Packaging Flashcards
(21 cards)
Tell us High speed packaging line controls
- Goods-In Check (Incoming Material Verification)
• Ensure correct bulk product (e.g., tablets) and packaging components (blister foil, cartons, leaflets) are received and meet specifications (identity, quantity, CoA review).
• Perform visual inspection of incoming materials for damage or contamination. - Line Clearance (Before Start-Up)
• Full clearance of the line prior to production start.
• Verification that no previous product or materials are present (to prevent mix-ups).
• Documented line clearance performed by trained operators and independently checked by QA or production supervisors. - Environmental and Temperature Controls
• Room temperature and humidity control in packaging areas.
• Temperature control at blister sealing stations to ensure proper adhesion without damaging the product (CPP for blister integrity). - Tablet/Blister Controls
• Camera Recognition/OCR System:
• Automated vision systems confirm the presence, position, and type of tablets/capsules in each blister pocket (detects wrong product, missing or broken tablets).
• OCR used to verify printed codes and information on blisters/foils.
• Blister/Hopper Speed Controls:
• Synchronisation between hopper feed and blister forming speed to prevent overfill, underfill, or misfeeds. - Sealing Foil Controls
• Temperature Control at Sealing Station:
• Critical for ensuring proper seal integrity without degrading product or foil (CPP).
• Cutting Mechanism Control:
• Accurate cutting aligned with printed foil patterns, ensuring readability of batch numbers, expiry dates, and regulatory markings. - Secondary Packaging Controls (Cartoning)
• Automated insertion of blisters and patient information leaflets (PIL) into cartons.
• Pharmacode/Barcode Verification:
• Ensures the correct PIL and blister are placed into the correct carton (avoids mix-ups).
• Carton printing (batch number/expiry date) and OCR/vision verification. - In-Process Controls (IPC)
• Weight Checks:
• Online checkweighers confirm each carton/blister meets expected weight parameters (detects missing components).
• Quality Control Sampling:
• Sampling at the beginning, middle, and end of the batch to check for defects (e.g., missing tablets, poor seals, print clarity). - Blister Integrity Testing
• Methylene Blue Dye Test or Pressure Dome Test:
• Verifies that blister seals are intact (critical for moisture-sensitive products). - Pin-Hole Detection System
• Online detection of microscopic holes in blister foils (prevents compromised barrier properties). - Material Reconciliation
• Ensure full reconciliation of bulk product and packaging components (account for rejects, samples, and waste).
• Prevents miscounts, overproduction, or diversion (aligns with Annex 16 and GMP Chapter 5). - Process Validation
• Ensure packaging processes consistently produce a product that meets predetermined quality attributes (e.g., sealing integrity, correct labelling).
• Ongoing Process Verification (OPV) or periodic review depending on risk and criticality. - Regular Audits and Line Inspections
• Internal audits of the packaging process and equipment.
• Regular maintenance and calibration of line equipment (vision systems, temperature sensors, weighers).
⸻
Extra Tips (QP Viva):
• Regulatory Expectations:
• Cite Annex 15 for validation and ongoing process verification.
• Refer to Annex 1 (if sterile products involved) for contamination controls.
• Highlight Annex 16 for reconciliation and QP certification.
• CCPs (Critical Control Points):
• Vision systems, sealing temperatures, weight checks, and blister integrity testing are often CCPs.
• You could reference ICH Q9 for risk management.
Let me know if you’
Why is pin hold detection important?
Pinhole detection is critical to ensure the integrity of primary packaging, particularly blister foils, which serve as the protective barrier for the product.
• Purpose:
Detects microscopic holes or defects in the packaging material (usually foil) that are not visible to the naked eye.
• Why important:
• Maintains product protection: Prevents exposure to environmental factors such as moisture, oxygen, or microbial contamination, which could degrade product quality (e.g., stability, potency).
• Ensures product shelf-life: Any breach in packaging integrity could compromise the product’s shelf-life and efficacy.
• Patient safety: Prevents potential harm due to degraded or contaminated product reaching patients.
• Regulatory compliance: Required as part of GMP (EU GMP Chapter 5 and Annex 16 for certification) to ensure packaging integrity before batch release.
• Relation to Packaging Integrity Testing:
• Pinhole detection complements other integrity tests like methylene blue dye or pressure dome tests, but is often done online during the high-speed packaging process to detect defects in real-time.
• Critical Control Point (CCP):
• Pinhole detection is typically considered a CCP for packaging lines, especially for products sensitive to moisture or oxygen (align with ICH Q9 risk management principles).
You are changing your PIL – can you talk through how this would be managed?
- Initiate Change Control:
• Raise a formal change control within the Pharmaceutical Quality System (PQS), documenting the rationale for the PIL change (e.g., safety update, new indication, readability improvements).
• Conduct a risk assessment to evaluate potential patient impact.- Determine Regulatory Pathway:
• Engage the Regulatory Affairs team to assess the type of variation required under the Human Medicines Regulations 2012 (or EU Variations Guidelines if applicable):
• Type IB or Type II Variation depending on the criticality of the change (e.g., safety-related changes would typically require a Type II variation).
• Reference: EMA Guideline on the Categorisation of Variations (C(2013) 2804 final). - Generate Updated PIL Mock-up:
• The artwork/design team prepares a mock-up of the updated PIL, ensuring compliance with readability requirements (e.g., font size, layout).
• If significant changes are made, a readability user testing (per EU Directive 2001/83/EC Article 59(3)) may be required to ensure patient comprehension. - Review and Approval:
• The updated PIL mock-up is reviewed and approved by both QA and Regulatory Affairs to ensure compliance with the Marketing Authorisation (MA) and regulatory guidelines. - Artwork Implementation and Packaging Coordination:
• Coordinate with the packaging/artwork team to implement the new PIL into the packaging process.
• Ensure that the new leaflet version is controlled, and the old version is superseded in the document management system and at the packaging site. - Regulatory Approval and Final Release:
• No implementation of the updated PIL is permitted until regulatory approval of the variation is received (for MA variations).
• Final QA approval ensures that the correct version of the PIL is released alongside the product. - Post-Implementation Monitoring (Optional):
• Monitor for any feedback from healthcare professionals or patients following the introduction of the new PIL to assess its effectiveness.
- Determine Regulatory Pathway:
There was a rogue tablet found which was blue in color after the manufacturing of 4 batches of white tablets.
This led to discussion about line clearance packaging machine challenge testing.
Open Deviation
* Correct. This is a Major deviation due to a suspected product mix-up (cross-contamination risk), which is critical under GMP.
- Description of Situation
* Good structure. Ensure you capture:
o When: Date and time of discovery.
o Where: Manufacturing area (specific room/line/equipment).
o During which process: e.g., packaging, line clearance, visual inspection.
o Who: Name and role of the person who found it.
o How found: Routine in-process check, visual inspection, or through QA walk-around?
Tip: Capture photos, retain the rogue tablet as evidence, and document the chain of custody.
________________________________________
3.
What is the White Tablet?
* Include:
o Product name, strength, indication, dosage form.
o Patient group: Especially if it’s a vulnerable population (e.g., pediatric, oncology).
o Market alternatives: Any available substitutes (important for risk to patient supply if recall occurs).
o Campaign manufacturing: Confirm if the 4 batches were produced consecutively (relevant for cleaning validation and cross-contamination control).
________________________________________
4.
What is the Blue Tablet?
* Include:
o Product name, strength, indication.
o Is the product manufactured on the same line/equipment?
o When was the last blue tablet batch manufactured?
o Cleaning validation status between product changeovers?
Tip: Check shared components (punches/dies) if applicable.
________________________________________
5.
Impact Assessment
* Excellent breakdown:
o MA (Marketing Authorisation): Deviation from specification due to foreign tablet presence (breach of product identity, purity).
o GMP: Product mix-up suggests potential failure in cleaning, line clearance, or segregation – may impact robustness of process controls.
o Patient safety: Consider potential adverse effects if the blue tablet has a different API/dose (especially high-risk products like cytotoxics).
o Batch status: Confirm whether any batches have been released or are still under QC. This drives recall scope.
Tip: Reference ICH Q9 (risk to product quality and patient safety).
________________________________________
6.
Immediate Actions
* Good.
o Quarantine all 4 batches.
o Inspect reference samples for foreign tablets.
o Extend checks to packaging components (blisters, labels) to confirm no mix-up at the packaging stage.
o Halt production line pending investigation.
________________________________________
7.
Root Cause Investigation (RCI)
* Well-structured Ishikawa (fishbone) approach. Expand on:
o QMS: Include deviations, complaints, OOS, OOT, change controls, PQR trends related to the product or line.
o Personnel: Check operator logs, shift patterns, any anomalies in staffing (fatigue, new staff, contractors).
o Documents: Review BMR/BPR for errors, incomplete sections, or deviations noted but uninvestigated.
o Facility/Equipment:
Cleaning validation (dirty hold times, residual testing).
PPM schedules.
Environmental monitoring trends.
CCTV review: Especially during cleaning, line clearance, and batch start-up.
o Process:
Line clearance: Confirm thoroughness, reconciliation (are counts accurate?).
Material reconciliation: Raw materials, intermediates, packaging components.
Goods-in checks: Ensure no cross-contamination of incoming materials.
Validation: Review process validation status for both products.
________________________________________
8.
Recall Considerations
* Spot on.
o Recall decision should involve:
Multidisciplinary Recall Committee (QP, QA, RA, Pharmacovigilance).
Risk classification (MHRA Guide to Defective Medicinal Products: Class 1, 2, or 3).
o DMRC contact before recall execution – MHRA requires early notification for potential Class 1 or 2 recalls.
________________________________________
9.
CAPA
* Yes, open CAPA based on investigation findings.
o Ensure effectiveness checks are defined (e.g., repeat line clearance audits, revalidation of cleaning procedures, retraining outcomes).
o Link CAPA to QMS review (PQR, deviation trending).
________________________________________
10.
Closing Report to DMRC
* Correct.
o Include:
Summary of deviation, investigation, CAPA.
Root cause and preventive actions.
Impact on other batches.
Patient safety evaluation.
o Follow the MHRA expectations for timelines and closure (as per the DMRC guide.
You received a complaint regarding a batch of sachets. Complainant stated that the pack contains white powder instead of the normal granules. What do you do?
Point - identify if the issue is related to product mix up or granulation or quality/formulation (e.g, API, excipients):
Gather Initial Information:
* Product Details:
o Identify the product name, indication, target patient group, and formulation (granules).
o Confirm if your facility manufactures any product in white powder form (risk of cross-contamination/mix-up?).
o Determine granulation method used (wet vs. dry), as this informs potential manufacturing defects.
o Identify market alternatives and potential clinical risks (e.g., excipient changes, bioavailability concerns).
* Complaint Details:
o Register the customer complaint per your Complaints SOP, in line with EU GMP Chapter 8.
o Request sample (if feasible) or at least batch number and expiry date for traceability.
________________________________________
2.
Immediate Assessment:
* Quarantine any impacted batch still under your control (Annex 16 – product not to be certified/released until resolved).
* Check distribution records to identify the quantity and destinations of released product.
* Evaluate if additional batches may be affected (shared equipment, line clearance history).
________________________________________
3.
Investigation:
* Perform QC testing on the complaint sample:
o Compare with retention samples (appearance, particle size, ID tests).
o Investigate whether the white powder is an incorrect product (product mix-up) or a granulation failure.
* Search for similar complaints in the complaints database (trend analysis, recurring issues).
________________________________________
4.
Impact Assessment:
* Market Authorization (MA):
o Does the issue lead to an Out-of-Specification (OOS) product or a product mix-up?
o Consider variability in granulation potentially impacting bioavailability (CQA).
* GMP Non-Compliance:
o Assess if this is indicative of a process failure, cross-contamination, or mix-up (Annex 1, Annex 15, Chapter 5).
* Patient Safety:
o Risk of adverse effects or reduced efficacy due to incorrect dosage form (granules vs. powder).
o Classify defect severity according to MHRA Defective Medicines Report Centre (DMRC) guidelines.
________________________________________
5.
Root Cause Analysis (RCA)
– Use
Ichikawa (Fishbone) Method
:
* PQS:
o Review deviations, change controls, PQRs for trends.
* Personnel:
o Check training records, operator qualification status for granulation/blending/packaging.
* Documentation:
o Review Batch Manufacturing Records (BMRs) for anomalies/comments.
* Facility/Equipment:
o Check preventive maintenance (PPM), calibration, cleaning records, EM data (humidity/temperature).
* Process Parameters:
o Review granulation speed, blending duration, binder quality, sieving/milling speed, drying parameters.
* Suppliers:
o Assess API/excipient suppliers (recent changes, CoAs, quality trends in PQRs).
o Investigate packaging materials for potential changes or non-conformance.
* Internal Audits/Self-Inspection:
o Review findings that might relate (Annex 1, Chapter 9).
________________________________________
6.
Regulatory Reporting & Recall Decision:
* Convene a recall committee (Quality, Regulatory Affairs, Medical).
o Use MHRA recall classification:
Likely Class 2 (may affect bioavailability).
Confirm recall level (likely patient level due to self-administered formulation).
* Notify DMRC (before executing recall) per Orange Guide and MHRA expectations.
________________________________________
7.
Corrective and Preventive Actions (CAPA):
* Raise CAPA aligned with ICH Q10 principles:
o Address root cause (e.g., equipment issue, supplier failure, operator error).
o Ensure effectiveness check to prevent recurrence.
________________________________________
8.
Closure and Reporting:
* Submit final report to DMRC detailing:
o Complaint nature, investigation outcome, root cause, impact assessment, recall actions, CAPA implementation, and effectiveness checks.
________________________________________
Key Regulatory References:
* EU GMP Chapter 8 – Complaints and Product Recall.
* Annex 16 – Certification by a QP.
* ICH Q9/Q10 – Risk Management, Pharmaceutical Quality System.
* MHRA Defective Medicines Report Centre (DMRC) guidelines.
* MHRA Inspectorate Blog – Recall expectations, risk classifications.
R&D manager tells you they want to pack an IMP and placebo for stage IV clinical trial on high speed packing line used for commercial, what do you say?
- What are the controls on a high speed packing line?
- Open communication (CC):
• Initiate dialogue with the R&D Manager, QA, Production, and other stakeholders to fully understand the clinical trial requirements (e.g., blinding, batch size, timelines, packaging specifics).- Impact assessment on Licence:
• Confirm that the site holds a Manufacturer’s Authorisation for Investigational Medicinal Products (MIA(IMP)) covering the packaging activities for the IMP and placebo.
• Ensure the QP release for IMP is performed in line with UK SI 2004/1031 (The Medicines for Human Use [Clinical Trials] Regulations).
• If this is a Phase IV trial using a marketed product, consider the application of Annex 16 for commercial product certification alongside Annex 13 requirements for clinical packaging. - Impact assessment on GMP (QMS considerations):
• QMS readiness: Ensure that the Quality Management System includes procedures (SOPs) specific to IMP packaging activities, covering areas such as:
• Deviation management,
• Change control (CC),
• Complaint handling,
• Batch documentation and reconciliation.
• Personnel:
• Confirm that personnel involved in IMP packaging receive GMP training, with additional training in GCP principles (as per ICH E6) to understand the clinical trial context, particularly blinding requirements and patient safety implications.
• Documentation:
• Verify that labeling and artwork comply with Annex 13 (or EU CTR) requirements, including statements like “For Clinical Trial Use Only,” randomisation codes, storage conditions, trial reference numbers, and expiry dates.
• Process controls:
• Implement contamination control measures, including:
• Cleaning validation of the high-speed packing line to prevent cross-contamination between commercial and clinical products.
• Goods-in checks for all packaging components and labels.
• Line clearance procedures to prevent mix-ups and maintain blinding integrity.
• Segregation of materials (IMP, placebo, commercial products) during storage and packing.
• Outsourced activities:
• Ensure a Quality Technical Agreement (QTA) is in place with the sponsor (or any third-party vendors), clearly defining responsibilities for packaging, labeling, and batch certification/release activities.
• Self-inspection:
• Confirm that periodic self-inspections of the IMP packaging operations are conducted as part of the QMS to ensure continued compliance with GMP and GCP.
- Impact assessment on Licence:
Rogue tablet found on packaging line. It is aspirin, found during packing salbutamol. Aspirin last manufactured 2 batches back. What do you do? Different shape and colour. Would you contact DMRC ?
- Product Understanding (Initial Risk Context)
• What is the product being packed?
• Formulation: Salbutamol tablets.
• Indication: Asthma, COPD.
• Patient population: Respiratory patients — risk of aspirin hypersensitivity (e.g., aspirin-exacerbated respiratory disease).
• Market alternative: Assess if alternative salbutamol supply exists.
⸻
- Register Deviation & Gather Key Information
• Register as major deviation due to potential product mix-up/contamination.
• Gather details:
• Who found the rogue tablet?
• When and where exactly (on line, post-pack, pre-label)?
• How was it detected? (e.g., visual inspection, automated detection)
• Proximity to point of fill (hopper)? Any chance of ingress upstream?
• Manufacturing context: Were the previous aspirin and current salbutamol batches part of campaign manufacturing? Cleaning status in between?
⸻
- Impact Assessment (Regulatory, GMP, Patient Risk)
• Regulatory risk (MA breach):
• Possible cross-contamination and misbranding — mixing of products in contravention of MA.
• GMP impact:
• Potential failure in line clearance, cleaning validation, contamination control strategy.
• Patient safety risk:
• Severe risk due to aspirin hypersensitivity in respiratory patients (salbutamol’s target population).
⸻
- Immediate Actions
• Quarantine:
• Affected batch (current salbutamol), last two batches (post-aspirin), and any work-in-progress.
• Reference sample inspection:
• Review retention samples for evidence of rogue tablets or contamination.
• Check distribution status:
• Identify if batches are still on site or already released/distributed.
• Complaint trend review:
• Check pharmacovigilance/complaints database for signals (e.g., unexpected asthma events).
⸻
- Root Cause Analysis (RCA) – Ishikawa Method
• QMS:
• Review deviation, CC logs, prior near-miss events for rogue tablet issues.
• Facility/Equipment:
• Line clearance records, cleaning records, recent PPM/service logs.
• Detection systems (camera recognition, metal detectors, vision systems) — verify qualification status, last challenge test performance.
• Equipment logbooks — confirm aspirin-to-salbutamol changeover steps were followed.
• Personnel:
• Review training, qualification records for operators involved in line clearance and batch setup.
• Documentation:
• Review BMRs for comments, non-standard events, or operator notes.
• Process:
• Verify cleaning validation reports for robustness, line clearance steps, visual inspection effectiveness.
⸻
- Escalation & Regulatory Contact
• If risk of product mix-up cannot be excluded (e.g., rogue tablet source unknown, potential batch contamination):
• Initiate recall risk assessment:
• Likely Class 1 or Class 2 recall due to potential for aspirin hypersensitivity in salbutamol patients.
• Determine recall level — likely patient level.
• Engage Medical, Regulatory Affairs, QP for formal risk classification.
• Contact DMRC (MHRA) proactively:
• Notify potential product defect per MHRA Defective Medicines Report Centre guidelines, before recall execution.
• Provide initial risk assessment, patient risk evaluation, and proposed actions.
⸻
- Corrective & Preventive Actions (CAPA)
• Corrective actions:
• Review/validate detection systems (e.g., camera, vision systems).
• Investigate line clearance procedures.
• Dispose quarantined batches if contamination cannot be ruled out.
• Preventive actions:
• Retraining personnel on line clearance.
• Enhance cleaning validation frequency, scope.
• Upgrade/validate detection capabilities (e.g., rogue tablet detection).
• Effectiveness checks post-CAPA before resuming production.
⸻
- Regulatory Closure
• Submit final defect report and CAPA summary to DMRC.
• Document QP decision-making process, regulatory engagement, and patient safety justification.
An operator tells you that they just focus a blister pack with an empty pocket, what do you do?
- Immediate Actions:
- Quarantine: Immediately quarantine the impacted batch and stop the packing line to prevent further processing.
- Record Details: Open a deviation (or non-conformance report), capturing:
- When it was found (date, time).
- Who found it (operator name).
- Where it was found (line, station).
- How it was detected (visual check, automated sensor, etc.).
⸻
- Initial Impact Assessment:
On the Product/Patient:
* Criticality: Missing tablet could lead to underdosing or treatment failure.
* Check product license (MA) – if it’s a critical dose product or narrow therapeutic index, the risk is higher.
* Check customer complaint records to identify if similar issues have occurred (signal detection).
On GMP:
* Indicates a potential failure of in-line controls (e.g., vision systems, weight checks).
* Check reference samples (retention samples) for evidence of similar defects.
* Conduct challenge testing to confirm if the detector/vision system can reliably detect empty pockets.
⸻
- Full Investigation (Ishikawa/Fishbone Analysis):
Root Cause Analysis Areas:
* Personnel:
* Was the operator trained and qualified?
* Any recent human error deviations?
* Equipment:
* Review Preventive Maintenance (PPM) records and calibration status of detection systems (e.g., vision systems).
* Any recent equipment breakdowns or outages?
* Confirm qualification status of equipment (e.g., line requalification after maintenance).
* Materials:
* Any issues with the blister material (e.g., feeding issues, supplier changes)?
* Methods/Processes:
* Review packaging process validation – is the method robust and are reject systems validated?
* Confirm that line clearance procedures were properly followed.
* Documentation:
* Any Batch Manufacturing Record (BMR) comments or anomalies noted?
* Review change controls – any recent process or equipment changes?
* QMS:
* Any related deviations, CAPAs, or self-inspection findings?
* External Factors:
* Any supplier issues with materials (e.g., tablet feeding consistency)?
* Outsourced equipment servicing – any concerns?
⸻
- Evaluate Wider Impact:
- If a systemic failure is identified (e.g., vision system non-functional), review other batches produced on the same line.
- If affected batches are released to market, convene a recall decision meeting:
- Assess recall classification (MHRA: Class 1, 2, 3).
- Determine recall level (e.g., wholesale, pharmacy, patient).
⸻
- Corrective and Preventive Actions (CAPA):
- Based on the root cause, implement CAPAs, for example:
- Increase equipment servicing frequency.
- Revalidate the detection system.
- Retrain personnel if human error is identified.
- Enhance process controls (e.g., introduce manual spot checks at intervals).
- Conduct effectiveness checks to ensure CAPAs resolve the root cause.
⸻
- Regulatory Reporting:
- If a recall is executed, submit a recall report to the DMRC (Defective Medicines Report Centre) and close the case following their guidance.
- Include a summary of the investigation, recall scope, CAPAs, and effectiveness checks.
⸻
- QP Consideration:
- As a QP, I would not certify the batch until the investigation concludes, root cause is identified, and CAPAs are implemented.
- I would ensure the product quality review (PQR) incorporates findings from this investigation to identify trend signals.
⸻
Final Suggestion:
* For viva, structure your answer:
1. Immediate actions (quarantine, record details).
2. Risk assessment (patient, GMP).
3. Investigation (Ishikawa).
4. Wider impact (other batches, recall).
5. CAPA.
6. Regulatory reporting.
7. QP decision-making.
What controls are in place on a high-speed packaging line?
Model Answer: Controls in Place on a High-Speed Packaging Line
1. Goods-In / Material Controls:
* Material identity and quality check (against COA/COC).
* Label reconciliation to prevent mix-ups.
* Line clearance to ensure removal of previous batch materials, supported by a documented line clearance checklist.
⸻
2. Blister Pocket Forming: * Process controls: * Belt conveyor speed monitored. * Temperature and pressure settings for thermoforming of blister pockets (PVC/Alu or Alu/Alu blisters). * Vacuum level monitoring to ensure consistent pocket formation. * Periodic checks on pocket integrity (visual inspection, depth check).
⸻
3. Tablet (or Capsule) Filling: * Product presence/absence sensors to detect missing tablets in pockets. * Vision systems for: * Shape/size verification. * Color recognition. * OCR (Optical Character Recognition) for printed tablets or capsule markings to prevent product mix-up.
⸻
4. Lidding / Sealing: * Temperature, pressure, and dwell time controls for the sealing station (aluminum foil to formed blister). * Sealing integrity checks (visual checks and/or inline sensor verification).
⸻
5. Cutting and Perforation: * Cutting mechanisms designed to avoid disturbing printed data (batch number, expiry date). * Camera systems may verify correct positioning/alignment of print post-cutting.
⸻
6. Cartoning: * Pharmacode/barcode scanners to verify correct carton. * In-process weight checks to confirm leaflet presence and correct blister count. * Tamper-evident features checked (if applicable).
⸻
7. Batch Printing (Batch/Lot & Expiry Date): * OCR/OCV systems (Optical Character Verification) to ensure correct batch and expiry details on cartons/blisters. * Reject mechanisms for illegible or incorrect prints.
⸻
8. Final Packaging Checks: * Aggregation systems (if serialization is required) for tracking packs at various packaging levels (blister → carton → shipper).
⸻
9. Quality Control (QC) Sampling and Testing: * In-process sampling at start, middle, and end of the batch for: * Blister integrity tests: * Blue dye ingress test (offline). * Vacuum/pressure decay tests (offline). * Inline pinhole detectors (electrical conductivity/laser-based systems). * Finished product checks: * Weight checks. * Visual inspections. * Label reconciliation.
⸻
10. Environmental Controls: * Temperature and humidity monitoring in the packaging area to protect product stability.
⸻
Additional Notes (QP Perspective):
* Alarm systems and interlocks ensure the line stops if critical parameters deviate (e.g., vision system failures, sealing temperature excursions).
* Calibration and qualification of critical equipment (e.g., vision systems, weighing scales) are part of Preventive Maintenance Programs (PPM).
* Periodic challenge tests on detection systems (e.g., introducing deliberately faulty packs) to confirm reject mechanisms work.
Your operators on your high-speed packaging line come to you with a problem that tablets are twinning? What does that mean? What would you do about it?
- Understanding the Issue:
- Twinning refers to two tablets sticking together, entering a single blister pocket.
- This poses a significant risk:
- Overdose for one pocket, underdose for another.
- Fails product specification (appearance, dose uniformity).
- Patient safety concerns (non-compliance, bioavailability).
⸻
- Immediate Actions:
- Stop the packaging line and quarantine the affected batch.
- Raise a deviation in the Quality Management System (QMS), documenting:
* When, where, and how the issue was detected.
* Operator details. - Conduct a challenge test on the detection system (vision/weight):
* Verify if twinned tablets can be detected and rejected.
* If detection capability is lacking, temporarily increase manual inspection frequency. - Physically confirm if twinned tablets can fit into blister pockets without detection.
* If so, consider the issue high-risk and escalate accordingly. - Quarantine any potentially affected bulk tablets (in storage or earlier in the process).
- Check customer complaint records and reference samples for any trends (e.g., coating defects, twinning issues).
⸻
- Impact Assessment:
- Product License (MA):
- Fails appearance specification.
- Risk of dose variability (critical defect).
- GMP Compliance:
- Potential process control failure (coating process, packaging detection systems).
- Patient Safety:
- Risk of underdosing/overdosing, treatment failure, or adverse effects.
- Visual defects may lead to patient non-compliance.
If released batches are potentially affected, convene a recall decision meeting (per MHRA recall classification).
⸻
- Root Cause Investigation (Ishikawa/Fishbone Analysis):
Materials:
* API/excipient CoAs: Check for hygroscopic properties.
* Storage conditions for bulk tablets: Review temperature/humidity logs.
* Supplier complaints or changes in material?
Equipment (Facility):
* Preventive maintenance (PPM) and qualification status of:
* Tablet coating equipment (spray nozzles, drying air).
* Packaging feeders (vibratory feeders, chutes).
* Vision/weight detection systems.
* Review environmental monitoring in packaging and storage.
Personnel:
* Review training records of operators.
* Confirm adherence to SOPs (e.g., line clearance, equipment cleaning).
Process:
* Review CPPs for tablet coating:
* Spray rate, pan speed, drying temperature/time, nozzle distance.
* Tablet compression process (hardness, friability).
* Batch records for deviations or anomalies.
Documentation/QMS:
* Review recent deviations, change controls, PQRs, and self-inspections for trends.
⸻
- Wider Impact and Regulatory Reporting:
- Determine if released batches are impacted.
- Recall decision meeting:
- Classify recall (MHRA: Class 1, 2, or 3).
- Determine recall level (wholesale, pharmacy, patient).
- Notify DMRC before executing the recall (if required).
- Submit recall reports and close the case with DMRC.
⸻
- Corrective and Preventive Actions (CAPA):
- Based on root cause, implement CAPAs, e.g.:
- Optimize coating parameters to prevent twinning.
- Upgrade detection systems (vision/weight) if challenge tests reveal deficiencies.
- Retrain staff on process controls.
- Enhance environmental controls (e.g., humidity monitoring in storage/packaging areas).
- Perform effectiveness checks to ensure CAPAs address the issue.
⸻
- QP Considerations:
- Withhold certification of the impacted batch until:
- Investigation concludes.
- Root cause is identified.
- CAPAs are implemented and effective.
- Ensure findings are fed into the Product Quality Review (PQR) process for trend analysis.
⸻
Regulatory References:
* EU GMP Annex 16 – QP responsibilities for batch certification.
* EU GMP Annex 15 – Qualification and validation.
* EU GMP Chapter 8 – Complaints and recalls.
* MHRA recall guidance – DMRC process.
What are the risks of twining??
- Patient Risks:
- Overdose/underdose:
- Twinned tablets may lead to double dosing in one pocket and no dose in another.
- Risk of adverse effects (overdose) or treatment failure (underdose).
- Non-compliance:
- Visual defects (twinned tablets, misshapen appearance) can lead to patient mistrust and non-adherence.
⸻
- Product Risks:
- Specification failure:
- Fails appearance, dose uniformity, or content uniformity tests.
- Stability issues:
- If coating defects occur (e.g., exposed core due to twinning), it can affect stability (especially for moisture-sensitive APIs).
- Non-conformance:
- Product may breach registered specifications (as per Marketing Authorization (MA)).
⸻
- GMP Risks:
- Indicates process failure:
- Poor coating process control.
- Inadequate environmental controls (humidity/temperature excursions).
- Equipment malfunction (e.g., feeders, coating pans, packaging line).
- Systemic failure potential:
- If undetected, may suggest ineffective detection systems (e.g., vision/weight control), leading to wider batch impact.
- Regulatory risk:
- Potential recall and regulatory reporting (e.g., DMRC in the UK).
What are controls on the line?
Model Answer: Controls on a Manual Packaging Line
1. Goods-In and Pre-Packaging Checks:
* Material identity verification against the Batch Manufacturing Record (BMR) and Certificates of Analysis (CoA).
* Line clearance checks to ensure removal of previous materials (manual line clearance with checklists).
* Cleaning verification of the packaging area and equipment.
⸻
2. Personnel Controls: * Training and qualification: * Operators trained in GMP, packaging procedures, and visual inspection techniques. * Periodic refresher training and requalification (especially for critical tasks like manual inspection). * Segregation of duties: Ensuring independent checks (e.g., one person packs, another verifies).
⸻
3. Manual Filling/Assembly Controls: * 100% visual inspection by two trained individuals: * First operator performs the packaging operation. * Second operator performs a 100% independent visual check to verify: * Correct product. * Correct quantity. * Correct packaging components (leaflet, carton, labels). * No defects (e.g., damage, contamination). * Material reconciliation: * Typically 100% reconciliation at the end of batch for manual lines, due to lack of automation. * All packaging components (leaflets, cartons, labels) are accounted for (issued vs used vs returned).
⸻
4. Weight Checks (if applicable): * Manual weighing of the final packaged product (including product, leaflet, primary and secondary packaging). * Typically performed on sampled units, unless 100% check is feasible.
⸻
5. In-Process Sampling: * QC sampling at: * Beginning, middle, and end of the packaging process. * Checks performed on samples: * Visual inspection for packaging integrity. * Label accuracy and legibility. * Correct components included (e.g., leaflet, correct batch/expiry details).
⸻
6. Packaging Integrity Testing: * Blister packs (if applicable): * Blue dye ingress test (offline sample-based). * Pressure dome or vacuum leak tests (depending on product type). * Sealed containers: * Visual seal integrity checks.
⸻
7. Label Control and Reconciliation: * Strict reconciliation of labels: * Account for all labels issued, used, and returned/destroyed. * Manual verification of label details (batch number, expiry date).
⸻
8. Documentation Controls: * Batch Packaging Record (BPR): * Completed in real-time. * Double-checked by a second person. * Deviation handling: * Any discrepancy (e.g., reconciliation failure) is raised as a deviation for investigation.
⸻
Key Differences from High-Speed Packaging:
* No automated detection systems (e.g., vision, weight sensors).
* Reliance on human checks (hence higher emphasis on training, double checks, and manual reconciliation).
* Lower batch sizes, but higher risk of human error if controls are weak.
⸻
QP Considerations:
* Ensure personnel competence through training records.
* Review line clearance, reconciliation, and sampling records thoroughly.
* Check that label reconciliation meets GMP Chapter 5 expectations.
* Validate manual inspection processes (as per Annex 15 for qualification).
While certifying a batch of commercial product you notice that the braille does not match the pack strength (braille – 50mg, pack 25mg). What are you concerns and what do you do?
- Concerns (Risk Assessment):
- Regulatory Risk:
- Non-compliance with the Marketing Authorization (MA): The approved artwork (including Braille) forms part of the Product License (Annex I SmPC and labeling).
- Potential recall: Mislabeling is a critical defect (GMP Chapter 8, MHRA recall guidance).
- GMP Risk:
- Indicates a failure in packaging artwork control or incoming goods check.
- Suggests potential process/system failure (e.g., artwork approval process, packaging line controls).
- Patient Risk:
- Misleading Braille information could affect visually impaired patients:
- Potential for wrong product selection or dosing error.
- Non-compliance risk due to confusion or mistrust.
⸻
- Immediate Actions:
- Quarantine the affected batch and stop the packaging line.
- Extend the quarantine to other batches packaged with the same artwork batch or component delivery.
- Check reference samples (retention samples) from other batches using the same packaging components.
- Review customer complaint records for any related issues (e.g., mislabeling, Braille complaints).
⸻
- Document the Issue:
- Raise a deviation/non-conformance in the Quality Management System (QMS), recording:
- When, where, and how the issue was detected.
- Batch numbers, packaging component lot numbers, and operators involved.
⸻
- Impact Assessment:
- Regulatory:
- Breach of MA (labeling and packaging requirements).
- Assess need for recall based on MHRA recall classification and regulatory advice.
- Engage Regulatory Affairs team to review labeling approval history.
- GMP:
- Process failure in artwork control, component release, or packaging verification systems (e.g., barcode, pharmacode, Braille detection if applicable).
- Patient Safety:
- Potential misuse by visually impaired patients.
- Risk might justify a Class 3 recall (e.g., pharmacy level), but risk assessment should involve Regulatory Affairs and clinical input.
⸻
- Regulatory Reporting:
- Convene a recall decision meeting:
- Determine recall class (Class 1, 2, 3) and recall level (e.g., wholesaler, pharmacy, patient).
- Likely a Class 3 pharmacy-level recall, but depends on risk assessment outcome.
- Notify the DMRC before executing the recall (per MHRA guidance).
⸻
- Root Cause Analysis (RCA - Ishikawa/Fishbone):
- QMS:
* Review previous deviations, change controls, PQRs for trends.
* Check label/artwork approval process for recent changes. - Personnel:
* Verify training/qualification records for staff involved in artwork approval, goods-in, and packaging operations. - Facility/Equipment:
* Review detector systems on the packaging line (e.g., pharmacode scanners, Braille detectors if present).
* Check maintenance logs and qualification status of equipment (Annex 15). - Documentation:
* Review Batch Packaging Records (BPRs) for this and other affected batches.
* Cross-check with approved artwork files and component specifications. - Process:
* Examine Goods-In checks, label reconciliation records, and packaging approval process.
* Assess artwork control system (e.g., supplier approval, artwork change management). - Supplier/Outsourced Activities:
* If artwork or Braille embossing is outsourced:
* Review supplier qualifications, CoCs, delivery notes, and goods-in checks.
* Review PQR data for any trends or previous findings related to packaging suppliers. - Self-Inspections/Audits:
* Check recent internal/external audits for findings related to packaging controls or artwork management.
- QMS:
⸻
- Corrective and Preventive Actions (CAPA):
- Based on root cause, potential CAPAs may include:
- Review/retrain staff on artwork control and approval.
- Enhance incoming goods checks (e.g., verify Braille at Goods-In).
- Upgrade or calibrate detection systems (e.g., Braille inspection tools, pharmacode scanners).
- Improve supplier oversight (e.g., audits, specifications for Braille embossing).
- Conduct effectiveness checks (e.g., additional batch reviews, supplier audits).
⸻
- Regulatory Close-out:
- If recall executed, submit recall report to DMRC for closure.
- Document all actions, RCA findings, and CAPAs in the QMS.
⸻
Regulatory References:
* EU GMP Chapter 5.58-5.60 – Labeling controls and reconciliation.
* EU GMP Annex 16 – QP certification responsibilities.
* EU GMP Chapter 8 – Complaints, quality defects, and recalls.
* MHRA Recall Guidance – DMRC reporting
Investigation revealed that the problem was with the artwork approval process.
Talk through the artwork approval process and indicate where this issue could have been identified and prevented.
Artwork Approval Process (Commercial Product):
1. Change Control (CC) Raised:
o A Change Control is initiated when any artwork or packaging component change is required (e.g., Braille strength update, regulatory text changes).
o The CC owner identifies the nature of the change (e.g., strength update, regulatory wording, design layout).
________________________________________
2. Impact Assessment (Regulatory, GMP, Patient):
- Regulatory Impact (MA/CTD Module 1):
o Artwork changes affecting patient-critical information (e.g., strength, indication, dosing) may require a variation submission to MHRA.
o Depending on the nature, this could be:
Type IB variation (minor but requires notification).
Type II variation (if critical safety, efficacy, or labeling info changes).
o The Regulatory Affairs team confirms the variation category. - GMP Impact:
o Affects packaging documentation (BMR/BPR updates), supplier communication, artwork version control, equipment settings (e.g., Braille embossing tools).
o Requires deactivation of old artwork versions. - Patient Impact:
o Ensures the patient receives accurate and compliant labeling, including Braille for visually impaired patients.
o Mislabeling can lead to non-compliance or incorrect dosing.
________________________________________
3. Artwork Mock-Up Design:
o A mock-up is designed by the Design Team (or outsourced artwork provider) based on:
The current MA requirements.
The approved template/layout (including Braille specification).
o Includes all regulatory information, product strength, legal texts, Braille embossing, and visual elements.
________________________________________
4. Artwork Approval (Internal Review):
o The mock-up undergoes multi-disciplinary review and approval, including:
Regulatory Affairs (compliance with MA/SmPC).
Quality Assurance (QA) (GMP compliance, version control).
Production/Packaging Team (feasibility for printing, embossing, equipment).
Marketing/Design Team (branding alignment).
o Approval records are maintained in controlled artwork management systems (e.g., electronic document control systems).
________________________________________
5. Variation Submission to MHRA (If Applicable):
o If required, Regulatory Affairs submit a variation (Type IB or II) to MHRA with the updated artwork.
o No change is implemented until MHRA approval is received.
________________________________________
6. Implementation Preparation (Post-Approval):
o Once MHRA and QA approve the updated artwork:
Suppliers are informed (e.g., printed component suppliers, embossing tool manufacturers).
Equipment updated (e.g., new Braille embossing plates installed).
Documentation updated (BMR/BPRs, SOPs).
Staff trained on new artwork version (focus on packaging operators, QA inspectors).
o Old artwork versions are deactivated in the Document Management System (DMS) to prevent accidental use.
________________________________________
7. Change Implementation:
o The updated artwork is released for production.
o QA line clearance verifies the correct artwork version, Braille embossing plates, and packaging components before starting the batch.
________________________________________
Where the Issue Could Have Been Identified and Prevented:
1. During the Internal Artwork Approval Review:
o The QA or Regulatory team should have cross-checked:
Braille content matches SmPC and MA strength.
Proofreading includes Braille verification (using Braille-reading tools or third-party checks).
o Failure here suggests a gap in artwork review controls.
2. During Goods-In Checks for Packaging Components:
o Packaging component deliveries (e.g., cartons with Braille) undergo incoming checks:
Visual verification.
Braille verification (if equipment or manual reading tools available).
o The issue could have been caught at Goods-In before packaging started.
3. At Line Clearance/Packaging Line Setup:
o Line clearance checks include verifying:
Correct packaging components.
Correct artwork version.
o If Braille verification is part of line clearance (e.g., embossing plate check), the discrepancy could have been caught here.
4. Supplier Approval/Qualification:
o If the Braille embossing plates are provided externally, supplier qualification and incoming quality checks (CoA/CoC for tooling) should confirm they match the approved artwork.
Where would you find the requirements for braille on packaging?
What standard is required for braille?
Drug name and strength have already been mentioned, what other information would a blind consumer need to know?
- Where to find the requirements for Braille on packaging:
• MHRA’s “Guideline for the Naming of Medicinal Products and Braille Requirements for Name on Label” outlines the requirements for Braille on outer packaging in the UK.
• The legal basis is from UK Human Medicines Regulations 2012 (SI 2012/1916), reflecting EU Directive 2001/83/EC, which remains part of retained EU law post-Brexit.- What must appear in Braille:
• The product name, including:
• Strength (if more than one strength exists).
• Pharmaceutical form (if more than one form exists).
• This must appear in Braille on the outer packaging (e.g., cartons, not blister packs).
• Example:
Paracetamol 500 mg tablets
The full name with strength and form must be represented. - What standard is required for Braille:
• The MHRA does not mandate a specific Braille font but recommends following national or European standards, such as:
• Marburg Medium Braille font (widely used across the EU/UK).
• The size, spacing, and formatting of Braille dots should comply with BS EN 15823:2010 (“Braille on packaging for medicinal products”), ensuring readability for visually impaired patients. - Additional information for blind or partially sighted consumers:
• While Braille is limited to the product name, strength, and pharmaceutical form, other key information (e.g., expiry date, batch number, patient information leaflet) is not required in Braille but should be accessible.
• The patient information leaflet (PIL) must indicate how to obtain versions in alternative formats (e.g., large print, audio, or electronic versions), helping ensure compliance with accessibility standards.
• This aligns with the Equality Act 2010, ensuring equitable access to information.
- What must appear in Braille:
Can you describe a high speed packaging line and the controls you would expect to see in place?
High-Speed Blister Packaging Line Process Flow:
1. Blister Pocket Forming:
• PVC or PVC/PVDC film is thermoformed into blister cavities using heat and vacuum or pressure.
2. Tablet Filling:
• Tablets are fed via a hopper system into each formed blister pocket. Vibratory feeders and precision tooling ensure correct positioning.
3. Sealing with Lidding Foil:
• An aluminum foil (pre-printed with product information like drug name, strength, batch number, expiry date) is sealed onto the filled blister using heat and pressure.
4. Perforation and Cutting:
• Blisters are cut into individual packs or strips while preserving printed information (batch/expiry details must remain visible).
5. Cartoning with PIL Insertion:
• Blisters are fed into cartons, and the Patient Information Leaflet (PIL) is inserted automatically.
6. Final QC Sampling and Testing:
• In-process checks and final sampling are conducted to verify product quality and packaging integrity.
⸻
Controls Expected at Each Step:
Pre-Production Controls:
1. Goods-In Inspection & Line Clearance:
• Verification of incoming packaging components (blister foil, cartons, PILs) for correct version and quality.
• Line clearance prior to batch start (to avoid mix-ups) with documented cleaning records.
In-Process Controls:
2. Blister Forming Controls:
• Temperature Control: Monitoring of forming station temperature (critical for proper blister shaping).
• Conveyor Speed Control: Line speed set and monitored to ensure synchronization across forming, filling, sealing, and cartoning.
3. Tablet Filling Controls:
• Camera Recognition System (OCR or Vision System): Verifies correct tablet placement, checks for broken or missing tablets, color consistency, and rejects defective blisters.
• Rejection Station: Automated rejection of incorrect blisters detected by the vision system.
4. Foil Sealing and Printing Controls:
• Sealing Parameters: Heat, pressure, and dwell time monitored for consistent seal integrity.
• Camera System for Foil Inspection: Ensures printed data (drug name, strength, batch number, expiry date) is correct and legible. Checks for misalignment or missing print.
• Cutting Control: Ensures blister cutting does not damage or remove printed foil information.
5. Cartoning and PIL Controls:
• Pharmacode Scanning: Ensures that the correct PIL and outer carton match the blister pack. A mismatch leads to rejection.
• PIL Presence Detection: Confirms PIL insertion into each carton.
6. Weight Check (Checkweigher):
• Ensures that the final packaged product (blister + PIL + carton) meets specified weight criteria. Deviations trigger automatic rejection.
7. Blister Seal Integrity Checks:
• Online Testing (Pin Hole Detection / Pressure Dome Method): Inline systems detect micro-leaks or pinholes.
• Offline Testing (e.g., Methylene Blue Dye Test): Conducted periodically or during validation to confirm seal integrity.
8. In-Process QC Sampling:
• Sampling at defined intervals (beginning, middle, and end of batch) for:
• Blister integrity.
• Print quality.
• Correct assembly (PIL, carton, blister).
• Weight compliance.
⸻
Additional Controls:
• Batch Record Documentation: All critical parameters, deviations, and interventions are recorded in the batch manufacturing record (BMR).
• Alarm Systems: For equipment malfunctions or deviations (e.g., filling errors, seal issues).
• Line Clearance at Changeover: Ensures no mix-up of products or components between batches.
During engineering maintenance 6 pink rogue tablets are found struck in the hopper and the position is just above the web. The maintenance schedule is every 6 months. The first batch manufactured after maintenance is pink tablets and rest of the batches are white tablets.
Immediate Actions:
1. Quarantine:
o Immediately quarantine the line, the current batch in process, and any potentially affected batches produced since the last confirmed line clearance (particularly white tablet batches).
o Place any released batches from this line on hold pending investigation.
2. Deviation Management:
o Raise a deviation promptly to document:
The date and time of discovery.
How and by whom the rogue tablets were found.
Exact location (hopper above the web).
Line and batch details (including pink and white tablet product codes).
o Initiate a full investigation in line with QMS procedures (referencing P22-D-002 for root cause analysis and P22-C-003 for CAPA management).
________________________________________
Investigation Strategy:
- Product Identification & History:
* Confirm identity of the pink tablets:
o Use visual inspection, printed codes, or, if needed, analytical methods (e.g., NIR, Raman spectroscopy) to confirm product.
* Review line usage history for the last 6 months:
o List all products processed, focusing on any pink tablet products.
* Check the line clearance records:
o Any deviations, comments, or issues reported during previous cleans/changeovers.
* Review engineering maintenance reports:
o Any maintenance issues or anomalies reported during last servicing. - Impact Assessment:
a) Regulatory (Annex 16):
* Potential cross-contamination or product mix-up.
* If contamination confirmed, recall may be required, in line with recall SOP (aligned to EudraLex Volume 4 Chapter 8, MHRA defect classification).
* Notify MHRA/DMRC if product defect confirmed.
b) Marketing Authorisation (MA):
* Possible breach of MA specification due to unintended presence of another product.
c) GMP Compliance:
* Possible failure in contamination control strategy (CCS), equipment design, or cleaning/line clearance processes.
* Review equipment qualification (DQ/IQ/OQ/PQ) and CCS alignment (per Annex 1 and ICH Q9/Q10).
d) Patient Safety (ICH Q9/QRM):
* Assess risk of incorrect product ingestion.
* Consider the worst-case scenario (e.g., hypersensitivity reactions to pink tablet ingredients).
* Discuss with QPPV whether any safety signals or adverse event reports exist related to these products.
________________________________________
Further Actions:
1. Reference Samples:
o Review retained reference samples of:
The quarantined batch.
Any released white tablet batches.
o Conduct visual inspection and, if required, analytical testing for presence of pink tablets.
2. Complaints/Pharmacovigilance:
o Review customer complaints for any related reports (misplaced tablets, unexpected effects).
o Consult QPPV for any relevant pharmacovigilance data.
3. Recall Consideration:
o If contamination confirmed, convene recall committee (including Regulatory, Medical, QA, QPPV).
o Perform risk classification (MHRA Class I/II/III) and recall level determination (e.g., patient, pharmacy).
o Communicate with MHRA/DMRC as per the defective medicines reporting SOP.
4. Root Cause Analysis (RCA):
o Apply structured tools (Ishikawa/Fishbone, 5 Whys).
o Investigate:
Personnel: Training and line clearance competency.
Facility/Equipment:
Qualification status of the packaging line.
Performance of tablet detection systems (metal detectors, vision systems):
Conduct challenge tests to verify functionality and ability to detect rogue tablets.
Equipment logs, PPM (Planned Preventative Maintenance) records, cleaning logs.
Documentation:
Review batch records (BPR) for recent batches: any anomalies, line clearance comments.
Process: Validate line clearance and cleaning procedures (align with Annex 1 CCS).
Suppliers/Outsourced: Competency and records of engineering contractors.
Self-inspection: Any findings related to line clearance or cleaning in recent audits.
5. CAPA Implementation:
o Based on RCA findings:
Increase frequency of line inspections or engineering maintenance.
Enhance line clearance verification (introduce additional checks).
Review and retrain personnel on line clearance and reconciliation processes.
Evaluate equipment upgrades (e.g., installing better detection systems).
o Define effectiveness checks to ensure CAPA is sustained.
6. QMS Review:
o Check for trend recurrence in deviations, change controls, PQRs.
o If systemic gaps identified, update SOPs or QMS processes accordingly.
7. Regulatory Closure:
o Submit defect report and closing summary to MHRA/DMRC if required.
________________________________________
Conclusion:
* Classify as a major deviation (potential product contamination).
* Follow through with risk assessment, patient safety evaluation, regulatory reporting, and systemic improvements.
* Ensure QP oversight throughout the process, aligned with Annex 16 and ICH Q10 expectations.
What is CCIT and how do you perform it?
Model Answer:
Container Closure Integrity Testing (CCIT) ensures the container (e.g., vial, prefilled syringe) maintains sterility over shelf-life. Methods include:
• Dye ingress
• Vacuum decay
• Helium leak detection
• High-voltage leak detection (for liquids)
Tips:
• Mention it’s a GMP expectation for sterile products (Annex 1 and USP <1207>).
• Essential for sterility assurance and product quality.
What are critical parameters to validate for a high-speed double-blind packaging line?
Model Answer:
• Speed control: Packaging line speed and reject mechanisms.
• Segregation: IMP vs placebo—prevent cross-mix.
• Line clearance and reconciliation: 100% checks, especially between arms of the study.
• Labeling: Blinded labeling accuracy and verification.
• Vision systems: To detect label presence, orientation, integrity.
• Weighing: If placebo and active differ in weight, use automated check-weighing.
Tips:
• Highlight importance of blinding integrity.
• Mention that validation includes mock runs simulating full study batches.
Can you describe a high-speed packaging line and what controls should be in place?
A high-speed packaging line is an automated system used for the efficient packaging of pharmaceutical products, often involving blister packs, cartons, leaflets, and serialization. It includes both automated and manual controls to ensure correct packaging, labelling, and compliance with GMP.
⸻
Main stages of a high-speed blister packaging line:
1. Foil Feeding and Blister Forming
• Forming foil (e.g. PVC/aluminium) is unwound and heated to create cavities (blisters).
• In-process check: Blister depth, pocket formation, seal integrity
2. Product Feeding (Tablet or Capsule Insertion)
• Product drops from a hopper into formed cavities.
• Vision system checks for:
• Missing product
• Broken tablets
• Double fills
• Colour mismatch
3. Lidding Foil Sealing
• Lidding foil (e.g. aluminium) is heat-sealed onto the filled blister.
• Camera check confirms sealing area, batch number, and expiry date (OCR/OCV)
4. Perforation, Cutting, and Printing
• Blisters are perforated and cut to size.
• Checkweigher confirms overall weight is within tolerance (important for detecting missing tablets or leaflets later)
5. Cartoning Machine (Secondary Packaging)
• Automatic leaflet folding and insertion.
• Blisters are inserted into cartons along with the leaflet.
• Pharmacode readers confirm correct leaflet and carton match.
6. Serialization and Labelling (if required)
• Unique serial number printing for each carton.
• Tamper-evident labels applied.
• Camera checks ensure correct code, placement, and legibility.
7. Reject Station
• Any units failing camera, weight, or code checks are automatically rejected using compressed air or diverter flaps.
⸻
Controls in place:
Automated controls:
• Vision systems for content verification
• OCR/OCV for batch/expiry date recognition
• Pharmacode readers for leaflet/carton ID
• Checkweighers for weight-based rejection
• Reject verification systems
Manual/IPC controls:
• Line clearance before and after batch
• IPC checks at predefined intervals
• Blue dye test (for blister seal integrity)
• Reconciliation of components (e.g. printed materials)
⸻
Regulatory considerations:
• Line validation (IQ/OQ/PQ)
• Batch record documentation
• Serialization per FMD or MHRA guidelines (post-Brexit)
• Audit trail for rejected units
High-Speed Packing Line Controls
High-speed packing lines have both automatic and manual controls:
• Automatic checks: camera vision systems, presence/absence detection (tablets, leaflet, cartons), pharmacode scanning, label verification, OCR for batch and expiry, checkweighers with reject systems.
• Manual IPCs: periodic checks for leak tests (e.g. blue dye), inspection of packaging integrity, label placement, etc.
• Process Flow: Foil feeding → blister forming → tablet filling → top foil sealing → batch/expiry printing → die-cutting → leaflet insertion → automatic cartoning → checkweigher → tertiary packaging.
• Validation considerations: Challenge tests with deliberate defects (e.g. missing leaflet), reject verification, and tight weight tolerance settings — especially for low-mass tablets where leaflet weight may be critical.