FDA Flashcards
(62 cards)
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.
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.
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.
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.
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).
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.
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.
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.
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.
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.
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).
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.
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.
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.
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.
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.
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.
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.
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
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.
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.
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 .
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.
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 .
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
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 .
- 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
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) .
- 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
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.
- 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 .
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.
- 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.
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 .
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
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.
- 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
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.
- 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
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.
- 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
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.
- 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
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 .
- 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
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.
- 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
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.