ICH Flashcards

(115 cards)

1
Q
  1. ICH S1A – Need for Carcinogenicity Studies: Which scenario does NOT typically require a long-term rodent carcinogenicity study under ICH S1A?
    A. A new antihypertensive drug intended for daily use over many years.
    B. A chronic therapy for diabetes to be taken continuously for >6 months.
    C. A short-course antibiotic intended for a single 10-day treatment.
    D. An antiviral for chronic hepatitis C given daily for one year.
A

Answer: C. Explanation: ICH S1A specifies that carcinogenicity studies are generally warranted if the clinical use is continuous for 6 months or longer (or in an intermittent/repeated manner of similar total duration) . Drugs used only for short durations (e.g. a single 1-2 week course) usually do not require 2-year rodent carcinogenicity bioassays, unless there is other cause for concern (such as positive genotoxicity or a suspicious drug class) . In this question, options A, B, and D all involve chronic long-term use (clearly exceeding 6 months), which would trigger carcinogenicity testing, whereas a short 10-day antibiotic course (option C) would not require such studies under normal circumstances.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
2
Q
  1. ICH S1B – Carcinogenicity Testing Approaches: Under ICH S1B guidance, which of the following is an acceptable approach to fulfill rodent carcinogenicity testing requirements for a new pharmaceutical?
    A. One long-term 2-year study in rats plus a 6-month study in a transgenic mouse model (instead of a 2-year mouse study).
    B. Two separate 6-month studies in rats (in place of any mouse study).
    C. A single 3-month study in one rodent species, if doses are high enough.
    D. A 1-year study in monkeys instead of any rodent carcinogenicity study.
A

Answer: A. Explanation: ICH S1B allows flexibility in how the carcinogenic potential is evaluated. The traditional approach is two long-term studies (usually 2-year studies in both rats and mice). However, ICH S1B introduced the option to replace the second rodent long-term study with a shorter study in a susceptible transgenic mouse model (approximately 6 months duration) . For example, a 2-year rat study plus a 6-month transgenic mouse assay (such as the Tg.rasH2 mouse) is an acceptable testing strategy that adheres to the 3Rs (reduce/refine/replace) by avoiding a full 2-year mouse study . Other choices listed are not aligned with ICH guidelines: two short rat studies or a single 3-month study would be insufficient, and substituting a primate study is not the recommended paradigm for carcinogenicity assessment.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
3
Q
  1. ICH S1C(R2) – High-Dose Selection in Carcinogenicity Studies: Which of the following is NOT a recommended criterion for selecting the high dose in rodent carcinogenicity studies according to ICH S1C(R2)?
    A. A dose that represents the Maximum Tolerated Dose (MTD) with only minimal toxicity observed .
    B. A dose that achieves approximately a 25-fold exposure (AUC) relative to the maximum human exposure .
    C. A dose that causes 50% mortality in a 90-day toxicity study (approximate LD50).
    D. A limit dose of about 1500 mg/kg/day in rodents, if no toxicity is seen and the human dose is low (≤500 mg/day) .
A

Answer: C. Explanation: ICH S1C(R2) outlines several science-based criteria for high-dose selection in carcinogenicity studies, including use of the MTD (Maximum Tolerated Dose) – defined as the highest dose causing only minimal toxic effects over the study duration , a pharmacokinetic exposure multiple (approximately 25× the human plasma AUC, if metabolism is similar) , and a limit dose (usually 1000–1500 mg/kg/day for rodents when the maximum human dose is ≤500 mg/day) in cases where toxicity does not occur even at very high doses . These approaches ensure an adequately high dose without excessive toxicity. In contrast, selecting a dose that causes 50% lethality (option C) is not recommended – lethality (LD50) is far beyond the minimal toxicity threshold and would confound the study (and raises ethical concerns). In fact, guidelines explicitly aim to avoid doses that cause significant mortality or moribundity in long-term studies .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
4
Q
  1. ICH S2(R1) – Standard Genotoxicity Test Battery: Which of the following is NOT part of the standard genotoxicity test battery recommended by ICH S2(R1)?
    A. A bacterial reverse mutation test (e.g., Ames test) to detect gene mutations in bacteria.
    B. An in vitro test for chromosomal damage, such as an in vitro micronucleus or metaphase chromosome aberration test in mammalian cells.
    C. An in vivo test for genetic damage (usually in rodent hematopoietic cells), such as an in vivo micronucleus assay.
    D. An in vivo dominant lethal mutation test in rodents to detect germ-cell mutations affecting fertility.
A

nswer: D. Explanation: The ICH S2(R1) guideline describes a standard battery of genotoxicity assays that typically includes: (i) a test for gene mutation in bacteria (the Ames test), (ii) a test for chromosomal aberrations or mutations in mammalian cells in vitro (e.g., in vitro chromosome aberration test, in vitro micronucleus, or mouse lymphoma tk assay), and (iii) an in vivo test for chromosomal damage (usually an in vivo rodent micronucleus assay in bone marrow or an in vivo chromosome aberration assay) . This battery aims to cover gene mutations, chromosomal breaks/clastogenicity, and aneugenicity. A dominant lethal test (option D) is an older assay detecting germ cell mutations by observing embryonic loss after mating treated males – it is not routinely included in the standard test battery for pharmaceuticals . Dominant lethal assays or other specialized germ-cell tests would only be conducted if there is a specific cause for concern (e.g., a positive finding that warrants further investigation of heritable mutations), not as a screening requirement for all compounds.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
5
Q
  1. ICH S3A – Toxicokinetics in Animal Studies: True or False – ICH S3A recommends that assessments of systemic exposure (toxicokinetics) be incorporated into repeat-dose toxicity studies in order to relate animal exposure to toxicological findings.
A

Answer: True. Explanation: ICH S3A (Toxicokinetics) emphasizes the importance of measuring drug concentrations in animals during toxicity studies. These toxicokinetic (TK) data are used to confirm systemic exposure, help interpret toxicity in relation to dose, and to compare exposure margins between animals and humans . In fact, S3A states that TK analysis is a required component of pivotal repeat-dose tox studies to ensure that observed toxic effects can be correlated with blood levels of the drug . By characterizing the pharmacokinetics in test species, one can determine if adequate exposure has been achieved, identify non-linear kinetics or accumulation, and support species selection and risk assessment.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q
  1. ICH S3B – When to Conduct Repeated-Dose Tissue Distribution Studies: ICH S3B outlines certain circumstances where dedicated repeated-dose tissue distribution studies are warranted. Which of the following situations would justify performing a repeated-dose tissue distribution study?
    A. Single-dose distribution data showed that drug (or metabolite) persists in certain organs with a half-life much longer than its plasma half-life, suggesting significant accumulation upon repeat dosing .
    B. The drug is rapidly eliminated and one-dose distribution indicates no specific organ retention.
    C. The compound is a small molecule with well-understood ADME and no unexpected toxicology findings.
    D. Routine performance of a 2-week distribution study in all new chemical entities (as a default requirement).
A

Answer: A. Explanation: ICH S3B (Guidance on Repeat-Dose Tissue Distribution) does not require tissue distribution studies for every compound; rather, it identifies specific triggers for when such studies may be informative. One trigger is if single-dose distribution studies reveal that a compound or its metabolites have an unusually long half-life in certain tissues – for example, if the apparent tissue half-life is much longer than the plasma elimination half-life and exceeds the dosing interval in toxicity studies . In such a case, a repeated-dose distribution study can determine whether the drug accumulates in those organs upon chronic dosing, which could explain or predict toxicity. Other triggers (not listed in the options) include unexpected target organ toxicity that wasn’t predicted by single-dose kinetics, or when the drug is designed for site-specific delivery (to verify targeting) . Options B and C describe scenarios of no particular concern (rapid elimination, predictable behavior), where additional distribution studies are usually unnecessary. Option D is incorrect because there is no blanket requirement for routine repeated-dose distribution studies; they are only done when justified by a weight-of-evidence that they would yield important safety information .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q
  1. ICH S4 – Duration of Chronic Toxicity Studies: According to ICH S4, what are the recommended durations for chronic toxicity studies in rodents and non-rodents (dogs/monkeys) for general pharmaceutical development?
    A. 6 months in rodents (e.g., rats) and 9 months in non-rodents .
    B. 12 months in rodents and 12 months in non-rodents.
    C. 3 months in rodents and 6 months in non-rodents.
    D. 6 months in rodents and 12 months in non-rodents (with a second 6-month study).
A

Answer: A. Explanation: ICH S4 harmonized the duration of long-term toxicity studies required in different regions. The guideline concluded that a 6-month study in rodents and a 9-month study in non-rodents are generally sufficient to evaluate chronic toxicity for pharmaceuticals intended for chronic use . This reduced previous regional disparities (for example, in some regions non-rodent studies of 12 months were once requested). Studies longer than 6 (rodent) or 9 (non-rodent) months are usually not necessary; a 12-month dog or monkey study, for instance, is not routinely needed if a 9-month study has been conducted . Options B, C, and D do not reflect the ICH-agreed durations: 12-month rodent studies are excessive (B), 3-month rodent is too short for a chronic setting (C), and 12 months in non-rodents (D) is beyond what is recommended in most cases, since 9 months is considered sufficient.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q
  1. ICH S5(R3) – Reproductive Toxicity Studies: Which of the following is NOT one of the standard study types included in ICH S5 for reproductive and developmental toxicity assessment?
    A. Fertility and early embryonic development study (to assess effects on adult male and female fertility and pre-implantation development).
    B. Embryo-fetal development study (teratology study, usually in two species, to assess developmental abnormalities in utero).
    C. Pre- and postnatal development study (to assess effects on late pregnancy, birth, and offspring growth to weaning).
    D. Juvenile animal toxicity study (to assess safety in post-weaning juvenile animals as they mature).
A

Answer: D. Explanation: ICH S5 covers the “reproductive and developmental toxicity” studies typically needed for new pharmaceuticals, which classically include: (1) a Fertility and Early Embryonic Development (FEED) study (Segment I) in adult males and females, (2) Embryo-Fetal Development (EFD) studies (Segment II) in pregnant animals – usually one rodent (rat) and one non-rodent (rabbit) – to detect teratogenicity and developmental toxicity, and (3) a Pre- and Postnatal Development (PPND) study (Segment III) in a rodent to assess effects on pregnancy, parturition, lactation, and offspring development . A juvenile animal study (JAS), on the other hand, is not part of this standard reproductive tox battery – it is a separate consideration addressed later by ICH S11. Juvenile toxicity studies are conducted only if pediatric development programs warrant them (to evaluate drug effects on growth and organ maturation outside the scope of S5 studies) . In summary, options A, B, and C correspond to the three core study segments required by ICH S5, whereas D (juvenile study) is governed by a different guideline (S11) and not automatically required for all drugs.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q
  1. ICH S6(R1) – Biotech Product Preclinical Testing: Identify the FALSE statement regarding preclinical safety evaluation of biotechnology-derived pharmaceuticals (per ICH S6):
    A. Conventional genotoxicity studies (mutagenicity tests) are usually not applicable to large protein biologics, since proteins are not expected to interact with DNA .
    B. The selection of animal species for toxicity testing should focus on pharmacologically relevant species – often only species that express the drug’s target (e.g., the receptor) are suitable . If no normal animal species is responsive, alternative approaches (using transgenic animals or homologous proteins) may be necessary .
    C. Immune responses (anti-drug antibodies) observed in animal studies do not reliably predict human immunogenicity; a biologic can cause antibodies in animals without the same occurring in humans (and vice versa) .
    D. Two-year rodent carcinogenicity studies are required for all biotechnology-derived proteins to evaluate tumorigenic risk (e.g. every monoclonal antibody must undergo a 2-year mouse study).
A

Answer: D. Explanation: ICH S6(R1) provides a tailored approach for biotech-derived drugs (such as peptides, proteins, monoclonal antibodies, etc.), acknowledging their unique properties. Statements A, B, and C are true reflections of S6 principles. Specifically, genotoxicity testing is generally not considered useful for high-molecular-weight proteins, as they do not penetrate cells or interact with DNA in the manner small molecules might . Likewise, species selection is critical: toxicity studies are usually done only in a species where the biologic is pharmacologically active (e.g., where the relevant epitope/receptor is present). Sometimes only one relevant species (like a non-human primate) can be used, and if no relevant species exists, one might use transgenic models or “surrogate” molecules that are active in animals . Immunogenicity is another consideration – animals often develop anti-drug antibodies to human proteins, but this is noted to not predict the clinical immunogenic response . The false statement is D: standard 2-year carcinogenicity bioassays are generally not required for biotech products. In fact, ICH S6 notes that such long-term carcinogenicity testing is usually inappropriate for protein therapeutics . Only in special cases (e.g., when a growth factor might have tumor-promoting activity or when there is particular cause for concern) might alternative tumorigenicity assessments be needed – otherwise, carcinogenicity studies can often be waived for biologics.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q
  1. ICH S7A – Safety Pharmacology Core Battery: ICH S7A defines a “core battery” of safety pharmacology studies that should be conducted to evaluate a drug’s effects on vital organ systems. Which set of functions is included in this core battery?
    A. Central Nervous System (CNS), Cardiovascular, and Respiratory system functions .
    B. Renal (kidney) function and hepatic (liver) function.
    C. Gastrointestinal motility and endocrine function.
    D. Immune system function (e.g., humoral and cellular immunity).
A

Answer: A. Explanation: The safety pharmacology “core battery” per ICH S7A focuses on the three critical life-support systems: CNS, cardiovascular, and respiratory . These correspond to evaluating a drug’s potential to cause neurofunctional changes (CNS effects on behavior, locomotion, reflexes, etc.), cardiovascular effects (especially on heart rate, blood pressure, and electrical conduction – including QT interval prolongation risk), and respiratory effects (on respiratory rate, airway resistance, etc.). Renal and hepatic effects (B) are generally assessed as part of general toxicology and are not part of the S7A core battery, though they can be investigated in supplemental studies if a concern arises. GI and endocrine effects (C) likewise may be done as supplemental safety pharmacology studies if warranted, but they are not in the mandatory core set . Immune function (D) falls under immunotoxicity (ICH S8) rather than safety pharmacology. Thus, option A is the correct answer, as it lists the vital systems that must be evaluated in the core battery before first-in-human trials .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q
  1. ICH S7B – Evaluating QT Interval Prolongation: ICH S7B provides guidance on nonclinical testing for delayed ventricular repolarization (QT prolongation). Which of the following approaches is recommended to assess a compound’s propensity to prolong the QT interval?
    A. A bacterial hERG gene mutation assay to detect hERG channel gene changes.
    B. An in vitro Ames test at high concentrations to see if QT prolongation occurs in bacteria.
    C. A 2-year rodent study with ECG monitoring at the end of study.
    D. An in vitro IKr (hERG) channel current assay to evaluate blockade of the hERG potassium channel, often supplemented by an in vivo QT study in animals .
A

Answer: D. Explanation: The propensity of a drug to delay cardiac repolarization (manifested as QT interval prolongation on the ECG) is assessed by a combination of in vitro and in vivo assays as outlined in ICH S7B. A key component is an in vitro hERG channel assay, which measures the drug’s ability to block the rapid delayed rectifier K^+ current (I_Kr) in cardiac cells (often using cloned hERG channels) . This is typically paired with an in vivo QT study in animals (such as telemetered dogs or primates, or conscious guinea pigs/rabbits) to see if QT interval is prolonged at high exposures . Together, these tests form an integrated risk assessment for QT prolongation risk, which is later correlated with clinical thorough QT (E14) studies. The other options are inappropriate: (A) A gene mutation assay is irrelevant to QT, (B) an Ames test won’t reveal electrophysiologic effects, and (C) a 2-year study is far too late and insensitive for detecting QT changes (cardiac safety pharmacology should be done much earlier). Thus, the correct approach involves targeted electrophysiology studies (like hERG) and dedicated in vivo cardiac safety studies as recommended by S7B .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q
  1. ICH S8 – Immunotoxicity Testing: If standard toxicity studies (which include exams of immune organ weights and histopathology) raise concerns that a new drug might suppress immune function, ICH S8 recommends additional specialized testing. Which of the following is a specific assay often used to investigate immunosuppressive effects in such cases?
    A. Rat micronucleus assay (bone marrow) to check immunosuppression.
    B. T-cell dependent antibody response (TDAR) assay – measuring antibody formation to a novel antigen in treated animals .
    C. hERG channel assay in lymphocytes.
    D. Bacterial endotoxin challenge test in vitro.
A

Answer: B. Explanation: Under ICH S8, a tiered approach is used for immunotoxicity. All drugs should be evaluated in routine tox studies for potential immune effects (via lymphoid organ histopathology, blood cell counts, etc.). If those or the drug’s pharmacology indicate a risk of immunosuppression, then functional immunotoxicity assays (second-tier tests) are conducted . One commonly recommended assay is the T-cell dependent antibody response (TDAR) assay, which evaluates the ability of an animal’s immune system to mount an antibody response to a harmless antigen (such as sheep red blood cells or KLH) as a proxy for intact immune function . Suppression of the TDAR indicates immunosuppressive effects on adaptive immunity. Other functional tests include natural killer (NK) cell activity, cytotoxic T-lymphocyte activity, macrophage phagocytosis, etc. . Option B (TDAR) is therefore correct. The distractors are not standard immunotoxicity assays: micronucleus tests detect chromosomal damage, not immune function; hERG assay examines cardiac channels, and an “endotoxin challenge” in vitro is not a typical ICH S8 method (host resistance models in vivo – challenging animals with pathogens – are a third-tier immunotoxicity approach, but this is much more involved than any option listed) .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q
  1. ICH S9 – Nonclinical Testing for Anticancer Drugs: ICH S9 addresses nonclinical study expectations for oncology drugs in patients with advanced cancer. Which nonclinical study can often be omitted or deferred for a drug intended to treat late-stage, life-threatening cancer, according to ICH S9 guidelines?
    A. A standard 2-year rodent carcinogenicity study prior to approval .
    B. Genetic toxicology (mutagenicity) tests before first-in-human trials.
    C. Short-term safety pharmacology (CV, CNS, respiratory assessments).
    D. 1-month repeat-dose toxicity studies in two species.
A

Answer: A. Explanation: ICH S9 recognizes that for drugs intended to treat patients with advanced, refractory cancers (who have serious, life-threatening disease and often limited life expectancy), certain long-term animal studies may not be necessary before approval. In particular, carcinogenicity studies are usually not required for such oncology drugs in the initial development for advanced disease . The reasoning is that the patients’ shortened lifespan and the severity of their illness make long-term cancer risk from the drug a lesser concern, and also many anticancer drugs are genotoxic by design (would cause tumors in rodents). Similarly, a “complete” reproductive toxicity program might be abbreviated or deferred – for example, embryo-fetal development studies might be done later or with a flexible timing – especially if patients are past reproductive age or can avoid pregnancy . In contrast, genotoxicity tests (B) are generally still required for anticancer drugs (to characterize mutagenic potential, since many chemotherapeutics are DNA-reactive). Basic safety pharmacology (C) and appropriate repeated-dose toxicology (D) are also conducted even for oncology agents, albeit with some flexibility in study design or duration. Thus, the 2-year rodent carcinogenicity bioassay is the study that ICH S9 most clearly states can usually be omitted for an oncology drug in the indicated population (unless the drug’s development moves into long-term adjuvant use or earlier-stage disease with cure potential, in which case additional studies would then be considered).

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q
  1. ICH S10 – Photosafety Evaluation: Under ICH S10, what triggers the need for photosafety (phototoxicity) testing of a pharmaceutical?
    A. Any compound that is intended for use in dermatology, regardless of structure.
    B. A compound (or its metabolite) that has significant light absorption in the UV–visible range (290–700 nm) – for example, a molar extinction coefficient >1000 L·mol^−1·cm^−1 – and reaches tissues like skin or eyes during exposure .
    C. Compounds that are colored or fluorescent.
    D. Any drug that will be administered to patients who go outdoors.
A

Answer: B. Explanation: ICH S10 defines a risk-based approach to photosafety. The first consideration is the drug’s photochemical properties: specifically, whether it absorbs light in the UV or visible spectrum (290–700 nm) above a certain threshold (a molar extinction coefficient >1000 L·mol^−1·cm^−1 is used as a guideline trigger) . If a compound does not significantly absorb in this range, it is incapable of photochemical excitation and no photosafety testing is needed . If it does absorb, the next question is whether it can distribute to light-exposed tissues (skin or eyes) at sufficient concentrations. Only if both conditions are met (substantial UV/Vis absorption and tissue exposure) is a phototoxicity assessment warranted. In such cases, an in vitro phototoxicity assay (like the 3T3 Neutral Red Uptake phototoxicity test) is typically performed first. Options A and D are overly broad – not all dermatology drugs need phototesting (e.g., if they don’t absorb UV) and obviously any drug might be used in patients who go outdoors, but we don’t test everything — we focus on those with photochemical properties. Option C (color/fluorescence) is not itself a criterion; it sometimes correlates with absorption, but the actual trigger is quantitative absorption above the specified threshold. Thus, option B correctly captures the ICH S10 trigger: intrinsic UV/visible absorption potential plus exposure of those tissues in vivo.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q
  1. ICH S11 – Pediatric Safety (Juvenile Animal Studies): When does ICH S11 indicate that a juvenile animal toxicity study (JAS) should be considered during drug development?
    A. In all cases for drugs that will eventually be given to any pediatric population (required automatically).
    B. Only if the drug is overtly genotoxic in adults.
    C. When children (particularly very young or infants) are a target population and the existing adult animal and human data do not adequately address developmental safety for critical growing organ systems .
    D. Only if requested by regulatory authorities post-market.
A

Answer: C. Explanation: ICH S11 provides guidance on when and how to conduct juvenile animal studies to support pediatric drug development. It does not mandate a juvenile study for every pediatric program. Instead, it advocates a weight-of-evidence (WoE) approach: consider the pharmacology of the drug, the age of the pediatric population, and what is already known from adult animal studies and any clinical data . A juvenile animal toxicity study is warranted if there are developmental safety concerns that cannot be resolved from existing data. For example, if a drug will be used in neonates or young children during periods of rapid organ development, and the adult animal studies didn’t cover those life stages, a JAS might be needed to evaluate effects on growth, maturation, neurobehavioral development, etc. . Option A is incorrect because not every pediatric drug needs a JAS (many drugs for older children or those with sufficient data can waive it). Option B is unrelated (genotoxicity isn’t the usual reason for a juvenile study). Option D is also incorrect – the intent is to perform necessary juvenile studies before or during clinical development in pediatrics, not after approval. In summary, ICH S11 calls for juvenile studies only when necessary to ensure pediatric safety, using a science-driven, case-by-case evaluation .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q
  1. ICH S12 – Gene Therapy Biodistribution: What is the primary focus of the ICH S12 guideline regarding gene therapy products?
    A. Guidelines for transgenic animal creation.
    B. Clinical management of gene therapy patients.
    C. Manufacturing specifications for viral vectors.
    D. Recommendations for nonclinical biodistribution (BD) studies, i.e., assessing the in vivo distribution, persistence, and clearance of the gene therapy vector and its genetic material in both target and non-target tissues .
A

Answer: D. Explanation: ICH S12 is specifically about nonclinical Biodistribution considerations for gene therapy products. It provides a harmonized framework for designing and conducting biodistribution studies in animals for gene therapies . The goal is to understand where the gene therapy vector (and transgene) travels in the body, which tissues it localizes in, how long it persists, and how it is cleared – in order to identify any potential safety risks such as off-target exposure or germline transmission. Biodistribution is defined as the in vivo distribution, persistence, and clearance of the gene therapy product, including detection of the vector’s DNA/RNA and expressed sequence in collected tissues . The other options are outside the scope of S12: it does not deal with clinical management or manufacturing details; those topics are covered by other guidelines or regulations. Instead, S12 helps ensure that before human trials, developers have characterized the gene therapy’s biodistribution profile in relevant animal models, thus supporting safety by highlighting any unintended organ exposure (e.g., detecting if a viral vector reaches germ cells, which could raise theoretical germline alteration concerns). The correct answer is therefore the conduct of nonclinical biodistribution studies for gene therapies , which is the essence of ICH S12.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q

According to ICH S6 (R1) on preclinical safety of biotechnology-derived pharmaceuticals, which scenario justifies using a single animal species for toxicity studies instead of the usual two species?
* A. The drug is well tolerated in rodents, so testing in a second species is considered redundant.
* B. The drug is an antibody known to cross-react with its target in two species; therefore only one species is needed.
* C. The drug is pharmacologically active only in non-human primates, and no other relevant species exists.
* D. The drug has low toxicity in preliminary studies, so one species is enough.

A

Correct Answer: C. Explanation: ICH S6(R1) emphasizes the use of relevant species for biopharmaceutical toxicity testing. A relevant species is one in which the product is pharmacologically active (expresses the appropriate receptor or epitope) . Usually, two relevant species (one rodent, one non-rodent) are recommended, but if only one relevant species can be identified, it may suffice with scientific justification. For example, many human-specific biologics are only active in primates, so a single-species program in non-human primates can be acceptable. Testing in non-relevant species is discouraged because it can be misleading. Options A, B, and D are incorrect: tolerance in rodents or low observed toxicity doesn’t waive the need for a second species if one exists, and if an antibody cross-reacts in two species, both are relevant and should generally be used.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
18
Q

In nonclinical studies of a therapeutic monoclonal antibody, anti-drug antibodies (ADAs) are detected in high titers. According to ICH S6(R1) guidance on immunogenicity, what is the primary purpose of assessing these ADAs in animals?
* A. To predict the likelihood of the drug causing immunogenic reactions in humans.
* B. To assist in interpreting altered pharmacodynamics or unexpected toxicology findings in the animals.
* C. To fulfill a regulatory requirement for immunogenicity data prior to human trials.
* D. To deliberately induce immune tolerance in animals for longer-term dosing.

A

Correct Answer: B. Explanation: Per ICH S6(R1), immunogenicity assessments in animal studies are conducted to aid in the interpretation of study results and the design of subsequent studies. Many biotechnology-derived products elicit antibodies in animals; measuring these anti-drug antibodies can explain altered exposure, loss of pharmacological activity, or unexpected toxicities in treated animals. Importantly, such animal immunogenicity data are not predictive of human immunogenicity for human or humanized proteins. Thus, option A is incorrect. Option C is not entirely accurate: while regulators expect you to monitor ADA in key studies when relevant, the focus is on scientific interpretation rather than a box-ticking requirement. Option D is false – the goal is not to induce tolerance; in fact, immunogenicity can limit the usable duration of animal studies, but one does not intentionally induce it as part of study design.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q

Question 3

Which statement correctly describes the “core battery” of safety pharmacology studies as defined by ICH S7A?
* A. It excludes central nervous system tests, since those are covered in separate neurotoxicity guidelines.
* B. It requires evaluation of each vital organ system in at least two mammalian species.
* C. It can be entirely replaced by observations from repeat-dose (general toxicity) studies.
* D. It focuses on assessing a drug’s effects on vital functions, typically cardiovascular, respiratory, and CNS systems.

A

Correct Answer: D. Explanation: ICH S7A defines a safety pharmacology core battery as studies that investigate the drug’s effects on vital functions – notably the cardiovascular, respiratory, and central nervous systems. These studies (e.g., measuring effects on blood pressure, heart rate and ECG for CV, respiratory rate for respiratory, and behavioral/neurofunctional observations for CNS) are usually done before first human use to ensure no acute, life-threatening pharmacodynamic effects were missed. Option A is incorrect because CNS assessments are part of the core battery (e.g., neurobehavioral testing). Option B is incorrect – typically one appropriate species is used per core system (for example, dog or monkey for cardiovascular telemetry, rodents for CNS behavior), not two species for each system. Option C is also incorrect: while some safety pharmacology endpoints can be integrated into general tox studies when properly measured, ICH S7A generally expects dedicated studies for the core battery or a robust justification why the tox studies suffice.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q

Question 4

A new small-molecule drug is entering development. According to ICH S7B, which nonclinical strategy is recommended to assess the potential for delayed ventricular repolarization (QT interval prolongation)?
* A. Rely solely on a human thorough QT (TQT) clinical study, as nonclinical models are poor predictors of QT risk.
* B. Perform an in vitro IKr (hERG) channel assay and an in vivo QT study in animals to evaluate proarrhythmic potential.
* C. Conduct a single high-dose ECG study in rodents to look for any QT changes.
* D. Use a cultured cardiomyocyte beating assay as the one definitive test for QT prolongation risk.

A

Correct Answer: B. Explanation: ICH S7B outlines a nonclinical testing strategy for QT prolongation risk that usually involves an in vitro IKr (hERG) assay plus an in vivo assessment of QT interval (typically in telemetered animals such as dogs). The in vitro hERG test identifies direct blockade of the potassium channel that can prolong cardiac repolarization, while the in vivo study can detect actual QT prolongation or proarrhythmias in an integrated system. These assays together inform the risk before human exposure and complement clinical ICH E14 guidance. Option A is incorrect because nonclinical data are expected prior to human trials; relying only on clinical TQT is not acceptable or in line with ICH guidelines. Option C is insufficient – rodents (e.g., guinea pigs or rats) are sometimes used, but a single high-dose study alone (especially in a less sensitive model) may miss issues; plus, non-rodents are often preferred for ECG telemetry. Option D: while emerging assays (like stem-cell derived human cardiomyocytes) can supplement, ICH S7B does not consider them a standalone replacement for the standard hERG plus in vivo paradigm.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q

Under ICH S8 (Immunotoxicity Studies), which finding in standard toxicology studies would most likely be considered a “cause for concern” that triggers additional immunotoxicity testing?
* A. Unexplained atrophy of the thymus and spleen observed in treated animals.
* B. Mild, transient elevation of liver enzymes with no corresponding histopathology.
* C. Slight increases in heart weight with no microscopic cardiac lesions.
* D. Occasional skin rash in high-dose animals, with no systemic illness.

A

Correct Answer: A. Explanation: ICH S8 recommends a weight-of-evidence approach to decide if further immunotoxicity testing (like functional assays) is needed. Significant changes in immune system organs – for example, lymphoid organ weight or histology changes (thymic atrophy, spleen depletion) – with no other explanation are a classic cause for concern. Such changes suggest potential immunosuppression or immune dysfunction and should prompt specific immune function testing . Options B, C, and D are either unrelated to the immune system or minor/indirect effects. A mild liver enzyme increase (B) usually points to liver effect, not an immune-specific issue. A heart weight change (C) without lesions isn’t a recognized immunotoxicity signal. A skin rash (D) could be immune-mediated (e.g., hypersensitivity), but the question context says “no systemic effects,” and ICH S8 actually excludes hypersensitivity and autoimmunity from its scope (those are addressed separately). So, the clear immunotoxicity alarm in this list is the thymus/spleen atrophy.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q

If additional immunotoxicity studies are warranted under ICH S8, which assay is commonly recommended to evaluate a drug’s effect on immune function?
* A. A bacterial Ames test to check for immunotoxic mutagenicity.
* B. A cardiac hERG channel assay to assess immune cell ion channel function.
* C. A T-cell dependent antibody response (TDAR) assay to measure adaptive immune function.
* D. A functional observational battery (Irwin test) for neurotoxicity as a surrogate for immune status.

A

Correct Answer: C. Explanation: The T-cell dependent antibody response (TDAR) assay is a key functional test recommended by ICH S8 when immunotoxicity concerns exist. In a TDAR, animals are immunized with a known antigen (like KLH, keyhole limpet hemocyanin) while on the test drug, and the amount of antibody produced is measured. A suppressed antibody response indicates immunosuppressive effects on adaptive immunity. Options A and B are unrelated to immune function (Ames is for genetic mutations; hERG is for cardiac repolarization). Option D (the Irwin test) evaluates neurobehavioral parameters in rodents and has nothing to do with immune function. Thus, the TDAR (option C) is the appropriate choice, as it specifically evaluates the integrated T-cell/B-cell response and is widely considered a sensitive indicator of immunosuppression in vivo.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q

ICH S9 provides guidance on nonclinical evaluation for anticancer pharmaceuticals. Which type of nonclinical study is often not required for drugs intended to treat patients with advanced life-threatening cancers?
* A. Core battery safety pharmacology studies (e.g., CNS, respiratory, cardiovascular).
* B. Long-term rodent carcinogenicity studies .
* C. Standard genotoxicity tests (mutagenicity/clastogenicity assays).
* D. Toxicokinetic measurements during animal toxicology studies.

A

Correct Answer: B. Explanation: For drugs targeting patients with advanced cancers (serious, life-threatening disease with limited treatment options), ICH S9 allows a reduced nonclinical package given the urgency and risk-benefit considerations. Notably, lifetime rodent carcinogenicity studies are usually not required in this setting . The rationale is that patients’ limited life expectancy and the nature of cancer treatment (often itself cytotoxic) make long-term cancer risk less relevant, and delaying development for a 2-year rodent study is not justified. Option A is incorrect – safety pharmacology (vital function assessment) is generally still needed before FIH trials, unless a strong justification is made otherwise. Option C is generally required; even cancer drugs undergo genotoxicity testing, since DNA-reactive properties are important to know for patient safety (and labeling), unless waived for a specific reason. Option D (toxicokinetics as part of tox studies) remains important in oncology drug development to interpret exposure vs. toxicity relationships. Thus, the carcinogenicity study is the one commonly waived for advanced cancer therapies, whereas the others are usually conducted or addressed by alternate data.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q

Which scenario best fits the intended scope of the ICH S9 guideline on nonclinical evaluation for anticancer pharmaceuticals?
* A. A chemotherapy used as a preventive treatment in healthy people at high risk of cancer (chemoprevention setting).
* B. An antibiotic being repurposed to treat infections in immunocompromised cancer patients.
* C. A novel drug for refractory late-stage cancer patients who have no remaining standard treatment options .
* D. A topical therapy for widespread pre-cancerous lesions (e.g., actinic keratosis) in the general population.

A

Correct Answer: C. Explanation: ICH S9 is specifically intended for oncology drugs in patients with advanced and/or refractory cancer – typically where the disease is life-threatening and current therapies are ineffective or nonexistent . The guideline’s recommendations (e.g., allowing certain nonclinical study waivers) are predicated on that high-risk context. Option C describes a drug for refractory late-stage cancer, which fits this scope. Option A (chemoprevention in healthy high-risk individuals) is outside S9’s scope because those individuals do not have life-threatening cancer yet – a much more conservative nonclinical package (similar to typical chronic use drugs) would be expected. Option B is about an anti-infective, not an anticancer agent, so S9 doesn’t apply. Option D involves a pre-cancer condition in a broad population; again, those patients aren’t in immediate life-threatening danger, so the full complement of nonclinical studies would usually be needed. Only the scenario in C matches the advanced cancer setting that ICH S9 is meant to address.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
Under ICH S10 (Photosafety Evaluation of Pharmaceuticals), which factor alone is generally sufficient to rule out the need for further phototoxicity testing of a systemically administered drug? * A. The drug is intended for short-term (acute) use only. * B. The drug is not applied topically to the skin. * C. The drug does not distribute to ocular tissues. * D. The drug lacks significant absorption in the UV-visible range (290–700 nm) .
Correct Answer: D. Explanation: A fundamental criterion in ICH S10 is whether the compound can absorb light between 290 and 700 nm. If a drug has no significant UV-visible absorption (typically defined as no molar extinction coefficient >1000 L·mol^−1·cm^−1 in that range), it is not considered photoreactive enough to cause direct phototoxicity . In such a case, no further photosafety testing is usually warranted. Options A, B, and C are not, by themselves, conclusive. A short duration of use (A) might lower risk but doesn’t categorically rule out phototoxicity if the drug absorbs strongly and reaches the skin. Not being a topical (B) is irrelevant for systemic phototoxicity – many drugs given orally or IV can cause skin photosensitization if they circulate to skin and absorb UV. Lack of ocular distribution (C) prevents photo-ocular toxicity but not skin phototoxicity. Therefore, the absence of UV/visible absorption (D) is the clear standalone disqualifier for phototoxic risk, per ICH S10’s decision flow.
26
According to ICH S11 (Nonclinical Safety Testing in Support of Pediatric Drug Development), in which situation is a dedicated juvenile animal study (JAS) most likely warranted? * A. The drug is a new treatment intended exclusively for a pediatric disease, with no prior adult use (pediatric-first development) . * B. The drug is a generic version of a well-studied adult medication. * C. The drug will be used only in ages 12–17 and has extensive adult safety data. * D. The drug showed no toxic effects in standard adult animal studies and will be used occasionally in children.
Correct Answer: A. Explanation: ICH S11 provides a framework for determining the need for juvenile animal studies using a weight-of-evidence approach. A pediatric-first or pediatric-only indication (with no adult data) is a prime example where a JAS is usually needed . In scenario A, since adults are not exposed, one must evaluate potential safety in immature organ systems directly; the drug’s effects on growth and development need characterization in juvenile animals. Option B: a generic of an adult drug can generally rely on existing data (including any prior juvenile studies or justified waivers) – no new JAS is usually needed if the original drug didn’t require one. Option C: if the drug’s youngest patients are adolescents (12+), often the adult tox data and perhaps existing clinical data can be extrapolated; adolescents are near-mature, so a JAS might be avoidable. Option D: “no toxicity in adults” is good news, but the need for a JAS depends on factors like the age of pediatric use, developmental vulnerabilities, and pharmacology of the drug – occasional use in children (especially older children) might not need a JAS if the exposure and biology aren’t concerning. In contrast, A clearly triggers a JAS due to lack of adult data and the unknowns of developing physiology.
27
Why are nonclinical biodistribution (BD) studies important in the development of gene therapy products, according to ICH S12? * A. To assess the risk of the vector integrating into the germline DNA of patients. * B. To replace general toxicology studies with a biodistribution assessment. * C. To measure shedding of the gene therapy vector into the environment (e.g., bodily fluids). * D. To determine where the gene therapy vector and transgene go in the body, aiding interpretation of toxicology results and clinical trial design .
Correct Answer: D. Explanation: ICH S12 emphasizes that understanding a gene therapy’s in vivo biodistribution – the tissue/organ distribution, persistence, and clearance of the vector and its expressed genetic material – is critical for safety evaluation. BD data help interpret nonclinical pharmacology and toxicology findings and inform the design of first-in-human trials . For example, if a viral vector spreads to ovaries or brain, regulators need to know that to monitor potential risks. Options A and C are outside the scope of S12: germline integration (A) and environmental shedding (C) are important considerations for gene therapies but are handled by separate guidances and assessments, not the BD guideline . Option B is incorrect because BD studies complement, not supplant, the toxicology studies – you still must do toxicity studies; BD tells you if observed toxicities correlate with where the gene therapy travels or expresses. In short, the BD study under S12 is about mapping distribution to help make sense of safety and efficacy in both animals and the subsequent clinical context.
28
Q1. Which study is typically not required for short-course therapy (e.g. 2-week antibiotic) per ICH S1A? A. Carcinogenicity study B. Repeat-dose tox C. Genotoxicity test D. Safety pharmacology
Answer: A Explanation: Carcinogenicity is not needed for short-term drugs unless there’s other concern.
29
Q2. What does the ICH S1B Weight-of-Evidence (WoE) approach permit? A. Justifying omission of 2-year rat carcinogenicity studies B. Replacing all tox with in vitro assays C. Ignoring positive genotox results D. Approving drugs post hoc
Answer: A Explanation: A WoE approach can avoid long-term studies if data supports low risk.
30
Q3. What alternative can be used for a 2-year mouse carcinogenicity study? A. Transgenic mouse model (e.g. Tg.rasH2) B. Guinea pig study C. Zebrafish embryo test D. In vitro hERG assay
Answer: A
31
Q4. What is the standard rodent carcinogenicity limit dose per ICH S1C(R2)? A. 1500 mg/kg/day B. 100 mg/kg/day C. No dose limit D. 10 mg/day
Answer: A Explanation: For human doses ≤500 mg/day, 1500 mg/kg/day is the default upper dose.
32
Q5. What is the MTD (Maximum Tolerated Dose)? A. Highest dose that doesn’t reduce lifespan B. NOAEL C. 50% tumor dose D. Lowest effective dose
Answer: A
33
Genotoxicity Testing (ICH S2) Q6. What is the standard genotoxicity battery? A. Ames, in vitro chromosomal assay, in vivo micronucleus B. 2-year rat study C. Bacterial endotoxin test D. Single-dose tox
Answer: A
34
Q7. Which test detects clastogenicity? A. In vitro chromosome aberration test B. Ames test C. hERG assay D. Rabbit irritation test
Answer: A
35
Q8. Which is a standard in vivo genotoxicity test? A. Rodent micronucleus assay B. In vivo Ames C. Dermal irritation D. ECG telemetry
Answer: A
36
Toxicokinetics & Distribution Q9. What is the purpose of toxicokinetics in tox studies? A. Correlate systemic exposure with toxicity findings B. Predict efficacy C. Determine MTD D. Test for phototoxicity
Answer: A
37
Q10. When are repeated-dose tissue distribution studies warranted? A. When unexpected tissue retention occurs B. Always C. Never D. Only if IV route is used
Answer: A ICH Safety Guidelines (continued)
38
Q11. What is the standard rodent chronic toxicity duration? A. 6 months B. 3 months C. 9 months D. 12 months
Answer: A
39
Q12. What is the standard chronic duration in non-rodents? A. 9 months B. 6 months C. 12 months D. 3 months
Answer: A
40
Q13. Why were rodent tox durations reduced to 6 months? A. Longer studies rarely reveal new findings B. Rats can’t survive longer C. 2-year data are always duplicated D. Non-rodents can’t match rodent data
Answer: A
41
Reproductive & Developmental Toxicity (ICH S5) Q14. Which study evaluates fertility and early development? A. Segment I B. Segment II C. Segment III D. Genotoxicity
Answer: A
42
Q15. What is Segment II designed to assess? A. Embryo-fetal development B. Fertility C. Postnatal behavior D. Bone growth
Answer: A
43
Q16. What does Segment III study evaluate? A. Pre-/postnatal development B. Brain function in adults C. Acute reproductive toxicity D. Kidney metabolism
Answer: A
44
Biotech Products (ICH S6) Q17. What makes an animal species “relevant” for biologics testing? A. Expresses human drug target B. Similar weight to humans C. Commonly used species D. Least expensive to test
Answer: A
45
Q18. Which study is generally not required for biologics? A. Genotoxicity B. Repeat-dose tox C. TK study D. PK profiling
Answer: A
46
Q19. Why is immunogenicity assessed in animals? A. To explain variability in exposure and toxicity B. To predict human allergies C. To eliminate the need for tox testing D. It isn’t assessed
Answer: A
47
Q20. When might carcinogenicity testing be needed for biologics? A. If it has growth-promoting activity B. Always C. Only in rabbits D. Never
Answer: A
48
Safety Pharmacology (ICH S7) Q21. What defines the core battery of safety pharmacology? A. CV, CNS, Respiratory systems B. Renal, Skin, GI C. Eye, Liver, Bone D. All systems tested equally
Answer: A
49
Q22. When should core safety pharmacology be completed? A. Before Phase I trials B. After Phase III C. Post-approval D. Only if tox shows CNS effects
Answer: A
50
Q23. What risk does ICH S7B focus on? A. QT prolongation B. Liver failure C. Carcinogenicity D. Fertility
Answer: A
51
Q24. What is the standard in vitro test for QT risk? A. hERG channel assay B. Ames test C. Chromosome aberration D. ECG telemetry
Answer: A
52
Immunotoxicity (ICH S8) Q25. Which finding would trigger immunotoxicity follow-up? A. Thymus atrophy B. Elevated AST C. Body weight change D. Local irritation
Answer: A
53
Q26. Which assay is standard for immune function? A. TDAR (T-cell Dependent Antibody Response) B. ECG C. Ames test D. Micronucleus test
Answer: A
54
ICH S9 – Oncology Q41. What study is usually NOT required for advanced cancer drugs? A. 2-year rodent carcinogenicity study B. Safety pharmacology C. Genotoxicity tests D. Toxicokinetics
Answer: A
55
Q42. What is the recommended chronic tox duration for cancer drugs under S9? A. 3 months in rodent and non-rodent B. 6 months in primates C. 2 years in dogs D. None
Answer: A
56
Q43. Are genotoxicity studies required for oncology drugs? A. Yes, standard tests (Ames + in vivo) are still required B. No C. Only for topicals D. Only if carcinogenic
Answer: A
57
ICH S9 – Oncology Q44. When can reproductive toxicity studies be deferred for oncology drugs? A. When effective contraception and labeling restrictions are in place B. Never C. Only if the drug is orally administered D. After approval only
Answer: A Explanation: ICH S9 allows deferral of reproductive studies if patients are not at risk of pregnancy (e.g. postmenopausal or using contraception) and the drug is for advanced cancer.
58
Q45. What patient population is ICH S9 guidance specifically intended for? A. Patients with advanced or life-threatening cancers B. Pediatrics C. Hypertensives D. General population
Answer: A
59
ICH S10 – Photosafety Q46. When is phototoxicity testing not required per S10? A. If the compound does not absorb in the UV–vis range (290–700 nm) B. If it is not injected C. If patient is advised to avoid light D. If no skin reactions are seen in Phase I
Answer: A
60
Q47. What in vitro assay is standard for phototoxicity screening? A. 3T3 NRU phototoxicity assay B. Ames test C. Micronucleus D. hERG
Answer: A
61
Q48. Which factor would prompt phototoxicity evaluation? A. Significant UV/Vis absorption and skin exposure B. GI irritation C. Slow metabolism D. High oral bioavailability
Answer: A
62
ICH S11 – Pediatric Q49. When is a juvenile animal study required? A. When the drug is for neonates or infants or affects developing systems B. For all patients under 18 C. Only for topicals D. Never
Answer: A
63
Q50. What species is commonly used in juvenile tox studies? A. Rat – due to postnatal organ development B. Rabbit C. Monkey D. Pig
Answer: A
64
Q51. What study evaluates long-term postnatal development? A. Segment III (pre- and postnatal development) B. Segment I C. TDAR D. 3-month tox
Answer: A
65
ICH S12 – Gene Therapy Q52. What is the purpose of a biodistribution (BD) study for gene therapies? A. Determine tissue distribution and persistence of vector and transgene B. Show efficacy C. Detect antibodies D. Measure LD50
Answer: A
66
Q53. What is a key tissue for BD studies in gene therapy? A. Gonads – to assess germline transmission risk B. Skin C. Liver only D. Kidney
Answer: A
67
Q54. What method is typically used in BD studies to detect vector? A. qPCR for vector genome B. Histology C. ELISA D. Western blot
Answer: A
68
Q55. Are BD studies expected before first-in-human gene therapy trials? A. Yes – to support safety and tissue targeting B. No – only needed after Phase III C. Not unless toxicity occurs D. Optional
Answer: A
69
ICH Quality – Q3A to Q3E Q56. What is the reporting threshold for impurities in APIs (≤2 g/day)? A. 0.05% B. 0.5% C. 0.1% D. 1%
Answer: A
70
Q57. What does the identification threshold mean? A. Above this level, impurity structure must be identified B. Above this, testing is skipped C. It’s the analytical sensitivity D. The highest safe level
Answer: A
71
Q58. What does the qualification threshold indicate? A. Above this level, toxicological data must support the impurity’s safety B. It’s the allowable specification C. The same as reporting D. Always 1%
Answer: A
72
Q59. Which impurity likely doesn’t require qualification? A. A known human metabolite already studied B. A novel degradant C. A known genotoxin D. An unknown at 1%
Answer: A
73
Q60. What does Q3B cover? A. Impurities in finished drug products (e.g. degradation products) B. Residual solvents C. Packaging D. Manufacturing steps
Answer: A
74
Q61. A degradant is found at 0.2% in a 50 mg/day product. What is the action? A. Report it; assess if identification is needed based on absolute daily intake B. Ignore C. Qualify automatically D. Eliminate
Answer: A
75
ICH Q3C – Residual Solvents Q71. Which solvent is a Class 1 residual solvent (to avoid)? A. Benzene B. Ethanol C. Acetone D. Methanol
Answer: A
76
Q72. What characterizes Class 2 solvents? A. Toxic solvents with PDEs – limited, but not banned B. Must be avoided C. Safe at any level D. Only used in topicals
Answer: A
77
Q73. What’s an example of a Class 3 solvent? A. Ethanol B. Benzene C. Chloroform D. Dichloromethane
Answer: A
78
Q74. What is the limit for Class 3 solvents? A. 50 mg/day or less B. 1.5 µg/day C. 0.05% D. 10 ppm
Answer: A
79
ICH Q3D – Elemental Impurities Q75. Which element is Class 1 (most toxic)? A. Lead (Pb) B. Iron C. Zinc D. Magnesium
Answer: A
80
80
Q76. Which is a Class 3 element (lower risk)? A. Zinc B. Cadmium C. Arsenic D. Mercury
Answer: A
81
Q77. What is the key principle of Q3D’s approach? A. Risk-based evaluation of elemental impurities B. Testing every batch for 24 elements C. Avoiding solvents D. Applying PDEs only in injectables
Answer: A
82
Q78. Which route typically has the lowest PDEs? A. Inhalation B. Oral C. Dermal D. Rectal
Answer: A
83
ICH Q3E – Extractables & Leachables Q79. What does Q3E address? A. Leachables from packaging and container systems B. Residual solvents C. Impurities from degradation only D. Metals
Answer: A
84
ICH M3 – Timing of Nonclinical Studies Q80. Which study is not required before Phase I? A. Carcinogenicity studies B. Repeat-dose tox C. Genotox D. Safety pharmacology
Answer: A
85
Q81. When should embryo-fetal development studies be done? A. Before large trials including women of childbearing potential without contraception B. After approval C. Before Phase I always D. Never
Answer: A
86
Q82. How long should tox studies be relative to clinical trials? A. At least as long as human exposure B. Always 6 months C. Only 2 weeks D. Irrelevant
Answer: A
87
Q83. For serious disease (e.g., cancer), how does M3 adjust requirements? A. Allows shorter tox durations and deferred studies B. Requires more species C. Eliminates all tox D. Requires genotox only
Answer: A
88
Q84. What does M3 say about single-dose tox studies? A. Can be omitted if adequate info from repeat-dose studies exists B. Always required C. Not discussed D. Must use LD50
Answer: A
89
Q85. What animals are usually required for tox studies? A. Rodent and non-rodent species B. Monkey and dog C. Human and rodent D. Rodent only
Answer: A
90
ICH M4 – CTD Format Q86. Which CTD module is region-specific? A. Module 1 B. Module 2 C. Module 3 D. Module 4
Answer: A
91
Q87. What is in Module 2? A. Overviews and summaries of Quality, Nonclinical, Clinical B. Full study reports C. Batch records D. Labeling
Answer: A
92
Q88. Where do nonclinical study reports go? A. Module 4 B. Module 2 C. Module 5 D. Module 1
Answer: A
93
Q89. What is in Module 3? A. Quality data: manufacturing, controls, stability B. Clinical trial data C. Genotoxicity D. Summary documents
Answer: A
94
Q90. What is in Module 5? A. Clinical study reports B. Nonclinical summaries C. Regional forms D. CMC data
Answer: A
95
ICH M7 – Mutagenic Impurities Q91. What impurities are covered by M7? A. DNA-reactive (mutagenic) impurities B. Metals C. All degradants D. Solvents
Answer: A
96
Q92. What is the default TTC (threshold of toxicological concern)? A. 1.5 µg/day for lifetime exposure B. 1.5 mg/day C. 150 µg/day D. 0.15 µg/day
Answer: A
97
Q93. What is the “cohort of concern”? A. Extremely potent mutagens like nitrosamines B. Class 3 solvents C. Salts D. Residual water
Answer: A
98
Q94. Can TTC be adjusted for short-term treatment? A. Yes, higher intakes are allowed for short durations B. No C. Only if injectable D. Never
Answer: A
99
Q95. How are multiple mutagenic impurities handled? A. Each must meet TTC individually B. Add all together C. Only test worst D. Ignore if <0.1%
Answer: A
100
Q96. What if an impurity has a structural alert but is negative in Ames? A. Can be treated as non-mutagenic B. Must be controlled to 1.5 µg/day C. Ignored D. Requires carcinogenicity study
Answer: A
101
Q97. If an impurity is mutagenic, what’s the mitigation? A. Limit to ≤1.5 µg/day or justify with risk assessment B. No action C. Wait for Phase III D. Test it in humans
Answer: A
102
Q98. What is a Class 1 impurity in M7? A. Known potent mutagenic carcinogens (e.g., N-nitrosamines) B. Ethanol C. Copper D. Water
Answer: A
103
Q99. What’s the recommended (Q)SAR approach in M7? A. Two complementary systems: expert rule-based and statistical B. One software C. None D. Always do in vivo first
Answer: A
104
Q100. When can TTC be exceeded? A. For short-term use or serious diseases, with justification B. Never C. For over-the-counter drugs D. For topicals
Answer: A
105
Question 1: ICH Q3A(R2) – Impurities in New Drug Substances Q1. In ICH Q3A(R2), what is the identification threshold for impurities in a new drug substance with a maximum daily dose of 500 mg? (This is the level above which an impurity should be identified and possibly characterized/qualified.) A. 0.15% or 1.0 mg per day, whichever is lower B. 0.05% (regardless of dose) C. 0.10% or 1.0 mg per day, whichever is lower D. 0.5% or 2.0 mg per day, whichever is lower
Answer: C. For a new drug substance dosed at ≤2 g/day, ICH Q3A(R2) sets the identification threshold at 0.10% of the drug substance or 1.0 mg total daily intake, whichever is lower . In this case (500 mg/day dose), 0.10% = 0.5 mg, and the alternative 1.0 mg/day limit is higher, so the threshold would be 0.5 mg (0.10%). Any impurity above that (~0.10%) in the drug substance must be identified. (The qualification threshold is higher at 0.15% or 1.0 mg , and the reporting threshold lower at 0.05% for ≤2 g doses.) Choices A and D reflect the qualification and higher-dose thresholds, respectively, not the identification threshold for 500 mg. Choice B (0.05% regardless of dose) is the general reporting threshold for >2 g/day cases and not applicable here.
106
Question 2: ICH Q3B(R2) – Impurities in New Drug Products Q2. For a new drug product with a maximum daily dose of 5 mg, which is the ICH Q3B(R2) identification threshold above which a degradation impurity should be identified? (This refers to impurities in the finished product where very low daily doses apply.) A. 1.0% or 5 µg per day, whichever is lower B. 0.2% or 2 mg per day, whichever is lower C. 0.10% of the drug substance (regardless of dose) D. 0.5% or 20 µg per day, whichever is lower
Answer: D. For drug products, ICH Q3B(R2) uses a sliding scale based on daily dose. A 5 mg/day dose falls in the “1 mg–10 mg” range, for which the identification threshold is 0.5% of the drug substance or 20 µg total daily intake, whichever is less . Twenty micrograms is the lower of those two values (since 0.5% of 5 mg is 25 µg), so any impurity above 20 µg/day (0.4% in this case) must be identified. This is a much tighter threshold than for high-dose drugs, reflecting the greater concern when the drug itself is low-dose. Choice A (1.0% or 5 µg) applies only to extremely low dose products <1 mg/day . Choice B (0.2% or 2 mg) is for higher dose ranges (>10 mg up to 2 g) . Choice C (0.10%) is the threshold for very high dose drugs >2 g/day (100 mg impurity) , not for a 5 mg dose. Thus, 0.5% or 20 µg is correct for a 5 mg/day product, demonstrating how Q3B’s thresholds include absolute µg limits for low-dose medications.
107
Question 3: ICH Q3C(R9) – Residual Solvents Classification Q3. Which one of the following solvents is classified as a Class 1 residual solvent that should be avoided according to ICH Q3C? A. Chloroform B. Benzene C. Methanol D. Acetone
Answer: B. Benzene is a Class 1 solvent under ICH Q3C, meaning it is a known human carcinogen and environmental hazard that should not be used (or should be eliminated to <2 ppm if unavoidable) . Class 1 solvents (which also include carbon tetrachloride, 1,2-dichloroethane, 1,1-dichloroethene, and 1,1,1-trichloroethane) have no safe exposure level and are to be avoided in drug manufacturing. In contrast, chloroform (A) is Class 2 (a toxic solvent that should be limited to a PDE of 0.6 mg/day, ~60 ppm) . Methanol (C) is Class 2 as well (with a higher permissible daily exposure of 30 mg/day, ~3000 ppm) . Acetone (D) is Class 3, a solvent with low toxic potential that is less tightly regulated (acceptable at levels up to 0.5% or 50 mg/day without justification) . Knowing the classes: Class 1 = avoid (e.g. benzene), Class 2 = limit (e.g. chloroform, methanol with specific ppm limits), Class 3 = low toxic concern (e.g. acetone, ethanol, requiring only GMP-controlled levels).
108
Question 4: ICH Q3D(R2) – Elemental Impurities Categories Q4. Which element is NOT included among ICH Q3D’s Class 1 (most toxic) elemental impurities that require stringent control? A. Lead (Pb) B. Cadmium (Cd) C. Nickel (Ni) D. Mercury (Hg)
Answer: C. Nickel is not a Class 1 elemental impurity – it is classified as Class 2A under ICH Q3D . Class 1 elements (the most toxic with ubiquitous potential presence) are Lead, Cadmium, Mercury, and Arsenic, which have the tightest Permitted Daily Exposures (e.g. Pb and Cd ~5 µg/day, As ~15 µg/day, inorganic Hg ~30 µg/day for oral route) . These require stringent risk assessment and control in virtually all drug products. Nickel, on the other hand, is Class 2A – toxic, but typically a catalyst or raw material impurity with a lower likelihood of presence unless introduced; its PDE for oral intake is much higher (200 µg/day) . Choices A, B, and D are all Class 1 metals (Pb, Cd, Hg) with very low safety thresholds. This distinction is important because Class 1 elements must always be considered in a risk assessment, whereas Class 2A elements are considered if likely present (e.g. from manufacturing equipment or catalysts) .
109
Question 5: ICH Q3E (Draft) – Extractables & Leachables Q5. Which statement correctly defines “extractables” in the context of the forthcoming ICH Q3E guideline on extractables and leachables (E&L)? A. Extractables are chemical compounds that migrate into the drug product under normal storage conditions. B. Extractables are exclusively metal ions that leach from manufacturing equipment into the product. C. Extractables are volatile impurities from the drug substance that become entrapped in packaging materials. D. Extractables are compounds that can be pulled out of container/closure or processing materials under aggressive solvent conditions, serving as a worst-case indicator of potential leachables.
Answer: D. Extractables are defined as compounds that can be extracted from packaging or manufacturing materials under forceful (accelerated) conditions (e.g. strong solvents, high temperature) . They represent a “worst-case scenario” of what could migrate into the product. In other words, extractables studies deliberately use harsh conditions to identify substances that a container closure system or single-use manufacturing component might release. These identified extractables help predict leachables, which are the subset of those compounds that actually do leach into the drug product under normal conditions. Choice A describes leachables (what migrates into the product during shelf-life or use), not extractables. Choice B is too narrow – extractables are any chemical (organic or inorganic); metals are just one category and are often assessed separately (e.g. via Q3D for elemental leachables). Choice C is incorrect – volatile impurities from the drug substance (e.g. residual solvents) are covered under Q3C, not considered extractables from packaging. The key point: extractables come from container/closure or process materials, and are discovered via aggressive extraction studies, whereas leachables are actually found in the drug product during its shelf life .
110
Question 6: ICH M3(R2) – Nonclinical Safety Study Durations for Trials Q6. ICH M3(R2) provides recommendations on the duration of animal toxicity studies needed to support clinical trial phases. What durations of repeat-dose toxicity studies in rodents and non-rodents, respectively, are generally recommended to support a clinical trial intended to last longer than 6 months (e.g. a Phase 3 trial or chronic use)? A. 3-month studies in rodents and 3-month studies in non-rodents B. 6-month studies in rodents and 9-month studies in non-rodents C. 9-month studies in rodents and 6-month studies in non-rodents D. 12-month studies in rodents and 12-month studies in non-rodents
Answer: B. For clinical dosing >6 months, ICH M3(R2) recommends chronic toxicity studies of 6 months in rodents and 9 months in non-rodents (such as dogs or monkeys) . This guideline ensures that the longest animal studies cover the duration of human exposure (and then some). Specifically, shorter clinical trials (≤6 months) can be supported by toxicity studies of equal length in both species, but for trials beyond 6 months, a 6-month rodent and 9-month non-rodent study are expected . (Regulators consider 6/9 months the maximum needed for marketing authorization for chronic indications in ICH regions, with no added value seen in 12-month rodent studies in most cases.) Choice A (3/3 months) is sufficient only for up to 3-month clinical studies. Choice C swaps the durations (non-rodents generally require the longer study due to their typically slower physiology and to better detect late-occurring toxicities). Choice D (year-long studies) is not the standard per M3(R2); 12-month studies are not routinely required before approval, except in special cases or if triggered by specific concerns. In summary, 6 months rodent/9 months non-rodent is the conventional package to support chronic use in humans , aligning with the principle that toxicity studies should equal or exceed the planned clinical exposure duration.
111
Question 7: ICH M4 & M4Q – CTD Quality Module vs. Summary Q7. In the Common Technical Document (CTD) format for drug applications, where should the detailed data on drug substance and drug product quality (CMC – Chemistry, Manufacturing, Controls) be placed, and where should the summary of quality information be placed? A. Detailed quality data in Module 2, and the summary in Module 3. B. Detailed quality data in Module 5, and the summary in Module 2. C. Detailed quality data in Module 4, and the summary in Module 1. D. Detailed quality data in Module 3, and the summary (Quality Overall Summary) in Module 2.
Answer: D. In the CTD structure, Module 3 is the Quality module containing the full body of data for drug substance and drug product (all the CMC details, such as specifications, analytical methods, batch data, stability, etc.), while Module 2 includes the Quality Overall Summary (QOS) . The QOS is a concise summarization of the Module 3 data, following a defined outline, and it highlights critical quality attributes and justifications. Module 2 also contains the nonclinical and clinical overviews/summaries, essentially summarizing Modules 3, 4, and 5 . Choices A, B, C are incorrect placements – for example, Module 4 is for nonclinical study reports, Module 5 is for clinical study reports, and Module 1 is region-specific administrative information. Knowing the CTD: * Module 2: Overviews and Summaries (QOS, Nonclinical Overview/Summaries, Clinical Overview/Summaries). * Module 3: Quality (CMC full data). * Module 4: Nonclinical (tox, pharmacology study reports). * Module 5: Clinical (clinical trial reports).
112
Question 8: ICH M4E(R2) – Clinical Overview Benefit-Risk Assessment Q8. ICH M4E(R2) introduced a significant revision to the CTD Efficacy module. What key enhancement did M4E(R2) make to the format of the Clinical Overview (Module 2.5)? A. It added a new requirement to integrate clinical pharmacology data into Module 2.7. B. It provided a structured format for the benefit–risk assessment in Section 2.5.6 of the Clinical Overview. C. It introduced a tabular listing of adverse events to Module 5 clinical study reports. D. It moved the Quality Overall Summary from Module 2 to Module
Answer: B. ICH M4E(R2) specifically revised Section 2.5.6 of the Clinical Overview to standardize the Benefits and Risks Conclusions section . This update gives a much more structured framework for sponsors to present the benefit–risk assessment of the product. It includes subsections like Therapeutic Context (e.g. disease background and current therapies) and separate discussions of benefits and risks, leading to an integrated benefit–risk conclusion . The goal was to improve the clarity and consistency of benefit–risk evaluations across applications. Choice A is irrelevant to M4E(R2) – it deals with Module 2.5 (Clinical Overview) rather than Module 2.7 (Clinical Summary). Choice C (tabular AEs) and D (moving QOS) are distractors; no such changes were made. In summary, the core enhancement of M4E(R2) is improved guidance on writing the Clinical Overview’s benefit-risk section , reflecting regulators’ increasing focus on clear benefit–risk reasoning in drug approval.
113
Question 9: ICH M7(R2) – Threshold of Toxicological Concern (TTC) Q9. According to ICH M7, what is the acceptable daily intake of a DNA-reactive (mutagenic) impurity that is considered to pose a negligible risk for lifetime exposure in humans? (Assume no compound-specific cancer data; use the default Threshold of Toxicological Concern.) A. 1.5 µg per person per day B. 5 µg per person per day C. 20 µg per person per day D. 120 µg per person per day
Answer: A. 1.5 µg/day is the default TTC (Threshold of Toxicological Concern) for mutagenic impurities, corresponding to a theoretical excess cancer risk of approximately 1 in 100,000 over a lifetime . ICH M7(R2) specifies that if an impurity is known or expected to be DNA-reactive (and no compound-specific carcinogenicity data exist), limiting it to no more than 1.5 µg per day is considered to ensure negligible carcinogenic risk for >10 years to lifetime exposure . Choices B, C, D reflect TTC limits for shorter duration exposures: M7 provides higher allowable intakes when exposure is transient or limited (e.g. up to 1 month: 120 µg/day; 1–12 months: 20 µg/day; 1–10 years: 10 µg/day) . After 10 years, the limit tightens to the 1.5 µg/day lifetime TTC. Thus, 1.5 µg/day is the correct lifetime limit. It’s worth noting that certain potent “cohort of concern” mutagens (like aflatoxin-like, N-nitrosamines, and azoxy compounds) are excluded from the 1.5 µg TTC approach – they require even lower limits due to their high carcinogenic potency . But for general mutagenic impurities, the 1.5 µg/day TTC is the default cut-off in ICH M7.
114
Question 10: ICH M7(R2) – Scope and Exemptions Q10. Which scenario is exempted from the impurity assessment and control requirements of ICH M7 (mutagenic impurities guideline)? A. An impurity in a medicine intended to treat advanced cancer, where the drug itself is genotoxic. B. A mutagenic impurity in an antihypertensive drug intended for lifelong chronic use. C. A potential DNA-reactive degradant in a tablet formulation with a 2-year shelf life. D. An impurity with a positive Ames test alert present in a Phase 3 trial material for a non-oncology indication.
Answer: A. ICH M7 explicitly does not apply to compounds used in advanced cancer treatment when genotoxicity is part of the therapeutic mode of action (as covered by ICH S9) . In other words, if a drug is intended for late-stage cancer patients (short life expectancy) and is itself genotoxic (or the treatment involves genotoxic modality), the stringent limits of M7 for impurities do not need to be applied. Such patients and therapies tolerate higher risks, and other guidance (ICH S9) addresses their development. All other choices (B, C, D) are within M7’s scope: * B: Lifelong use for a common disease – any mutagenic impurity must meet TTC or compound-specific limits. * C: Degradants that are DNA-reactive fall under M7 and need assessment/controlled to acceptable intakes. * D: Impurities in late-phase clinical development (except advanced cancer drugs) are covered by M7; by Phase 3, companies are expected to have a control strategy for mutagenic impurities in place, even if some limits are adjusted by less-than-lifetime (LTL) considerations. In summary, M7 primarily exempts advanced cancer therapies and a few other special cases (e.g. impurities in certain biological products, like peptides, where mutagenic impurity risk is not relevant) . For most other drugs, including chronic-use medications, ICH M7’s risk assessment (structural alerts, Ames testing, TTC limits, etc.) for genotoxic impurities applies.