National Academies Press: OpenBook
« Previous: 4 Hazard Identification
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

5

Susceptibility Factors

Several factors are known to influence susceptibility to the adverse effects of arsenic. This chapter addresses susceptibility factors discussed in the committee’s workshop by Beck (2013) and Hansen (2013)—life stage, genetics, nutrition, and pre-existing disease—and the additional factors of sex, smoking, alcohol consumption, and exposure to mixtures. The sections on these factors reflect a preliminary survey of the literature that the US Environmental Protection Agency (EPA) should consider more comprehensively and systematically as it conducts its toxicologic assessment of inorganic arsenic.

LIFE STAGES

Life stages refers to different ages and developmental stages throughout life, including prenatal development (pregnancy) and aging. There is increasing evidence that arsenic exposure early in life affects fetal and child health and development and health later in life. However, there seems to be a lack of data on the influence of aging.

Early-life development is a critical window of susceptibility for multiple toxic agents. The main focus has been on developmental neurotoxicity—recently reviewed by Bellinger (2013)—but there is increasing evidence that other organs and functions may be particularly susceptible during development, such as the immune system (Dietert 2011) and the lungs (Ramsey et al. 2013a). Thus, it is essential to evaluate whether early-life exposure to arsenic may affect the risk of the numerous arsenic-related health effects observed in adults and to consider this question in the health risk assessment.

Concerning prenatal arsenic exposure, it seems clear that all metabolites of inorganic arsenic easily cross the placenta to the fetus. Strong correlations between arsenic in maternal blood and in cord blood have been found in women exposed to arsenic-contaminated drinking water (Concha et al. 1998a; Jin et al. 2006; Hall et al. 2007). Epidemiologic studies, including prospective cohort studies, have provided evidence that arsenic exposure through drinking water during pregnancy may cause dose-dependent impairment of fetal and infant growth and survival (see section “Pregnancy Outcomes” in Chapter 4). In particular, the developing immune system and central nervous system seem to be susceptible to arsenic exposure, and adverse effects seem to appear even after relatively low arsenic exposure (see sections “Neurotoxicity” and “Immune Effects” in Chapter 4).

Besides the prenatal and perinatal stages, early childhood may be a period of susceptibility to inorganic arsenic exposure. A few epidemiologic studies have indicated that continued exposure after birth may impair children’s growth. A cross-sectional study in China suggested that increased water arsenic concentrations were inversely associated with body weight of children (720) 8–12 years old (S.X.Wang et al. 2007). In a prospective cohort study in Bangladesh, measures of 2,372 infants were related to concentrations of arsenic metabolites in the urine of mothers in early and late pregnancy and of the children at the age of 18 months (Saha et al. 2012). The study adjusted for age (within each age group), sex, maternal body-mass index, socioeconomic status, and birth weight or length. Observed inverse associations of maternal urinary arsenic with children's weight and length at the age of 3–24 months were markedly attenuated after adjustment for relevant covariates. However, the associations of urinary arsenic at the age of 18 months with weight and length at the age of 18–24 months were more robust, particularly in girls. The

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

effects seemed to appear after low arsenic exposure. Compared with girls in the first quintile of urinary arsenic (less than 16 μg/L, adjusted for specific gravity), those in the fourth quintile (26–46 μg/L) were almost 300 g lighter and 0.7 cm shorter. In a followup of 1,505 mother–infant pairs on whom there were data on concentrations of arsenic, cadmium, and lead in maternal and child urine, the effects of the combined exposure to these metals on children’s weights and heights up to the age of 5 years were evaluated (Gardner et al. 2013). The investigators adjusted for family socioeconomic status, maternal tobacco chewing during pregnancy, cooking with indoor fires, maternal education, season of birth, parity, and exposure to cadmium and lead. In the longitudinal analysis, the multivariable-adjusted attributable difference in children’s weight at the age of 5 years was -0.33 kg (95% CI -0.60 to -0.06) for high arsenic exposure (95th percentile or higher) compared with the lowest exposure (5th percentile or lower). The corresponding multivariable-adjusted attributable difference in height was -0.50 cm (95% CI -1.20 to 0.21). As in the earlier study, the associations were apparent primarily in girls

Those findings are supported by experimental studies in which mice were exposed to very low concentrations of arsenic (10 μg/L in drinking water) prenatally via the dams’ drinking water or postnatally (Kozul-Horvath et al. 2012). Birth outcomes, including litter weight and number of pups, were unaffected, but exposure in utero and postnatally resulted in impaired growth of the offspring. The growth deficits resolved afer cessation of exposure in male mice but not in female mice up to the age of 6 weeks.

Adverse effects of arsenic on child health, especially increased risk of infectious diseases and signs of immunosuppression, have been reported in several cross-sectional epidemiologic studies (see section “Immune Effects” in Chapter 4). Whether the underlying effect was initiated before birth or during childhood is not clear. Similarly, impaired cognitive function has been reported in several epidemiologic studies and indicates that children, especially of preschool age, are susceptible to arsenic-induced neurotoxicity (see section “Neurotoxicity” in Chapter 4).

The increasing evidence that early-life exposure to inorganic arsenic increases the risk of adverse health effects later in life was discussed in the committee’s workshop by Waalkes (2013). Important evidence comes from the studies in northern Chile, showing considerably increased risk of cancer and noncancer effects in young adults who were born in Antofagasta during or shortly before the period (1958–1970) of high arsenic concentrations in drinking water (Smith et al. 2006; Yuan et al. 2007; Dauphine et al. 2011; Smith et al. 2012). In particular, the risk of death from chronic bronchiectasis was higher in those who had been exposed in utero and during early childhood and higher in those who had been exposed in utero compared with the rest of Chile (Smith et al. 2006) (see section “Respiratory Effects” above). The increased susceptibility to respiratory effects of early-life exposure has support from experimental studies that indicated impairment of lung development caused by in utero exposure to arsenic (Ramsey et al. 2013a,b). Recent epidemiologic studies of arsenic-related developmental immunotoxicity have also indicated long-term consequences for susceptibility, for example, to infections (Moore et al. 2009; Ahmed et al. 2012).

The human evidence of later-life effects of low-dose early-life arsenic exposure is supported by a series of experimental studies in mice that were given drinking water that contained arsenic at 50 μg/L (mainly arsenate) during gestation (or before mating) and thereafter until weaning. That exposure regimen resulted in marked dysregulation of the hypothalamic–pituitary–adrenal axis in male mice at the age of 35 days (Goggin et al. 2012), interactions with the glucocorticoid receptor and associated signaling pathway (Martinez et al. 2008; Martinez-Finley et al. 2011), and impaired learning and memory (Martinez-Finley et al. 2009).

Perhaps the most striking animal studies are those of Waalkes and co-workers, who demonstrated that arsenic exposure in utero, with no additional postnatal exposure, leads to the development of liver, lung, ovary, and adrenal tumors in rats (Waalkes et al. 2003). They have gone on to show that prenatal arsenic exposure increases the carcinogenicity of other agents in the offspring when they reach adulthood (Tokar et al. 2011a).

A few recent studies have indicated that maternal arsenic exposure during pregnancy may influence epigenetic markers, specifically those related to DNA methylation, in DNA isolated from the blood cells of newborn children. Two studies in Bangladesh of 113 and 101 mother–newborn pairs reported positive

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

associations between maternal arsenic exposure and global methylation (methyl-incorporation or LINE-1 assays) in DNA isolated from cord blood (Kile et al. 2012; Pilsner et al. 2012), whereas no significant difference in in cord blood LINE-1 methylation was observed between arsenic-exposed (55) and -unexposed (16) newborns (as assessed by arsenic in hair and nails) in a cross-sectional study in Thailand (Intarasunanont et al. 2012). The study by Pilsner et al. (2012), which also included the Alu and LUMA assays, indicated that the effects on DNA methylation were largely sex-specific, with arsenic-related increased methylation in newborn boys but decreased methylation in girls. Evaluation of methylation in cord-blood DNA of 134 infants in a prospective birth cohort in New Hampshire using the Illumina Infinium Methylation 450K array found evidence of differential patterns of DNA methylation in relation to fairly low maternal urinary arsenic concentrations in late pregnancy (median 4.1 μg/L, interquartile range 1.8–6.6 μg/L, maximum value about 300 μg/L) (Koestler et al. 2013). Among the 100 loci that had the strongest association with arsenic, those in CpG promoter islands (44) showed mostly (75%) higher methylation in the highest-exposed group (>6.6 μg/L) than in the lowest-exposed group. However, cg08884395 (associated with estrogen receptor 1, ESR1) and cg27514608 (peroxisome proliferator–activated receptor-γ coactivator 1-α [PPARGC1A]) showed inverse associations between arsenic exposure and methylation and had statistically significant trends. Although changes in DNA methylation have been observed and associated with arsenic exposure, their biologic effects and meaning at a functional level in the cell and as related to health effects are unknown.

Key Considerations for the IRIS Assessment

Collectively, the highly suggestive evidence that early-life exposure to arsenic, even at low concentrations, increases the risk of adverse health effects and impaired development in infancy and childhood and later in life leads the committee to suggest that the timing of exposure, particularly early-life exposure, be considered in evaluating epidemiologic studies for dose–response assessment. The increasing body of data on mechanisms, including hormone interactions and epigenetic alterations, in support of those effects warrants consideration.

GENETICS OF ARSENIC METABOLISM AND TOXICITY

Genetic factors probably confer susceptibility or resistance to inorganic arsenic exposure. Genetic variants may have biologic effects of their own that act synergistically with inorganic arsenic or may have normal function in the absence of inorganic arsenic. Genetics may interact with inorganic arsenic ion multiple ways. Three possible interactions are as follows:

• Genetic variants may increase or decrease the absorbed dose of inorganic arsenic or alter the excretion of inorganic arsenic or its metabolites. The committee knows of no examples of this type of susceptibility.

• Genetic variants may alter the metabolism of inorganic arsenic independently of excretion or absorption. Most of the arsenic literature deals with this type of susceptibility.

• Genetic variants may increase or decrease the organ or cellular toxicity of inorganic arsenic. There is a small body of literature on this type of susceptibility.

As described below, there are many examples in the literature regarding genetic susceptibility factors, but their role in risk assessment is still unclear; the prevalence of these factors will vary in different populations that have different underlying ethnicities. Furthermore, the function of the genetic variants will probably vary with dose and may not be relevant in some exposure scenarios. Finally, most of the published reports are of case–control studies that provide few or no data on timing of exposure. An often overlooked underlying aspect of genetic susceptibility to environmental factors is the timing of exposure.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

Regardless of the type of environmental factor (for example, chemical, social, or nutritional), the timing of exposure probably plays a critical role in whether a genetic factor will interact with it.

Critical Developmental Windows and Gene–Environment Interactions

The field of gene–environment interactions is surprisingly underdeveloped. Most environmental health studies of gene–environment interactions have used a small-scale candidate-gene approach and have not planned for replication in independent populations. Underlying the problem is an often neglected aspect of genetic epidemiology studies: gene expression changes from one life stage to another. Genetic variants that produce gene–environment interactions might do so only when the exposure corresponds to a critical developmental window during which the gene is highly expressed (Wright and Christiani 2010). Most gene–environment interaction studies are case–control studies and cannot address critical developmental windows.

Genomewide Approach vs Candidate-Gene Approach

Most human studies of genetic variation and arsenic have focused on a few candidate single nucleotide polymorphisms (SNPs) that may modify arsenic toxicity or predict its metabolism. Although such an approach has strengths, including biologic plausibility and clear a priori hypotheses, there are limitations in selecting only a few SNPs for a study (Rebbeck et al. 2004). The potential functional effect of many polymorphisms is sometimes undefined or even controversial. In addition, it is unlikely that one SNP of one gene can account fully for the complexity of function of an entire biologic pathway. Finally, some genes or SNPs that have important roles in arsenic pathogenesis may not have been identified yet, and a candidate approach would not be able to identify such genes. Thus, bias is a concern in the selection of SNPs in a candidate approach (Wright and Christiani 2010).

An alternative approach is to use genomewide scans, which capitalize on advances in high-throughput technology and on the data from the completed HapMap Project (Hirschhorn 2005; Hirschorn and Daly 2005). Genomewide scans allow screening of the genome in an unbiased manner with respect to genetic risks factors (Kronenberg 2008). The greatest strength of such an approach is that it allows new biologic relationships to be discovered. Its primary weakness is that well-known biologic associations are ignored. When genetic associations, or gene–environment interactions, are simply ranked, a gene that may have greater biologic plausibility a priori is considered equal to all other genes—even genes that may not be expressed in the target tissue (Wright and Christiani 2010). To date, genomewide association studies have been conducted to identify genetic risk factors for diseases as varied as age-related macular degeneration (Haines et al. 2006), cardiac diseases (Cupples et al. 2007; Vasan et al. 2007), diabetes (Meigs et al. 2007; Wellcome Trust Case Control Consortium 2007), amyotrophic lateral sclerosis (van Es et al. 2007), rheumatoid arthritis (Plenge et al. 2007; Thomson et al. 2007; Wellcome Trust Case Control Consortium 2007), and cancer (Broderick et al. 2007; Murabito et al. 2007; Spinola et al. 2007; Zanke et al. 2007). Whole-genome sequencing is an approach that has the advantage of discovering rare variants that may modify arsenic toxicity. Overall, the field of genomics has undergone a paradigm shift from a hypothesis-driven approach to a hypothesis-generating approach. Because genomic approaches generate multiple comparisons on a grand scale, tests of statistical significance need to take into account that genes are not necessarily expressed independently of each other and still control for overall experimentwide error. One procedure is a correction for multiple comparisons, such as Bonferroni correction or control of false discovery rates. Such methods are purely statistical, however, and ignore biology. Another approach is to conduct the research in multiple populations sequentially. Researchers generate hypotheses in one population and then test them in separate populations as a second-line screening and validation or replication method (Rodriguez-Murillo and Greenberg 2008). At least one whole-genome interaction study of inorganic arsenic has been conducted (Pierce et al. 2012). The region proximal to the arsenic 3+ methyltransferase enzyme (AS3MT) gene was identified as containing variants associated with the percentage of

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

monomethyl arsenic (MMA) and dimethyl arsenic (DMA) metabolites and is associated with the risk of a disease outcome (skin lesions). Replication in an independent population is still needed, however.

Recently, a linkage analysis was conducted in the Strong Family Heart Study, a family-based study of mostly American Indian families in Arizona and Oklahoma (Tellez-Plaza et al. 2013). Urinary arsenic and its metabolites were considered as quantitative traits. Linkage studies differ from association studies (typically of case–control design) in studying family members and estimated heritability (the percentage of variance in urinary arsenic metabolites due to genetics). Regions in the genome that have suggestive logarithm of the odds of disease (LOD) scores (over 1.9) were described on chromosomes 5, 9, and 11. AS3MT is found on chromosome 10, and a locus that had an LOD score of 1.8 was found proximal to this site. Like genomewide-association studies, linkage studies identify genomic regions of interest, not specific genes. Further study, typically deep sequencing and the identification of functional variants, is needed before any genes can be considered as susceptibility factors. Finally, in any study of genetic contributions to arsenic metabolism, estimates of heritability are probably influenced by the degree of exposure in the underlying populations (that is, in the absence of arsenic exposure, a genetic trait cannot exhibit heritability), and this influence may explain the modest LOD score for the region proximal to AS3MT.

Studies of Genetic Susceptibility to Arsenic

Arsenic Metabolism: Role of Methylation

The metabolism of inorganic arsenic is critical for its toxicity and has been studied extensively in humans and animals. Two processes are involved: reduction and oxidation reactions that interconvert arsenate and arsenite and methylation reactions, which convert arsenite to MMA and DMA (Tam et al. 1979; Buchet et al. 1981a,b; Marcus and Rispin 1988). Inorganic arsenic is methylated, mainly in the liver, to form monomethylated and dimethylated metabolites (Vahter and Marafante 1987; Hopenhayn-Rich et al. 1993). There is also evidence of genetically determined differences in arsenic methylation (Vahter et al. 1995b; Engström et al. 2013; Harari et al. 2013; Schlebusch et al. 2013). The biomethylation process involves both the arsenic 3+ methyl transferase gene and and the glutathione S-transferase (GST) enzyme systems (particularly the omega class) (Styblo et al. 1995). Further upstream, several genes related to one-carbon metabolism (such as folate metabolism genes, which provide methyl groups as substrate for methyl transferase enzymes) are also involved. Once arsenic is absorbed into the blood, it undergoes reduction to arsenite and then methylation to yield MMA and DMA, which, in their pentavalent forms, are more readily excreted in urine. Studies have shown that animal species and individuals vary markedly with respect to arsenic methylation efficiency and that efficient methylation of inorganic arsenic is associated with a high rate of excretion (Vahter 2002). Gamble et al. (2005) reported associations between onecarbon donor status and urinary methylated arsenic species in Bangladeshi adults. The percentage of DMA in urine was positively associated with plasma folate, although the percentages of inorganic arsenic and MMA were negatively associated with folate. In a followup study, folate supplementation led to enhanced arsenic methylation and excretion and lower blood arsenic concentrations (Gamble et al. 2007).

Glutathione S-transferase

Glutathione could be a necessary catalyst for arsenic methylation through its role in the reduction of arsenate to arsenite. The GST-omega isozyme family is critical in this reaction although purine nucleoside phosphorylase can also reduce arsenate. There are eight classes of GST in mammalian cells, each of which may have subclasses of isozymes. These enzymes catalyze the nucleophilic attack on electrophilic chemicals, including xenobiotics, by glutathione. Various studies have addressed polymorphisms in GST isozymes and either arsenic speciation or toxicity. Chung et al. (2013) found an association between GST null variant (GSTM1 MspI) with increased risk of urothelial cancer. A GST-omega nonsynonymous SNP (Ala140Asp) modified the association between hair arsenic and metabolic syndrome (Wang et al. 2007).

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

Lesseur at al. (2012) assessed polymorphisms in five GST isozymes (P, M, O, T, and Z). A coding SNP in GST P1 (Ile105Val) modified the association of bladder cancer with toenail arsenic concentrations. Main effects were also noted in the risk of bladder cancer and GSTO2 SNP Asn142Asp and GSTZ1 SNP Glu32Lys. In a Vietnamese population, GST P1 Ile105Val was associated with lower MMA; this suggested a protective effect of the SNP (Agusa et al. 2012). Such a finding would be counterintuitive given the study by Lesseur et al. (2012) of this SNP with respect to toenail arsenic and bladder cancer; however, Lesseur et al. did not report arsenic speciation. L.I. Hsu et al. (2011) reported that the GSTT1 null variant modified the association between cumulative arsenic estimated by water concentrations and bladder cancer. Polymorphisms in other GST genes (GSTO1, GSTO2, GSTP1, and GSTM1) did not modify that association. Similarly, Hsieh et al. (2011) did not find an association or evidence of effect modification between arsenic exposure and GSTO1 and O2 polymorphisms and carotid atherosclerosis but did find evidence of effect modification between a haplotype of purine nucleoside phosphoralase SNPs and well-water arsenic. Finally, Chung et al. (2011) reported that subjects who had the GSTO2 Ala140Asp SNP had lower proportions of MMA in urine than subjects who were homozygous for the Ala allele; this suggests a protective effect of the SNP. That SNP was also assocated with a reduced risk of urothelial carcinoma. Collectively, those studies suggest that the genetic variability of the GST enzymes may influence susceptibility to arsenic toxicity, but the ability to use this information in the IRIS assessment is not clear.

Arsenic 3+ Methyl Transferase

The enzyme AS3MT catalyzes the methylation of arsenite by using S-adenosyl methionine as the methyl donor. The AS3MT gene has been the target of several candidate-gene studies of genetic susceptibility to arsenic. A nonsynonymous SNP M287T and an intronic SNP in the AS3MT gene were both associated with reduce methylated arsenic species in urine in American Indians (Gomez-Rubio et al. 2012). Lower levels of methylation of arsenic also predicted higher risk of metabolic syndrome in women in that study. Beebe-Dimmer et al. (2012) did not find an association between AS3MT SNPs and bladder cancer, but they found evidence of an interaction between arsenic in drinking water and the Met287Thr SNP with an odds ratio of 1.17 per 1-μg/L increase in water arsenic concentration restricted to people who had at least 1 copy of the Thr allele. The previously described study by Lesseur et al. (2012) did not find evidence that interactions between AS3MT SNPs and arsenic exposure affected bladder-cancer risk. Agusa et al. (2011) found relationships between urinary arsenic methylation and two noncoding SNPs. Engström et al. (2011) genotyped a suite of SNPs in the AS3MT gene in Bangladesh and Argentinian Andean populations that had high water arsenic exposures. They found a haplotype (series of SNPs on the same allele) that was associated with lower urinary percentage of MMA and one that was associated with higher urinary percentage of MMA in both populations. The haplotype with the higher percentage of MMA was the major allele in Bangladesh (52% allele frequency), whereas the haplotype associated with the lower percentage of MMA was the major allele in Argentina (70% allele frequency). In addition, four polymorphisms in DNA methylation enzymes DNMT1a and DNMT3b were associated with a higher urinary percentage MMA in the pregnant women in Bangladesh.

Genomic Screens

At least two larger-scale genomic screens have been conducted in populations characterized for arsenic exposure. These studies differ from the previously described linkage study in that they were not conducted in related people and used SNPs rather than short tandem-repeat markers to tag genomic regions of interest. Karagas et al. (2012) screened 10,000 nonsynonymous SNPs in a subset of subjects in a case–control study of bladder cancer conducted in New Hampshire (832 cases and 1,191 controls). The top hits were then validated in the larger case–control study. Variants in the FSIP1 gene (for a fibrous sheath interacting protein) and the SLC39A2 gene (for solute carrier protein 39A2, a metal transporter) modified the association between arsenic in drinking water and bladder cancer.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

In a genomewide genotyping analysis conducted in 1,313 Bangladeshi subjects, a significant association between arsenic metabolism and toxicity phenotypes and five SNPs proximal to the AS3MT gene was found (Pierce et al. 2012). An expression array conducted in a subset of 950 subjects demonstrated increased AS3MT expression associated with the SNPs, and this suggests that these are cis-quantitative trait loci. Finally, one of the SNPs modified the association between urinary arsenic and premalignant skin disorders in a case–control study in Bangladesh (Pierce et al. 2012).

Key Considerations for the IRIS Assessment

A number of studies have demonstrated that genetic factors can modify the response to inorganic arsenic. Most have been candidate-gene studies that focused on arsenic-metabolism genes. A small number have also addressed whether these genes modify the relationship between inorganic arsenic exposure and health effects but only for cancer, metabolic syndrome, cardiovascular disease, and skin lesions. There has been one genomewide scan (of 300,000 SNPs) in a population characterized for inorganic arsenic exposure, and it validated previous research on AS3MT gene variants as potential risk factors (Pierce et al. 2012). A family-based linkage study provided moderate evidence that the same region modifies arsenic metabolism (Tellez-Plaza et al. 2013). Although results have not been validated in independent populations and deep sequencing of the region has not been performed, this information, when considered with the results of candidate-gene studies of AS3MT, suggests that this gene is a modifier of inorganic arsenic metabolism. Nonetheless, further work is needed to characterize genetic susceptibility to arsenic. If the methods for incorporating genetics into risk assessment are sufficient at this stage, AS3MT would be a logical choice for incorporation. Candidate-gene studies have identified genetic variants that modify arsenic toxicity in base-repair genes (Applebaum et al. 2007; Breton et al. 2007; Ebert et al. 2011). These studies were specific to cancer end points. Overall, genetic risk factors for inorganic arsenic toxicity are still not fully understood, and for most genes the genetic risk probably varies too greatly with inorganic arsenic dose, age at exposure, and ethnicity of the underlying population to allow reasonable estimates of increased or decreased risk.

SEX DIFFERENCES IN ARSENIC METABOLISM AND HEALTH EFFECTS

There is evidence of marked sex differences in the metabolism and toxicity of arsenic. It is essential to evaluate such differences in the IRIS assessment to protect the most susceptible people in the population. Evaluation of sex differences may also provide information on mechanisms and modes of action of arsenic.

It is well documented that the biotransformation of arsenic differs by sex. Other factors that may influence arsenic metabolism, to be considered in evaluating the sex-dependent metabolism, include magnitude of exposure, age, pregnancy, liver diseases, and smoking (Vahter 2002, 2009). In general, women excrete a greater percentage of DMA and a lower percentage of MMA in urine than do men after similar exposures (Hopenhayn-Rich et al. 1996; Hsueh et al. 2003;Lindberg et al. 2007, 2008a; Tellez-Plaza et al. 2013). That is probably related to women’s having more efficient one-carbon metabolism than men, especially a potential of remethylating homocysteine via the choline–betaine pathway (Zeisel 2011). In addition, arsenic methylation efficiency increases during pregnancy (Concha et al. 1998a; Gardner et al. 2011).

Results of a number of studies have indicated that men are more affected by arsenic-related skin effects, including skin cancer, than women (Watanabe et al. 2001; Kadono et al. 2002; Chen et al. 2003; Rahman et al. 2006; Ahsan et al. 2007; Lindberg et al. 2008b; Leonardi et al. 2012). That does not seem to be the case for all arsenic-related cancers, however, and should be studied in more detail. As discussed in the committee’s workshop by Cantor (2013), the indicated differences might be influenced by the background prevalence of the end point under study, and epidemiologic studies have rarely evaluated the differences in relative or absolute risk of arsenic-related health effects. Recent data indicate that girls may

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

be at higher risk for developmental effects after early-life arsenic exposure (Hamadani et al. 2011; Saha et al. 2012; Gardner et al. 2013) and boys after prenatal exposure (Kippler et al. 2012).

There may be several mechanisms of the observed sex difference in arsenic toxicity. It may be mediated at least partly by a difference in arsenic metabolism, inasmuch as the reduced form of the monomethylated metabolite, MMA(III), is highly toxic. Efficient methylation to DMA increases the excretion of arsenic from the body (Vahter 2002). Arsenic-related skin lesions, for example, are influenced by methylation efficiency in a sex-dependent manner (Lindberg et al. 2008b). There may also be sex differences in toxicodynamics. For example, when pregnant mice were exposed to arsenic in drinking water at 42 or 85 mg/L on gestation days 8–18, female offspring developed ovarian and lung tumors and uterine and oviduct hyperplasia whereas male offspring had a highly increased incidence of liver and adrenal tumors later in life (Waalkes et al. 2003). Liver tumors were induced in female offspring only when the in utero arsenic exposure was combined with skin application of a tumor-promoting phorbol ester, 12-O-tetradecanoylphorbol-13-acetate (Waalkes et al. 2004a; Liu et al. 2006; Tokar et al. 2010b). Gene-expression analysis of the liver of male mice with hepatocellular carcinoma induced by exposure to arsenic in utero showed overexpression of ER-α and cyclin D1 and a feminized expression pattern of several cytochrome P450 genes (Waalkes et al. 2004b). In addition, hepatic DNA from male offspring showed a significant reduction in methylation in GC-rich regions (Xie et al. 2007). Interaction of arsenic with ER-α and estrogen-associated functions has previously been reported (Lopez et al. 1990: Chattopadhyay et al. 1999; Stoica et al. 2000; Chen et al. 2002; Du et al. 2012; Treas et al. 2012). Mice that were exposed postnatally showed fewer sex differences in cancer development; arsenic increased lung adenocarcinoma and hepatocellular carcinoma in both sexes, but gallbladder tumors occurred only in males (Tokar et al. 2011b).

In studies of mice exposed to more environmentally relevant doses of arsenic (0.05, 0.5, 5.0, or 50 mg/L in drinking water for 4 months), monthly assessment of locomotor activity showed that female mice in all treatment groups exhibited hyperactivity at every test. In contrast, male mice exhibited hyperactivity in the group exposed at 0.5 mg/L and hypoactivity in the highest-dose group after 4 months of exposure (Bardullas et al. 2009).

Key Considerations for the IRIS Assessment

There is clear evidence of sex differences in the metabolism of inorganic arsenic. Because arsenic metabolism is a recognized susceptibility factor, it seems likely that the toxicity of arsenic could also differ between men and women. However, only a few arsenic-related health outcomes have been evaluated by sex. It is essential to evaluate sex differences in the IRIS assessment to provide protection for the most susceptible people in the population.

NUTRITIONAL DEFICIENCIES

As discussed in the committee’s workshop by Beck (2013), there is considerable evidence from studies in animal models and in humans that nutritional factors may influence arsenic metabolism and toxicity. For example, studies of animals fed diets deficient in methionine, choline, or proteins have demonstrated decreased methylation and increased tissue retention of arsenic (Vahter and Marafante 1987). In West Bengal, India, poor nutritional status (low body weight) was associated with an increased risk of skin lesions (Mazumder et al. 1998). Similarly, in Bangladesh, lower body-mass index has been associated with an increased risk of arsenicosis skin lesions (Milton et al. 2004; Ahsan et al. 2006). In addition, using a validated dietary food-frequency survey in the large prospective Bangladesh Health Effects of Arsenic Longitudinal Study, Heck et al. (2009b) found that higher intakes of protein, methionine, and cysteine were associated with 10-15% greater urinary arsenic excretion after controlling for numerous covariates. Those are but a few examples of the extensive literature identifying poor nutrition as a susceptibility factor in arsenic toxicity.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

Essentially, the most important dietary factors aside from protein and amino acid intake appear to fall into two categories—vitamins and nutrients that are involved in one-carbon metabolism and the synthesis of SAM, the universal methyl donor; and selenium, an element whose antagonism of arsenic toxicity has long been known (Levander 1977; Zheng et al. 2005).

Nutritional Effects on One-Carbon Metabolism

Nutritional effects on one-carbon metabolism on arsenic metabolism and toxicity were discussed in the committee’s workshop by Gamble (2013). The synthesis of SAM, which is required for each of the two methylation steps of arsenic metabolism, is influenced by many nutrients that directly or indirectly contribute methyl groups to the one-carbon metabolic pathway (such as folate, choline, and betaine) or serve as cofactors for enzymes in the pathway (such as vitamins B2, B6, and B12) (reviewed in Hall and Gamble 2012). In particular, deficiencies of folate and the B vitamins appear to exacerbate the associations between inorganic arsenic exposure and its health effects in humans. For example, in a population-based cross-sectional study in Bangladesh, the association between arsenic exposure and hypertension was stronger in participants who had lower than average dietary intakes of folate and other B vitamins (Y. Chen et al. 2007b). A nested case–control study of incident skin-lesion cases and matched controls (Pilsner et al. 2009) in Bangladesh and a case–control study in India (Mitra et al. 2004) reported that folate deficiency is a risk factor for arsenic-induced skin lesions. That folate deficiency might modify the risks of arsenic-induced health outcomes can be explained by the observation that people who are folate-deficient are poor methylators of arsenic and have more of the more toxic inorganic arsenic and MMA metabolites in blood and urine than those who are folate-replete (Gamble et al. 2005). Moreover, folic acid supplementation of folate-deficient study participants facilitated the synthesis and urinary elimination of DMA (and total arsenic) and lowered blood MMA and total arsenic concentrations (Gamble et al. 2007). Another study of the influence of nutritional status on arsenic metabolism, carried out in pregnant women in Bangladesh, found only a marginal effect of plasma folate (after adjustment for the degree of arsenic exposure), which influences the methylation markedly (Li et al. 2008). That effect was due at least partly to the strong effect of pregnancy on arsenic methylation, starting very early in pregnancy and possibly related to the betaine-mediated induction of one-carbon metabolism in early pregnancy (Gardner et al. 2011).

Influence of Selenium Status on Arsenic Toxicity

Selenium and arsenic antagonize each other’s toxicity, probably because they facilitate the excretion of one another in bile (Levander 1977; Zeng et al. 2005). A selenium–arsenic–glutathione conjugate has been identified in rabbits and mice (Gailer et al. 2002; Burns et al. 2008) but has not been validated in humans. Epidemiologic studies have described inverse relationships between blood or plasma concentrations of selenium and the percentage of MMA in blood or urine or both (Basu et al. 2011; Pilsner et al. 2011). A case–control study in Bangladesh also reported inverse relationships between blood selenium concentrations and urinary arsenic and the risk of arsenic-induced skin lesions (Y. Chen et al. 2007a).

Key Considerations for the IRIS Assessment

It is clear that folate and selenium nutritional status have important effects on arsenic metabolism and toxicity. Therefore, the IRIS assessment should consider the nutritional status of study populations when examining dose–response relationships reported in the epidemiologic literature. Although a nutritional deficiency should not exclude an epidemiologic study from inclusion in a systematic review, nutritional factors could be qualitatively noted when the weight of evidence is considered. However, concerns about deficiencies affecting arsenic toxicity in the United States should be muted because of the nutrition-

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

al status of the population with regard to folate and selenium. The United States mandated the fortification of foods with folic acid in 1998, thereby decreasing the prevalence of folate deficiency (and rates of adverse birth outcomes associated with deficiency during pregnancy). However, some segments of the population rely on maize rather than wheat and thus do not benefit from supplemented foods. People exposed to arsenic in Bangladesh and much of South Asia have a high prevalence of folate deficiency, which exacerbates arsenic toxicity (Gamble et al. 2005, 2007; Hall and Gamble 2012). The selenium content of foods varies with the selenium content of soils in which they are grown. In the United States, soil selenium concentrations vary widely (USGS 2012). However, comprehensive reviews of blood and serum selenium concentrations in the United States (e.g., Combs 2001) have led to the conclusion that “the risk of selenium deficiency in the general US population is negligible” (Laclaustra et al. 2009).

PRE-EXISTING DISEASE, SMOKING, AND ALCOHOL CONSUMPTION

Pre-existing Disease

Risk assessment is increasingly concerned about the vulnerability that stems from interaction between a chemical’s mode of action and disease processes that can occur in segments of the population. With respect to inorganic arsenic, the evidence summarized in Chapter 4 that arsenic increases the risk for several major diseases—including cardiovascular, respiratory, and renal disease and diabetes—suggests that this source of vulnerability (pre-existing disease) may be particularly pertinent, especially given the high rates of these diseases in the United States. Because this is an emerging field of vulnerability assessment, several underlying principles and implications for quantitative risk assessment are outlined below in relation to considerations for inorganic arsenic. Chemical interactions with background disease processes may be of several types as shown in Figure 3:

•  The disease alters chemical action by altering toxicokinetics so as to change internal dose materially.

•  The disease alters chemical action by affecting host defense mechanisms, as can occur when a pathologic condition is associated with chronic inflammation and oxidative stress.

•  The chemical increases the likelihood of disease by damaging systems that are also affected by the disease process.

In the first two interactions, the presence of the disease process in an exposed person can be expected to shift the dose–response curve because the chemical is more effective in a diseased person. In the third, the disease is more likely to occur in an exposed person because of the chemical’s contribution to the pathologic process.

Those interactions may occur in many exposed people, but at low dose they are most pertinent in those who are furthest along the disease spectrum and thus make up a group that is highly vulnerable both to the disease and to chemical toxicity. The interactions are most likely to be important on a population level if the vulnerable group constitutes a sizable fraction of the population. For example, if the disease is rare, few people are at heightened risk; this might be important for these few, but the population risk may not be greatly affected. In contrast, if the disease is common, many people will have it, and many others will have risk factors and subclinical biomarkers indicative of it that can in time lead to it. That implies a vulnerability distribution in which healthy members of the population are resistant to the chemical and those on the threshold of disease are most vulnerable to a chemical-induced shift to clinical disease. From a dose–response perspective, such a distribution may cause effects to be seen at exposures well below the threshold in a typical person because sensitive members of the population will respond at lower doses and

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

image

FIGURE 3 Potential interaction between chemical exposure and disease process. Interaction could lead to altered chemical potency as disease process affects host toxicokinetics or defense mechanisms (downward arrows) creating a vulnerability to chemical effect. Interaction could also affect disease risk especially if the chemical and disease have similar upstream pathways and clinical outcomes (upward arrows). In this case, chemical exposure creates an additional risk factor for disease to occur.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

highly sensitive members at even lower doses. Such variability in threshold related to disease or other host factors may make it difficult to identify a population threshold at levels of exposure commonly encountered by the population even if a threshold exists at the individual level (Schwartz et al. 2002; NRC 2009). This is a likely explanation for the linear-appearing relationship between fine particulate matter (PM) and cardiovascular mortality (White et al. 2009). Inhalation of fine PM is a source of inflammation and oxidative stress that is associated with a variety of cell-signaling and physiologic changes that are common to the pathogenesis of cardiovascular disease. Alterations in heart-rate variability and telomere shortening are two such mechanisms whereby some members of the population (such as the elderly and those with impaired cardiopulmonary function) have values of these risk biomarkers in the direction of clinical disease and thus have a lowered threshold for PM-induced clinical effects relative to healthy people (Pope et al. 2004; Grahame and Schlesinger 2012; Langrish et al. 2012). Although fine-PM–induced cardiovascular effects may be widespread in the population at high dose, they might be experienced only by those who have preclinical risk biomarkers at low dose; the presence of these vulnerable people tends to push the observable dose–effect relation to lower doses (NAS 2009). If the dose–response relationship appears linear on that basis, it may have a different slope at low doses, at which the chemical is primarily adding to underlying disease, and at high doses, at which the chemical can exert its adverse effects even in healthy people.

Thus, the factors most important in evaluating the potential for arsenic to interact with background disease processes on a populationwide basis are these:

• Overlap between toxic mode of action and disease mechanisms in terms of similar target organs, similar upstream signaling pathways, common biomarkers (a biomarker of chemical effect is also a preclinical biomarker of disease), and common defense mechanisms involved in fending off the effect.

• Prevalence of the overlapping disease.

• Prevalence of preclinical disease.

Regarding disease prevalence, it is important to take note of whether particular segments of the population that may be exposed to arsenic are also more susceptible to an interacting disease; for example, black Americans have higher risk of hypertension and heart disease (Kurian and Cardarelli 2007).

Assessment of the potential for background disease interaction starts with evaluation of arsenic itself, its toxic outcomes, upstream indicators of effect, other relevant mode-of-action information, and governing toxicokinetic factors. The first avenue to explore is whether arsenic’s toxic outcomes are peculiar to arsenic or are similar to acute or chronic illness experienced by the population. For example, arsenic exposure is associated with skin lesions that are peculiar to arsenic overdose and are uncommon in the general population. Although it is possible that arsenic interacts with other skin conditions, it has not been documented except in the case of psoriasis, in which arsenic has a therapeutic effect (Farber 1992).

In contrast, epidemiologic studies show that exposure to arsenic increases the risk of cardiovascular disease, diabetes, renal disease, respiratory disease, neurodevelopmental impairment, and cancer (Naujokas et al. 2013). That is evidence that arsenic can contribute to some diseases that are prevalent in the US population. In some cases, the data indicate that arsenic affects subclinical markers of those diseases, and this provide mechanistic support for the epidemiologic associations. For example, a study in two Romanian towns found that people exposed to arsenic at 40 μg/L (on the average) in drinking water were more likely to have abnormal blood-pressure responses to psychologic or cold-induced stress (Kunrath et al. 2013). Those end points are predictive of hypertension (Wood et al. 1984). Arsenic exposure is associated with increases in plasma adhesion molecules that are indicative of endothelial dysfunction, vascular inflammation, and increased risk of cardiovascular disease (Y. Chen et al. 2007c). Thus, arsenic appears to influence cardiovascular-disease risk by at least two pathways at relevant doses. Comparable evidence on other disease outcomes (such as renal dysfunction, diabetes, and cancer) would be helpful in establishing the nature and extent of the interaction between arsenic and the disease process. Given that those diseases are prevalent in the US population, consideration should be given to whether many people may be vulnerable to the ef-

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

fects of arsenic exposure because the disease processes impair defense mechanisms or otherwise act in concert with arsenic’s mode of action.

The potential for arsenic to contribute to ongoing disease processes caused by other factors means that it has the potential to shift the distribution of a clinical biomarker toward a disease cutpoint so that a greater percentage of the population will be over the cutpoint and classified as having the disease. That effect is illustrated in Figure 4, in which the dose–response relationship for cadmium-induced decrease in renal function (glomerular filtration rate, GFR) is shown as a leftward shift in the baseline GFR distribution and more people are classified as having chronic renal disease (GFR <60 mL/min per 1.73 m2) (Ginsberg 2012). Cadmium risk can then be expressed as the increase in population risk of chronic renal disease rather than merely as a quantitative change in GFR or protein leakage. That is similar to how small decreases in IQ caused by lead exposure can appear minor on an individual level but be best understood on a population level by analyzing the effect on the IQ distribution of the population (Schwartz 1994; Grosse et al. 2002; Jakubowski 2011). It is important to consider that inferences about how changing the mean of the distribution affects the number of people in the tails are subject to a number of conditions related to population variability and heterogeneity (Bellinger 2007).

Given the evidence that arsenic affects the rate of various diseases, as well as upstream preclinical biomarkers of the diseases, it may be possible for a noncancer assessment to describe the increased disease risk associated with any particular arsenic dose. If a reference dose (RfD) is derived, it can be described as the dose associated with a de minimis increase in disease risk, whose statistical bounds are based on the degree of uncertainty and variability in the analysis. The determination of what is de minimis risk can take into consideration the severity of the end point and the size of the uncertainty distribution associated with the risk at the RfD (NRC 2009). That information will help risk managers to understand the implications of arsenic exposure and the meaning of the RfD if one is derived.

The practical implications of arsenic’s interaction with background disease are as follows:

• Those who have pre-existing disease (such as cardiovascular disease, diabetes, or renal dysfunction) may be more vulnerable to arsenic toxicity. Evaluating the evidence of susceptibility on the basis of pre-existing disease requires sufficient disease and nondisease subjects who have well-defined arsenic exposures and assessment of arsenic-related end points. Most epidemiologic studies, however, have not evaluated differences in arsenic susceptibility, so it is difficult to evaluate the importance of the interaction between arsenic and background disease.

image

FIGURE 4 Cadmium-induced shift in GFR distribution at a chronic exposure of 1 μg/kg per day relative to a 47.8- year-old female baseline. Source: Ginsberg 2012. Reprinted with permission; 2012, Journal of Toxicology and Environmental Health, Part A: Current Issues.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

• Those who are at high risk for a disease (they are genetically predisposed or preclinical markers are present) may be more likely to get the disease if exposed to arsenic. Evidence of that would ideally involve prospective studies that include assessment of a preclinical biomarker, arsenic exposure, and disease incidence over time. Without such evidence, it may still be possible to estimate the increased risk of disease (such as chronic renal disease or myocardial infarction) by evaluating how arsenic shifts the population distribution of preclinical biomarkers (such as GFR or heart-rate variability).

• It may be impossible to find a threshold of an arsenic-induced increase in disease in the population because a large number of people are in a preclinical state and would be sensitive to the low end of the dose–response curve. For that to be evident, the epidemiology data would need to characterize the relationship between arsenic exposure and risk of disease in a broad cross-section of the population (or look at precursor lesions or key events) and allow a robust examination of a low–dose slope.

• Whether the potential for arsenic interaction with disease processes differs between the populations from which the dose–response data are derived and the US population as a whole needs to be considered.

On the basis of the degree of evidence on a vulnerable subpopulation, consideration should be given to whether dose–response assessment will focus on the population as a whole or will involve separate assessments for the general population and susceptible subgroups. If it is the population as a whole, the traditional approach is to address variability with uncertainty factors; it may also be possible to analyze the effect of variability on risk by evaluating how the risk distribution of the disease shifts in response to arsenic. In essence, the risk distribution based on a subclinical biomarker is an expression of toxicodynamic variability that can be captured in dose–response assessment (NRC 2009; Ginsberg 2012).

The alternative approach is to address vulnerable subpopulations as separate from the general population and assign them unique potencies via dose–response modeling specific to the groups that might be based on actual–dose response data for the groups, on adjustments for specific toxicokinetic or toxicodynamic factors, or on more generic adjustment or uncertainty factors. An example of a generic approach is the application of age-dependent adjustment factors for heightened cancer potency in young children compared with adults in the case of mutagenic carcinogens (EPA 2005a). For arsenic, if it is known that a particular age group, disease (or disease-related end point), genetic variant, or coexposure creates unique vulnerability, efforts should be made to estimate the potency differences relative to the general population and on that basis to consider developing separate potency values or basing a single IRIS value on the most sensitive group or on the overall population with adjustments for vulnerable groups.

Potential Interaction with Smoking

Several epidemiologic studies have assessed the interaction between arsenic ingestion via drinking water and tobacco-smoking in Bangladesh (Chen et al. 2006; Melkonian et al. 2011), in Taiwan (Chen et al. 2004), or in occupational settings (Hertz-Piccioto et al. 1992). The studies have reported a synergistic interaction in which the occurrence of skin lesions (Chen et al. 2006; Melkonian et al. 2011) and the occurrence of lung cancer (Hertz-Piccioto et al. 1992) have been found to be greater than additive. Chen et al. (2004) found a dose–response trend for arsenic and lung-cancer risk that was more prominent among smokers. There is also evidence of an interaction between arsenic and smoking for cardiovascular disease (Y. Chen et al. 2011a; Moon et al. 2013) and bladder cancer (Karagas et al. 2004). The most detailed interaction study was done prospectively in nearly 4,000 Bangladeshi men with a 6-year followup. The synergistic effect between tobacco-smoking and arsenic exposure on skin lesions was most evident in the higher quintiles of arsenic exposure (Melkonian et al. 2011). For lung cancer, a reanalysis of six studies that reported on occupational arsenic exposure, smoking status, and lung cancer found a synergistic interaction that was also most evident in the higher arsenic exposure categories (Hertz-Piccioto et al. 1992). As summarized in Chapter 4, several lines of evidence suggest an interaction between smoking and bladder-cancer risk in which the arsenic-related bladder-cancer increase is more evident in smokers. No data

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

are available for determining whether exposure to environmental tobacco smoke interacts with arsenic with respect to skin, lung, or other end points. There are numerous potential mechanisms for the synergism, including inhibition of DNA repair by arsenic, which increases smoking-induced genetic damage. Most of the abovementioned studies of interactions between arsenic and smoking in Bangladesh and in occupationally exposed people involved men. There appears to be much less information on women. It was reported, however, that tobacco-chewing, which is prevalent in women in Bangladesh, was associated with a considerably higher risk of skin lesions in women than that in women who did not use tobacco (Lindberg et al. 2010). It was particularly obvious in women who had the lowest efficiency of arsenic methylation.

Because smoking is still relatively common, the finding of synergism with arsenic for at least four disease outcomes will be an important consideration. Indeed, of all susceptibility factors evaluated, smoking is probably the one on which the findings at both high and low to moderate arsenic exposure were most consistent. However, several uncertainties and data gaps with respect to this interaction could be acknowledged in attempting a quantitative assessment. The data gaps include lack of smoking–arsenic interaction studies for other end points, limitations of the information on the effects of secondhand tobacco smoke, the incomplete description of interaction dose–response relationships, and the lack of mechanistic understanding. Regarding interaction dose–response relationships, the interaction between two agents will depend on the relative size of the doses: the nature of the interaction may change as the ratio of exposures varies (e.g., see Chou and Talalay 1984). For arsenic, the interaction matrix for skin lesions suggests that the best chance to see synergism is at higher doses of arsenic (Melkonian et al. 2011), and this might suggest that the interaction is less likely to affect the dose–response relationship at low arsenic doses. However, that is uncertain given the limitatins of the statistical power in the epidemiologic data to find low-dose arsenic effects and interactions. For cardiovascular disease, for instance, there is evidence of effect modification at low to moderate concentrations of arsenic (Y. Chen et al. 2011a; Moon et al. 2013). Furthermore, the literature has generally evaluated smoking status (current, former, or never) without defining the amount of smoking needed to begin to see an arsenic interaction.

A plausible quantitative approach is a sensitivity analysis in which the smoking-interaction synergism size effect reported for skin lesions, lung cancer, bladder cancer, and cardiovascular disease is applied to the dose–response relationship for these end points to determine the degree to which it would change the potency calculation. If that degree is found to be influential, consideration can be given to providing a separate potency estimate for smokers. A separate estimate could be useful in standard-setting and particularly in risk–benefit analyses when the burden of disease related to arsenic exposure is assessed in the general population in light of the proportion of people who smoke. Public education regarding radon includes the cancer risk in nonsmokers vs smokers because of the synergistic interaction between these agents. If the data are sufficient, perhaps a similar approach could be used for public education concerning arsenic.

Potential Interaction with Alcohol Consumption

Although it is likely that many people have dual exposure to arsenic and ethanol, the potential for an interaction has not been extensively studied (Bao and Shi 2010). Results of a study of rats indicate that ethanol increases arsenic retention in the liver and kidneys and increases arsenic-induced hepatotoxicity (Flora et al. 1997). The epidemiologic data indicate higher concentrations of arsenic in urine in conjunction with alcohol ingestion, and this suggests a toxicokinetic interaction, higher arsenic concentrations in alcoholic drinks, or increased water intake after alcohol intake (Tseng et al. 2005). Ethanol may augment arsenic-induced oxidative stress and induction of angiogenic factors that would promote tumor growth (Klei and Barchowsky 2008; L. Wang et al. 2012). There are additional mechanistic avenues through which an interaction could occur and potentially affect the outcome of epidemiologic studies (Bao and Shi 2010). That may prove to be a subject of productive research but has not been sufficiently developed to understand the nature and dose–response relationship of coexposure interaction on key end points.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

Key Considerations for the IRIS Assessment

Interaction with some disease processes or chemical coexposures, particularly smoking, may increase vulnerability to the effects of arsenic. Evidence that arsenic alters disease incidence or interacts with upstream disease biomarkers shows which end points are most likely to involve disease-related vulnerability. Evaluation of the size and nature of vulnerable population groups will help to determine whether available epidemiologic studies adequately capture these groups. That consideration will also show how the response at doses below the range of observation might be affected with respect to the feasibility of defining a population threshold or dose-dependent transition. Quantitative approaches may involve separate analysis of vulnerable groups if such group-specific data are available or adjustment of the overall population response to account for specific chemical interactions or vulnerability factors.

MIXTURES AND COEXPOSURES

Coexposures or mixtures that include inorganic arsenic complicate the risk evaluation of arsenic in at least two ways: arsenic may interact with other agents that potentially modify the effect of arsenic, and there may be a combination effect of arsenic with other similarly acting chemicals (such as other metals).

Arsenic may interact with other agents because it perturbs pathways and end points shared by the agents. The effect can be a “zero interaction” called additivity (for example, anticholinesterase pesticides have a cumulative effect on key enzymes [Timchalk et al. 2005]), synergism (greater than additive, such as arsenic or radon plus smoking), or antagonism (less than additive, such as the antagonistic relationship between arsenic and selenium). Those interactions are typically dose-dependent for each agent, so assessing how one chemical alters the response to another can be complicated. Thus, the nature of the interaction may depend on the dose ratio and require testing multiple dose combinations of the interacting chemicals to explore fully the types of interaction possible (Jonker et al. 2005); or interactions that may occur at high doses of one or both chemicals may not occur at lower doses (an interaction threshold; Yang and Dennison 2007). The main concern at low doses is that an exposure to a chemical that is well tolerated in the population (for example, below its threshold dose in most or all people) can become an important risk contributor through additive or synergistic interaction. Zero interaction, or additivity, is typically seen as shifting the dose–response curve to the left (more potent) according to simple dose additivity, whereas a synergistic interaction may have a different dose–response curve. Dose additive or synergistic interactions are likely to be highly variable in the population because exposure to the agents is not uniform and leads to a range of responses and vulnerabilities. That added variability may be important to capture in a risk assessment for chemicals with which prominent interactions are likely to occur at doses experienced in the population or with which coexposure to multiple similarly acting chemicals is likely. Additional uncertainty factors are possible, but background interaction with similarly acting chemicals can be a reason that thresholds can be difficult to define at the population level and can lead to the potential for linear-appearing low-dose slopes and related modeling (NRC 2009).

Inorganic arsenic has varied modes of action and numerous end points, so the potential for chemical–chemical interaction or combination effects is large (Hong et al. 2004; Hore et al. 2007; Islam et al. 2011; Naujokas et al. 2013). Two types of interaction that have been identified beyond those previously discussed with selenium and tobacco smoke are interactions with other metals and with polycyclic aromatic hydrocarbons.

Coexposure and Potential Interaction with Other Metals

Like arsenic, a number of metals are pro-oxidants and share underlying effects on proteins, signaling pathways, and genetic instability; thus, numerous arsenic–metal interactions are plausible (Hartwig 2013).

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

Candidates for such interaction with arsenic include lead, mercury, cadmium, chromium, nickel, and cobalt (Yao 2008; Valko et al. 2005). For example, cadmium and arsenic appear to have cumulative effects on renal-tubule leakage, as evidenced in humans exposed at moderate environmental doses (Huang et al. 2009). Those effects appear to be mediated by oxidative stress inasmuch as oxidative biomarkers increased in a pattern that appears additive among these agents. Interactions involving arsenic and other metals have not been well researched.

Coexposure and Potential Interaction with Polycyclic Aromatic Hydrocarbons

Another possible interaction is between inorganic arsenic and carcinogenic polycyclic aromatic hydrocarbons (PAHs). PAH-related DNA adducts and mutagenesis are increased by coexposure to arsenic, and this interaction appears synergistic in at least some experimental systems (Maier et al. 2002; Fischer et al. 2005). The most plausible mechanism is that arsenic-induced inhibition of DNA repair allows the PAH adducts to be longer-lived and thus to build up in host DNA. Several studies that have tested that hypothesis, however, have failed to find impaired removal of PAH–DNA adducts in the presence of arsenic (Maier et al. 2002; Chiang and Tsou 2009). Other mechanisms are possible, including arsenic-induced impairment of the p53 response to DNA damage (Chen et al. 2005; Shen et al. 2008). The fact that arsenic also potentiates ultraviolet-induced genetic damage (Wiencke et al. 1997; Chen et al. 2005) demonstrates the potential for interactive effects beyond PAHs. Although those interactions remain to be explained, they raise the possibility that at least some of the arsenic-induced tumors seen in human studies are comutagenic effects. That theoretically points to an alternative cancer-potency estimate based on the dose–response relationship in which arsenic potentiates the toxicity–genotoxicity–carcinogenicity of other agents. However, that may be somewhat speculative given limitations in quantitative dose–response data and in the mechanistic information needed to explore such possible potentiation fully. Therefore, this is a subject worthy of mention by EPA as an additional mechanistic consideration that may help to explain some of the end points associated with arsenic exposure.

For interactions in which there is arsenic coexposure with other carcinogens, the standard risk-assessment assumption of dose additivity is reasonable.

Key Considerations for the IRIS Assessment

In its evaluation of inorganic arsenic, EPA should consider coexposure to other metals and PAHs because these are the most documented. In accumulating the evidence on mixtures that include arsenic, it would be helpful to take note of which chemicals arsenic tends to occur with in the environment. Coexposure to some metals (such as lead and cadmium) is particularly likely because of their similar sources, persistence, and retention in topsoil (ATSDR 2007; Wang and Fowler 2008). Arsenic in drinking water sometimes occurs with manganese, uranium, radon, and other elements (Ayotte et al. 2011). (Arsenic, cadmium, chromium, lead, and mercury are among the most common metals on the basis of site frequency as tabulated by the Agency for Toxic Substances and Disease Registry (ATSDR 2009). Arsenic also occurs with other environmental chemicals, including PAHs (Mori et al. 2011).

Three cases of possible interactions could guide EPA’s assessment of coexposures in the context of their affecting the interpretation of dose–response relationships in key epidemiologic studies.

Case 1. The key epidemiologic study does not have subjects who are more vulnerable as a result of coexposure to interacting metals or PAHs. For example, some Superfund sites might have hot spots of metal contamination involving arsenic and other metals. That could represent a special exposure scenario that is not captured by statistical sampling of a general population. By not counting or identifying such vulnerable people, an epidemiologic study might not capture the risk in this subgroup, and that could lead

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

to potential underestimation of risks to those who are subject to the coexposure. The concern is partially mitigated by recommendations made later in this report (see Chapter 7) to calculate potency values for multiple end points, not just the most sensitive one. The risk of each end point associated with arsenic could then be considered cumulatively with similarly acting metals that co-occur in that exposure groups.

Case 2. The key epidemiologic study has arsenic as a covariate (it is positively correlated) with other important metals and PAHs, but they are not measured. In such a study, the health effects might be completely (and incorrectly) attributed to arsenic without considering the contribution of other chemicals. That could lead to overestimation of the effects of arsenic.

Case 3. The key epidemiologic study has interacting metals or PAHs distributed by chance across the arsenic exposure groups; this would increase the unexplained variability in each group. The additional variability would tend to obscure or weaken the effect of arsenic.

In considering those scenarios, it would be helpful for EPA to assess, to the extent possible, how interacting metals or PAHs might co-occur in the epidemiologic study populations in comparison with the target population of the risk assessment. That could lead to a better understanding of potential underestimation or overestimation of risk and a description of how coexposures might contribute to overall potency on the one hand and uncertainty on the other and of whether cumulative risk posed by metals, PAHs, or other chemicals is important to consider.

SUMMARY

As described above, populations may be susceptible to the effects of arsenic because of their sex, life stage, nutritional status, metabolism-related polymorphisms, disease status, smoking status, and exposure to other chemicals that could interact with arsenic to produce a cumulative or synergistic effect. Consideration of susceptible subpopulations and life stages in deriving risk-based values is a complex task. Developing a potency adjustment for such populations is feasible if the appropriate dose–response data are available for a given subgroup in comparison with the general population. With respect to inorganic arsenic, it is plausible that those in pre-existing or high-risk categories for chronic diseases (such as cardiovascular disease, diabetes, and renal disease) could be more vulnerable to arsenic toxicity inasmuch as arsenic has been associated with increased risks of those conditions in at least some cases or has been shown to act on upstream biomarkers of disease risk (e.g., Wu et al. 2012; Kunrath et al. 2013). Thus, the available literature should be examined to assess whether such information is available in existing epidemiologic studies or systematic reviews. Windows of susceptibility appear to occur in connection with in utero or early-life exposure with respect to ensuing cancer, cardiovascular, and respiratory risk. Nutritional factors (such as low folate intake), genetic deficiency in arsenic methylation, and tobacco smoke might also predispose some people to arsenic toxicity.

The committee supports EPA’s plans to use systematic reviews to evaluate susceptibility factors. For many of the factors considered in this chapter, it is unlikely that adequate data will be available to incorporate the considerations quantitatively in estimating risks. The factors that appear to have the strongest evidence are in utero and early-life exposures, sex differences, and smoking. If adequate data are found, consideration could be given to determining a separate potency calculation for a vulnerable subgroup that would be carried through the IRIS process to the extent possible and to determining whether the vulnerability is appropriately represented in the study population relative to the target (US) population. In the absence of adequate data, the size of the relevant subgroup (if large, likely to contribute to population risk) and whether the mechanism for vulnerability (if known) is likely to be operable at low dose could be evaluated. At high dose, both vulnerable and less vulnerable people can be affected; but at low dose, only the most vulnerable are likely to be affected. If that is a small percentage of the population, there might not be enough people to permit recording a statistically significant effect at low dose. The larger the fraction of the population that has a particular vulnerability, the greater the chance that the population will continue to have a significant response at low dose and thereby extend the dose range of ob-

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

served effects. That is the apparent explanation of the linear-appearing population response to particulate matter and cardiovascular mortality in spite of the likelihood that a threshold for the effect occurs at the individual level (NRC 2009). As the dose is lowered, fewer people are responsive, so statistical significance is harder to achieve, but this does not mean that the risk at low dose is zero. In cases in which a sizable subpopulation is vulnerable to a particular effect, it is reasonable to extend the dose–response relationship below the range of observation by a modest extrapolation.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

6

Mode of Action

The Environmental Protection Agency (EPA) should evaluate the biochemical and systems-biology information available on arsenic to determine how it can help to explain the pattern of adverse health outcomes and the mode of action that best matches the underlying biology. Similarly, variability and its effect on the low end of the dose–response curve and the distinction between adaptive changes and adverse responses can be addressed in the context of mode of action.

Mode-of-action analysis provides a framework for data integration around a common theme for an agent and a specific health outcome (Andersen et al. 2000; Carmichael et al. 2011). The strategic vision for mode-of-action analysis in risk assessment has been laid out in EPA’s cancer-risk guidelines (EPA 2005b) and has been successfully applied to the assessment of chloroform (EPA 2001) and trichloroethylene (EPA 2011). EPA’s draft plans indicate that this mode-of-action framework will be included in its analysis of adverse-outcome pathways. Mode-of-action data can be extended to a number of uses beyond the cancer-risk guidelines, including noncancer health outcomes and sensitive population assessments. Mode-of-action analysis is a systematic approach to understand the relationship between exposure to an agent and biologic outcomes and can affect interpretation of events at the low end of the dose–response curve. It uses the identified key instigating events to drive dose–response analyses. An acknowledged challenge for EPA in using this approach is that many of the existing mode-of-action data have been developed at relatively high exposure to arsenic and the objective is to understand attributable risk at lower exposures. In addition, strong data on actual systemic exposure to the various forms of arsenic after ingestion are often difficult to obtain. Identifying those data gaps and their potential effect on the ability to extrapolate to potential effects at low exposures wil be an important part of the IRIS assessment process.

The mode-of-action framework (Boobis et al. 2006, 2008; Carmichael et al. 2011) in conjunction with the human-relevance framework (Meek et al. 2003) provides a transparent method of organizing information for hazard identification and risk assessment that includes exposure information, dose–response information, a clear conclusion, identified data gaps, and potentially susceptible populations. It permits the integration of cancer and noncancer risk assessment (Carmichael et al. 2011) and provides a transparent mechanism to share results with stakeholders. A similar approach that uses a graphical presentation of how epidemiologic and toxicologic data intersect is described by Adami et al. (2011). Similar discussions of new approaches to hazard identification and risk assessment are provided in Science and Decisions: Advancing Risk Assessment (NRC 2009) and Toxicity Testing in the 21st Century: A Vision and a Strategy (NRC 2007) and are meant to extend the concepts provided in the “Red Book” (NRC 1983).

IMPORTANT ASPECTS OF MODE-OF-ACTION ANALYSIS

The key aspect of mode-of-action analysis is that it provides an evidence-based assessment, integration, and synthesis of all the available data on a chemical, its adverse health effects, and its biology. The data integration and synthesis of a mode-of-action analysis incorporate various types of data, including epidemiologic data, standard toxicity-testing data, data from mechanistic biochemical and metabolism studies, data from gene-expression studies, and data on epigenetic changes. As discussed in the committee’s workshop by Clewell (2013), those data can come from in vivo or in vitro studies involving a variety

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

of species, systems, and cell types whose relevance to humans could be assessed. A chemical can modulate numerous upstream events, and determining whether these converge to form one or more modes of action for a specific adverse outcome can be challenging. Therefore, mode-of-action analysis endeavors to provide a plausible hypothesis for an adverse outcome (Andersen et al. 2000; Meek et al. 2003); there can be uncertainties in the sequence of events, in the causal nature of the events, and in whether more than one pathway is involved.

Mode-of-action analysis is distinct from a detailed understanding of mechanism of action and places into perspective all relevant scientific data that link key events and pathways to an adverse health outcome that is consistent with the underlying biology of the target tissue. It is that aspect of mode-of-action analysis that places available data into a context that can be used to interpret vulnerability among species and among individuals so that changes in dose–response relationships in going from high to low exposures can be understood. Thus, mode-of-action analysis is an essential step in Integrated Risk Information System (IRIS) assessment, and the committee applauds EPA’s efforts to organize the in vitro and in vivo mechanistic data and its plan to use this analysis in interpreting dose–response relationships and intersubject variability in the inorganic arsenic assessment.

Mode-of-action analysis should be applied to organize and synthesize data and to harmonize cancer and noncancer end points, especially in the case of arsenic, on which there are large data gaps in regions of the dose–response relationship in key areas and low-dose exposures of regulatory concern. Although association studies alone are not sufficient for quantitative risk assessment, they provide a qualitative guide for hazard identification. One should rigorously examine each (epidemiology) study in the context of the modified Bradford Hill criteria to determine appropriateness for risk assessment (Boobis et al. 2006, 2008). Coexposures are particularly important to take into account because differences in cancer tumor spectrum or other health outcomes (such as diabetes, cardiovascular disease, immune effects, and developmental effects) between populations may be due to the presence of different contaminants (such as lead, mercury, cadmium, and silica) or different dietary inadequacies (such as zinc, selenium, folate, and protein). Similarly, toxicology studies are often performed to detect hazards and may not be designed for description of mode of action. Mechanistic studies performed in vitro and in vivo can inform understanding of the mode of action. An emphasis on hazard characterization is warranted and can be achieved by using a mode-of-action approach in which the necessary key events for the pathogenesis of each health outcome are examined in a dose-dependent and time-dependent manner.

Not all proposed modes of action will be supported by adequate data, and there will not always be complete data on the health outcomes under consideration, but the mode-of-action framework should be worked through for the proposed modes of action for each adverse health outcome of concern. It is understood that for many of the health outcomes, a detailed mode-of-action analysis will not be possible, because of lack of information. Providing the available data and identifying data gaps and the effect of the lack of information on confidence in the assessment of risk, particularly at low exposures, will be important parts of the evaluation. When approached in this manner, a proposed mode of action provides a unifying hypothesis for interpreting and integrating multiple studies and for explaining differences between the sexes, among populations, among different ages, and among species. Several test cases for cancer mode of action have been provided, and this framework has been extended to noncancer end points (Sonich Mullin et al. 2001; Meek et al. 2003; Boobis et al. 2006, 2008; Rhomberg et al. 2011). The mode-of-action framework provides a transparent method for examining the weight of scientific evidence separate from policy considerations but with an eye to informing science policy (Carmichael et al. 2011).

In beginning a mode-of-action analysis, the committee recommends that the IRIS program use an approach similar to that followed in the IRIS assessments of chloroform (EPA 2001) and trichloroethylene (EPA 2011). The process needs to begin with the question to be answered about the health effects of exposue to arsenic. Identifying the adverse outcomes of concern and determining the strengths and weaknesses of the available data are important steps in this process. Toxicokinetic data are essential for understanding how exposure corresponds to internal dose and how they vary among individuals. Exposure assessment will influence the dose-dependent and time-dependent response to an agent and is important for dose–response analysis and for risk assessment. Understanding exposure data related to each

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

health outcome throughout the dose range is a particularly important aspect of the process and is an acknowledged difficulty that IRIS assessors will face in light of the existing arsenic database.

EPA’s draft plan for the inorganic arsenic assessment describes proposed evidence tables that will be organized by health category and will capture exposure and outcome information in human studies. A modified form is needed for animal or in vitro studies to record such information as the form of arsenic used, the species and strain, and the tissue or cell line. Understanding how the assessment was performed will be greatly facilitated by the use of additional tables that list modes of action for each health outcome (both cancer and noncancer) and include exposure, tissue dose (if possible), and key events. The analysis should further demonstrate how the documented exposure and time course of exposure lead to the key events. The tables should cite literature that was used to construct the analysis and discuss supporting and conflicting evidence. Linkage of adverse health outcomes to the key events in a mode-of-action analysis—when coupled with exposure, metabolism, tissue accumulation of parent and metabolites, and rate-limiting steps (such as protein binding, reactive oxygen signaling, cytotoxicity, proliferation, apoptosis, receptor effects, and immune suppression)—provides a strong hazard identification and characterization analysis. This evidence map of the exposure–response relationship, exposure frequency, and duration—when coupled with the time dependence of the sequential progression and time course of the observed health effects—should match the proposed key events predicted by the mode of action. Such a mode-ofaction assessment can be particularly helpful in the analysis of potential confounding effects, of the role of simultaneous exposure to other agents or actions, and of individual risks and population susceptibilities.

Mode-of-action analysis is a transparent method for examining data and interpreting data gaps and for providing an integrated assessment of hazard identification and risk characterization. The linkage of in vitro to in vivo animal to human dose and dose duration response to temporal onset of key events is necessary (Andersen et al. 2000; Tsuda et al. 2003; Slikker et al. 2004a,b). The data can then be compared to see whether they are qualitatively and quantitatively similar. That comparison could be facilitated by creating concordance tables in the range of observations for in vitro, in vivo animal, and human data. The tables should take into account the number of cases and the incidence, the actual exposure (on the basis of a common unit, such as micrograms per liter of plasma, micrograms per gram of tissue, current exposure, total cumulative exposure, or tissue concentrations), species or population (and subpopulation) differences, coexposures, and the timing and route of exposure in the analysis. It would be helpful to document intervention studies that perturb the adverse-outcome pathway either upstream or downstream of the proposed effect and thus provide evidence of the relative importance to the outcome in question. In addition, it would be useful to document how the observed health outcomes and mode of action attributed to arsenic could be modulated by other potentially causal agents, such as micronutrient deficiency, other metal excess, host vulnerability factors, life stages, pre-existing disease, diet, sex, genetic background, and smoking. Box 6 lists the major steps of mode-of-action analysis.

BOX 6 Steps of Mode-of-Action Analysis

1.  Provide problem formulation statement

2.  Tabulate adverse outcomes with supporting and conflicting data

3.  Provide pharamacokinetic data throughout the exposure and temporal range for each adverse health outcome and its precursors

4.  List modes of action for each adverse outcome, linking pharmacokinetic and pharmacodynamic information to health outcomes in an exposure and temporal manner

5.  Construct a concordance table to provide strengths and weaknesses of each proposed mode of action for each species, population, and subpopulation

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

POTENTIAL MODES OF ACTION OF ARSENIC

An important aspect of the IRIS assessment of inorganic arsenic will be provision of a proposed mode of action for each observed health outcome, including the associated supporting and contradictory evidence. If after execution of a mode-of-action framework analysis a cohesive mode of action is not apparent or it is clear that multiple modes of action may be involved, a mode-of-action summary statement should indicate that while elaborating the data and hypotheses assessed. Determination of the key events in an adverse-outcome pathway is in the context of the exposure and duration of exposure leading to the key intermediary steps in the pathogenesis of the health outcome. The key events must be linked in an exposure and temporal pattern for each of the steps in the pathway (cf Andersen et al. 2000; Tsuda et al. 2003; Slikker et al. 2004a,b). Where possible, data from multiple studies should be integrated to link tissue dose and biologically effective dose with the pathogenesis of the adverse outcome of concern. Thus, both dose response and temporal response must be consistent for a given hypothesized mode of action. For each primary health outcome, the precursor events (the rate-limiting or key events) can be determined as a function of time and exposure relative to the health outcome. Defining the key events consists of describing the absorption, metabolism, accumulation, and retention of arsenic or its metabolites in conjunction with any biochemical or functional change during the pathogenetic process that leads to the health outcome of concern (Boobis et al. 2009). For arsenic, defining the exposure at which tissue metabolism and accumulation occur at levels sufficient to trigger the next key event is important and could be informed by pharmacokinetic and pharmacodynamic models. The temporal and exposure relationship for the next key event for each health outcome in pathogenesis would permit linkage to the adverse outcome. The time course and dose dependence should be coherent with respect to dose, time, species, age, sex, and population for each adverse health outcome of concern.

The IRIS assessment should consider the plausible modes of action for each adverse outcome that might result from oral exposure to arsenic. Several modes of action have been described in the literature were discussed in the committee’s workshop by Cohen (2013) and detailed in Chapter 4 as potentially associated with arsenic action. They include binding to protein sulfhydryl groups, reactive oxygen generation, reactive oxygen species (ROS) signaling and the oxidative stress that ensues, and perturbation of DNA methylation (epigenetic factors) (Kitchin and Wallace 2008; Kitchin and Conolly 2010). Tissuespecific binding to protein sulfhydryl groups could be explored as a mode of action that can lead to inhibition of a specific protein activity (Kitchin and Wallace 2008). Sulfhydryls (such as glutathione), vicinal thiols (such as pyruvate dehydrogenase and some zinc finger proteins), or selenium may be the cellular target of arsenic and its metabolites.

An example of how the arsenic mode-of-action analysis could be structured is provision of what is known about the dose-dependent interactions of arsenic with protein sulfydryls, vicinal dithiols, and zinccontaining proteins and how the interactions might lead to biochemical changes (such as ROS generation), functional changes (such as mitochondrial damage), and epigenetic changes as a function of exposure. How those effects could culminate in the development of a specific health outcome can then be described to the extent possible on the basis of the underlying data (see, for example, Snow et al. 2005), especially with respect to their dose dependence. That approach is supported by the temporal associations that are observed in mice exposed to a high concentration of arsenic in drinking water (Kitchin and Conolly 2010). The temporal analysis is useful and when coupled with a dose-dependent and exposure-dependent profile may provide an integrated sequence of events that define arsenic adverse-outcome pathways. Such a framework may highlight the induction of ROS and redox imbalance as an integrative mode of action for lung cancer, bladder cancer, and cardiovascular disease.

Arsenic’s action is complex, and many mechanisms of action and several possible modes of action have been assessed in the context of exposure of humans to high doses through drinking water. It is generally accepted that arsenic is not directly mutagenic but rather acts via an indirect mechanism that involves secondary mediators (such as oxidant damage, modified proteins, and immune suppression). Other research has provided data to support proliferation as a mode of action of bladder carcinogenesis associated with high-dose exposure to inorganic arsenic. In adult rats given arsenic directly, cytotoxicity is ob-

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

served and is followed by regenerative repair (Nascimento et al. 2008; Suzuki et al. 2008a,b, 2009a,b, 2010, 2012; Yokohira et al. 2010). Similarly, an increased incidence of bladder neoplasms is observed in adult mice exposed to high doses of arsenic during the period of rapid cell proliferation in utero (Tokar et al. 2010b). Another hypothesized mode of action is epigenetic modulation that may influence arsenic-induced signaling response. The most studied aspects of the epigenome are DNA methylation, covalent posttranslational histone modifications, and microRNAs, and they have been associated with both cancer and noncancer effects. Epigenetic modifications are believed to influence signaling events and phenotypic variation in cells. DNA methylation is the most widely studied epigenetic modulation in humans and other mammals; it can influence gene silencing by directly affecting the affinity of transcription factors for their DNA binding sites. Arsenic exposure has been associated with decreased S-adenosyl methionine concentrations (and increased concentrations of S-adenosyl homocysteine), altered methyltransferase activity, and changes in DNA methylation patterns in animals and humans (Reichard and Puga 2010). Dose and temporal association of DNA methylation changes with key initiating events would need to be derived to support its role in mode of action analyses.

Gene-expression and other -omic data can support the determination of mode of action. Several groups have performed gene expression in cell lines, in primary human cells, and in tissues from animals treated with arsenic, and the results need to be organized by health outcome and dose, exposure duration, and time course of response as reviewed by Ankley et al. (2010). Analysis of the quality of the data and the normalization methods need to be considered. Several additional caveats need to be considered in that driver genes may be expressed at low levels and not detected, the relevant change may not be at the gene transcription level, many changes are simply responses and not causal mechanisms, and not all changes are adverse. In addition, the genomic profile should be obtained of the tissue type of origin (specifically, primary cells from this tissue for in vitro studies) for the health outcome of concern. Because genes are not independent variables, one should focus on rate-limiting steps for the proposed mode of action and key events. Integration of -omic data permits linkage among tissues, and the Bayesian network-analysis approaches may extend to comparison of populations (Subramanian et al. 2005; Geneletti et al. 2011; Schadt and Björkegren 2012). Pathway mapping can be a useful place to start in such qualitative analyses (Clewell et al. 2011; Yager et al. 2013), but current datasets need to be critically assessed with regard to normalization procedures and statistical control of batch effects and multiple comparisons. In addition to pathway mapping, one might focus on ligand through receptor to signaling to transcription-factor analysis or on the transcription-factor set (for example, zinc finger proteins with vicinal thiols). It is important to remember that genes are not independent variables and that statistical significance is not equivalent to biologic relevance (Moggs et al. 2004; Mootha et al. 2004). Several papers provide a framework for the use of gene-expression data to inform understanding of key events (Bercu et al. 2010; Gentry et al. 2010); however, they have focused on perturbation of pathways, not on approaches to determining mode of action or key events (Moggs et al. 2004; Mootha et al. 2004). Gene-expression analyses permit hypotheses—such as selenium depletion, zinc finger protein inactivation, or interruption of mitochondrial respiration—to be explored to support assessment of mode of action and key events.

Modified Bradford Hill criteria for assessing causality have been used to provide a structure for assessing causality systematically in toxicologic and epidemiologic studies (Adami et al. 2011). Causality assessment is a key defining principle in mode-of-action analyses and is an integral part of a toxicologic assessment. The application of the modified Bradford Hill criteria in the integration of mechanistic, toxicologic, and epidemiologic data is necessary to determine which adverse health outcomes are causally associated with a specific exposure, exposure duration, and timing of exposure. The application of these criteria to combined mechanistic and epidemiologic data has been conceptualized by Adami et al. (2011), and they have been applied to the effects of atrazine on breast cancer by Simpkins et al. (2011). EPA proposes the use of adverse-outcome-pathway analysis in a separate construct that is similar to mode-ofaction analysis but does not consider that some changes are physiologic or even adaptive and not adverse. The utility of mode-of-action and adverse-outcome-pathway analysis lies in the fact that their dependence on the modified Bradford Hill criteria for causality assessment relies on exposure and temporal dependence for coherence of any observed effects; thus, this emphasizes attributable risk.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×

SUMMARY

Mode-of-action analyses permit a transparent assessment of the data supporting or refuting the human health effects of exposure to oral inorganic arsenic in the US population. Both cancer and noncancer effects can be integrated through a mode-of-action analysis. Such analysis permits an unbiased use of all the available data to examine the effects of arsenic exposure and exposure duration at different doses. A mode-of-action analysis needs to be compiled for each health outcome that has been ascribed to arsenic exposure to inform the risk assessment. Numerous mechanisms of action have been proposed for arsenic-associated health outcomes (including protein binding, ROS generation, epigenetic effects, and cytotoxicity), and it is unlikely that a single mode of action is responsible for all the observed adverse health outcomes. Mode-of-action analysis permits the integration of data to advance understanding of the coherence, biologic plausibility, and human relevance of findings throughout the exposure–response continuum and provides a transparent means of synthesizing the data. Mode-of-action analysis may be particularly useful in understanding low-dose exposure and dose-dependent transitions that may occur with increasing dose. The exposure (the tissue or biologic effect level) is considered in the context of metabolism, transport, and accumulation in the tissues of interest. Where nonlinearities in the pharmacokinetics or pharmacodynamics are identified, they can be used to understand the time and dose dependences of the key rate-limiting events and of the proposed adverse health outcome. The mechanistic, toxicologic, and epidemiologic data can then be interpreted in a comprehensive mode-of-action framework that facilitates an improved understanding of exposure–response relationships and interhuman variability of response.

Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 51
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 52
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 53
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 54
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 55
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 56
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 57
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 58
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 59
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 60
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 61
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 62
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 63
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 64
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 65
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 66
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 67
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 68
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 69
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 70
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 71
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 72
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 73
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 74
Suggested Citation:"5 Susceptibility Factors." National Research Council. 2013. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report. Washington, DC: The National Academies Press. doi: 10.17226/18594.
×
Page 75
Next: 7 DoseResponse Analysis »
Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic: Interim Report Get This Book
×
Buy Paperback | $39.00 Buy Ebook | $31.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

The US Environmental Protection Agency (EPA) Integrated Risk Information System (IRIS) program develops toxicologic assessments of environmental contaminants. IRIS assessments provide hazard identification and dose-response assessment information. The information is then used in conjunction with exposure information to characterize risks to public health and may be used in risk-based decisionmaking, in regulatory actions, and for other risk-management purposes. Since the middle 1990s, EPA has been in the process of updating the IRIS assessment of inorganic arsenic. In response to a congressional mandate for an independent review of the IRIS assessment of inorganic arsenic, EPA requested that the National Research Council convene a committee to conduct a two-phase study. Critical Aspects of EPA's IRIS Assessment of Inorganic Arsenic is the report of the first phase of that study. This report evaluates critical scientific issues in assessing cancer and noncancer effects of oral exposure to inorganic arsenic and offers recommendations on how the issues could be addressed in EPA's IRIS assessment.

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!