Preclinical Toxicity Endpoints for Large Molecule Drugs in FDA Approval
By Rhizome Team
Preclinical toxicity testing is a critical component of large molecule drug development, providing essential safety data to support FDA approval. Understanding which toxicity endpoints are most commonly evaluated helps sponsors design comprehensive preclinical programs that meet regulatory expectations.
For biologics, toxicity endpoints must be carefully selected to assess potential adverse effects while accounting for the unique characteristics of large molecule drugs, including their immunogenicity, species-specific activity, and complex pharmacology. The FDA expects toxicity studies to follow Good Laboratory Practices (GLP) and align with ICH guidelines for biotechnology-derived pharmaceuticals.
Here we examine the toxicity endpoints used in preclinical testing for large molecule drugs seeking FDA approval, including assessments of organ toxicity, clinical pathology, histopathology, and other safety parameters. This analysis provides practical guidance for biologics developers designing preclinical safety studies.
Want to read the citations or ask Rhizome a follow-up question? Schedule a call
what toxicity endpoints are used in preclinical testing for large molecule drugs for FDA approval?
Answer
Core toxicity endpoints FDA expects for large molecule drugs (biologics)
FDA and ICH guidances for biotechnology-derived pharmaceuticals (e.g., ICH S6 and S6(R1)) outline a consistent set of nonclinical safety endpoints, tailored to the biology of the product and informed by pharmacology, species specificity, and mechanism of action 192021222324253254256257258. Across large molecules, the following endpoints are central:
General and organ toxicity
- Repeat‑dose toxicity with standard clinical observations, mortality, body weight and food consumption, clinical pathology (hematology, clinical chemistry, urinalysis), organ weights, gross necropsy, and comprehensive histopathology to identify target organs and reversibility 2224124234323631194. Examples include multi‑system clinical pathology and organ/tissue histology in ipilimumab and golimumab programs 34323631241242, and full necropsy/histopathology in bezlotoxumab 194.
Safety pharmacology (vital organ system functions)
- Effects on the cardiovascular, respiratory, and central nervous systems, often built into repeat‑dose studies; ECG/QTc and hemodynamics are typical cardiovascular endpoints 22461793651. Guidance for exploratory INDs and severe hematologic disorders specifies CNS/respiratory/cardiovascular assessments, with ECGs generally captured in nonrodents 46179. Atezolizumab programs included QT/QTc evaluations 51.
Immunotoxicity and immunogenicity
- Immune system endpoints are prominent for biologics. FDA’s immunotoxicity guidance and ICH S6 call for assessing immune stimulation/suppression, humoral and cell‑mediated immunity, and potential immunogenicity (anti‑drug antibodies, ADA), with additional endpoints such as cytokine release and developmental immunotoxicity when warranted 2122122121123124. S6(R1) further recommends ADA assessments to interpret altered exposure, pharmacodynamics, or immune‑mediated reactions (e.g., immune complex disease, vasculitis, anaphylaxis) and to characterize neutralizing potential when ADA are detected, noting that animal ADA is not predictive of human immunogenicity 258.
- Concrete implementations include ADA/neutralizing antibody detection and immune function tests (e.g., TDAR), immunophenotyping, and immune response challenges in NHP studies of mAbs (golimumab, atezolizumab, rituximab‑pvvr) 2412422435140. Programs often integrate ADA sampling with TK and pathology to interpret findings (e.g., bezlotoxumab, ipilimumab) 196195194273271.
Reproductive and developmental toxicity (DART)
- Embryo‑fetal development (EFD) and pre/postnatal development (PPND) studies are conducted when clinically relevant, frequently in a pharmacologically relevant species (often NHPs) for mAbs and other biologics due to species specificity 2119256. Endpoints include fetal/offspring viability, external, skeletal, and visceral morphology, and maternal/offspring toxicity 256258. Product examples and guidance highlight case‑by‑case needs for biologics, including embryo‑fetal loss with PD‑1/PD‑L1 inhibition and NHP EFD studies for mAbs 54241242243256.
Genotoxicity
- Routine genotoxicity testing is generally not applicable to most proteins/mAbs per ICH S6; nevertheless, it may be considered for certain modalities or when concerns arise (e.g., oligonucleotides, some imaging biologics) 192321474556. Exploratory IND guidance notes standard genotox batteries while recognizing they may be inappropriate for proteins 4745. Oligonucleotide therapeutics guidance describes in vitro genotoxicity assays (e.g., Ames, mammalian cell gene mutation) for these large molecules 56.
Carcinogenicity
- Standard 2‑year rodent bioassays are often not informative for mAbs; carcinogenic risk is addressed using product‑specific, weight‑of‑evidence approaches (e.g., receptor expression, proliferation indices) and performed only when concerns arise 2319254. Many antibody programs do not conduct rodent carcinogenicity, consistent with ICH S6/S6(R1) 241242254. In contrast, for oligonucleotide therapeutics, in vivo carcinogenicity studies are generally expected, guided by ICH S1 and M3(R2) 5760. Immunosuppression‑related tumor risk can be a consideration in immunotoxicity assessments 122121123124.
Local tolerance
- Assessment of adverse effects at the administration site (e.g., inflammation, erythema, edema) through gross and microscopic evaluation, or via specialized local tolerance paradigms for certain biologic classes (e.g., vaccines) 2332248183. Ipilimumab and canakinumab programs documented local injection‑site findings histologically 32248, and vaccine guidances emphasize reactogenicity endpoints (induration, erythema, granuloma, sterile abscess) 183.
Tissue cross‑reactivity and off‑target binding (for protein therapeutics)
- Tissue cross‑reactivity (TCR) studies in human tissues (and sometimes in relevant animal tissues) are used to assess potential off‑target binding and inform species selection and toxicity risk 151134. Examples include TCR packages for monoclonal antibodies and inclusion of in vitro TCR to support bezlotoxumab 151134194.
Toxicokinetics/pharmacokinetics and exposure–response
- TK/PK assessments are integral to interpreting toxicity, selecting doses, and establishing exposure margins in repeat‑dose studies; S6/S6(R1) recommend dose selection based on PK/PD principles with TK sampling aligned to toxicity endpoints 22253257. Programs routinely integrate TK with ADA to interpret exposure changes (e.g., bezlotoxumab) 194196.
Modality-specific nuances
Monoclonal antibodies and Fc‑fusion proteins
- Emphasis on relevant species (often NHP) for general tox and DART due to species specificity; standard safety pharmacology embedded in GLP tox; TCR to assess off‑target binding; immunogenicity/TDAR/immunophenotyping as needed. Genotoxicity and carcinogenicity are generally not performed unless specific concerns exist 192122151253254. Product examples (golimumab, ipilimumab, atezolizumab, rituximab‑pvvr) illustrate immunogenicity, immune function, and organ tox endpoints used in practice 241242243343236315140.
Antibody–drug conjugates (ADCs)
- In addition to the biologic’s endpoints, ADCs require attention to payload‑related toxicities and metabolites; FDA highlights QTc risk evaluations focusing on the unconjugated payload/metabolites and comprehensive immunogenicity assessments to detect ADAs to any component (antibody, payload, linker) 169171172173. Organ impairment studies are used to correlate exposure changes with toxicity signals 171172.
Oligonucleotide therapeutics
- Expectation for general toxicity in two species (rodent and nonrodent), reproductive/developmental toxicity per ICH S5(R3), in vivo carcinogenicity in many programs, and targeted genotoxicity assays; juvenile studies when indicated (ICH S11). Nonhuman primates are commonly the nonrodent due to sequence conservation and known pharmacology 555657585960.
Vaccines (biologics)
- Repeat‑dose toxicity, local tolerance/reactogenicity, DART as applicable, and immunopathology risks (e.g., enhanced respiratory disease for respiratory pathogens); animal immunogenicity studies support characterization but are typically considered separately from “toxicity” per se 183157158.
Cellular and gene therapies (including genome editing)
- Safety packages emphasize local and systemic toxicities, biodistribution/persistence, off‑target effects (e.g., on/off‑target editing, chromosomal rearrangements), and immune responses to vector and transgene; organ‑specific pathology (e.g., neuropathology for neurotropic products) is important 11711961629495. These endpoints contextualize toxicity for large molecule modalities beyond conventional proteins.
Medical imaging biologics
- Repeat‑dose toxicity, genotoxicity (as appropriate), reproductive/developmental toxicity, immunotoxicity, and local tolerance/irritancy are considered, with special attention to immunogenicity and species specificity for biological imaging agents 6768697072.
Endocrine‑related concerns (when applicable)
- For products with potential endocrine interactions, additional endpoints address endocrine‑sensitive organs (thyroid, adrenal, reproductive organs, pituitary), developmental endpoints (e.g., nipple retention, anogenital distance), and carcinogenicity assessments tailored to endocrine effects 798180.
Developmental immunotoxicity and pediatric considerations
- FDA’s immunotoxicity guidance recognizes developmental immunotoxicity (DIT) when exposure occurs during fetal, neonatal, or juvenile windows; endpoints include immunophenotyping, immune function testing, and anatomical integrity of the immune system, integrated with a weight‑of‑evidence approach 122121123124.
- ICH S11 outlines juvenile animal study endpoints relevant to biologics, including detailed organ histopathology, clinical pathology, bone endpoints, ophthalmology, CNS/behavioral testing, and TK, with ADA collection for biopharmaceuticals when appropriate 104105106107108103.
How these endpoints are applied in practice: Illustrative examples
- Ipilimumab (CTLA‑4 mAb): Broad general tox panel (hematology, coagulation, clinical chemistry, urinalysis), organ weights, gross and microscopic pathology, cardiovascular safety pharmacology (ECG intervals), injection‑site local tolerance, and immunotoxicity/antigenicity sections in the dossier; reproductive and carcinogenicity sections included in the nonclinical evaluation 343236313335.
- Golimumab (anti‑TNF mAb): Chronic repeat‑dose tox in cynomolgus monkeys and mice, reproductive tox (EFD, PPND), local tolerance, immunogenicity (APA) and TDAR functional assays, macroscopic/histopath evaluations; genotoxicity/carcinogenicity generally not performed per ICH S6 241242243246.
- Atezolizumab (anti‑PD‑L1 mAb): ADA and neutralizing antibody assay development and impact assessments; multi‑organ inflammation and immune‑mediated tissue damage in animals; cardiovascular, respiratory, and neurologic functional evaluations; developmental/reproductive toxicity concerns including embryo‑fetal loss with PD‑1/PD‑L1 inhibition; QT/QTc evaluation 514954.
- Rituximab‑pvvr (biosimilar): NHP comparative studies assessed potential toxicity, local tolerance, hematology, immunophenotyping (e.g., B‑cell PD), TK/PK, and immunogenicity 40.
- Bezlotoxumab (anti‑toxin mAb): Repeat‑dose tox included mortality/clinical signs, body weight, food consumption, clinical pathology, organ weights, histopathology, ophthalmology, TK, ADA assessments, and in vitro tissue cross‑reactivity; NOAEL established from these endpoints 193194195196197.
- Canakinumab (anti‑IL‑1β mAb): Chronic/subchronic NHP tox with organ histopathology, local tolerance at injection sites, immunogenicity via validated assays, reproductive tox, limited utility of genotox/carcinogenicity unless concerns arise, and cardiovascular safety pharmacology (e.g., QT) 247248249251252.
Notes on species and interpretation that affect endpoint strategy
- Species selection shapes feasible endpoints (e.g., TDAR, reproductive studies). ICH S6 emphasizes a relevant species for biotechnology‑derived products; NHPs are frequently used when rodent cross‑reactivity is lacking 192120256257.
- Animal immunogenicity informs study interpretation (exposure loss, immune complex disease), but is not predictive of human immunogenicity; still, sponsors are advised to collect ADA in nonclinical studies when immune‑mediated changes are observed 258.
- For certain product classes (e.g., oligonucleotides), FDA expects broader toxicology (including carcinogenicity) than for classical protein therapeutics 5760.
Why these endpoints matter for FDA decision-making
- FDA’s approach integrates pharmacology, exposure, and a mechanism‑informed selection of toxicity endpoints to ensure the nonclinical package anticipates human risks. Endpoints like TCR and immunotoxicity reduce unforeseen immune‑mediated toxicities; DART and juvenile assessments protect at‑risk populations; and consistent general tox and safety pharmacology enable identification of target organs, dose‑limiting toxicities, and reversibility, supporting safe first‑in‑human dosing and risk management 192212225325617946.
Practical checklist: What a preclinical large molecule program typically includes
- General/organ toxicity: clinical observations, clinical pathology, organ weights, gross and microscopic pathology; recovery assessments 2234323631194.
- Safety pharmacology: cardiovascular (including ECG/QTc as needed), respiratory, and CNS endpoints, often within repeat‑dose tox 461793651.
- Immunotoxicity/immunogenicity: ADA (binding/neutralizing) and interpretation, immune function tests (e.g., TDAR), immunophenotyping, and cytokine release assays when relevant; developmental immunotoxicity if indicated 1222122258.
- Reproductive/developmental toxicity: EFD and PPND when clinically relevant, often in a pharmacologically relevant species (commonly NHP for mAbs) 21256.
- Genotoxicity: generally not required for proteins/mAbs; consider for modalities like oligonucleotides per guidance 1923474556.
- Carcinogenicity: weight‑of‑evidence for proteins/mAbs; often not performed unless concerns arise; expected for many oligonucleotide programs 2542412425760.
- Local tolerance/reactogenicity: injection‑site assessments, including histology; vaccine‑specific local/systemic reactogenicity 32248183.
- Tissue cross‑reactivity: human (± animal) tissue panels to assess off‑target binding for mAbs and related proteins 151134194.
- Toxicokinetics/pharmacokinetics: exposure, exposure–response and dose selection integrated with toxicity endpoints 22253257.
- Modality‑specific needs: biodistribution/off‑target editing for gene therapies; QTc and metabolite‑focused assessments for ADC payloads 1179495169171.
Taken together, these endpoints reflect FDA’s and ICH’s biotechnology guidances, tailored by product class, mechanism, and patient population, and are exemplified across approved biologics’ nonclinical programs 192021222324122253254256257258466769151134241242343236315140194247248249.