首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 78 毫秒
1.
Two assumptions used in risk assessment are investigated: (1) the assumption of fraction of lifetime dose rate assumes that the risk from a fractional lifetime exposure at a given dose rate is equal to the risk from full lifetime exposure at that same fraction of the given dose rate; (2) the assumption of fraction of lifetime risk assumes that the risk from a fractional lifetime exposure at a given dose rate is equal to that same fraction of the risk from full lifetime exposure at the same dose rate. These two assumptions are equivalent when risk is a linear function of dose. Thus both can be thought of as generalizations of the assumption that cancer risk is proportional to the total accumulated lifetime dose (or average daily dose), which is often made to assess the risk from short-term exposures. In this paper, the age-specific cumulative hazard functions are derived using the two-stage model developed by Moolgavkar, Venzon, and Knudson for situations when the exposure occurs during a single period or a single instant. The two assumptions described above are examined for three types of carcinogens, initiator, completer, and promoter, in the context of the model. For initiator and completer, these two assumptions are equivalent in the low-dose region; for a promoter, using the fraction of lifetime risk assumption is generally more conservative than that of the fraction of lifetime dose rate assumption. Tables are constructed to show that the use of either the fraction of lifetime dose rate assumption or the fraction lifetime risk assumption can both underestimate and overestimate the true risk for the three types of carcinogens.  相似文献   

2.
The paper applies classical statistical principles to yield new tools for risk assessment and makes new use of epidemiological data for human risk assessment. An extensive clinical and epidemiological study of workers engaged in the manufacturing and formulation of aldrin and dieldrin provides occupational hygiene and biological monitoring data on individual exposures over the years of employment and provides unusually accurate measures of individual lifetime average daily doses. In the cancer dose-response modeling, each worker is treated as a separate experimental unit with his own unique dose. Maximum likelihood estimates of added cancer risk are calculated for multistage, multistage-Weibull, and proportional hazards models. Distributional characterizations of added cancer risk are based on bootstrap and relative likelihood techniques. The cancer mortality data on these male workers suggest that low-dose exposures to aldrin and dieldrin do not significantly increase human cancer risk and may even decrease the human hazard rate for all types of cancer combined at low doses (e.g., 1 g/kg/day). The apparent hormetic effect in the best fitting dose-response models for this data set is statistically significant. The decrease in cancer risk at low doses of aldrin and dieldrin is in sharp contrast to the U.S. Environmental Protection Agency's upper bound on cancer potency based on mouse liver tumors. The EPA's upper bound implies that lifetime average daily doses of 0.0000625 and 0.00625 g/kg body weight/day would correspond to increased cancer risks of 0.000001 and 0.0001, respectively. However, the best estimate from the Pernis epidemiological data is that there is no increase in cancer risk in these workers at these doses or even at doses as large as 2 g/kg/day.  相似文献   

3.
Applications of methods for carcinogenic risk assessment often focus on estimating lifetime cancer risk. With intermittent or time-dependent exposures, lifetime risk is often approximated on the basis of a lifetime average daily dose (LADD). In this article, we show that there exists a lifetime equivalent constant dose (LECD) which leads to the same lifetime risk as the actual time-dependent exposure pattern. The ratio C = LECD/LADD then provides a measure of accuracy of risk estimates based on the LADD, as well as a basis for correcting such estimates. Theoretical results derived under the classical multistage model and the two-stage birth-death-mutation model suggest that the maximum value of C, which represents the factor by which the LADD may lead to underestimates of risk, will often lie in the range of 2- to 5-fold. The practical application of these results is illustrated in the case of astronauts subjected to relatively short-term exposure to volatile organics in a closed space station environment, and in the case of the ingestion of pesticide residues in food where consumption patterns vary with age.  相似文献   

4.
There has been considerable discussion regarding the conservativeness of low-dose cancer risk estimates based upon linear extrapolation from upper confidence limits. Various groups have expressed a need for best (point) estimates of cancer risk in order to improve risk/benefit decisions. Point estimates of carcinogenic potency obtained from maximum likelihood estimates of low-dose slope may be highly unstable, being sensitive both to the choice of the dose–response model and possibly to minimal perturbations of the data. For carcinogens that augment background carcinogenic processes and/or for mutagenic carcinogens, at low doses the tumor incidence versus target tissue dose is expected to be linear. Pharmacokinetic data may be needed to identify and adjust for exposure-dose nonlinearities. Based on the assumption that the dose response is linear over low doses, a stable point estimate for low-dose cancer risk is proposed. Since various models give similar estimates of risk down to levels of 1%, a stable estimate of the low-dose cancer slope is provided by ŝ = 0.01/ED01, where ED01 is the dose corresponding to an excess cancer risk of 1%. Thus, low-dose estimates of cancer risk are obtained by, risk = ŝ × dose. The proposed procedure is similar to one which has been utilized in the past by the Center for Food Safety and Applied Nutrition, Food and Drug Administration. The upper confidence limit, s , corresponding to this point estimate of low-dose slope is similar to the upper limit, q 1 obtained from the generalized multistage model. The advantage of the proposed procedure is that ŝ provides stable estimates of low-dose carcinogenic potency, which are not unduly influenced by small perturbations of the tumor incidence rates, unlike 1.  相似文献   

5.
A cancer risk assessment methodology based upon the Armitage–Doll multistage model of cancer is applied to animal bioassay data. The method utilizes the exact time-dependent dose pattern used in a bioassay rather than some single measure of dose such as average dose rate or cumulative dose. The methodology can be used to predict risks from arbitrary exposure patterns including, for example, intermittent exposure and short-term exposure occurring at an arbitrary age. The methodology is illustrated by applying it to a National Cancer Institute bioassay of ethylene dibromide in which dose rates were modified several times during the course of the experiment.  相似文献   

6.
Many models of exposure-related carcinogenesis, including traditional linearized multistage models and more recent two-stage clonal expansion (TSCE) models, belong to a family of models in which cells progress between successive stages-possibly undergoing proliferation at some stages-at rates that may depend (usually linearly) on biologically effective doses. Biologically effective doses, in turn, may depend nonlinearly on administered doses, due to PBPK nonlinearities. This article provides an exact mathematical analysis of the expected number of cells in the last ("malignant") stage of such a "multistage clonal expansion" (MSCE) model as a function of dose rate and age. The solution displays symmetries such that several distinct sets of parameter values provide identical fits to all epidemiological data, make identical predictions about the effects on risk of changes in exposure levels or timing, and yet make significantly different predictions about the effects on risk of changes in the composition of exposure that affect the pharmacodynamic dose-response relation. Several different predictions for the effects of such an intervention (such as reducing carcinogenic constituents of an exposure) that acts on only one or a few stages of the carcinogenic process may be equally consistent with all preintervention epidemiological data. This is an example of nonunique identifiability of model parameters and predictions from data. The new results on nonunique model identifiability presented here show that the effects of an intervention on changing age-specific cancer risks in an MSCE model can be either large or small, but that which is the case cannot be predicted from preintervention epidemiological data and knowledge of biological effects of the intervention alone. Rather, biological data that identify which rate parameters hold for which specific stages are required to obtain unambiguous predictions. From epidemiological data alone, only a set of equally likely alternative predictions can be made for the effects on risk of such interventions.  相似文献   

7.
A Monte Carlo simulation is incorporated into a risk assessment for trichloroethylene (TCE) using physiologically-based pharmacokinetic (PBPK) modeling coupled with the linearized multistage model to derive human carcinogenic risk extrapolations. The Monte Carlo technique incorporates physiological parameter variability to produce a statistically derived range of risk estimates which quantifies specific uncertainties associated with PBPK risk assessment approaches. Both inhalation and ingestion exposure routes are addressed. Simulated exposure scenarios were consistent with those used by the Environmental Protection Agency (EPA) in their TCE risk assessment. Mean values of physiological parameters were gathered from the literature for both mice (carcinogenic bioassay subjects) and for humans. Realistic physiological value distributions were assumed using existing data on variability. Mouse cancer bioassay data were correlated to total TCE metabolized and area-under-the-curve (blood concentration) trichloroacetic acid (TCA) as determined by a mouse PBPK model. These internal dose metrics were used in a linearized multistage model analysis to determine dose metric values corresponding to 10-6 lifetime excess cancer risk. Using a human PBPK model, these metabolized doses were then extrapolated to equivalent human exposures (inhalation and ingestion). The Monte Carlo iterations with varying mouse and human physiological parameters produced a range of human exposure concentrations producing a 10-6 risk.  相似文献   

8.
Experimental animal studies often serve as the basis for predicting risk of adverse responses in humans exposed to occupational hazards. A statistical model is applied to exposure-response data and this fitted model may be used to obtain estimates of the exposure associated with a specified level of adverse response. Unfortunately, a number of different statistical models are candidates for fitting the data and may result in wide ranging estimates of risk. Bayesian model averaging (BMA) offers a strategy for addressing uncertainty in the selection of statistical models when generating risk estimates. This strategy is illustrated with two examples: applying the multistage model to cancer responses and a second example where different quantal models are fit to kidney lesion data. BMA provides excess risk estimates or benchmark dose estimates that reflects model uncertainty.  相似文献   

9.
Previous applications of carcinogenic risk assessment using mathematical models of carcinogenesis have focused largely on the case where the level of exposure remains constant over time. In many situations, however, the dose of the carcinogen varies with time. In this paper, we discuss both the classical Armitage-Doll multistage model and the Moolgavkar-Venzon-Knudson two-stage birth-death-mutation model with time-dependent dosing regimens. Bounds on the degree of underestimation of risk that can occur through the use of a simple time-weighted average dose are derived by means of comparison with an equivalent constant dose corresponding to the actual risk under the time-dependent dosing regimen.  相似文献   

10.
Quantitative Cancer Risk Estimation for Formaldehyde   总被引:2,自引:0,他引:2  
Of primary concern are irreversible effects, such as cancer induction, that formaldehyde exposure could have on human health. Dose-response data from human exposure situations would provide the most solid foundation for risk assessment, avoiding problematic extrapolations from the health effects seen in nonhuman species. However, epidemiologic studies of human formaldehyde exposure have provided little definitive information regarding dose-response. Reliance must consequently be placed on laboratory animal evidence. An impressive array of data points to significantly nonlinear relationships between rodent tumor incidence and administered dose, and between target tissue dose and administered dose (the latter for both rodents and Rhesus monkeys) following exposure to formaldehyde by inhalation. Disproportionately less formaldehyde binds covalently to the DNA of nasal respiratory epithelium at low than at high airborne concentrations. Use of this internal measure of delivered dose in analyses of rodent bioassay nasal tumor response yields multistage model estimates of low-dose risk, both point and upper bound, that are lower than equivalent estimates based upon airborne formaldehyde concentration. In addition, risk estimates obtained for Rhesus monkeys appear at least 10-fold lower than corresponding estimates for identically exposed Fischer-344 rats.  相似文献   

11.
Dale Hattis 《Risk analysis》1990,10(2):303-316
Neither experimental animal exposures nor real-life human exposures are delivered at a constant level over a full lifetime. Although there are strong theoretical reasons why all pharmacokinetic processes must "go linear" at the limit of low dose rates, fluctuations in dose rate may produce nonlinearities that either increase or decrease actual risks relative to what would be expected for constant lifetime exposure. This paper discusses quantitative theory and specific examples for a number of processes that can be expected to give rise to pharmacokinetic nonlinearities at high dose rates–including transport processes (e.g., renal tubular secretion), activating and detoxifying metabolism, DNA repair, and enhancement of cell replication following gross toxicity in target tissues. At the extreme, full saturation of a detoxification or DNA repair process has the potential to create as much as a dose2 dependence of risk on dose delivered in a single burst, and if more than one detoxification step becomes fully saturated, this can be compounded. Effects via changes in cell replication rates, which appear likely to be largely responsible for the steep upward turning curve of formaldehyde carcinogenesis in rats, can be even more profound over a relatively narrow range of dosage. General suggestions are made for experimental methods to detect nonlinearities arising from the various sources in premarket screening programs.  相似文献   

12.
Reassessing Benzene Cancer Risks Using Internal Doses   总被引:1,自引:0,他引:1  
Human cancer risks from benzene exposure have previously been estimated by regulatory agencies based primarily on epidemiological data, with supporting evidence provided by animal bioassay data. This paper reexamines the animal-based risk assessments for benzene using physiologically-based pharmacokinetic (PBPK) models of benzene metabolism in animals and humans. It demonstrates that internal doses (interpreted as total benzene metabolites formed) from oral gavage experiments in mice are well predicted by a PBPK model developed by Travis et al. Both the data and the model outputs can also be accurately described by the simple nonlinear regression model total metabolites = 76.4x/(80.75 + x), where x = administered dose in mg/kg/day. Thus, PBPK modeling validates the use of such nonlinear regression models, previously used by Bailer and Hoel. An important finding is that refitting the linearized multistage (LMS) model family to internal doses and observed responses changes the maximum-likelihood estimate (MLE) dose-response curve for mice from linear-quadratic to cubic, leading to low-dose risk estimates smaller than in previous risk assessments. This is consistent with the conclusion for mice from the Bailer and Hoel analysis. An innovation in this paper is estimation of internal doses for humans based on a PBPK model (and the regression model approximating it) rather than on interspecies dose conversions. Estimates of human risks at low doses are reduced by the use of internal dose estimates when the estimates are obtained from a PBPK model, in contrast to Bailer and Hoel's findings based on interspecies dose conversion. Sensitivity analyses and comparisons with epidemiological data and risk models suggest that our finding of a nonlinear MLE dose-response curve at low doses is robust to changes in assumptions and more consistent with epidemiological data than earlier risk models.  相似文献   

13.
In light of the Armitage-Doll multistage carcinogenesis theory, this paper examines the assumption that an additive relative risk relationship is indicative of two carcinogens that affect the same stage in the cancer process. We present formulas to compute excess cancer risks for a variety of patterns for limited exposure durations to two carcinogens that affect the first and penultimate stages; and using an index of synergy proposed by Thomas (1982), we find a number of these patterns to produce additive, or nearly additive, relative risk relationships. The consistent feature of these patterns is that the two exposure periods are of short duration and occur close together.  相似文献   

14.
The traditional multistage (MS) model of carcinogenesis implies several empirically testable properties for dose-response functions. These include convex (linear or upward-curving) cumulative hazards as a function of dose; symmetric effects on lifetime tumor probability of transition rates at different stages; cumulative hazard functions that increase without bound as stage-specific transition rates increase without bound; and identical tumor probabilities for individuals with identical parameters and exposures. However, for at least some chemicals, cumulative hazards are not convex functions of dose. This paper shows that none of these predicted properties is implied by the mechanistic assumptions of the MS model itself. Instead, they arise from the simplifying "rare-tumor" approximations made in the usual mathematical analysis of the model. An alternative exact probabilistic analysis of the MS model with only two stages is presented, both for the usual case where a carcinogen acts on both stages simultaneously, and also for idealized initiation-promotion experiments in which one stage at a time is affected. The exact two-stage model successfully fits bioassay data for chemicals (e.g., 1,3-butadiene) with concave cumulative hazard functions that are not well-described by the traditional MS model. Qualitative properties of the exact two-stage model are described and illustrated by least-squares fits to several real datasets. The major contribution is to show that properties of the traditional MS model family that appear to be inconsistent with empirical data for some chemicals can be explained easily if an exact, rather than an approximate model, is used. This suggests that it may be worth using the exact model in cases where tumor rates are not negligible (e.g., in which they exceed 10%). This includes the majority of bioassay experiments currently being performed.  相似文献   

15.
In recent years physiologically based pharmacokinetic models have come to play an increasingly important role in risk assessment for carcinogens. The hope is that they can help open the black box between external exposure and carcinogenic effects to experimental observations, and improve both high-dose to low-dose and interspecies projections of risk. However, to date, there have been only relatively preliminary efforts to assess the uncertainties in current modeling results. In this paper we compare the physiologically based pharmacokinetic models (and model predictions of risk-related overall metabolism) that have been produced by seven different sets of authors for perchloroethylene (tetrachloroethylene). The most striking conclusion from the data is that most of the differences in risk-related model predictions are attributable to the choice of the data sets used for calibrating the metabolic parameters. Second, it is clear that the bottom-line differences among the model predictions are appreciable. Overall, the ratios of low-dose human to bioassay rodent metabolism spanned a 30-fold range for the six available human/rat comparisons, and the seven predicted ratios of low-dose human to bioassay mouse metabolism spanned a 13-fold range. (The greater range for the rat/human comparison is attributable to a structural assumption by one author group of competing linear and saturable pathways, and their conclusion that the dangerous saturable pathway constitutes a minor fraction of metabolism in rats.) It is clear that there are a number of opportunities for modelers to make different choices of model structure, interpretive assumptions, and calibrating data in the process of constructing pharmacokinetic models for use in estimating "delivered" or "biologically effective" dose for carcinogenesis risk assessments. We believe that in presenting the results of such modeling studies, it is important for researchers to explore the results of alternative, reasonably likely approaches for interpreting the available data--and either show that any conclusions they make are relatively insensitive to particular interpretive choices, or to acknowledge the differences in conclusions that would result from plausible alternative views of the world.  相似文献   

16.
Uncertainty in Cancer Risk Estimates   总被引:1,自引:0,他引:1  
Several existing databases compiled by Gold et al.(1–3) for carcinogenesis bioassays are examined to obtain estimates of the reproducibility of cancer rates across experiments, strains, and rodent species. A measure of carcinogenic potency is given by the TD50 (daily dose that causes a tumor type in 50% of the exposed animals that otherwise would not develop the tumor in a standard lifetime). The lognormal distribution can be used to model the uncertainty of the estimates of potency (TD50) and the ratio of TD50's between two species. For near-replicate bioassays, approximately 95% of the TD50's are estimated to be within a factor of 4 of the mean. Between strains, about 95% of the TD50's are estimated to be within a factor of 11 of their mean, and the pure genetic component of variability is accounted for by a factor of 6.8. Between rats and mice, about 95% of the TD50's are estimated to be within a factor of 32 of the mean, while between humans and experimental animals the factor is 110 for 20 chemicals reported by Allen et al.(4) The common practice of basing cancer risk estimates on the most sensitive rodent species-strain-sex and using interspecies dose scaling based on body surface area appears to overestimate cancer rates for these 20 human carcinogens by about one order of magnitude on the average. Hence, for chemicals where the dose-response is nearly linear below experimental doses, cancer risk estimates based on animal data are not necessarily conservative and may range from a factor of 10 too low for human carcinogens up to a factor of 1000 too high for approximately 95% of the chemicals tested to date. These limits may need to be modified for specific chemicals where additional mechanistic or pharmacokinetic information may suggest alterations or where particularly sensitive subpopu-lations may be exposed. Supralinearity could lead to anticonservative estimates of cancer risk. Underestimating cancer risk by a specific factor has a much larger impact on the actual number of cancer cases than overestimates of smaller risks by the same factor. This paper does not address the uncertainties in high to low dose extrapolation. If the dose-response is sufficiently nonlinear at low doses to produce cancer risks near zero, then low-dose risk estimates based on linear extrapolation are likely to overestimate risk and the limits of uncertainty cannot be established.  相似文献   

17.
Vinyl chloride (VC) was used as a propellant in a limited percentage of aerosol hairspray products in the United States from approximately 1967 to 1973. The question has arisen whether occupational exposures of hairdressers to VC-containing hairsprays in hair salons were sufficient to increase the risk for developing hepatic angiosarcoma (HAS). Transient two-zone and steady-state three-zone models were used to estimate the historical airborne concentration of VC for individual hairdressers using hairspray as well as estimated contributions from other hairdressers in the same salon. Concentrations of VC were modeled for small, medium, and large salons, as well as a representative home salon. Model inputs were determined using published literature, and variability in these inputs was also considered using Monte Carlo techniques. The 95th percentile for the daily time-weighted average exposure for small, medium, and large salons, assuming a market-share fraction of VC-containing hairspray use from the Monte Carlo analysis, was about 0.3 ppm, and for the home salon scenario was 0.1 ppm. The 95th percentile value for the cumulative lifetime exposure of the hairdressers was 2.8 ppm-years for the home salon scenario and 2.0 ppm-years for the small, medium, and large salon scenarios. If using the assumption that all hairsprays used in a salon contained VC, the 95th percentile of the theoretical lifetime cumulative dose was estimated to be 52–79 ppm-years. Estimated lifetime doses were all below the threshold dose for HAS of about 300 to 500 ppm-years reported in the published epidemiology literature.  相似文献   

18.
U.S. Environment Protection Agency benchmark doses for dichotomous cancer responses are often estimated using a multistage model based on a monotonic dose‐response assumption. To account for model uncertainty in the estimation process, several model averaging methods have been proposed for risk assessment. In this article, we extend the usual parameter space in the multistage model for monotonicity to allow for the possibility of a hormetic dose‐response relationship. Bayesian model averaging is used to estimate the benchmark dose and to provide posterior probabilities for monotonicity versus hormesis. Simulation studies show that the newly proposed method provides robust point and interval estimation of a benchmark dose in the presence or absence of hormesis. We also apply the method to two data sets on carcinogenic response of rats to 2,3,7,8‐tetrachlorodibenzo‐p‐dioxin.  相似文献   

19.
Increased cell proliferation increases the opportunity for transformations of normal cells to malignant cells via intermediate cells. Nongenotoxic cytotoxic carcinogens that increase cell proliferation rates to replace necrotic cells are likely to have a threshold dose for cytotoxicity below which necrosis and hence, carcinogenesis do not occur. Thus, low dose cancer risk estimates based upon nonthreshold, linear extrapolation are inappropriate for this situation. However, a threshold dose is questionable if a nongenotoxic carcinogen acts via a cell receptor. Also, a nongenotoxic carcinogen that increases the cell proliferation rate, via the cell division rate and/or cell removal rate by apoptosis, by augmenting an existing endogenous mechanism is not likely to have a threshold dose. Whether or not a threshold dose exists for nongenotoxic carcinogens, it is of interest to study the relationship between lifetime tumor incidence and the cell proliferation rate. The Moolgavkar–Venzon–Knudson biologically based stochastic two-stage clonal expansion model is used to describe a carcinogenic process. Because the variability in cell proliferation rates among animals often makes it impossible to detect changes of less than 20% in the rate, it is shown that small changes in the cell proliferation rate, that may be obscured by the background noise in rates, can produce large changes in the lifetime tumor incidence as calculated from the Moolgavkar–Venzon–Knudson model. That is, dose response curves for cell proliferation and tumor incidence do not necessarily mimic each other. This makes the use of no observed effect levels (NOELs) for cell proliferation rates often inadmissible for establishing acceptable daily intakes (ADIs) of nongenotoxic carcinogens. In those cases where low dose linearity is not likely, a potential alternative to a NOEL is a benchmark dose corresponding to a small increase in the cell proliferation rate, e. g., 1%, to which appropriate safety (uncertainty) factors can be applied to arrive at an ADI.  相似文献   

20.
Life cycle assessment (LCA) is a framework for comparing products according to their total estimated environmental impact, summed over all chemical emissions and activities associated with a product at all stages in its life cycle (from raw material acquisition, manufacturing, use, to final disposal). For each chemical involved, the exposure associated with the mass released into the environment, integrated over time and space, is multiplied by a toxicological measure to estimate the likelihood of effects and their potential consequences. In this article, we explore the use of quantitative methods drawn from conventional single-chemical regulatory risk assessments to create a procedure for the estimation of the cancer effect measure in the impact phase of LCA. The approach is based on the maximum likelihood estimate of the effect dose inducing a 10% response over background, ED10, and default linear low-dose extrapolation using the slope betaED10 (0.1/ED10). The calculated effects may correspond to residual risks below current regulatory compliance requirements that occur over multiple generations and at multiple locations; but at the very least they represent a "using up" of some portion of the human population's ability to accommodate emissions. Preliminary comparisons are performed with existing measures, such as the U.S. Environmental Protection Agency's (U.S. EPA's) slope factor measure q1*. By analyzing bioassay data for 44 chemicals drawn from the EPA's Integrated Risk Information System (IRIS) database, we explore estimating ED10 from more readily available information such as the median tumor dose rate TD50 and the median single lethal dose LD50. Based on the TD50, we then estimate the ED10 for more than 600 chemicals. Differences in potential consequences, or severity, are addressed by combining betaED10 with the measure disability adjusted life years per affected person, DALYp. Most of the variation among chemicals for cancer effects is found to be due to differences in the slope factors (betaED10) ranging from 10(-4) up to 10(4) (risk of cancer/mg/kg-day).  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号