Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
1.
Regul Toxicol Pharmacol ; 142: 105415, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37257751

ABSTRACT

Low levels of N-nitrosamines (NAs) were detected in pharmaceuticals and, as a result, health authorities (HAs) have published acceptable intakes (AIs) in pharmaceuticals to limit potential carcinogenic risk. The rationales behind the AIs have not been provided to understand the process for selecting a TD50 or read-across analog. In this manuscript we evaluated the toxicity data for eleven common NAs in a comprehensive and transparent process consistent with ICH M7. This evaluation included substances which had datasets that were robust, limited but sufficient, and substances with insufficient experimental animal carcinogenicity data. In the case of robust or limited but sufficient carcinogenicity information, AIs were calculated based on published or derived TD50s from the most sensitive organ site. In the case of insufficient carcinogenicity information, available carcinogenicity data and structure activity relationships (SARs) were applied to categorical-based AIs of 1500 ng/day, 150 ng/day or 18 ng/day; however additional data (such as biological or additional computational modelling) could inform an alternative AI. This approach advances the methodology used to derive AIs for NAs.


Subject(s)
Nitrosamines , Animals , Nitrosamines/toxicity , Carcinogens , Structure-Activity Relationship , Pharmaceutical Preparations
2.
Regul Toxicol Pharmacol ; 135: 105247, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35998738

ABSTRACT

Under ICH M7, impurities are assessed using the bacterial reverse mutation assay (i.e., Ames test) when predicted positive using in silico methodologies followed by expert review. N-Nitrosamines (NAs) have been of recent concern as impurities in pharmaceuticals, mainly because of their potential to be highly potent mutagenic carcinogens in rodent bioassays. The purpose of this analysis was to determine the sensitivity of the Ames assay to predict the carcinogenic outcome with curated proprietary Vitic (n = 131) and Leadscope (n = 70) databases. NAs were selected if they had corresponding rodent carcinogenicity assays. Overall, the sensitivity/specificity of the Ames assay was 93-97% and 55-86%, respectively. The sensitivity of the Ames assay was not significantly impacted by plate incorporation (84-89%) versus preincubation (82-89%). Sensitivity was not significantly different between use of rat and hamster liver induced S9 (80-93% versus 77-96%). The sensitivity of the Ames is high when using DMSO as a solvent (87-88%). Based on the analysis of these databases, the Ames assay conducted under OECD 471 guidelines is highly sensitive for detecting the carcinogenic hazards of NAs.


Subject(s)
Dimethyl Sulfoxide , Nitrosamines , Animals , Bacteria , Biological Assay , Carcinogens/toxicity , Cricetinae , Mutation , Nitrosamines/metabolism , Nitrosamines/toxicity , Pharmaceutical Preparations , Rats , Rodentia/metabolism , Solvents
3.
Regul Toxicol Pharmacol ; 122: 104895, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33610610

ABSTRACT

As per the ICH Q3A(R2) and Q3B(R2) regulatory guidelines, safety studies may be needed when an impurity in new drug substances or products is above the qualification threshold, and such qualification studies should be conducted in one nonclinical species for a duration of 14-90 days. However, the guidelines do not specify details about species selection, recommended study design, and the exact study duration that would support clinical use of a specific duration. This lack of guidance leads to ambiguity and sponsors have used various study designs to qualify impurities. In 2018, the European Medicines Agency provided a draft reflection paper encouraging the incorporation of 3Rs (Replacement, Reduction, and Refinement) principles for animal use into impurity qualification. As a response, the IQ DruSafe Impurity Working Group (WG) surveyed the IQ member companies to capture the current practices for impurity qualification, and evaluate study designs for a potential reduction in animal testing. This article summarizes the results and learnings from the survey. Additionally, the WG leveraged the survey learnings and provided harmonized study design considerations aimed towards achieving the study objectives, while supporting the 3Rs initiative in reducing the total number of animals used (up to 90%) for impurity qualification.


Subject(s)
Animal Use Alternatives/standards , Drug Contamination , Drug Industry/standards , European Union , Guidelines as Topic
4.
Regul Toxicol Pharmacol ; 123: 104926, 2021 Jul.
Article in English | MEDLINE | ID: mdl-33862169

ABSTRACT

The ICH M7(R1) guideline describes a framework to assess the carcinogenic risk of mutagenic and carcinogenic pharmaceutical impurities following less-than-lifetime (LTL) exposures. This LTL framework is important as many pharmaceuticals are not administered for a patient's lifetime and as clinical trials typically involve LTL exposures. While there has been regulatory caution about applying LTL concepts to cohort of concern (COC) impurities such as N-nitrosamines, ICH M7 does not preclude this and indeed literature data suggests that the LTL framework will be protective of patient safety for N-nitrosamines. The goal was to investigate if applying the LTL framework in ICH M7 would control exposure to an acceptable excess cancer risk in humans. Using N-nitrosodiethylamine as a case study, empirical data correlating exposure duration (as a percentage of lifespan) and cancer incidence in rodent bioassays indicate that the LTL acceptable intake (AI) as derived using the ICH M7 framework would not exceed a negligible additional risk of cancer. Therefore, controlling N-nitrosamines to an LTL AI based on the ICH M7 framework is thus demonstrated to be protective for potential carcinogenic risk to patients over the exposure durations typical of clinical trials and many prescribed medicines.


Subject(s)
Diethylnitrosamine/toxicity , Mutagens/toxicity , Carcinogens , Dose-Response Relationship, Drug , Humans , Mutagenesis , Nitrosamines/toxicity , Toxicity Tests
5.
Int J Toxicol ; 40(3): 285-298, 2021.
Article in English | MEDLINE | ID: mdl-33525949

ABSTRACT

A workshop entitled "Deriving Compound-Specific Exposure Limits for Chemicals Used in Pharmaceutical Synthesis" was held at the 2018 Genetic Toxicology Association annual meeting. The objectives of the workshop were to provide an educational forum and use case studies and live multiple-choice polling to establish the degree of similarity/diversity in approach/opinion of the industry experts and other delegates present for some of the more challenging decision points that need to be considered when developing a compound-specific exposure limit (ie, acceptable intake or permissible or permitted daily exposure). Herein we summarize the relevant background and case study information for each decision point topic presented as well as highlight significant polling responses and discussion points. A common observation throughout was the requirement for expert judgment to be applied at each of the decision points presented which often results in different reasoning being applied by the risk assessor when deriving a compound-specific exposure limit. This supports the value of precompetitive cross-industry collaborations to develop compound-specific limits and harmonize the methodology applied, thus reducing the associated uncertainty inherent in the application of isolated expert judgment in this context. An overview of relevant precompetitive cross-industry collaborations working to achieve this goal is described.


Subject(s)
Environmental Exposure/standards , Guidelines as Topic , Pharmaceutical Preparations/standards , Risk Assessment/standards , Toxicology/standards , Case-Control Studies , Decision Making , Humans
6.
Regul Toxicol Pharmacol ; 110: 104524, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31734179

ABSTRACT

Regulatory Guidance documents ICH Q3A (R2) and ICH Q3B (R2) state that "impurities that are also significant metabolites present in animal and/or human studies are generally considered qualified". However, no guidance is provided regarding data requirements for qualification, nor is a definition of the term "significant metabolite" provided. An opportunity is provided to define those categories and potentially avoid separate toxicity studies to qualify impurities. This can reduce cost, animal use and time, and avoid delays in drug development progression. If the concentration or amount of a metabolite, in animals or human, is similar to that of the known, structurally identical impurity (arising from the administered test material), the qualification of the impurity on the grounds of it also being a metabolite is justified. We propose two complementary approaches to support conclusions to this effect: 1) demonstrate that the impurity is formed by metabolism in animals and/or man, based preferably on plasma exposures or, alternatively, amounts excreted in urine, and, where appropriate, 2) show that animal exposure to (or amount of) the impurity/metabolite is equal or greater in animals than in humans. An important factor of both assessments is the maximum theoretical concentration (or amount) (MTC or MTA) of the impurity/metabolite achievable from the administered dose and recommendations on the estimation of the MTC and MTA are presented.


Subject(s)
Drug Contamination , Pharmaceutical Preparations/metabolism , Animals , Biotransformation , Humans , Toxicity Tests
7.
Regul Toxicol Pharmacol ; 79 Suppl 1: S48-56, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27233925

ABSTRACT

The Acceptable Daily Exposure (ADE) derived for pharmaceutical manufacturing is a health-based limit used to ensure that medicines produced in multi-product facilities are safe and are used to validate quality processes. Core to ADE derivation is selecting appropriate point(s) of departure (PoD), i.e., the starting dose of a given dataset that is used in the calculation of the ADE. Selecting the PoD involves (1) data collection and hazard characterization, (2) identification of "critical effects", and (3) a dose-response assessment including the determination of the no-observed-adverse-effect-level (NOAEL) or lowest-observed-adverse-effect-level (LOAEL), or calculating a benchmark dose (BMD) level. Compared to other classes of chemicals, active pharmaceutical ingredients (APIs) are well-characterized and have unique, rich datasets that must be considered when selecting the PoD. Dataset considerations for an API include therapeutic/pharmacological effects, particularities of APIs for different indications and routes of administration, data gaps during drug development, and sensitive subpopulations. Thus, the PoD analysis must be performed by a qualified toxicologist or other expert who also understands the complexities of pharmaceutical datasets. In addition, as the pharmaceutical industry continues to evolve new therapeutic principles, the science behind PoD selection must also evolve to ensure state-of-the-science practices and resulting ADEs.


Subject(s)
Drug Industry , No-Observed-Adverse-Effect Level , Occupational Exposure/prevention & control , Occupational Health , Pharmaceutical Preparations , Animals , Benchmarking , Dose-Response Relationship, Drug , Drug Industry/legislation & jurisprudence , Drug Industry/standards , Guidelines as Topic , Health Policy , Humans , Occupational Exposure/adverse effects , Occupational Exposure/legislation & jurisprudence , Occupational Exposure/standards , Occupational Health/legislation & jurisprudence , Occupational Health/standards , Pharmaceutical Preparations/classification , Pharmaceutical Preparations/standards , Pharmacokinetics , Policy Making , Risk Assessment , Toxicity Tests
8.
Regul Toxicol Pharmacol ; 79 Suppl 1: S28-38, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27233926

ABSTRACT

This manuscript discusses the different historical and more recent default approaches that have been used to derive an acceptable daily exposure (ADE). While it is preferable to derive a health-based ADE based on a complete nonclinical and clinical data package, this is not always possible. For instance, for drug candidates in early development there may be no or limited nonclinical or clinical trial data. Alternative approaches that can support decision making with less complete data packages represent a variety of methods that rely on default assumptions or data inputs where chemical-specific data on health effects are lacking. A variety of default approaches are used including those based on certain toxicity estimates, a fraction of the therapeutic dose, cleaning-based limits, the threshold of toxicological concern (TTC), and application of hazard banding tools such as occupational exposure banding (OEB). Each of these default approaches is discussed in this manuscript, including their derivation, application, strengths, and limitations. In order to ensure patient safety when faced with toxicological and clinical data-gaps, default ADE methods should be purposefully as or more protective than ADEs derived from full data packages. Reliance on the subset of default approaches (e.g., TTC or OEB) that are based on toxicological data is preferred over other methods for establishing ADEs in early development while toxicology and clinical data are still being collected.


Subject(s)
Drug Industry , No-Observed-Adverse-Effect Level , Occupational Exposure/prevention & control , Occupational Health , Pharmaceutical Preparations , Toxicity Tests/methods , Animals , Dose-Response Relationship, Drug , Drug Industry/history , Drug Industry/legislation & jurisprudence , Drug Industry/standards , Guidelines as Topic , Health Policy , History, 20th Century , History, 21st Century , Humans , Lethal Dose 50 , Occupational Exposure/adverse effects , Occupational Exposure/legislation & jurisprudence , Occupational Exposure/standards , Occupational Health/history , Occupational Health/legislation & jurisprudence , Occupational Health/standards , Pharmaceutical Preparations/classification , Pharmaceutical Preparations/history , Pharmaceutical Preparations/standards , Policy Making , Reproducibility of Results , Risk Assessment , Toxicity Tests/history , Toxicity Tests/standards
9.
Regul Toxicol Pharmacol ; 79 Suppl 1: S39-47, 2016 Aug.
Article in English | MEDLINE | ID: mdl-27267172

ABSTRACT

A European Union (EU) regulatory guideline came into effect for all new pharmaceutical products on June 1st, 2015, and for all existing pharmaceutical products on December 1st, 2015. This guideline centers around the use of the Acceptable Daily Exposure (ADE) [synonymous with the Permitted Daily Exposure (PDE)] and operational considerations associated with implementation are outlined here. The EU guidance states that all active pharmaceutical ingredients (API) require an ADE; however, other substances such as starting materials, process intermediates, and cleaning agents may benefit from an ADE. Problems in setting ADEs for these additional substances typically relate to toxicological data limitations precluding the ability to establish a formal ADE. Established methodologies such as occupational exposure limits or bands (OELs or OEBs) and the threshold of toxicological concern (TTC) can be used or adjusted for use as interim ADEs when only limited data are available and until a more formal ADE can be established. Once formal ADEs are derived, it is important that the documents are routinely updated and that these updates are communicated to appropriate stakeholders. Another key operational consideration related to data-poor substances includes the use of maximum daily dose (MDD) in setting cross-contamination limits. The MDD is an important part of the maximum allowable/safe concentration (MAC/MSC) calculation and there are important considerations for its use and definition. Finally, other considerations discussed include operational aspects of setting ADEs for pediatrics, considerations for large molecules, and risk management in shared facilities.


Subject(s)
Drug Industry , No-Observed-Adverse-Effect Level , Occupational Exposure/prevention & control , Occupational Health , Pharmaceutical Preparations , Animals , Dose-Response Relationship, Drug , Drug Industry/legislation & jurisprudence , Drug Industry/standards , Guidelines as Topic , Health Policy , Humans , Occupational Exposure/adverse effects , Occupational Exposure/legislation & jurisprudence , Occupational Exposure/standards , Occupational Health/legislation & jurisprudence , Occupational Health/standards , Pharmaceutical Preparations/classification , Pharmaceutical Preparations/standards , Policy Making , Risk Assessment , Toxicity Tests
10.
Regul Toxicol Pharmacol ; 72(3): 602-9, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26025210

ABSTRACT

Pharmaceutical companies develop specialized therapies to treat late stage cancer. In order to accelerate life-saving treatments and reduce animal testing, compounds to treat life-threatening malignancies are allowed modified requirements for preclinical toxicology testing. Limited data packages in early drug development can present product quality challenges at multi-product manufacturing facilities. The present analysis established an endpoint-specific threshold of toxicological concern (TTC) for developmental and reproductive toxicity (DART) for anticancer compounds. A comprehensive database was created consisting of over 300 no-observed adverse effect levels (NOAELs) for DART of 108 anticancer compounds. The 5th percentile NOAEL for developmental and reproductive toxicity was 0.005 mg/kg/day (300 µg/day), resulting in a human exposure threshold of 3 µg/day assuming standard uncertainty factors and a 60 kg human bodyweight. The analysis shows this threshold is protective for developmental and reproductive toxicity of highly potent groups of anticancer compounds. There were similar TTC values calculated for direct-acting and indirect-acting anticancer compounds.


Subject(s)
Antineoplastic Agents/toxicity , Risk Assessment/methods , Animals , Dose-Response Relationship, Drug , Embryonic Development/drug effects , Fetal Development/drug effects , Humans , Reproduction/drug effects , Toxicity Tests
11.
PDA J Pharm Sci Technol ; 78(3): 214-236, 2024 Jun 28.
Article in English | MEDLINE | ID: mdl-38942477

ABSTRACT

Leachables in pharmaceutical products may react with biomolecule active pharmaceutical ingredients (APIs), for example, monoclonal antibodies (mAb), peptides, and ribonucleic acids (RNA), potentially compromising product safety and efficacy or impacting quality attributes. This investigation explored a series of in silico models to screen extractables and leachables to assess their possible reactivity with biomolecules. These in silico models were applied to collections of known leachables to identify functional and structural chemical classes likely to be flagged by these in silico approaches. Flagged leachable functional classes included antimicrobials, colorants, and film-forming agents, whereas specific chemical classes included epoxides, acrylates, and quinones. In addition, a dataset of 22 leachables with experimental data indicating their interaction with insulin glargine was used to evaluate whether one or more in silico methods are fit-for-purpose as a preliminary screen for assessing this biomolecule reactivity. Analysis of the data showed that the sensitivity of an in silico screen using multiple methodologies was 80%-90% and the specificity was 58%-92%. A workflow supporting the use of in silico methods in this field is proposed based on both the results from this assessment and best practices in the field of computational modeling and quality risk management.


Subject(s)
Computer Simulation , Drug Contamination , Drug Contamination/prevention & control , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/analysis , Antibodies, Monoclonal/chemistry
12.
Regul Toxicol Pharmacol ; 65(1): 162-7, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22732128

ABSTRACT

In the manufacture of pharmaceuticals, if a multiproduct facility shares equipment amongst drug substances/products it is incumbent upon the manufacturer to demonstrate removal of the pharmaceutical through a robust cleaning validation/verification program. Removal must be to below limits considered acceptable from a quality and toxicological perspective. In order to address the toxicological concerns, an acceptable daily exposure (ADE) was developed which is the "dose that is unlikely to cause an adverse effect if...exposed, by any route...at or below this dose every day for a lifetime" (ISPE, 2010). For compounds in development, defaulted ADEs were proposed by Dolan et al. (2005) and adopted by the International Society of Pharmaceutical Engineers (ISPE) as conservative cutoffs for compounds with limited data. In Phase 1 clinical trials, exposure is typically short-term (single dose or repeated doses for ≤30 days) compared to the chronic doses used to derive ADE and defaulted ADEs. An analysis of publicly available databases for toxicological and pharmacological effects supports the use of 10-fold higher defaulted values when the residual drug substance is in a developmental pharmaceutical intended for Phase 1 clinical trials (exposure ≤30 days).


Subject(s)
Drug Industry/standards , Pharmaceutical Preparations/standards , Toxicology/methods , Clinical Trials, Phase I as Topic/methods , Databases, Factual , Drug Contamination/prevention & control , Drug Design , Drug Industry/instrumentation , Drug Industry/methods , Drug-Related Side Effects and Adverse Reactions , Equipment Contamination , Humans , International Agencies , Pharmaceutical Preparations/administration & dosage , Risk Assessment/methods
13.
Regul Toxicol Pharmacol ; 65(1): 157-61, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22921793

ABSTRACT

Cleaning validation programs are developed to demonstrate acceptable carryover of drug substances/products when multiple drug substances are manufactured in shared process equipment. The International Society of Pharmaceutical Engineers (ISPE) developed a guidance document in 2010 describing the Risk-Based Manufacture of Pharmaceutical Products (referred to as RiskMAPP) (ISPE, 2010). This guidance document developed the concept of an acceptable daily exposure (ADE), which is the toxicologically acceptable daily dose for the first drug substance used in processing drug equipment (DS(A)) without prior knowledge of the subsequent drug substance (DS(B)). This paper discusses an extension of the ADE methodology called the product-specific ADE (PSADE) which is derived when DS(B) is known. Four case studies demonstrate examples in which the PSADE can be scientifically supported in lieu of the ADE and highlight some limitations in its application. The PSADE approach can be used to justify higher acceptance limits for cleaning validation when the ADE based acceptance limits are below the process capability limit of the cleaning process or limit of quantitation of the analytical method.


Subject(s)
Drug Contamination/prevention & control , Drug Industry/standards , Pharmaceutical Preparations/standards , Animals , Drug Industry/instrumentation , Drug Industry/methods , Drug-Related Side Effects and Adverse Reactions , Equipment Contamination/prevention & control , Humans , International Agencies , Pharmaceutical Preparations/administration & dosage , Risk Assessment/methods , Toxicology/methods
14.
Article in English | MEDLINE | ID: mdl-37770142

ABSTRACT

N-Nitrosodiethylamine (NDEA), a well-studied N-nitrosamine, was tested in rats to compare the dose-response relationship of three genotoxicity endpoints. Mutant / mutation frequencies were determined using the transgenic rodent (TGR) gene mutation assay and error corrected next generation sequencing (ecNGS) (i.e., duplex sequencing (DS)), and genetic damage was detected by the alkaline comet assay. Big Blue® (cII Locus) animals (n = 6 per dose group) were administered doses of 0.001, 0.01, 0.1, 1, 3 mg/kg/day NDEA by oral gavage. Samples were collected for cII mutation and DS analyses following 28-days of exposure and 3 days recovery. In a separate study, male Sprague-Dawley (SD) rats (n = 6 per dose group) were administered the same doses by oral gavage for two consecutive days and then samples collected for the alkaline comet assay. A dose-related increase in mutant / mutation frequencies of the liver but not duodenum was observed using the TGR assay and DS with DS resulting in a slightly more sensitive response, with a lower benchmark dose (BMD). In addition, a dose-related increase in percent tail DNA was observed in the liver using the alkaline comet assay. Therefore, DS and comet assays showed good utility for hazard identification and dose-response analysis of a representative N-nitrosamine comparable to the TGR gene mutation assay.


Subject(s)
Diethylnitrosamine , Nitrosamines , Rats , Animals , Male , Comet Assay/methods , Diethylnitrosamine/toxicity , Rodentia , Rats, Sprague-Dawley , Mutation , Animals, Genetically Modified , DNA Damage , High-Throughput Nucleotide Sequencing , Mutagenicity Tests/methods , Dose-Response Relationship, Drug
15.
PDA J Pharm Sci Technol ; 76(5): 369-383, 2022.
Article in English | MEDLINE | ID: mdl-35031541

ABSTRACT

The threshold of toxicological concern (TTC), i.e., the dose of a compound lacking sufficient experimental toxicity data that is unlikely to result in an adverse health effect in humans, is important for evaluating extractables and leachables (E&Ls) as it guides analytical testing and minimizes the use of animal studies. The Extractables and Leachables Safety Information Exchange (ELSIE) consortium, which consists of member companies that span biotechnology, pharmaceutical, and medical device industries, brought together subject matter expert toxicologists to derive TTC values for organic, non-mutagenic E&L substances when administered parenterally. A total of 488 E&L compounds from the ELSIE database were analyzed and parenteral point of departure (PPOD) estimates were derived for 252 compounds. The PPOD estimates were adjusted to extrapolate to subacute, subchronic, and chronic durations of nonclinical exposure and the lower fifth percentiles were calculated. An additional 100-fold adjustment factor to account for nonclinical species and human variability was subsequently applied to derive the parenteral TTC values for E&Ls. The resulting parenteral TTC values are 35, 110, and 180 µg/day for human exposures of >10 years to lifetime, >1-10 years, and ≤1 year, respectively. These parenteral TTCs are expected to be conservative for E&Ls that are considered non-mutagenic per ICH M7(R1) guidelines.


Subject(s)
Biotechnology , Parenteral Nutrition , Animals , Humans , Pharmaceutical Preparations
16.
Crit Rev Toxicol ; 41(6): 507-44, 2011 Jul.
Article in English | MEDLINE | ID: mdl-21591905

ABSTRACT

Quantitative methods for estimation of cancer risk have been developed for daily, lifetime human exposures. There are a variety of studies or methodologies available to address less-than-lifetime exposures. However, a common framework for evaluating risk from less-than-lifetime exposures (including short-term and/or intermittent exposures) does not exist, which could result in inconsistencies in risk assessment practice. To address this risk assessment need, a committee of the International Life Sciences Institute (ILSI) Health and Environmental Sciences Institute conducted a multisector workshop in late 2009 to discuss available literature, different methodologies, and a proposed framework. The proposed framework provides a decision tree and guidance for cancer risk assessments for less-than-lifetime exposures based on current knowledge of mode of action and dose-response. Available data from rodent studies and epidemiological studies involving less-than-lifetime exposures are considered, in addition to statistical approaches described in the literature for evaluating the impact of changing the dose rate and exposure duration for exposure to carcinogens. The decision tree also provides for scenarios in which an assumption of potential carcinogenicity is appropriate (e.g., based on structural alerts or genotoxicity data), but bioassay or other data are lacking from which a chemical-specific cancer potency can be determined. This paper presents an overview of the rationale for the workshop, reviews historical background, describes the proposed framework for assessing less-than-lifetime exposures to potential human carcinogens, and suggests next steps.


Subject(s)
Carcinogens/toxicity , Environmental Exposure/standards , Mutagens/toxicity , Biological Assay/methods , Carcinogens/administration & dosage , Databases, Factual , Decision Trees , Dose-Response Relationship, Drug , Endpoint Determination , Food Contamination/analysis , Guidelines as Topic , Household Products/adverse effects , Humans , Mutagens/administration & dosage , National Institute of Environmental Health Sciences (U.S.) , Neoplasms/chemically induced , Pesticides/adverse effects , Risk Assessment , Time Factors , United States , United States Environmental Protection Agency , United States Food and Drug Administration
17.
Regul Toxicol Pharmacol ; 58(3): 369-81, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20801182

ABSTRACT

In order to determine a threshold for nongenotoxic carcinogens, the traditional risk assessment approach has been to identify a mode of action (MOA) with a nonlinear dose-response. The dose-response for one or more key event(s) linked to the MOA for carcinogenicity allows a point of departure (POD) to be selected from the most sensitive effect dose or no-effect dose. However, this can be challenging because multiple MOAs and key events may exist for carcinogenicity and oftentimes extensive research is required to elucidate the MOA. In the present study, a microarray analysis was conducted to determine if a POD could be identified following short-term oral rat exposure with two nongenotoxic rodent carcinogens, fenofibrate and methapyrilene, using a benchmark dose analysis of genes aggregated in Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways and Gene Ontology (GO) biological processes, which likely encompass key event(s) for carcinogenicity. The gene expression response for fenofibrate given to rats for 2days was consistent with its MOA and known key events linked to PPARα activation. The temporal response from daily dosing with methapyrilene demonstrated biological complexity with waves of pathways/biological processes occurring over 1, 3, and 7days; nonetheless, the benchmark dose values were consistent over time. When comparing the dose-response of toxicogenomic data to tumorigenesis or precursor events, the toxicogenomics POD was slightly below any effect level. Our results suggest that toxicogenomic analysis using short-term studies can be used to identify a threshold for nongenotoxic carcinogens based on evaluation of potential key event(s) which then can be used within a risk assessment framework.


Subject(s)
Carcinogens/toxicity , Fenofibrate/toxicity , Methapyrilene/analysis , Methapyrilene/toxicity , Neoplasms/chemically induced , Toxicogenetics/methods , Animals , Carcinogens/administration & dosage , Carcinoma, Hepatocellular/chemically induced , Carcinoma, Hepatocellular/genetics , Dose-Response Relationship, Drug , Female , Fenofibrate/administration & dosage , Gene Expression , Liver Neoplasms/chemically induced , Liver Neoplasms/genetics , Methapyrilene/administration & dosage , Neoplasms/genetics , No-Observed-Adverse-Effect Level , Oligonucleotide Array Sequence Analysis , Rats , Risk Assessment
18.
Regul Toxicol Pharmacol ; 57(2-3): 300-6, 2010.
Article in English | MEDLINE | ID: mdl-20363275

ABSTRACT

The current risk assessment approach for addressing the safety of very small concentrations of genotoxic impurities (GTIs) in drug substances is the threshold of toxicological concern (TTC). The TTC is based on several conservative assumptions because of the uncertainty associated with deriving an excess cancer risk when no carcinogenicity data are available for the impurity. It is a default approach derived from a distribution of carcinogens and does not take into account the properties of a specific chemical. The purpose of the study was to use in silico tools to predict the cancer potency (TD(50)) of a compound based on its structure. Structure activity relationship (SAR) models (classification/regression) were developed from the carcinogenicity potency database using MultiCASE and VISDOM. The MultiCASE classification models allowed the prediction of carcinogenic potency class, while the VISDOM regression models predicted a numerical TD(50). A step-wise approach is proposed to calculate predicted numerical TD(50) values for compounds categorized as not potent. This approach for non-potent compounds can be used to establish safe levels greater than the TTC for GTIs in a drug substance.


Subject(s)
Drug Contamination , Models, Theoretical , Mutagens/toxicity , Neoplasms/chemically induced , Pharmaceutical Preparations , Animals , Databases, Factual , Forecasting , Mice , Pharmaceutical Preparations/chemistry , Pharmaceutical Preparations/classification , Pharmaceutical Preparations/standards , Rats , Risk Assessment , Software , Structure-Activity Relationship
19.
Chem Res Toxicol ; 22(2): 348-56, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19170655

ABSTRACT

The role of metabolism in genotoxicity and carcinogenicity of many chemicals is well established. Accordingly, both in vitro metabolic activation systems and in vivo assays are routinely utilized for genotoxic hazard identification of drug candidates prior to clinical investigations. This should, in most cases provide a high degree of confidence that the genotoxic potential of the parent and associated metabolites have been characterized. However, it is well known that significant differences can exist between human metabolism and that which occurs with in vitro and in vivo genotoxicity tests. This poses challenges when considering the adequacy of hazard identification and cancer risk assessment if a human metabolite of genotoxic concern is identified during the course of drug development. Since such challenges are particularly problematic when recognized in the later stages of drug development, a framework for conducting a carcinogenic risk assessment for human genotoxic metabolites is desirable. Here, we propose a risk assessment method that is dependent upon the availability of quantitative human and rodent ADME (absorption, distribution, metabolism, excretion) data, such that exposures to a metabolite of genotoxic concern can be estimated at the intended human efficacious dose and the maximum dose used in the 2-year rodent bioassay(s). The exposures are then applied to the risk assessment framework, based on known cancer potencies, that allows one to understand the probability of a known or suspect genotoxic metabolite posing a carcinogenic risk in excess of 1 in 100,000. Practical case examples are presented to illustrate the application of the risk assessment method within the context of drug development and to highlight its utility and limitations.


Subject(s)
Mutagenicity Tests/methods , Mutagens/metabolism , Mutagens/toxicity , Animals , Body Burden , Carcinogens/metabolism , Carcinogens/toxicity , Humans , Metabolic Networks and Pathways , Rats , Risk Assessment
20.
Int J Toxicol ; 28(6): 468-78, 2009.
Article in English | MEDLINE | ID: mdl-19966139

ABSTRACT

This symposium focuses on the management of genotoxic impurities in the synthesis of pharmaceuticals. Recent developments in both Europe and United States require sponsors of new drug applications to develop processes to control the risks of potential genotoxic impurities. Genotoxic impurities represent a special case relative to the International Conference on Harmonisation Q3A/Q3B guidances, because genotoxicity tests used to qualify the drug substance may not be sufficient to demonstrate safety of a potentially genotoxic impurity. The default risk management approach for a genotoxic impurity is the threshold of toxicological concern unless a more specific risk characterization is appropriate. The symposium includes descriptions of industry examples where impurities are introduced and managed in the synthesis of a pharmaceutical. It includes recent regulatory developments such as the "staged threshold of toxicological concern" when administration is of short duration (eg, during clinical trials).


Subject(s)
Drug Contamination , Drug Discovery , Mutagens/toxicity , Pharmaceutical Preparations/chemical synthesis , Animals , Carcinogens/toxicity , Chemistry, Pharmaceutical , DNA/drug effects , DNA/genetics , DNA Damage , Dose-Response Relationship, Drug , Ethyl Methanesulfonate/toxicity , European Union , Humans , Legislation, Drug , Pharmaceutical Preparations/analysis , Pharmaceutical Preparations/chemistry , Risk Assessment , United States
SELECTION OF CITATIONS
SEARCH DETAIL