Investigational medicinal products Flashcards
(55 cards)
For the following IMP Supply chain, can you explain the requirements:
IMP (DP) QP Certified in Germany - Imported and Stored at GB hub - sent to NHS study site
- Regulatory Framework
The requirements for IMP oversight in this scenario are defined in:
• UK Clinical Trials Regulations 2004 (as amended)
• EU GMP Annex 13 and Annex 16
• MHRA Inspectorate Guidance on IMP Importation (Post-Brexit)
Germany is classified as an approved country for import (listed country) — therefore, QP certification performed in Germany (EU MIA(IMP)) is recognised by the MHRA, but the UK QP retains legal responsibility for verification before release to the trial site.
⸻
- Licensing Requirements
• Germany MIA(IMP) licence must cover manufacture, testing, certification.
• UK MIA(IMP) licence required for importation, storage, and distribution within GB.
• Importation testing (ID test) required at the UK MIA(IMP) site.
• WDA(H) licence would apply only for non-IMPs or unmodified comparators.
⸻
- UK QP Oversight Responsibilities
a) Verification Before Release to NHS Site
UK QP must verify the following before IMP is released to the GB trial site:
• Evidence of EU QP Certification (acceptable forms include):
• QP Certification Statement (Article 62(1) of EU CTR 536/2014)
• Batch Certificate
• Verified access to EU MIA(IMP) ERP system
• Transport temperature log and any deviations managed appropriately.
• UK Clinical Trial Authorisation (CTA) and Ethics approval are in place.
• IMP is only supplied to approved NHS sites listed in the ethics application.
⸻
b) Required Documentation for UK QP Oversight
• Manufacturing and supply chain details.
• CTA form and approval letters (including amendments).
• Evidence that the EU MIA(IMP) site holds a valid GMP certificate (EudraGMDP).
• Shipment details for each batch (addresses verified).
• Temperature excursion reports and assessment records.
• Quality Technical Agreements (QTA):
• Between Sponsor & EU MIA(IMP) holder.
• Between Sponsor & UK MIA(IMP) holder.
• Between Sponsor & GB Storage Hub (if applicable).
Step
Control Measure
Import to GB Hub
Segregate IMP (physically or electronically) until UK QP verification complete.
Storage
Facility must be named on UK MIA(IMP). Ensure GDP-compliant storage conditions.
Shipment to NHS Site
UK QP verification of certification & documentation required before release.
Reference & Retention Samples
Not mandatory to store in GB — but location must be visible to UK QP and defined in the QTA. MHRA must have timely access.
- Additional Considerations
• Deviations (e.g., temperature excursions) must be assessed with documented justification from EU QP and Sponsor, reviewed by UK QP.
• The UK QP can delegate the routine verification task to trained personnel within the MIA(IMP) Quality System but retains ultimate responsibility.
• IMP should not be available for use at NHS trial sites until full verification and confirmation of appropriate QP certification is complete.
What would be the additional requirements or differences if the IMP was imported directly from Germany to NHS study site?
“If IMP is imported directly from the EU manufacturing site to the NHS site without going through a GB storage hub, the UK QP responsibilities remain unchanged. There must be a robust QTA between the Sponsor, UK MIA(IMP) holder, and NHS site to ensure control of the IMP at the clinical site. The UK QP must verify EU QP certification, transport conditions, and approve the IMP for use before administration to patients. The IMP must remain segregated at the NHS site until this approval is received. This ensures compliance with Annex 13, MHRA guidance, and maintains the integrity of the IMP supply chain.
What are the high level changes in the Clinical Trial Regulation?
- Risk-Based Approach to Clinical Trial Approvals
• Introduction of Low-Intervention Trials (Low-Risk Trials):
• Clinical trials using IMPs that are UK-licensed (or EU-authorised if Northern Ireland) and used within their marketing authorisation (SmPC).
• These trials no longer require MHRA Clinical Trial Authorisation (CTA) — only Research Ethics Committee (REC) approval is needed.
• Sponsor responsibility remains to assess IMP risk and notify MHRA if they believe a trial qualifies.
⸻
- Single Application Portal via IRAS
• Clinical trial and ethics applications now submitted together via the Integrated Research Application System (IRAS).
• MHRA and REC conduct a combined review — reducing duplication and streamlining approval processes.
• Single decision letter provided (CTA + Ethics Approval together for standard-risk trials).
⸻
- Clearer Definitions Introduced
More detailed definitions for:
• IMP — Investigational Medicinal Product
• Non-IMP — Non-Investigational Medicinal Product (e.g., rescue medication)
• Auxiliary Medicinal Product (AxMP) — Product used in a trial but not being studied as an IMP (e.g., background treatment, challenge agents).
This provides clarity for sponsors and QPs on what products fall under IMP handling and certification requirements.
⸻
- Flexible Labelling Requirements for Non-IMP Products
• For authorised Non-IMPs or AxMPs (with valid UK or EU marketing authorisation), labelling flexibility is permitted:
• Reduced labelling requirements apply (no need for full Annex 13 clinical trial labelling).
• Particularly applies where product is used within licensed indications.
⸻
- Additional Key Updates
Change
Impact
Streamlined Approval Process
Faster regulatory review timelines (maximum 30-60 days).
Increased Transparency
Requirement for public registration and result reporting of clinical trials (aligns with EU CTR approach).
Focus on Proportionality
Oversight and monitoring expectations scaled to trial risk (risk-adapted approach).
Summary Statement for Viva:
“The recent UK changes aligning with the EU Clinical Trial Regulation introduced a risk-based approach to trial approvals, with low-risk trials using authorised medicines no longer requiring MHRA CTA but still needing ethics approval. The process is now streamlined via IRAS for a single application and combined review. The regulation also clarifies definitions for IMP, non-IMP, and Auxiliary Medicinal Products, and introduces flexible labelling requirements for authorised non-IMPs, reducing regulatory burden for certain trials while maintaining patient safety.”
Wha t is a Imp Protocol, why it is needed, what are the content in it and where will you find it.
What is a Clinical Trial Protocol?
• The Clinical Trial Protocol is a core document that defines how a clinical trial will be conducted.
• It ensures the scientific validity, patient safety, and GCP compliance of the trial.
• It describes the trial design, objectives, methodology, statistical considerations, and operational details.
⸻
Why is the Protocol Needed?
• It provides clear instructions to:
• Clinical investigators (Doctors, Nurses, Pharmacists).
• The Sponsor.
• QA/QP (ensures GMP & GCP compliance).
• Required by ICH GCP E6(R2) and UK Clinical Trials Regulations.
• Allows consistency in trial conduct across multiple sites.
• Used for regulatory approval (MHRA/REC submission).
• Used by the QP during certification to ensure:
• IMP dosage form matches protocol requirements.
• Packaging/Label content aligns with protocol (dosing, patient number).
• Shelf-life/use-by dates support treatment duration.
Section
Description
-Title Page
Study title, protocol number, version, sponsor details.
-Background & Rationale
Summary of IMP, pre-clinical/clinical data.
-Trial Objectives
Primary & secondary objectives.
-Inclusion / Exclusion Criteria
Defines eligible patient population.
-Randomisation & Blinding
Method of assigning patients to treatment arms.
-Treatment Plan
IMP dose, dosing schedule, route of administration, dose titration rules.
-IMP Details
Storage conditions, shelf-life, accountability, labelling requirements (Annex 13).
-Safety Monitoring
Adverse event reporting, stopping rules.
-Statistical Considerations
Sample size calculation, data analysis plan.
-Ethics & Regulatory
Informed consent process, ethical approval.
What is IMPD and PSF, what is the difference in both and why do you need it.
- What is the IMPD (Investigational Medicinal Product Dossier)?
• The IMPD provides quality, non-clinical, and clinical information to support a Clinical Trial Application (CTA).
• It is equivalent to Module 3 of the CTD (Common Technical Document) for commercial products but specific for clinical trials.
IMPD Structure:
• Quality Section includes:
• Active Substance (API) information — manufacture, specifications, stability.
• IMP (Finished Product) information — manufacture, packaging, testing, stability.
• Non-clinical and clinical sections — depending on the development phase.
⸻
- What is the PSF (Product Specification File)?
• The PSF is a GMP document specifically required under Annex 13 of EU GMP (for IMPs).
• It provides all essential quality information that a QP needs to perform certification of IMP batches.
Typical PSF Content:
• IMP composition (formulation).
• Manufacturing and packaging process overview.
• Specifications for starting materials, API, and IMP.
• Storage conditions and shelf-life.
• Primary container information.
• Labelling requirements (per protocol).
• Reference to manufacturing instructions (Master Batch Record).
• Testing methods and acceptance criteria.
Feature
IMPD
PSF
Purpose
Regulatory dossier for CTA submission (MHRA/REC).
Operational GMP document for QP batch certification.
Content
CTD-style quality, non-clinical, clinical data.
GMP practical information for IMP manufacture, testing, storage, labelling.
Regulatory Reference
EU CTD guidance, UK SI 2004/1031, Volume 10.
EU GMP Annex 13, ICH Q7 (for IMP APIs).
Owner
Sponsor regulatory team.
Sponsor Quality Unit / Manufacturer’s QA/QP team.
Document
Why Needed
IMPD
Required for initial and amended CTA submission to MHRA and REC. Demonstrates product quality, safety, and suitability for clinical use.
PSF
Required by Annex 13 for QP certification. Acts as the “Bible” for the QP — contains all batch-specific GMP requirements to ensure IMP is made, tested, and stored appropriately before release.
Summary Statement for Viva:
“The IMPD is a regulatory document submitted to the MHRA as part of the CTA to support the safety, quality, and efficacy of the IMP in a clinical trial. It includes quality information equivalent to CTD Module 3, covering both API and IMP details. The PSF is a GMP operational document required under Annex 13 that provides the QP with all the necessary manufacturing, testing, storage, and packaging information to certify a batch of IMP for use in a trial. Both documents are essential — the IMPD supports regulatory approval, and the PSF supports QP release in compliance with EU GMP and UK SI 2004/1031.”
When you have the PSF why do you need IMPD for certification and vice-versa.
Question:
If you have the PSF (Product Specification File), why do you need the IMPD (Investigational Medicinal Product Dossier) for IMP batch certification? And vice versa?
⸻
Model Answer (QP Viva Style Response):
As a QP, both the PSF and IMPD serve important but different purposes during the certification of an IMP batch — and they complement each other.
PSF (Product Specification File):
• The PSF is site-specific and supports routine GMP compliance for manufacturing and packaging of IMP at the site.
• It contains:
• The manufacturing process as performed at the site.
• Specifications for starting materials (excluding API), excipients, packaging components.
• In-process controls, finished product specifications.
• Approved manufacturing instructions.
• Storage conditions and shelf-life assigned to the batch.
• However, critically, the PSF does not contain API details — particularly:
• The API impurity profile.
• Control strategy for API impurities.
• API synthesis route.
Why is IMPD also required?
• The IMPD contains detailed development information about the product submitted to the Competent Authority (MHRA) and Ethics Committee during clinical trial application (CTA).
• It contains:
• Full API details — synthesis, impurity profile, specifications.
• Justification for specifications based on clinical development stage.
• Stability data and shelf-life justification.
• Data supporting risk-based control strategy.
⸻
Why do I need both for certification?
Document
Purpose for Certification
Example Usage
PSF
Ensures manufacturing is in line with site’s approved GMP documentation
Verify packaging process, release specs, lab methods, labels.
IMPD
Provides essential quality data not in PSF
API impurity profile — critical for assessing OOS or non-conformance. Confirm that the assigned shelf-life and storage is justified.
Example Scenario (to demonstrate understanding in the viva):
“If I encountered an OOS related to an unknown impurity in the drug product, I would need to refer to the IMPD to understand the API impurity profile, synthesis route, and acceptance criteria — this is not available in the PSF.
Conversely, if I were certifying a batch for release, I would use the PSF to ensure compliance to the site’s GMP documentation, e.g., correct packaging instructions, specifications, and testing methods.”
What is the investigation Brochure and whats the content in it. Why is this needed
The IB is a regulatory document prepared by the Sponsor for a clinical trial.
It is used primarily by clinical investigators (doctors, nurses, pharmacists involved in the trial) to understand the safety, pharmacology, and dosing of the Investigational Medicinal Product (IMP).
⸻
Why is the IB needed?
The IB provides essential information to support:
• Safe administration of the IMP to human subjects. • Risk-benefit assessment by investigators. • Training and information for clinical trial staff. • Compliance with GCP (Good Clinical Practice) — ICH E6(R2).
⸻
What is the content of the IB?
The content is similar to an SmPC (Summary of Product Characteristics) — but adapted for a product under investigation (i.e., not yet authorised).
Typical Sections of IB (ICH E6 R2 — Section 7):
Section
Content
Physical, Chemical, Pharmaceutical Properties
API name, formulation, excipients, appearance, storage.
Pharmacology
Pharmacodynamics (PD), Mechanism of Action, Pharmacokinetics (PK).
Toxicology
Preclinical studies, safety data (animal studies).
Clinical Data
Summary of clinical experience — previous trial results, known ADRs.
Posology
Dosing information — route of administration, frequency, special warnings.
Contraindications
Risks, precautions, monitoring parameters.
Safety Monitoring
Adverse event reporting, stopping rules, special precautions.
Why does a QP need to know about the IB?
As a QP, my responsibility is product quality — but I must understand the IB because:
• It guides correct storage and handling conditions of the IMP at site.
• It gives context for risk assessment (e.g., if an excipient has specific handling risks).
• It may be referred to during OOS/OOT investigations (especially if safety impact).
• It provides awareness of the intended dosage and route of administration (important for labelling checks and release).
The requirement of IMP labels, how it is different wrt commercial products. What are the contents, where will you find the details wrt UK and EU.
- the imp labels have some apecirfic informations such as
- trial use only, trial refer MDR number, pi, sponsor. Cro imformation.
- the requirements described in annex 13.
- as uk has deepened from EU, uk still adopte eu 2001/20/ec and old version of annex 13.
Eu follow new annex 13 in line with 536/2014.
What system of wordings would you use to prevent unblinding?
Labelling Controls:
Control Measure
Purpose
Identical Label Design
Same label material, font style, font size, layout, spacing.
Use of Neutral Code or Batch Number
Avoid product-specific identifiers visible to patients/investigators.
Pre-approved Label Text
Approved via PSF and Sponsor to ensure blinding maintained.
Blinding Code Management
Controlled via Sponsor’s code break procedure, separate from manufacturer.
GMP Labelling Process Controls:
GMP Control
Purpose
Line Clearance
To prevent mix-up of active/placebo labels/components.
100% Visual Inspection
For manual labelling — each unit checked by trained operators.
Automated Camera Inspection
On high-speed lines — vision systems to verify label presence, correctness, and alignment.
Label Reconciliation
Ensure 100% accountability of issued vs. used labels — critical GMP requirement.
Your company informs you that they want to transfer their R&D unit next to your manufacturing site. What do you need in place?
As a QP, I would first raise a Change Control (CC) to assess the potential regulatory, GMP, and product quality impacts of integrating the R&D unit near or within the manufacturing site.
Step 1: Impact Assessment
a) Regulatory Impact — MIA(IMP) Licence
• Confirm what type of activities R&D will perform:
• Non-GMP R&D activities? → No licence impact if fully segregated.
• GMP-related activities? (e.g., manufacture of IMP analytical testing) → May require:
• Licence variation to existing MIA(IMP).
• New MIA(IMP) licence if imp manufacturing .
b) GMP Impact Assessment
Assess risk to existing GMP operations, focusing on:
Area of Consideration
Questions to Ask
GMP Risk
Shared Areas/Equipment
Will any rooms, equipment, or HVAC be shared?- Risk of cross-contamination or mix-up.
Personnel Flow-Will R&D staff enter GMP areas?- Need GMP training and restricted access control.
Material Flow-Will materials move between R&D and GMP?
-Risk of misidentification or contamination.
c) Finished Product Impact
• Assess whether the R&D activities could have any impact on product quality or patient safety — e.g., contamination risks, data integrity, unintentional use of R&D material.
⸻
Step 2: Control Measures and Mitigations
Regulatory Controls:
• Licence variation application to MHRA — if required.
• Update Site Master File (SMF).
• Define scope of QP responsibilities — e.g., IMP QP release oversight if R&D is manufacturing IMPs.
GMP Controls:
Ensure QMS is in place to control the interface between R&D and GMP operations:
Control Measure
Purpose
CCS (Contamination Control Strategy)
Address risks from shared facility or utilities.
Cleaning Validation/Verification
For any shared equipment or facility.
Segregation
Physical separation of R&D and GMP activities.
SOPs for Shared Areas
Define roles, responsibilities, and cleaning requirements.
GMP Training for R&D Staff
Especially if they enter GMP areas or handle GMP equipment.
Environmental Monitoring Review
Ensure EM regime covers any impacted areas. Update EM strategy if required.
Final QP Summary Statement:
“As a QP, my role is to ensure that any integration of R&D activities does not compromise GMP compliance or patient safety. I would ensure a robust risk assessment is performed, regulatory requirements are clarified (including licence variation if required), and that appropriate controls — including CCS, cleaning validation, segregation, and GMP training — are implemented to mitigate any potential risks to product quality.”
What are the differences between commercial and IMPs?
- Legislation / Regulatory Framework
Product Type
UK Legislation
Notes
Commercial Product
UK SI 2012/1916 (Human Medicines Regulations 2012)
Governs manufacturing, MA, distribution, sale of licensed medicines.
IMP (Investigational Medicinal Product)
UK SI 2004/1031 (Medicines for Human Use Clinical Trials Regulations 2004)
Governs clinical trial manufactuing
CTA approval process.
Product Type
Regulatory Approval
Responsible Body
Commercial
Marketing Authorisation (MA)
MHRA (UK) or EMA (EU)
IMP
Clinical Trial Authorisation (CTA) + Ethics Committee Approval
MHRA (CTA) + REC (Research Ethics Committee)
- GMP Application
Product Type
GMP Expectation
Notes
Commercial
Full GMP per EU GMP Vol 4 (all chapters and annexes apply)
Mature, validated processes.
IMP
Phase-Appropriate GMP per Annex 13 and ICH Q7/Q10 principles
Flexibility allowed based on risk and trial phase — but patient safety always paramount.
- Batch Size / Manufacturing Scale
Product Type
Batch Size
Reason
Commercial
Large-scale
Market demand, cost efficiency.
IMP
Small-scale
Limited to clinical trial need, often bespoke or patient-specific (e.g., ATMPs).
- Labelling Requirements
Product Type
Label Requirements
Governing Regulation
Commercial
Per MA and SmPC
HMR 2012 / EU Labelling Directive.
IMP
Annex 13 (UK) or EU CTR Annex VI
Includes trial-specific information — protocol number, “For clinical trial use only”, Sponsor details. - QP Role and Release
Product Type
QP Release
Notes
Commercial
QP certifies each batch per MA requirements.
Full compliance to MA dossier.
IMP
QP certifies each batch per CTA & GMP.
Additional responsibility to ensure trial-specific conditions (protocol, label, blinding) are met.
Is GMP for API required for IMPs?
GMP for APIs in Investigational Medicinal Products (IMPs)
Relevant Guidelines and Regulations (EU & UK)
When manufacturing an Investigational Medicinal Product (IMP), both EU and UK regulations require that the IMP be made under Good Manufacturing Practice (GMP). However, specific guidance addresses how GMP applies to the Active Pharmaceutical Ingredient (API) used in IMPs:
• ICH Q7 (EU GMP Part II) – This is the international GMP guide for APIs. Section 19 of ICH Q7 provides guidance specifically for APIs intended for clinical trials  .
• EU GMP Annex 13 – This annex (Manufacture of IMPs) outlines GMP expectations for investigational products. It acknowledges differences from commercial production and emphasizes certain controls (e.g. for sterile products) .
• MHRA (UK) Guidance – The UK’s MHRA aligns with EU principles. MHRA explicitly states that while full compliance with GMP Part II (ICH Q7) is not strictly required for IMP APIs, the IMP manufacturer must ensure the API is of appropriate quality . In other words, even if an IMP API isn’t made under full commercial API GMP, there is still an obligation to have it made under appropriate controls.
Phase-Appropriate GMP for IMP APIs
Phase-appropriate GMP means that the stringency of GMP controls should correlate with the stage of development:
• Early-phase clinical trial materials are produced on a small scale with evolving processes. Regulators do not expect these production processes to be fully validated as they would be for a marketed product . This flexibility acknowledges practical constraints during development.
• ICH Q7 Section19 confirms that not all GMP controls for commercial APIs are applicable to investigational APIs. Instead, controls should be appropriate to the stage of development of the drug . For example, impurity limits or process validation steps may be less defined in Phase I than by Phase III.
• Annex 13 (EU) reinforces this principle. It states: “Production processes for investigational medicinal products are not expected to be validated to the extent necessary for routine production, but premises and equipment are expected to be qualified.” . This means basic GMP infrastructure (qualified equipment, trained personnel, documentation, etc.) must be in place even if the process is still being optimized.
• In the United States (FDA), a similar phase-based approach exists (e.g. Phase1 investigational drugs are exempted from certain GMP requirements), but under EU/UK rules all IMP manufacturing is governed by GMP – with allowances for development stage.
When Full API GMP is Expected (Sterile or High-Risk IMPs)
There are situations where full GMP controls for the API are expected even in clinical trial manufacture:
• Sterile Products: If the IMP or its API is sterile or intended for a sterile dosage form, full sterile GMP standards apply to the API stage as well. Annex13 makes clear that for sterile investigational products, sterilization processes must be validated to the same standard as for marketed products . Likewise, if an API is manufactured and supplied as sterile, it must comply with EU GMP Annex1 requirements for sterility assurance . There is no relaxation of sterility requirements due to clinical trial status.
• High-Risk Routes of Administration: For IMPs administered via high-risk routes (e.g. intravenous injections, intrathecal, inhalation), regulators expect stringent GMP controls on the API. The safety margin is low in these cases, so the API’s purity, sterility (if applicable), and consistency should be ensured with near-commercial level rigor. In practice this means applying full or near-full GMP (e.g. thorough impurity control, robust cleaning, validated aseptic handling if needed) even in early trials.
• Biological APIs and ATMPs: For biologics or Advanced Therapy IMPs (gene therapies, cell therapies), full GMP from the earliest steps is required. MHRA notes that for biological/ATIMP APIs, EU GMP Part I/IV applies to all manufacturing steps from the cell bank or vector onward . Early steps like cell culture or gene vector production are critical to the final product, so they must be under GMP control as if they were part of a licensed process. (This is outlined in EU GMP Annex2 and PIC/S Annex2A for ATMPs.)
• Later Phase Trials: By Phase III (or when an IMP is approaching marketing), expectations approach full commercial GMP for both API and finished product. Any scale-up or process changes are tightly managed under change control, and the API manufacturing process should be essentially validated. While not explicitly “required” by law until marketing, a Phase III IMP API would be held to a very high GMP standard to ensure a smooth transition to commercialization.
QP Assessment of API Quality and GMP Compliance
Each batch of an IMP must be certified by a Qualified Person (QP) before release for a clinical trial. The QP must assure that all materials, including the API, have been produced in compliance with appropriate GMP and the trial authorization. Key points on how a QP assesses API controls:
• Supplier Qualification: The QP will verify that the API supplier/manufacturer has been appropriately qualified. This typically includes auditing the API manufacturing site or reviewing audit reports to confirm they follow ICH Q7 principles (Section19) for clinical trial materials . Even if full PartII GMP certification isn’t in place, there should be evidence of a quality system and GMP-like controls at the API site.
• Documentation Review: The QP reviews the Certificate of Analysis (CoA) and specifications of the API against the IMP dossier (IMPD/CTA file). They ensure that the API meets the purity, identity, and quality parameters expected. Any anomalies in analytical results or impurity levels must be understood and justified before proceeding.
• GMP Declaration (Import of API): If the API (or IMP manufacture involving the API) is sourced from outside the UK/EU, the QP needs to pay special attention to its GMP status. For IMPs, formal API import authorizations and registrations are not required as they are for commercial APIs . However, when an IMP’s supply chain includes manufacturing sites outside the EU/UK, a QP declaration is usually required in the clinical trial application. The QP must attest that those non-EU sites comply with standards equivalent to EU GMP . This often means the QP has a declaration letter on file (or as part of the CTA) confirming the API was made under acceptable GMP.
• Risk Assessment: The QP uses a risk-based approach (as per ICH Q9 principles) to judge if the level of controls on the API is commensurate with its criticality. For example, if an API has a novel synthesis with potential toxic impurities, the QP will ensure enhanced controls or testing are in place to mitigate patient risk. The MHRA expects QPs to exercise “due diligence in understanding the risks to the product and subject/patient” when assessing an IMP supply chain .
• Additional Testing or Controls: If the API’s manufacturing data are limited or GMP controls were light (common in very early development), the QP might require additional measures before certification. This could include additional testing of the API or drug product for impurities, sterility, or endotoxins, tighter release specifications, or a technical agreement with the supplier to address any gaps.
• Compliance with CTA/IMPD: Importantly, the QP ensures that everything stated in the Clinical Trial Authorisation (CTA) or IMP dossier about the API’s manufacture and quality has been followed. Any deviation from the described process or specifications would need regulatory notification or amendment. The QP will check that the current IMPD (including any amendments) is on hand and that the API’s details in it (such as manufacturer, synthesis route, grade, etc.) match reality.
The QP’s overall goal is to be confident in the API’s fitness for human use before allowing the IMP to be used in trial subjects. They will document their assessment as part of batch certification records. In summary, the QP will not certify the IMP batch unless satisfied that the API was made under suitable GMP controls and its quality is verifiably acceptable .
Summary – Viva-Style Response
Question: “Is GMP for APIs required in the manufacture of IMPs?”
Answer: In principle, yes – GMP control is required for an IMP’s API, but in a “phase-appropriate” manner. For early-phase investigational products, full ICH Q7 compliance for the API is not strictly mandatory, but the API must be manufactured under appropriate GMP measures to ensure quality . Key guidelines like ICH Q7 Section19 and EU GMP Annex13 acknowledge that requirements can be scaled to development stage. For example, initial batches might not have fully validated processes, except in critical cases like sterile products where full GMP standards (e.g. sterile process validation per Annex1) are expected even for IMPs . A Qualified Person will review the API’s source and quality for each IMP batch – making sure the API was made by a suitably qualified manufacturer with proper controls, even if not holding a formal GMP certificate. Ultimately, the QP must be satisfied that the API’s purity, potency, and safety are assured by adequate GMP before certifying the IMP batch for trial use. This approach ensures subject safety while permitting flexibility during drug development.
(UK/EU Perspective: Both UK and EU authorities follow these principles – the MHRA and EMA expect appropriate GMP for IMP APIs, with more stringent controls applied as risk increases, aligning with the EU and national GMP guidelines.) 
IMP sent to Canada clinical trial site, wrong IMP sent to UK IMP instead, what do you do?
QP Viva Model Answer — Wrong IMP Sent to Canada (UK-labelled IMP Sent)
Immediate Action:
1. Raise Deviation / Non-Conformance.
2. Quarantine affected stock:
• At Canada clinical site (prevent administration).
• At UK site (prevent further distribution).
3. Notify Sponsor immediately — as they are responsible for the clinical trial conduct.
⸻
Impact Assessment:
a) Regulatory Impact:
• Check CTA/IMPD for both UK and Canada — IMP may not comply with Health Canada labelling requirements (e.g., language, storage conditions, regulatory statements).
• Confirm applicable national law for IMP use in Canada (Health Canada GCP/GMP guidance).
• If product labelling does not meet Canadian CTA — product may be non-compliant for use without relabelling.
⸻
b) GMP Impact:
• Distribution error — failure in segregation, shipping verification, or labelling control.
• Check PSF/SOPs for dispatch controls — what failed?
• Review whether IMPs for multiple countries are stored together without sufficient segregation.
⸻
c) Patient Safety Impact:
• Assess risk of unblinding:
• Are there visible differences between UK and Canadian labels?
• Are critical label elements (strength, instructions) affected?
• Check if product has been administered:
• If yes — assess clinical risk and notify Sponsor/Regulator.
• If no — quarantine and plan for recovery or relabelling.
⸻
Corrective & Preventive Actions (CAPA):
Immediate Controls:
1. Quarantine affected stock in Canada and UK.
2. Review Canadian label requirements — if labels meet essential requirements (dose, storage, expiry), product may still be used after risk assessment and Sponsor agreement.
3. Communicate with Sponsor — they are responsible for regulatory communication with Health Canada.
⸻
If No Patient Risk:
• Sponsor may authorise use under deviation, with documentation and risk justification.
• Deviation investigation to be completed with root cause analysis (RCA) and CAPA.
⸻
If Patient Risk Identified:
• Consider recall of affected stock.
• Relabelling must be performed at a licensed MIA(IMP) site.
• Relabelled product must undergo full GMP controls — line clearance, label reconciliation, and final QP certification before release.
• Report to MHRA Defective Medicines Report Centre (DMRC) — as required by GMP Chapter 8 if a recall is initiated.
⸻
Preventive Actions:
• Review and strengthen distribution controls:
• Segregation of IMPs by country.
• Verification of shipping labels against destination country.
• SOP update for international supply chain controls.
• Additional GMP training for staff.
⸻
Final QP Statement for Viva:
“As a QP, my priority is to ensure patient safety, GMP compliance, and regulatory adherence. Wrong IMP distribution is a significant deviation that must be investigated thoroughly. Depending on the outcome of the risk assessment and regulatory consultation, the product may be used under deviation or may require recall and relabelling under GMP controls. I would not certify any further batches until robust CAPA is in place to prevent recurrence.”
You work in a facility and they are packing aspirin, however they want to do a clinical trial and they want to pack in the same line IMP and placebo, what do you need? What are the risks ?
Initial Action:
As a QP, I would initiate a formal Change Control to assess the regulatory, GMP, and patient safety impact of introducing IMP (aspirin) and placebo packaging into an existing commercial packaging line.
⸻
- Licence Considerations:
• Confirm whether the site holds an MIA(IMP) licence.
• If the site currently operates under MIA (commercial), packaging IMP requires:
• Licence variation to add IMP activity; or
• New MIA(IMP) licence.
• IMP QP certification will be required for clinical trial material release — this must be defined in the licence.
⸻
- GMP Impact & Risks:
Key GMP Risks:
Risk
Impact
Controls Required
Cross-contamination (aspirin is pharmacologically active)
Potential patient harm — allergic reactions or unintended dosing.
Cleaning validation / cleaning verification. Product contact surfaces must be adequately cleaned between aspirin IMP and other products.
Mix-up between IMP and Placebo
Risk to trial integrity and patient safety.
Strict segregation during packaging — physical or procedural. Label controls to Annex 13 expectations. 100% visual inspection / automated checks.
Risk of Unblinding
Double-blind trial risk.
Identical presentation of IMP and placebo — same label material, font, layout, packaging.
- Patient Safety Risks:
• Aspirin is known to cause allergic reactions in sensitive individuals — therefore, cross-contamination controls are critical.
• IMP must be packaged to prevent any possible unblinding — strict adherence to GMP Annex 13 labelling controls.
⸻
- Control Measures Required:
Regulatory Controls:
• MIA(IMP) licence in place (or variation submitted and approved).
• QP oversight and certification of each IMP batch before release.
GMP Controls:
• Review and update Contamination Control Strategy (CCS):
• Include specific controls for packaging IMP and placebo.
• Segregation of materials (active vs placebo).
• Cleaning validation of the line for aspirin removal — with acceptance criteria based on toxicological risk assessment (PDE/EMA guideline).
• Line clearance procedures enhanced — GMP Chapter 5 compliance.
• Label controls per Annex 13 — trial-specific details, sponsor info, randomisation code protection.
• Staff training on GCP awareness and IMP handling.
⸻
Additional Controls:
• Goods-in checks for IMP materials (aspirin and placebo).
• Batch documentation updated to reflect IMP-specific requirements.
• Quality oversight of blinding control measures — check label proofs, ensure placebo and active appearance are identical.
⸻
Final QP Summary Statement for Viva:
“As a QP, I would ensure that before packaging any IMP and placebo on a commercial line, a thorough change control process is completed, the site holds the correct MIA(IMP) licence, and that robust GMP controls are in place to prevent cross-contamination, mix-up, or unblinding. The patient safety risk from aspirin necessitates a stringent contamination control strategy, validated cleaning, and strict segregation of materials and product. I would not certify any IMP batch until all controls, documentation, and quality oversight meet GMP and regulatory expectations.”
A sterile IMP product having a light yellow colour solution compared to previous batch.
*Aseptic technique/terminal sterilisation
* Depyrogenation technique
* PSF
* Single blinded/double blinded
What are the challenges for manufacturing of IMP products?
As a QP, manufacturing Investigational Medicinal Products (IMPs) presents several challenges compared to commercial products — due to limited product knowledge, evolving processes, and clinical trial-specific requirements.
⸻
- Blinding Control and Randomisation
IMPs often require double-blind or placebo-controlled designs — therefore, manufacturing controls must prevent unblinding:
Challenge
Control Measures
Maintaining blind integrity
Use of randomisation codes, independent blinding team, blinded packaging SOP.
Label controls
Compliance with Annex 13 (UK) / EU CTR Annex VI — neutral labels, consistent design for active/placebo.
Risk of mix-up
Strict line clearance, segregation of IMP vs placebo, label reconciliation, 100% visual inspection.
- Phase-Appropriate GMP and Manufacturing Scale-Up
Challenge
QP Consideration
Small batch sizes in early phase
Greater variability, manual processes, risk of operator error.
Scale-up in later phases
Changes to process, equipment, API manufacturer, excipients — requiring comparability assessment and possible re-validation.
Manufacturing flexibility
Annex 13 allows phase-appropriate GMP — but patient safety must never be compromised.
- Limited Quality Data Availability
Challenge
Impact for QP Decision Making
Limited stability data
Shorter shelf-life assigned, need to monitor stability closely during the trial.
Incomplete impurity profile (API or product)
Requires risk-based approach when assessing OOS or OOT results.
Developing process knowledge
Manufacturing changes may occur during clinical development — need for clear change control and regulatory updates (amend CTA if required).
- Supply Chain Complexity and Distribution Challenges
Challenge
GMP Controls
Global clinical trials
Need to comply with multiple regulatory labelling requirements (different countries).
Temperature-sensitive IMP
Supply chain must ensure transport and storage conditions maintained (GDP principles).
Small stock availability
Risk of product shortage if batch failure occurs — may require expedited deviation assessment or extension of shelf-life based on available stability data.
“Manufacturing of IMP products presents unique challenges due to the need to maintain blinding, comply with phase-appropriate GMP, manage evolving manufacturing processes, and make quality decisions based on limited data. As a QP, I must ensure patient safety and product quality at all times — using risk-based assessment, ensuring robust GMP controls, and maintaining oversight of the supply chain and manufacturing changes throughout the product’s clinical development lifecycle.
You’re packaging a blinded, randomised IMP, what extra considerations are there?
o What is needed on the packaging line to ensure no mix ups?
o What should you put on the packaging to ensure there is no cross contaminatin?
Main Risks:
• Risk of unblinding — e.g., mix-up of active vs placebo, differences in appearance, labelling, packaging configuration.
• Risk of cross-contamination — between active and placebo during packaging operations.
⸻
Controls on the Packaging Line (Prevent Mix-ups & Unblinding):
1. Goods-In Checks:
• Ensure correct receipt of placebo and active materials.
• Visual checks and QC verification for appearance similarity (if intended to be visually identical).
2. Segregation & Physical Separation:
• Segregated storage and staging areas for active vs placebo materials pre-packaging.
• Dedicated line or campaign approach (usually placebo first, then active).
3. Randomisation Control:
• Randomisation code securely controlled and approved prior to packaging.
• Blinded staff vs unblinded staff segregation — only designated unblinded personnel handle randomisation data.
4. Line Clearance & Checks:
• Full line clearance between placebo and active packaging operations.
• Cleaning verification and documentation before re-use of equipment or areas.
5. Staff Controls:
• Specific training on blinding risk and procedures.
• Restricted access to blinded areas.
• Use of CCTV where appropriate (for IMP handling or re-labelling activities).
6. Label Controls:
• Labels must not indicate treatment arm.
• Consider use of identical label stock, font, text layout, batch coding formats across arms.
7. Reconciliation:
• 100% reconciliation of labels and IMP units.
• Any discrepancy is a potential critical deviation.
8. Periodic QA Oversight:
• Line audits during packaging.
• QA review of packaging records, reconciliation, deviation management.
⸻
Controls to Prevent Cross-Contamination:
• Cleaning validation of equipment (if re-used for placebo/active).
• Cleaning verification swabs or visual inspection between product changes.
• Use of closed packaging lines where possible.
• Environmental monitoring during packaging of aseptic products (if applicable).
• Manufacturing order — generally placebo first (less risk of carryover), then active.
• Prevent cross-contamination via labelling material — ensure no loose labels or adhesive transfer.
⸻
Final Considerations as a QP:
• Review and approve packaging protocols and batch records.
• Confirm segregation, cleaning, line clearance, reconciliation, and randomisation controls have been applied.
• Any deviation — assess potential unblinding or cross-contamination risk prior to certification.
• Ensure robust technical/quality agreements if packaging is outsourced (e.g., CMO).
⸻
This structured response demonstrates:
• Risk identification.
• GMP controls.
• QP oversight duties.
• Reference to typical Annex 13 / IMP guidance expectations.
Its placebo controlled. How would you ensure blinding?
Excellent points — here’s a polished model answer suitable for QP viva style for the question:
⸻
Model Answer:
It is a placebo-controlled clinical trial — how would you ensure blinding of IMP during manufacture, packaging, and supply?
⸻
Risks:
• Risk of unintentional unblinding during manufacture, packaging, or at the clinical site due to differences between active and placebo products.
⸻
Controls to Ensure Blinding:
- Randomisation & Blinding Strategy
• Randomisation code generated by Sponsor or 3rd party (e.g., CRO).
• Access to randomisation code restricted to unblinded personnel only (not to packaging operators or clinical site staff).
• Randomisation list securely stored, controlled, and version-controlled.
⸻
- Appearance Matching of IMP
• Active and placebo must be visually indistinguishable (unless justified in protocol).
• Checks at Goods-In stage to confirm matching appearance (size, shape, colour, weight, packaging configuration).
⸻
- Labelling Controls
• Label content must not reveal treatment allocation.
• Label design (text, format, font) should be identical between active and placebo.
• Annex 13 compliant labelling — no product-specific identifiers unless required by protocol.
⸻
- Segregation of Materials
• Physical or electronic segregation of active vs placebo materials during storage, packaging, and handling.
• Controlled access to unblinded materials if required (e.g., QC samples).
⸻
- Packaging Line Controls
• Line clearance between different operations (e.g., active packaging vs placebo).
• Cleaning verification if shared equipment.
• Operator training on blinding procedures and criticality.
• Visual checks post-cleaning and prior to start of next batch.
⸻
- Full Reconciliation
• 100% reconciliation of:
• Printed labels.
• Packed IMP units (active and placebo).
• Rejected/destroyed materials.
• Any discrepancy is a potential critical deviation and must be investigated.
⸻
- QA Oversight
• QA checks during packaging operations to verify blinding controls.
• Periodic audit of blinding processes (Sponsor oversight or self-inspection).
What is needed on the packaging line to ensure no mix ups
GMP Risk:
High-speed automated packaging lines increase the risk of mix-up (wrong component, wrong product, mislabelling).
Especially critical for products like IMPs, where patient safety and trial integrity are impacted.
Key Controls on the Packaging Line:
- Line Clearance (Annex 15 requirement)
Comprehensive line clearance before starting packaging:
Removal of all materials from previous product/campaign.
Visual inspection of equipment, conveyors, printers, and collection areas.
Documented verification by trained operator and QA check.
Critical GMP step — prevents cross-contamination and mix-up.
- Goods-In Check of Packaging Components
Verification of correct receipt of packaging components (labels, leaflets, cartons).
Appearance check — correct batch-specific details (e.g., variable data, expiry date, batch number).
Cross-check against Bill of Materials (BOM) and packaging order.
- Camera Recognition / Vision Systems
Automated in-line vision systems used for high-speed lines to detect:
Correct label application.
Barcode/2D Data Matrix verification.
Pharmacode checks for correct carton orientation.
Rejection of defective or non-conforming units.
Provides real-time rejection and prevents mispackaging.
- Pharmacode / 2D Barcode Control
Pharmacode (colour bars or numerical code) used to verify correct printed component during operation.
2D Data Matrix or GS1 barcode used for product traceability and verification.
Prevents component mix-up during packaging at high speed.
- QP Sampling (If Applicable)
Manual or automated sampling of packed product for visual verification and testing (e.g., leaflet presence, correct assembly).
Part of batch certification evidence.
May be done at in-process or post-packaging stage depending on the process.
Other Supporting Controls:
Segregation of components and finished goods.
Reconciliation of all packaging components at batch end (Annex 13 & Annex 15).
Operator training on criticality of GMP controls.
Line-specific procedures and validated automated systems.
Summary Statement for Viva:
“On a high-speed packaging line, controls to prevent mix-up include full line clearance, goods-in checks of packaging materials, use of automated vision systems and camera recognition for label and code verification, pharmacode or 2D barcode controls, and reconciliation of packaging components. Additionally, QP sampling may be performed for verification. All these controls work together within a GMP quality system to prevent packaging errors and protect patient safety.”
What should you put on the packaging to ensure there is no cross contamination?
MP Risk:
Cross-contamination on a packaging line can occur due to:
Residual product from previous batch.
Incorrect material (e.g., leaflet, carton, label).
Operator error.
Poor cleaning or ineffective line clearance.
Controls to Prevent Cross-Contamination on a Packaging Line:
- Line Clearance
Mandatory GMP requirement (Annex 15).
Removal of all previous product materials (labels, cartons, leaflets, product).
Visual verification of cleanliness.
Documented check by trained operators and QA.
No start of new batch until line clearance is signed off.
- Cleaning Validation & Cleaning Certification
Cleaning Validation:
Validated cleaning procedure to demonstrate removal of product residues and particles to acceptable limits.
Cleaning Certification:
Cleaning records signed and verified before start of next batch.
Includes verification of equipment, contact parts, conveyor belts, and floors.
- Goods-In Check of Materials
Verification that only correct materials are received for the batch.
Packaging components segregated and controlled.
Visual appearance check to prevent material mix-up.
- Material Reconciliation
100% reconciliation of:
Labels.
Leaflets.
Cartons.
Blisters/bottles.
Any discrepancy is a potential critical deviation.
- Staff Training
GMP training for all packaging operators:
Importance of preventing cross-contamination.
Following line clearance and cleaning procedures.
Reporting any deviation immediately.
- Periodic Audit & CAPA System
Routine self-inspection of packaging areas.
CAPA system to address any findings from deviations, complaints, or audit.
Review of cleaning and line clearance practices.
- CCTV (Optional, Site Dependent)
CCTV monitoring may support GMP compliance and security.
Not mandatory — but useful for:
Oversight of critical GMP activities.
Investigation of deviations or complaints.
What is double blinded study? What controls you would expect to see in primary packing of such type of trial? How would you ensure that blind is maintained throughout the trial? What documentation you would require? Q
QP Viva Model Answer — Double-Blind Study & GMP Controls for IMP Primary Packaging
- What is a Double-Blind Study?
A double-blind study is a clinical trial design where neither the patient nor the clinical investigator knows whether the subject is receiving the active product or the placebo.
• This is to prevent bias in the trial results and maintain scientific integrity.
⸻
- What GMP Controls Would I Expect During Primary Packaging of a Double-Blind IMP?
As a QP, I would expect strict controls during packaging to prevent unblinding or product mix-up:
GMP Control
Purpose
Line Clearance
To ensure packaging line is free from previous materials (per GMP Chapter 5).
Goods-in Check
Verification of correct receipt of both active and placebo materials.
Material Segregation
Physical and procedural segregation of active and placebo during packaging.
Use of Randomisation Code
Blinding maintained by assigning unique identifiers or codes linked to treatment allocation — managed by an independent function (not manufacturing team).
Label Controls
Labels must be identical in appearance (material, font, layout) for both active and placebo — in line with Annex 13 requirements.
100% Visual Inspection
For manual packaging — ensure correct label applied, verify integrity of blinding.
Automated Camera Systems
On high-speed lines — to detect presence, correctness, and positioning of labels.
- How Would I Ensure Blind is Maintained Throughout the Trial?
• Ensure an effective blinding SOP is in place — describing roles, responsibilities, randomisation code handling, and blinding control measures.
• Randomisation code list should be securely stored — access-controlled and only available to unblinded personnel (e.g., Sponsor or designated person).
• Define procedures for code break in medical emergency — available at clinical sites but controlled.
• Robust QA oversight — verification of label proofs, randomisation schema, packaging batch records.
• Control of any post-packaging activities — e.g., relabelling, returns handling, reconciliation, destruction — to ensure blinding is not compromised.
• Ensure clear documentation trail is maintained in the Trial Master File (TMF).
⸻
- What Documentation Would I Require as a QP?
Document
Purpose
Randomisation Code / Blinding List
To confirm control of treatment allocation — should be maintained by unblinded personnel.
Blinding SOP
To ensure clear control strategy is defined.
Packaging Batch Records
Including line clearance record, label reconciliation, 100% inspection record.
Certificate of Analysis (CoA)
For both active and placebo — confirming they meet specifications.
Approved Label Text
As per PSF and CTA submission — in line with Annex 13 requirements.
Deviation Reports
If any deviation occurs during packaging or blinding process — full impact assessment required.
Final QP Closing Statement for Viva:
“As a QP, my role is to ensure that the blinding integrity is maintained throughout the packaging, storage, distribution, and use of the IMP in the clinical trial. I would expect to see robust GMP controls in place, appropriate documentation available for review, and clear procedures for managing randomisation codes and potential code breaks. Any deviation with a risk of unblinding would require thorough investigation and Sponsor involvement before I could certify the batch for release.”
Scinario: You’re are a QP at an IMP site. You have noticed that your example pack in your Batch Packaging Record has a carton with label with information missing. Would you release the batch? (about 15 mins)
No, I would not proceed to certify the batch without further investigation, because IMP labelling must comply with Annex 13 requirements (UK) or EU CTR Annex VI, depending on the regulatory framework.
As a first step, I would clarify: What specific information is missing on the label?
IMP labelling is critical to ensure patient safety, regulatory compliance, and trial integrity. Missing information such as product name, strength, storage conditions, protocol number, or mandatory statements like ‘For Clinical Trial Use Only’ would be considered a GMP-critical issue.
Therefore, I would raise a deviation, quarantine the batch, and investigate fully before making any decision to release.”
Follow up: ap. The information “for clinical trial use only” missing
Follow-up ap — The Information “For Clinical Trial Use Only” is Missing
Model QP Viva Answer:
“This statement is a mandatory label requirement under Annex 13 for IMPs in the UK — or EU CTR Annex VI for EU trials. It is critical because it identifies the product as investigational and helps prevent its use outside the clinical trial.
Finding this missing triggers a significant GMP concern — therefore, I would:
Actions:
1. Open a deviation immediately.
2. Conduct a full impact assessment, covering:
Area
Impact
Licence / Regulatory
Breach of Annex 13 label requirement — may not comply with approved label (lavender copy / PSF).
GMP
Failure of label verification process — risk of repeat across batches if systemic issue.
Patient Safety
High risk — without the statement, product could be mistaken for a commercial product, increasing risk of incorrect administration or misuse.
Next Step:
“I would review whether this error is isolated to the example pack or if it has affected other product in this or other batches. Until the investigation is complete and product is confirmed compliant, I would not release the batch.”
Follow up: aq. You have found this information missing on the pack in BPR itself.
“This raises concern about either a printing error or labelling process failure. My next steps would be:
• Quarantine the batch.
• Review batch documentation fully.
• Physically inspect the entire batch on the shop floor.
• Confirm if this is an isolated documentation error (dummy carton) or an actual GMP failure affecting released stock.”