Pharmaceutical microbiology Flashcards
(126 cards)
FTell me the mfg process for ATMP?
- Apheresis: Cell Isolation from patient (autologous) or donner (Allogenic)
- Cell isolation
- Genetic modification (Transduction) with virus vector
- Cell expansion
- Harvest and Formulation
- Cryopreservationan preservation and storage
What are the CCPs in ATMP manufactuig?
- Apheresis - patient/donor identity, transport time/condition, documentation of chain identity
- Transduction - transduction condition (Multiplicity of infection, time, temp)
- Vector Identity, potency, sterility
- Absence of replication-competent virus - Cell Expansion - Culture condition (Temperature, pH, oxgen, co2)
- EM (Grade A/B)
- Monitoring of Growth kinetics and contamination - Formulation and Cryopreservation
* Why critical: Affects product stability and post-thaw viability.
Controls:
* Dimethyl Sulfoxide (preventing Ice Crystal formation) concentration
* Freezing rate and container integrity
* Final product sampling for QC (viability, identity, potency) - Labelling and Traceability
* Why critical: Risk of mismatch or misidentification.
Controls:
* Use of unique identifiers (e.g., barcodes)
* Reconciliation against source and recipient data
* Verification at each transfer step - Storage and Transport
* Why critical: Product viability is highly sensitive to temperature excursions.
Controls:
* Use of validated cryo-storage and dry shippers
* Continuous temperature monitoring
* Transport chain of custody documentation
What are the key QC tests performed for a cell-based Advanced Therapy Medicinal Product (ATMP)?
- Appearance / Visual Inspection
* Purpose: To ensure no particulate matter or visible contamination.
* Check: Colour, clarity, presence of aggregates. - Cell Count and Viability
* Purpose: Confirm therapeutic cell dose and ensure viable cells are delivered.
* Methods: Trypan blue exclusion or flow cytometry.
* Acceptance: Typically ≥70% viability. - Identity
* Purpose: Confirm that the correct cell type is present (e.g., T-cells).
* Method: Flow cytometry using specific surface markers (e.g., CD3 for T-cells).
* Also confirms: Patient/donor match (traceability). - Potency
* Purpose: Demonstrate biological activity of the product.
* Methods:
* Cytotoxicity assay (e.g., killing of target cancer cells)
* Cytokine release (e.g., IFN-γ, IL-2)
* Expression of CAR (e.g., via flow cytometry or qPCR) - Sterility
* Purpose: Ensure product is free from bacterial or fungal contamination.
* Method: Compendial method (Ph. Eur. 2.6.1) or validated rapid sterility test.
* Note: May not be available at release due to short shelf-life—parametric release may be applied. - Endotoxin (Bacterial Pyrogens)
* Purpose: Prevent harmful inflammatory responses in patients.
* Method: LAL test (Limulus Amebocyte Lysate).
* Acceptance: Below threshold based on dose volume (e.g., <5 EU/kg/hour). - Mycoplasma Testing
* Purpose: Detect common contaminants in cell cultures.
* Methods: Culture-based or PCR-based validated methods. - Replication-Competent Virus (RCV) Testing (for viral vector-based ATMPs like CAR-T)
* Purpose: Confirm absence of infectious virus from vector production.
* Timing: Typically performed on the vector and sometimes on the final product. - Adventitious Virus Testing
* Purpose: Detect unexpected viral contaminants.
* Usually performed: On vector seed banks or cell banks, not every batch. - pH / Osmolality / DMSO Concentration
* Purpose: Ensure the product is within physiological limits and formulation is accurate.
Difference between large and small molecule tests?
The main difference between large and small molecule QC tests lies in their structural complexity and testing approach. Large molecules such as proteins, monoclonal antibodies, or hormones have complex 3D structures and are inherently variable. Their QC tests often include immunological or functional assays, such as ELISA or bioassays, to assess potency, binding affinity, or biological activity. These tests require tight control of temperature, pH, and handling, as biologics are sensitive to denaturation. In contrast, small molecules are chemically synthesised, with well-defined structures and higher stability. Their testing focuses on physicochemical properties, using techniques like HPLC, Karl Fischer titration, acid-base titration, or oxidation-reduction assays. Method validation differs too — small molecules often rely on ICH Q2(R2), whereas large molecules follow both ICH Q2 and Q6B for biological characterisation.
You’re validating an autoclave for both porous and fluid loads. Can you draw the load types, and explain the purpose of the pulses in the cycle?
Sure. In autoclaving, we typically validate two types of loads:
- Porous loads – such as instruments, filter assemblies, stainless steel parts, rubber bungs, wraps, textiles
- Fluid loads – like water for injection (WFI), growth media, or product in final containers
Draw up pulses.
- Porous Load (e.g. surgical instruments, textiles):
Beginning of the cycle:
Pre-vacuum pulses (alternating negative and positive pressure) are applied to remove trapped air from occluded areas and replace it with saturated steam.
This ensures effective steam penetration to all parts of the load.
Middle (Sterilisation phase):
The plateau (e.g. at 121°C) is typically shorter than for fluid loads, since moist heat contact with surfaces is sufficient once air is fully removed.
End of cycle:
Post-vacuum pulses may be applied to remove residual steam and moisture, aiding in the drying phase.
- Fluid Load (e.g. IV bags, ampoules, vials):
No vacuum pulses are applied — to avoid boiling or product loss due to pressure drop.
Longer sterilisation plateau is required to allow time for heat to uniformly penetrate the liquid in the containers.
Air overpressure is often used during cooling to prevent container breakage due to internal vapor pressure (in the vials).
What are the pulses in the cycle for?
The pulses refer to vacuum and pressure phases in the pre-conditioning stage of a pre-vacuum autoclave cycle.
- Their purpose is to remove air from the chamber and load — especially from porous materials and lumens where air can be trapped.
- Air inhibits steam contact, and even a small air pocket can prevent sterilisation.
- Pulsing alternates vacuum and steam injections to create pressure changes that ‘suck out’ air from the deepest parts of the load.
This is why porous loads need pre-vacuum cycles with pulsing.
Fluid loads, on the other hand, typically use gravity or slow exhaust cycles, with air removal via steam displacement.
What’s the difference between porous and fluid loads in validation?
- Porous Loads
Use pre-vacuum cycles with multiple pulses
Validated using Bowie-Dick test for air removal
Challenged using biological indicators (BIs) in wrapped instruments
Risk = air pockets - Fluid Loads
Often use gravity or slow-exhaust cycles
Validated using FP sensors to confirm time/temp
Challenged using BIs and temp probes inside liquid containers
Risk = delayed heating/cooling (cold spots)
What’s the risk of air pockets?”
“Air pockets prevent direct contact between saturated steam and the item’s surface, which is necessary for efficient heat transfer and microbial kill.
A single air pocket can result in an unsterilised zone. That’s why:
- Air removal is critical in porous load sterilisation
- We verify this with Bowie-Dick tests and thermometric mapping
- Load configurations must minimise crevices and ensure proper drainage”
For an Aseptic filling line, it was given a schematic diagram of a Grade C cleanroom with Isolator, showing location of environmental monitoring in both the Grade C room and the Grade A isolator. The product being filled was a biologic (mab) not subjected to terminal sterilisation.
You are a QP at this site (MIA) reviewing the batch docs for certification. 6 cfu had been recovered from the finger dab (Gram +ve Cocci/skin commensals) contact plate for 1 of the operators, all other Ems were within limits. How do you proceed?
spent a great length asking questions about the Product, and Process (PRICE-PRISM and drew out Ishikawa analysis to identify the issue). Turned out that root cause was faulty VHP transfer sanitisation system. I was challenged/questioned for almost all the questions I asked in this scenario i.e., why did I want to know that, what was the relevance to my decision making?
Micro OOS for a dispensing booth in non-sterile manufacture, what do you do? Micro OOS due to increased capacity, impact on batches; same scenario but in sterile manufacture what would you do;
Micro OOS due to increased capacity, impact on batches
Same scenario but in sterile manufacture what would you do?
Grade D micro limits as per Annex 1?
A
Air sample CFU /m3 - No growth
Settle plates CFU /4 hours - No growth
Contact plates CFU / plate - No growth
Glove print, Including 5 fingers on
both hands CFU / glove - No growth
B
Air sample CFU /m3 - 10
Settle plates CFU /4 hours - 5
Contact plates CFU / plate - 5
Glove print, Including 5 fingers on
both hands CFU / glove - 5
C
Air sample CFU /m3 - 100
Settle plates CFU /4 hours - 50
Contact plates CFU / plate - 25
Glove print, Including 5 fingers on
both hands CFU / glove - not required
D
Air sample CFU /m3 - 200
Settle plates CFU /4 hours - 100
Contact plates CFU / plate - 50
Glove print, Including 5 fingers on
both hands CFU / glove - not required
Grade of rooms for different types of activities i.e. dispensing in sterile vs non sterile?
Sterile:
Grade A: Critical zone for aseptic operations (e.g. open vial filling, aseptic connection) — usually achieved with LAF or isolator.
Grade B: Background environment for Grade A (e.g. background to an open RABS or LAF zone).
Grade C: Preparation of sterile components (e.g. compounding, weighing under laminar flow), background for sterilised items prior to aseptic transfer (e.g. autoclave unloading, isolator loading).
Grade D: Handling of non-sterile starting materials, washing of components, background for less critical aseptic support operations if justified by risk assessment.
Non-sterile
Grade C: Non-sterile product filling (e.g. oral liquids, metered dose inhalers) where low bioburden is required.
Grade D: Weighing and dispensing of non-sterile materials. May also be used for manufacturing steps where environmental control is needed but sterility is not required.
What do you understand about media fills?
A media fill is an aseptic process simulation required by EU GMP Annex 1. It validates the entire aseptic filling operation using sterile growth medium such as TSB in place of product. It must simulate worst-case conditions — including the longest duration, highest operator involvement, and critical interventions — to demonstrate that no microbial contamination occurs. Media fills are typically conducted at least every 6 months per shift and are essential for operator qualification, process validation, and sterility assurance.
Scenario1: You are doing your routine media fill and have 4 vials with growth – what would you do?
OOS Investigation as per MHRA guidance and ID test
Scenario1: If the microorganism was confirmed to be gram positive can you suggest any potential sources of contamination? Do you know the latin names ?
- human skin/mucosa: gwoning, intervention near the point of fill, glove damaged or poor aseptic skills.
Latin names:
Staphylococcus aureus, epidermidis, micrococcus leuteus, corynebacterium spp.
Scenario: If it was confirmed as a gram negative can you suggest any potential sources of contamination?
- water system: drain pipes
-wet surface, improper dries or sterilisaed equipment - raw materials with high bioburden
Pseudomonas aeruginosa
Escherichia coli
Enterobacter cloacae
Turns out it was a skin organism caused by a new operator – he hadn’t been present in any routine manufacture so no impact to lots already manufactured – what would you need to do before returning to routine manufacture?
- the line quarantine until 3 consequtive media fill pass
- the operatore training and 3 concequtive pass of media fill
You are a QP at the sight and came to know that the personal finger dabs failed which was taken before the aseptic processing. What are the limits for finger dabs in Tablets and creams manufacturing?
- Quarantine and Hold the Aseptic Filling Line
“The filling line must remain quarantined until the aseptic process is successfully revalidated.”
No sterile product may be manufactured or certified until process validation is complete.
This aligns with EU GMP Annex 1 (Section 10.12) and Annex 16.
- Requalify the Aseptic Process via 3 Consecutive Media Fills
“Three consecutive successful media fills must be completed under worst-case conditions to re-establish sterility assurance.”
Each run should simulate normal interventions and represent the full shift duration.
All critical parameters must be met: no contamination, all EM in limits, and full documentation.
- Dequalification and Retraining of the Operator
“The new operator must be dequalified and retrained in aseptic technique, gowning, and interventions.”
Review and reinforce training in:
Gowning procedure
Aseptic manipulations
Cleanroom behaviour
Assess gowning qualification, EM/personnel results, and intervention technique.
- Requalification of the Operator via 3 Successful Media Fill Runs
“The operator must successfully complete 3 consecutive media fills as part of requalification.”
These should be:
Individual or team-based simulations
Monitored and documented
Representative of actual process involvement
- Update Contamination Control Strategy (CCS) if Needed
You are a QP at a sterile manufacturing site and you received a call from QC informing you of a Micro OOS for a unit going turbid. This was still incubation and at day 5. This was part of Media Fill. What are your concerns? 8 batches manufactured since last Media Fill. 4 onsite and 4 on the market. What do you do? You have been told that the Micro is Micrococcus.
IMMEDIATE ACTIONS (Containment and Risk Identification)
1. Confirm Media Fill Type:
* I would first confirm whether the media fill is a process simulation (routine requalification of the aseptic process) or an operator qualification.
* If it is a process simulation, this represents the entire line’s sterility assurance and puts all batches manufactured since the last successful media fill at risk.
* If it is an operator qualification, the impact may be limited to that operator’s involvement, but still requires escalation.
2. Send Contaminated Unit for Microbial Identification:
* Although it’s only Day 5, I would immediately remove the turbid unit for microbial ID, understanding that this terminates its incubation.
* The remaining media fill units would continue incubation to Day 14, as per EU GMP Annex 1 (2022), Section 10.9.
* Additional positives during incubation would further confirm process failure.
3. Initiate a Formal OOS Investigation:
* An OOS investigation is opened per SOP, covering:
* Turbidity confirmation and visual inspection
* Incubation chamber integrity
* Potential for lab handling error or cross-contamination
4. Interpret Organism Identified (Micrococcus):
* Micrococcus is typically a human skin flora, indicating potential operator-related contamination, such as inadequate gowning, poor aseptic technique, or barrier integrity failure.
5. Quarantine the Aseptic Manufacturing Line:
* I would immediately halt all aseptic operations and quarantine the isolator, associated equipment, and any components used during the media fill until investigation and root cause analysis are complete.
⸻
PRODUCT IMPACT AND REGULATORY RISK
6. Raise a Deviation and Assess Product/Patient Impact:
* I would initiate a GMP deviation to assess potential impact on sterility assurance and patient safety.
* I would evaluate the robustness of our Contamination Control Strategy (CCS) and identify any lapses in controls.
* I would also check for any customer complaints, ADRs, or pharmacovigilance signals via the QPPV to support a comprehensive impact assessment.
7. Identify and Quarantine All Potentially Affected Batches:
* 4 batches on-site: Quarantine immediately.
* 4 batches released to market: Compile product details (batch number, expiry, distribution, status).
* Begin a recall risk assessment, considering the media fill failure as a potential signal of compromised product quality.
⸻
INVESTIGATION STRATEGY
8. Conduct Root Cause Analysis Using Ishikawa (Fishbone):
* Personnel: Review operator training, aseptic qualifications, gowning logs, CCTV footage.
* Method: Assess media fill design, interventions, aseptic manipulations, filling technique.
* Environment: Analyse EM data before, during, and after the session, including any alert/action level excursions.
* Equipment: Review isolator integrity, leak test results, pressure differentials, airflow verification.
* Materials: Check transfer technique, RABS/RTP performance, sanitisation, and component integrity.
⸻
DISPOSITION AND RECALL DECISION
9. Escalate if Contamination Risk to Product Cannot Be Ruled Out:
* Convene a Recall Committee and conduct a recall classification as per MHRA guidance.
* If sterility assurance is compromised, initiate a Class 2 recall for the 4 market batches, and reject/dispose of the 4 batches on-site.
* Notify the Defective Medicines Report Centre (DMRC) and maintain communication with MHRA throughout the process.
* Provide a full QP decision justification with risk assessment, referencing Annex 16 and the Orange Guide.
⸻
REMEDIATION AND PREVENTION
10. Implement CAPA Based on Investigation Findings:
* Reinforce aseptic technique training, focusing on hand hygiene, gowning, and line interventions. * Requalify all operators involved. * Strengthen EM monitoring frequency and review CCS effectiveness. * Evaluate the design of the media fill and increase challenge robustness if needed. 11. Verify Effectiveness of CAPA Before Restarting Operations: * Require three consecutive successful media fills, covering both process simulation and operator requalification. * QA and QP must review and approve all investigation outcomes and CAPA prior to resuming routine aseptic manufacturing.
⸻
Conclusion (For Viva Closure):
“As QP, I would lead a science-based and risk-driven investigation into the media fill failure, act conservatively regarding product disposition to protect patients, and ensure full GMP compliance and MHRA engagement throughout. I would not allow any resumption of aseptic manufacturing until a thorough investigation and CAPA implementation is complete, with demonstrable sterility assurance re-established.”
What different types of sterilisation methods are there?
- Terminal Sterilisation
Sterilisation of the product in its final container.
a.
Moist Heat Sterilisation (Autoclaving)
Uses saturated steam (e.g. 121°C for 15 mins)
Mechanism: Coagulation and denaturation of proteins
Suitable for: Aqueous solutions, surgical instruments, certain injectables
Preferred method per EU GMP Annex 1 where applicable
b.
Dry Heat Sterilisation
Uses hot air (e.g. 160–170°C for 2+ hours)
Mechanism: Oxidative damage
Suitable for: Glassware, metal instruments, oils, powders
Also used for depyrogenation
c.
Filtration (Sterilising-Grade)
Membrane filters (typically 0.22 μm pore size)
Mechanism: Physical removal of microorganisms
Suitable for: Heat-sensitive solutions
Must be followed by aseptic filling in Grade A environment
d.
Gas Sterilisation
Ethylene Oxide (EtO): Alkylates microbial DNA and proteins
Used for heat/moisture-sensitive medical devices
Requires long aeration to remove toxic residues
Vapour Hydrogen Peroxide (VHP): Used for room and equipment decontamination, not typically used for product sterilisation
e.
Radiation Sterilisation
Gamma irradiation (Cobalt-60), E-beam, or X-ray
Mechanism: Ionises microbial DNA
Suitable for: Single-use medical devices, plastics, some pharmaceuticals
How would you validate a steam autoclave?
Talk about equipment qualification, validation protocol, load cycles, mapping, Bis, F0 calculations, drew cycles, routine checks and water quality
Daily- bowie dick, chamber, probe, door seal, load inspection, alarms, log book, drain
weekly- leak rate test, air detector function, bowie dick test
yearly - leak rate test, bowie dick, steam quality test (non-condensable gasses <3.5%, Steam dryness >0.95%, super heat not exceed , air detector
Why is non condensable gas important?
Non condensable gases like air or CO2 do not condense and can reduce sterilisation efficiency.