FDA Flashcards

(62 cards)

1
Q

FDA’s Least Burdensome provisions encourage regulators and industry to use only the minimum amount of information necessary to address regulatory questions, avoiding unnecessary studies or data requirements . Which of the following best exemplifies this principle?
* A. Designing a development program to eliminate extraneous tests while still demonstrating safety and efficacy.
* B. Delaying all nonclinical studies until after product approval to reduce upfront work.
* C. Seeking FDA approval with no supporting data if studies are costly or time-consuming.
* D. Prioritizing speed over safety by skipping critical trials to get to market faster.

A

Answer: A.
Explanation: “Least burdensome” means using the most efficient and least onerous approach that still meets regulatory standards. Answer A reflects providing the minimum necessary data — no more and no less — to prove safety/effectiveness . This might involve leveraging prior evidence, validated alternatives, or narrower scopes for testing to avoid unnecessary burdens . In contrast, B and D go too far, jeopardizing thoroughness or safety, and C is not allowed – some valid supporting data must be provided. The principle is about removing undue burdens while still “maintaining the statutory requirements” for approval , not about skipping essential tests.

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

FDA’s Redbook 2000 uses a tiered approach for food additive testing based on chemical structure category (toxic potential) and estimated human dietary exposure. Suppose a new additive is classified as Category B (intermediate toxic potential) and the estimated human intake is 30 ppb (parts-per-billion) in the diet. Into which Concern Level would this fall, and what is the implied testing tier?
* A. Low Concern (CL I) – Minimal toxicity testing needed (very low exposure).
* B. Intermediate Concern (CL II) – Moderate tier testing recommended.
* C. High Concern (CL III) – Extensive toxicity testing required.
* D. No concern – Exposure is negligible, testing can be waived entirely.

A

Answer: B.
Explanation: Category B structures are assigned to Concern Level II once human exposure exceeds 25 ppb but is below 500 ppb . In this case, 30 ppb falls into CL II (intermediate concern). Redbook’s framework combines structure Category B with exposure (~30 ppb) to set an intermediate concern level . CL II triggers a moderate testing battery – more than just basic tests, but not as extensive as CL III. (By comparison, CL I covers Category B exposures ≤25 ppb, and CL III covers >500 ppb .) Answer D is incorrect because 30 ppb is not negligible; some testing is expected at CL II. Answer C overshoots (CL III corresponds to much higher exposure), and A underestimates the concern.

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

Under Redbook 2000, the recommended toxicity tests increase with higher Concern Levels. For a food additive assigned to the lowest concern level (CL I), which one of the following studies is NOT typically required?
* A. Bacterial gene mutation assay (Ames test for genotoxicity).
* B. Short-term (28-day) oral toxicity study in rodents.
* C. Two-year rodent carcinogenicity bioassay.
* D. In vivo micronucleus assay (rodent bone marrow cytogenetics test).

A

Answer: C.
Explanation: A long-term carcinogenicity study is not required for CL I additives. At the lowest exposure tier, Redbook recommends primarily genetic toxicology tests (like the Ames test and in vivo micronucleus) and possibly a short-term repeat-dose study . For CL I (low concern), no chronic or carcinogenicity studies are expected. Options A, B, and D are part of the basic battery for even low concern additives . In fact, Redbook’s summary table shows that genetic toxicity tests are recommended for all concern levels (I–III) . A 28-day rodent study (short-term toxicity) is also indicated even at CL I (often with limited scope) . However, two-year cancer bioassays (and other long-term studies like one-year dog studies or multi-generation reproduction studies) come into play only at higher concern levels (CL II/III) , making C the correct answer.

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

Triggers for Reproductive & Developmental Toxicology Studies

In drug development, certain conditions trigger the need for dedicated reproductive and developmental toxicity studies (per ICH S5 and related guidances). All of the following scenarios would typically prompt such studies EXCEPT:
* A. A chronic medication intended for women of childbearing potential (WOCBP).
* B. Evidence of testicular or ovarian toxicity in repeated-dose animal studies.
* C. A new drug planned for use in pregnant women (e.g. to treat gestational conditions).
* D. A short-term (<2 week) therapy intended only for men with a terminal illness.

A

Answer: D.
Explanation: Reproductive and developmental toxicology studies are generally not needed (or can be deferred) for a short-duration therapy in a population that excludes women of childbearing potential, especially in a terminal illness setting. In such a case (Answer D), the drug wouldn’t be used by individuals at risk of pregnancy or effects on fertility, so full reprotox testing may be waived or delayed. In contrast, Answer A is a classic trigger: before exposing large numbers of WOCBP to a chronic drug, regulators require fertility and embryo-fetal development studies. Answer B: if general tox studies show potential reproductive organ toxicity, it triggers focused studies to assess effects on fertility or development. Answer C: any drug intended for use during pregnancy (e.g. to treat pregnant patients) absolutely requires embryo-fetal development studies (and possibly peri/postnatal studies) to ensure safety for the fetus. Thus, A, B, C all represent scenarios where reproductive/developmental tox data are expected, whereas D does not.

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

FDA’s guidance “Nonclinical Studies for the Safety Evaluation of Pharmaceutical Excipients” (2005) allows leveraging prior human experience to support a new excipient’s safety and potentially waive certain studies. Which factor is NOT considered by FDA as a valid basis for bridging toxicology data from prior use of an excipient?
* A. The excipient was previously used in an approved drug or is GRAS for food, under similar exposure conditions.
* B. The route of administration and duration of exposure in the prior use are comparable to the proposed new use.
* C. The exposure level in humans (dose) in prior use is of the same order as in the new product (or higher).
* D. The excipient is listed in the USP/National Formulary (USP–NF) monograph as a pharmaceutical ingredient.

A

Answer: D.
Explanation: Inclusion in the USP–NF monograph is not evidence that an excipient is safe for a new use – FDA explicitly notes that a USP listing or mention in a non-FDA document “is not an indication that the substance has been reviewed by the FDA and found safe” . In contrast, factors A, B, and C are specifically considered when using prior human use to support safety. FDA’s excipient guidance says that if an excipient has documented human exposure (e.g. used in foods or approved drugs) with a similar route, exposure level, population, and duration, it may not require the full battery of new toxicology studies . Prior GRAS or drug use (Answer A) is relevant, as are matching route and exposure (B, C). These real-world data can justify a “least burdensome” approach for the excipient. However, a USP monograph alone, which is more about quality standards, does not address safety data – so D is not a sufficient basis to waive tox studies.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
6
Q

The FDA’s GRAS (Generally Recognized as Safe) concept allows certain food substance uses to bypass premarket approval if safety is widely recognized by experts. Which of the following statements about GRAS is FALSE?
* A. Color additives (substances used solely to impart color) are eligible for GRAS classification.
* B. General recognition of safety can be based on published scientific evidence or a history of common use in food prior to 1958.
* C. GRAS status applies to a specific use of a substance – a substance might be GRAS for one purpose but not necessarily for another.
* D. A GRAS determination typically requires that safety data are widely available and accepted by qualified experts (not kept as trade secrets).

A

Answer: A.
Explanation: It is not true that color additives can be GRAS – in fact, food dyes and color additives are explicitly excluded from the GRAS provision. By law, the GRAS exemption applies to “food additives” but there is no parallel GRAS clause in the definition of a color additive . (So any substance added solely to impart color in food must go through the color additive approval process, not GRAS.) The other statements are accurate: B – GRAS can be supported either by a history of safe use in food (e.g. common food ingredients used pre-1958) or by published studies such that experts unanimously agree on safety. C – GRAS is use-specific; a substance can be GRAS for one intended use but not for a different use or higher level (each intended condition requires evaluation ). D – A core tenet of GRAS is that safety information is “generally available” and there’s a consensus among qualified experts on safety . Proprietary, unpublished data that only one company has seen would not meet the “generally recognized” standard. Thus, A is the false statement.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
7
Q

When selecting a safe starting dose for initial human trials, FDA guidance (2005) recommends converting the animal NOAEL to a Human Equivalent Dose (HED) using appropriate scaling. According to this guidance, what is the preferred method for extrapolating an animal dose to an equivalent human dose?
* A. Use a straight mg/kg body weight conversion (1:1 from animal mg/kg to human mg/kg).
* B. Scale doses according to peak plasma concentrations (Cmax) in animals vs. humans.
* C. Normalize doses to body surface area (mg/m²) to account for interspecies size differences.
* D. Use whichever species had the highest mg/kg NOAEL as the basis for HED.

A

Answer: C.
Explanation: The FDA recommends using body surface area (BSA) scaling for most systemically administered drugs when converting animal doses to human equivalent doses . In practice, this means converting the animal NOAEL (in mg/kg) to an HED by normalizing to mg/m² (since larger animals have relatively lower metabolic rates per body weight). This approach better accounts for interspecies differences in physiology than a simple mg/kg ratio. Answer A (1:1 on body weight) can overestimate safe human doses for small animals because it doesn’t account for metabolic scaling – e.g. a mouse can tolerate many mg/kg relative to a human. B (Cmax scaling) is not the general default; exposure (AUC) or BSA is used unless pharmacokinetic data suggest otherwise. D is incorrect; one should not automatically choose the highest mg/kg NOAEL species – the most appropriate species is usually the one with the lowest HED, not the highest mg/kg (to be most protective) . Thus, BSA normalization is the standard method for HED.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
8
Q

During risk assessment, multiple animal studies may yield different NOAELs. FDA’s starting dose guidance says the “most sensitive species” (the one resulting in the lowest HED) usually drives the first-in-human dose . For example, imagine the NOAELs for a new drug are: 50 mg/kg/day in rats and 20 mg/kg/day in dogs. Using standard km factors (rat km≈6, dog km≈20, human km≈37), which species is most likely to be considered the most sensitive, and why?
* A. The rat, because its HED is lower (~8 mg/kg) than the dog’s HED (~10.8 mg/kg) after body surface area scaling.
* B. The rat, because 50 mg/kg is a much higher dose than 20 mg/kg (indicating more toxicity).
* C. The dog, because 20 mg/kg in a larger animal yields a lower HED than the rat’s (20 mg/kg always sounds smaller).
* D. The dog, because canine toxicology is generally more predictive for human risk.

A

Answer: A.
Explanation: Converting the NOAELs to HED: For rats, 50 mg/kg * (6/37) ≈ 8.1 mg/kg HED; for dogs, 20 mg/kg * (20/37) ≈ 10.8 mg/kg HED. The rat’s HED is lower, meaning the rat is effectively the more sensitive species on a dose-normalized basis. According to FDA, one should generally use the species that gives the lowest HED (most conservative) to set the starting dose . Thus, the rat’s NOAEL leads to the more cautious human dose. Answer B is wrong reasoning: while 50 mg/kg is numerically higher, it’s in a small animal – after scaling, it’s actually the more limiting dose. C is incorrect because the actual calculation shows the opposite. D is a generalization – dog data can be very important, but here the numbers show the rat is more sensitive. In summary, Option A is correct: the rat’s NOAEL yields the lower HED (~8 mg/kg), so the rat is the most sensitive species for dose-setting.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
9
Q

FDA and ICH guidances (e.g., ICH S1C(R)) outline how to choose the highest dose in rodent carcinogenicity studies. Which of the following is NOT a recommended criterion for selecting the top dose in a 2-year rodent carcinogenicity bioassay?
* A. A dose that induces minimal toxicity (e.g. slight <10% body weight decrease) — the Maximum Tolerated Dose (MTD).
* B. A dose that is 50 times the human AUC exposure if lower doses show no toxicity.
* C. A dose at which further absorption or exposure plateaus (“saturation of absorption”).
* D. A predefined limit dose (e.g. 1000–1500 mg/kg/day in rodents) if the compound is nontoxic even at very high doses.

A

Answer: B.
Explanation: 50× human exposure is too high – current guidelines do not recommend using a fifty-fold multiple of human AUC. The ICH S1C(R) guidance identifies several acceptable criteria for the high dose, including: the MTD (minimal toxicity such as ≤10% body weight suppression), a dose that yields roughly 25× the human systemic exposure (AUC) if toxicity is not reached, a dose at which absorption or pharmacodynamic effect is maxed out, or a limit dose (usually 1000 mg/kg/day for rodents, or 1500 mg/kg in some cases) . Using ~25× exposure is meant to ensure a large margin without going to an unreasonably high multiple. Option B’s 50× exceeds what is considered necessary (and could needlessly increase animal toxicity). In contrast, A, C, and D are all aligned with ICH recommendations: (A) using the MTD is a standard approach; (C) if higher doses don’t increase exposure due to saturation, that dose can serve as the top; (D) if a compound is super nontoxic, a limit dose is used to avoid absurdly high dosing . Thus, the 50× AUC criterion is not recommended, making B the correct answer.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
10
Q

For statistical robustness in detecting tumor incidence, chronic rodent carcinogenicity studies typically use relatively large group sizes. What is the standard number of animals per sex per group in a 2-year rat or mouse carcinogenicity study (as historically recommended by FDA/ICH)?
* A. ~10 animals/sex/group
* B. ~20 animals/sex/group
* C. ~50 animals/sex/group
* D. ~100 animals/sex/group

A

Answer: C.
Explanation: Fifty rodents of each sex per group is the standard group size for the classic two-year carcinogenicity bioassay. Using ~50 males and 50 females per dose group provides enough power to detect a modest increase in tumor incidence. Regulatory guidelines (ICH S1) and FDA’s protocol expectations have long settled on 50/sex/group for the main study groups, plus potentially additional satellite animals for interim evaluations. Options A and B (10 or 20 per sex) would lack power – tumors are usually rare events, so you need a larger N to see statistically significant increases. Option D (100 per sex) is higher than necessary and is not the usual practice (it would be very costly and is not required under guidelines). Thus, answer C (50/sex) is correct. (For perspective, FDA’s “Redbook” also used 50 animals/sex for chronic studies in food additives.) This number balances statistical detection with practical feasibility.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
11
Q

FDA’s guidance Safety Testing of Drug Metabolites (MIST) calls for additional evaluation of disproportionate metabolites. Which scenario best defines a human disproportionate metabolite that may warrant separate safety testing?
* A. A metabolite present only in humans, but at <5% of total drug-related exposure in human plasma.
* B. A metabolite that comprises >10% of drug-related exposure in humans and similarly >10% in at least one animal species.
* C. A metabolite formed in animal studies but not detected in humans at clinically relevant doses.
* D. A metabolite present at >10% of total exposure in humans and at a higher plasma level in humans than in any of the animal test species.

A

Answer: D.
Explanation: A “disproportionate drug metabolite” is one that is observed in human circulation at a significant level (generally exceeding 10% of total drug-related exposure) without adequate exposure in the test species used in the nonclinical program . In other words, it’s either unique to humans or present at much higher concentrations in humans than in animals. Answer D captures both criteria: >10% of total exposure in humans and higher than any animal – this is exactly the kind of metabolite the guidance flags.

Options A, B, C are incorrect:
* A: A metabolite under 10% of total exposure in humans is below the threshold of concern (no additional testing typically needed ).
* B: If a metabolite is >10% in humans and also >10% in an animal, that means the animal was exposed sufficiently. It’s not disproportionate because the animal data likely covered it. The concern is when humans have a major metabolite that animals did not see at similar levels.
* C: A metabolite seen in animals but not in humans is actually the inverse situation (an “animal-only” metabolite) – that might be interesting for animal tox interpretation, but it doesn’t pose unknown human risk.

Therefore, D is the correct description of a disproportionate human metabolite that may require its own safety assessment .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
12
Q

According to FDA’s metabolite safety testing guidance, if a human metabolite is identified as disproportionate (unique or much higher in humans) and exceeds the exposure threshold of concern, what additional nonclinical testing is typically recommended?
* A. No additional studies – the parent drug’s toxicity tests suffice for all metabolites.
* B. Conduct a dedicated toxicity study (e.g. 1-month or 3-month repeat-dose) on the isolated metabolite, plus supporting studies as needed.
* C. Perform only in vitro assays (like an Ames test) for the metabolite, but no animal studies.
* D. Initiate a small clinical trial administering the pure metabolite to healthy volunteers.
According to FDA’s metabolite safety testing guidance, if a human metabolite is identified as disproportionate (unique or much higher in humans) and exceeds the exposure threshold of concern, what additional nonclinical testing is typically recommended?
* A. No additional studies – the parent drug’s toxicity tests suffice for all metabolites.
* B. Conduct a dedicated toxicity study (e.g. 1-month or 3-month repeat-dose) on the isolated metabolite, plus supporting studies as needed.
* C. Perform only in vitro assays (like an Ames test) for the metabolite, but no animal studies.
* D. Initiate a small clinical trial administering the pure metabolite to healthy volunteers.

A

Answer: B.
Explanation: If a metabolite is disproportionate in humans (high levels not tested in animals), FDA recommends a separate safety assessment for that metabolite. Typically this means synthesizing the metabolite (if feasible) and conducting targeted studies, such as repeat-dose toxicity in one or two species and genotoxicity assays, to characterize its safety profile . For example, a 1-month rodent study might be done to see if the metabolite causes any effects on its own. Genotoxicity (Ames test and/or chromosomal assays) is also often done on the metabolite if it structurally differs significantly from the parent. Option A is incorrect because the parent’s tests might not have revealed effects of a metabolite that wasn’t present in animals. Option C – solely doing in vitro tests – would be insufficient if the metabolite exposure is large; in vivo data are usually needed to assure safety (though genotoxicity testing is one component). Option D (testing the metabolite in humans) is not ethically or logistically appropriate as a first step – nonclinical safety should be established before any such human exposure. In summary, the guidance calls for additional animal toxicity testing for the metabolite to ensure that the human is not exposed to an untested, potentially toxic entity .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
13
Q

The Least Burdensome Provisions (Feb 2019)
1. What best describes FDA’s definition of the “least burdensome” approach in medical device regulation?
a. Reducing safety or effectiveness requirements for faster approvals
b. Using the minimum information necessary to address regulatory questions in an efficient manner
c. Requiring sponsors to prove their device is absolutely risk-free
d. Applying fewer regulations only to novel breakthrough devices

A

Explanation: Option b is correct. FDA defines “least burdensome” to mean the minimum amount of information necessary to adequately address a regulatory question at the right time, using the most efficient method . It does not lower the statutory safety or effectiveness standards; instead, it streamlines how those standards are met. Options a and c are incorrect because least burdensome is not about compromising safety or demanding risk-free proof, but about avoiding unnecessary data collection. Option d is false because the least burdensome principles apply to all device submissions (not just breakthrough devices) throughout the product lifecycle .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
14
Q
  1. Which of the following actions by FDA staff best exemplifies the “least burdensome” principles?
    a. Asking for extensive additional clinical trials even when existing data are sufficient
    b. Leveraging existing data or foreign regulatory decisions to avoid duplicate testing
    c. Insisting on rigid, one-size-fits-all requirements for all devices
    d. Delaying any postmarket data collection until after approval is granted
A

Explanation: Option b is correct. A key least burdensome principle is that FDA will use the most efficient means to resolve issues, which includes leveraging existing evidence and even data from other regulators to avoid redundant trials . Option a is opposite to least burdensome (it increases burden unnecessarily). Option c contradicts the tailored approach of least burdensome—regulatory requirements should fit the technology and context (not one-size-fits-all) . Option d is incorrect because least burdensome encourages using postmarket data when appropriate (i.e. collecting certain data after approval to lighten premarket burden) .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
15
Q
  1. Congress first introduced the “least burdensome” provisions for medical devices in which law?
    a. Food and Drug Administration Modernization Act (FDAMA) of 1997
    b. Medical Device Amendments of 1976
    c. 21st Century Cures Act of 2016
    d. Safe Medical Devices Act of 1990
A

Explanation: Option a is correct. The concept of least burdensome was initially added by FDAMA in 1997 . This law directed FDA to streamline device premarket review by eliminating unnecessary burdens. Later laws (e.g. 2012 and 2016 amendments) built upon the 1997 provisions but were not the first introduction. Options b and d are earlier device laws that did not specifically include least burdensome provisions. Option c (21st Century Cures 2016) expanded upon least burdensome principles but FDAMA 1997 was the original source.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
16
Q
  1. True or False: The least burdensome principles allow FDA to lower the required level of device safety or effectiveness in order to expedite product availability.
    a. True – “Least burdensome” means FDA can accept greater uncertainty about safety/effectiveness.
    b. False – Statutory standards for safety/effectiveness remain unchanged; only the means of obtaining evidence are streamlined .
A

Explanation: Option b is correct. Least burdensome does not alter the fundamental requirements for demonstrating device safety and effectiveness . It simply means FDA and industry should focus on the most efficient way to get the evidence needed, avoiding any unnecessary effort. The FDA still requires a reasonable assurance of safety and effectiveness; it just commits to not ask for extraneous data . Therefore, it is false that FDA lowers the standard; the standards stay the same, but how one meets them can be optimized.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
17
Q
  1. Which of the following is NOT one of the guiding principles of FDA’s least burdensome approach?
    a. FDA requests only the minimum necessary information to address the regulatory question .
    b. Industry should submit well-organized, relevant documentation (and omit irrelevant data) .
    c. FDA and industry utilize interactive communication and consider postmarket data when appropriate .
    d. FDA insists on a single pre-specified pathway for all submissions to maintain consistency.
A

Explanation: Option d is correct (the false statement). A hallmark of least burdensome is flexibility – tailored approaches rather than a single rigid pathway . In fact, FDA explicitly intends to adapt to individual circumstances and use interactive communications. Options a, b, and c are all true principles from the guidance: FDA will ask only for information truly needed ; sponsors should keep submissions concise and focused ; and both parties should aim for efficient processes, including use of postmarket surveillance to lighten premarket requirements .

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

Nonclinical Safety for Pharmaceutical Excipients (May 2005)
6. In FDA’s excipient guidance, a “new excipient” is defined as an inactive ingredient that:
a. Has never been used in any FDA-regulated product before
b. Is intended to exert therapeutic effects at its use level
c. Lacks sufficient safety data for the proposed exposure, duration, or route of use
d. Is a substance like water or sugar with well-known safety profiles

A

Explanation: Option c is correct. The guidance defines new excipient as an inactive ingredient not fully qualified by existing safety data for the intended level, duration, or route of exposure . It may have been used before, but not at the proposed dose/formulation or route. Option a is too strict – even previously used substances can be “new excipients” if used in a new way without sufficient data. Option b is wrong because excipients are not intended to have therapeutic effects . Option d describes well-known excipients (not “new” ones) which usually wouldn’t require extensive new safety evaluation.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
19
Q
  1. FDA’s excipient guidance divides recommendations by intended duration of use. What maximum clinical use duration defines a “short-term use” excipient?
    a. Up to 14 days of consecutive use
    b. 1 month
    c. 3 months
    d. 6 months
A

Explanation: Option a is correct. Short-term use excipients are those used in products labeled for 14 or fewer consecutive days of use . This typically covers excipients in drugs for acute conditions or short courses of therapy. Intermediate use generally refers to longer than 2 weeks up to ~3 months, and long-term would be more than 3 months, but the guidance explicitly sets ≤14 days as the short-term threshold.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
20
Q
  1. Which nonclinical studies does FDA recommend even for excipients intended only for short-term (≤14 days) clinical use?
    a. Acute toxicity in rodent and non-rodent, genotoxicity battery, and even reproductive toxicity studies
    b. Only acute toxicity testing, nothing further
    c. A 2-year carcinogenicity study due to potential risk
    d. No studies at all if use is short and exposure is low
A

Explanation: Option a is correct. For short-term use excipients, FDA still recommends a comprehensive safety evaluation including: acute toxicity in at least two species, standard genetic toxicology tests, one-month repeat-dose studies, and an assessment of reproductive toxicity (fertility and embryo-fetal development) . Even short exposure could involve women of child-bearing potential, so teratogenicity and fertility studies are advised. Options b and d are too minimal – the guidance does not exempt short-term excipients from all testing. Option c (a full 2-year carcinogenicity) is generally not required for short use unless other signals warrant it; carcinogenicity studies are typically reserved for long-term use excipients.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
21
Q
  1. For an excipient intended in a chronic-use medication (labeled for >3 months of use), which additional nonclinical studies are recommended beyond those for short- and intermediate-use excipients?
    a. Longer-term toxicity studies (e.g. 6-month rodent and 9–12 month non-rodent studies) and an evaluation of carcinogenicity potential
    b. No additional studies; short-term data are sufficient
    c. Only genotoxicity testing, nothing else
    d. Just a 1-month study since humans will be monitored clinically
A

Explanation: Option a is correct. For excipients in drugs used longer than 3 months, FDA recommends performing chronic toxicity studies (typically a 6-month study in rodents and a chronic 9–12 month study in a non-rodent species) . Additionally, the carcinogenic potential should be evaluated — either via two-year rodent bioassays or justified alternative approaches . Options b, c, and d are incorrect because for long-term exposure, short-term studies alone are not sufficient. Chronic exposure raises concerns like carcinogenicity that must be addressed, so a comprehensive long-term tox program is needed.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
22
Q
  1. FDA’s excipient guidance outlines scenarios for evaluating carcinogenicity of a new excipient. Which approach is NOT an acceptable option mentioned for excipients intended for long-term use?
    a. Conducting two-year carcinogenicity bioassays in two species (e.g. rat and mouse)
    b. A single two-year study in one rodent species plus a transgenic or alternative assay in a second species
    c. Providing a scientific justification to forgo dedicated carcinogenicity studies (e.g. negative genotox, low exposure, no pre-neoplastic lesions)
    d. Ignoring carcinogenicity assessment entirely for a long-term use excipient, with no justification
A

Explanation: Option d is correct (not acceptable). For excipients in long-term use, FDA expects sponsors to address carcinogenicity either by studies or robust justification. The guidance allows multiple acceptable approaches: conducting two 2-year rodent studies (option a), doing one long-term rodent study plus an alternative assay (option b), or justifying why carcinogenicity testing isn’t needed based on weight-of-evidence (option c) . Simply not evaluating carcinogenicity at all (option d) is not acceptable for a chronic-use excipient – if one chooses not to do bioassays, a science-based rationale must be provided .

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
23
Q
  1. Excipients intended for injectable (parenteral) use often warrant special safety tests. Which test does FDA recommend for an IV excipient to assess local effects?
    a. In vitro hemolysis assay at the intended intravenous concentration
    b. Ames test for mutagenicity
    c. Skin sensitization (guinea pig maximization) test
    d. Respiratory irritation study in rodents
A

Explanation: Option a is correct. For excipients used in IV formulations, FDA suggests performing an in vitro hemolysis test at the clinical concentration to evaluate potential blood cell lysis . Similarly, for intramuscular or subcutaneous routes, measuring creatine kinase (muscle enzyme) can help detect local muscle damage . Options b (Ames) is a general genotox test done for all excipients (not specifically for injectables’ local safety). Option c (skin sensitization) is more relevant for topical excipients (though a sensitization assay is recommended generally as part of safety). Option d (respiratory irritation) would apply to inhaled excipients. The key parenteral-specific test is hemolysis for IV excipients.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
24
Q
  1. FDA recommends evaluating photosafety (phototoxicity) for excipients under what circumstance?
    a. For any excipient that significantly absorbs UV/visible light, similar to an active drug
    b. Only for topical excipients, not orally administered ones
    c. Never – excipients are inert and don’t require phototoxicity evaluation
    d. Only if the excipient is derived from a botanical source
A

Explanation: Option a is correct. The guidance advises considering photosafety testing for excipients if there is a concern (e.g. the excipient or formulation has light-absorbing properties), using the same principles as for active ingredients . This is determined on a case-by-case basis. Option b is incorrect because even oral excipients might distribute to skin; if they absorb light, phototoxicity could be relevant. Option c is false – “inert” excipients can sometimes cause photoreactions, so FDA recommends evaluating the need for photosafety testing . Option d is irrelevant; source alone (botanical or not) isn’t the criterion – it’s the chemical’s photochemical properties that matter.

How well did you know this?
1
Not at all
2
3
4
5
Perfectly
25
GRAS (Generally Recognized as Safe) for Food Substances – FAQ Guidance 13. What does it mean for a substance to be classified as GRAS (Generally Recognized as Safe) under U.S. FDA regulations? a. The substance is exempt from the definition of “food additive” and not subject to premarket approval because experts generally agree it’s safe under its intended use . b. FDA has formally approved the substance through a food additive petition. c. The substance is natural, so it is automatically safe. d. It only applies to animal feed ingredients, not human food ingredients.
Explanation: Option a is correct. A substance that is GRAS is by definition not a food additive and does not require FDA premarket approval, because it is generally recognized, by qualified experts, to be safe for its intended use . This general recognition is what differentiates GRAS substances from other food additives that need petitions. Option b is incorrect because GRAS is an alternative to formal approval (GRAS substances don’t go through the additive petition process). Option c (“natural = safe”) is a myth – natural substances can still be hazardous; GRAS requires scientific consensus on safety, not just being natural. Option d is wrong because GRAS applies to human food and animal food substances (both are covered in the guidance).
26
14. Which of the following is NOT a criterion for a use of a substance to be eligible for GRAS status? a. Safety is supported by either scientific procedures or common use in food prior to 1958 . b. A panel of qualified experts would agree the intended use is safe based on available data. c. The data establishing safety are generally available (e.g. published) and widely accepted . d. The substance is patented, and its data are kept confidential within one company.
Explanation: Option d is correct (not a GRAS criterion). For a substance’s use to be GRAS, the evidence of safety must be publicly available and generally accepted by the scientific community . A patented substance with only secret, proprietary safety data cannot meet the “generally recognized” standard. Options a, b, and c are true: Under the law, a substance can be GRAS either through scientific data or through common food use before 1958 ; it requires consensus among qualified experts; and the safety information must be widely available and vetted (often published in the literature).
27
15. How do the safety evidence requirements for a GRAS determination compare to those for a food additive approval? a. GRAS requires the same quantity and quality of scientific evidence as needed for a food additive, but with general recognition by experts . b. GRAS requires less evidence since it’s informal. c. Food additives need toxicology data, but GRAS substances do not require toxicological studies. d. GRAS decisions can be based on anecdotal history alone, unlike food additives.
Explanation: Option a is correct. FDA’s regulation (21 CFR 170.30) states that general recognition of safety through scientific procedures requires the same quality/quantity of evidence as a formal food additive approval – the difference is that the evidence is already published or known such that experts generally accept it . The GRAS route is not a shortcut on evidentiary standard, it’s about consensus and publicity of data. Options b, c, and d are incorrect: GRAS is not meant to be a weaker standard; toxicological and other relevant studies are still required as evidence (option c is false). Pure anecdotal history isn’t sufficient unless it constitutes a substantial history of safe food use prior to 1958 with documentation (option d is misleading – even common use must be evidenced and scientifically interpreted).
28
16. True or False: A company must submit a GRAS notification to FDA and wait for FDA approval before marketing a GRAS substance. a. True – FDA must review and affirm each GRAS determination before use. b. False – The GRAS notification process is voluntary, and FDA’s response (“no questions”) is not a formal approval .
Explanation: Option b is correct. Submitting a GRAS notice to FDA is voluntary – a company can self-conclude that a use is GRAS and market the substance without prior FDA approval . FDA encourages notification, but it’s not mandatory. When FDA reviews a GRAS notice, it issues a letter (e.g. “no questions”) but this is not an approval or license; it simply means FDA has no objections based on the notifier’s evidence. Therefore, a company does not have to wait for FDA’s explicit approval if they are confident in their GRAS conclusion (though they bear responsibility for that safety determination). Option a is false.
29
17. If you have determined a substance is GRAS for its intended use and you notify FDA, do you need to wait for FDA’s response before marketing the substance? a. No – You may market the substance immediately, provided your GRAS conclusion is correct, even if FDA has not yet responded . b. Yes – You must wait for FDA to issue a GRAS approval letter. c. Yes – Marketing before FDA feedback is illegal for GRAS substances. d. No – But you must at least receive confirmation of receipt from FDA.
Explanation: Option a is correct. If a manufacturer has made a valid GRAS determination, they do not need to wait for FDA’s review outcome to begin marketing . FDA’s GRAS notification program is voluntary and its response is not an approval. In fact, the FAQ guidance explicitly says that if you are correct in your GRAS conclusion, you can proceed with marketing while FDA’s review is pending . Option b and c are wrong because FDA doesn’t “approve” GRAS substances the way it does food additives. (Of course, if FDA later disagrees with the GRAS conclusion, the product would have regulatory issues, so companies usually prefer to get the “no questions” letter for assurance.)
30
18. Under GRAS criteria, “common use in food prior to 1958” can support GRAS status. Which statement is true about this pathway? a. It requires a substantial history of consumption by a significant number of people before 1958, with documented safety in that use . b. If usage was only outside the U.S. before 1958, it cannot be considered. c. Any substance used in any manner before 1958 qualifies as GRAS automatically. d. It no longer applies because all GRAS must now be based on scientific studies.
Explanation: Option a is correct. GRAS via experience (common use in food) means that before 1958 the substance was consumed as food by a large population and deemed safe (this needs to be documented with data on the history and extent of use) . Option b is not entirely true – common use outside the U.S. can be considered, but the proponent should provide evidence and possibly notify FDA if only foreign use data exist (U.S. experts must be able to review it and reach consensus). Option c is incorrect because not every pre-1958 use is “substantial” or safe; it must meet significance and safety recognition. Option d is false – the law still allows the common-use pathway, though in practice most modern GRAS determinations rely on scientific data.
31
19. When determining that a substance is GRAS for use in animal food (feed for food-producing animals), what additional consideration is important? a. The safety of humans consuming products (meat, milk, eggs) from those animals must also be evaluated . b. Only the target animal’s safety matters, since GRAS is separate for human vs animal. c. GRAS cannot be used for animal feed ingredients. d. Palatability to the animal is the sole concern.
Explanation: Option a is correct. For a GRAS conclusion in animal food, one must consider both the target animal’s safety and any human food safety aspects if that animal enters the human food supply . For example, a GRAS determination for a cattle feed additive must address residues in milk or meat and ensure they are safe for people. Option b is wrong because human exposure via food products is explicitly part of the GRAS evaluation for animal feed uses . Option c is incorrect – GRAS applies to animal food as well, and FDA’s Center for Veterinary Medicine handles those notices. Option d (palatability) is not the key safety consideration (it’s more of an efficacy/usage concern, not directly related to GRAS safety which focuses on health risk).
32
20. Which statement about FDA’s GRAS notification program is true? a. It is a voluntary program where anyone can notify FDA of a GRAS conclusion . b. FDA’s “no questions” letter for a GRAS notice is equivalent to formal approval. c. If FDA disagrees with a GRAS notice, it will still list the substance as GRAS with caveats. d. The GRAS notification process is only for human food; there is no mechanism for animal food.
Explanation: Option a is correct. The GRAS notification procedure is explicitly voluntary – the guidance encourages companies to use it, but it’s not mandatory . Any person can notify FDA of their GRAS conclusion for either human or animal food. Option b is wrong: FDA’s response is not an approval, it’s just an acknowledgement (or sometimes an objection). Option c is incorrect because if FDA disagrees (issues e.g. a “GRAS notice does not provide a basis for GRAS” letter), the substance would not be added to the GRAS listings. Option d is false – there is a parallel GRAS notification process for animal food (21 CFR 570 subpart E) handled by CVM .
33
Redbook 2000: Toxicological Principles for Food Ingredient Safety 21. FDA’s “Redbook 2000” uses a Concern Level (CL) approach for food additives. What two key factors determine a food additive’s Concern Level? a. The substance’s toxicological potential (structure) and the estimated human exposure to the substance . b. Whether it’s natural or synthetic and its cost of manufacture c. The flavor and color of the additive d. The number of prior FDA approvals for the substance
Explanation: Option a is correct. The Concern Level (I, II, or III) is assigned based on (1) the additive’s chemical structure (which is categorized as A, B, or C for low to high toxic potential) and (2) the estimated human dietary exposure to the additive . These two factors combined determine if an additive is low, intermediate, or high concern. Options b, c, and d are irrelevant to the CL classification. Natural vs synthetic or cost has no bearing on inherent toxicity. Flavor/color are unrelated to safety concern per se. Prior approvals don’t factor into new use’s concern level (each use is considered on its own exposure and structure merits).
34
22. Suppose a new food additive is structurally related to known carcinogens (Category C structure) and the estimated human intake is 20 ppb. What FDA Concern Level would this correspond to? a. Low Concern (CL I) b. Intermediate Concern (CL II) c. High Concern (CL III) d. Not enough information
Explanation: Option b is correct. Category C (high toxic potential) substances have the most stringent exposure cutoffs for concern levels. According to Redbook guidelines, a Category C structure with human exposure of 20 ppb falls into Concern Level II (for Category C: CL I is 0–12 ppb, CL II is >12 up to 250 ppb, CL III is >250 ppb) . Therefore 20 ppb, being above 12 but well below 250, puts it in intermediate concern. We have enough information (structure category and exposure) to assign the CL. (For comparison: a Category A (low hazard) could go up to 50 ppb and still be CL I, but Category C hits CL II at anything above 12 ppb.)
35
23. Which testing is generally sufficient to establish safety for a food additive assigned to Concern Level I (low concern)? a. Genetic toxicity assays and short-term toxicity testing (acute studies) b. A full 2-year carcinogenicity study in two species c. Chronic (1-year) dog studies d. Reproductive and developmental studies in multiple species
Explanation: Option a is correct. For a low concern additive (CL I), FDA’s Redbook recommends a minimal toxicity testing package: typically acute toxicity studies and a standard genotoxicity battery . Often this might include an Ames test and one in vivo genotoxicity assay, and acute or short-term repeat-dose studies to confirm no unexpected toxicity. In contrast, options b, c, and d are much more extensive than required for CL I. Carcinogenicity studies (b) and year-long studies (c) are generally reserved for CL III (high concern) situations . Reproductive/developmental studies (d) are introduced at CL II and CL III, not typically needed for CL I if exposure is truly minimal .
36
24. As Concern Level increases from I to III, the recommended toxicity testing expands. Which study becomes specifically recommended at Concern Level III that is not typically needed at CL I or II? a. Long-term carcinogenicity studies (e.g. 2-year rodent bioassays) b. Bacterial mutagenesis (Ames) test c. 90-day subchronic rodent study d. Acute oral toxicity in rodents
Explanation: Option a is correct. High concern (CL III) additives require the full battery of tests, including carcinogenicity studies in two rodent species (usually 2-year bioassays) and chronic toxicity studies (e.g. 1-year in non-rodent) . These are not generally called for at CL I or II. Options b, c, and d are all tests that are done even at lower concern levels: for instance, genotoxicity (Ames test) is done at all levels (including CL I) ; a 90-day subchronic is typically part of CL II; acute tox is done even for CL I. The big additions at CL III are chronic/carcinogenicity studies and often multi-generation reproductive studies.
37
25. Which of the following correctly matches a Concern Level with tests typically recommended by FDA at that level? a. CL II (Intermediate): Genotoxicity, 90-day subchronic in rodent & non-rodent, and reproductive/developmental studies . b. CL I (Low): 1-year chronic studies in two species and a carcinogenicity bioassay. c. CL II: Only acute toxicity tests and Ames assay, nothing more. d. CL III (High): Just a 90-day study since exposure is higher.
Explanation: Option a is correct. For a Concern Level II additive, the recommended tests include everything in CL I (acute and genotoxicity) plus 90-day repeat-dose studies in rodent and non-rodent, and appropriate reproductive and developmental toxicity studies . Option b is wrong because those extensive chronic/carcinogenicity studies correspond to CL III, not CL I. Option c is too minimal for CL II; CL II requires more than CL I (which itself already included acute and genotox). Option d is incorrect – CL III demands the most testing (chronic and carcinogenicity, not just a 90-day study) .
38
26. What is the safety standard that a food additive or GRAS substance must meet for FDA to consider it acceptable? a. A “reasonable certainty of no harm” under the conditions of intended use . b. Zero risk to any consumer c. An adequate margin of safety of 10,000-fold in all studies d. No observed effects in any animal test at any dose
Explanation: Option a is correct. By regulation, food additives (and GRAS uses) must satisfy the standard of reasonable certainty of no harm under their intended conditions of use . This acknowledges that absolute zero risk is not attainable, but safety must be reasonably assured. Option b (“zero risk”) is not the standard – FDA does not require proving zero risk, but rather a high degree of confidence in safety. Option c is an exaggerated safety factor not universally required (typical safety factors might be 100x, etc., but not a fixed 10,000). Option d is incorrect because some effects (non-harmful or minor) might be acceptable; the key is no significant adverse effects at expected exposure, which is encapsulated in “no harm”.
39
27. In FDA’s concern level scheme, what does a Category C structural designation imply about a food additive’s structure? a. It is structurally related to compounds known to be carcinogenic or mutagenic (high toxic potential) . b. It contains only common nutrients and thus has low toxicity concern. c. It’s a completely new chemical class with no toxicity information at all. d. It was approved in the past (Category C stands for “Certified”).
Explanation: Option a is correct. Category C structures are those that raise a high level of toxicological concern, often because they resemble known carcinogens or mutagens . For example, a molecule with a structural alert (like a nitrosamine or aflatoxin-like structure) would likely be Cat C. Option b describes Category A (low concern, e.g. common nutrients) not Category C. Option c (completely unknown) would still be assessed but Category C specifically denotes known “bad actor” structural features, not just lack of info. Option d is unrelated – “Category C” in this context has nothing to do with prior approvals; it’s about chemical structure risk.
40
Maximum Safe Starting Dose in Initial Clinical Trials (July 2005) 28. According to FDA’s guidance on first-in-human (FIH) starting dose, which animal study result is typically used as the basis for calculating the Maximum Recommended Starting Dose (MRSD) in humans? a. The no-observed-adverse-effect level (NOAEL) from the most sensitive relevant animal species b. The highest dose tested in animals (regardless of toxicity) c. The LD50 (lethal dose 50%) in rodents d. The dose that caused mild pharmacological effects in animals
Explanation: Option a is correct. FDA recommends identifying the NOAEL in each animal species tested and then using the lowest HED (human equivalent dose) among those, which comes from the most sensitive species, as the basis for MRSD . The NOAEL is defined as the highest dose that did not produce adverse effects in the animal studies . Options b and c are incorrect because we do not use the highest dose or lethal dose — we use the highest non-toxic dose. Option d (a pharmacologically active dose) might be considered in special cases (if it’s much lower than the NOAEL and could cause effect in humans), but generally the NOAEL is the key for safety.
41
29. How is a Human Equivalent Dose (HED) usually derived from animal data for the purpose of setting a starting dose in humans? a. By scaling the animal NOAEL dose to humans based on body surface area (mg/m²) using FDA’s conversion factors . b. By taking the animal NOAEL in mg/kg and using the same mg/kg value in humans c. By dividing the animal NOAEL by 10, directly (a simplistic approach) d. By matching blood concentration levels exactly between species
Explanation: Option a is correct. The guidance instructs to convert the NOAEL from each animal species to a Human Equivalent Dose using body surface area (BSA) normalization (mg/m² scaling) . FDA provides standard conversion factors (based on species body weight and BSA relationships) to do this. Option b (using mg/kg equally) does not account for interspecies differences in metabolism and size – smaller animals tolerate higher mg/kg, so mg/m² is preferred. Option c (NOAEL/10) is actually the safety factor step, not the interspecies scaling step. Option d (matching concentrations) isn’t the default method for initial dose; allometric BSA scaling is the default for simplicity and conservatism .
42
30. FDA’s default safety factor for calculating a safe human starting dose from the animal NOAEL is: a. 10-fold, to account for uncertainties between animals and humans b. 1 (no safety factor; use HED directly) c. 100-fold by default d. It depends entirely on the compound, with no general recommendation
Explanation: Option a is correct. The guidance suggests using a safety factor of 10 by default when dividing the HED (from the most sensitive species) to arrive at the MRSD . This 10× factor provides a margin for human variability and uncertainties. Options b and c are incorrect extremes – 1 would often be too risky, and 100 is larger than generally necessary (though in rare cases a higher factor might be used, 100 is not the default for most drugs). Option d is partially true that it can vary, but FDA does give a general recommendation of 10 as a starting point, adjusting up or down if needed by compound-specific considerations .
43
31. Which scenario might prompt using a larger than 10× safety factor when setting the first human dose? a. The animal dose-response curve is steep, with serious toxicity appearing just above the NOAEL . b. The drug’s pharmacologic class is well-understood and human data exist c. The NOAEL was far below the lethal dose with a wide therapeutic index d. Animals showed only mild reversible effects at high doses
Explanation: Option a is correct. A steep dose-response or other concerning findings in animals (e.g., toxicity that escalates rapidly beyond the NOAEL, or irreversibility of toxic effects) would justify increasing the safety factor above 10 . Other reasons include non-linear pharmacokinetics or unresolved toxicity signals – all situations where additional caution is warranted . Options b, c, and d would actually lean towards possibly smaller safety factors: if the drug class is well-known (b) or the safety margin in animals is very large (c, d), one might consider a factor <10 in some cases . So those are opposite of the question’s premise.
44
32. In what case might FDA accept a safety factor less than 10 for the MRSD calculation? a. If there is extensive human clinical experience or data on closely related compounds, reducing uncertainty . b. If the animal study was done in non-GLP conditions c. If the drug is intended for a life-threatening condition d. Never; FDA always requires at least 10× safety margin for FIH dosing
Explanation: Option a is correct. FDA acknowledges that a lower safety factor (e.g., <10) might be justified when uncertainty is reduced – for instance, if the drug belongs to a well-characterized class with human data or if other information makes the animal-to-human translation more certain . In such cases the sponsor must justify scientifically why a smaller margin is sufficient . Options b and c are not appropriate reasons to lower the factor; a non-GLP study would reduce confidence (arguing for more caution, not less), and a life-threatening indication might encourage starting dose conservatism or higher initial risk acceptance but not arbitrarily shrinking the safety factor without data. Option d is incorrect – 10 is default, but not unchangeable; FDA allows flexibility with rationale .
45
33. FDA’s guidance defines NOAEL for starting dose selection. Which of the following best describes NOAEL? a. The highest tested dose at which no adverse effects were observed in animals . b. A dose that has no effect of any kind (adverse or not) c. The dose that causes the least burdensome effect d. The dose that causes death in 50% of animals
Explanation: Option a is correct. The No-Observed-Adverse-Effect Level (NOAEL) is defined as the highest dose that does not produce a statistically or biologically significant increase in adverse effects compared to control . It specifically focuses on adverse (toxicologically relevant) effects, not any effect. Option b is describing a NOEL (no observed effect level), which is slightly different – the NOAEL may allow for some pharmacological or benign effects as long as they are not harmful . Option c is irrelevant wording. Option d is the definition of LD₅₀, not NOAEL. The NOAEL is a key benchmark for safety and starting dose calculations.
46
34. If a certain drug shows a pharmacologically active effect in animals at doses much lower than the NOAEL, how might this influence the first-in-human starting dose? a. The starting dose might be set based on the lower pharmacologically active dose (PAD) instead of the NOAEL, if that effect could pose risk . b. It would not influence the starting dose at all – only toxicity matters. c. FDA would ignore the pharmacologic effect and use NOAEL/HED as usual. d. It means the drug is unsafe to proceed to human trials.
Explanation: Option a is correct. In cases where a drug’s pharmacologic action occurs at doses below the NOAEL, and that action might itself be risky in humans (e.g., excessive PD effect), the guidance says the starting dose calculation can consider a pharmacologically active dose (PAD) as the limiting factor . Essentially, if the PAD is significantly lower than the toxic dose, it can dictate a lower safe starting dose. Option b and c are too simplistic – while toxicity drives the MRSD in most cases, FDA does note that an exception can be made to use a PAD (with justification) . Option d is wrong; it doesn’t mean the drug is unsafe, just that you might start at a dose that avoids a strong pharmacologic effect.
47
35. What is the “most sensitive species” in the context of selecting a starting dose for human trials? a. The animal species whose NOAEL, when converted to HED, is the lowest, thus giving the most conservative safe dose . b. The species that had the most severe toxic response c. The species phylogenetically closest to humans d. The species with the highest mg/kg tolerated dose
Explanation: Option a is correct. After converting NOAELs from each tested species to human equivalent doses (HEDs), the most sensitive species is the one with the lowest HED NOAEL – that species’ NOAEL implies humans might tolerate the least, so it governs the starting dose . This ensures a conservative approach. It’s not necessarily the species with the most dramatic toxicity overall, but the one that yields the smallest safety margin to humans. Options b and c are not the formal definition; sometimes a primate might be closest to human (c), but if a rodent has a lower HED, the rodent is “most sensitive” for dose-setting. Option d is opposite – highest tolerated dose would be least sensitive.
48
Safety Testing of Drug Metabolites (MIST Guidance) 36. Under FDA’s “Metabolites in Safety Testing” (MIST) guidance, when is additional toxicity testing of a drug’s metabolite generally warranted? a. When the metabolite’s exposure in humans exceeds 10% of total drug-related exposure and is significantly higher than exposure in the animal studies . b. Whenever any human metabolite is detected at any level c. Only if the metabolite is found to be pharmacologically active d. Only if the metabolite is present in human urine
Explanation: Option a is correct. The guidance (and subsequent ICH) established that a human metabolite should be considered for dedicated safety evaluation if it makes up more than ~10% of total drug-related exposure in humans and that level is not adequately covered in the toxicity tests in animals . This “>10% and disproportionate” criterion is the key trigger. Option b is too broad; minor metabolites don’t need separate testing. Option c (pharmacologically active) — activity might raise interest, but the guidance criterion is mainly exposure-based (though activity could be a factor even below 10% in some cases). Option d is irrelevant; excretion route doesn’t by itself mandate testing – it’s about systemic exposure levels.
49
37. Which scenario is MOST likely to require conducting a separate safety study for a metabolite? a. A human-only metabolite comprises 15% of drug exposure, while in rats it was 2% (i.e., much higher in humans) . b. A metabolite is 5% of total exposure in humans and 5% in dogs. c. A metabolite is 20% in humans and 25% in mice. d. A human metabolite is 12% of exposure, and rodents had 15% exposure to the same metabolite.
Explanation: Scenario a is correct. A metabolite present at 15% of total exposure in humans and disproportionately lower in animals (only 2%) meets the “greater than 10% and under-represented in animals” criterion . This would likely prompt additional safety studies on that metabolite. In scenario b, 5% is below the 10% threshold (no action generally needed). In c, even though 20% in humans is above threshold, the fact that mice had 25% (comparable or higher) means the metabolite was already evaluated in mice at equal/higher exposure, so separate testing might not be necessary (the animal studies likely covered its safety). In d, 12% in humans is >10% but rodents had similar exposure (15%), so again the animal tests probably covered it. The unique risk is when humans have a major metabolite that animals did not see at similar levels.
50
38. What is a typical approach to evaluate the safety of a metabolite that warrants testing? a. Synthesizing the metabolite and performing dedicated toxicity studies (e.g. 90-day animal studies) on that metabolite b. Assuming it’s covered by parent compound testing and doing nothing c. Testing it only in in vitro assays, never in vivo d. Doing a human clinical trial with the isolated metabolite before Phase I of the parent drug
Explanation: Option a is correct. If a metabolite is deemed to need separate evaluation (per MIST criteria), the sponsor would usually isolate or synthesize that metabolite and then test it in appropriate animal toxicity studies (often a subchronic duration) similar to how one would test a new drug . Option b is exactly what MIST tries to avoid — one must ensure it’s actually covered; if not, you can’t ignore it. Option c (only in vitro) may be part of the strategy (e.g. genotoxicity on the metabolite), but in vivo data are generally expected for a major unmetabolized compound. Option d is not typical or ethical – one wouldn’t trial an isolated metabolite in humans prior to ensuring its safety in animals. The usual course is animal testing of the metabolite.
51
39. According to the FDA metabolite safety guidance, which metabolite would be considered “major” for further evaluation? a. One that accounts for ≥10% of the parent drug’s AUC in humans . b. One present at 3% of total drug exposure in humans c. Any metabolite detected in any animal species d. A metabolite only found in vitro at trace levels
Explanation: Option a is correct. The guidance identifies a “major metabolite” generally as one exceeding ~10% of total drug-related exposure (AUC) in humans . Those are the ones that, if not seen in animals at comparable levels, may need additional study. Options b, c, and d describe minor metabolites. A 3% metabolite (b) is usually too low to worry about. Option c is irrelevant because just being present in animals doesn’t automatically flag it; it’s about human exposure. Option d (trace in vitro) is also not major. The threshold is the ~10% of systemic exposure in humans – above that is a rule of thumb for “major.”
52
40. True or False: If animal studies have exposed test animals to a metabolite at levels similar to or greater than human exposure levels, separate safety testing of that metabolite is generally not required. a. True – Adequate “exposure coverage” in the toxicology species means the metabolite’s safety has been indirectly evaluated . b. False – Each human metabolite must be tested in standalone studies regardless of animal data.
Explanation: Option a is correct. The MIST approach says that if a metabolite is formed in animals at sufficient levels (comparable to humans), then the standard animal studies on the parent drug have effectively already assessed that metabolite’s toxicity, and no separate studies are usually needed . It’s when a metabolite is “disproportionate” (much higher in humans than in any animal) that additional tests are called for. Therefore, it’s true that adequate exposure in animal tests obviates extra testing. Option b is false: FDA does not require every metabolite to be individually tested; only those meeting the criteria (major and lacking coverage) need it.
53
Carcinogenicity Study Protocol Submissions (FDA Guidance) 41. FDA’s guidance on carcinogenicity study protocol submissions recommends sponsors engage FDA prior to starting 2-year rodent carcinogenicity studies. What is the advised timing? a. Notify FDA about 30 days in advance that a carcinogenicity protocol will be submitted, and seek a Special Protocol Assessment review before initiating the study . b. Submit the protocol after completing the 2-year study c. Only send the protocol if FDA specifically requests it d. Include the carcinogenicity protocol in the NDA submission rather than beforehand
Explanation: Option a is correct. FDA encourages sponsors to interact early: at least 30 days before submitting the carcinogenicity study protocol, let the Agency know it’s coming, then submit the protocol with a request for Special Protocol Assessment (SPA) for FDA to review it within 45 days . The goal is to reach agreement on the study design prospectively. Options b and d are too late in the process (waiting until after the study or until NDA means missed opportunity for FDA input). Option c is incorrect because sponsors should be proactive in seeking protocol concurrence; it’s an offered FDA process, not only on FDA’s solicitation.
54
42. Who within FDA reviews and comments on proposed animal carcinogenicity study protocols? a. The CDER review division with input from the Carcinogenicity Assessment Committee (CAC) . b. The Food Advisory Committee c. Only the local FDA field office d. No one – FDA does not review study protocols for carcinogenicity studies
Explanation: Option a is correct. In CDER, the primary review of a carcinogenicity protocol is by the drug’s Review Division, which consults with the specialized CAC or Executive CAC – expert committees that evaluate carcinogenicity study designs . They provide written comments on dose selection, species choice, study duration, etc., to ensure the protocol is adequate. Options b and c are unrelated; the Food Advisory Committee deals with food, not drug carcinogenicity, and FDA field offices are for inspections, not protocol review. Option d is false – FDA actively offers to review and concur on carcinogenicity protocols via the SPA process.
55
43. FDA’s carcinogenicity protocol guidance lists information sponsors should include to facilitate FDA’s review. Which of the following is one of those recommended information elements? a. Results of a 90-day dose-rangefinding toxicity study under conditions comparable to the planned carcinogenicity study (same species, route, etc.) . b. A list of all manufacturing vendors for the drug c. Patient clinical trial data demonstrating efficacy d. Marketing plans for the drug after approval
Explanation: Option a is correct. FDA expects to see a supporting toxicity study report (usually ~90 days) that justifies dose selection for the 2-year bioassay, using the same species/strain, route, and possibly diet that will be used in the carcinogenicity study . This is critical to show what doses can be tolerated chronically. The guidance also asks for other info such as metabolic profiles (human vs animal), toxicokinetics, etc., but not the items in b, c, d. Options b, c, d are outside the scope of a protocol submission (which is focused on preclinical study design, not manufacturing or marketing or clinical efficacy).
56
44. Which of the following data is FDA expecting in a carcinogenicity protocol submission to justify the dose selection for the rodent bioassay? a. Toxicokinetic data showing estimated exposure (AUC) in animals at the candidate doses, compared to human exposure . b. Only a hypothesis with no data c. A list of safety pharmacology assays performed d. Epidemiology data in humans for similar drugs
Explanation: Option a is correct. FDA wants toxicokinetic (TK) data from the animal studies to ensure the doses chosen provide sufficient exposure multiples relative to human exposure . The submission should estimate steady-state AUC in the test species for the parent drug and major metabolites at the proposed doses, and compare those to human AUC at the maximum recommended human dose . This helps in deciding if a dose is high enough (e.g., achieving ≥10× human exposure or reaching an exposure plateau). Options b and d are not applicable; FDA expects actual preclinical data, not just guesses or unrelated human epidemiology. Option c (safety pharmacology tests) isn’t specifically part of the carcinogenicity protocol package – the key is dose justification data like TK and metabolism.
57
45. FDA’s guidance suggests including metabolic profile information in the protocol submission. What is the purpose of providing human and animal metabolism data before a carcinogenicity study? a. To ensure the animal model produces similar major metabolites so that any human-specific metabolites aren’t missed in the long-term study . b. To prove the drug is effective (via metabolites) c. To identify impurities in the drug d. It’s not necessary to include metabolism data
Explanation: Option a is correct. Sponsors should include metabolic profiles in humans and in the species to be used for the carcinogenicity study . This helps FDA evaluate whether the chosen animal species is a good surrogate (i.e., does it form the major human metabolites?). If a major human metabolite is absent in the rodent, that might influence study design or the need for a second species/alternative assays. Options b and c are unrelated to protocol design (efficacy and impurity info are handled elsewhere). Option d is incorrect – metabolism data are explicitly called for as part of the protocol review package to justify the model and dose selection .
58
46. The guidance highlights that positive genotoxicity findings for a drug or its metabolites can influence the carcinogenicity study approach. What impact might a positive genotox signal have? a. It may indicate that a standard 2-year rodent bioassay is not appropriate and that an alternative carcinogenicity assay or different dose selection approach should be considered . b. None – genotoxicity results don’t affect long-term study design c. It automatically cancels the need for a carcinogenicity study d. It means the highest doses should be increased because genotoxic compounds need higher exposure testing
Explanation: Option a is correct. Positive genotoxicity (e.g., a mutagenic result) is a red flag that can preclude certain dose rationales (like using high exposure multiples or limit doses) and might suggest that the standard 2-species/2-year protocol could be reconsidered in favor of alternative approaches (such as shorter-term transgenic models) . Essentially, if something is genotoxic, it might be assumed carcinogenic, and special study designs (like rasH2 mouse studies) might be used instead of a full 2-year rat study. Option b is false – genotox definitely influences plans. Option c is also wrong; a positive genotox doesn’t cancel carcinogenicity assessment, it heightens concern (sometimes a carcinogenicity study might still be done but perhaps only one species or a different design). Option d is the opposite of FDA’s advice – you wouldn’t push doses higher if you already know the compound is genotoxic; the focus would be on understanding risk, possibly at lower exposures.
59
47. FDA’s carcinogenicity protocol guidance allows use of a limit dose in rodents under certain conditions. What is the limit dose typically cited, and what condition must be met? a. 1500 mg/kg/day, provided that dose yields an exposure at least 10 times the human AUC at the maximum recommended human dose (MRHD) . b. 150 mg/kg/day, if it’s well tolerated c. 5000 mg/kg/day for any drug, universally d. 1500 mg/m² in rodents, regardless of human exposure
Explanation: Option a is correct. ICH S1C(R) and the FDA guidance recognize 1500 mg/kg/day in rodents as a potential limit dose for carcinogenicity testing, but only if at that dose the systemic exposure (AUC) in rodents is at least 10-fold higher than the exposure in humans at the MRHD . In other words, if 1500 mg/kg achieves a huge margin over human exposure, it’s unnecessary to test higher doses. Options b and c are incorrect numeric values (150 mg/kg is not the cited limit, and 5000 mg/kg is beyond practical limits and not in guidance). Option d is wrong unit/application; 1500 mg/kg (not mg/m²) is specified, and the condition is tied to exposure multiples, not just the dose amount .
60
48. Why does FDA recommend including plasma protein binding data for the parent drug and metabolites in the protocol package? a. To help interpret differences in free drug exposure between species, which can affect dose selection and relevance . b. Because protein binding determines if a compound is carcinogenic c. To decide what type of diet to feed animals d. It actually does not recommend providing protein binding info
Explanation: Option a is correct. The guidance asks for plasma protein binding data in the rodent species vs humans for the parent and major metabolites . This is to assess whether differences in protein binding could lead to different free (active) drug exposures. If, for example, the drug is much less protein-bound in animals than humans, animals effectively get higher active exposure at the same total concentration. Such info can influence dose setting (to ensure a fair safety margin). Options b and c are unrelated to the role of protein binding. Option d is incorrect as the guidance explicitly lists protein binding data as helpful information .
61
49. The guidance advises sponsors to document when reduced food consumption and body weight gain in a dietary carcinogenicity study are due to palatability. Why is this important? a. Because if weight loss is due to poor taste (palatability) of the drug in feed, the max tolerated dose may not have truly been reached, and switching to gavage might allow higher doses . b. Because FDA is concerned about animal welfare more than study results c. Because palatability issues invalidate the study d. It isn’t important; any weight loss is treated the same
Explanation: Option a is correct. In feed studies, sometimes the highest dose is limited by animals not eating enough (due to the drug making the feed unpalatable), causing weight loss. The guidance says sponsors should show that weight loss is truly due to toxicity and not just refusal to eat . If it’s a palatability issue, a different administration route (like gavage) might allow testing a higher dose that animals would tolerate taste-wise. This ensures the chosen top dose is a genuine MTD (maximum tolerated dose), not an artifact of poor flavor. Option b is tangential – while welfare is always a concern, the context here is scientific validity. Option c is extreme; palatability issues don’t invalidate the study, but they need to be managed/documented. Option d is false because the cause of weight loss matters for dose interpretation.
62
50. What is the rationale for FDA encouraging sponsors to submit prior study reports or summaries that are critical to dose selection along with the carcinogenicity protocol? a. It helps FDA reviewers verify the basis for the chosen high dose (e.g. seeing the 90-day study results), speeding up review and agreement . b. It is legally required to resubmit all data c. FDA does not want to cross-reference IND files d. To check if the sponsor misreported any data in the protocol write-up
Explanation: Option a is correct. Including the actual reports or at least detailed summaries of key supporting studies (like the range-finding tox study) allows FDA to quickly evaluate and concur with the proposed doses without having to dig through earlier files . It’s about efficiency – if the pharmacologist reviewing the protocol can see the relevant data in one package, they can more readily give feedback or agreement. Option b is not a strict requirement; it’s a recommendation for efficiency, not a law to resubmit. Option c (not wanting to cross-reference) is basically the reason behind (a) – providing the data upfront is easier than making FDA retrieve IND submissions – but the phrasing c is off. Option d is not the stated rationale (FDA isn’t presuming dishonesty; they just need the info to evaluate the protocol properly).