Pharmaceutical formulation and processing Flashcards
(100 cards)
Give examples of products where the API is derived from a cell bank?
Monoclonal antibodies, recombinant proteins like EPO and insulin, certain viral and protein-based vaccines, and enzyme replacement therapies all rely on a defined MCB and WCB system to ensure batch consistency and regulatory compliance.
Q1:
What are the main types of Advanced Therapy Medicinal Products (ATMPs)?
Q2:
Can you give an example of a cell-based ATMP and briefly explain how it works?
Q3:
What is considered the API in CAR-T cell therapy?
- General Understanding of ATMPs
Q1: What are the main types of Advanced Therapy Medicinal Products (ATMPs)?
A1:
ATMPs include:
• Gene therapy products – deliver genes to treat or prevent disease.
• Somatic cell therapy products – use cells that have been manipulated ex vivo.
• Tissue-engineered products – use cells and scaffolds to regenerate, repair, or replace tissues.
• Combined ATMPs – contain a medical device as an integral part.
⸻
Q2: Can you give an example of a cell-based ATMP and briefly explain how it works?
A2:
Yes, CAR-T therapy is a cell-based ATMP. Patient T-cells are collected, genetically modified using a viral vector to express a Chimeric Antigen Receptor (CAR) targeting tumor-specific antigens, expanded, and re-infused to attack cancer cells.
⸻
Q3: What is considered the API in CAR-T cell therapy?
A3:
The genetically modified T-cells themselves are the API, as they provide the therapeutic effect.
- Process Knowledge
Q4:
Walk me through the key manufacturing steps for an autologous CAR-T cell therapy.
Q5:
What are the critical differences in manufacturing autologous vs allogeneic cell therapies?
Q6:
What are the potential risks during the transduction step, and how are they controlled?
Q7:
Why is cryopreservation important in ATMP manufacture, and what are the challenges it introduces?
- Process Knowledge
Q4: Walk me through the key manufacturing steps for an autologous CAR-T cell therapy.
A4:
1. Apheresis – patient T-cells collected.
2. Cell isolation – T-cells separated from leukocytes.
3. Transduction – T-cells genetically modified with a viral vector.
4. Expansion – modified cells multiplied under controlled conditions.
5. Formulation – formulated and cryopreserved for delivery.
6. Thaw & Infusion – thawed at site and infused into patient.
⸻
Q5: What are the critical differences in manufacturing autologous vs allogeneic cell therapies?
A5:
• Autologous: patient-specific, one batch = one patient, higher variability, more logistical control needed.
• Allogeneic: donor-derived, one batch for multiple patients, allows for cell banking and standardization.
⸻
Q6: What are the potential risks during the transduction step, and how are they controlled?
A6:
Risks include:
• Incomplete transduction
• Insertional mutagenesis
• Contamination with replication-competent virus
Controls:
• Use of GMP-grade viral vectors
• In-process monitoring of transduction efficiency
• QC testing for replication-competent virus
⸻
Q7: Why is cryopreservation important in ATMP manufacture, and what are the challenges it introduces?
A7:
It preserves cell viability for transport and scheduling flexibility. Challenges include:
• Cell viability upon thawing
• Cold chain integrity
• DMSO toxicity (cryoprotectant)
- GMP and QP Certification Perspective
Q8:
What GMP guidelines apply to ATMPs?
Q9:
As a QP, what specific documentation or evidence would you review before certifying an ATMP batch for release?
Q10:
If sterility test results are not available due to short shelf-life, how would you justify release?
Q11:
Would you consider releasing an ATMP batch that is out of specification for sterility? Under what circumstances?
- GMP and QP Certification Perspective
Q8: What GMP guidelines apply to ATMPs?
A8:
EudraLex Volume 4, Part IV – GMP for ATMPs. It requires a risk-based approach, tailored control strategies, and specific traceability for human tissues. Also, relevant sections of Annex 1 apply for aseptic processing.
⸻
Q9: As a QP, what specific documentation or evidence would you review before certifying an ATMP batch for release?
A9:
• Batch manufacturing record
• Apheresis documentation
• QC results (sterility, endotoxin, viability, identity, potency)
• Chain of identity and custody records
• Cold chain monitoring
• Any deviations/investigations
• Certificate of analysis
• Confirmation of traceability
⸻
Q10: If sterility test results are not available due to short shelf-life, how would you justify release?
A10:
Justify release based on parametric release:
• A validated aseptic process
• Environmental monitoring data
• Filter integrity test
• Endotoxin test passed
• A predefined risk assessment approved by QP and inspectorate
⸻
Q11: Would you consider releasing an ATMP batch that is out of specification for sterility? Under what circumstances?
A11:
In exceptional cases only.
• Requires clinical need and no alternative therapy
• Physician and QP agreement
• MHRA notified
• Organism identified and risk assessed
• Administered under a “specials” exemption—not certified by QP
- Regulatory and Traceability
Q12:
How does traceability differ between ATMPs and traditional biologics?
Q13:
What role does the Human Tissue Authority (HTA) play in ATMP manufacturing in the UK?
Q14:
What agreements must be in place if ATMP manufacturing steps are performed at different sites?
- Regulatory and Traceability
Q12: How does traceability differ between ATMPs and traditional biologics?
A12:
ATMPs require full bidirectional traceability from donation to administration due to patient-specific material. This includes donor, processing, and distribution data—especially under Directive 2004/23/EC and Part IV GMP.
⸻
Q13: What role does the Human Tissue Authority (HTA) play in ATMP manufacturing in the UK?
A13:
HTA ensures consent, procurement, testing, and traceability for human tissues and cells. For ATMPs involving tissues (e.g., stem cells), HTA licensing is required under the Human Tissue (Quality and Safety for Human Application) Regulations 2007.
⸻
Q14: What agreements must be in place if ATMP manufacturing steps are performed at different sites?
A14:
• Technical/Quality Agreements defining responsibilities (GMP compliance, documentation, deviations)
• Clear traceability between sites
• QP oversight of outsourced steps
• Annex 16 compliance (QP must have visibility and final decision-making authority)
⸻
- Comparison with Biologics/Biotech
Q15: How does the use of cell banks in biologics differ from the approach used in ATMPs?
A15:
Biologics use Master and Working Cell Banks (MCB/WCB) for consistency across multiple batches.
ATMPs (especially autologous) use fresh patient-derived cells with no banking; variability is higher, and each batch is unique.
⸻
Q16: Explain why master and working cell banks are not typically used in autologous ATMP manufacturing.
A16:
Because each autologous product is derived from an individual patient’s own cells, which are not banked or reused. The process is batch-specific and non-replicable, unlike standardised cell lines in biologics.
⸻
Q17: How do the shelf-life and storage requirements of ATMPs affect the QP decision-making process?
A17:
Short shelf-life (often hours to days) limits time for QC release. QPs may rely on in-process controls, parametric release data, and real-time review of batch records to make timely certification decisions.
- Comparison with Biologics/Biotech
Q15:
How does the use of cell banks in biologics differ from the approach used in ATMPs?
Q16:
Explain why master and working cell banks are not typically used in autologous ATMP manufacturing.
Q17:
How do the shelf-life and storage requirements of ATMPs affect the QP decision-making process?
Q15. In biologics (e.g., monoclonal antibodies), a master cell bank (MCB) and working cell bank (WCB) are established during development. The WCB is used as a consistent and qualified source of the production cell line that generates the active substance (API) in large-scale manufacturing. These banks are central to ensuring consistency, traceability, and control of the biological source material over time.
In contrast, ATMPs, especially autologous gene therapies like CAR-T cells, do not use traditional cell banks for product manufacturing, since the starting material (e.g., patient’s own T-cells) is unique to each batch. However, cell banks may still be used in the QC lab to prepare reference materials, such as:
• Positive controls for potency assays (e.g., engineered T-cells from a healthy donor),
• Or target cell lines (e.g., CD19+ tumour cell lines) used in cytotoxicity-based potency tests.
These reference banks are typically developed in-house and are assay-specific, not product-specific, and are not used to produce the final product itself — unlike in biologics.
Q16.
Can you describe the process of manufacturing Pressurised Metred Dose Inhalers?
- Overview
Pressurised Metered Dose Inhalers (pMDIs) are combination products containing an active substance, excipients, and a liquefied gas propellant. Manufacturing must ensure:
* Content uniformity
* Dose reproducibility
* Container closure integrity
Manufacture involves handling of liquefied gases, pressure filling, and precise valve assembly, requiring strict control under Annex 10.
⸻
- Dispensing and Formulation
- Dispensing is performed in controlled conditions appropriate to the material (typically Grade D).
- The formulation may be:
- A suspension (API suspended in propellant)
- A solution (API dissolved in propellant)
- Common propellants include hydrofluoroalkanes (HFA-134a or HFA-227).
- Accurate control of API concentration and suspension uniformity is critical.
⸻
- Filling Methods
Annex 10 describes two compliant filling approaches:
a. One-Shot Filling (Single-Stage Pressure Filling)
* The bulk formulation (API + propellant) is prepared in a pressure vessel with mixing.
* The product is filled directly into the canister using a pressure filling head.
* The metering valve is crimped after filling.
b. Two-Shot Filling (Split Filling)
* A portion of the formulation is pressure-filled into the canister.
* The valve is then crimped onto the canister.
* The remaining formulation or propellant is pressure-filled through the valve stem.
* This allows for more accurate dose control in certain designs.
⸻
- Crimping
- Per Annex 10, crimping must be controlled and validated.
- Checks include crimp diameter, depth, and seal integrity.
- Poor crimping can cause leakage and dose inconsistency.
⸻
- Assembly
- The filled and crimped canister is inserted into a plastic actuator body (inhaler device).
- The assembled unit is then labelled and packaged.
⸻
- In-Process Controls (as per Annex 10)
Critical in-process controls include:
* Weigh checking (net fill weight or pressure fill volume)
* Crimp integrity tests
* Valve function and leakage
* Suspension uniformity (if applicable, may require mixing prior to filling)
⸻
- QC Testing
Finished product testing must demonstrate:
* Uniformity of delivered dose (per actuation)
* Content uniformity
* Valve performance and dose reproducibility
* Leak testing (e.g. crimp and valve seal integrity)
* Microbiological quality, if applicable (especially for multi-dose or aqueous-containing systems)
* Stability testing to ensure valve and suspension performance over shelf life
⸻
- GMP Considerations under Annex 10
- Equipment must be suitable for high-pressure operation and regularly qualified.
- Liquefied gases must be handled safely and stored appropriately.
- Cleaning and maintenance of filling lines must prevent cross-contamination and maintain product quality.
- Manufacturing operations must be performed by trained personnel with validated processes.
⸻
Viva-Ready Summary:
“Under Annex 10, the manufacture of pMDIs involves accurate dispensing, pressure filling via either one-shot or two-shot methods, valve crimping, device assembly, and stringent in-process controls. Propellants like HFA-134a are used, and crimp integrity and fill weight are critical control points. Finished product testing must confirm delivered dose uniformity and leak integrity. All processes must be validated and comply with Annex 10 due to the specialised nature of aerosol manufacturing.”
What are the CPPs and facility requirements?
Facility Requirements (per EU GMP Annex 10)
1. Closed Filling Systems
* Required to prevent exposure to liquefied gas propellants and avoid contamination.
* Must be pressure-rated, well-maintained, and validated.
2. Environmental Classification
* Minimum Grade D is acceptable for:
* Dispensing of starting materials
* Filling of pressurised canisters in closed systems
* Open handling (if any) must be justified and may require Grade C.
3. Temperature-Controlled Storage
* APIs and propellants must be stored at validated temperatures to ensure stability and dosing accuracy.
4. Explosion-Proof and ATEX-Compliant Equipment
* Required due to the flammable nature of propellants (e.g., HFAs).
* Electrical installations must comply with safety regulations.
5. Validated HVAC and Pressure Controls
* To prevent ingress of particulates or moisture.
* Maintain environmental conditions for sensitive materials.
6. Personnel and Operator Protection
* Ensure containment and exposure controls are in place during pressure filling.
* Training and PPE for handling propellants and high-pressure equipment.
⸻
Viva-Ready Summary:
“Key CPPs for pMDIs include content and dose uniformity, suspension homogeneity, valve and crimping integrity, leak testing, and propellant temperature/pressure control. Facilities must support closed-system filling with a minimum of Grade D environment, temperature-controlled storage, validated pressure-rated equipment, and ATEX compliance for flammable propellants — all as required under Annex 10.”
How the CPPs were relevant/influential to the CQAs?
CPP
Linked CQAs
Why It Matters
Bulk formulation mixing time and speed
Content uniformity, DDU
Prevents API sedimentation and ensures homogeneity
Suspension hold time
Content uniformity
Extended hold can lead to sedimentation or stratification
Agitation during filling
Dose uniformity
Keeps suspension consistent during pressure filling
Filling temperature and pressure
Fill weight, spray characteristics
Propellants are highly temperature-sensitive
Shot weight / fill volume control
Delivered dose, DDU
Inaccurate fill impacts actuation dose and number of doses per unit
Crimping pressure and diameter
Leak rate, stability
Critical for container closure integrity
Valve component quality (metering chamber volume)
DDU, APSD
Variability affects dose volume and particle delivery
Environmental conditions (Grade D)
Micro quality, particulate control
Supports low bioburden manufacture (as per Annex 10)
Cleaning validation of lines and valves
Micro quality
Please give a summary of the key specifications of a PMDi?
Summary: Key Specifications for a Pressurised Metered Dose Inhaler (pMDI)
- Content Uniformity
- Each inhaler must deliver API content within ±15% of label claim
- Acceptance criteria often based on Ph. Eur. 2.9.40 / USP <601>
- Based on assay per container (e.g. 85–115% of stated content)
- Delivered Dose Uniformity (DDU)
- Measured over multiple actuations from each unit
- Each delivered dose must be within 75–125% of label claim
- Mean dose typically required to be within 85–115%
- Test conducted after shaking, at different canister positions (beginning, middle, end)
- Aerodynamic Particle Size Distribution (APSD)
- Measured by cascade impaction or laser diffraction
- Mass Median Aerodynamic Diameter (MMAD) should be:
- Typically <5 microns for pulmonary delivery
- Ensures lung deposition
- Fine Particle Fraction (FPF): portion of emitted dose <5 µm — often reported
- Microbiological Quality (for non-sterile inhalers)
- TAMC (Total Aerobic Microbial Count):
- <100 CFU/g or mL
- TYMC (Total Yeast and Mould Count):
- <10 CFU/g or mL
- Absence of specified pathogens (per Ph. Eur. 5.1.4 / BP Vol. 5):
- Staphylococcus aureus: Absent
- Pseudomonas aeruginosa: Absent
bile torerwnt gram negatives - absent
Note: pMDIs with aqueous components or intended for immunocompromised patients may require tighter controls or sterile assurance.
⸻
- Additional Tests
- Leak test / crimp integrity
- Actuation force
- Priming / re-priming performance
- Spray pattern and plume geometry
- Stability testing under ICH conditions
You are a QP at a PMDi manufacturing site that has been dedicated to the manufacture of a salbutamol inhaler. Site leadership wants to introduce the capability to manufacture steroid inhalers too. How would you advise the site to make this introduction?
Step 1: Initiate Change Control (CC)
Open a formal change control to capture all proposed activities.
Involve cross-functional teams: QA, manufacturing, regulatory, validation, engineering, and H&S.
Assign risk rating and timescales, and define deliverables (e.g. cleaning validation, facility upgrade).
Step 2: Impact Assessment
a. Licence (MIA/MA)
Review the MIA scope – does it include manufacturing of steroid-based pMDIs?
If the new activity is not covered, variation to MIA may be required.
For IMPs or commercial products, ensure Marketing Authorisation (MA) includes the new formulation, process, and shelf life specs.
b. GMP and Facility Design
Steroids (e.g. beclomethasone, fluticasone) are typically high potency APIs.
Evaluate need for:
Dedicated equipment or fully validated cleaning (per EU GMP Chapter 5.19–5.21)
Local exhaust ventilation, airlocks, or containment systems
Appropriate operator protection and PPE
Environmental monitoring controls for airborne particles
c. Cross-contamination / Mix-up Risks
Salbutamol is a reliever, steroids are preventers — mix-ups could have critical patient impact.
Update your Contamination Control Strategy (CCS) (Annex 1, 2022) to reflect:
Product segregation
Line clearance procedures
Equipment and valve differentiation (colour coding, labelling)
Step 3: Validation and Qualification
a. Cleaning Validation
If shared equipment is used, demonstrate robust cleaning to acceptable MACO limits.
Consider worst-case matrix (e.g. low-dose, highly potent steroid vs. high-volume salbutamol)
b. Process Validation
Perform traditional or lifecycle-based process validation for:
Bulk suspension preparation
Filling process (shot weight, homogeneity)
Crimping and leak integrity
Delivered dose and particle size distribution
c. Analytical Method Validation
For both actives and preservatives (if applicable)
Ensure methods are sensitive enough to detect cross-contaminants
Step 4: Update QMS and QP Oversight
Revise:
SOPs
Batch documentation
Personnel training
Product-specific risk assessments
As QP, ensure all changes are assessed and documented under Annex 16 obligations.
Viva-Ready Summary:
“I would advise opening a change control and conducting a full impact assessment on the MIA, GMP compliance, and contamination risks. Since steroids are high potency, I’d evaluate the need for dedicated facilities or cleaning validation, update the CCS, and implement strong segregation measures. I’d also ensure process and cleaning validations are performed, and the QMS is updated to support the new product. No batch can be released until the facility and process are shown to be under control and regulatory requirements are met.”
How Cleaning validation is conducted?
Reference: EU GMP Annex 15, EMA PDE Guideline, PIC/S PI 006
Cleaning validation ensures that manufacturing equipment is cleaned effectively between product changes to prevent cross-contamination and ensure patient safety.
- Risk-Based Approach and Planning
Conduct a risk assessment to define the scope:
Use an equipment train map to identify shared surfaces
Assess worst-case product based on toxicity, potency, solubility, and cleanability
Identify hard-to-clean areas (dead legs, gaskets, mixer shafts)
Map cleaning agent compatibility and historical cleaning performance
Use a Cleaning Validation Matrix to determine if the new product is the “worst-case”:
Most difficult to clean?
Most toxic or lowest PDE?
Least soluble in cleaning agent?
- Establishing Acceptance Criteria
Use the PDE-based MACO (Maximum Allowable Carryover) approach:
PDE (mg/day) = derived from toxicological evaluation (e.g. NOAEL × human weight ÷ safety factors)
MACO calculation (shared equipment):
MACO = (PDE × Minimum batch size of next product) / Maximum daily dose of next product
Acceptance limits also defined for:
Visual cleanliness
Microbiological limits (if applicable)
Residue levels (API, cleaning agent, bioburden)
- Sampling and Analytical Methodology
Choose validated sampling methods:
Swab sampling: for hard-to-clean, direct-contact areas
Rinse sampling: for large, cleanable areas or inaccessible internal surfaces
Sometimes both methods are used for completeness
Validate the analytical method:
Ensure specificity, sensitivity, and reproducibility
Must achieve LOQ below the MACO
Recovery rate from surfaces should be ≥70%
- Cleaning Validation Protocol Must Include:
Scope, rationale, and worst-case rationale
Sampling plan and sampling sites
Acceptance criteria (MACO, visual, micro)
Analytical methods (validated, recovery rates stated)
Number of cleaning runs (typically 3 consecutive successful runs)
Cleaning agent and parameters
Staff training and operator qualification
Cleaning verification worksheets
Plan for deviation management and CAPA
Revalidation triggers (e.g. new product, equipment changes)
- Ongoing Verification and Review
Include cleaning verification during batch cleaning operations
Define periodic revalidation frequency based on risk and cleaning history
Trending and periodic review of swab/rinse data and deviations to confirm state of control
Can you tell me the formulation of a tablet and the purpose of each excipient?
Excipient
Function / Purpose
Active Pharmaceutical Ingredient (API)
The drug substance that provides the therapeutic effect
Binder (e.g., Povidone, HPMC)
Promotes adhesion of powder particles during granulation; ensures tablet integrity
Colorant (e.g., Iron oxides, Titanium dioxide)
Provides identification and aesthetic appeal; helps avoid medication errors
Coating Agent (e.g., Hypromellose, Sugar)
Masks taste, protects from moisture/light, improves swallowability
Diluent / Filler (e.g., Lactose, Mannitol, MCC)
Increases bulk to allow proper tablet size for low-dose APIs
Disintegrant (e.g., Crospovidone, Sodium Starch Glycolate)
Facilitates tablet breakup to ensure drug release
Enteric Coating Polymer (e.g., Cellulose acetate phthalate)
Protects the tablet from stomach acid; releases in the intestine
Flavor Agent (e.g., Peppermint oil, Vanilla)
Masks unpleasant taste in chewable or dispersible tablets
Glidant (e.g., Talc, Colloidal silica)
Improves powder flow properties during tablet compression
Lubricant (e.g., Magnesium stearate, Stearic acid)
Reduces friction during compression and ejection from die
Preservative (e.g., Methylparaben, Propylparaben)
Inhibits microbial growth in moisture-sensitive or liquid-containing formulations
Solubiliser (e.g., Polysorbate 80)
Enhances solubility of poorly soluble drugs
Sweetener (e.g., Aspartame, Sucralose)
Improves palatability in chewable/dispersible tablets
Surfactant (e.g., Sodium lauryl sulfate)
Improves wetting and dissolution
What could cause ‘sticking’ during tablet manufacture?
Root Causes of Sticking
- Formulation-Related Factors
Hygroscopic ingredients: Absorb moisture (e.g. sorbitol, mannitol, certain APIs), making them sticky.
Insufficient or inappropriate lubricant: E.g., inadequate magnesium stearate or incorrect mixing time.
Low melting point or waxy excipients: Soften under compression heat and adhere to tooling.
Poor flow or cohesion: Causes inconsistent fill and compaction behaviour.
- Environmental Conditions
High humidity in the manufacturing area: Increases moisture uptake, especially by hygroscopic excipients.
Inadequate drying of granules: Residual moisture can lead to tackiness and sticking.
- Process Parameters
Over-compression or low turret (rotation) speed: Generates more heat, softening the material.
Tooling (rotating part of tablet machine) temperature rise: Continuous compression without cooling can heat the punches.
Over-wet granules in wet granulation: Improper drying leads to moisture retention.
- Coating-Related Factors (if coated)
Over-wet coating solution: Causes sticking during the film-coating phase.
Improper drying during coating: Tackiness not resolved before pan rotation or air supply.
Can you give some reasons why you might coat a tablet?
⸻
- Taste Masking
- To mask the bitter or unpleasant taste of the active ingredient.
- Especially important for paediatric, chewable, or orally disintegrating tablets.
⸻
- Improved Appearance and Identification
- Provides a smooth, uniform surface and enhanced colour.
- Helps with product branding and dose differentiation.
- Improves patient acceptance and reduces risk of medication errors.
⸻
- Enteric Coating
- Protects the stomach lining from irritating drugs (e.g. aspirin).
- Prevents degradation of acid-labile drugs (e.g. omeprazole) in gastric fluid.
- Allows targeted release in the small intestine.
⸻
- Modified or Controlled Release
- Allows sustained, delayed, or timed release of the drug.
- Reduces dosing frequency, improves patient adherence, and maintains steady plasma levels.
⸻
Other Reasons (if asked):
* Moisture/light protection (e.g. for hygroscopic or light-sensitive APIs)
* Mechanical strength (to resist abrasion and chipping)
* Improved swallowability
What is classed as low and high pH and where in GI tract you get these pH ?
Low pH (Acidic): pH 1–4
* Location: Stomach
* The gastric environment has a pH of around 1.5 to 3.5 due to hydrochloric acid (HCl).
* This acidic environment is important for:
* Enzyme activation (e.g. pepsin)
* Digestion
* Killing pathogens
Neutral to Slightly Basic pH: pH 6–7
* Location: Duodenum (first part of the small intestine)
* Bile and pancreatic secretions neutralise gastric acid.
* Important for enzymatic digestion and absorption.
High pH (Alkaline): pH 7–8.5
* Location: Jejunum and ileum (distal small intestine)
* Slightly alkaline environment supports:
* Enzymatic function
* Absorption of most nutrients
High level controls / concerns when making a cream?
CQA - Why It’s Critical
Appearance- Ensures uniformity, absence of phase separation, colour consistency
Homogeneity (Content Uniformity)- Confirms even distribution of API throughout the product (no hot spots or API clumps)
pH- Ensures skin compatibility and API stability (typically pH 4.5–7.5 for dermal products)
Viscosity / Rheology- Affects spreadability, dose uniformity, and patient acceptability
Particle Size (if API is suspended)- Impacts texture, bioavailability, and physical stability
Microbial Quality- For non-sterile creams: TAMC < 100 CFU/g, TYMC < 10 CFU/g; no S. aureus, P. aeruginosa
Preservative Efficacy (PET)- Required for multi-use products; ensures resistance to microbial contamination
API Assay / Potency- Must meet specification (typically 90–110% of label claim)
Phase Separation / Stability- Ensures emulsion or suspension remains physically stable over shelf life
Water Content (if applicable)- Too high: microbial risk; too low: phase changes, thickening
Packaging Compatibility- Ensures container does not leach or absorb components; maintains integrity
Do creams have to be sterile? What facility classification is needed?
Not all creams require sterility. The need for sterility depends on the intended route of administration and clinical use. For example:
Yes – creams intended for ocular use, wound application, or use on broken/compromised skin must be sterile, due to the high risk of infection.
No – creams for intact skin (e.g. emollients or cosmetic creams) do not require sterility, but must still comply with microbial limits as per Ph. Eur. 5.1.4 or ICH Q6A.
Facility classification required:
For non-sterile creams:
Manufacturing should be performed in a closed system with appropriate environmental controls to minimize contamination.
A minimum of Grade D cleanroom is generally expected for manufacturing, especially at the filling stage, in accordance with WHO Annex 9 and risk-based approach.
For sterile creams:
Must comply with EU GMP Annex 1 requirements.
Final filling should be performed in Grade A (e.g. LAF or isolator), with a Grade B background (if open system) or Grade D/C background (if using isolators).
The facility should support aseptic processing, with validated sterilisation or aseptic compounding processes depending on the product type.
Can you think of any reasons why you would want a sterile cream?
Indication for occular, wound
Later another batch manufacturing lead again holding to repair mixing equipment. - Detail about qualification validation of the equipment, led to viscosity change led to Analytical method change, and AMV
Scenario: Mixing Equipment Failure → Viscosity Change → Analytical Method Change and AMV
Root Cause: Equipment Fault
The deviation arose due to failure of a mixing vessel, leading to viscosity deviation in a semisolid product. This had downstream impact on product quality, analytical method, and validation status.
- Immediate Containment
* Quarantine the line and affected batches.
* Halt further manufacturing until investigation and resolution are complete.
* Initiate urgent equipment repair. - Deviation and Investigation
* Open a deviation and initiate full root cause analysis:
o Determine onset of failure (e.g. motor wear, sensor drift).
o Identify all potentially impacted batches since the fault.
o Review:
Reference sample testing
Retrospective QC results
Customer complaints
Signal detection input from QPPV (if product is on market) - Equipment Qualification / Requalification (Annex 15)
If significant repair made:
* Initiate change control and requalification:
o Design Qualification (DQ) – if parts were redesigned or replaced.
o Installation Qualification (IQ) – confirm correct reassembly.
o Operational Qualification (OQ) – test RPM, temperature, pressure.
o Performance Qualification (PQ) – 3 consecutive compliant validation batches. - Impact on Product Quality
* Viscosity is a CQA — changes could affect:
o Dose uniformity
o Spreadability and stability
o Patient safety
- Conduct risk and impact assessment on batches previously released.
- Analytical Method Review and AMV
* If viscosity shift invalidates the current test method:
o Initiate method change via change control
o Conduct full method revalidation (ICH Q2(R2)):
Accuracy, precision, linearity, specificity, LOQ
o Update analytical SOPs and release specs - Regulatory Impact
* If the change affects:
o Finished product spec
o Analytical method
o Manufacturing process
* Submit a Type II variation to MHRA or EMA with supporting data:
o AMV, stability, validation results - Drug Shortage Risk
* If investigation or requalification causes supply delay:
o Liaise with the MA holder immediately
o If no suitable market alternative, notify MHRA via the DASH (Drug Shortages and Alerts) portal
o Document interim risk mitigation (e.g. staggered release, risk-based patient prioritisation)
What are the ideal properties of a cream how could you manage?
- Texture, Viscosity & Spreadability
Target: Smooth, non-gritty, easy to spread with acceptable consistency.
Controlled by:
Oil-to-water ratio (O/W or W/O system)
Choice of thickening agents (e.g., cetostearyl alcohol, carbomer)
Mixing speed and shear (CPP during emulsification)
Temperature control during manufacturing
- Appearance
Target: Uniform, homogenous cream with no phase separation or discoloration.
Controlled by:
Proper emulsification and preservative system
Use of colorants (e.g., titanium dioxide) if permitted
Addition of antioxidants (e.g., BHT, tocopherol) to prevent oxidative degradation
Microbial control to prevent spoilage or discoloration
- Phase Ratio (Oil/Water Balance)
Target: Stable emulsion with desired skin feel and drug release profile.
Controlled by:
Typical water phase content: ~20–80% (depending on O/W or W/O type)
Use of humectants (e.g., glycerol, propylene glycol) and emollients (e.g., white soft paraffin, isopropyl myristate)
Selection of appropriate oil phase excipients and co-surfactants
- Dosage Uniformity / API Distribution
Target: Homogeneous distribution of API throughout the cream matrix.
Controlled by:
Adequate mixing time and shear (CPP)
Use of emulsifiers (e.g., Tween 80, Span 60) to stabilise the system
In-process checks (sample homogeneity and content uniformity testing)
- Microbial Control / Preservation
Target: Free from objectionable organisms, with low bioburden.
Controlled by:
Use of preservatives (e.g., methylparaben, propylparaben) especially for multi-dose creams
Validated preservative efficacy test (PET) per Ph. Eur. 5.1.3
Hygienic processing, closed systems, and Grade D cleanroom conditions
Water quality: Purified Water or better
- Stability and Shelf-Life
Target: Maintains quality over intended shelf-life under defined storage.
Controlled by:
Inclusion of stabilisers, antioxidants, and proper packaging
Conducting stability studies under ICH conditions
Avoiding leachables from inappropriate container materials
How could you manage the consistency of the cream?
To ensure batch-to-batch consistency of a cream (a semisolid emulsion), a QP must rely on a combination of robust formulation design, validated processes, qualified systems, and controlled materials, all embedded within a compliant Pharmaceutical Quality System (PQS).
- Quality Management System (QMS)
Ensure a strong QMS is in place with:
Deviation, OOS, and Change Control procedures
Product Quality Reviews (PQRs) for trend analysis
CAPA and risk-based decision-making (ICH Q9/Q10)
- Facilities, Equipment, and Utilities
All systems must be:
Qualified (DQ/IQ/OQ/PQ) according to Annex 15
Maintained through a Preventive Maintenance Plan (PPM)
Monitored via EM programme (airborne particles, viable counts)
Purified Water system: validated and tested per Ph. Eur. specs (TAMC, TYMC, TOC, conductivity)
- Raw Material and Packaging Control
Approved suppliers with full technical agreements
Periodic supplier audits
Goods-in checks (ID, assay, bioburden, certificates of analysis)
Material traceability must be documented
- Manufacturing Process Controls
Control Critical Process Parameters (CPPs) such as:
Mixing speed, temperature, shear force
Water phase quality
Holding time before filling
Implement process validation (Annex 15 – traditional or continuous)
Ensure cleaning validation/verification between product changeovers
- In-Process and Final QC Testing
Perform in-process controls for:
Appearance
Viscosity
Weight/volume
pH
Uniformity of content
Final batch testing must confirm conformance to specifications
- Documentation and Review
All manufacturing and QC data must be:
Documented in real time
Reviewed and verified by trained personnel
Approved by QA before QP certification
- Supplier and Change Management
Ongoing supplier qualification
Evaluate impact of raw material variability
Control formulation/process changes via structured change control
What types of flows are there in a semisolid preparation?
In semisolid preparations such as creams, ointments, and gels, the materials typically exhibit non-Newtonian flow behaviour, meaning their viscosity changes under applied shear stress. The key types of flow include:
- Pseudoplastic Flow (Shear-Thinning)
Most common in creams and gels
Viscosity decreases as shear stress increases
(e.g., when rubbed or spread, the product becomes thinner and easier to apply)
Seen in emulsions and polymer gels
Example: topical corticosteroid cream
- Plastic Flow
Material behaves as a solid until a yield stress is exceeded, after which it flows like a liquid.
Once flow begins, viscosity decreases with increasing shear (similar to pseudoplastic).
Common in ointments (e.g., white soft paraffin-based)
Example: zinc oxide ointment
- Thixotropic Flow
A time-dependent shear-thinning behaviour
Viscosity decreases under constant shear but recovers when shear is removed.
Useful for semisolids that need to be easily applied but regain structure afterward.
Enhances physical stability
What will be generalized to the cream and what measures would you take in your manufacturing steps for consistency and flow?
-process validation and continuous Verification
A:
In cream manufacturing, generalised expectations include achieving a stable emulsion, consistent viscosity, homogeneity, and flow behaviour across all batches. These are directly linked to Critical Quality Attributes (CQAs) such as content uniformity, rheology, appearance, and phase stability.
To ensure batch-to-batch consistency and flow, I would implement the following controls throughout the product lifecycle:
⸻
- Pharmaceutical Quality System (PQS)
- A robust PQS must be in place to govern:
- Deviation, Change Control, OOS, and Complaint Handling
- Annual Product Quality Reviews (PQRs) to identify trends
- Ensures a lifecycle approach per ICH Q10
⸻
- Personnel
- Ensure GMP training, aseptic awareness (if applicable), and process-specific training
- Documented training records for manufacturing and QC staff
⸻
- Facilities and Equipment
- Qualification (DQ/IQ/OQ/PQ) of mixing vessels, homogenisers, and filling lines per Annex 15
- Preventive maintenance programme (PPM) in place
- Environmental Monitoring (EM) in production rooms (if Grade D or classified)
⸻
- Documentation and Batch Records
- Controlled SOPs, BMRs and QC worksheets in use
- All in-process and QC checks reviewed and signed by trained, authorised staff
⸻
- Manufacturing Process Controls
- Process validation for mixing speed, temperature, shear force, pH, and order of addition
- Implement continuous process verification (CPV), where justified, as per ICH Q13 for semi-continuous or batch processes
- Use in-process controls to monitor viscosity, appearance, and homogeneity
⸻
- Analytical Method Validation (AMV)
- Validated methods for:
- Viscosity
- pH
- Content uniformity
- Preservative content
- AMV aligned with ICH Q2(R2)
⸻
- Supplier Qualification and Material Control
- Approved suppliers under technical agreements
- Periodic supplier audits
- Goods-in checks on APIs, excipients, and packaging for identity and specification compliance
⸻
- Complaint and Recall System
- Evaluate customer complaints for trends linked to consistency or flow
- If needed, trigger recall procedure with root cause analysis and CAPA
⸻
- Internal and External Audits
- Conduct regular GMP audits of internal operations and third-party manufacturers
- Use findings to drive continuous improvement