You have 3 free guides left 😟
Unlock your guides
You have 3 free guides left 😟
Unlock your guides

1.4 Toxicological endpoints

7 min readaugust 20, 2024

Toxicological endpoints are crucial measures used to assess how substances affect living organisms. They help determine the safety and risks of chemicals, drugs, and pollutants by evaluating various effects on health and biological systems.

These endpoints cover a wide range of potential impacts, from to long-term effects like cancer. They involve different types of studies, including in vitro and in vivo experiments, to provide a comprehensive understanding of a substance's toxicity profile.

Toxicological endpoints

  • Toxicological endpoints are measurable outcomes used to assess the potential adverse effects of a substance on living organisms
  • These endpoints help determine the safety and risk associated with exposure to various chemicals, drugs, or environmental pollutants
  • Different types of toxicological endpoints are used depending on the duration of exposure, the specific organ or system affected, and the mechanism of toxicity

Acute toxicity endpoints

Top images from around the web for Acute toxicity endpoints
Top images from around the web for Acute toxicity endpoints
  • Assess the immediate effects of a single or short-term exposure to a substance
  • Commonly measured endpoints include , which is the dose that causes death in 50% of the test animals
  • Other acute endpoints include signs of toxicity such as changes in behavior, body weight, or clinical chemistry parameters (blood glucose, liver enzymes)

Subchronic toxicity endpoints

  • Evaluate the effects of repeated exposure to a substance over a portion of the lifespan, typically 90 days in rodents
  • Endpoints include changes in body weight, organ weights, histopathology, and clinical chemistry parameters
  • Subchronic studies help identify target organs and establish dose levels for studies

Chronic toxicity endpoints

  • Assess the effects of long-term, repeated exposure to a substance over a significant portion of the lifespan (1-2 years in rodents)
  • Endpoints include changes in body weight, organ weights, histopathology, and tumor incidence
  • Chronic studies are used to establish the and the

In vitro toxicity endpoints

  • Measure the effects of a substance on isolated cells, tissues, or organs in a controlled laboratory setting
  • Common endpoints include cell viability, cytotoxicity, , and specific cellular functions (enzyme activity, receptor binding)
  • In vitro assays are often used for high-throughput screening and to investigate mechanisms of toxicity

In vivo toxicity endpoints

  • Assess the effects of a substance on whole, living organisms, typically animals such as rodents or non-human primates
  • Endpoints include changes in body weight, organ weights, histopathology, behavior, and clinical signs of toxicity
  • In vivo studies are required for regulatory approval of drugs and chemicals to ensure safety in humans

Carcinogenicity endpoints

  • Evaluate the potential of a substance to cause cancer after long-term exposure
  • Endpoints include tumor incidence, multiplicity, and time-to-tumor formation
  • studies are typically conducted in rodents over a significant portion of their lifespan (2 years in mice, 2-3 years in rats)

Genotoxicity endpoints

  • Assess the ability of a substance to damage DNA, which can lead to mutations and potentially cancer
  • Endpoints include DNA damage, chromosomal aberrations, and gene mutations
  • Genotoxicity assays can be conducted in vitro (bacterial reverse mutation assay, micronucleus test) or in vivo (rodent bone marrow micronucleus test)

Reproductive toxicity endpoints

  • Evaluate the effects of a substance on male and female reproductive function and fertility
  • Endpoints include changes in reproductive organ weights, histopathology, sperm parameters (count, motility, morphology), and fertility indices
  • studies are conducted in rodents over multiple generations to assess potential impacts on future offspring

Developmental toxicity endpoints

  • Assess the effects of a substance on embryonic and fetal development during pregnancy
  • Endpoints include malformations, variations, and developmental delays in the offspring
  • studies are typically conducted in pregnant rodents or rabbits during the critical period of organogenesis

Neurotoxicity endpoints

  • Evaluate the effects of a substance on the structure and function of the nervous system
  • Endpoints include changes in behavior, motor activity, sensory function, and neuropathology
  • studies can be conducted in adult animals or during critical periods of nervous system development (pre- and postnatal)

Immunotoxicity endpoints

  • Assess the effects of a substance on the immune system's ability to defend against pathogens and foreign substances
  • Endpoints include changes in immune organ weights (thymus, spleen), lymphocyte subpopulations, antibody production, and immune function tests
  • studies are often conducted in rodents exposed to the substance for 28 days or longer

Endocrine disruption endpoints

  • Evaluate the potential of a substance to interfere with the normal function of the endocrine system, which regulates hormones
  • Endpoints include changes in hormone levels, reproductive organ weights, and development of hormone-sensitive tissues (mammary glands, prostate)
  • studies may involve in vitro assays (receptor binding, gene expression) or in vivo studies in rodents or aquatic organisms

Organ-specific toxicity endpoints

  • Assess the effects of a substance on specific target organs, such as the liver, kidney, or heart
  • Endpoints include changes in organ weights, histopathology, and organ-specific biomarkers (liver enzymes, kidney function tests)
  • studies are often conducted as part of subchronic or chronic toxicity studies

Dose-response relationships

  • Describe the relationship between the dose of a substance and the magnitude of the observed toxic effect
  • Dose-response curves are used to determine the for toxicity and to establish safe exposure levels
  • The shape of the can provide insights into the mechanism of toxicity (linear, threshold, hormetic)

NOAEL vs LOAEL

  • The no-observed-adverse-effect level (NOAEL) is the highest dose of a substance that does not cause any detectable adverse effects
  • The lowest-observed-adverse-effect level (LOAEL) is the lowest dose of a substance that causes a detectable adverse effect
  • NOAEL and LOAEL are used to establish safe exposure levels for humans by applying uncertainty factors to account for interspecies and intraspecies differences

Benchmark dose modeling

  • A statistical approach to estimate the dose of a substance that causes a predetermined level of adverse response (benchmark response)
  • uses dose-response data to calculate the lower confidence limit of the dose that produces the benchmark response (BMDL)
  • BMDL is used as a point of departure for and setting

Toxicokinetic considerations

  • describes the absorption, distribution, metabolism, and excretion (ADME) of a substance in the body
  • Understanding toxicokinetics is crucial for determining the internal dose of a substance at the target site and the duration of exposure
  • Toxicokinetic parameters include absorption rate, bioavailability, plasma half-life, and clearance

Toxicodynamic considerations

  • describes the molecular and cellular events that occur after a substance reaches its target site
  • These events include receptor binding, enzyme inhibition, oxidative stress, and cellular damage
  • Understanding toxicodynamics helps elucidate the mechanism of toxicity and identify potential targets for intervention

Mechanistic vs apical endpoints

  • Mechanistic endpoints are early, subcellular events that precede overt toxicity, such as gene expression changes or protein modifications
  • Apical endpoints are observable, whole-organism outcomes, such as changes in body weight, organ toxicity, or mortality
  • Mechanistic endpoints can provide insights into the mode of action of a substance, while apical endpoints are used for risk assessment and regulatory decision-making

Biomarkers of toxicity

  • Measurable indicators of a toxic effect or exposure to a substance, such as changes in gene expression, protein levels, or metabolite profiles
  • Biomarkers can be used to detect early signs of toxicity, monitor disease progression, or assess the efficacy of interventions
  • Examples of biomarkers include liver enzymes (ALT, AST) for hepatotoxicity, kidney function tests (creatinine, BUN) for nephrotoxicity, and DNA adducts for genotoxicity

Adverse outcome pathways (AOPs)

  • Conceptual frameworks that describe the causal linkages between a molecular initiating event (MIE) and an adverse outcome (AO) at the individual or population level
  • AOPs integrate mechanistic and apical endpoints to provide a comprehensive understanding of the toxicity of a substance
  • AOPs can be used to guide the development of predictive toxicity testing strategies and support regulatory decision-making

Regulatory toxicity testing requirements

  • Toxicity testing requirements for chemicals, drugs, and other regulated substances are established by government agencies such as the US Environmental Protection Agency (EPA) and the Food and Drug Administration (FDA)
  • Testing requirements vary depending on the intended use of the substance, the potential for human exposure, and the expected toxicity profile
  • Common regulatory toxicity tests include acute, subchronic, and chronic toxicity studies, reproductive and developmental toxicity studies, and carcinogenicity studies

Alternative methods to animal testing

  • Non-animal methods that aim to reduce, refine, or replace the use of animals in toxicity testing
  • Examples include in vitro assays using human cells or tissues, in silico models based on structure-activity relationships, and read-across from similar substances
  • Alternative methods are increasingly being used to screen large numbers of substances, prioritize testing, and provide mechanistic insights

High-throughput screening assays

  • Automated, miniaturized assays that enable the rapid testing of large numbers of substances for potential toxicity
  • High-throughput assays often use in vitro models, such as human cell lines or primary cells, and measure endpoints such as cell viability, gene expression, or receptor binding
  • These assays are used to prioritize substances for further testing, identify potential mechanisms of toxicity, and support the development of predictive toxicity models

Computational toxicology approaches

  • The use of computer models and algorithms to predict the toxicity of substances based on their chemical structure, physicochemical properties, and biological activity
  • Approaches include quantitative structure-activity relationship (QSAR) models, pharmacophore modeling, and machine learning algorithms
  • Computational toxicology can help prioritize substances for testing, guide the design of safer chemicals, and support risk assessment and regulatory decision-making
© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.


© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.

© 2024 Fiveable Inc. All rights reserved.
AP® and SAT® are trademarks registered by the College Board, which is not affiliated with, and does not endorse this website.
Glossary
Glossary