closely for efficacy/safety changes

Case B: Drug not stable at -5°C
- Withdraw affected product
- Destroy on-site (witnessed, documented)
- Return to manufacturer (if fewer than dispensed)
- Continue study with new lot

STEP 5: RECONCILIATION ADJUSTMENT
- Excursion-destroyed product = reduces inventory
- Formula: Beginning + Receipts - Dispensed - Returned - Destroyed (excursion) = Ending
- Example: If 50 capsules destroyed due to excursion:
  100 received - 60 dispensed - 50 destroyed (excursion) = -10 ???
  Problem: This would show negative balance!
  Reality: Destroyed capsules came from beginning inventory or returns
  Adjustment: 100 received - 60 dispensed - 50 destroyed = -10
  Actually: 50 (beginning) + 100 (received) - 60 (dispensed) - 50 (destroyed) = 40

Regulatory Consequence:


Return & Destruction Documentation

Critical Point: Every dose of investigational product is regulated. Return or destruction must be thoroughly documented.

RETURN OF DRUG TO MANUFACTURER

Trigger Examples:

Documentation Requirements:

DRUG RETURN LOG ENTRY:

Subject ID: 101
Visit: Week 4 (completion visit)
Quantity Returned: 8 capsules (of 10 dispensed, subject retained 2 unused)
Lot Number: ABC-12345-001
Expiration Date: 06/2024
Condition: Good condition, no visible damage
Returned by: Sarah Jones, Study Coordinator (Signature)
Witnessed by: Robert Smith, Investigator (Signature)
Date: March 15, 2024
Time: 2:45 PM

Return to Manufacturer:
Packed in: Cold pack (maintained 15-25°C during shipment)
Shipping method: FedEx Express overnight
Tracking #: 123456789
Shipped: March 16, 2024 at 4:00 PM
Received by manufacturer: March 17, 2024 at 8:30 AM
Manufacturer receipt signature: Pharmacy Manager, ABC Pharma
Return documented: Completion of accountability cycle

DESTRUCTION AT SITE

Trigger Examples:

Documentation Requirements:

DESTRUCTION LOG ENTRY:

Drug Destroyed: Study X Investigational Product (Lot DEF-54321-002)
Quantity: 30 capsules
Expiration Date: 12/2023
Reason for Destruction: Expired—study completion on 11/30/2023, inventory not used
Date of Destruction: December 4, 2023
Time: 1:30 PM
Method: Incineration (controlled waste incineration facility, Phoenix Waste Co.)
Facility confirmation #: PW-2023-4567

Destruction Witnessed By:
- Principal Investigator: Dr. Michael Johnson (Signature)
- Study Coordinator: Jennifer Lee (Signature)
- Facility Technician: Tom Brown, Phoenix Waste (Signature)

Facility Certificate of Destruction: Attached
- Confirms drug weight before incineration
- Confirms incineration at safe temperature (>1000°C)
- Confirms complete destruction
- Facility license/accreditation verified

Documentation Retained: Original destruction form + facility certificate
Location: Study file, Drug Accountability Section

Drug Accountability Audit Trail

FDA Inspectors Will Verify:

Question 1: "Show me all drug receipts"
→ Shipment records with dates, quantities, shipper documentation

Question 2: "How was this drug dispensed?"
→ Dispensing records showing subject ID, date, quantity, lot, signature

Question 3: "Where is the drug that wasn't dispensed?"
→ Inventory count records with dates and signatures

Question 4: "What happened to product at the end?"
→ Return/destruction documentation with witnesses and facility confirmation

Question 5: "Do your records match your physical inventory?"
→ Reconciliation showing: Beginning + Receipts - Dispensed - Destroyed = Ending
→ Physical count matching expected count

If Records Are Incomplete:


Drug Accountability Calculation Practice

Formula Reminder:

Beginning Inventory + Receipts - Dispensed - Returned - Destroyed = Ending

Practice Problem 1:

Started study with: 75 capsules
Received shipment: 100 capsules
Dispensed to subjects: 95 capsules
Subjects returned: 18 capsules
Destroyed (expired): 7 capsules

Expected ending: ? 
Physical count: 48 capsules
Is it balanced?

ANSWER:
75 + 100 - 95 - 18 - 7 = 55 expected
Physical count = 48
DISCREPANCY: 7 capsules missing
Not balanced - requires investigation

Practice Problem 2:

Starting inventory: 50 capsules
Two shipments received:
  - Shipment 1: 125 capsules
  - Shipment 2: 75 capsules
Dispensed: 180 capsules
Returned: 22 capsules
Destroyed: 12 capsules

Expected ending: ?
Physical count: ?
(Assume balanced—what should count be?)

ANSWER:
50 + 125 + 75 - 180 - 22 - 12 = 36 capsules expected
Physical count should be: 36 capsules (balanced)

SECTION 8: INTERACTIVE CASE STUDIES

Case Study 1: Subject Eligibility Challenge

Protocol Snapshot:

Scenario:

WEEK 1: Subject screening
- Age: 52 years ✓
- BP: 145/92 ✓
- Labs (obtained 3 days ago):
  • eGFR: 42 mL/min/1.73m² ← KEY FINDING
  • Creatinine: 1.7 mg/dL
  • Sodium, potassium: Normal

Site coordinator notes: "eGFR is 42, protocol excludes <45. That's only 3 points different. Could we enroll anyway? Patient really wants to participate."

Investigator response: "We can probably interpret it flexibly. He's clinically stable."

Decision: ENROLL SUBJECT
Subject ID: 101
Randomized: Arm 1 (receives treatment)
Dispensed: 5 capsules per visit
Completion: Subjects visits Week 1, 2, 3

WEEK 3: Monitoring visit
Monitor audits enrollment documentation
Finds: Subject 101 has eGFR 42 at enrollment
Alert: "Subject does not meet inclusion criteria—eGFR 42 <45 exclusion"

YOUR ANALYSIS:

Question 1: Is this a deviation or violation?

ANSWER: VIOLATION (at minimum MAJOR DEVIATION)

Why?
- eGFR <45 was explicitly excluded (protocol clearly states "eGFR <45")
- Subject's eGFR: 42 (below 45)
- Subject enrolled despite exclusion = deviation
- Safety implication: eGFR 42 indicates reduced kidney function
- Study drug likely renally metabolized (most drugs are)
- If renally metabolized: Reduced clearance → higher drug levels → toxicity risk
- Investigator and staff enrolled subject knowing (or should have known) they didn't meet criterion
- This is VIOLATION of informed consent + safety + data integrity

Question 2: What actions must be taken?

ACTIONS:

Immediate (within 24 hours):
1. Notify sponsor clinical monitor
2. Sponsor determines: Is subject at safety risk?
   - Is study drug renally metabolized? YES (most likely)
   - Is eGFR 42 high-risk for toxicity? YES (reduced clearance)
   - Recommendation: Assess subject for toxicity, consider withdrawal

Site Actions:
3. Assess subject for adverse events
   - Any signs of drug toxicity? (Depends on drug class)
   - If hypertension drug: Signs of excessive BP lowering? Renal dysfunction progression?
4. Draw baseline safety labs if not recently done
5. Document assessment in subject's study record

Reporting:
6. Prepare violation report documenting:
   - Who enrolled subject (investigator/coordinator)
   - When discovered (Week 3)
   - Why enrolled despite exclusion (investigator overrode criterion)
   - Assessment of safety risk
7. Submit to IRB within 24 hours (safety violation)
8. Sponsor considers FDA reporting

CAPA (Corrective Action Plan):
9. Root cause: Why did this happen?
   → Site staff didn't understand numeric thresholds
   → Thought "eGFR 42 is close to 45, probably okay"
10. Corrective action:
   → Retraining on I/E criteria
   → Specific emphasis: "42 is LESS than 45. Not eligible."
   → Quiz staff to verify understanding
11. Preventive action:
   → Implement I/E checklist with numeric values
   → Pharmacist independent review of inclusion criteria before randomization
   → Weekly audit of screening/enrollment

Question 3: Can the subject continue in the study?

DECISION: Likely WITHDRAWAL (but depends on assessment)

Safety assessment:
- eGFR 42 = moderate-to-severe renal impairment
- If drug renally metabolized: Toxicity risk confirmed
- If subject showing any toxicity signs: Withdraw to protect safety
- If no toxicity yet: Could continue with enhanced monitoring + frequent renal assessments
  (But withdrawal is safer/more appropriate)

Data integrity assessment:
- Subject's data may not represent intended population
- eGFR 42 subjects weren't studied (excluded by design)
- Data from this subject may be excluded from primary efficacy analysis

Informed consent assessment:
- Subject didn't know they had exclusionary condition (eGFR 42)
- Subject didn't know their reduced kidney function was a risk
- Offer re-consent OR withdrawal

Most likely: WITHDRAWAL
- Protects subject safety (reduces toxicity risk)
- Preserves study integrity (removes non-qualifying subject)
- Documents violation appropriately

Documentation:
"Subject 101 withdrawn due to baseline eGFR 42 (exclusion criterion: eGFR <45). 
Violation identified at Week 3 monitoring visit. Subject enrolled despite not meeting inclusion criteria. 
Kidney function compromised; drug clearance likely reduced. Risk of toxicity. 
Subject offered re-enrollment if eGFR improves to ≥60 in future. 
Final safety assessment: No abnormal labs at withdrawal."

Case Study 2: Drug Accountability Reconciliation

Study Context:

Scenario:

INVENTORY ACCOUNTING:

Jan 1: Received 100 capsules
  Lot: ABC-12345-001
  Exp: 09/2024
  Condition: Good ✓
  Stored: Room temperature ✓

Jan 1 - March 25: Dispensing ongoing
Jan 30: Week 1 checkpoint - received 25 capsules = count 75 remaining ✓
Feb 15: Week 4 checkpoint - received 32 capsules = count 43 remaining ✓
Mar 15: Week 8 checkpoint - received 28 capsules = count 15 remaining ✓
Mar 26: Study closeout - total dispensed tally = 95 capsules

Subject Returns (during study):
- Subject 101: 3 unused capsules (returned Jan 15)
- Subject 102: 2 unused capsules (returned Feb 5)
- Subject 103: 0 returned
- Subject 104: 1 unused capsule (returned Feb 28)
Total returned: 6 capsules

Destroyed:
- During study: None (no expiration yet)
- At closeout: Remaining inventory (to be destroyed per protocol)

WEEK 12 (STUDY CLOSEOUT):
Calculation of expected ending inventory:
Beginning: 100 capsules
Received: 0 (only one shipment)
Dispensed: 95 capsules
Returned: 6 capsules
Destroyed: 0 (to be calculated)

Expected: 100 + 0 - 95 - 6 - 0 = -1 capsule ???
Problem: This shows NEGATIVE inventory!

Investigation:
1. Recount physical inventory: 5 capsules
   (Not -1, not 0, but 5 remaining)

2. Review dispensing records:
   - Carefully audit who received what
   - Subject 101: 5 dispensed ✓
   - Subject 102: 5 dispensed ✓
   - Subject 103: 5 dispensed ✓
   - Subject 104: 5 dispensed ✓
   - Subject 105: 5 dispensed ✓
   - ... (continue for all 19 subjects)
   - Total: 95 capsules ✓

3. Review return records:
   - Subject 101: 3 returned ✓
   - Subject 102: 2 returned ✓
   - Subject 104: 1 returned ✓
   - Total returned: 6 capsules ✓

4. Interview staff:
   "During Week 2 recount, one capsule fell on floor. We discarded it because it couldn't be used."

Expected adjusted: 100 + 0 - 95 - 6 - 1 (destroyed accidental) = -2 ???
Still doesn't match!

DEEPER INVESTIGATION:
5. Review original receipt:
   "Received 100 capsules"
   Recount at receipt: 100 ✓

6. Review shipping documentation:
   "Shipped 101 capsules" ← FOUND IT!
   Receiving team only recorded 100
   One capsule missing from receipt count

CORRECTED ACCOUNTING:
Actually received: 100 (what was counted and signed for)
OR: Received 101 but only 100 recorded?

Ask sponsor: "Did you ship 100 or 101?"
Sponsor confirms: "Shipped 100. Lab had counting error (reported 101 but was 100)."

FINAL RECONCILIATION:
Beginning: 100
Received: 0
Dispensed: 95
Returned: 6
Destroyed (accidental, dropped): 1
Missing (unaccounted): 0

Calculation: 100 - 95 - 6 - 1 = -2 (still wrong!)

Actually:
Dispensed: 95 capsules (in subjects or consumed)
Returned: 6 capsules (usable)
Destroyed: 1 capsule (accidental, documented)
Remaining inventory: Should be 100 - 95 = 5 PLUS the 6 returned = 11 total available?
NO—returned capsules are SEPARATE from dispensed
Think of it differently:
- 95 went to subjects
- 6 of those were returned (so really 89 consumed, 6 available)
- 1 destroyed (accident)
- Stock: 5 remaining

Total accounting: 89 + 6 + 1 + 5 = 101?
Wait, we only had 100...

CORRECT ACCOUNTING LOGIC:
"Dispensed" means "given to subjects" (whether consumed or returned)
So: 95 capsules given to subjects
Of those 95: 89 consumed, 6 returned
Separate from those 95: 1 destroyed accidental, 5 in stock

Wait, 6 returned + 5 remaining = 11, but 100 - 95 = 5?
The returned capsules CAME FROM the dispensed 95!

CORRECT FORMULA:
Beginning: 100
Received: 0
Consumed by subjects: 89 (95 dispensed - 6 returned)
In stock (not yet destroyed): 5
Destroyed: 6 (1 accidental + 5 end of study destruction)

Calculation: 100 - 89 - 1 (accidental) - 5 (destroyed at closeout) = 5 in stock ✓

Actually, using the formula correctly:
100 + 0 - 95 (dispensed total) - 6 (returned) - 1 (destroyed accidental) + 6 (returned back to inventory) = ? 
This is confusing because returned capsules are complicated

CLEAREST WAY:
Received: 100
Dispensed: 95 (including the 6 that were returned)
- Of the 95: 89 ultimately consumed, 6 returned
Destroyed: 1 (accidental drop)
Remaining: 5

Reconciliation: 100 - 89 (consumed) - 1 (accidental) - 5 (destroyed at closeout) = 5 remaining ✓

Wait, that's circular (5 - 5 = 0).

SIMPLEST:
All 100 capsules accounted for as:
- 89 used by subjects (consumed)
- 6 returned by subjects (available for destruction)
- 1 destroyed accidentally
- 4 remaining in stock at closeout?

Actually, if 6 returned + 1 destroyed + remaining = what wasn't dispensed
100 total - 95 dispensed = 5 not dispensed
Of those 5: 1 destroyed accidental, 4 remaining
Plus: 6 returned (which came from the 95)

Total: 89 consumed + 6 returned + 1 destroyed + 4 remaining = 100 ✓
But wait, the 6 returned are part of the 95 dispensed

CORRECT FINAL ACCOUNTING:
100 capsules received
95 capsules dispensed to subjects
  - 89 consumed by subjects
  - 6 returned by subjects (still available)
5 capsules never dispensed
  - 1 destroyed accidentally
  - 4 remaining in stock

Total: 89 + 6 + 1 + 4 = 100 ✓

At closeout: 
6 returned capsules: destroyed
4 remaining capsules: destroyed
Final total destroyed: 10 capsules + 1 accidental = 11? No...

Actually: 6 + 4 = 10, plus 1 accidental = 11, minus...
This is getting circular.

CLEAREST FORMULA APPLICATION:
Beginning: 100
Receipts: 0
Dispensed: 95 [This includes everything given out, even returns]
Returned: 6 [Subtracted from dispensed]
Destroyed at closeout: 4 [From the never-dispensed inventory]
Destroyed accidental: 1 [From the never-dispensed inventory]

Calculation: 100 + 0 - 95 - 6 - 4 - 1 = -6 ???

Hmm, the issue is that "returned" shouldn't be subtracted separately if using "dispensed"...

CORRECT APPLICATION:
Beginning Inventory + Receipts - Dispensed - [Destroyed] = Ending

Where:
"Dispensed" = ultimately left site in subject hands (not returned)
"Returned" = came back and is either in stock or destroyed
"Destroyed" = all destruction (accidental + closeout)
"Ending" = inventory at end of study

So:
Beginning: 100
Receipts: 0
Dispensed (consumed): 89
Destroyed (accidental + returned + stock): 1 + 10 = 11

Check: 100 - 89 - 11 = 0 ✓

Total destroyed at closeout: 
- 1 accidental
- 6 returned
- 4 remaining
= 11 total destroyed

Status: BALANCED ✓

Your Analysis:

Question 1: Is the study balanced?

ANSWER: YES, balanced

Reconciliation: 100 - 89 - 11 = 0 (all accounted for)

Detailed accounting:
- 89 capsules consumed by subjects (net)
- 11 capsules destroyed/disposed
  - 1 accidental (documented)
  - 6 from subject returns
  - 4 from stock

Question 2: What must be documented?

DOCUMENTATION REQUIRED:

1. Accidental Destruction (1 capsule):
   Date: Week 2 (specific date needed)
   Circumstance: Fell on floor during inventory count
   Action taken: Discarded (could not be used per protocol)
   Documented by: [Staff name]
   Witnessed by: [Second staff member]

2. Return from Subjects (6 capsules):
   - Subject 101: 3 capsules returned Jan 15 (reason: unused)
   - Subject 102: 2 capsules returned Feb 5 (reason: end of visit)
   - Subject 104: 1 capsule returned Feb 28 (reason: subject preference)
   Each return: Date, subject ID, quantity, condition, signed

3. Closeout Destruction (10 capsules):
   All unused inventory destroyed at study closeout
   Date: March 31, 2024
   Method: Incineration (Phoenix Waste Co. facility)
   Witnessed by: Site Coordinator + Investigator + Facility tech
   Facility destruction certificate: Attached

4. Final Reconciliation:
   Beginning: 100
   Dispensed: 89 (consumed)
   Destroyed/Returned: 11 (accidental 1 + returns 6 + stock 4)
   Balance: 0 ✓

Question 3: Any issues or deviations?

FINDINGS:

Minor issue: 1 capsule dropped and destroyed without witness documentation
Status: MINOR DEVIATION
  - One capsule unaccounted in formal destruction log
  - But documented in site coordinator's notes
  - Sponsor informed and accepted

Action taken:
- Documented in drug accountability record
- Noted in closeout report
- No regulatory issue (minor loss with explanation)

Recommendation:
- Implement procedure: All accidents/spills must have witness signature
- Document in destruction log, not just notes
- Include reason in log: "Accidental loss during recount"

Final Status: RECONCILIATION BALANCED with minor documentation gap noted

SECTION 9: GLOSSARY & KEY TERMINOLOGY

ADMINISTRATIVE CHANGE: Documentation/contact/format update not affecting study conduct

AMENDMENT: Change to scientific/regulatory content of protocol requiring approval

BASELINE: Initial assessment value used for comparison

BLINDING: Keeping treatment assignment hidden from subjects/investigators/sponsor

CASE REPORT FORM (CRF): Document for collecting study data from each subject

CHAIN OF CUSTODY: Documentation of product location/handler throughout study

CLINICAL HOLD: FDA action stopping study due to safety/integrity concerns

CORRECTIVE ACTION PLAN (CAPA): Systematic response to deviation/violation

DEVIATION: Any noncompliance with protocol

DEVIATION REPORT: Formal documentation of deviation incident

DOUBLE-BLIND: Neither subject nor investigator knows treatment assignment

EFFICACY: Ability of treatment to produce intended effect

EMERGENCY UNBLINDING: Revealing treatment code when medically necessary

ENDPOINT: Specific measurement defining study success

EXCLUSION CRITERIA: Characteristics preventing subject enrollment

EXPOSURE-RESPONSE RELATIONSHIP: How drug levels relate to effects

FDA (Food and Drug Administration): U.S. regulatory body

GCP (Good Clinical Practice): International standards for clinical research

HALF-LIFE: Time for drug concentration to reduce by 50%

ICH (International Council for Harmonisation): Global standards organization

ICH E6(R2): Guideline defining clinical trial protocol requirements

IND (Investigational New Drug): FDA application for study authorization

INFORMED CONSENT: Subject's voluntary agreement to participate with full information

INFORMED CONSENT FORM (ICF): Document describing study and obtaining consent

INCLUSION CRITERIA: Characteristics allowing subject enrollment

IRT (Interactive Response Technology): System managing randomization/supply

IWRS (Interactive Voice Response System): Phone-based randomization system

MEDICAL NECESSITY: Deviation justified by subject safety/health emergency

MINOR DEVIATION: Non-compliance unlikely to affect safety/data integrity

MAJOR DEVIATION: Non-compliance potentially affecting safety/data

MONITOR: Sponsor representative overseeing study conduct

MONITORING VISIT: On-site assessment of protocol compliance

NONCLINICAL STUDIES: Laboratory/animal studies preceding human trials

OBJECTIVE: Research question the study answers

OUTCOME: Result or effect being studied

PHARMACOKINETICS: How body processes drug (absorption, distribution, metabolism, excretion)

PHARMACODYNAMICS: How drug affects body (mechanism, effects)

PHASE 1: First human study (small, safety-focused)

PHASE 2: Early efficacy testing (moderate subjects, disease-specific)

PHASE 3: Efficacy confirmation (large, randomized, controlled)

PHASE 4: Post-marketing surveillance

POPULATION: Group of subjects with defined characteristics

PROTOCOL: Detailed study plan describing objectives, design, procedures

PROTOCOL DEVIATION: Unplanned non-compliance with protocol

PROTOCOL VIOLATION: Serious deviation affecting safety/integrity/consent

RANDOMIZATION: Random assignment to treatment groups

REGULATORY: Relating to government approval/oversight

RTSM (Randomization & Trial Supply Management): System managing randomization and drug

SAFETY: Absence of harmful effects; monitoring for adverse events

SAMPLE SIZE: Number of subjects needed for statistical power

SCREENING: Initial assessment determining if subject meets criteria

SCREENING FAILURE: Subject not meeting inclusion/exclusion criteria

SECONDARY ENDPOINT: Supportive measurement of study question

SINGLE-BLIND: Subject doesn't know treatment; investigator does

SOCRA (Society of Clinical Research Associates): Professional organization

SPONSOR: Organization funding/responsible for study

STABILITY: Drug remaining effective at specified conditions

STATISTICAL ANALYSIS PLAN: Pre-specified analysis methodology

SUBJECT: Human participant in study

TRIPLE-BLIND: Subject, investigator, sponsor unblinded

VIOLATION: Serious deviation compromising safety/integrity/consent

WASH OUT: Period allowing drug elimination before next treatment

WITHDRAWAL: Subject discontinuing study participation


SECTION 10: EXAM PREPARATION STRATEGY

Week 8 Study Plan

Time Allocation (10 hours total):


Memory Aids & Mnemonics

14 Protocol Sections (6.1-6.14): Remember: "WHO does WHAT and WHY" + "HOW" + "DOCUMENTATION"

Amendment vs. Administrative: "When it AFFECTS the study → Amendment. When it's just PAPERWORK → Administrative."

Deviation vs. Violation: "ALL violations are deviations. Some deviations become violations when SAFETY/DATA/CONSENT is compromised."

I/E Application: "Read the NUMBER. Use the NUMBER. Don't interpret the NUMBER."

Drug Reconciliation: "Beginning + In - Out = Ending"


Exam Question Types & Strategies

Question Type 1: "What's Missing?"

Question: "A protocol is missing... [description]. Which section is incomplete?"
Strategy: Match the content to the section (6.1-6.14)
Example answers:
- Missing primary endpoint → 6.3 (Objectives)
- Missing inclusion criteria → 6.4.3 (I/E)
- Missing monitoring plan → 6.4.8 (Safety assessments)
- Missing budget → 6.9 (Financing)

Question Type 2: "Classify This Change"

Question: "Protocol amendment to [describe change]. Is this an amendment or administrative change?"
Strategy: Does it affect who can enroll / what's measured / how it's measured?
- YES → Amendment
- NO → Administrative

Question Type 3: "Deviation or Violation?"

Question: "A subject [scenario]. Is this a deviation, major deviation, or violation?"
Strategy: Use decision framework
- Step 1: Non-compliance? YES → Deviation
- Step 2: Safety risk? YES → Major deviation or violation
- Step 3: Data integrity? YES → Major deviation or violation
- Step 4: Consent issue? YES → Violation
- Step 5: Minor? YES → Minor deviation

Question Type 4: "Apply I/E Criteria"

Question: "Subject has [characteristics]. Does I/E allow enrollment?"
Strategy: Match each characteristic to criteria
- Look for numeric thresholds (eGFR, age, BP)
- Calculate washout periods
- Determine eligibility YES/NO with reason

Question Type 5: "Drug Accountability Calculation"

Question: "[Scenario]. Calculate ending inventory. Does it balance?"
Strategy: Apply formula
- Beginning + Receipts - Dispensed - Returned - Destroyed = Ending
- Compare to physical count
- Identify discrepancy if applicable

High-Yield Topics for Final Review

MUST KNOW for exam:

  1. The 14 protocol sections (names and contents)
  2. Deviation vs. violation distinction (testable on 6-8 exam questions)
  3. Amendment vs. administrative (testable on 2-3 exam questions)
  4. I/E criteria application (testable on 2-3 exam questions)
  5. Drug accountability reconciliation formula (testable on 2-3 exam questions)
  6. IRT/IWRS basic functionality (testable on 1-2 exam questions)

SHOULD KNOW:

  1. Specific inclusion/exclusion examples
  2. CAPA components
  3. Reporting timelines
  4. Subject continuation decisions after violations
  5. Temperature excursion management

NICE TO KNOW:

  1. Historical protocols (what old protocols looked like)
  2. Alternative designs (rarely tested)
  3. International variations in protocol requirements

Common Exam Mistakes to Avoid

Mistake 1: Confusing "Dispensed" with "Consumed"

WRONG: "95 dispensed means 95 consumed"
RIGHT: "95 dispensed includes returns. Some consumed, some returned."
Formula impact: Must account for returns separately

Mistake 2: Thinking All Deviations are Violations

WRONG: "Late visit = violation"
RIGHT: "Late visit = deviation. If it affects primary efficacy data = major deviation. If time-critical = violation."
Strategy: Assess IMPACT, not just classify as deviation

Mistake 3: eGFR = "close enough"

WRONG: "eGFR 42 is close to 45, probably okay"
RIGHT: "eGFR 42 is < 45. Protocol says ≥60. Not eligible."
Strategy: Use the NUMBER. Don't interpret.

Mistake 4: Thinking Planned Deviations are Always Violations

WRONG: "Subject took prohibited medication = violation"
RIGHT: "Subject took prohibited medication for medical necessity, documented = deviation (not violation)"
Strategy: Assess whether DOCUMENTED and JUSTIFIED

Mistake 5: Confusing Amendment with Regulatory Submission

WRONG: "Amendment = must go to FDA immediately"
RIGHT: "Amendment = approved by IRB first. FDA notification only if safety-related."
Strategy: Amendment pathway ≠ FDA submission pathway (usually)

Last-Minute Review Checklist

Before test, verify you can:

If you're not confident on any of these, review before test.


FINAL EXAM STRATEGY

During the Exam:

  1. Read each question carefully—don't assume (multiple choice are often tricky)
  2. Look for KEY WORDS:
    • "Deviation" vs. "Violation" language
    • Numeric values for I/E criteria
    • "Amendment" vs. "Administrative"
  3. Use decision frameworks you memorized
  4. Don't rush—allocate 1-2 min per question
  5. Skip hard questions, come back to them
  6. Watch for "which is NOT true?" questions (reverse logic)

Time Management:


END OF COMPREHENSIVE LEARNING PACKET

This packet contains all essential Week 8 content for SOCRA CCRP exam preparation. Use alongside:

  1. ICH E6(R2) Section 6 (direct reading)
  2. Weekly Quiz #7 (practice)
  3. Practice question bank (reinforcement)
  4. Office hours (remediation)
  5. Instructor clarification (complex topics)

Good luck with your exam preparation. Protocol mastery is foundational to clinical research excellence.