Fact-checked by Grok 2 weeks ago

Preclinical development

Preclinical development is the phase of pharmaceutical that evaluates potential drug candidates through laboratory-based and to assess their pharmacological effects, , , and preliminary prior to initiating human clinical trials. This stage bridges target identification and lead optimization in with the regulatory submission of an (IND) application, focusing on generating data to support safe progression to Phase 1 trials. Key activities include testing in cell cultures to examine mechanisms of action and experiments in animal models—typically and non-rodents—to investigate , , , (ADME), and dose-dependent toxicities. These studies adhere to (GLP) standards to ensure data reliability for regulatory review. Preclinical development is critical for filtering out unsafe or ineffective compounds, as only a small fraction of candidates advance, reflecting empirical realities of biological complexity and interspecies differences that limit perfect prediction of human outcomes. Notable challenges include the imperfect translatability of animal data to humans, contributing to high attrition rates—over 90% of drugs fail post-preclinical—despite rigorous testing, which underscores ongoing needs for advanced models like organoids or computational simulations to enhance causal inference and reduce reliance on traditional paradigms. While animal testing remains indispensable for establishing dose safety margins and identifying off-target effects through direct causal observation, it faces scrutiny over ethical costs, though evidence affirms its foundational role in averting human harm from unvetted agents.

Definition and Objectives

Role in the Drug Development Pipeline

Preclinical development serves as the critical intermediary stage in the drug development pipeline, positioned after the initial phase—where potential therapeutic compounds are identified and synthesized—and before the initiation of human clinical trials. This phase focuses on rigorously evaluating candidate molecules through non-human testing to establish foundational evidence of , potential , and, most importantly, profiles that justify progression to human studies. Regulatory bodies, such as the U.S. (FDA), require comprehensive preclinical data as part of the (IND) application, which must demonstrate that the compound is reasonably safe for initial dosing in humans and unlikely to cause serious harm under proposed conditions. The primary role of preclinical development is to mitigate risks inherent in advancing unproven compounds, thereby optimizing in a characterized by high failure rates. By conducting (cell-based) and (animal) studies, researchers generate data on , , , (), thresholds, and dose-response relationships, enabling the refinement or elimination of candidates that exhibit unacceptable liabilities early on. This filtering function is essential, as only approximately 10-20% of compounds entering preclinical testing ultimately receive regulatory approval, with preclinical often exceeding 80-90% when accounting for broader discovery-to-IND transitions, primarily due to inadequate signals or concerns. Adherence to (GLP) standards, mandated under 21 CFR Part 58, ensures data integrity and reproducibility, forming the evidentiary basis for IND review, where the FDA typically responds within 30 days. In economic terms, preclinical development typically spans 1-3 years and incurs costs ranging from $15 million to $100 million per candidate, representing a fraction of the total $1-2 billion average for successful drugs but serving as a cost-effective checkpoint to avoid the far higher expenses of clinical phases, which account for the majority of R&D outlays. Despite these efforts, limitations in translational fidelity—such as differences between animal models and —mean that even promising preclinical results predict clinical success imperfectly, underscoring the phase's role not as a guarantee but as a probabilistic reducer informed by empirical testing. Successful completion enables the pipeline's advancement to Phase 1 trials, where initial and safety are confirmed, while failures inform iterative improvements in discovery methodologies or target selection.

Primary Goals and Metrics of Success

The primary goals of preclinical development encompass establishing a preliminary profile to identify potential and target organs, determining an initial starting dose and scheme for human trials, and characterizing pharmacological activity to support proof-of-concept in relevant biological models. These efforts also aim to generate data on , , , and (ADME) properties to predict human and inform clinical dosing strategies. Additionally, preclinical studies seek to flag parameters for clinical monitoring, such as biomarkers of , and to exclude certain patient populations based on observed adverse effects in nonclinical models. Metrics of success are quantitative and qualitative benchmarks that gauge whether a candidate merits advancement to (IND) submission. Key among these is the (NOAEL), derived from repeat-dose studies in and non-, which establishes the highest dose devoid of significant adverse effects and underpins human equivalent dose calculations—often scaled by a factor of 1/50 for interspecies based on differences. Favorable , including exceeding 20-30% in preclinical and half-lives supporting once-daily dosing, signal viable drug-like properties. metrics include potent dose-response relationships, with effective concentrations (EC50) in cellular or animal models aligning with achievable exposures, and a (ratio of toxic dose to effective dose, ideally >10) indicating an acceptable safety margin. Absence of disqualifying findings, such as in Ames assays or via channel inhibition ( >10 μM preferred), further defines success, as these endpoints predict clinical risks and regulatory hurdles. Overall, a candidate's progression hinges on integrated data demonstrating translatability to humans, with success rates historically low—around 50-70% of INDs advancing from preclinical stages—underscoring the need for robust, predictive models.

Core Methodologies

In Vitro Testing

In vitro testing encompasses laboratory experiments performed on isolated biological components, such as cells, tissues, , or biomolecules, outside of a living , often in multi-well plates or bioreactors to simulate controlled physiological conditions. These assays form an initial phase of preclinical development, enabling of drug candidates for preliminary , , and toxicity profiles before advancing to more complex models. Typically conducted after identification, in vitro studies help prioritize candidates by assessing target engagement, such as receptor binding or inhibition, and basic pharmacokinetic properties like and . Efficacy profiling in vitro often involves cell-based assays measuring outcomes like , , or functional responses in relevant lines; for instance, cancer drug candidates may be tested for in tumor cultures using metrics such as values, which quantify the concentration required to inhibit 50% of growth. Toxicity evaluations include genotoxicity assays (e.g., for mutagenicity), hERG channel assays for cardiac risk, and cultures for metabolic liability, aiming to identify off-target effects early. Advanced models, such as 3D organoids or systems, enhance predictivity by mimicking tissue architecture and multi-cellular interactions, though 2D monolayers remain dominant for initial high-throughput efforts, accounting for nearly half of screening in oral . These tests comply with (GLP) standards when generating data for regulatory submissions, such as (IND) applications. Advantages of in vitro testing include its cost-effectiveness, scalability for screening thousands of compounds rapidly, and ethical benefits by minimizing animal use in early stages. It allows precise control over variables, facilitating mechanistic insights unattainable in whole-organism models. However, limitations persist: in vitro systems often fail to replicate systemic , immune responses, or multi-organ interactions, leading to discrepancies where promising candidates underperform ; for example, in vitro potency correlates imperfectly with clinical exposure due to absent , , , and dynamics. Such gaps underscore the need for tiered approaches integrating in vitro data with in silico predictions and confirmatory studies to improve translatability.

In Silico Modeling

In silico modeling refers to the use of computational algorithms and simulations to predict drug behavior, molecular interactions, and biological outcomes during preclinical development, enabling the evaluation of candidates prior to resource-intensive in vitro or in vivo studies. These approaches leverage mathematical models, bioinformatics, and increasingly to analyze vast datasets, forecast properties such as binding affinity, , and , and optimize . Originating from early quantitative structure-activity relationship (QSAR) models in the , in silico methods have evolved with advances in computing power and , becoming integral for high-throughput of chemical libraries exceeding millions of compounds. Key techniques include ligand-based modeling, such as QSAR and pharmacophore mapping, which correlate chemical structures with observed activities using statistical regressions or without requiring target structures; and structure-based methods like molecular and dynamics simulations, which predict how small molecules fit into protein binding sites derived from or . Physiologically based pharmacokinetic (PBPK) models further simulate absorption, distribution, metabolism, and excretion () profiles by integrating anatomical and physiological parameters, aiding dose predictions across species. Recent integrations of AI, including for , have accelerated hit identification, as demonstrated by Insilico Medicine's AI-generated anti-fibrotic candidate ISM001-055, which advanced from discovery to Phase I trials in 30 months by 2023. In preclinical contexts, tools support target validation by simulating pathway perturbations, toxicity forecasting via models like Derek Nexus for reactive identification, and efficacy profiling through multi-scale quantitative systems (QSP) simulations that bridge molecular to organismal levels. For instance, has identified inhibitors for targets, prioritizing compounds for synthesis based on predicted binding energies. These methods reduce failure rates in later stages; AI-discovered molecules exhibit 80-90% success in Phase I trials, surpassing historical industry averages of around 70%. Despite advantages in speed and cost—potentially screening 10^6 compounds in days versus weeks for assays—in silico predictions carry limitations, including reliance on training data quality, which can propagate biases or overlook off-target effects not captured in simplified models. Accuracy varies, with success rates often 50-70% for pose but lower for novel scaffolds, necessitating experimental validation to mitigate false positives. Regulatory bodies like the FDA endorse qualified models for ADMET extrapolation but require bridging to empirical data under frameworks like model-informed (MIDD). Ongoing refinements, such as hybrid physics-based and data-driven approaches, aim to enhance reliability for broader acceptance in submissions.

In Vivo Animal Studies

In vivo animal studies in preclinical drug development involve the administration of candidate compounds to live animals to evaluate their , , (PK), and pharmacodynamics (PD) within intact physiological systems, bridging the gap between and human data. These studies assess whole-body responses, including absorption, distribution, metabolism, excretion (), target engagement, and potential toxicities that may not manifest in isolated cell or computational models. Typically conducted under (GLP) standards, they provide critical data for determining the no observable adverse effect level (NOAEL) and establishing safe initial doses for first-in-human trials. Common animal models include rodents such as mice and rats for initial screening due to their small size, rapid reproduction, genetic engineering capabilities, and lower costs, enabling high-throughput testing of efficacy in disease models and basic toxicity profiles. For more advanced assessments requiring closer physiological resemblance to humans—particularly for PK in larger body sizes or specific organ functions—non-rodent species like dogs, minipigs, or non-human primates (e.g., cynomolgus monkeys) are employed, especially in safety pharmacology studies evaluating cardiovascular or neurological effects. Selection of species follows regulatory guidance prioritizing relevance to human biology while minimizing animal use per the 3Rs principles (replacement, reduction, refinement). Key study types encompass acute (single-dose) toxicity tests lasting up to 14 days to identify immediate hazards; subchronic (repeated-dose, 14-90 days) and (6-12 months) studies to detect cumulative effects like organ damage or ; developmental and reproductive toxicology (DART) to assess impacts on , embryofetal , and perinatal outcomes; and safety pharmacology core battery tests for functional toxicities in cardiovascular, respiratory, and central nervous systems. Efficacy studies model human diseases, such as xenograft tumors in immunodeficient mice for candidates or genetically modified rodents for metabolic disorders, measuring endpoints like tumor regression or modulation. Doses are escalated from therapeutic levels to multiples of the expected human exposure (e.g., 10-100-fold) to probe margins of . Historically mandated for Investigational New Drug (IND) submissions to the U.S. FDA, these studies supplied and data justifying clinical progression, with requirements including at least two species (one , one non-rodent) for repeated-dose toxicity. The FDA Modernization Act 2.0, enacted December 29, 2022, eliminated the statutory requirement for in preclinical safety assessments, allowing non-animal alternatives like organ-on-chip or advanced models if they demonstrate equivalent predictivity. Nonetheless, data remains standard in most IND packages as of 2025, given ongoing validation needs for alternatives and the causal insights from systemic exposures, though critiques highlight poor translatability—e.g., animal models predict human toxicity with only 70-80% concordance for some endpoints, contributing to high rates (over 90% of candidates fail post-preclinical).

Pharmacological Assessments

Pharmacokinetics and ADME

(PK) encompasses the study of how an organism affects a , primarily through , , , and (), which is evaluated early in preclinical development to forecast systemic exposure, guide dosing regimens, and mitigate risks such as suboptimal or unexpected accumulation. These assessments inform whether a candidate can achieve therapeutic concentrations without excessive , using both and models to bridge species differences and human predictions. In vitro ADME assays provide high-throughput screening for key properties: solubility is measured via UV spectrophotometry across physiological pH ranges (e.g., 5.0–7.4) to ensure adequate dissolution; permeability employs parallel artificial membrane permeation assay (PAMPA) or cell monolayers to gauge intestinal absorption potential; metabolic stability uses liver microsomes or hepatocytes to quantify clearance rates, often via LC/MS/MS after incubation periods like 60 minutes; and via equilibrium dialysis assesses free fraction availability. (CYP) inhibition screens for five major isoforms (e.g., ) to flag drug-drug interaction risks, with benchmarks targeting low values for viability. These assays, requiring minimal compound (1–7 mg), enable rapid iteration during lead optimization, prioritizing candidates with high permeability, stability, and solubility per Lipinski's rule extensions. In vivo PK studies, typically in rodents and non-rodents like dogs or monkeys, involve intravenous and oral dosing (5–50 mg/kg) followed by serial blood sampling to derive parameters such as area under the curve (), maximum concentration (Cmax), half-life (), clearance (CL), volume of distribution (), and oral bioavailability (). Toxicokinetics (TK), integrated into repeated-dose toxicity studies per ICH M3(R2) guidelines, monitors drug in the same animals to correlate levels with adverse effects, ensuring to trials by comparing species-specific . For instance, comprehensive PK profiles collect nine time points up to 24 hours post-dose to model accurately. Regulatory frameworks, including FDA and ICH M3(R2), mandate / data prior to () submissions: metabolic profiling and protein binding in humans and animals, plus systemic exposure in toxicity species, must precede clinical trials, with full characterization (including human-unique metabolites exceeding 10% exposure) required before phase 3. This ensures safe , as poor preclinical correlates with 40–50% of clinical failures due to exposure inadequacies, emphasizing empirical validation over assumptions.

Pharmacodynamics and Efficacy Profiling

In preclinical development, (PD) profiling evaluates the biochemical, physiological, and molecular effects of a candidate on its intended and biological pathways, establishing dose-response relationships essential for predicting therapeutic potential. This process quantifies parameters such as the half-maximal effective concentration (50), defined as the drug concentration producing 50% of the maximum response, and Emax, the peak effect attainable, often using sigmoidal models to fit data from concentration-effect curves. These assessments occur primarily through assays, including radioligand binding for affinity (e.g., Ki values) and functional assays like calcium or inhibition to measure potency and selectivity against off-targets. PD data inform decisions by linking drug exposure to , often integrated with via PK/PD modeling to simulate exposure-response profiles. Efficacy profiling extends PD characterization to disease-relevant contexts, employing validated models to demonstrate therapeutic benefits such as symptom alleviation or modulation. efficacy is probed via lines engineered to mimic , measuring endpoints like viability or production, while studies use animal models—e.g., xenografts for , where tumor volume reduction quantifies response, or collagen-induced models for anti-inflammatories, tracking swelling and histological scores. These models aim to replicate human pathophysiology, though indicates variable translatability; for instance, preclinical in CNS disorders correlates poorly with clinical outcomes due to species differences in blood-brain barrier penetration and receptor expression. Regulatory bodies like the FDA require such data to support applications, emphasizing proof-of-concept in relevant species to justify human trials. Challenges in PD and efficacy profiling include assay variability and model limitations, with high attrition rates—over 90% of candidates failing to advance from preclinical to clinical stages—partly attributable to overestimated efficacy in non-human systems. Advanced approaches, such as models or organ-on-chip systems, seek to enhance predictivity by incorporating human-specific elements, though their routine adoption remains limited by validation needs. Overall, robust PD/efficacy data, corroborated across orthogonal models, underpin calculations (efficacy relative to toxicity thresholds) and guide dosing strategies for phase I trials.

Toxicological and Safety Evaluations

Toxicity Study Types

Toxicity studies in preclinical development evaluate the potential adverse effects of drug candidates on biological systems, primarily through controlled animal and assays to identify dose-response relationships, target organ toxicities, and margins of safety before human exposure. These studies are guided by international harmonized standards, such as those from the International Council for Harmonisation (ICH), which specify requirements for various study types to support (IND) applications. The design emphasizes dose escalation, multiple species (typically rodents and non-rodents), and endpoints like , , and mortality to establish no-observed-adverse-effect levels (NOAELs). Acute toxicity studies assess immediate or short-term effects following a single high-dose administration, often via oral, intravenous, or , to determine approximate lethal doses (e.g., LD50) and acute organ liabilities, though their standalone requirement has diminished in favor of repeated-dose data under ICH M3(R2) guidelines, as single exposures rarely predict chronic risks. These are conducted in over 14 days post-dosing, monitoring overt signs like convulsions or . Repeated-dose toxicity studies, including subchronic (typically 14-90 days) and chronic (6-12 months), evaluate cumulative effects from daily or intermittent dosing, mimicking intended clinical regimens to detect delayed toxicities such as hepatic enzyme induction or nephropathy. Subchronic studies in two species support early clinical trials, while chronic studies, required for chronic-use drugs, involve larger cohorts (20-50 animals/group) with recovery phases to assess reversibility. Endpoints include body weight changes, hematology, and necropsy, with non-rodent species like dogs or monkeys providing metabolic insights absent in rodents. Genotoxicity studies screen for DNA damage potential using batteries like the Ames bacterial reverse mutation assay, in vitro mammalian cell tests (e.g., chromosomal aberration), and micronucleus assays in , as per ICH S2(R1), to flag mutagens early and halt development of high-risk candidates. Positive findings trigger mechanistic follow-up, given false positives from metabolic differences between . Reproductive and developmental toxicity studies are segmented into three phases: fertility (mating/ in ), embryofetal (dosing during in rabbits/), and pre/postnatal (full cycle in rats), per ICH S5, to detect impacts on gametes, fetuses, or offspring viability, with two-species testing for species-specific placental transfer. These support trials in fertile populations, revealing effects like teratogenesis at doses below therapeutic levels. Carcinogenicity studies, conducted in (rats and mice) over 18-24 months per ICH S1, involve high-dose lifetime exposure to predict oncogenic via tumor incidence, though translation to humans is limited by species metabolic variances, with transgenic models sometimes supplementing for targeted therapies. These are typically deferred until late preclinical unless early signals warrant them. Additional specialized types, such as immunotoxicity (e.g., T-cell phenotyping) or (UV-exposed skin assays), are integrated when suggests , ensuring comprehensive without over-reliance on any single modality.

Determination of No Observable Adverse Effect Level (NOAEL)

The No Observable Level (NOAEL) is defined as the highest dose of a test substance at which there is no statistically or biologically significant increase in the severity or incidence of in the exposed animals relative to the concurrent control group. This metric is derived from nonclinical studies and serves as a for establishing safe starting doses in clinical trials by providing a below which no is anticipated. In practice, NOAEL determination requires careful evaluation of dose-response relationships across multiple endpoints, prioritizing the most sensitive species and study to reflect potential risk conservatively. NOAEL is typically identified in repeated-dose toxicity studies conducted under (GLP) standards, involving (e.g., rats) and non- (e.g., dogs or cynomolgus monkeys) species for durations scaled to the planned exposure, as outlined in ICH M3(R2) guidelines. Animals are allocated to dose groups (often including low, mid, high, and control), with exposures ranging from subchronic (e.g., 28 days) to chronic (e.g., 6-12 months) based on phases. Comprehensive assessments include daily clinical observations, body weight and food intake monitoring, , , clinical biochemistry, , gross necropsy, organ weights, and histopathological examinations of major organs. Adverse effects qualifying for NOAEL assessment encompass overt (e.g., mortality or severe clinical signs), target organ (e.g., histopathological lesions in liver or ), and effects on reproductive performance or embryofetal from dedicated studies. Non-adverse findings, such as adaptive responses (e.g., liver without functional impairment) or reversible physiological changes, are distinguished through weight-of-evidence , ensuring only causally linked, biologically relevant toxicities influence the NOAEL. The NOAEL is selected as the highest dose lacking such effects, equivalent to one level below the Lowest Observed Level (LOAEL), with statistical tests (e.g., ANOVA followed by post-hoc ) confirming where applicable. For extrapolation to humans, the species-specific NOAEL (in mg/kg) is normalized to Human Equivalent Dose (HED) using allometric scaling factors based on (e.g., divide rat NOAEL by 6.2, dog by 1.8), selecting the lowest HED from the most sensitive . An additional intraspecies safety factor of at least 10 is applied to account for pharmacokinetic and pharmacodynamic uncertainties, yielding the Maximum Recommended Starting Dose (MRSD); higher factors (e.g., 100-fold total) may apply for severe toxicities or steep dose-response curves. Simulations indicate inherent uncertainties in NOAEL estimation due to dose spacing and variability, with translation errors potentially exceeding 10-fold across , underscoring the need for robust study design. Regulatory acceptance requires integration of NOAEL data from pivotal GLP-compliant studies, often the most sensitive findings, to support (IND) applications.

Regulatory and Compliance Framework

Good Laboratory Practice (GLP) Standards

Good Laboratory Practice (GLP) standards comprise a set of regulations and principles designed to ensure the quality, reliability, and integrity of data generated from nonclinical laboratory studies, particularly those supporting regulatory submissions for pharmaceuticals, chemicals, and pesticides. These standards mandate systematic planning, execution, monitoring, recording, and reporting of studies to minimize errors, , and inconsistencies that could undermine safety assessments. In the context of preclinical development, GLP compliance is essential for toxicological and safety studies, as it provides regulators with assurance that the data accurately reflect the conducted experiments and are suitable for evaluating potential human risks before clinical trials. Noncompliance can invalidate study results, delaying drug development or leading to regulatory rejection. The origins of GLP trace back to the mid-1970s, when revelations of and poor practices in for food additives and pesticides prompted the U.S. (FDA) to propose regulations in 1978 under 21 CFR Part 58. These addressed systemic issues, such as inadequate documentation and uncontrolled study environments, identified in FDA audits that revealed up to 10-20% of submitted data as unreliable in some cases. The FDA's final regulations took effect in 1979, with amendments in 1987 to refine scope and procedures. Concurrently, the Organisation for Economic Co-operation and Development () developed harmonized GLP principles in 1981, revised in 1997, to facilitate mutual acceptance of data across member countries and reduce duplicative testing. These principles, now adopted by over 40 countries, emphasize comparable for international regulatory purposes. Core GLP requirements encompass several interrelated elements. Test facilities must maintain qualified personnel, including a study director responsible for overall conduct and a unit (QAU) to independently verify compliance through audits and inspections. Standard operating procedures (SOPs) are mandatory for all routine operations, from equipment to animal handling, ensuring . Studies require detailed protocols outlining objectives, methods, and acceptance criteria, with preserved in original form to allow reconstruction. Equipment must be suitable, calibrated, and maintained, while test systems—such as animals or models—demand characterization and humane treatment per ethical guidelines. Data handling protocols prevent alteration or loss, with computerized systems requiring validation for accuracy and . Final reports must include all amendments, deviations, and QAU statements, signed by the study director. In preclinical development, GLP applies selectively: exploratory studies may be non-GLP for efficiency, but pivotal safety studies—such as repeat-dose toxicity, , and reproductive —must adhere to GLP to support () applications. The FDA enforces compliance through bioresearch inspections, which in fiscal year 2023 examined over 100 facilities, issuing warnings for violations like inadequate SOPs or falsification. OECD compliance programs similarly promote acceptance via mutual joint visits and advisory documents. While GLP enhances trustworthiness, critics note it does not guarantee scientific validity, as it focuses on procedural integrity rather than study design flaws, yet empirical audits show GLP studies exhibit lower variability and higher rates compared to non-GLP counterparts.

Preparation for Investigational New Drug (IND) Submission

The preparation for an (IND) submission centers on compiling a comprehensive preclinical data package to demonstrate that the investigational product is reasonably safe for initial human testing, as required by FDA regulations in 21 CFR Part 312. This process involves integrating results from animal , , , and safety studies to permit an adequate , with the goal of identifying potential hazards without unreasonable exposure in early clinical phases. Sponsors must ensure that nonclinical laboratory studies comply with (GLP) standards under 21 CFR Part 58, as FDA may refuse to consider non-GLP data for IND review. Key preclinical components include detailed reports on animal and , encompassing acute and subchronic toxicity tests, assays, and reproductive/developmental toxicity studies where relevant, often conducted in two species (typically and non-rodent) to establish the no observable adverse effect level (NOAEL). These data must support dose selection for Phase 1 trials, with integrated summaries addressing , distribution, metabolism, excretion (), pharmacodynamic effects, and any observed adverse events, including dose-response relationships and species-specific differences. For biotechnology-derived products, additional and biodistribution data may be required. Gaps in data, such as incomplete exposure margins or unresolved safety signals, necessitate further studies before submission to avoid FDA holds. Sponsors typically perform an initial data review and against FDA IND requirements, evaluating existing results for completeness and conducting any bridging studies, such as 28-day repeat-dose in pivotal species if not already completed. Pre-IND meetings with FDA, requested via the Center for Drug Evaluation and Research (CDER) or Center for Biologics Evaluation and Research (CBER), are advisable to clarify data needs, discuss pivotal study designs, and resolve interpretive issues, with submissions ideally occurring 60 days in advance. The , submitted electronically via the Electronic Submissions Gateway, includes FDA Form 1571 (covering details), investigator statements, and an environmental assessment under the if the drug may alter the human environment. Following submission, FDA has 30 days to review for safety concerns; if no hold is placed, clinical trials may proceed, though amendments for new preclinical findings are required under 21 CFR 312.30.

Ethical Considerations and Necessity of Animal Testing

Principles of the 3Rs and Welfare Standards

The principles of the 3Rs—Replacement, Reduction, and Refinement—originated in the 1959 book The Principles of Humane Experimental Technique by William Russell and Rex Burch, providing a framework to minimize animal use in scientific research while preserving data quality. Replacement involves substituting animal models with non-animal alternatives, such as in vitro cell cultures, organoids, computational simulations, or physicochemical analyses, wherever scientifically valid for preclinical endpoints like initial toxicity screening. Reduction focuses on minimizing the number of animals required through optimized study design, including statistical power calculations, pilot studies, and sharing of control data across experiments, as applied in pharmaceutical pharmacokinetic assessments to avoid redundant dosing cohorts. Refinement entails minimizing pain, distress, and welfare impacts via techniques like analgesia, non-invasive imaging, enriched housing environments, and humane endpoints that terminate studies before severe suffering occurs, thereby enhancing animal well-being in toxicity evaluations. In preclinical drug development, the 3Rs are implemented through integrated strategies, such as leveraging modeling for dose prediction to reduce animal cohorts in studies and adopting for repeated physiological measurements without surgical invasion. These principles not only address ethical concerns but also improve research reproducibility by standardizing procedures and reducing variability from animal distress. Regulatory bodies, including the FDA and , endorse 3Rs adherence, requiring justification of animal use in submissions and encouraging alternatives where predictive equivalence is demonstrated, though full replacement remains limited by the need for systemic physiological data unobtainable from isolated models. Animal welfare standards complement the 3Rs by enforcing baseline protections, such as those outlined in the U.S. (amended) and the 8th Edition of the Guide for the Care and Use of Laboratory Animals (2011), which mandate veterinary oversight, species-appropriate caging, and in preclinical facilities. Institutional Animal Care and Use Committees (IACUCs) review protocols for 3Rs compliance, ensuring procedures like repeated blood sampling in pharmacodynamic studies incorporate refinements such as vascular access ports to limit handling stress. Accreditation by organizations like AAALAC International verifies adherence to these standards, with global harmonization via ICH guidelines promoting consistent welfare in multinational preclinical trials. Non-compliance risks regulatory delays, underscoring welfare as integral to valid preclinical outcomes.

Empirical Evidence for Predictive Value Despite Criticisms

A of 150 pharmaceutical compounds from 12 companies revealed that preclinical animal studies predicted with an overall concordance rate of 71%, demonstrating substantial empirical support for their negative predictive value in identifying safe candidates. Non-rodent species showed higher predictivity, detecting 63% of toxicities, compared to 43% for alone, underscoring the importance of multi-species testing in enhancing reliability. This concordance exceeds random chance and has informed regulatory decisions, as evidenced by the low rate of severe, unanticipated toxicities in early clinical phases for drugs advancing past rigorous preclinical safety evaluations. A systematic scoping review of 121 studies on animal-to-human further corroborated these findings, reporting a 71% concordance rate for across all considered, with non-rodents achieving 63% predictivity for outcomes. For adverse events, correlations between animal findings and human gastrointestinal, hepatic, and renal toxicities were more frequent than discrepancies, indicating that preclinical models effectively flag risks that manifest clinically. These data counter criticisms of negligible predictivity by quantifying how filters out compounds likely to cause harm, thereby reducing ethical and financial costs of proceeding to human trials with unsafe agents. While efficacy predictivity remains lower—often below 50% in broad analyses due to species-specific physiological differences—targeted preclinical models have succeeded in forecasting clinical responses in domains like , where patient-derived xenografts (PDX) retrospectively aligned with outcomes in approximately 90% of cases for cytotoxic and targeted therapies. Such successes highlight causal links between validated animal endpoints and human efficacy, particularly when models incorporate human-relevant biomarkers and dosing regimens. Regulatory bodies, including the FDA, continue to mandate these studies precisely because empirical evidence shows they mitigate risks more effectively than unproven alternatives, despite ongoing refinements toward integrated approaches.

Challenges, Limitations, and

Species Translation Failures and High Failure Rates

Species translation failures occur when pharmacological or toxicological effects observed in preclinical animal models do not replicate in s, primarily due to interspecies differences in , , receptor expression, and pathology. For instance, and non-human primates exhibit variations in enzyme profiles critical for , leading to discrepancies in clearance and metabolite formation that can result in false negatives for toxicity. These mismatches contribute significantly to the high rates in , where approximately 90-95% of candidates that advance past preclinical stages fail in clinical trials, with and issues accounting for the majority of terminations. Empirical data highlight the predictive limitations: animal models detect only about 50% of toxicities that emerge in clinical testing, while overpredicting others irrelevant to humans, such as certain rodent-specific carcinogens. In , for example, many anticancer agents effective in xenografts fail in trials due to differences in tumor microenvironments and immune responses, with II failure rates exceeding 70% for such translations. Similarly, in , preclinical success in animal or Alzheimer's models translates to efficacy in fewer than 10% of cases, attributed to species-specific anatomy and protein aggregation dynamics. These failures underscore causal disconnects, where animal physiology proxies inadequately capture causal pathways for progression and drug response. Notable case studies exemplify these pitfalls. TGN1412, a for autoimmune diseases, showed no adverse effects in cynomolgus monkeys but induced cytokine storms and multi-organ failure in a 2006 phase I human trial, necessitating for six participants due to human-specific T-cell activation absent in the primate model. Another instance involves fialuridine, an antiviral that progressed through without hepatotoxicity signals but caused fatal in human phase II trials in 1993, linked to differences in mitochondrial between . Such events, while rare, amplify scrutiny on reliance, with overall translation failure rates from animal efficacy data hovering around 86-92% across therapeutic areas. The cumulative effect manifests in pipeline attrition: from preclinical nomination, only about 10% of compounds reach market approval, with species-related predictive gaps exacerbating costs estimated at $2.6 billion per successful drug, including sunk investments in non-translating candidates. While some analyses attribute only 14% of preclinical failures to liver mismatches, broader discordance in and drives 30-40% of early clinical discontinuations. Addressing these requires integrating human-relevant earlier, though animal mandates persist under regulatory frameworks despite acknowledged limitations.

Cost, Time, and Resource Demands

Preclinical development imposes substantial demands on time, financial resources, and personnel, contributing significantly to the overall high attrition and expense of bringing new drugs to market. The duration of this phase, encompassing lead optimization, pharmacokinetics, pharmacodynamics, and toxicology studies required for Investigational New Drug (IND) submission, typically spans 1 to 3 years for IND-enabling activities, though the full process from target identification to candidate selection can extend to 4-7 years when including early discovery. These timelines are influenced by iterative testing cycles, regulatory feedback, and the need for comprehensive data generation under Good Laboratory Practice (GLP) standards, with delays often arising from unexpected toxicity findings necessitating additional studies. Financial costs for preclinical development vary widely by therapeutic area, molecule complexity, and decisions, but estimates place average outlays at $5 million to $100 million or more per candidate, primarily driven by GLP-compliant programs, development, and early scale-up. studies alone, including acute, subchronic, and chronic dosing in and non-rodents, can account for 40-60% of these expenses due to specialized protocols and analytical requirements. Capitalized costs, factoring in opportunity and failure risks across portfolios, amplify effective expenditures, as only a fraction of candidates advance, underscoring the phase's role in the broader $1-2 billion average per approved drug. Resource demands include multidisciplinary teams of pharmacologists, toxicologists, pathologists, and veterinarians, often numbering dozens per project, operating in GLP-certified facilities equipped for high-containment handling, analytical instrumentation, and maintenance. Animal usage constitutes a major input, with 10,000 to 20,000 specimens typically required per drug candidate for efficacy and safety profiling across species like mice, rats, dogs, and , necessitating dedicated and husbandry to meet and regulatory standards. These elements strain specialized infrastructure, with contract research organizations (CROs) frequently employed to mitigate in-house limitations, though coordination overhead further elevates demands.

Emerging Alternatives and Regulatory Shifts

Non-Animal Models: Organ-on-Chip, AI, and Human-Relevant Systems

Non-animal models in preclinical development encompass advanced and computational approaches designed to simulate human physiological responses more accurately than traditional , potentially reducing translation failures where up to 90% of drugs succeeding in animals fail in human trials. These systems prioritize human-derived cells, tissues, and data-driven predictions to assess , , and , addressing interspecies differences that contribute to high attrition rates. Organ-on-chip (OOC) devices, (AI) algorithms, and organoid-based human-relevant platforms represent key innovations, though their integration remains limited by gaps, scalability issues, and incomplete replication of systemic interactions. Organ-on-chip technology utilizes microfluidic chips to recapitulate organ-level functions, such as fluid flow, mechanical stresses, and cellular interactions, using human cells to model tissue microenvironments. Developed since the early 2010s, OOC systems have demonstrated utility in predicting drug-induced and , with studies showing higher concordance to human outcomes than animal models in specific endpoints like . For instance, lung-on-chip models have replicated cigarette smoke-induced inflammation and viral infections, enabling evaluation of therapeutics without animal use. Multi-organ chips linking liver, heart, and gut compartments further simulate , offering insights into compound that correlate better with clinical data in case studies. However, OOC lacks standardized protocols and full-body integration, including immune responses and vasculature, preventing it from fully supplanting at present. Artificial intelligence models leverage on vast datasets to forecast and , analyzing chemical structures, genomic profiles, and historical preclinical outcomes to identify risks earlier. In toxicity prediction, has achieved accuracies exceeding 80% for endpoints like and by integrating quantitative structure-activity relationship (QSAR) models with , outperforming some rule-based animal assays in retrospective validations. Tools such as those from Schrödinger combine physics simulations with to predict adverse drug reactions, contributing to reduced animal use in lead optimization phases. Despite these advances, 's reliance on training data introduces biases from incomplete or animal-centric datasets, and it struggles with novel mechanisms or long-term effects, necessitating hybrid approaches with experimental validation. Approximately 30% of preclinical candidates still fail due to unanticipated , underscoring 's role as a supportive rather than standalone tool. Human-relevant systems, including (iPSC)-derived organoids, provide three-dimensional, self-organizing structures that mimic organ architecture and function for personalized testing. Organoids from patient-specific cells have predicted drug responses in diseases like and cancer, with brain organoids revealing neurotoxicity patterns absent in rodent models. In drug safety assessments, kidney organoids have identified nephrotoxic compounds with 70-90% sensitivity to human clinical outcomes, surpassing two-dimensional cultures. These models enable while incorporating genetic variability, potentially lowering the 5% success rate of candidates from preclinical stages. Limitations persist, however, as organoids lack vascularization, innervation, and interactions, restricting their ability to capture whole-body dynamics or chronic exposures. Ongoing efforts focus on co-culturing with endothelial cells to enhance maturity, but regulatory acceptance requires prospective validation against clinical endpoints.

Recent FDA Initiatives (2024-2025) and Validation Hurdles

In April 2025, the U.S. (FDA) released a "Roadmap to Reducing Animal Testing in Preclinical Safety Studies," outlining a stepwise strategy over 3–5 years to reduce, refine, or replace animal use in drug safety assessments through New Approach Methodologies (NAMs), including computational modeling, human cell-based assays, and organs-on-chips. This initiative builds on the FDA Modernization Act 2.0 (enacted 2022), which permits for investigational new drug applications, and responds to congressional pressures, including the proposed FDA Modernization Act 3.0 introduced in February 2024 to accelerate implementation. The roadmap prioritizes high-impact areas like monoclonal antibodies and gene therapies, where animal data has shown limited translatability to human outcomes, aiming for case-by-case waivers supported by robust NAM evidence. Complementing this, the FDA launched its New Alternative Methods Program in July 2025 to facilitate regulatory adoption of NAMs by developing qualification pathways, fostering public-private collaborations, and integrating human-relevant data from and -driven predictions. Enhanced FDA-NIH partnerships, announced in mid-2025, emphasize tools and human-specific models to supplant animal extrapolations, with pilot programs testing for in preclinical phases. These efforts align with executive directives under the Trump administration to minimize across federal research, projecting potential reductions of 20–30% in mandatory studies by 2028 if validation benchmarks are met. Despite momentum, validation of NAMs faces significant hurdles, including the need for standardized protocols to ensure across labs, as current organ-on-chip systems vary in design and lack unified performance metrics for endpoints like or . validation—comparing NAM outputs to historical human clinical data—remains resource-intensive, with FDA requiring demonstration of predictive accuracy exceeding animal models' 50–70% in translation, yet few NAMs have accumulated sufficient longitudinal datasets for regulatory qualification. models, while accelerating hypothesis generation, encounter challenges in interpretability ("" limitations) and generalizability beyond training cohorts, necessitating approaches with mechanistic assays to address causal gaps unresolvable by correlation-based alone. Regulatory acceptance is further impeded by liability concerns and the absence of harmonized international standards; for instance, while FDA guidance on organs-on-chips is anticipated by late 2025, discrepancies with requirements could delay global submissions, increasing costs for bridging studies. Economic barriers persist, as developing validated platforms demands upfront investments estimated at $50–100 million per modality, deterring smaller biotech firms despite potential long-term savings from reduced . Critics, including some pharmacologists, argue that over-reliance on unproven NAMs risks underestimating rare toxicities, underscoring the FDA's emphasis on tiered evidence hierarchies where animal data serves as a benchmark until NAMs prove equivalent human predictivity.

References

  1. [1]
    Drug Development and Review Definitions - FDA
    Aug 20, 2015 · During preclinical drug development, a sponsor evaluates the drug's toxic and pharmacologic effects through in vitro and in vivo laboratory ...
  2. [2]
    The basics of preclinical drug development for neurodegenerative ...
    Jun 12, 2009 · Preclinical development encompasses the activities that link drug discovery in the laboratory to initiation of human clinical trials.
  3. [3]
    Step 2: Preclinical Research - FDA
    Jan 4, 2018 · After preclinical testing, researchers review their findings and decide whether the drug should be tested in people. Content current as of: 01/ ...
  4. [4]
    Investigational New Drug (IND) Application - FDA
    During a new drug's early preclinical development, the sponsor's primary goal is to determine if the product is reasonably safe for initial use in humans ...Investigator-Initiated INDs · IND Forms and Instructions · Guidances
  5. [5]
    [PDF] Guidance for Industry - S6 Preclinical Safety Evaluation of ... - FDA
    The objectives of the preclinical safety studies are to define pharmacological and toxicological effects not only prior to initiation of human studies but ...
  6. [6]
    Preclinical Testing Techniques: Paving the Way for New Oncology ...
    1. Introduction. Preclinical testing is a cornerstone of the drug development process and is responsible for preliminary studies of numerous compounds with the ...2. Spheroids · 4. Organ-On-A-Chip · Organoids For Preclinical...
  7. [7]
    General Principles of Preclinical Study Design - PMC - NIH
    Preclinical studies using animals to study the potential of a therapeutic drug or strategy are important steps before translation to clinical trials.
  8. [8]
    Why 90% of clinical drug development fails and how to improve it?
    If drug candidates in the preclinical stage are also counted, the failure rate of drug discovery/development is even higher than 90%. Analyses of clinical trial ...
  9. [9]
    Lost in translation: the valley of death across preclinical and clinical ...
    Nov 18, 2019 · According to the National Institutes of Health (NIH), 80 to 90% of research projects fail before they ever get tested in humans and for every ...<|control11|><|separator|>
  10. [10]
  11. [11]
    The Staggering Cost of Drug Development: A Look at the Numbers
    Aug 10, 2023 · Preclinical Phase: Costs here, primarily due to laboratory studies and animal testing, can range from $15 million to $100 million. · Clinical ...
  12. [12]
    Step 3: Clinical Research - FDA
    Jan 4, 2018 · Researchers design Phase 3 studies to demonstrate whether or not a product offers a treatment benefit to a specific population.
  13. [13]
    Preclinical Pharmacokinetics in Drug Development - Allucent
    The primary goal of the pre-clinical program is to support the estimation of a safe and effective dose range for testing in clinical studies. The toxicology ...<|separator|>
  14. [14]
    Principles of early drug discovery - PMC - PubMed Central
    This review will look at key preclinical stages of the drug discovery process, from initial target identification and validation, through assay development.
  15. [15]
    Successful Drug Development Despite Adverse Preclinical Findings ...
    Over 90% of withdrawals of marketed drugs are due to clinical toxicity, particularly hepatotoxicity and cardiovascular toxicity 40 – 43 , which underlines the ...Missing: metrics | Show results with:metrics
  16. [16]
    R&D Time and Success Rate | Knowledge Portal
    Success rates for MMV were estimated at: Preclinical: 50%, Phase I: 70%, Phase IIa: 78%, Phase IIb: 75%, Phase III: 67% and Registration: 100%. Other technology ...
  17. [17]
    In Vitro vs In Vivo Preclinical Studies - News-Medical
    Feb 23, 2021 · Before a drug candidate can be tested in humans, its safety and efficacy must be explored in in vitro or in vivo preclinical studies.
  18. [18]
    Advanced In Vitro Models for Preclinical Drug Safety - NIH
    Dec 26, 2024 · Cell-based in vitro models are widely used for screening and account for nearly half of HTS efforts, especially for orally available drugs.
  19. [19]
    Building a Tiered Approach to In Vitro Predictive Toxicity Screening
    The challenge is to incorporate methods for evaluating and understanding potential liabilities of NCEs into multiple phases of the drug discovery process.
  20. [20]
    [PDF] Preclinical Assessment of Investigational Cellular and Gene ... - FDA
    The resulting data from preclinical studies should address these objectives in order to guide the design of early-phase clinical trials, as well as establish a ...
  21. [21]
    Innovations in Drug Development: The Increased Utility of In Vitro ...
    Mar 7, 2024 · Faster drug development: In vitro testing has the potential to speed up research significantly and allow novel therapies to reach patients ...
  22. [22]
    In Vivo vs In Vitro: Definition, Pros and Cons | Technology Networks
    Dec 18, 2023 · In vitro models provide a starting point for researchers to gather insights into how a cell responds to a new drug in a controlled, isolated ...
  23. [23]
    Translatability of in vitro potency to clinical efficacious exposure - NIH
    May 12, 2023 · The present study shows that in vitro potency is predictive to estimate the therapeutic drug exposure to some extent, whereas the general trend of overexposure ...
  24. [24]
    Preclinical Predictors of Anticancer Drug Efficacy - NIH
    In vitro assays are also inadequate in the evaluation of the potential toxicity of a novel agent arising from unexpected off-target effects. Finally, many ...<|separator|>
  25. [25]
    A Guide to In Silico Drug Design - PMC - PubMed Central
    In this review, we present an overview of the important CADD methods and applications, such as in silico structure prediction, refinement, modelling and target ...
  26. [26]
    In silico models in drug development: where we are - ScienceDirect
    These in silico approaches are demonstrating their ability to generate reliable predictions as well as new knowledge on the mode of action of drugs and the ...
  27. [27]
    Model-Informed Drug Development: In Silico Assessment of ... - NIH
    This comprehensive review aims to explain the mechanisms that control the dissolution and/or release of drugs and their subsequent permeation through ...3. Drug Release Modeling · 5.1. Qspr/qsar Models · 5.2. Md Simulations
  28. [28]
    From Start to Phase 1 in 30 Months | Insilico Medicine
    From Start to Phase 1 in 30 Months: AI-discovered and AI-designed Anti-fibrotic Drug Enters Phase I Clinical Trial.
  29. [29]
    How successful are AI-discovered drugs in clinical trials? A first ...
    In Phase I we find AI-discovered molecules have an 80–90% success rate, substantially higher than historic industry averages.
  30. [30]
    Scientific and regulatory evaluation of mechanistic in silico drug and ...
    Nov 19, 2021 · This type of in silico model aims at studying a pathophysiological system by focusing on various possible biological scales. The scale may vary ...
  31. [31]
    Potential Benefits of In Silico Methods: A Promising Alternative in ...
    In silico methods not only facilitate drug development by closing gaps in experimental research but also enhance our comprehension of viral pathogenesis and ...1. Introduction · 6. Qsar And Admet In Natural... · 13. Conclusions
  32. [32]
    Limitations of In Silico Methods | proventainternational.com
    Nevertheless, in silico trials will be able to facilitate faster and more cost-effective risk assessments, reducing the total number of human participants. How ...
  33. [33]
    In silico methods for drug-target interaction prediction - ScienceDirect
    Sep 24, 2025 · In summary, the limitations of early in silico methods for DTI prediction, such as their dependency on 3D structural data, inadequacy in ...Feature Engineering · Conclusions And Future... · Strategies (insights)
  34. [34]
    Modernizing Preclinical Drug Development: The Role of New ...
    May 29, 2025 · Integrating computational and in vitro NAM approaches can optimize preclinical drug development, improving translational accuracy and reducing clinical trial ...Missing: pipeline | Show results with:pipeline
  35. [35]
    Preclinical Studies in Drug Development | PPD
    Once a lead drug candidate is identified, a typical preclinical development program consists of six major segments: Manufacture of drug substance / active ...
  36. [36]
    The Role of Mouse Models in Drug Discovery | Taconic Biosciences
    Mar 20, 2025 · Discover how mouse models drive drug discovery, from disease modeling to preclinical testing, enhancing research efficiency and accelerating ...Key Takeaways · Target Validation · Summary
  37. [37]
    The (misleading) role of animal models in drug development
    Apr 7, 2024 · This article explains for a general audience how animal research is used to develop new medicines, its benefits and limitations, and how more accurate and ...
  38. [38]
    Different Species of Animal Models in Preclinical Testing
    Feb 19, 2021 · While preclinical studies into cancer have utilized canine and primate participants, the mouse model is currently the most common animal model ...
  39. [39]
    6 Types of Toxicology Studies for IND & NDA Programs - WuXi AppTec
    May 2, 2024 · #1. General Toxicology Testing · #2. Carcinogenicity Testing · #3. Safety Pharmacology Testing · #4. DART Testing · #5. In Vitro Toxicology Testing.
  40. [40]
    Toxicology Studies - Pacific BioLabs
    Pre-clinical / Non-clinical In Vivo Toxicology ; In Vivo Toxicology Test Categories. Acute Systemic Toxicity (3 to 14 days); Subacute Toxicity (14 to 30 days) ...
  41. [41]
    Guide to NCL In Vivo Studies: Efficacy, Pharmacokinetics & Toxicology
    Each NCL in vivo study is tailored to the particular compound being tested, so each toxicity study is different, but in general we conduct single or repeat-dose ...Efficacy Studies · Toxicity Studies · Pharmacokinetic Studies
  42. [42]
    FDA Announces Plan to Phase Out Animal Testing Requirement for ...
    Apr 10, 2025 · The FDA's animal testing requirement will be reduced, refined, or potentially replaced using a range of approaches.
  43. [43]
    [PDF] Roadmap to Reducing Animal Testing in Preclinical Safety Studies
    Apr 10, 2025 · This roadmap outlines a strategic, stepwise approach for FDA to reduce animal testing in preclinical safety studies with scientifically ...
  44. [44]
    In Vitro and In Vivo Assessment of ADME and PK Properties During ...
    Sep 9, 2015 · We provide guidelines for ADME and PK assessments, benchmarks and practical “rules of thumb” for selecting compounds with sufficient PK to be viable ...
  45. [45]
    [PDF] M3(R2) Nonclinical Safety Studies for the Conduct of Human ... - FDA
    Harmonization of the guidance for nonclinical safety studies will help to define the current recommendations and reduce the likelihood that substantial ...
  46. [46]
    A Guide to In Vitro ADME Testing in Drug Development - WuXi AppTec
    Jun 23, 2022 · The in vitro testing of ADME at well-controlled conditions provides insights into the DMPK properties of a drug candidate. Obtaining this ...
  47. [47]
    [PDF] Pharmacodynamic principles and the time course of immediate drug ...
    Dec 15, 2017 · The two key parameters of pharmacodynamics are the maximum response (Emax) and the concentration producing 50% of Emax (C50).
  48. [48]
    Basic Concepts in Population Modeling, Simulation, and Modelâ ...
    Jan 2, 2014 · where Emax is the maximum effect a drug can elicit, EC50 is the concentration at half-maximal response, Cp is the con- centration of drug, and E ...
  49. [49]
    Implementation of pharmacokinetic and pharmacodynamic ... - NIH
    The purpose of this publication is to serve as a guide for drug discovery scientists toward optimal design and conduct of PK/PD studies in the research phase.
  50. [50]
    Pharmacokinetic/Pharmacodynamic Modeling for Drug ...
    Oct 29, 2018 · PK/PD modeling allows us to address a number of key questions at the various stages of the drug discovery and development process (i.e., PhAT).
  51. [51]
    How to design robust preclinical efficacy studies that make a difference
    Below are some fundamental steps to develop robust experimental designs when using mouse models of human diseases.<|separator|>
  52. [52]
    The successes and limitations of preclinical studies in predicting the ...
    To improve drug development outcomes, it is important to review when preclinical pharmacodynamic and safety models have successfully predicted human ...
  53. [53]
    Pharmacokinetic/Pharmacodynamic-Driven Drug Development - PMC
    This review will address past and current deficencies in how PK/PD studies are conducted and offer new strategies that might bridge the gap between preclinical ...
  54. [54]
    Safety Guidelines - ICH
    This document covers the preclinical safety testing requirements for biotechnological products. It addresses the use of animal models of disease, determination ...
  55. [55]
    Redbook 2000: IV.B.1. General Guidelines for Toxicity Studies - FDA
    Oct 26, 2017 · Guidelines for specific recommended toxicity studies are found in Chapter IV.C., including: genetic toxicity studies (Chapter IV.C.1.), acute ...
  56. [56]
    [PDF] Regulatory Toxicology and Pharmacology - FDA
    Apr 20, 2020 · The FDA follows the recommendations outlined in the ICH gui- dances for assessing genotoxic risk (ICH S2(R1), 2012) and overall cancer risk for ...
  57. [57]
    Preclinical Toxicology of Vaccines - PMC - PubMed Central - NIH
    In addition to toxicity studies, in vivo and in vitro assays play a significant role in assessing critical safety characteristics of vaccines. Testing ...
  58. [58]
    [PDF] Guidance for Industry - FDA
    Jul 6, 2005 · There are essentially three types of findings in nonclinical toxicology studies that can be used to determine the NOAEL: (1) overt toxicity (e.g ...
  59. [59]
    Practical Considerations in Determining Adversity and the No ...
    Mar 1, 2022 · Simply, the NOAEL is the highest dose at which there are no observed adverse effects. In practice, it is one dose level below the lowest dose at ...
  60. [60]
    [PDF] ICH guideline M3(R2) on non-clinical safety studies for the conduct ...
    determining the recommended starting dose in humans. In general, the No Observed Adverse Effect Level (NOAEL) determined in nonclinical safety studies.
  61. [61]
    The estimation and translation uncertainties in applying NOAEL to ...
    The objective of the work reported here was to assess, via simulation, the level of uncertainty in the NOAEL estimated from an animal species.
  62. [62]
    [PDF] Good Laboratory Practice (GLP) 101 – Regulations and Basic Studies
    www.fda.gov. 11. OECD Principles on GLP. • Developed in 1978 and revised in 1997. – FDA GLP Draft Rule (1976) provided the basis for OECD Principles on GLP. – ...
  63. [63]
    OECD Principles on Good Laboratory Practice
    The Principles of Good Laboratory Practice (GLP) have been developed to promote the quality and validity of test data used for determining the safety of ...
  64. [64]
    Nonclinical Laboratories Inspected under Good Laboratory Practices
    May 7, 2025 · The FDA conducts careful inspections of facilities that perform nonclinical laboratory studies to determine compliance with Part 58 (Good Laboratory Practice)
  65. [65]
    [PDF] Good Laboratory Practice Regulations - FDA
    SUPPLEMENTARY INFORMATION: FDA discussed the need for regulations on good laboratory practice for nonclinical laboratory studies (GLP's) in the preamble to the ...
  66. [66]
    [PDF] Good Laboratory Practice | OECD
    The OECD Principles of Good Laboratory. Practice (GLP) have been developed to promote the quality and validity of test data used for determining the safety of ...
  67. [67]
    Good Laboratory Practice and Compliance Monitoring - OECD
    The OECD Principles of Good Laboratory Practice (GLP) establish quality standards for the organisation and management of test facilities.
  68. [68]
    21 CFR Part 312 -- Investigational New Drug Application - eCFR
    This part contains procedures and requirements governing the use of investigational new drugs, including procedures and requirements for the submission to, and ...
  69. [69]
  70. [70]
    [PDF] IND content and format for Phase 1 studies - FDA
    CLARIFICATIONS OF PRESENT IND REGULATIONS. An IND submission for Phase 1 studies is required by regulation to contain the sections enumerated below ...<|separator|>
  71. [71]
    Preclinical Data Review And Preparation For IND Filing
    Aug 19, 2025 · Initial Data Review – Evaluate existing preclinical results against FDA IND requirements. · Gap Analysis – Identify missing studies or incomplete ...
  72. [72]
    Division of Anti-Infectives (DAI) Information for Pre-IND Submissions
    Jun 24, 2025 · Considerations for Preparing a Pre-IND Submission · Chemistry, Manufacturing and Controls · Pharmacology/Toxicology · Clinical Microbiology.
  73. [73]
    Prepare the Initial IND Submission | Clinical Research Resource HUB
    The initial IND submission should be accompanied by a cover letter, an IND Application Form - FDA 1571 (see instructions) and a Certification of Compliance - ...
  74. [74]
    The Three Rs - Norecopa
    William Russell and Rex Burch developed the concept of the 3Rs during the 1950s, and described them in their book The Principles of Humane Experimental ...
  75. [75]
    Strategies to apply 3Rs in preclinical testing - PMC - PubMed Central
    Oct 5, 2021 · The three Rs (Replacement, Reduction, and Refinement), seek to ensure the rational and respectful use of laboratory animals and maintain an ...Abstract · 1. Introduction · 3. Virtual Compound...
  76. [76]
    Russell and Burch's 3Rs Then and Now: The Need for Clarity in ...
    Reduction minimizes research animal distress by decreasing the number of animals that can experience distress. Refinement is, by definition, diminution or ...
  77. [77]
    Enhancing Research Quality with the 3Rs: Replace, Reduce, Refine
    Feb 7, 2025 · Designing animal studies with the 3Rs—Replace, Reduce, Refine—not only enhances animal welfare but also improves research quality and efficiency ...
  78. [78]
    [PDF] FDA, ICH, and the 3Rs
    Promotion of public health through international harmonization that contributes to: – Prevention of unnecessary duplication of clinical trials and.
  79. [79]
    Animal Use Alternatives (3Rs) | National Agricultural Library - USDA
    The 3Rs alternatives refers to the replacement, reduction, and refinement of animals used in research, teaching, testing, and exhibition.
  80. [80]
    Animal Welfare and IACUC - Pacific BioLabs
    Pacific BioLabs has zero tolerance for inhumane treatment of animals. We promote adherence to the three R's – Refine, Reduce, Replace – and generally reserve ...
  81. [81]
    Ethical use of animals in medicine testing
    These principles encourage alternatives to the use of animals in the testing of medicines while safeguarding scientific quality and improving animal welfare ...
  82. [82]
    Concordance of the toxicity of pharmaceuticals in humans ... - PubMed
    The main aim of this project was to examine the strengths and weaknesses of animal studies to predict human toxicity (HT).
  83. [83]
    Animal to human translation: a systematic scoping review of ...
    Jul 15, 2019 · The concordance rates between animal and human toxicity were 71% when all species were considered, with nonrodents alone predictive for 63% and ...
  84. [84]
    Preclinical Success to Clinical Failure: Do We Have a Model ...
    Mar 29, 2020 · In a retrospective analysis of cytotoxic and targeted therapies, PDX models were clearly predictive (~90% accurate) of clinical outcome when ...
  85. [85]
    Novel Technologies to Improve Predictivity of Non-clinical Studies
    Sep 6, 2022 · FDA is working to replace, reduce, and refine (the 3 Rs) dependence on animal studies by advancing development of, and evaluating new, fit-for-purpose non- ...
  86. [86]
    Poor Translatability of Biomedical Research Using Animals - PubMed
    Mar 7, 2023 · The failure rate for the translation of drugs from animal testing to human treatments remains at over 92%, where it has been for the past few decades.Missing: species | Show results with:species
  87. [87]
    Limitations of Animal Studies for Predicting Toxicity in Clinical Trials
    Nov 25, 2019 · Percentages of drugs that fail in preclinical trials (due to drug toxicity or failure of efficacy in animal testing) and in clinical trials (due ...
  88. [88]
    Limitations of Animal Studies for Predicting Toxicity in Clinical Trials
    Animal testing is used in pharmaceutical and industrial research to predict human toxicity, and yet analysis suggests that animal models are poor predictors of ...
  89. [89]
    Pre-clinical animal models are poor predictors of human toxicities in ...
    Sep 1, 2020 · Our objective was to determine the correlation between preclinical toxicity found in animal models (mouse, rat, dog and monkey) and clinical ...
  90. [90]
    [PDF] Examples of drugs which have failed after being considered ...
    Examples of drugs which have failed after being considered successful in animal tests. 1. In 2006, six men took a drug (known as TGN1412) in a clinical trial ...
  91. [91]
    Drugs That Work In Mice Often Fail When Tried In People - NPR
    Apr 10, 2017 · Medical Research Using Animals Often Fails To Produce Drugs That ... "Imagine you were doing a human drug trial and you said to the FDA ...
  92. [92]
    Why Does Drug Development Take So Long? | Eureka blog
    Oct 28, 2021 · It takes about 12-15 months to complete the preclinical safety tests that Sponsors need when determining whether to submit an Investigational ...
  93. [93]
    Drug development – ​​The four phases - Biostock
    Altogether, the discovery phase and the preclinical phase can take four to seven years. After completion of the preclinical tests, provided the results ...
  94. [94]
    R&D Costs | Knowledge Portal
    This estimation covers preclinical development ($4.9 million to $5.3 million), pharmaceutical development (at least $5.3 million), and Phases I through III of ...
  95. [95]
    Costs of Drug Development and Research and Development ...
    Jun 28, 2024 · Studies have estimated that the R&D cost for a new drug ranges from $314 million to $4.46 billion, depending on the therapeutic area, data, and ...
  96. [96]
    Research and Development in the Pharmaceutical Industry
    Apr 8, 2021 · In recent studies, estimates of the average R&D cost per new drug range from less than $1 billion to more than $2 billion per drug. Those ...
  97. [97]
    Organization & Personnel - FDA
    Apr 7, 2015 · (a) A testing facility shall have a quality assurance unit which shall be responsible for monitoring each study to assure management that the ...Missing: preclinical | Show results with:preclinical
  98. [98]
    New Approach Methodologies in Drug Development - ScienceDirect
    Sep 12, 2025 · Nearly 90% of drugs that pass animal tests fail in human trials. •. New Approach Methodologies (NAMs) offer human-relevant insights.<|separator|>
  99. [99]
    Organ-on-chip technology: Opportunities and challenges
    Organ-on-chip (OOC) technology is an innovative approach that reproduces human organ structures and functions on microfluidic platforms.
  100. [100]
    Organ-on-a-Chip: A new paradigm for drug development - PMC
    Mar 24, 2021 · This review discusses how the Organ-on-a-Chip technology can play critical roles at different preclinical stages of drug development and ...
  101. [101]
    Human organs-on-chips for disease modelling, drug development ...
    Mar 25, 2022 · Here, I review how single and multiple human organ chip systems have been used to model complex diseases and rare genetic disorders.
  102. [102]
    Review Organ-on-a-Chip: A New Paradigm for Drug Development
    In this review, we discuss how the Organ-on-a-Chip technology can have critical roles in different preclinical stages of drug development and highlight the ...
  103. [103]
    Human Organ-On-A-Chip: Technologies Offer Benefits Over Animal ...
    May 21, 2025 · Currently, OOCs cannot replace animal testing but may be used alongside animals. The next generation of OOC is focused on developing “body-on-a ...
  104. [104]
    Recent advances in AI-based toxicity prediction for drug discovery
    Today, AI models are capable of predicting wide range of toxicity endpoints, such as hepatotoxicity, cardiotoxicity, nephrotoxicity, neurotoxicity, and ...Abstract · Introduction · Endpoint-specific toxicity... · Emerging AI innovations in...
  105. [105]
    AI-driven drug discovery picks up as FDA pushes to reduce animal ...
    Sep 2, 2025 · New York-based Schrodinger combines physics-based simulations with AI to predict drug toxicology. But industry experts say in the near ...<|separator|>
  106. [106]
    Artificial intelligence in preclinical research: enhancing digital twins ...
    AI revolutionizes preclinical research, offering alternatives to animal testing. Techniques like ML, DL, OoC, and DTs enable precise drug safety simulations.
  107. [107]
    applications of artificial intelligence in ADMET and toxicity prediction
    Oct 6, 2025 · It has been reported that ~30% of preclinical candidate compounds (PCCs) fail due to toxicity issues, making adverse toxicological reactions the ...
  108. [108]
    Replacing Animal Testing with Stem Cell-Organoids - PubMed Central
    Apr 19, 2024 · The aim of the review is to focus on stem cell related methodologies, such as organoids, that can serve as an alternative to animal testing and discuss its ...
  109. [109]
    Mini organs offer alternative method for predicting drug safety and ...
    Organoids, a 'new approach methodology', could forecast how drug candidates perform and help developers make informed decisions.
  110. [110]
    Beyond animal models: how organoids could transform drug ...
    One of the clearest use cases for organoids is in cancer drug development, where only around 5% of drug candidates that pass preclinical testing show positive ...
  111. [111]
    Stem Cells and Organoids: A Paradigm Shift in Preclinical Models ...
    Jul 1, 2025 · By offering models that closely mimic native tissue physiology and pathology, organoids serve as a bridge between traditional cell culture and ...
  112. [112]
    Implementing Alternative Methods - FDA
    Jul 31, 2025 · FDA's New Alternative Methods Program is intending to spur the adoption of alternative methods for regulatory use that can replace, reduce, and refine animal ...
  113. [113]
    A Turning Point: FDA and NIH Commit to Reducing Animal Testing
    Jul 14, 2025 · The event builds on the recent release of the FDA's Roadmap to Reduce Animal Testing in Preclinical Safety Studies—a plan that signals a growing ...
  114. [114]
    FDA and NIH Initiatives Part of Move Away From Animal Testing
    Jun 3, 2025 · The Trump Administration is using executive actions, agency initiatives, and policy directives to reduce or eliminate animal testing in medical research.
  115. [115]
    FDA NAMs Roadmap: Economic and Scientific Impact on Preclinical ...
    Sep 9, 2025 · The FDA roadmap outlines specific validation requirements, including retrospective analyses comparing NAM predictions to known human outcomes, ...
  116. [116]
    Organ-On-A-Chip: Upcoming US FDA Guidance Could Help Flesh ...
    Jul 16, 2025 · “FDA's April 2025 roadmap signals a more proactive stance toward replacing and reducing animal testing, where appropriate, using alternatives ...Missing: preclinical | Show results with:preclinical
  117. [117]
    FDA Roadmap to Reducing Animal Testing: A New Regulatory Era
    Jul 8, 2025 · This initiative builds on the 2022 FDA Modernization Act 2.0, which legally removed the requirement for animal testing in IND and BLA ...
  118. [118]
    FDA Roadmap Accelerates High-Tech Alternatives to Animal Tests
    Apr 24, 2025 · Organs-on-chips could make drug testing more accurate, but hurdles remain. A 2024 Nature Communications perspective points to major barriers, ...Missing: preclinical | Show results with:preclinical