Medchem Flashcards
(131 cards)
What is ADME of your product?
ADME – Therapeutic Radiopharmaceutical Injections (Aseptic, IV)
Absorption
- 100% bioavailability
Immediate systemic circulation
- Aseptic pyrogen-free
- No GI/barrier absorption phase
- Distribution
*Bloodstream → perfused organs
*Target-specific uptake (e.g. somatostatin, PSMA, thyroid)
*Vd depends on size/binding
*Free radionuclide = off-target (thyroid, bone, liver)
*Co-admin agents (e.g. amino acids for renal protection)
*Radiochemical purity = critical for correct biodistribution
Metabolism
*Minimal metabolism
*Stays chemically intact
*Iodide: organification in thyroid
*Peptides/antibodies: lysosomal degradation
*In vivo stability = QP release check
Excretion
*Renal (urine) or hepatobiliary (faeces)
*Effective half-life = physical + biological
*Rapid clearance (most in 24–48h)
*Radiation safety: excreta handling, shielding, patient isolation (if gamma)
*QP: ensure safe disposal, labeling, ARSAC/IRMER compliance
What are the control points for the manufacturing?
Critical Control Points – Aseptic Radiopharmaceutical Manufacturing
- Radioisotope Receipt & Handling
*Identity & radionuclidic purity check
*Shielding & contamination control
*Correct activity & calibration (traceable to national standards) - Aseptic Processing
*Grade A environment (e.g. isolator, LAF hood)
*Validated sterile filtration (0.22 μm filter)
*Media fill validation
*Cleanroom qualification (Grade B/C/D as applicable)
*Gowning, operator aseptic technique (training + requalification) - Radiolabeling Reaction
*Controlled temperature/time
*pH, reducing agent, buffer = optimal conditions
*Radiochemical purity critical for targeting
*Use of validated synthesis module/equipment - Radiochemical Purity Testing
*Instant TLC, HPLC
*Specification: typically ≥95%
*Detects free radionuclide (off-target risk)
*Must be complete before QP release - Final Sterile Filtration & Filling
*Aseptic technique
*Filter integrity test (pre/post)
*Closed vial systems, proper crimping
*Visual inspection: particulates, color, damage - Time-Critical Release & Transport
*Time from production to use is short (short half-life)
*Real-time release by QP (parametric, if sterility pending)
*Transport in approved, shielded containers
*Maintain chain of identity, temperature if needed - Batch Record & Traceability
*Full documentation: materials, process, QC
*Dose/activity traceable to patient
*Deviations investigated before release
Please talk through ADME of a product I had on my form.
Then proceeded to ask me about ADME in general (theory) and what is the importance of this knowledge for the OP. I talked about choices of formulation
Model Answer: ADME of My Product + Theoretical ADME + Relevance to QP
- ADME of My Product – Ga-68 Aseptically Prepared Injection
A – Absorption
* Administered intravenously, so 100% bioavailability — it enters the systemic circulation immediately, bypassing absorption barriers like the GI tract.
D – Distribution
* Ga-68 is chelated to a peptide ligand that targets somatostatin receptors overexpressed on neuroendocrine tumour cells.
* It shows receptor-mediated localisation at the tumour site after systemic distribution.
M – Metabolism
* The radiolabelled compound remains largely intact in plasma; minimal hepatic metabolism.
* This stability is important to ensure clear imaging signal at the target site.
E – Excretion
* Primarily via the renal route, with the compound largely excreted unchanged in urine.
* Rapid clearance from non-target tissues supports a favourable target-to-background ratio in PET imaging. - ADME in Theory (e.g. for oral tablet products)
A – Absorption
* Absorbed through the gastrointestinal tract, depending on solubility, permeability, and formulation factors (e.g. excipients, pH).
D – Distribution
* Drug distributes via systemic circulation, often binds to plasma proteins (e.g. albumin) before reaching target tissues.
M – Metabolism
* Mainly occurs in the liver by cytochrome P450 enzymes (e.g. CYP3A4, CYP2D6), converting the parent drug into active/inactive metabolites.
E – Excretion
* Eliminated via renal (urine) or hepatic (bile) pathways, sometimes as unchanged drug or metabolite. - Why ADME Knowledge is Important for a QP
As a QP, understanding ADME is essential for:
* Product knowledge: Understanding how the drug behaves in the body informs risk-based decision-making in manufacturing and QA.
* Deviation or defect investigation: If a product defect occurs (e.g. out-of-spec impurity, incorrect strength), knowing the ADME helps assess whether it may affect efficacy or patient safety.
* Shelf-life and impurity assessment: Helps assess the clinical impact of impurity profiles or stability trends, particularly for systemic vs locally acting drugs.
* Bioavailability and route of administration: Guides understanding of why certain specs or manufacturing steps are critical (e.g. particle size for oral absorption, sterility for IV injection).
Summary:
“ADME gives me, as a QP, the necessary background to assess product quality in the context of patient risk. It helps me bridge GMP issues with pharmacological impact — particularly important when dealing with deviations, shelf-life decisions, or novel formulations like radiopharmaceuticals.”
Please talk about the controls around the manufacturing of the product?
As a QP, I would expect comprehensive controls covering the facility, personnel, equipment, materials, and process parameters to ensure aseptic assurance and product quality, in line with EU GMP Annex 1, ISO 14644, and radiation safety guidance.
- Environmental and Facility Controls
* Grade A environment (typically in an isolator or RABS) for aseptic filling, qualified as per ISO 14644-1 (≤3,520 particles ≥0.5 µm/m³ in operation).
* Pressure differentials of 15–20 Pa maintained between rooms of different grades to ensure directional airflow from cleaner to less clean areas.
Air change rate:
o ≥20 ACH (air changes per hour) in Grade B and C areas to maintain air cleanliness.
* Laminar airflow velocity:
o Typically 0.45 m/s ±20% in Grade A areas to ensure unidirectional flow over critical zones.
* Isolator integrity testing and glove integrity testing performed routinely to prevent breach of sterile barrier. - Personnel and Operational Controls
* Line clearance:
o Performed and documented before each batch to confirm that the area, equipment, and materials are free from previous product, labels, or contaminants.
o It is a critical GMP step to prevent cross-contamination and mix-ups.
* Operator aseptic technique:
o All personnel are trained, qualified, and routinely assessed.
* Transfer technique:
o Materials transferred into Grade A via validated sporicidal wiping, pass-through hatches with unidirectional flow.
* Gowning procedures:
o Grade B gowning for entry into clean areas; strict procedures aligned with Annex 1.
* Media fill validation:
o Conducted under worst-case conditions to demonstrate the aseptic process can reliably produce sterile product. - Process and Material Controls
* Sterile filtration:
o Final filtration using validated 0.22 µm filter, with integrity testing (e.g. bubble point) before and/or after use.
* Low bioburden starting materials:
o Ensures pre-filtration microbial load is within defined limits.
* Temperature control:
o Controlled room temperature and validated temperature of reaction vessel during synthesis.
* Radioactivity in-process checks:
o Ensures correct activity per dose, labelling efficiency, and radiochemical purity. - Environmental Monitoring and Trending
* Non-viable particle monitoring:
o Continuous in Grade A; routine in Grade B/C.
- Viable monitoring:
o Air sampling, settle plates, and contact plates during and after operations. - Excursions investigated as deviations; data are trended for early detection of control loss.
Critical Process Parameters (CPPs) Include:
- Line clearance verification
- Isolator and glove integrity
- Filter integrity (pre-/post-use)
- Operator aseptic technique
- Transfer disinfection steps
- Pressure differentials and air velocity
- Environmental monitoring results
- In-process radioactivity checks
- Room and process temperature control
Summary:
“In aseptic radiopharmaceutical manufacturing, strict control of the environment, operators, materials, and process is essential. Line clearance is a key GMP step to prevent cross-contamination. As a QP, I would review these controls, batch records, and any deviations to ensure product sterility and compliance before certifying the batch.”
What do you do when MACO is breached?
- I would raise a deviation immediately and quarantine any potentially affected product.
- I’d assess the extent of the failure and the products involved, conduct a risk assessment to evaluate the impact on patient safety and GMP compliance.
- Determine if any product released prior to the result could be affected — if so, initiate a recall assessment and engage with the Medical and Regulatory teams.
- Root cause investigation would follow — for example, ineffective cleaning, equipment design, operator error, or analytical method issue.
- Implement CAPAs: this may include revalidation, retraining, adjustment of procedures or cleaning agents, or redesign of equipment.”
Cleaning validation – Multi product oral suspension manufacturing site. MACO levels breached on annual cleaning validation.
What is cleaning validation, and why is it critical in a multi-product facility?
Cleaning validation is a documented process that demonstrates the effectiveness and reproducibility of cleaning procedures to prevent cross-contamination.
It’s especially critical in a multi-product facility where different APIs may have varying potencies, toxicity profiles, and carryover risks.
As a QP, I must ensure that cleaning procedures consistently remove product residues, excipients, and cleaning agents to levels that are safe, regulatory compliant, and not detectable beyond the MACO limit.
What are recovery studies, and how are they used in cleaning validation?
Recovery studies are performed to determine the efficiency of the sampling method, typically swabbing or rinsing.
Coupons made of the same material as the equipment are spiked with known amounts of residue, allowed to dry, and then sampled.
The percentage of recovered residue indicates how effective the sampling is.
The recovery factor is then applied to actual sample results. For example, if you recover 80%, the actual residue may be higher — so your result is adjusted by dividing by 0.8.
Low recovery can either invalidate the result or require more stringent cleaning targets
How to MACO calculations?
MACO stands for Maximum Allowable Carryover. It can be calculated in several ways, but the traditional approach is:
MACO = (PDE of previous product x Minimum batch size of the next product /Max daily dose of product B
The calculated value is then converted into a surface-specific acceptance limit based on equipment surface area or swabbed area, and cleaning validation results must fall below this value.
What is the difference between swab and rinse sampling? When would you use each?
Swab sampling is used to test small, hard-to-clean areas like joints, valves, and dead-legs. It provides a direct, localized measure of surface residue.
Rinse sampling captures residues from the entire internal surface area of equipment, especially where swabbing is impractical, such as pipes or reactors.
Both methods are complementary. In a multi-product facility, I would typically use both — swabbing for worst-case locations and rinses for overall system assurance
A Clean-in-Place (CIP) system failure was discovered in a multi-product facility — the spray ball in a critical vessel failed to rotate, resulting in an ineffective clean. Batches were released before this failure was identified. You’re the QP — what do you do?
My immediate priority is to assess the impact of the cleaning failure on product quality and patient safety.
I would:
1. Quarantine any impacted or in-process batches.
2. Initiate a deviation and investigation.
3. Assess how long the spray ball had been failing — i.e. what batch(es) may be affected.
4. Review cleaning validation and visual inspection records.
5. Evaluate whether the failure led to cross-contamination, microbial risk, or cleaning agent residues.
6. Assess if any affected batches have already been released
What validation aspects would you consider here?
This incident highlights a weakness in the cleaning system’s design qualification and routine verification.
As part of cleaning validation lifecycle per Annex 15, I would review:
* Installation Qualification (IQ) of the spray ball and CIP system.
* Operational Qualification (OQ) — was the rotation verified during validation?
* Performance Qualification (PQ) — was spray coverage assessed?
* Preventive maintenance frequency and whether spray ball rotation was part of routine checks.
* Visual inspection training and documentation for cleaning operators.
The outcome is a Class 1/Class 2 recall. What’s the difference between them?
- Class 1 recall: Life-threatening risk. Immediate recall needed. Examples: contamination with a toxic compound, undeclared allergen, microbial contamination in sterile product.
- Class 2 recall: Could cause illness or mistreatment, but not life-threatening. Still serious, but not immediate threat.
In this case, if the cleaning failure involved potential contamination with high-risk materials (e.g. penicillin, cytotoxic, or microbial residue in a sterile product), a Class 1 recall is appropriate.
What is your role as a QP in this recall situation?
As the QP, I must:
* Lead the product quality risk assessment.
* Initiate internal notification processes.
* Halt further distribution.
* Assess whether any other batches may be affected.
* Contribute to the recall classification discussion.
* Ensure appropriate communication with the QPPV if the product is licensed and patient safety is involved.
* Work with regulatory colleagues to notify DMRC (Defective Medicines Report Centre).
* Provide technical input for recall documentation and risk evaluation.
What would you present to the DMRC (what would you include in my proposal)?
Reference: MHRA blog
- Description of defect
- Risk/Impact assessment
- Recall class (1-4)
- Recall extend (wholesaler, pharmacy, patient)
- Close report submited after complete of CAPA
Can you explain the concept of Pharmacokinetics and ADME?
Explain the Concept of Pharmacokinetics and ADME
Pharmacokinetics (PK) is the study of how the body affects a drug - drug absoebed, distributed, metabolised and excreted - over time.
It describes the movement of a drug through the body using four key processes, summarised by the acronym ADME:
________________________________________
ADME – Key Components of Pharmacokinetics:
1. Absorption
o How the drug enters the bloodstream after administration.
o Depends on the route (e.g. oral, IV, IM) and formulation (e.g. tablet, injection).
o Influenced by solubility, permeability, and first-pass metabolism.
2. Distribution
o How the drug spreads through the body from the blood to tissues.
o Affected by blood flow, tissue binding, and plasma protein binding (e.g. to albumin).
3. Metabolism
o How the drug is chemically modified, primarily in the liver by enzymes (e.g. cytochrome P450s).
o Produces active or inactive metabolites.
4. Excretion
o How the drug or its metabolites are removed from the body, typically via the kidneys (urine) or bile (faeces).
Model Answer: What is Pharmacodynamics?
Pharmacodynamics (PD) is the study of how a drug affects the body — in contrast to pharmacokinetics, which is how the body affects the drug. (such as recepter binding and therapeuticresponce)
________________________________________
Key Elements of Pharmacodynamics:
1. Mechanism of Action (MoA)
o How the drug interacts with its biological target (e.g. receptor, enzyme, ion channel)
o E.g., beta-blockers binding to β-adrenergic receptors to reduce heart rate
2. Dose–Response Relationship
o Describes how the drug effect changes with dose
o Includes parameters like:
EC50: concentration at which 50% of maximal effect is observed
Emax: maximum achievable effect
3. Therapeutic Effect vs Side Effects
o PD helps define the therapeutic window — the range between effective and toxic doses
o Also helps understand on-target vs off-target effects
Can you give examples of the different routes of excretion?
Drugs and their metabolites can be excreted from the body through several primary and secondary (minor) routes:
⸻
Primary Routes:
1. Renal (Kidney – Urine)
• Most common route for water-soluble drugs and metabolites
• Examples: aminoglycosides, penicillin
• Can involve glomerular filtration, tubular secretion, or reabsorption
2. Hepatic (Biliary – Faeces)
• Lipophilic drugs/metabolites are excreted into bile and passed in faeces
• May undergo enterohepatic recycling
• Examples: rifampicin, some steroids, digoxin
⸻
Secondary/Minor Routes:
3. Saliva
• Can be used in drug monitoring
• Some drugs (e.g. lithium, phenytoin) may be measurable in saliva
4. Sweat glands
• Minor excretion route
• Can cause skin irritation or odour (e.g. garlic, some chemotherapy drugs)
5. Tears
• Very minimal excretion
• Some drugs may cause ocular side effects due to presence in tear fluid
6. Breast milk
• Clinically important in lactating patients
• Lipophilic, weakly basic drugs can accumulate
• Examples: diazepam, codeine, some antibiotics
• Important for QP to assess during SmPC review if labelling error or exposure occurs
7. Exhalation (Lungs)
• Volatile or gaseous drugs (e.g. anaesthetics, alcohol)
• Also important for radiopharmaceuticals using gaseous isotopes
⸻
Summary:
“The main excretion routes are renal and biliary, but other routes like sweat, saliva, breast milk, and breath may be relevant for certain drugs. Understanding excretion is important for assessing the impact of impurities, drug accumulation, or off-label use — all relevant for QP batch release and risk assessment.”
Can you describe wat bioavailability is vs bioequivalence?
- Bioavailability
Bioavailability refers to the proportion (%) of the active pharmaceutical ingredient (API) that is absorbed into the systemic circulation after administration and is available to exert its therapeutic effect.
• For intravenous (IV) administration: bioavailability is 100%
• For oral and other non-IV routes: bioavailability is often less than 100% due to factors like:
• Incomplete absorption
• First-pass metabolism in the liver or gut wall
• Degradation in the GI tract
Example: Oral morphine has lower bioavailability (~30%) compared to IV morphine due to significant first-pass metabolism.
⸻
- Bioequivalence
Bioequivalence refers to the comparison between two drug products (usually a generic vs reference product) to show that they have similar bioavailability, and therefore similar efficacy and safety profiles.
• It is established by comparing key pharmacokinetic parameters:
• Cmax (peak concentration in plasma)
• Tmax (time to reach peak)
• AUC (overall exposure over time)
• Regulatory standards typically require the 90% confidence interval for the ratio (test/reference) of Cmax and AUC to fall within 80%–125%
Example: A generic simvastatin must demonstrate bioequivalence to the branded product (Zocor) to be approved for substitution.
⸻
Summary for Viva:
“Bioavailability is the percentage of the API that reaches systemic circulation. Bioequivalence compares two products to confirm they have similar bioavailability and are therefore interchangeable in terms of clinical effect.”
What products have good bioavailability and which have bad?
Products with Good Bioavailability:
1. Intravenous (IV) injection or infusion
• 100% bioavailability by definition — drug is delivered directly into systemic circulation
• Example: morphine IV, radiopharmaceutical injections (e.g. Ga-68)
2. Intramuscular (IM) or Subcutaneous (SC) injections
• High bioavailability (often >90%), but slower onset than IV
• Example: insulin SC
3. Buccal or sublingual (oromucosal) dosage forms
• Bypasses first-pass metabolism; fast absorption
• Example: glyceryl trinitrate sublingual tablets, buprenorphine films
4. Inhaled products
• Rapid absorption through pulmonary route
• Example: salbutamol inhalers
⸻
Products with Poor or Variable Bioavailability:
1. Oral dosage forms (e.g. tablets, capsules)
• Subject to first-pass metabolism, pH effects, solubility, and GI motility
• Bioavailability can range from very low to high depending on the drug
• Examples:
• Low: propranolol (~30%), bisphosphonates (~1%)
• High: paracetamol (~80–90%)
2. Rectal dosage forms
• Variable bioavailability depending on placement and formulation
• Partial avoidance of first-pass metabolism
• Example: diazepam rectal gel (moderate bioavailability)
3. Topical formulations
• Generally poor systemic absorption unless designed for transdermal delivery
• Example: hydrocortisone cream (local), vs fentanyl patch (systemic)
⸻
Summary:
“Products administered IV have the highest bioavailability. Oromucosal and parenteral routes also tend to have high absorption. Oral and rectal forms show variable or reduced bioavailability due to first-pass metabolism and formulation factors. As a QP, understanding this helps assess formulation performance and risk during deviation or change control.”
Can you explain which Phase of Clinical Trials is likely to carryout Pharmacokinetics studies?
Pharmacokinetic (PK) studies are primarily carried out during the preclinical and Phase 1 stages of drug development.
⸻
- Preclinical Phase (Non-clinical Studies)
• PK studies are conducted in animal models to understand:
• Absorption, Distribution, Metabolism, and Excretion (ADME)
• Dose–exposure relationships
• Potential accumulation or toxicity
• Data is used to design safe starting doses for first-in-human studies.
⸻
- Phase 1 (First-in-Human Studies)
• Conducted in healthy volunteers (except for oncology or high-risk drugs)
• Aim is to assess:
• PK parameters: Cmax, Tmax, AUC, half-life (t½), clearance, volume of distribution
• Safety and tolerability
• Bioavailability and dose proportionality
This phase provides human PK data to support dose selection for later phases.
⸻
PK Studies in Later Phases (if applicable):
• Phase 2/3: PK studies may still be performed to:
• Assess PK in specific patient populations (e.g. renal/hepatic impairment)
• Evaluate drug interactions or food effects
• Phase 4: May include population PK or special subgroup analysis
⸻
Summary:
“Pharmacokinetic studies are conducted in the preclinical phase in animals and in Phase 1 in humans to understand how the drug behaves in the body. This supports safe dose selection and informs formulation development — which is critical knowledge for a QP during IMP certification.”
Can you explain the concept of Therapeutic Index (Narrow vs wide therapeutic window) and give examples of drugs that have a narrow therapeutic window?
The therapeutic index (TI) is a measure of a drug’s safety margin. It describes the range between the dose that produces a therapeutic effect and the dose that causes toxic or adverse effects.
⸻
Definition:
• Therapeutic Index (TI) = TD₅₀ / ED₅₀
• TD₅₀ = dose that causes toxicity in 50% of the population
• ED₅₀ = dose that produces the desired effect in 50% of the population
In clinical practice, we often refer to the therapeutic window, which is the plasma concentration range where the drug is effective without being toxic.
⸻
Narrow Therapeutic Index (NTI) Drugs:
• These drugs have a small margin between effective and toxic doses, so close monitoring is essential to avoid under- or overdosing.
• Require plasma concentration monitoring, dose adjustment, or patient-specific management.
Examples:
• Warfarin – risk of bleeding if too high, clotting if too low (monitored by INR)
• Digoxin – small margin between therapeutic and toxic cardiac effects
• Carbamazepine – seizure control vs CNS toxicity
• Lithium – used in bipolar disorder, requires regular serum level monitoring
• Theophylline – used in asthma/COPD, narrow margin of safety
⸻
Wide Therapeutic Index Drugs:
• Large margin between effective and toxic doses
• Less risk if plasma levels fluctuate
Examples:
• Paracetamol (within standard dosing)
• Penicillin
• Ibuprofen (in healthy adults, within dosing guidelines)
⸻
Why It Matters for a QP:
“Understanding therapeutic index helps the QP assess the impact of deviations or OOS results. For NTI drugs, even small manufacturing or dosing errors can have clinical consequences, so the risk-based approach to batch certification must be stricter.”
What is NOEL?
NOAEL stands for No-Observed-Adverse-Effect Level.
It is the highest dose or exposure level of a substance at which no adverse effects are observed in the test population during non-clinical (toxicology) studies, typically in animals.
⸻
Key Points:
• It is established from repeat-dose toxicity studies.
• Represents the threshold below which no harmful effects are seen, even with prolonged exposure.
• Reported in mg/kg/day for the tested species.
⸻
Why is NOAEL important?
In the pharmaceutical industry, NOAEL is used to calculate the Permitted Daily Exposure (PDE) as per ICH Q3C and EMA guidelines.
PDE =
NOAEL × Body weight (kg)
÷
F1 × F2 × F3 × F4 × F5
Where F1–F5 are uncertainty (safety) factors based on:
• Species differences
• Study duration
• Route of administration
• Nature of toxicity
• Quality of data
⸻
QP Relevance:
“As a QP, I would use NOAEL-derived PDE values to evaluate cross-contamination risks in shared facilities and to ensure cleaning validation meets health-based exposure limits (HBELs), in line with Annex 15 and EMA Q&A on cross-contamination.”
What is ICH M7 and TTC?
What is difference between TTC and PDE?
ICH M7
ICH M7(R1) is a guideline titled:
“Assessment and Control of DNA Reactive (Mutagenic) Impurities in Pharmaceuticals to Limit Potential Carcinogenic Risk”
It provides a risk-based approach for evaluating and controlling mutagenic impurities (MIs) that may pose a carcinogenic risk to patients, even at low levels.
⸻
Key Concepts in ICH M7:
• Focuses on DNA-reactive impurities (e.g. alkylating agents, aromatic amines)
• Encourages the use of QSAR tools and bacterial mutagenicity assays (e.g. Ames test)
• Supports use of TTC (Threshold of Toxicological Concern) as an acceptable limit for unknown or unqualified mutagenic impurities
⸻
TTC – Threshold of Toxicological Concern:
• TTC is a risk-based exposure limit used when specific toxicological data are lacking
• For most pharmaceuticals, ICH M7 defines the TTC for genotoxic impurities as:
• 1.5 µg/day
• This corresponds to a theoretical lifetime cancer risk of <1 in 100,000
• The limit may vary:
• Higher for short-term exposure (≤1 month)
• Lower for high-potency carcinogens (e.g. nitrosamines, aflatoxins)
⸻
Why This Is Important for a QP:
“As a QP, I must ensure that any known or potential mutagenic impurities are assessed under ICH M7. If no toxicological data are available, I would apply the TTC limit of 1.5 µg/day and ensure that analytical methods are suitably sensitive. This is critical for patient safety and batch release decisions.”
What is the difference between PDE and TTC?
- PDE – Permitted Daily Exposure
• Defined in ICH Q3C, Q3D, and EMA cross-contamination guidelines
• Represents the safe daily exposure limit to a substance based on actual toxicological data (usually NOAEL)
• Used for known, well-characterised substances
• Applied in:
• Cleaning validation (e.g. shared equipment)
• Elemental impurity limits (ICH Q3D)
• Residual solvents (ICH Q3C)
• Cross-contamination risk assessment (Annex 15)
Calculated from NOAEL using safety factors (F1–F5)
⸻
- TTC – Threshold of Toxicological Concern
• Defined in ICH M7(R1)
• Used for DNA-reactive (mutagenic) impurities when substance-specific toxicology data are unavailable
• TTC is a generic conservative limit, set to ensure lifetime cancer risk is <1 in 100,000
• For most drugs:
• TTC = 1.5 µg/day
• Not suitable for:
• High-potency carcinogens (e.g. nitrosamines, aflatoxins)
• Paediatric populations
PDE is a data-driven, substance-specific limit used for general toxicity. TTC is a default conservative limit used in ICH M7 for genotoxic impurities when no data exist. Both are important tools for ensuring patient safety — as a QP, I must apply the appropriate one based on the impurity type and data availability.”
New product in multipurpose facility, how would you do it?
- Initiate Change Control (CC)
• Raise a formal change control to assess and document the full impact of introducing the new product across the site.
• This would trigger risk assessments across quality, operations, regulatory, validation, and safety functions.
⸻
- Regulatory Impact Assessment
• Marketing Authorisation (MA):
• Assess if changes affect the MA (e.g. site of manufacture, equipment, testing, batch size).
• Submit variation if required.
• MIA(Manufacturing Licence):
• Check whether the MIA includes the formulation and dosage form of the new product.
• Submit an MIA variation to the MHRA if needed.
⸻
- GMP Impact Assessment
a. Facility & Equipment
• Is it a potent or sensitising product?
• Assess whether a dedicated facility or closed system is needed.
• Review HVAC segregation, pressure differentials, and airflow patterns.
b. Personnel
• Training needs for new product, process, and safety risks
• Perform COSHH assessment for operator exposure and PPE requirements
c. Documentation
• Update or create:
• CCS (Contamination Control Strategy)
• Cleaning SOPs
• Master Batch Records
• Validation protocols
d. Manufacturing Process
• Assess cleaning validation requirements:
• Perform risk assessment using worst-case approach across product matrix
• Use equipment train mapping to identify shared surfaces
• Assess cleanability, solubility, and potency
• If still worst-case: execute cleaning validation runs
• Line clearance procedures may need updating or strengthening
e. Quality Control (QC)
• Assess analytical method validation needs for new product
• Review equipment suitability and detection limits for residual testing
f. Outsourced Activities
• If any part is outsourced (e.g. testing, intermediate manufacture), verify:
• Supplier approval
• Technical agreements
• Audit reports
⸻
- Cross-Contamination Risk and Cleaning Validation
• Conduct a cleaning validation risk assessment using:
• Toxicological data (e.g. NOAEL)
• Calculate PDE and MACO (Maximum Allowable Carryover)
• Evaluate cleaning method effectiveness (e.g. using swab/rinse recovery studies)
• Compare new product’s cleanability to existing worst-case
• If it becomes the new worst-case, full cleaning validation is required
⸻
- Impact on Other Products
• Assess risk of cross-contamination or product mix-up
• Reassess product-to-product sequence rules
• Review EM data and environmental capability to handle the new product
⸻
Summary for Viva:
“I would initiate a formal change control and perform a comprehensive impact assessment across regulatory, facility, process, and quality systems. I’d evaluate cross-contamination risk using PDE-based cleaning validation principles, ensure training and documentation updates, and verify that the site remains compliant with GMP and the MIA.”
What is first pass metabolism?
First-pass metabolism (also called first-pass effect) refers to the metabolism of a drug before it reaches the systemic circulation, primarily when the drug is administered orally.
⸻
How it works:
• After oral administration, the drug is absorbed from the gastrointestinal (GI) tract into the portal vein, which carries blood directly to the liver.
• In the liver, the drug undergoes enzymatic metabolism (mainly by cytochrome P450 enzymes).
• As a result, a significant portion of the drug may be inactivated or transformed into metabolites before reaching systemic circulation.
⸻
Impact on Bioavailability:
• First-pass metabolism reduces the bioavailability of the drug — meaning less active drug reaches the bloodstream.
• Drugs with high first-pass effect may require:
• Higher oral doses
• Alternative routes (e.g. sublingual, rectal, or IV)
⸻
Examples of drugs with high first-pass metabolism:
• Propranolol
• Glyceryl trinitrate (GTN) — given sublingually to avoid first-pass effect
• Morphine
• Lidocaine
⸻
Summary for Viva:
“First-pass metabolism refers to the liver’s enzymatic breakdown of a drug after oral administration and before it reaches systemic circulation. It reduces bioavailability and must be considered when designing dosage forms or evaluating formulation changes — which is highly relevant for QP decision-making.”