Skip to main content
Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
EFSA J. 2024 Jan; 22(1): e8488.
Published online 2024 Jan 18. doi: 10.2903/j.efsa.2024.8488
PMCID: PMC10794945
PMID: 38239496

Update of the risk assessment of inorganic arsenic in food

Associated Data

Supplementary Materials

Abstract

The European Commission asked EFSA to update its 2009 risk assessment on arsenic in food carrying out a hazard assessment of inorganic arsenic (iAs) and using the revised exposure assessment issued by EFSA in 2021. Epidemiological studies show that the chronic intake of iAs via diet and/or drinking water is associated with increased risk of several adverse outcomes including cancers of the skin, bladder and lung. The CONTAM Panel used the benchmark dose lower confidence limit based on a benchmark response (BMR) of 5% (relative increase of the background incidence after adjustment for confounders, BMDL05) of 0.06 μg iAs/kg bw per day obtained from a study on skin cancer as a Reference Point (RP). Inorganic As is a genotoxic carcinogen with additional epigenetic effects and the CONTAM Panel applied a margin of exposure (MOE) approach for the risk characterisation. In adults, the MOEs are low (range between 2 and 0.4 for mean consumers and between 0.9 and 0.2 at the 95th percentile exposure, respectively) and as such raise a health concern despite the uncertainties.

Keywords: benchmark dose (BMD), epidemiological studies, inorganic arsenic (iAs), margin of exposure (MOE), risk assessment

SUMMARY

In 2009, the EFSA Panel on Contaminants in the Food Chain (CONTAM) adopted a Scientific Opinion on the presence of arsenic in food. The European Commission (EC) has asked the European Food Safety Authority (EFSA) for an update of the hazard assessment of inorganic arsenic (iAs) because new studies have become available on the toxic effects of iAs since the publication of the 2009 Opinion. Using the revised exposure assessment on iAs issued by EFSA in 2021 an updated risk assessment of iAs for human health should be performed. The EC has also asked for risk assessments on small and complex organo arsenic species and on the combined exposure to inorganic and organic arsenic which will be provided in separate Opinions.

Arsenic is a metalloid, widely present in the environment because of both natural occurrence and anthropogenic activity. Arsenic occurs in various organic and inorganic forms. In food and feed, inorganic arsenic species are predominantly in the +3 or + 5 oxidation state, present as thio complexes or, as the oxo anions arsenite and arsenate. During sample preparation and analysis iAs bound to thio groups in peptides or proteins in food is converted to arsenite and arsenate. Hence, data on occurrence are nearly exclusively recorded as these two analysed species. The sum of the quantified arsenite and arsenate in food (in mg As/kg wet weight) is often referred to as iAs. In natural water and drinking water, arsenic habitually appears as arsenite and arsenate, whereas organic arsenic forms are rare in water. In humans, iAs is rapidly and to a great extent (45%–80%) absorbed after ingestion, widely distributed in the body to almost all organs. It readily crosses the placental barrier. iAs is metabolised by reduction, oxidative methylation, thiolation and glutathiolation. The degree of methylation is crucial for its toxic effects in humans. Inorganic arsenic is eliminated via the urine in the form of iAs and its methylated metabolites, with a half‐life of around 2–3 days.

The toxicokinetics of iAs in laboratory animals differ considerably from those in humans, particularly regarding their methylation capacity. Therefore, toxicity data resulting from animal experiments are not a suitable basis for human health risk assessment.

There is no universally accepted biomarker for chronic exposure to iAs. Measurements of total arsenic in blood or urine represent not only iAs but also organic As species and are thus of limited value. Specific measurements of iAs, and its methylated metabolites, or the sum of iAs, and its methylated metabolites, also named total urinary inorganic arsenic (u‐tiAs), provide more appropriate estimates of iAs exposure. Inorganic arsenic accumulates in hair and nails. Thus, As in hair and nails is also used as biomarker for chronic iAs exposure.

Chronic exposure to iAs is associated with increased DNA damage (DNA breaks and oxidatively induced DNA base modifications) and clastogenic events in somatic cells from exposed individuals. Chronic exposure to iAs in utero or adult life is also associated with epigenetic changes that may lead to aberrant gene expression.

Inorganic arsenic is itself a weak inducer of gene mutations, but efficiently induces chromosomal aberrations, micronuclei and aneuploidy in vitro and in vivo. Inorganic arsenic does not directly interact with DNA, but it induces oxidative stress, which is believed to play a role in the formation of DNA base oxidation as well as both DNA single and double strand breaks. The production of oxidative clustered DNA lesions is likely responsible for iAs induced double strand breaks.

Inorganic arsenic and its metabolites interfere with the DNA damage response by inhibiting DNA repair and by interfering with cell cycle control‐ and apoptotic pathways. The inhibition of DNA repair systems leads to accumulation of DNA damage induced by both exogenous and endogenous sources. This phenomenon likely contributes to the co‐mutagenic effects observed when iAs is combined with exposure to other DNA‐damaging agents. Methylation of iAs, particularly to trivalent methylated species, should be regarded as an activation process that forms more reactive species, which exert stronger cyto‐ and genotoxic effects.

In animal studies, increases in tumour incidence upon exposure to iAs via drinking water have been observed. However, the results of these studies with respect to tumour sites and effective doses are inconsistent and do not provide a robust basis for risk assessment.

For hazard assessment only human studies were considered. The studies encompassed study subjects with exposure to long‐term low to moderate levels of iAs, defined as concentrations of arsenic in water of less than ~ 150 μg/L, or biomarker concentrations estimated to result from equivalent doses.

The evidence from epidemiological studies can be considered as sufficient and causal for associations between low to moderate exposure to iAs (defined as above) and cancers of the skin, bladder and lung, skin lesions other than skin cancer, spontaneous abortion, stillbirth, infant mortality, congenital heart disease, respiratory disease, chronic kidney disease, neurodevelopmental effects, ischemic heart disease and carotid artery atherosclerosis. However, since the epidemiological studies on skin lesions were performed in low and medium income regions (Bangladesh, China) and nutrition and health status are important modifying factors, it is difficult to translate these risks to populations with more adequate nutrition such as in Europe.

The evidence from epidemiological studies performed in Bangladesh is sufficient to assume a causal association between low to moderate exposure to iAs and decreased birth weight. However, the results for this effect from other countries (Chile, Taiwan, Mongolia, Mexico, US) are inconsistent. Moreover, the average birth weight in Europe is much higher and undernutrition is less common, putting the relevance of the evidence from Bangladesh for a European population in doubt.

There is insufficient evidence for an association between low to moderate exposure to iAs, and breast, prostate, kidney, liver, pancreatic and gallbladder cancer, male fertility, neurotoxicity, stroke and hypertension, glucose metabolism, diabetes and metabolic syndrome.

Data from studies were considered for dose–response modelling if the studies had a low level of overall risk of bias. In addition, data had to show a statistically significant association with iAs as a continuous variable and/or a statistically significant trend test and/or a statistically significant increase of risk in the upper exposure category. For modelling of quantile data, results for at least three exposure categories in total were required.

For studies meeting these criteria, iAs concentrations in drinking water (measured as total As) were transformed into total daily iAs dietary exposures (applying default or reported water intakes and adding region specific exposure estimates to account for the contribution from food). When total urinary iAs concentrations (sum of iAs and its methylated metabolites) were reported these were transformed into daily iAs dietary exposures per kg bw per day by applying default factors for urine volume or creatinine excretion.

The results used for dose–response analysis are the adjusted incidences and resulting number of cases based on the adjusted risk ratios reported in these studies and the provided (cohort studies) or estimated (case–control studies) population sizes. As benchmark response (BMR), a relative increase of 5% of the background incidence (estimated from the lowest exposure category) after adjustment for confounders was used.

The results from 20 epidemiological studies fulfilled the validity criteria for BMD modelling and the results from these benchmark dose (BMD) calculations were considered further for the selection of an appropriate Reference Point. For the six cohort studies the follow‐up time varied between 3 and 12 years.

Low benchmark dose lower confidence limits (BMDLs) (≤ 0.17 μg iAs/kg bw per day) based on a BMR of 5% relative increase of the background incidence after adjustment for confounders were calculated for skin cancer, lung cancer, bladder cancer, respiratory disease, skin lesions, chronic kidney disease and ischemic heart disease.

The CONTAM Panel used the BMDL based on a BMR of 5% (relative increase of the background incidence after adjustment for confounders, BMDL05) of 0.06 μg iAs/kg bw per day obtained from a case–control study on skin cancer (squamous cell carcinoma) carried out in the US, as a reference point (RP) because the study was considered to be of good quality with a low risk of bias. Exposure categories were based on u‐tiAs. The results from a second skin cancer case–control study on basal cell carcinoma with a population from Hungary, Romania and Slovakia support the choice for an RP based on skin cancer for hazard assessment.

The CONTAM Panel concluded that an RP of 0.06 μg iAs/kg bw per day should also be considered to cover lung cancer, bladder cancer, skin lesions, ischemic heart disease, chronic kidney disease, respiratory disease, spontaneous abortion, stillbirth, infant mortality and neurodevelopmental effects.

Inorganic As is a genotoxic carcinogen. Both thresholded and non‐thresholded mechanisms could apply for the different genotoxic effects of iAs and its trivalent and pentavalent methylated metabolites. Therefore, the CONTAM Panel concluded that it is appropriate to apply a margin of exposure (MOE) approach for risk characterisation rather than establishing a health‐based guidance value. There are no precedents in EFSA for identification of an MOE of low concern, when using a BMDL derived from human cancer data. Therefore, the Panel decided not to determine a value for an MOE of low concern.

The RP of 0.06 μg iAs/kg bw per day for skin cancer is in the range of the mean dietary exposure estimates for iAs in adults (0.03–0.15 μg iAs/kg bw per day), and below any of the 95th percentile exposure estimates in adults (range = 0.07–0.33 μg iAs/kg bw per day). Therefore, in adults, the MOEs range between 2 and 0.4 for mean consumers and between 0.9 and 0.2 at the 95th percentile exposure, respectively.

Based on the conditional uncertainty analysis and considering both studies on skin cancer, the probability that the mean exposure scenario exceeds the associated BMDs range from unlikely (likelihood ≈ 0.17) to likely (likelihood ≈ 0.86).

An MOE of 1 would correspond to an exposure level that is associated with a 5% increase relative to the background incidence for skin cancer, based on the available data. The CONTAM Panel concludes that this MOE raises a health concern.

The CONTAM Panel notes that dietary iAs exposure is higher in the younger age groups and therefore the respective MOEs are smaller. However, this does not necessarily indicate that children are at greater risk, because the effects are due to long‐term exposure and most of the epidemiological studies are conducted in adults who would also have had higher dietary exposure during early life. Therefore, the CONTAM Panel concludes that children are covered by this risk characterisation.

Although risk characterisation is based on the results of relatively large epidemiological studies, susceptible individuals of higher genetic risk may not be adequately represented in these studies. Therefore, dietary exposure to arsenic may be of greater concern for such individuals than for the general population.

According to the report from EFSA from 2021, across the different age classes, the main contributors to the dietary exposure to iAs (LB) were ‘Rice’, ‘Rice‐based products’, ‘Grains and grain‐based products (no rice)’ and ‘Drinking water’. Particular foodstuffs indicated for the young population (e.g. ‘Cereal‐based food for infants and young children’ and ‘Biscuits, rusks and cookies for children’) made a relevant contribution in the dietary exposure to iAs in this age group.

The CONTAM Panel notes that an EFSA guidance on the use of human data for risk assessments is needed, in particular on BMD modelling of epidemiological data and for a quantitative risk assessment for genotoxic carcinogens based on epidemiological data. The Panel recommends investigating the relevance of arsenic‐induced epigenetic alterations for iAs associated disease risk. The mechanisms of induction of DNA double strand breaks by arsenic should be investigated to clarify its mode of interaction with DNA. Additional research should explore the mechanisms underlying genomic instability caused by iAs. The health effects of pre‐ and perinatal exposure to arsenic and how arsenic‐induced alterations occurring during early life can impact the development of certain diseases in adult life should be further elucidated. The role of inter‐individual variations in susceptibility to arsenic‐related health conditions should be investigated with a focus on arsenic biotransformation and differences in DNA repair. Moreover, the CONTAM Panel notes that several recommendations regarding the dietary exposure assessment for iAs, which are still valid, were made in the 2021 EFSA scientific report.

1. INTRODUCTION

1.1. Background and Terms of Reference as provided by the requestor

1.1.1. Background

In 2009, the EFSA Panel on Contaminants in the Food Chain (CONTAM) adopted a Scientific Opinion on the presence of arsenic in food. Arsenic occurs naturally in soil and ground water. Inorganic arsenic may cause cancer of the skin, urinary bladder and lungs and EFSA calculated BMDL01 values for these effects between 0.3 and 8 μg/kg bw per day, highlighting a possible risk to consumers on the basis of the estimated exposure. However, for the organic arsenic forms EFSA indicated that arsenobetaine, which is the major form of arsenic in fish and most seafood, is widely assumed to be of no toxicological concern. Arsenosugars and arsenolipids are mainly metabolised in humans to dimethylarsinate (DMA(V)), but sparse information is available regarding their toxicity. For other organoarsenic compounds no human toxicity data are available. Because of the lack of data, arsenosugars, arsenolipids, methylarsenate (MMA(V)) and dimethylarsinate (DMA(V)) could not be considered in the risk characterisation.

In its 2014 scientific report on dietary exposure to inorganic arsenic in the European population, 1 EFSA identified grain‐based products as the main contributor to the exposure and also rice, milk and dairy products as important contributors. The heterogeneity of the food consumption data, the conversion of total arsenic to inorganic arsenic and the treatment of left censored data represented important uncertainties in the exposure assessment.

Commission Recommendation (EU) 2015/1381 2 recommended Member States to monitor during 2016, 2017 and 2018 the presence of arsenic, preferably by determining the content of inorganic and total arsenic and, if possible, other relevant arsenic species, in a wide variety of food, and to provide these data to EFSA on a regular basis at the latest by October 2018.

The newly available occurrence data have been used for an updated consumer exposure assessment for inorganic arsenic, which was endorsed by the CONTAM Panel of 24–25 November 2020.

Since the publication of the 2009 Opinion on arsenic in food new studies have become available on the toxic effects of inorganic arsenic and it is therefore appropriate to update the hazard characterisation. Taking into account the new exposure assessment and new information on the adverse health effects, a new risk assessment should be performed for the risks for human health related to inorganic arsenic in food. Since the publication of the 2009 Opinion on arsenic in food new studies have become available on the toxic effects of organic arsenic. Furthermore, it has become clear that dimethylmonothioarsenate (DMMTA, thio‐DMA(V)) is formed from methylarsenate (MMA(V)) due to the application of sulphate fertilisers, and that this substance could be more toxic than methylarsenate (MMA(V)). It is therefore appropriate to update the hazard identification and characterisation of organic and inorganic arsenic. On the basis of the available occurrence data for organic arsenic or literature information on organic arsenic in food, the exposure could be estimated in order to carry out an assessment for the risks for human health related to organic arsenic in food. In view of different properties of the organoarsenic species, arsenobetaine, arsenosugars and arsenolipids, it is appropriate to assess the organic arsenic species in 2 further separate opinions: an Opinion on small organoarsenic species and an Opinion on arsenobetaine, arsenosugars and arsenolipids, which should also include other relevant complex organoarsenic species. After finalising the risk assessments on inorganic and organic arsenic, also the combined exposure to inorganic and organic arsenic should be assessed.

1.1.2. Terms of Reference

In accordance with Art. 29 (1) of Regulation (EC) No 178/2002 the Commission asks EFSA for

  • an updated consumer risk assessment for inorganic arsenic in food, taking into account
    • the updated exposure assessment, endorsed by the Panel in November 2020.
    • newly available scientific information
  • a consumer risk assessment for organic arsenic in food, taking into account
    • newly available scientific information
    • occurrence data for organic arsenic in food
    • literature data on the occurrence of organic arsenic in food
  • a consumer risk assessment on the combined exposure to inorganic and organic arsenic, taking into account
    • the risk assessment on inorganic arsenic in food
    • the risk assessments on organic arsenic in food

1.1.3. Interpretation of the Terms of Reference

The present Opinion is the first of in total four scientific opinions covered by the ToR from the EC (see Section 1.1). It is an update of the previous EFSA Opinion on inorganic arsenic (iAs) in food (EFSA CONTAM Panel, 2009) which has been used as a starting point for the assessment. In this Opinion the adverse effects of the food relevant iAs species arsenite and arsenate are evaluated. In addition, potential genotoxic effects of their major human metabolites (see Table 1 in Section 1.2.1 on Chemistry for a list of compounds covered) are assessed. The risks of iAs in food are characterised using the scientific report on chronic dietary exposure to arsenic (EFSA, 2021).

TABLE 1

Inorganic arsenic species and their major human metabolites relevant for the present assessment.

Name a AbbreviationChemical structure b
Inorganic arsenic (sum of arsenite and arsenate)
ArseniteiAs(III)As(O)3
ArsenateiAs(V)O = As(O)3
Methylarsonate, monomethylarsonic acidMMA(V)CH3AsO(OH)2
Methylarsonite, monomethylarsonous acidMMA(III)CH3As(OH)2
Dimethylarsinate, dimethylarsinic acid c DMA(V)(CH3)2AsO(OH)
Thio‐dimethylarsinate, thio‐dimethylarsinic acid, dimethylmonothioarsinateThio‐DMA(V), DMMTA(V), DMTA(V)(CH3)2AsS(OH)
Dimethylarsinite, dimethylarsinous acidDMA(III)(CH3)2AsOH
MMA (sum of MMA(III) and MMA(V))
DMA (sum of DMA(III) and DMA(V))
a The names for the fully protonated forms (monomethylarsonic acid and dimethylarsinic acid) are often used in the literature although the species are usually measured as the anion, namely monomethylarsonate or dimethylarsinate).
b Arsenic species are drawn in their fully protonated form.
c Also named cacodylic acid.

1.2. Additional information

1.2.1. Chemistry of inorganic arsenic relevant to its presence in food

The metalloid arsenic (atomic number, 33; relative atomic mass, 74.92) occurs in group 15 of the periodic table along with nitrogen and phosphorus. Consequently, the chemistry of arsenic is similar in many respects to that of these two elements. The most stable arsenic species found under normal environmental conditions contain the arsenic atom in oxidation state +5; under reducing conditions arsenic species can occur with the arsenic atom in oxidation state −3 and + 3.

In food and feed, arsenic is usually found in combination with other elements such as oxygen, sulfur, carbon and hydrogen. From a chemical point of view arsenic combined with oxygen and sulfur is called inorganic arsenic, whereas arsenic combined with carbon and hydrogen is referred to as organic arsenic.

Inorganic arsenic in food and feed comprises species mainly in the +3 or + 5 oxidation state, present as thio complexes or, primarily, as the oxo anions arsenite and arsenate. During sample preparation and analysis iAs bounds to thio groups in peptides or proteins in food is converted to arsenite and arsenate and hence data are nearly exclusively recorded as these two actually measured species. The sum of the quantified arsenite and arsenate in food (in mg As/kg wet weight) is often referred to as iAs. In oxygenated conditions, such as found in drinking water, the arsenic is present mainly as arsenate.

In natural water and drinking water, arsenic habitually appears in inorganic forms (arsenite and arsenate); organic arsenic forms are rare in water, as they are the result of biologically activity (EFSA, 2021).

After ingestion iAs metabolised by humans (see Section 3.1.1) by a series of reduction, methylation and conjugation processes. Arsenate, as well as human iAs metabolites relevant to this Opinion are listed in Table 1 below.

1.2.2. Analytical methods

In its previous Opinion (EFSA CONTAM Panel, 2009) the CONTAM Panel summarised that several suitable methods are available for the measurement of total arsenic and arsenic species in food and biological samples. For the determination of total arsenic following mineralisation, hydride generation atomic absorption spectrometry (HG‐AAS) and inductively coupled plasma mass spectrometry (ICP–MS) are sensitive, reliable and commonly used methods. Speciation analysis for iAs consists of three main aspects: extraction, separation and detection (e.g. Petursdottir et al., 2015). Inorganic arsenic species are frequently extracted with methanol, water or a mixture of both. The higher extraction efficiencies observed for mildly acidic extractions might be due to acid hydrolysis causing a release of degradation products from arsenic species in the protein and lipid fractions. As an alternative the extraction procedure can be based on microwave‐assisted alkaline solubilisation of the sample. Here arsenate and arsenite are solubilised and at the same time arsenite is oxidised to arsenate allowing subsequent determination of (combined) iAs as arsenate by anion exchange high‐performance liquid chromatography (IEX‐HPLC). Concentrations of iAs in food are generally presented in mg As/kg wet weight. After each extraction method separation by anion exchange columns are routinely applied to efficiently separate iAs from DMA and MMA. Due to excellent compatibility with ion exchange HPLC, excellent detection limits and linear range capable of quantifying low ng/L as well as high mg/L concentrations in the same run ICP–MS is the detection technique most widely used. Since arsenic has only one isotope, isotope dilution methods cannot be applied to improve detection specificity. However, ICP–tandem mass spectrometer is increasingly applied combining high sensitivity with high specificity. Here arsenic is detected in the MS/MS mode with oxygen introduced into the collision/reaction cell to shift As+ from mass 75 to mass 91 through the formation of 75As16O+ in the cell (e.g. Stetson et al., 2020).

1.2.3. Previous assessments

In 2009, the EFSA CONTAM Panel assessed the risks to human health related to the presence of arsenic in food (EFSA CONTAM Panel, 2009). The Panel noted that the main adverse effects reported to be associated with long‐term ingestion of iAs in humans are skin lesions, cancer, developmental toxicity, neurotoxicity, cardiovascular diseases, abnormal glucose metabolism and diabetes. A provisional tolerable weekly intake (PTWI) of 15 μg/kg bw was established by the Joint FAO/WHO Expert Committee on Food Additives (JECFA) in 1983 (FAO/WHO, 1983). Since then, data have shown that oral exposure to iAs causes cancers of the lung and urinary bladder in addition to skin, and a range of other adverse effects had also been reported at exposures lower than in the studies reviewed by JECFA. Therefore, the CONTAM Panel concluded that the JECFA PTWI was no longer appropriate. The CONTAM Panel identified cancers of the urinary bladder, lung and skin, and skin lesions, as the most relevant endpoints for providing an appropriate Reference Point to be used in the risk characterisation. Dose–response data from key epidemiological studies were modelled using a benchmark response of 1% extra risk. A limitation in all the available studies was that total dietary exposure to iAs was not measured, and in most studies the concentration of arsenic in drinking water was used as the exposure metric. A range of benchmark dose lower confidence limit (BMDL01) values between 0.3 and 8 μg/kg bw per day was identified for cancers of the lung, skin and bladder, as well as for skin lesions. Since dietary exposure to iAs was within the range of the BMDL01 values, the possibility of a risk to some consumers could not be excluded. In 2009, the CONTAM Panel also evaluated organic forms of arsenic, concluding that arsenobetaine is not of toxicological concern and that arsenosugars, arsenolipids, methylarsonate and dimethylarsinate could not be considered in the risk characterisation, because of the lack of data.

In 2011, the JECFA reviewed its PTWI of 15 μg/kg bw set in 1983 for iAs (FAO/WHO, 2011). The JECFA noted that the quantitative assessment of cancer risk from iAs is limited, for instance because of the lack of information on total exposure in epidemiological studies. The JECFA calculated a BMDL for a 0.5% increased incidence of lung cancer seen in an epidemiological study (Chen, Chiou, Hsu, Hsueh, Wu, & Chen, 2010a) using several assumptions to estimate exposure from drinking‐water and food with differing concentrations of iAs. A BMDL0.5 of 3.0 μg/kg bw per day (range of 2.0–7.0 μg/kg bw per day) for lung cancer based on the range of estimated total dietary exposure was calculated. The Committee noted that since the PTWI of 15 μg/kg bw established previously was in the region of the BMDL0.5, it was no longer appropriate, and in consequence the PTWI was withdrawn.

In 2012, the International Agency for Research on Cancer (IARC) assessed the carcinogenicity of arsenic and arsenic compounds (IARC, 2012). They noted that trivalent arsenicals do not react directly with DNA, but cause oxidative DNA damage, DNA strand breaks and/or alkali labile sites. Pentavalent arsenic causes DNA strand breaks and DNA‐protein crosslinks attributed to the dimethylarsinic (DMA) peroxy radical. Other possible modes of action include induction of genomic instability, probably via reactive oxygen species (ROS), inhibition of DNA‐repair and co‐mutagenesis with other compounds. The IARC concluded that there was sufficient evidence in humans for the carcinogenicity of mixed exposure to iAs compounds, including arsenic trioxide, arsenite and arsenate, which cause cancer of the lung, urinary bladder and skin. Arsenic and iAs compounds were also associated with cancer of the kidney, liver and prostate. There was sufficient evidence in experimental animals for the carcinogenicity of dimethylarsinic acid, calcium arsenate, sodium arsenite and limited evidence in experimental animals for the carcinogenicity of sodium arsenate, gallium arsenide, arsenic trioxide and trimethylarsine oxide. There was inadequate evidence in experimental animals for the carcinogenicity of monomethylarsonic acid and arsenic trisulfide. Overall, there was sufficient evidence in experimental animals for the carcinogenicity of iAs compounds. Taking into account all of the evidence the IARC classified arsenic and inorganic arsenic compounds as carcinogenic to humans (Group 1), dimethylarsinic acid and monomethylarsonic acid as possibly carcinogenic to humans (Group 2B), and arsenobetaine and other organic arsenic compounds not metabolised in humans, as not classifiable as to their carcinogenicity to humans (Group 3). The IARC noted that elemental arsenic and inorganic arsenic species share the same metabolic pathway namely: arsenate → arsenite → methylarsonate → dimethylarsenite. Thus, independent of the mechanisms of action and the metabolite being the actual carcinogen, different iAs species should be considered as carcinogenic.

In 2015, the German Federal Institute for Risk Assessment evaluated the risks for consumers related to arsenic in rice and rice products (BfR, 2014). The estimated exposures of the different age groups were compared with the lower value of the BMDL01 value range of 0.3–8 μg/kg bw per day calculated for iAs by EFSA (EFSA CONTAM Panel, 2009) to characterise possible risks. Comparing chronic exposure estimates of adults with the EFSA BMDL01 value resulted in MOEs of 37–1000 (average consumption) and 12–320 (95th percentile) for the adult population. For adult consumers only, the MOEs ranged from 11 (average) to 889 (95th percentile). For children aged over 0.5 years, MOEs of 9 to 500 and 2–143 were calculated at the mean and 95th percentile, respectively. For children aged from 0.5 to 1 year, MOE values ranged from 1 to 22. Based on the outcome of the assessment, the BfR concluded that health impairments regarding the risk of cancer are possible and that iAs in food should be reduced to the unavoidable minimum.

In 2016, the UK Committee on Toxicity of Chemicals in Food, Consumer Products and the Environment (COT, 2016) published a statement on potential risks from arsenic in the diet of infants aged 0 to 12 months and children aged 1 to 5 years. The COT noted that the EFSA had calculated BMDL01 values ranging from 0.3 to 8.0 μg/kg bw per day based on cancers of the lung, skin and urinary bladder, and on skin lesions (EFSA CONTAM Panel, 2009), while the JECFA established a BMDL of 3.0 μg/kg bw per day for a 0.5% increased incidence of lung cancer (FAO/WHO, 2011). The COT concluded that the JECFA BMDL should be used in the risk characterisation because it was based on more robust and recent data than that available to EFSA. Taking into account the quality of the key data, the COT concluded that an MOE of 10 or more compared to the JECFA BMDL would be considered as a low health concern.

In 2016, the U.S. Food and Drug Administration (FDA) issued an assessment of health risks from iAs in rice and rice products for public comment (US FDA, 2016). The quantitative estimates of cancer risk focused on lung and bladder cancer, which provided the best evidence of cancer at relatively low levels of exposure. The predicted cancer risks attributed to lifetime consumption of arsenic in rice and rice products was 39 cases per million people (10 cases of bladder, 29 of lung cancer) as compared to combined lung and bladder cases from all causes over a lifetime of 90,000 cases per million people. The modelling predicted that risk increases almost proportionally with daily rice servings and if the amount of rice would be increased by one serving daily, lifetime cancer risk would increase to 74–183 cases per million people, depending on the type of rice consumed. Eliminating rice and rice products from the diets of infants and children (until age 6) could reduce lifetime cancer risk by 6% and 23%, respectively. The FDA also noted evidence that fetuses may have increased susceptibility to adverse effects via maternal uptake of iAs. Exposure during infancy and early childhood could have neurotoxic effects, although it was not clear if these effects would be lasting.

In 2021, the EFSA again assessed chronic dietary exposure to iAs (EFSA, 2021). The results from this assessment are used for the present Opinion and are described in detail in Section 3.4.1 on Occurrence data used in the present assessment and Section 3.5.1 Exposure assessment used for the present Opinion.

1.2.4. Legislation

Council Regulation (EEC) No 315/933 3 stipulates that food containing a contaminant in an amount unacceptable for public health shall not be placed on the market, that contaminant levels should be kept as low as can reasonably be achieved and that, if necessary, the European Commission may establish maximum levels (MLs) for specific contaminants. These maximum levels are laid down in the Annex I of Commission Regulation (EU) 2023/915. 4 Table 2 presents an excerpt of the table where the MLs for iAs as listed in Annex I to Regulation 2023/915.

TABLE 2

Maximum levels for inorganic arsenic in food as laid down in Regulation (EU) No 2023/915.

3.4ArsenicMaximum level (mg/kg)Remarks
Inorganic arsenic (sum of As(III) and As(V))The maximum level for inorganic arsenic applies to products listed in 3.4.1–3.4.4
3.4.1Cereals and cereal based productsRice, husked rice, milled rice and parboiled rice as defined in Codex Standard 198–1995
3.4.1.1Non‐parboiled milled rice (polished or white rice)0.15
3.4.1.2Parboiled rice and husked rice0.25
3.4.1.3Rice flour0.25
3.4.1.4Rice waffles, rice wafers, rice crackers, rice cakes, rice flakes and popped breakfast rice0.30
3.4.1.5Rice destined for the production of food for infants and young children a 0.10
3.4.1.6Non‐alcoholic rice‐based drinks0.030
3.4.2Infant formulae, follow‐on formulae and food for special medical purposes intended for infants and young children a and young child formulae b The maximum level applies to the product as placed on the market
3.4.2.1Placed on the market as powder0.020
3.4.2.2Placed on the market as liquid0.010
3.4.3Baby food a 0.020The maximum level applies to the product as placed on the market
3.4.4Fruit juices, concentrated fruit juices as reconstituted and fruit nectars c 0.020
Total arsenic The maximum level for total arsenic applies to products listed in 3.4.5
3.4.5Salt0.50
a Food as defined in Article 2 of Regulation (EU) No 609/2013 of the European Parliament and of the Council of 12 June 2013 on food intended for infants and young children, food for special medical purposes and total diet replacement for weight control and repealing Council Directive 92/52/EEC, Commission Directives 96/8/EC, 1999/21/EC, 2006/125/EC and 2006/141/EC, Directive 2009/39/EC of the European Parliament and of the Council and Commission Regulations (EC) No 41/2009 and (EC) No 953/2009. OJ L181, 29.6.2013, p. 35.
b ‘Young‐child formulae’ refers to milk‐based drinks and similar protein‐based products intended for young children. These products are outside the scope of Regulation (EU) No 609/2013 (Report from the Commission to the European Parliament and the Council on young‐child formulae (COM(2016) 169 final) https://eur‐lex.europa.eu/legal‐content/EN/TXT/?uri=CELEX%3A52016DC0169&qid=1620902871447).
c Food as defined in Council Directive 2001/112/EC of 20 December 2001 relating to fruit juices and certain similar products intended for human consumption. OJ L 10, 12.1.2002, p. 58.

Harmonised requirements for arsenic in drinking water are set by Council Directive 98/83/EC 5 on the quality of water intended for human consumption and by the new Directive (EU) 2020/2184 6 of the European Parliament and of the Council on the quality of water for human consumption, which entered into force on 12 January 2022 and which will repeal Council Directive 98/83/EC from 13 January 2023.

Both directives stipulate that Member States shall set limit values of 10 μg/L for arsenic in water intended for human consumption.

In Commission Directive 2003/40/EC 7 establishing the list, concentration limits and labelling requirements for the constituents of natural mineral waters and the conditions for using ozone‐enriched air for the treatment of natural mineral waters and spring waters, a maximum limit of 0.010 mg/L is established for total arsenic.

The Codex Alimentarius Commission (Codex) has adopted maximum levels for iAs of 0.2 mg/kg in polished rice 8 and of 0.35 mg/kg in husked rice (paddy rice from which the husk only has been removed, also known as brown rice or cargo rice). 9

2. DATA AND METHODOLOGIES

The current update of the EFSA risk assessment on iAs, was developed applying a structured methodological approach, which implied developing a priori the protocol or strategy of the full risk assessment. The protocol in Annex A of this Opinion contains the method that was proposed for covering all the steps of the risk assessment process.

The CONTAM Panel used its previous risk assessment on iAs in food (EFSA CONTAM Panel, 2009) as a starting point for drafting the current Opinion.

2.1. Collection and appraisal of data collected from public literature

2.1.1. Collection of data

Several comprehensive literature searches were conducted to identify scientific literature relevant to the assessment. Web of Science (Core collection: Science Citation, Index, Emerging Sources Citation Index, Current Chemical Reactions, Index Chemicus) 10 and Pubmed (NLM) 11 were identified as sufficient and appropriate to serve as sources of information for literature for the present assessment. The structure, syntax and terms used for the searches have been designed and conducted by the Working Group (WG) with the support of an information specialist. Controlled vocabulary, when available, (i.e. MeSH) and natural vocabulary were used to represent the key elements of the searches. The language of the original studies has been limited to English. The obtained references were imported to EndNote (Clarivate Analytics). 12 Deduplication was performed using automatic and manual procedures. Since this Scientific Opinion is an update of the Scientific Opinion of EFSA on arsenic in 2009, the literature search was restricted to papers published from 1 January 2009 onwards. In addition, a search for previous assessments (risk and exposure assessments of governmental and international bodies) was carried out. During the development of the Opinion, additional publications for the different sections were collected by applying a ‘snowballing approach’ 13 and considered for the assessment where relevant.

Finally, a public consultation on the draft Opinion was carried out from 24 July 2023 to 10 September 2023 and the comments and documents were considered for finalising the Opinion.

The first literature search was carried out on 14 of April 2021 to identify papers on the following key topics:

  1. ADME: Arsenic AND ADME AND (Humans OR Animals) AND Reviews
  2. Toxicity:
    • Arsenic AND (Toxicity AND Animal)
    • Arsenic AND (Cytotoxicity OR Genotoxicity) AND (In vitro OR In vivo)
  3. Epidemiological studies: Arsenic AND Epidemiological studies AND Humans

ADME results have been limited to review papers as this was deemed sufficient to address the terms of reference. For all other sections primary research was considered as well. The period covered in this search was 1st of January 2010 to 14th of April 2021. The search strings for each database are described in detail in Table B.1 of Annex B. The number or results of the searches are reported in Table 3 below.

TABLE 3

Number of hits of the search on key topics covering the period of 2010–2021.

SearchPubMedWeb of sciencePubMed and web of science combined after de‐duplication
ADME 701434858
Toxicity 407934304536
Epidemiological studies 4,468 28224870

Abbreviation: ADME, Absorption, Distribution, Metabolism and Excretion.

An additional search was performed in September 2021 to identify papers on epigenetic and transgenerational effects of arsenic not sufficiently covered with the search terms previously applied. This was done as the WG agreed the search terms applied in the first overall search were insufficient to cover all publications relevant for this field. The search was conducted covering the period of 1st of January 2010 to 20th of September 2021. The search string is reported in Annex B in Table B.2. The number of results of the searches is reported in Table 4 below.

TABLE 4

Number of hits of the search on epigenetic and transgenerational covering the period of 2010–2021.

SearchPubMedWeb of sciencePubMed and web of science combined after de‐duplication
Epigenetic and transgenerational effects 54178169

The first comprehensive literature search carried out on 14th April 2021 was repeated on 10 December 2021 to cover also the publication period of 1st of January 2009 to 31st of December 2009 (not covered in the first literature search). It was repeated again on 15 July 2022 to cover also the period 14th of April 2021 to 18th of July 2022. Notably, the key topics of these two repeated searches were the same except that only studies related to epidemiology were considered (search terms and key topics of these searches are therefore not presented in the Annex B). This is because it was agreed at the 119th CONTAM Panel that for the risk characterisation of iAs animal data do not need to be considered. This was decided because of the known toxicokinetic differences between laboratory animals and humans which limit their use for assessing human health effects of iAs (EFSA CONTAM Panel, 2009). The abundance of human data available together with the notion that for human health risk assessment human data should be used preferably were further arguments for basing crucial parts of the assessment on human data only.

The results of the two repeated searches are reported in Tables 5 and and66 below.

TABLE 5

Number of hits of the search on epidemiological studies covering the period of 2009–2010.

SearchPubMedWeb of sciencePubMed and web of science combined after de‐duplication
Epidemiological studies 125277170

TABLE 6

Number of hits of the search on epidemiological studies covering the period of 2021–2022.

SearchPubMedWeb of sciencePubMed and web of science combined after de‐duplication
Epidemiological studies 563465727

2.1.2. Evaluation of data

For the pre‐evaluation, the scientific literature obtained was screened based on title and abstract, applying expert judgement. Publications obviously irrelevant for the present mandate were excluded from further consideration. These were publications for example dealing with the use of arsenic in medical or technical applications, publications on arsenic in geology, effect on arsenic on plants, wild animals or ecosystems or publications where arsenic was used as a compound to validate test systems. From the first search the pre‐evaluation resulted in a total of 4539 potentially relevant studies (human and animal). From these only human data, and animal data on carcinogenicity, genotoxicity and epigenetic changes and in vitro data on genotoxicity and epigenetics were considered further (see Section 2.1.1 above). Publications dealing with arsenic trioxide were also not further considered because of the difference in the toxicokinetics between arsenic trioxide and arsenic present in food and drinking water. Human studies covering mixtures of arsenic and other contaminants or compounds potentially ameliorating arsenic mediated effects (which were in particular intervention studies) were also excluded from analysis as these were considered not relevant for the risk characterisation of iAs. In the updated search on key topics covering the period 2009–2010, 81 studies were considered possibly relevant in the pre‐evaluation. In the updated search on key topics covering the period 14th of April 2021 to 18th of July 2022, 160 studies were considered possibly relevant after the pre‐evaluation. For the search on epigenetic and transgenerational effects carried out in 2021, the pre‐evaluation by experts resulted in 64 possibly relevant papers on the specific topic. For the final evaluation and refinement of the scientific literature to be considered for the assessment, the abstracts were again assessed and evaluated by relevant domain experts from the CONTAM working group.

2.1.3. Transformation of data from epidemiological studies and benchmark dose calculations

The transformation of data from epidemiological studies for use in benchmark dose (BMD) assessments is described in detail in Section 3.2.4 Dose–response analysis.

2.1.4. Overview of methodology applied

Figure 1 provides an overview of the methodology applied for the present risk assessment. After collection and evaluation of the literature (see Sections 2.1.1 and 2.1.2) selection criteria were applied, after which causal association was scrutinised, the suitability for BMD modelling was evaluated after which dose response modelling was carried out. Before conducting BMD modelling the relative risk estimates were transformed to quantal data. Additionally, the iAs concentrations were converted to exposures, taking into account default values and estimating source populations for case–control studies. This was followed by a risk of bias analysis of the studies from which the most relevant valid BMDL could be derived. Finally, a Reference Point based on the BMD modelling, expert knowledge and an MOE to estimated exposure levels in Europe were established.

An external file that holds a picture, illustration, etc.
Object name is EFS2-22-e8488-g001.jpg

Flowchart on the methodology applied for risk assessment.

3. ASSESSMENT

3.1. Hazard identification and characterisation

3.1.1. Toxicokinetics

This chapter gives a short summary of the crucial steps in the toxicokinetics of iAs upon ingestion by humans. A more detailed review can be found in the previous CONTAM Opinion on arsenic (EFSA CONTAM Panel, 2009) as well as recent reviews (e.g. Roy et al., 2020; Thomas, 2021).

In humans, soluble iAs is rapidly and well absorbed after ingestion (45%–80%). After absorption, arsenic is widely distributed to almost all organs and readily crosses the placental barrier. In blood arsenic is distributed between plasma and erythrocytes. Here it is mainly bound to SH‐groups in proteins and low‐molecular‐weight compounds such as glutathione (GSH) and cysteine. The major metabolic pathways of iAs in humans include oxidation, reduction, methylation, thiolation and glutathiolation of arsenic. Methylation capacity is crucial for arsenic toxicity. Elimination through urine in the form of iAs and its methylated metabolites occurs with a half‐life of around 2–3 days (Buchet et al., 1981b). Average values for the major urinary species following iAs intake are 10%–30% iAs (sum of arsenite and arsenate), 10%–20% MMA (sum of MMA(III) and MMA(V)) and 55%–75% DMA (sum of DMA(III) and DMA (V)). Notably there is high inter‐species, inter‐population and inter‐individual variability for arsenic methylation and also other aspects of toxicokinetics. Susceptibility factors include life stage, sex, nutritional status (e.g. Kurzius‐Spencer et al., 2017; López‐Carrillo et al., 2016; Xu, Drobná, et al., 2016), microbiota (e.g. Coryell et al., 2019) and genetic polymorphisms related to arsenic metabolism genes and transporter genes (e.g. EFSA CONTAM Panel, 2009; Schuhmacher–Wolz et al., 2009). Most studies concentrate on the polymorphisms of arsenite‐methyltransferase (AS3MT) and glutathione‐S‐transferases (GST) especially GSTO1 (glutathione S‐transferase omega 1) (e.g. Antonelli et al., 2014; Caceres et al., 2010; De Loma et al., 2018; Engström et al., 2011; Fu et al., 2014; Gao et al., 2015; González‐Martínez et al., 2018).

3.1.1.1. Metabolism

The metabolism of iAs in the humans has been proposed through three different, likely interlinked, pathways (Figure 2). The classical pathway suggests a process of sequential reduction and oxidative methylation steps. Following ingestion, the first step of metabolism is a reduction of As(V) to As(III): 50%–70% of arsenate is rapidly reduced to arsenite (most likely facilitated by GSH) in blood and the liver (Aposhian et al., 2004; EFSA CONTAM Panel, 2009; Rehman & Naranmandura, 2012). Then As(III) undergoes a cycle of oxidative methylation reactions and reduction reactions that sequentially couple a methyl group to an As atom in the trivalent form (Figure 2A). Methylation takes place mainly in the liver, although most organs are capable of methylating arsenic (e.g. bladder, kidneys). Arsenite methyltransferase (AS3MT) catalyses the transfer of the methyl group from S‐Adenosylmethionine (SAM) to arsenite and MMA(III), resulting in MMA(V) and DMA(V).

An external file that holds a picture, illustration, etc.
Object name is EFS2-22-e8488-g002.jpg

Simplified schemes of the originally proposed human iAs metabolism pathway (A), the two more recently proposed alternative pathways (B, C) as well as a possible conjunction of the pathways. ATG, Arsenic triglutathione; MMA(V), monomethylarsonic acid; MMA(III), monomethylarsonous acid; DMA(V), dimethylarsinic acid); DMA(III), dimethylarsinous acid; MMAG, monomethylarsonic diglutathione; DMAG, dimethylarsinic glutathione; thio‐DMA(V), thio‐dimethylarsinic acid; GSH, glutathion‐S‐transferases; AS3MT, arsenite methyltransferase; SAM, S‐Adenosylmethionine; SAH, S‐Adenosylhomocysteine. Scheme modified from Hartwig and Schwerdtle (2009), © Blackwell Publishing Ltd/Wiley.

The second proposed pathway suggests that arsenic either binds to certain proteins (Figure 2C) or conjugates with glutathione (Figure 2B) and these intermediates are subsequently methylated by AS3MT. Arsenic triglutathione (ATG) is methylated to monomethylarsonic diglutathione (MMAG) and dimethylarsinic glutathione (DMAG). MMAG and DMAG are further reduced to MMA(III) and DMA(III) and then oxidised to the respective pentavalent forms.

Thio‐DMA(V) is the pentavalent sulfur analogue of DMA(V) and a metabolite of organic as well as inorganic arsenicals. Thio‐DMA(V) was first identified as a mammalian arsenic metabolite in urine from a sheep naturally consuming large amounts of arsenosugars through seaweed (Hansen et al., 2004). It has been discussed that thio‐DMA(V) may have been misidentified as DMA(III) in human urine samples before and, therefore, might have escaped detection in many samples to date (Hansen et al., 2004). Indeed, thio‐DMA(V) has later been identified a later study in human urine after exposure to iAs‐contaminated drinking water (Raml et al., 2007). In this study investigating the urine of 75 inorganic arsenic‐exposed women in Bangladesh, thio‐DMA(V) has been shown to be a common metabolite, being detected in 44% of the samples. Although thio‐DMA(V) has been identified as human iAs metabolite the metabolic mechanism of the formation is not fully understood so far. Like DMA and MMA, thio‐DMA(V) also directly occurs in food, e.g. in rice.

3.1.1.2. Interspecies variability in inorganic arsenic toxicokinetics

In 2009, the CONTAM Panel concluded that experimental animals differ considerably from humans with regard to iAs metabolism and other aspects of toxicokinetics. Thus, the results of toxicity studies in animals do not provide a suitable basis for hazard assessment (EFSA CONTAM Panel, 2009).

This paragraph highlights briefly important interspecies differences in iAs toxicokinetics.

Animals differ in their ability to methylate inorganic As (Vahter, 1999). Additionally, Vahter (1999) showed that the methylated metabolites are produced in different ratios in humans compared to most mammals and that humans excrete more MMA(V). Marmoset monkeys do not methylate iAs, whereas rodents (hamsters, mice), rabbits and dogs show high methylating capacity, reaching up to 80% urinary DMA(V). The lower urinary excretion of DMA in rats is not an indication of low iAs methylation capacity but results from the specific retention of DMA(V) in rat erythrocytes. Primary rat hepatocytes have been shown to methylate iAs better than cultured primary human hepatocytes and keratinocytes (Styblo et al., 2000).

Since 2009, no toxicokinetic studies contradicting the previous conclusions have been identified. Thus, the conclusion still stands that results of toxicity studies in animals do not provide a suitable basis to assess the risks to human health related to iAs ingestion because of considerable interspecies differences in the toxicokinetics of arsenic.

3.1.1.3. Transfer to human milk

In contrast to the rapid transfer of As to the fetus, little As is excreted in human milk following iAs ingestion (EFSA, 2021; EFSA CONTAM Panel, 2009). A small study reported median levels around 0.31 μg/L of tAs in US mothers, with a maximum of 0.62 μg/L and detectable arsenic in only five of the nine samples of breast milk analysed. Arsenic exposure of the mothers was not assessed (Carignan et al., 2015). The average concentration of arsenic in human milk of 60 Swedish mothers was 0.55 μg/L (SD = 0.70 μg/L) with the highest concentration measured being 4.6 μg/L (Björklund et al., 2012). Arsenic concentration in human milk was positively correlated with fish consumption, but external arsenic exposure was not assessed. Speciation analysis of milk from this study showed no content of inorganic As or its methylated metabolites in the human milk. Other studies have shown that following the consumption of fish, a small percentage of arsenolipids are transported to the milk of nursing women (Stiboller et al., 2017; Xiong et al., 2020).

Different studies support the assumption that even in areas with drinking water containing relatively high to moderate concentrations of arsenic, only small amounts appear to pass through mammary glands to human milk (Carignan et al., 2015; Concha, Vogler, Lezcano, et al., 1998; Concha, Vogler, Nermell, & Vahter, 1998; Signes‐Pastor et al., 2018). Overall, the available studies indicate that after ingestion of inorganic arsenic, little arsenic is transferred to human milk.

3.1.2. Biomarkers

3.1.2.1. Biomarkers of exposure

In its previous Opinion (EFSA CONTAM Panel, 2009) the CONTAM Panel noted that total arsenic in urine is a common biomarker of arsenic exposure as most arsenic compounds originating from oral arsenic intake of food and water are excreted via urine within a few days. Urine collected over 24 h is preferred for biomonitoring of arsenic. However, because of difficulties in obtaining 24‐h urine samples, first‐morning or spot urine samples are usually used for biomonitoring of arsenic. Fish and other seafood can contain relatively high concentrations of arsenobetaine and other organic arsenic compounds, which are considered much less toxic than iAs and are also excreted in the urine (Taylor, Li, et al., 2017). Consequently, consumption of even small amounts of seafood can result in high urinary concentrations of total arsenic making it difficult to assess iAs exposure. Total urinary arsenic is therefore often not a suitable biomarker of exposure. Specific measurements of iAs, and its methylated metabolites (e.g. MMA and DMA), or the sum of iAs and its methylated metabolites, also named total urinary inorganic arsenic (u‐tiAs), provide more appropriate estimates of exposure (e.g. Middleton et al., 2016). In urine samples where arsenobetaine levels are low (e.g. arsenic intake is not from fish consumption), DMA typically contributes to > 50% of total arsenic in the urine (Buchet et al., 1981a; Colín‐Torres et al., 2014; Cubadda et al., 2012; Fillol et al., 2010; Tam et al., 1979). However, the sum of iAs and related metabolites (u‐tiAs) might be affected by ingestion of DMA which can occur as such in food including rice but also by ingestion of arsenosugars via seaweed, which can be metabolised into DMA and other methylated arsenic compounds compromising the assessment of iAs exposure (Taylor, Li, et al., 2017). Thus, study participants need to refrain from consumption of all seafood for several days before sampling to achieve reliable results. Urinary arsenic concentrations are often adjusted to creatine to control for variations in urine flow rates. This may lead to incorrect conclusions because of differences in muscle mass between individuals and, in particular, between women and men (Nermell et al., 2007). Therefore, it has been proposed to normalise to the specific gravity of the urine rather than adjusting for creatinine levels (Nermell et al., 2007).

Levels of total arsenic in blood are also of limited value when estimating exposure to iAs because it is cleared from the blood within a few days after ingestion (Buchet et al., 1981a; Middleton et al., 2016). The exception to this is where there is constant exposure to arsenic, for example by using the same water supply or where rice is a staple food in the diet (Meharg et al., 2014; Signes‐Pastor, Gutiérrez‐González, et al., 2021).

Under conditions of constant high exposure to iAs, the concentration of total arsenic in the blood reaches a steady state, better reflecting exposure to iAs although diurnal variation also exists (Signes‐Pastor, Gutiérrez‐González, et al., 2021). High levels of iAs in the drinking water correlate with elevated total arsenic concentrations in blood (Arikan et al., 2015; Rodrigues et al., 2015). Unlike urinary arsenic, which reflects excretion, arsenic in blood is a measure of internal exposure and thus reflects actual tissue burdens. However, blood arsenic concentration also correlates positively with frequency of fish consumption, making it unsuitable as a biomarker of exposure to iAs (Miklavcic et al., 2013; Snoj Tratnik et al., 2019).

Inorganic arsenic is the major form in hair and nails with MMA and DMA combined making up < 20% of total arsenic in this biomatrix. Because trivalent arsenicals have high affinity for sulfhydryl groups they bind to keratin in hair and nails (Shen et al., 2013). Inorganic arsenic therefore tends to accumulate in hair and nails and because they grow slowly, arsenic concentrations in hair and nails are considered reasonable biomarkers for long‐term exposure to iAs (Signes‐Pastor, Gutiérrez‐González, et al., 2021).

The CONTAM Panel reviewed the literature published since the previous Opinion (EFSA CONTAM Panel, 2009) and concluded that there is no new information that alters the view that there is no universally accepted biomarker for chronic iAs exposure. If continuous iAs exposure is stable over time, urinary arsenic may be used as a biomarker provided that arsenic species or u‐tiAs are analysed. Hair and, in particular, toenails are probably the best biomarkers of long‐term exposure to iAs. However, also these are imperfect because arsenic in hair and nails may not exclusively originate from iAs and there are studies showing a correlation between seafood consumption and total arsenic in hair and nails (Taylor, Goodale, et al., 2017).

3.1.2.2. Markers of genotoxicity

Since the previous Opinion, additional biomonitoring studies investigating genotoxic damage in cells from individuals either occupationally or environmentally exposed to arsenic have been published using different markers. The marker most frequently investigated was the frequency of micronuclei (MN) in peripheral blood lymphocytes. In this Section only relevant studies conducted on populations exposed orally to iAs via an environmental source were reviewed. Details of the studies are presented in Table 7.

TABLE 7

Markers of genotoxicity in cells from exposed individuals.

ReferenceTest systemPopulation size (n) case/controlAs exposure (mean)ResultsAdditional information
Banerjee et al. (2010)

CA in lymphocytes

Serum oxidative stress enzymes activities

50 exposed (34 with skin lesions; 16 without)

41 unexposed from West Bengal (India)

Concentration in water (μg/L): exposed, 218.17; unexposed, 6.92; chronic exposure: more than 10 years

Significantly higher incidence of CA in exposed versus unexposed individuals

Catalase and myeloperoxidase activity higher in exposed versus unexposed individuals at low levels of exposure

Levels of As measured in urine, nails and hair
Bartolotta et al. (2011)MN assay in buccal cells27 exposed to high levels, 32 exposed to low levels from rural and urban areas of ArgentinaConcentration in water (μg/L): exposed to high levels > 50 versus exposed to low levels < 50, serving as controlsSignificant increase of MN frequency in the exposed group of both rural and urban populations versus controlsNo effect of age and gender
Sampayo‐Reyes et al. (2010)Alkaline comet assay in peripheral blood leukocytes84 exposed (adults and children): low exposed (n = 42); medium exposed (n = 26); high exposed (n = 19) from Northern Mexico

Concentration in water (μg/L)

Low exposed, 12.1; medium exposed, 16; high exposed, 45.6

A positive association was found between the level of exposure and DNA breaks (% of DNA in tail)

Correlation of the total urinary As content of the exposed individuals with the As content in drinking water

Influence of AS3MT Met287Thr polymorphism on DNA break induction

Banerjee et al. (2013)MN assay in urothelial cells

304 exposed

113 unexposed from West Bengal (India)

Concentration in cooked rice (mg/kg): exposed from > 100 up to > 300; unexposed ≤ 100; not significantly exposed to arsenic through drinking waterSignificantly higher MN frequency in all groups with a mean As concentration in cooked rice > 200 μg/kg versus lower exposure group (≤ 100–≤ 200)

Strong correlation between grouped urinary As and cooked rice As data

Information on possible confounding factors available

Hinhumpatch et al. (2013)

8‐oxodG in salivary DNA and urines by HPLC

hOGG1 gene expression by RT‐PCR in saliva

Children: 40 exposed/20 unexposed from Ron Phibul District (Thailand)

Concentration in water (μg/L): drinking water: exposed 5.66/unexposed 0.4;

Non‐drinking water: exposed 39.96/unexposed 2.97;

In utero and continuous exposure

Significantly higher levels of salivary 8‐oxodG in exposed children; levels of urinary 8‐oxodG excretion and salivary hOGG1 expression significantly lower in exposed childrenAnalysis of As in drinking water, saliva, nails and urines. Information on possible confounding factors available
Paul, Das, et al. (2013)MN assay in urothelial cells and lymphocytes189 arsenicosis/171 unexposed from West Bengal (India) recruited at two time points, (2005–06 and 2010–11)

Concentration in water (μg/L):

Exposed: 190.1 (2005–2006)/37.94 (2010–2011)

Unexposed: < 10

Decrease of As exposure resulted in significant decline of MN frequency in urothelial cells and lymphocytes

No changes of MN frequency in unexposed individuals at the two recruitment times

Decrease of As exposure resulted in significant decline in the number of individuals having dermatological disorders and in the severity of each dermatological outcome
Paul, Bhattacharjee, et al. (2013)MN assay in urothelial cells145 exposed/60 unexposed (2004–2005); 128 exposed/54 unexposed (2010–2011)Concentration in water (μg/L): 348.23 (2004–2005)/ 5.60 (2010–2011)

Decrease of As exposure resulted in a significant decline in the MN frequency

No changes of MN frequency in unexposed individuals at the two recruitment times

Pei et al. (2013)8‐oxodG by Elisa in urines and leukocytes75 exposed/12 unexposed from ChinaConcentration in water (μg/L): exposed from 27.2 to 35.6 from normal to severe skin lesionsChronic exposure to low levels selectively induces oxidatively induced DNA damage of peripheral blood polymorphonuclear cellsUrinary As levels were increased in the severe skin lesion group compared with the normal group. Information on confounding factors available
Harper et al. (2014)

8‐oxodG in urines by Elisa

Protein carbonyls by Elisa in plasma

378 participants from BangladeshConcentration in water (μg/L): five exposure categories from < 10 to > 300None of the measures of As exposure was significantly associated with protein carbonyl or 8‐oxodG levelsWater, blood and urinary As concentrations were measured. Information on possible confounding factors available
Paul et al. (2014)MN assay in lymphocytes157 exposed (75 WOSL; 82 WSL)/ 88 unexposed individuals from West Bengal (India)Concentration in water (μg/L): exposed WOSL 186.74; WSL 195.66 / unexposed: 4.37Significant increase of MN frequency in both subpopulations of exposed individuals (higher in WSL versus WOSL) versus unexposed individuals

As levels measured in water, urine, nails, hair and blood samples

Promoter hypomethylation of ERCC2 and decrease in CAK activity

Dutta et al. (2015)

Alkaline comet assay in airway cells

8‐oxodG in plasma by Elisa

142 chronically exposed women/ 131 unexposed women from West Bengal, IndiaConcentration in water (μg/L): exposed 11–50; unexposed < 10Significantly higher levels of DNA breaks (comet tail moment) in airway cells from exposed versus unexposed women. Increased levels of 8‐oxodG in plasma of exposed womenElevated inflammation markers in exposed women
Bandyopadhyay et al. (2016)MN assay in lymphocytesChildren (5–15 years): 67 exposed/49 unexposed from West Bengal (India)Concentration in water (μg/L): exposed, 45.01; unexposed, 6.22; chronic exposure: since birthMN frequency significantly higher in exposed versus unexposed individualsSignificant association of MN frequency in exposed individuals with reduced LINE‐1 methylation
Roy et al. (2016)

MN assay in exfoliated buccal cells

Comet assay in lymphocytes

138 individuals divided into four groups.

Group I (n = 54) non chewer, unexposed. Group II (n = 32) chewers, unexposed. Group III (n = 24) exposed, non‐chewers. Group IV (n = 28) chewers, exposed

Concentration in water (μg/L) exposed Group III, 43; Group IV, 43; unexposed Group I, 2 and Group II, 3Statistically significant increase of MN frequency for Group II, III and IV as compared to controls (Group I); Group IV the highest incidence of MN. DNA breaks (% of tail DNA) gradual significant increase among the groups. Strong positive correlation between the exposed population and the frequency of binucleated cellsAmount of chewing tobacco had significant positive correlation with MN frequency and in the percentage of tail DNA
Tovar‐Sanchez et al. (2016)Alkaline comet assay and CA in whole blood samples22 exposed, 20 unexposed from MexicoConcentration in water (μg/L): exposed 60, unexposed (not detected)DNA breaks (tail length) and CA were positively and significantly correlated with As concentrations in whole blood samples. Terminal deletions registered the highest determination coefficientCheck for metal concentration (Pb, As, Cu, Zn, Cd) in drinking water. As was the only metal with a significant difference in blood samples between exposed and control. Information on confounding factors available
Jiménez‐Villarreal et al. (2017)Two‐tailed comet assay in lymphocytes76 exposed, 112 unexposed from MexicoConcentration in water (μg/L): exposed 14.3/unexposed 7.7Significantly higher frequency of DSB (comet tail length) in exposed to higher levels of As versus control groupInformation on nutritional status and lifestyle variables
Phookphan et al. (2017)8‐nitroguanine by Elisa in urinesChildren (6–9 years): 40 exposed, 41 unexposed; prenatally and early childhood exposed, from ThailandConcentration in water (μg/L): 8.38 and 78.05 for drinking and non‐drinking water, respectively. Levels of As in drinking and non‐drinking water in the control sites were 12.5‐ and 13.4‐fold lowerThe level of urinary 8‐nitroguanine was significantly higher in exposed newborns and children, by 1.4‐ and 1.8‐fold, respectivelyHypomethylation of inflammatory genes (COX2, EGR1 and SOCS3)
Chatterjee et al. (2018)MN assay in buccal and urothelial cells, and lymphocytesChildren (5–15 years): 68 exposed/52 non exposed from West Bengal (India)Concentration in water (μg/L) exposed 50.8 and unexposed 6.2; chronic exposure since birthSignificantly higher MN frequency in all the three cell types in As exposed children versus unexposed childrenCorrelation between urinary As and MN frequency in lymphocytes and urothelial cells
Mar Wai et al. (2019)8 − oxodG in urines by Elisa198 pregnant women in the third trimester of pregnancy from MyanmarConcentration in water (μg/L): 0.02–198, exposure for at least 6 monthsHigher urinary As concentrations were significantly associated with higher 8 − oxodG levelsUrinary As concentrations were significantly correlated with drinking water As concentrations
Navasumrit et al. (2019)8‐oxodG, 8‐nitroguanine by immunoassays, alkaline comet assay and MN assay in cord blood205 pregnant women recruited during 2010–2012 from Haman Province, Vietnam

Concentration in toenail (μg/g): three groups were identified: low < 0.5, medium 0.5–1 and high > 1

Concentration in water (μg/L): low exposed group < 10; high exposed group ~ 60

Maternal toenail As level was significantly associated with all markers of early genetic effects. Cord blood As levels associated with DNA strand breaks (tail length, tail moment, %DNA in tail) and MN frequency

Cord blood As level significantly increased with maternal As exposure

The group of women with high exposure had a significant reduction in arsenic methylation capacity

Tsai, Kuo, et al. (2021)8‐oxodG and N7‐MeG by LC–MS/MS in urines788 participants were enrolled in a prospective cohort study in Taiwan between 1991 and 1994, with follow‐up between 2011 and 2014Concentration in well water (μg/L): low < 24.30, high > 24.30; cumulative exposure (concentration in well‐water multiplied by the duration of artesian water consumption) low < 874.2, high > 874.2The levels of 8‐oxodG and N7‐MeG have a significantly positive association with cumulative artesian well‐water As exposure and urinary As species levels after adjusting for age, sex and cigarette smokingNon‐statistically significant mediation effects of 8‐oxodG were observed. Higher risk of bladder cancer for participants with As exposure and urinary 8‐OHdG levels higher than the median

Abbreviations: 8‐OHdG, 8‐hydroxy‐2′‐deoxyguanosine; 8‐oxodG, 8‐hydroxydeoxyguanosine; As, arsenic; AS3MT, arsenic(III)methyltransferase; CA, chromosomal aberration; CAK, CDK‐activating kinase; Cd, cadmium; COX2, cytochrome c oxidase subunit II; Cu, copper; DNA, deoxyribonucleic acid; DSB, double strand break; EGR1, early growth response factor 1; ERCC2, excision repair cross‐complementation group 2; Elisa, enzyme‐linked immunosorbent assay; HPLC, high‐performance liquid chromatography; hOGG1, human 8‐oxoguanine DNA glycosylase 1; LC–MS/MS, liquid chromatography–tandem mass spectrometry; LINE‐1, long interspersed nucleotide element‐1; MN, micronucleus; n, number; N7‐MeG, N7‐methylguanine; Pb, lead; RT‐PCR, reverse transcriptase‐polymerase chain reaction; SOCS3, suppressor of cytokine signalling 3; WOSL, without skin lesions; WSL, with skin lesions; Zn, zinc.

Most biomonitoring studies have been conducted in areas containing high levels of arsenic in soils and waters because of their geological constitution and/or vicinity to mining sites. In these studies, the drinking water arsenic concentration reported for the control population (so called ‘unexposed’ individuals) is < 10 μg/L. Chronic ingestion of contaminated drinking water (arsenic concentration > 50 μg/L) in some populations, including children, from Argentina (Bartolotta et al., 2011) and India, particularly from West Bengal (Bandyopadhyay et al., 2016; Chatterjee et al., 2018; Paul et al., 2014; Paul, Bhattacharjee, et al., 2013; Paul, Das, et al., 2013) and Assam (Roy et al., 2016), was reported to be associated with significant increases of MN frequency in lymphocytes and also in urothelial (Bartolotta et al., 2011; Chatterjee et al., 2018; Paul, Bhattacharjee, et al., 2013; Paul, Das, et al., 2013) and buccal (Chatterjee et al., 2018) cells. A cross‐sectional study (Paul, Das, et al., 2013) was conducted in West Bengal where individuals with arsenicosis 14 and unexposed individuals were recruited before and after reduction in the level of exposure via drinking water. In the arsenicosis group, a five‐fold decrease in arsenic concentrations in drinking water led to a significant decline of MN frequency in urothelial cells (three‐fold decrease) and lymphocytes (2.5‐fold decrease) and of the number of individuals with dermatological disorders. In a cross‐sectional study from exposed and unexposed individuals from the same area (Paul, Bhattacharjee, et al., 2013) an ~ 60‐fold decrease in arsenic concentrations in drinking water led to a significant decline (~ three‐fold decrease) of MN frequency in urothelial cells of exposed individuals. Still there was a significant difference compared to the control population (1.3–1.5‐fold increase). In these two cross‐sectional studies the follow‐up individuals had consumed drinking water with reduced levels of arsenic for at least 5 years indicating that a partial reduction of cytogenetic damage may occur when the oral exposure decreases. Two studies investigated chromosomal aberrations (CA) in populations from West Bengal (Banerjee et al., 2010) and a mining district in Mexico (Tovar‐Sanchez et al., 2016), exposed to arsenic via drinking water, showing a significantly higher frequency of CA in lymphocytes as compared to unexposed individuals.

One study (Banerjee et al., 2013) examined the genotoxic impact of exposure to high arsenic in rice in humans. This cohort study from West Bengal (India) involved 417 subjects not otherwise significantly exposed to arsenic through drinking water (not more than 20% of total dietary exposure). Significantly higher frequencies of MN in urothelial cells were associated with consumption of cooked rice with > 200 μg/kg arsenic as staple food as compared to lower exposure groups. Several potential confounders were analysed (age, gender, tobacco usage, bodyweight, drinking water arsenic, drinking water intake and study sub‐area). The only covariates of significance in predicting MN, were cooked rice arsenic (p < 2.2 e‐16) and, to a much lower extent, tobacco usage (p = 0.022). It is of note that for this study population the authors estimated that 200 μg/kg total arsenic is equivalent to ~ 180 μg/kg iAs in rice and, on the basis of the mean cooked rice intake (560 g/day) and mean body weight of participants (51.3 kg), to a mean daily intake of iAs of ~ 2.0 μg/kg bw, raising concerns for the health risks of populations consuming rice as a staple food.

MN and CA are frequently associated with cancer and inflammation‐driven diseases (reviewed in Fenech et al., 2021). The study by Banerjee et al. (2008) was the first to provide evidence that individuals chronically exposed to arsenic via drinking water with hyperkeratosis 15 present a significantly higher frequency of CA than exposed individual without hyperkeratosis. No significant difference in the level of DNA damage as detected by the alkaline elution method was observed between the two groups but an in vitro challenge assay 16 showed that upon induction of DNA damage, the repair capacity in the exposed individuals with premalignant hyperkeratosis was significantly less than that of individuals without skin lesions. A deficiency in DNA repair was proposed as the main driver of arsenic carcinogenicity. More recently, Paul et al. (2011, 2014) confirmed these findings by showing a correlation between higher incidence of CA and higher arsenic concentration in the biological tissues (hair and nails) of patients with blood cancer from areas heavily contaminated with arsenic when compared to healthy controls from non‐contaminated areas (Paul et al., 2011) and a higher frequency of MN in exposed individuals with skin lesions as compared to subjects without skin lesions (Paul et al., 2014). These studies highlight the possibility that MN and CA formation in humans is not only a marker of induced DNA damage but also of genetic susceptibility and disease initiation and progression (Bonassi et al., 2008; Fenech et al., 2020).

Navasumrit et al. (2019) carried out a birth cohort study on 205 pregnant women residing in arsenic‐contaminated areas in Hanam province (Vietnam) to evaluate potential genotoxic effects of in utero arsenic exposure in newborns. Toenails and urine samples were collected during pregnancy and umbilical cord blood samples immediately after birth. Oxidative/nitrative DNA damage (8‐hydroxydeoxyguanosine (8‐oxodG) and 8‐nitroguanine), DNA strand breaks and MN were measured in cord blood. Evidence has been provided that cord blood arsenic concentrations reflect chronic As exposure of the fetus during pregnancy (Rodrigues et al., 2015; Guan et al., 2012). In the study by Navasumrit et al. (2019) cord blood arsenic concentration was significantly associated with that of maternal toenail arsenic suggesting an association between fetal and maternal iAs exposure via drinking water. Maternal toenail arsenic level was significantly associated with all markers of genetic effects measured in cord blood while cord blood arsenic levels were associated with increased DNA strand breaks and MN frequency but not with oxidative DNA damage. All these effects were dose dependent.

Two meta‐analyses (Annangi et al., 2016; Dong et al., 2019) were carried out to review the biomonitoring studies where MN frequency was used as marker of genotoxicity in human populations exposed environmentally or occupationally to arsenic. Annangi et al. (2016) limited the review to studies (three on occupational and seven on environmental exposure) where peripheral blood lymphocytes were analysed while Dong et al. (2019) considered studies (25 on different sources of exposure) dealing with three human cell types, i.e. lymphocytes, buccal and urothelial cells. Both analyses, concluded that environmental chronic exposure to iAs via contaminated water is associated with increased MN frequency among exposed individuals. The subgroup analysis carried out by Dong et al. (2019) showed that when the exposure source was occupational or other than drinking water, no increase in MN frequency was observed in all three types of cells tested.

Telomere dysfunction is an important mechanism to generate chromosomal instability commonly found in several disease processes including cancer (McNally et al., 2019). Some studies addressed the potential association of arsenic exposure with alteration of telomere structure but with equivocal results. Significant increase in telomere length in leukocytes was reported in arsenic‐exposed communities from Mexico (Villarreal et al., 2019) and Northern Argentina (Li et al., 2012); a slight increase, not statistically significant, in children from West Bengal (Chatterjee et al., 2018) and lack of association in people from Bangladesh (Zhang et al., 2018). Future studies are needed to establish the mechanisms underlying lengthening of telomeres and its potential contribution to arsenic‐induced cancer risk.

Besides clastogenic effects, other markers of DNA damage were positively associated with the level of exposure via arsenic‐contaminated water. DNA breaks (both single and double‐strand breaks) as detected by alkaline elution were significantly higher in airway cells (Dutta et al., 2015) and in lymphocytes (Roy et al., 2016) of exposed populations from Indian regions and in lymphocytes from environmentally exposed Mexican children and adults (Jimenez‐Villarreal et al., 2017; Sampayo‐Reyes et al., 2010; Tovar‐Sanchez et al., 2016) as compared to control groups. Elevated levels of oxidative stress‐induced damage (e.g. 8‐oxodG, 8‐nitroguanine) in urine, plasma or saliva, were detected in populations from highly contaminated areas (Dutta et al., 2015; Hinhumpatch et al., 2013; Mar Wai et al., 2019; Navasumrit et al., 2019; Pei et al., 2013). 8‐oxodG is considered a marker for oxidative stress derived from oxidatively generated damage to 2′‐deoxyguanosine via a mechanism that has not yet been clarified (Evans et al., 2016). Only one study found no evidence that exposure to arsenic increases levels of plasma protein carbonyls or 8‐oxodG (Harper et al., 2014). In a Thai population, arsenic exposure in utero was associated with increased levels of urinary 8‐nitroguanine in newborns which significantly correlated with increased expression of inflammatory genes in cord blood (Phookphan et al., 2017). A follow‐up study showed that these arsenic‐exposed children had increased urinary 8‐nitroguanine and significantly higher levels of 8‐oxoguanine in saliva DNA corresponding well to decreased expression of human 8‐oxoguanine DNA glycosylase 1 (hOGG1), the major DNA glycosylase for the excision of 8‐oxoguanine DNA base lesions (Hinhumpatch et al., 2013). In a large prospective cohort study (788 participants) in Taiwan (Tsai, Kuo, et al., 2021) the levels of urinary 8‐oxodG and N7‐methylguanine showed a positive association with cumulative well‐water exposure to arsenic and urinary arsenic species.

Inorganic arsenic induces DNA damage and the formation of MN and CA in various cell types in vitro as well as in vivo (see Section ‘Genotoxicity’). Here, a large number of studies where these genotoxicity markers have been investigated in somatic cells (i.e. lymphocytes, urothelial and buccal cells) of people environmentally exposed to high levels of arsenic in adulthood as well as in early life were reviewed. Although some studies present limitations (e.g. small population size, lack of dose–response relationship, no evaluation of potential confounders), overall, they consistently indicate that chronic environmental exposure via contaminated water is associated with increased DNA damage (DNA breaks and oxidatively induced DNA damage) (levels of As in the range of 10–40 μg/L) and clastogenic events (levels of As > 50 μg/L) in somatic cells from exposed individuals. A well conducted birth‐cohort study (Navasumrit et al., 2019) showed that these effects are also induced in newborns after in utero arsenic exposure. Table 7 shows an overview of the studies on markers of genotoxicity published since 2009.

3.1.2.3. Markers of epigenetic changes

An extensive body of research highlights the epigenetic effects of iAs and its human metabolites. These effects have been shown in in vitro cell cultures and in in vivo animal models exposed to iAs (see comprehensive reviews from Bustaffa et al., 2014; Bhattacharjee et al., 2020; Cardoso et al., 2020). Here, we provide a brief overview of the most relevant studies published since 2009 on ex vivo data, obtained in human populations exposed to iAs primarily via contaminated drinking water.

Changes in DNA methylation have been the most extensively studied epigenetic alterations while changes in histone post‐translational modifications (PTMs), chromatin structure and micro‐RNA (miRNA) expression have begun to receive increasing attention.

S‐adenosylmethionine (SAM) is the main donor of methyl groups during the methylation of both trivalent arsenic species and DNA. DNA demethylation events upon iAs exposure are therefore expected because of the high demand imposed on DNA methyltransferase (DNMT) enzymes during the arsenic biotransformation process while the occurrence of localised hypermethylation is more challenging to explain mechanistically (reviewed in Reichard & Puga, 2010). In recent years, technological advancements, including methylation‐sensitive deep sequencing and microarrays, have allowed to characterise DNA methylation changes globally with sequence‐level sensitivity. Epigenome‐wide association studies (EWAS) have confirmed significant associations between in utero or adult life arsenic exposure and epigenetic dysregulation of global DNA methylation levels in blood. These EWAS include birth cohorts in the United States (Bozack et al., 2018; Green et al., 2016; Koestler et al., 2013), Bangladesh (Broberg et al., 2014; Cardenas, Houseman, et al., 2015; Gliga et al., 2018; Kile et al., 2014), Mexico (Rojas et al., 2015) and Taiwan (Kaushal et al., 2017), and studies in adults in the United States (Bozack et al., 2020; Liu et al., 2014), Bangladesh (Argos et al., 2015; Demanelis et al., 2019), Argentina (Ameer et al., 2017) and China (Guo et al., 2018) where exposure to iAs was primarily through drinking water. However, when analysed in detail, the results of these EWAS are largely inconsistent. As reviewed in Argos et al. (2015) and Bozack et al. (2020), the analysis of these studies shows great differences in the identity and number of CpGs identified and a very limited overlap between significant loci. In addition, some of these studies present small sample sizes resulting in limited statistical power particularly when interrogating a large number of CpG sites. Differences between the populations studied (e.g. age, sex, genetic background, nutritional status), the type of tissue examined (e.g. whole blood, cord blood), levels of iAs exposure (ranging from 23 μg/g creatinine [Kaushal et al., 2017] to > 302 μg/g creatinine urinary arsenic [Argos et al., 2015]), differential residual confounding and methods for quantifying DNA methylation might explain the limited replication among EWAS. Moreover, most studies lack data on functional changes in gene expression.

Most of the investigations into the effects of arsenicals on histone PTMs have been performed in vitro, in which changes in both global and gene‐specific histone PTM patterns have been observed (reviewed in Bailey & Fry, 2012). Few studies have been conducted in humans. Changes in both methylation and acetylation of histones have been shown in association with arsenic exposure and these associations differed in a sex‐specific manner (Chervona et al., 2012; Howe et al., 2016). A critical role of miRNA in iAs‐induced tumorigenesis and carcinogenesis has been proposed based on many in vitro studies showing that dysregulated expression of miRNA occurs in arsenic‐induced malignant cell transformation (reviewed in Cardoso et al., 2018). A few studies conducted in human populations confirm that arsenic species may modulate miRNA expression (Banerjee et al., 2017, 2019; Pérez‐Vázquez et al., 2017; Zeng et al., 2019).

Despite the limitations presented above, some relevant aspects of iAs effects on the epigenome have emerged which are:

  • Changes in DNA methylation are reported in populations exposed to a wide range of iAs levels including low levels (ranging from 23 to > 302 μg/g creatinine urinary As). It is of note that a genome‐wide examination of cord blood isolated from newborns from a birth cohort in New Hampshire (Koestler et al., 2013) showed that low levels of prenatal As exposure (maternal urinary total As: median 4.1 μg/L, interquartile range 1.8–6.6) may affect the methylation of CpG islands. The DNA methylation status of several of these CpG islands had linear relationships with maternal urinary total inorganic As.
  • There is a growing body of evidence indicating that iAs induces epigenetic dysregulation in a sex‐specific manner (Broberg et al., 2014; Niedzwiecki et al., 2015; Pilsner et al., 2012). These alterations, if functional, may account for differential susceptibility to iAs‐induced disease in males and females, a feature observed in various cohorts exposed to iAs.
  • Several reports indicate that in utero arsenic exposure alters DNA methylation potentially altering fetal developmental programming, which may result in a higher risk of disease later in life. For instance, Rager et al. (2014), by analysing the methylation profile of newborn cord blood samples from a Mexican pregnancy cohort exposed to arsenic by drinking water (from 0.456 to 236 μg/L), identified 16 genes that showed a correlation between iAs‐associated changes in DNA methylation and mRNA expression, seven of which were associated with differences in birth outcomes such as gestational age (GA) and head circumference (HC).
  • iAs‐induced DNA methylation changes have been detected in critical genes for disease development such as tumour suppressor genes and DNA repair genes (Bhattacharjee et al., 2018; Hossain et al., 2012; Intarasunanont et al., 2012, Kile et al., 2012; Lambrou et al., 2012, Majumdar et al., 2010; Pilsner et al., 2012; Smeester et al., 2011). However, in many cases, the functional consequences of these changes or their causal relationship with disease have not been established.
  • Altered epigenetic marks can be metastable, and thus the effects of altered gene expression and disease risk have the potential to be transgenerational. In a rat model Nava‐Rivera et al. (2021) have recently provided multigenerational (up to F3) evidence of changes in global DNA methylation in gonadal tissue upon parental arsenic chronic exposure. It is of concern that iAs‐induced epigenome alterations in the early life of an individual may affect the health of later generations.

The existing reports of epigenetic changes mediated by iAs in exposed human populations highlight the need for further research. In particular, the relevance of these alterations to disease risk in exposed populations needs to be addressed. The interplay between multiple epigenetic components and non‐epigenetic alterations that likely work together to mediate the toxic effects of arsenicals also requires to be further investigated.

3.1.3. Genotoxicity

As summarised in the previous EFSA Opinion (EFSA CONTAM Panel, 2009) the evidence available at that time on the genotoxic effects of iAs included:

  • negative mutation assays in bacterial cells; weak (or insignificant) mutagenic activity in mammalian cells in vitro;
  • clastogenic (micronuclei, chromosomal aberrations, sister chromatid exchanges) and aneugenic effects mostly at high doses in mammalian cells in vitro;
  • induction of DNA damage (DNA strand breaks, oxidative base modifications and apurinic/apyrimidinic (AP) sites) in mammalian cells in vitro;
  • enhancement of genotoxicity, mutagenicity and clastogenicity of other DNA damaging agents in mammalian cells in vitro by interfering with DNA damage response processes;
  • in vivo induction of micronuclei and chromosomal aberrations in mouse peripheral blood lymphocytes and in mouse bone marrow.

The data published since 2009 mainly refer to further analysis of DNA damage and clastogenic effects induced by iAs both in vitro and in vivo. The findings are summarised below, and further details are presented in Table 8 (in vitro studies) and Table 9 (in vivo studies).

TABLE 8

In vitro genotoxicity tests with arsenic species.

ReferenceTest systemCellsConcentration/treatment timeResultsAdditional information
Ding et al. (2009)

8‐oxodG measurement by HPLC‐ED and in situ immune fluorescence;

PARP‐1 activity by in situ immune fluorescence

Human keratinocyte cell line (HaCaT)

Sodium arsenite

2 μM (24 h)

Enhancement of UVR‐induced 8‐oxodG and suppression of UVR‐induced PARP‐1 activation (lowest dose tested 0.2 μM)

No data on cytotoxicity. Exposure to 1–3 μM arsenite: no increase of 8‐oxodG

Inhibition of PARP‐1 activity significantly increases UVR‐induced 8‐OHdG formation

Mass spectrometry analysis reveals that arsenite can occupy the first zinc finger of PARP‐1 (PARPzf)

Nollen et al. (2009)Western blotting and gene expression analysis of cell extractsTert‐immortalised human diploid skin fibroblasts (VH10hTert)

Sodium arsenite 1, 5, 10 μM (24 h)

MMA(III) 0.1, 1, 1.3 μM (24 h)

Inhibition of XPC expression and protein levels for both arsenicals. Diminished association of XPC to sites of local UVC damage

Viability > 80%

Cell viability: cell number and colony forming ability

Inhibition of XPE expression

Ying et al. (2009)

γ‐H2AX foci

Pulse field gel electrophoresis (PFGE)

Chinese hamster cells (AA8)Sodium arsenite from 5 to 50 μM (24 h)

Positive: concentrations as low as 5 μM could induce gH2AX foci

(IC50 = 18.6 μM)

Cell viability: clonogenic assay

50 μM arsenite produces a similar amount of DSBs as a 6 Gy dose of ionising radiation as estimated by PFGE. The occurrence of homologous recombination (HR) confirmed by Rad 51 foci and cell recombination assay

HR defective cells are hypersensitive to arsenite

Arsenite inhibits SSB repair

Catanzaro et al. (2010)Alkaline comet assayPrimary cultures of rat astrocytes

Sodium arsenite

2.5, 5, 10, 30 μM (24 h)

Positive: significant increase of DNA breaks (tail length) at 10 μM for 24 h (> 70% viability)

Cell viability: trypan blue exclusion test

Cells treated with 30 μM for 24 h were all ghost (small heads and large tails), likely apoptotic cells

Absence of ROS production by CM‐H2DCFDA

Dopp et al. (2010)Alkaline comet assayNon‐methylating cells: UROtsa transformed human urothelial cells; methylating cells: primary human hepatocytesAs(III), MMA(III), iAs(V), MMA(V) for 1 h; range of doses tested not reported

Positive: significant induction of DNA breaks (tail moment): primary human hepatocytes at 5 μM MMA(III) (LC50 = 20μM) and 50 μM iAs(III) (not cytotoxic up to 5 mM); UROtsa cells at 5 μM MMA(III) (LC50 = 83μM) and 5 μM iAs(III) (LC50 = 5000 μM) for 1 h

Negative: in human hepatocytes iAs(V) and MMA(V) did not induce DNA breaks at doses up to 500 μM; they were not tested in urothelial cells

Cell viability: trypan blue exclusion test

Induction of free radicals, measured by the thiobarbituric acid test, showed cell‐type specific differences

Kligerman et al. (2010)Chromosome aberration assayFreshly prepared splenic lymphocyte culturesMMA(III), 0.3 and 0.5 μM (12–17 h) Positive: significant increase of CA at both concentrations. MMA(III) (0.5 μM) positive when cells were treated in late G1‐ or S‐phase; negative when treatment was confined to the G0‐ or G1‐phase of the cell cycle

Replication index indicates that both concentrations are cytotoxic

DNA lesions produced by MMA(III)I that can lead to cytogenetic damage are short‐lived

Bartel et al. (2011)

MN assay

Alkaline unwinding

Human A549 lung cells

Arsenite, MMA(III), DMA(III), MMA(V), DMA(V), thio‐DMA(V)

(1, 24 h)

Positive: significant increase of MN frequency at non‐ cytotoxic/slightly cytotoxic concentrations for 24 h (< IC70): MMA(III) (0.5, 1 μM), thio‐DMA(V) (5 μM), MMA(V) (250 μM) and DMA(V) (250 μM) At cytotoxic concentrations significant increases also for arsenite (≥ 50 μM for 24 h) (≥ IC70)

Negative: thio‐DMA(V) did not induce DNA strand breaks up to high cytotoxic doses (100 μM for 1 h, 75 μM for 24 h)

Cell viability: cell number and colony forming ability. Based on the effective cellular arsenic concentrations, the cytotoxic order was: thio‐DMA(V) ∼ arsenite ∼ MMA(III) > DMA(III) MMA(V) ∼ DMA(V)

Thio‐DMA(V) and especially DMA(III) increased the formation of multinucleated cells (significant effects at cytotoxic concentrations, 15 μM, IC70 = 7.2 μM)

Ebert et al. (2011)DNA repair enzymes cleavage assay, gene expression and western blotting of cell extractsA549 human epithelial lung adenocarcinoma cells

Arsenite: up to 100 μM

DMA(III): up to 7.5 μM

MMA(III): up to 7.5 μM

MMA(V): up to 500 μM

DMA(V): up to 500 μM

24 h exposure followed by preparation of cell extracts

Specific inhibitory effects on OGG1 activity by DMA(V), on LigIII by arsenite, on XRCC1 by MMA(V) (starting at ≥ 3.2 μM cellular arsenic; cytotoxicity < 30%); the trivalent methylated metabolites effective only at cytotoxic concentrations

Cytotoxicity, cell number and colony forming ability

Cytotoxic effects correlate with cellular uptake in the decreasing order DMA(III), MMA(III), arsenite, MMA(V), DMA(V)

Lai et al. (2011)

Alkaline comet assay

Micronucleus assay

Mouse embryonic fibroblasts (MEF) Pol β+/+ and Pol β −/− Sodium arsenite: 2.5, 5, 10, μM (24 h)

Positive: accumulation of DNA breaks (comet rate%) at 2.5, 5 and 10 μM in both cell lines, more pronounced in Pol β −/−cells

Positive: significantly increased number of micronuclei at 2,5, 5 and 10 μM in both cell lines, more pronounced in Pol β−/− cells Viability > 80%

Pol β +/+IC50 = 58.3 μM

Pol β−/‐IC50 = 46.6 μM

Cell viability: MTT assay

Comet rate (%) = (total number of cells with tails/total number of counted cells) x 100.

Slower DNA repair kinetics in Pol β−/− cells

Naranmandura, Xu, et al. (2011)Intracellular ROS generation measured by oxidation‐sensitive fluorescent probe (DCFH‐DA)At liver RLC‐16 cellsMMA(III) (IC50 = 1 μM), DMA(III) (IC50 = 2 μM), iAsIII (IC50 = 18 μM) (24 h)MMA(III), ROS generated primarily in mitochondria; DMA(III), ROS generation in other organelles; iAS(III), no ROS generation

Cytotoxicity: MMA(III) (IC50 = 1μM) > DMA(III) (IC50 = 2μM) > iAs(III) (IC50 = 18 μM)

Mitochondria are the primary target for MMA(III)‐induced cytotoxicity

Naranmandura, Carew, et al. (2011)Alkaline comet assayHuman bladder cancer cells (EJ‐1)iAsIII (75 μM), DMA(III) (12 μM), Thio‐DMA(V) (here named DMMTA(V)) (17 μM) (3–6‐24 h). All tested at IC50 Positive: iAs(III), significant increase of DNA breaks (tail length) after 6 and 24 h exposure; DMA(III) and thio‐DMA(V) after 3,6 and 24 h exposure; all tested at IC50 (MTS assay)

Concomitant measurement of ROS levels: iAs(III), increase only after 24 h exposure; DMA(III) and thio‐DMA(V) after 3, 6, 24 h exposure

iAs(III), increased GSH levels; DMA(III) and thio‐DMA(V), reduction of GSH(5μM for 24 h). iAs(III) increased expression of p21 and p53; DMA(III) and thio‐DMA(V) decreased expression of p21 and p53 (dose dependent)

Singh et al. (2011)

Alkaline comet assay

Mitochondrial DNA mutation assay by multiplex PCR

Immortalised normal prostate epithelial cells (RWPE‐1)Sodium arsenite: 100 pg/mL (90 days)

Positive: increased DNA breaks (tail length) as compared to control cells (no tails)

No major deletions or insertions in the sequence of mitochondrial DNA; insertion mutation (a ‘G’) in the mitochondrial ATPase gene

Cell viability: cell count

Chronic exposure to 100 pg/mL: Increase in cell proliferation (cells become resistant to As)

Jiang et al. (2013)

Alkaline comet assay

Micronucleus assay

Human lung adenocarcinoma cells (A549)Sodium arsenite: 5, 10, 15, 20, 25 μM (24 h)

Positive: significant increase of DNA breaks (tail moment) in the full concentration range.

(cell viability > 70% up to 15μM)

Positive: significant (p < 0.05) increase of MN frequency from 10 μM (cell viability > 70%)

Cell viability measured by the MTT assay and Trypan blue exclusion.

Induction of cell cycle arrest at G2/M phase and apoptosis.

Leffers et al. (2013)Micronucleus assayImmortalised human urothelial cells (UROtsa)

Sodium arsenite: 0.1, 0.5, 1, 2, 3.5 μM (48 h)

DMA(V): 50, 100, 170, 200 μM (48 h)

Thio‐DMA(V): 0.1, 0.5, 1, 2 μM (48 h)

Positive: Arsenite: significant increase (p < 0.001) of induced MN from 0.5 to 3.5 μM (including a subtoxic concentration range)

DMA(V) and thio‐DMA(V): significant increase (p < 0.001) of bi‐ and multi‐nucleated cells from 100 to 200 μM (DMA(V)) and at 2 μM (thio‐DMA(V)), at incipient cytotoxic concentrations.

30% reduction of CFA:

Arsenite 2 μM; DMA(V) 150 μM; thio‐DMA(V) 1.5 μM

Cell viability: cell number and colony forming ability.

Arsenite: No increase in the number of bi‐ or multinucleated cells.

Tokar et al. (2013)

Cell transformation (agar assay and invasion assay)

ROS generation by the immuno‐spin trapping analysis of protein radicals (immunochemical quantification)

Arsenic methylation‐proficient TRL1215 liver cell line and methylation‐deficient RWPE1 prostate cell lineMMA(III): 0.25–1.0 μM (20 weeks or more) Positive: increased matrix metal‐ loproteinase secretion, colony formation and invasion at 18–22 weeks in both cell lines

Similar alterations in arsenic and oxidative stress adaptation factors in both cell lines

MMA(III) and iAs cause an acquired malignant phenotype in methylation‐deficient cells, yet iAs does not induce oxidative DNA damage

Bach et al. (2014)FPG‐modified comet assayMouse embryo fibroblasts (MEF) Ogg1 +/+ and Ogg1 −/−

Sodium arsenite:

Short‐term studies: up to 20 μM for 24 or 48 h; Long‐term studies: 0.5, 1 and 2 μM for up to 17 weeks

Positive: significant levels of oxidative DNA lesions (%DNA in tail) in Ogg1 /− cells at subtoxic concentrations (> 80% survival) in both short‐term (5 μM for 1.5, 3, 24 h) and long‐term (0.5 μM for 4, 10, 17 weeks) studies; in Ogg1 wt at 20 μM for 24 h and 1 and 2 μM for 17 weeks

Cell viability: Beckman counter method (up to 80 μM for 24 or 48 h)

Analysis of arsenic metabolism in the cells; similar in MEF wt and Ogg1 −/−

Ebert et al. (2014)Alkaline unwinding with FPGImmortalised human urothelial cells (UROtsa)Sodium arsenite and thio‐DMA(V): up to 5 μM (1, 24 and 48 h)

Negative: arsenite and thio‐DMA(V) significant increase of DNA breaks only at cytotoxic concentration (5 μM)

Arsenite and thio‐DMA(V): CFA, IC70 = 5.2 μM

Cell viability: cell number and colony forming ability

Inhibition of damage‐induced poly (ADP)‐ribosylation: arsenite at high concentration (3.5 μM), thio‐DMA(V): at 35,000‐fold lower concentration

Thio‐DMAV or arsenite induced significant S phase and G2/M phase arrest; higher induction of apoptosis by thio‐DMA(V)

Li et al. (2014)Alkaline comet assayRat pheochromocytoma cell line (PC12)Sodium arsenite: 5, 20 μM for 24 h Positive: increase of DNA breaks (%tail DNA) at both doses, dose‐related (cell viability 5 μM, 97%; 20 μM, 81%)

Cell viability: MTT assay

Increased ROS levels

Rehman et al. (2014)Intracellular ROS generation measured by oxidation‐sensitive fluorescent probe (DCFH‐DA)Human myeloid leukaemia HL‐60 cells

MMA(III), DMA(III) and iAs(III): (1 μM for 12 h)

MMA(III): (IC50 3 μM)

DMA(III): (IC50 2 μM)

iAs(III): (IC50 10 μM)

MMA(III) and DMA(III) but not iAs(III) increased oxidative stress, loss of mitochondrial membrane potential and apoptosis
Unterberg et al. (2014)Micronucleus assayNon‐tumorigenic urothelial cell line (UROtsa)

Arsenite and thio‐DMA(V): 0.005, 0.01, 0.1, 1, 10, 100, 250, 500 nM (7, 14 and 21 days)

Positive: Arsenite, 100 nM, 14 days and 250 and 500 nM all exposure times (100 nM subtoxic dose, > 80% CFA).

Thio‐DMA(V): negative in the full concentration range. No cytotoxicity up to 500 μM

Cell viability: cell number and colony forming ability.

Induction of global DNA methylation

Xie et al. (2014)

Chromosomal aberration assay

Neutral comet assay and gH2AX foci forming assay

Human primary bronchial fibroblast (NHBF) and epithelial (NHBE) cells.

Sodium arsenite: 0.5, 1, 5, 10 μM (24 and 120 h)

Positive: increase in chromosome damage in fibroblasts but not in epithelial cells, significant at 5, 10 μM with both exposure times High cytotoxicity at both concentrations (≤ 50% CFA)

Induction of aneuploidy and mitotic abnormalities in fibroblasts only.

Positive: induction of DNA double strand breaks in both fibroblast and epithelial cells Range 1–10 μM, 24 h, 120 h) including sub‐toxic doses in the case of epithelial cells

Cell viability: colony forming ability
Meyer et al. (2015)Micronucleus assayHuman liver HepG2 cellsDMA(V): 0, 1, 10, 100, 500 μM (48 h) Positive: significant increase of MN at 100 and 500 μM, IC70 155 μM
Benhusein et al. (2016)Alkaline comet assayHepG2 Liver CellsArsenate, sodium arsenite and DMA(V): 10 μM for 24 h

Positive: increase of DNA breaks (tail moment) with arsenate and arsenite

Negative: with DMA(V)

Cell viability: 85%–90%

Cell viability: trypan blue exclusion test

Increase of cellular levels of glutathione with arsenate and arsenite

Okamura and Nohara et al. (2016)γ‐H2AX by western blottingMouse B lymphoma cells (A20)Sodium arsenite: 10 μM (up to 14 days) Positive: γ‐H2AX was increased following exposure for 8 and 14 daysActivation of the p53‐p21 pathway. Decreased expression of DNA repair genes. Increased expression of cytidine deaminases. Increased Bcl6 mutations (point mutations, deletions and insertions). Induction of senescence
Xu et al. (2016)Alkaline and hOGG1‐modified (FLARE) comet assayIsolated mouse bone marrow, spleen, thymus cellsSodium arsenite or MMA(III): 5, 50 and 500 nM (4 h) Positive: Arsenite: bone marrow cells significant increase of DNA breaks (% DNA in tail) from 5 to 500 nM, spleen and thymus cells only at 500 nM. MMA(III): cells from all the three organs showed significant increase of DNA breaks starting at 5 μM

No data on cytotoxicity

MMA(III) is more genotoxic than iAs(III) in vitro

Holcomb et al. (2017)NER capacity by slot‐blot assayHuman lung fibroblasts (IMR‐90 cells) and primary mouse keratinocytesSodium arsenite: 10–20‐40 μM (24 h)

Concentration‐dependent inhibition of the removal of 6–4 PPs and CPDs in both cell types

Significant reduction of XPC protein levels. Significant effects at 20 and 40 μM

Viability: 10 μM ~ 80%; 20 μM ~ 70%; 40 μM ~ 50% (from the graph)

Cell viability: trypan blue exclusion test
Jiang et al. (2017) In vitro binding assay, streptavidin agarose affinity assay, western blotting, fluorescence microscopy, mass spectrometry, clonogenic survivalHEK293T human embryonic kidney epithelial cells and HeLa cellSodium arsenite: 5 μM (cell exposure for 24 h)Arsenite binding to FANCL in vitro and in cells. Reduction of FANCD2 recruitment to chromatin and DNA damage sites in cells upon ICL induction. Increased cell sensitivity toward ICL‐inducing agentsNo data on arsenite cytotoxicity are presented
Xu et al. (2017)

Alkaline comet assay and γ‐H2AX foci forming assay

ROS detection by flow cytometry

Isolated mouse thymus cells. DN, double negative, do not express CD4 or CD8; DP, double positive, express both cell markers (obtained by cell sorting)MMA(III): 5, 50 and 500 nM (18 h) Positive: MMA(III) significant increase of DNA breaks (%DNA in tail) in DN cells in the whole concentration range; DP cells at 50 and 500 nM. Significant increase of gH2AX fluorescence and reactive oxygen species level (DN cells, 50 and 500 nM; DP cells, 500 nM)Cell viability: MTS assay. No quantitative data provided. Low (5 nM, ~ 80% survival) to moderate (500 nM ~ 50% srv)) cytotoxicity (from the graph)
Kopp et al. (2018)γ‐H2AX by western blotting

Human hepatoblastoma cells (HepG2)

Human epithelial colorectal adenocarcinoma cells (LS‐174T)

Sodium arsenite: 1, 10, 25, 50, 75, 100 μM (24 h) Arsenic pentoxide: 10, 50, 100, 250, 500 μM (24 h)

Positive: significant increase (p < 0.01) of gH2AX levels at subtoxic doses for both arsenicals

Arsenite: in HepG2 cells, LOAEC 25 μM, %RCC = 92; LS‐174T, LOAEC 10 μM, %RCC = 100.

Arsenic pentoxide: in HepG2 cells, LOAEC 250 μM, %RCC = 88; LS‐174T: LOAEC 100 uM, %RCC = 96

Cell viability: comparison of DNA content (related to the number of cells) in treated versus control cells and expressed as relative cell count, %RCC
Ganapathy et al. (2019)Aneuploidy testHuman lung epithelial BEAS‐2B cells and keratinocytesSodium arsenite: 0.5 μM (2 months) Positive: significant increase of cells with aneuploidy (~ 2% in untreated cells versus ~ 14% in arsenite‐treated cells)

No data on cytotoxicity are presented. 100 cells scored for each treatment, 5 experiments per point

Perturbation of mitosis via activation of Akt and upregulation of Plk1. Suppression of Akt or Plk1 inhibited aneuploidy

Abbreviations: 8‐OHdG, 8‐hydroxy‐2′‐deoxyguanosine; 8‐oxodG, 8‐hydroxydeoxyguanosine; ADP, adenosine diphosphate; Akt, protein kinase B or PKB; As(III), arsenite; BEAS‐2B, bronchial epithelial airway sensitive‐2B (normal human bronchial epithelial cell line); CA, chromosomal aberration; CD4 / CD8, cluster of differentiation 4 and 8; CFA, colony‐forming ability; CM‐H2DCFDA, chloromethyl derivative of H2DCFDA; CPD, cyclobutane pyrimidine dimer; DCFH‐DA, 2′,7′ dichloro‐dihydrofluorescein diacetate; DMA(III), dimethylarsinous acid; DMA(V), dimethylarsinic acid; DN, double negative; DNA, deoxyribonucleic acid; DP, double positive; DSB, double‐strand break; FANCD2, fanconi anaemia complementation group D2; FANCL, fanconi anaemia complementation group L; FLARE, fragment length analysis using repair enzymes; FPG, formamidopyrimidine DNA glycosylase; gH2AX, serine139‐phosphorylated histone H2AX; GSH, glutathione; Gy, Grey; HaCaT, human epidermal keratinocyte line; HeLa cells, human cell line named after Henrietta Lacks; HEK293T, human embryonic kidney epithelial cells; HepG2, human hepatoblastoma cells; hOGG1, human 8‐oxoguanine DNA glycosylase 1; HPLC‐ED, high‐performance liquid chromatography with electrochemical detection; HR, homologous recombination; iAs, inorganic arsenic; iAs(V), arsenate; IC50, inhibitory concentration 50%; ICL, inter‐strand DNA cross‐link; IMR‐90 cells, human lung fibroblasts; LC50, lethal dose killing 50% of the animals; LigIII, DNA ligase III; LOAEC, lowest‐observed‐adverse‐effect‐concentration; LS‐174T, human epithelial colorectal adenocarcinoma cell; MEF, mouse embryo fibroblast; MMA(III), monomethylarsonous acid; MMA(V), monomethylarsonic acid; MN, micronucleus; MTS, 3‐(4,5‐dimethylthiazol‐2‐yl)‐5‐(3‐carboxymethoxyphenyl)‐2‐(4‐sulfophenyl)‐2H‐tetrazolium; MTT, 3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyl‐2H‐tetrazolium bromide; NER, nucleotide excision repair; NHBE, normal human bronchial epithelial cells; NHBF, normal human bronchial fibroblast cells; OGG1, 8‐oxoguanine DNA glycosylase 1; PARPzf, the first zinc finger of PARP‐1; PARP‐1, poly (adenosine diphosphate‐ribose) polymerase 1; PCR, polymerase chain reaction; PFGE, pulse field gel electrophoresis; pg, picogram(s); PLK1, polo‐like kinase 1; Pol β, DNA polymerase beta; PP, photoproduct; RCC, reactive cell count; ROS, reactive oxygen species; RWPE‐1, immortalised normal prostate epithelial cells; SSB, single‐strand break; UROtsa, immortalised human urothelial cells; VH10hTert, tert‐immortalised human diploid skin fibroblasts; thio‐DMA(V), thio‐dimethylarsinic acid; u‐tiAs, total urinary inorganic arsenic (sum of inorganic arsenic and its methylated metabolites MMA and DMA); UVC, ultraviolet C light; UVR, ultraviolet radiation; wt, wild‐type; XPC, Xeroderma pigmentosum complementation group C; XPE, Xeroderma pigmentosum complementation group E; XRCC1, X‐ray repair cross‐complementing protein 1.

TABLE 9

In vivo genotoxicity of inorganic arsenic.

ReferenceTest systemAnimalsConcentration/treatment timeResultsComments/additional information
Inorganic arsenic species other than arsenic trioxide
Osmond et al. (2010)

Quantitative PCR transcript amplification

Lung

Mice (1, 8, 24 weeks old) (three to five animals/group)Sodium arsenite 2 or 50 mg/kg bw/day in drinking water (24 h or 2 weeks), oral route

Adult animals (24‐week‐old): decrease of all BER gene transcript levels (APE1, LIG1, LIG3, OGG1, PARP1 and POLΒ) in lung tissue upon 2 weeks treatment

Neonates whose mothers were exposed to 50 mg/kg. for 24 h or 2 weeks: increased levels of the APE1, LIG1, OGG1 and PARP1 transcripts in lung tissue

Number of animals not specified
Mehta & Hundal (2014)

Chromosomal aberrations

Bone marrow

Mature female rats (n = 48; 12 animals/group)Sodium arsenite 10, 30, 50 mg/kg (30 and 60 days), oral route Positive: significant increase of CA in bone marrow cells after 30 days of exposure to 30 and 50 mg/kg; after 60 days exposure to all three doses (10, 30 and 50 mg/kg), dose‐dependent increase

Significant loss of bw

Lack of data on cytotoxicity (mitotic index is not reported)

Poor experimental details and data quality (unknown number of metaphases scored)

Takumi et al. (2014)

gpt mutation assay

Liver

Electrochemical detection (EC)‐HPLC, measurement of 8‐oxodG

Male gpt delta mice (5 weeks old), n = 12; 6 animals/group

Sodium arsenite

85 mg/kg (85 mg/L) in drinking water ad libitum (3 weeks); estimated: 4.3 mg/kg per day per mouse

Positive: increased gpt mutation frequency in the liver by 1.5‐fold. Mutational spectra: predominance of G:C to T:A transversion and higher frequency of deletion mutations

Significantly higher levels of 8‐oxodG in the livers of arsenite‐exposed group

No changes in the average body weight, average relative liver weight significantly lower than in the control group

Fujioka et al. (2016)

gpt and Spi − mutation assay.

Urinary epithelium

liver

Old male gpt delta F344 rats, n = 36, 12 animals/group)Sodium arsenite 87 mg/L; DMA(V) 92 mg/L in drinking water (13 weeks) Negative: no effects on the mutant frequencies or mutation spectrum in urinary bladder epithelium or liver for both arsenicals

In the arsenite treatment group: the final body weights and absolute liver weights were significantly decreased; water intake and food consumption were significantly decreased from week 2.

In the DMA(V) group: no effects on body and liver weights; increased water intake, no effects on food consumption

A:T to T:A transversions in the gpt gene, being spontaneous mutations, excluded from mutation frequency and spectra.

Dash et al. (2018)Alkaline comet assay in uterine cellsFemale virgin albino Wistar strain rats (n = 24; six animals/group)Sodium arsenite 10 mg/kg bw/day (2 or 8 days) without and with N‐acetyl‐l‐cysteine (NAC), oral route

Positive: Arsenite: significant increase of DNA breaks (number of comets, tail length); decreased DNA damage by co‐treatment with NAC

No changes in bw

Significant weight loss for ovaries and uterus; ovarian and uterine tissue damage, disruption in steroidogenesis in the arsenic‐treated group

Nath Barbhuiya et al. (2020)Alkaline comet assay in ovary and uterusSwiss albino mice (6–8 week old; n = 30; six animals/group; five experimental points)

Sodium arsenite 2 mg/kg bw per day (30 days)

Exposure also to smokeless tobacco alone and in combination with sodium arsenite, oral route

Positive: significant increase of DNA breaks (head/tail DNA, tail moment, tail length)

Significant loss of bw

Data on target tissue toxicity: 50% decrease of relative organ weight and extensive histological abnormalities for both ovary and uterus

Arsenic trioxide
Kesari et al. (2012)CA in bone marrow cellsSwiss albino mice (6–8 week old, n = 30, six animals/group)Daily oral gavaging of aqueous solution of arsenic trioxide: arsenic content 3.0 mg/L, 1.5 mg/L, 150 μg/L and 30 μg/L (15 consecutive days) Doses tested: 0.0 0.3, 1.5, 15, 30 μg/kg bw per day

Positive: significant increase of structural chromosomal abnormalities upon exposure to the full range of doses tested; dose‐dependent increase

Numerical abnormalities were induced by doses higher than 0.3 μg/kg bw per day

Lack of data on cytotoxicity (mitotic index is not reported)

Too few metaphases scored per animal (50 instead of 200)

Khan et al. (2013)MN assay in erythrocytes from bone marrow cellsSwiss albino mice (6–8‐week‐old, four groups, number of animals/group is not specified)

Daily oral gavaging of aqueous solution of arsenic trioxide: arsenic content 1.5 mg/L, 150 μg/L and 30 μg/L (15 consecutive days)

Doses tested: 0.0–0.3‐1.5–15 μg/kg bw per day

Positive: significant increase in micronucleated cells al all doses tested, dose‐dependent

1000 erythrocytes scored (OECD protocol 2000)

About six‐fold increase in micronucleated cells at the human reference dose of arsenic (0.3 μg/kg per day)

Nava‐Rivera et al. (2021)Alkaline comet assayRats about 3 weeks old; males (n = 12) and females (n = 12)Arsenic trioxide in drinking water 1 mg/L of arsenic equivalent to 1 mg/kg bw, for an interrupted period of 16 weeks Positive: significant intra‐generational increase in the level of DNA breaks (% DNA in tail) in the arsenic lineage compared to the control lineage in all generations in both females and males. The inter‐genotoxic effect following the arsenic lineage, was in decrement across generations but with statistical significance in the F3 generation compared to the control groupThe F0 parental was directly exposed to As; the F1 offspring experienced indirect exposure in utero and trough being breastfed until weaning in 21st day, F2 had a multigenerational indirect exposure as F1 germinal line, F3 transgenerational lineage neither direct nor indirect exposure to arsenic

Abbreviations: 8‐oxodG, 8‐hydroxydeoxyguanosine; A, adenine; APE1, apurinic/apyrimidinic endonuclease 1; BER, base excision repair; bw, body weight; C, cytosine; CA, chromosomal aberration; DMA(V), dimethylarsinic acid; DNA, deoxyribonucleic acid; EC‐HPLC, high‐performance liquid chromatography with electrochemical detection; F0, F1, F2, F3, filial generation 0, 1, 2, 3; G, guanine; LIG1, DNA ligase 1; LIG3, DNA ligase 3; MN, micronucleus; n, number; NAC, N‐acetyl‐l‐cysteine; NAC, N‐acetyl‐l‐cysteine; OECD, Organisation for Economic Co‐operation and Development; OGG1, 8‐oxoguanine DNA glycosylase 1; PARP1, poly (adenosine diphosphate‐ribose) polymerase 1; PCR, polymerase chain reaction; POLB, DNA polymerase beta; T, thymine.

3.1.3.1. In vitro genotoxicity

Inorganic arsenic species

Most studies published after 2009 deal with the characterisation of DNA damage induced by iAs species. The comet assay has been widely applied because it is a rapid and sensitive technique able to measure multiple classes of DNA damage in individual cells. The standard alkaline comet assay, that allows to detect single (SSB) and double‐strand breaks (DSB) and alkali‐labile sites (apurinic/apyrimidinic sites), has been used in a variety of cells in culture of rodent or human origin, either primary or transformed. In general, arsenite and arsenate were confirmed as good inducers of DNA breaks and alkali‐labile sites at subtoxic concentrations (Benhusein et al., 2016; Catanzaro et al., 2010; Jiang et al., 2013; Li et al., 2014; Naranmandura, Carew, et al., 2011; Singh et al., 2011; Xu, McClain, et al., 2016). The induction of oxidative DNA base damage by iAs was further supported by studies where the formamidopyrimidine‐DNA glycosylase (Fpg)‐modified comet assay was associated with the use of mouse cell lines defective in the DNA glycosylase specific for removal of 8‐oxoguanine, OGG1 (Bach et al., 2014, 2015). The use of Fpg in the comet assay allows for the detection of 8‐oxoguanine in addition to certain imidazole ring‐opened purines and abasic sites (Karakaya et al., 1997). From these studies, OGG1 emerged as a relevant DNA repair enzyme for arsenic‐induced toxicity and genotoxicity. In addition, in OGG1 defective cells chronically exposed to arsenic, the overexpression of the metabolising enzyme arsenic‐methyltransferase (As3MT) was identified as an adaptive mechanism that protects from cell death but leads to accumulation of high levels of DNA damage due to their repair defect (Bach et al., 2015). The alkaline comet assay does not distinguish SSB from DSB. The induction of DSB by iAs was suggested by some studies where the level of phosphorylated H2AX, measured either by western blotting or by the foci assay, was used as a marker of DSB (Kopp et al., 2018; Okamura & Nohara, 2016; Xie et al., 2014). The induction of γH2AX foci can result from different types of DNA lesions including DNA adducts, abasic sites, strand breaks and replication or transcription blocking lesions (Watters et al., 2009). In 2009, Ying et al., provided unequivocal evidence that arsenite induces DSB by confirming the increase in the number of γH2AX foci with the analysis of DSB by pulse field gel electrophoresis (PFGE) that allows to look directly to DSB. Another confirming evidence was presented by Xie et al. (2014) by using the neutral comet assay that detects only DSB (Xie et al., 2014). DSB are the most lethal DNA lesions, and it is of note that arsenic induces this type of damage at subtoxic concentrations as low as 5 μM (Xie et al., 2014; Ying et al., 2009). In addition, Ying et al. (2009) showed that these DSB are replication‐dependent and homologous recombination (HR) is required for their repair. Based on these findings, they hypothesised that these breaks arise from collapse of replication forks due to the large number of lesions and inhibition of DNA repair induced by arsenic. A study in yeast (Litwin et al., 2013) confirms the induction of DSB by arsenic and shows that arsenic may also cause DSBs independent of replication and transcription. On the basis of these findings the authors suggested ‘a direct genotoxic mode of arsenic action’.

The source and mechanisms of ROS production by iAs have been addressed in some studies (Catanzaro et al., 2010; Ebert et al., 2014; Li et al., 2014; Naranmandura, Carew, et al., 2011). In general, long‐term cell exposure and/or high concentrations of sodium arsenite were required to measure a significant increase in the levels of intracellular ROS. In line with these findings, no production of ROS was detected in rat liver submitochondrial particles and liver cells (RLC‐16) upon exposure to low levels of arseniteI (Naranmandura, Xu, et al., 2011). Even if there is no direct evidence of arsenic‐induced ROS at subtoxic doses, indirect evidence has been reported. Arsenic was shown to increase glutathione levels upon short‐term exposure to low concentrations (Benhusein et al., 2016; Naranmandura, Carew, et al., 2011). Activation of p38/Nrf2 pathway and upregulation of glutamate‐cysteine ligase catalytic subunit (GCLC) expression have been proposed as the underlying mechanisms (reviewed in Ran et al., 2020). While these in vitro studies confirm that arsenic exposure has a regulatory role on glutathione synthesis they are not in agreement with in vivo studies showing that arsenic exposure can reduce glutamate and cysteine levels and inhibit glutathione synthesis (reviewed in Ran et al., 2020). Further experiments are required to assess the regulatory role of arsenic on glutathione synthesis.

Arsenic can also induce generation of reactive nitrogen species (RNS) (reviewed in Gurr et al., 2003). The mechanisms involved are still poorly understood. Recently, As‐induction of peroxynitrite through superoxide and nitric oxide production, has been shown to contribute to As‐induced DNA repair inhibition (Zhou et al., 2019).

The induction of cell cycle alterations and perturbations of mitosis with associated aneuploidy and/or chromosomal damage (CA and MN) by iAs was confirmed in a wide repertoire of immortalised cell lines (Ganapathy et al., 2019; Jiang et al., 2013; Leffers et al., 2013; Unterberg et al., 2014) and in primary fibroblasts but not in primary epithelial cells (Xie et al., 2014). Clastogenic effects were reported at subtoxic doses (Ganapathy et al., 2019; Leffers et al., 2013; Unterberg et al., 2014).

Several lines of evidence indicate that arsenic exposure leads to compromised DNA repair (reviewed in Tam et al., 2020). This is likely to account for the enhancement by arsenite of the mutagenicity of other DNA‐damaging agents. In the last decade the molecular mechanisms of arsenic‐induced disruption of DNA repair have been further investigated. Besides the nucleotide excision repair (NER), the first DNA repair pathway shown to be inhibited by arsenic and its metabolites, the base excision repair (BER) and several players of the DNA damage response (DDR) were shown to be additional targets of arsenicals. Nollen et al. (2009) showed that in human skin fibroblasts arsenite affects the expression of the DNA damage recognition protein, XPC, resulting in decreased protein level and diminished assembly of the NER machinery within the cells after local UVC‐irradiation. In line with this finding Holcomb et al. (2017) showed that in in vitro cultures of human fibroblasts and mouse keratinocytes arsenite exerts a concentration‐dependent inhibition of the removal of UV photoproducts and a significant reduction of XPC protein levels. Cells exposure to arsenite was shown to decrease the activity, expression or protein level of key players in BER such as the DNA glycosylase, OGG1, DNA ligase IIIa and XRCC1 as analysed in cell extracts (Ebert et al., 2011). In addition, the main DNA polymerase of BER, Pol β, was shown to play an important role in repairing arsenite‐induced DNA damage and maintaining chromosomal integrity (Lai et al., 2011). Global Poly(AP‐ribosyl)ation, predominantly mediated by PARP1 and critical for immediate initiation of DDR, was previously shown to be markedly inhibited by arsenite (Hartwig et al., 2003). Ding et al. (2009) showed that arsenite suppresses the repair of UVR‐induced 8‐oxoguanine, inhibits PARP‐1 activity in cells in culture and interacts with the PARP‐1 zinc finger domain (Ding et al., 2009). The authors proposed that arsenite‐induced inhibition of PARP‐1 activity by contributing to oxidative DNA damage might account for the reported co‐carcinogenic activities of arsenic in UVR‐induced skin carcinogenesis. Arsenite was also shown to compromise ubiquitination of FANCD2, a key player of inter‐strand DNA cross link (ICL) repair, thus causing diminished recruitment of FANCD2 to ICL DNA damaged sites in the cells and inhibition of DNA ICL repair in arsenite‐treated cells in culture as shown by sensitisation to DNA ICL agents (Jiang et al., 2017). The abilities of arsenic to disrupt zinc fingers, to dysregulate ubiquitination and suppress gene expression may all negatively impact on the DDR and potentially contribute to the enhancement of mutagenic and carcinogenic effects by arsenicals (reviewed in Muenyi et al., 2015).

Human metabolites of inorganic arsenic species

Our current understanding of the significance of iAs biomethylation indicates that at least some of the toxic effects of iAs exposure depend on formation of methylated metabolites containing trivalent arsenic. As summarised in the previous EFSA Opinion (EFSA CONTAM Panel, 2009), several studies indicate that the trivalent methylated arsenic species are more cyto‐ and genotoxic than both their pentavalent counterparts and the iAs species. Similarly, they are more potent inhibitors of the activities of GSH reductase and of thioredoxin reductase. The genotoxicity of methylated arsenic species does not involve direct covalent interaction with DNA. Indirect processes such as generation of ROS and inhibition of DNA repair might be the underlying mechanisms (reviewed in Thomas et al., 2001 and Thomas, 2021). Since 2009, additional evidence has been obtained regarding the generation of free radicals by these arsenical species. In rat liver RLC‐16 cells, MMA(III) generates ROS primarily in mitochondria likely accounting for its higher cytotoxic effect as compared to DMA(III) that generates ROS in other organelles (Naranmandura, Xu, et al., 2011). This contrasts with the lack of generation of ROS by iAs(III) that is less cytotoxic in this cell system. Similar results were reported by the same group (Rehman et al., 2014) in human myeloid leukaemia HL‐60 cells where MMA(III) and DMA(III), but not iAs(III), increased oxidative stress and induced loss of mitochondrial membrane potential and apoptosis. Consistent with these data ROS generation by MMA(III) was confirmed in a variety of cells (Dopp et al., 2010; Tokar et al., 2013; Xu et al., 2017). The decreasing order of cytotoxicity from trivalent to pentavalent arsenic species, MMA(III) ≥ DMA(III) > iAs(III) ≫ iAs(V) ≫ DMA(V) > MMA(V), was confirmed in two human cell lines, on the basis of the 24 h IC50 values, by using real time cell‐electronic sensing analysis on a large number of trivalent and pentavalent arsenic species (Moe et al., 2016). MMA(III) toxicity was also significantly higher than that of iAs(III) in human brain cells in vitro (Yoshinaga‐Sakurai et al., 2020).

Ebert et al. (2011) addressed the issue of potential interference of methylated arsenical species with DNA repair enzymes showing that they affect BER by several mechanisms. The analysis of cell extracts from treated human lung cells showed that OGG1 activity was mostly affected by DMA(V), DNA ligase III protein level by arsenite and XRCC1 protein content by MMA(V). The trivalent methylated metabolites exerted strong effects on the investigated BER proteins but only at cytotoxic concentrations.

To understand the role of speciation and methylation in the toxicity of arsenic, studies were conducted with arsenic methylation proficient and deficient cell lines (Dopp et al., 2010). The non‐methylating UROtsa cells were shown to accumulate higher amounts of MMA(III) in the cytosol and to be less prone to arsenic‐induced cytotoxicity than the methylating hepatocytes where arsenic compounds are more distributed into the cell organelles. Induction of DNA breaks was detected in both cell types at concentrations as low as 5 μM MMA(III). The authors noted that this concentration lies within the concentration range of arsenic in urine in humans (5 μM iAs (III) = 650 μg/L). A correlation between DNA damage induction and free radical formation was reported although a significant increase of DNA breaks was already detected at relatively low ROS levels. The use of the hOGG1‐modified comet assay confirmed the induction of guanine base oxidation upon MMA(III) exposure of mouse bone marrow, spleen, thymus cells (Xu, McClain, et al., 2016) and activation of the DNA damage response, as detected by increased γH2AX fluorescence, in mouse thymus cells (Xu et al., 2017). No significant induction of DNA breaks was reported in HepG2 liver cells after exposure to DMA(V) (Benhusein et al., 2016) in line with a lower DNA‐damaging capacity by pentavalent species. The clastogenic activity of methylated arsenic species, MMA(III), MMA(V) and DMA(V), was confirmed by the micronucleus assay at non cytotoxic/slightly cytotoxic concentrations (Bartel et al., 2011; Meyer et al., 2015) and resulted to be higher than that of iAs (Bartel et al., 2011). Interestingly, when MMA(III)‐induced chromosomal aberrations were measured in freshly prepared splenic lymphocyte cultures upon exposure in different cell cycle phases (Kligerman et al., 2010), a significant increase was only detected when cells were treated in late G1‐ or S‐phase but not in G0‐ or G1‐phase. The authors concluded that MMA(III)‐induced cytogenetic damage is short‐lived.

Among methylated As metabolites, thio‐DMA(V) was the latest to be identified as a human metabolite and shown to be present in food. Consequently, limited data on toxicity were available until 2009. Bartel et al. (2011) carried out a toxicological characterisation of thio‐DMA(V) in cultured human lung A549 cells showing that it is highly cytotoxic, does not induce DNA breaks and produces a small but significant increase of bi‐ and multi‐nucleated cells at slightly cytotoxic doses. Increased frequency of bi‐ and multi‐nucleated cells was also reported by Leffers et al. (2013) in UROtsa cells for both thio‐DMA(V) and DMA(V) at incipient cytotoxic concentrations, indicating cell cycle arrest and disturbance in mitosis. Neither increase of micronuclei frequency nor of bi‐ or multi‐nucleated cells was reported by Unterberg et al. (2014) after long exposure times (from 7 to 21 days) of UROtsa cells to subtoxic doses of thio‐DMA(V), while arsenite was confirmed to be clastogenic at subtoxic doses. The toxic mode of action of thio‐DMA(V) was further characterised in human bladder cells (Ebert et al., 2014) confirming its high cytotoxicity and the lack of induction of DNA breaks and oxidative base modifications. Interestingly, thio‐DMA(V) specifically inhibited H2O2‐induced cellular poly(ADP‐ribosyl)ation (at a 35,000‐fold lower concentration than arsenite), suggesting that, by inhibiting DNA repair, it might promote genotoxicity. Thio‐DMA(V) was also among the most toxic arsenic compounds in human bladder cancer EJ‐1 cells, similar to trivalent DMA(III) (Naranmandura, Carew, et al., 2011). Cellular exposure to thio‐DMA(V) resulted in reduced protein expression of p53 and p21, increased DNA breaks, as measured by the alkaline comet assay, and increased intracellular hydroxyl radicals. The authors concluded that in vitro thio‐DMA(V) is the most toxicologically potent arsenic species, and it might be relevant to arsenic‐induced carcinogenicity in the urinary bladder.

Cell transformation

Previous work showed that MMA(III) and DMA(III) can transform human urothelial cells in culture (Bredfeldt et al., 2006). Waalkes' laboratory confirmed the oncogenic potential of MMA(III) in a cell transformation assay in both methylation‐deficient (prostate) and proficient (liver) cells that acquired cancer cell characteristics concurrently with oxidative stress induction during chronic MMA(III) exposure (up to 30 weeks) (Tokar et al., 2013). In a previous work the same laboratory (Kojima et al., 2009) showed that iAs can also induce a malignant phenotype in prostate methylation‐deficient cells but without generation of oxidative damage while MMA(III) does so more rapidly and with ROS induction, suggesting different underlying mechanisms. Several studies report that cell transformation induced by long‐term arsenic exposure involves epithelial‐to‐mesenchymal transition (EMT) (Chang & Singh, 2019; Wang et al., 2013), which plays a role in tumour progression and metastasis.

Table 8 provides an overview of the in vitro genotoxicity tests with arsenic.

3.1.3.2. In vivo genotoxicity

In 2014, Takumi et al. (2014) published the first study addressing the mutational activity and mutational spectrum of iAs in animal models. By using the gpt mutation assay in gpt delta mice they showed that oral administration of arsenite induces a significant increase in mutation frequency in the liver and sequence analysis revealed a predominance of GC to TA transversions which is a hallmark of 8‐oxoguanine mutagenesis (Moriya & Grollman, 1993). To support the suggestion that DNA base oxidation might underly the mutagenic activity of arsenic they showed that under the same experimental conditions increased levels of 8‐oxoguanine are induced in the livers of arsenite‐exposed mice. A successive study (Fujioka et al., 2016) was unable to confirm the mutational activity of arsenic in vivo by analysing gpt mutations in urinary bladder epithelium and liver of rats exposed orally to similar arsenite doses for longer exposure times. This discrepancy might be explained by differences in iAs toxicokinetics between rats and mice (see Section 3.1.1.1 Metabolism). In addition, the exposure regimen was poorly tolerated by the test animals as shown by the significant reduction in water intake and food consumption from week 2 and the significant reduction in body weight. No effects on the gpt mutant frequencies or mutation spectrum were reported. In this study, in addition to the gpt mutational assay, the Spi assay was used. This assay efficiently detects intra‐chromosomal deletions but not inter‐ chromosomal translocations or intrachromosomal megabase deletions. No long DNA fragment deletions were detected in the bladder or livers of treated rats. Negative results were also obtained following administration of DMA(V).

A few studies published since 2009 were identified regarding the genotoxic effects of iAs in animal cell/tissues. Mehta and Hundal (2014) addressed the question of whether iAs induces chromosomal aberrations in bone marrow cells of female rats after chronic exposure to low doses of arsenite (10–50 μg/L) for 30–60 days. The results show a statistically significant dose‐related increase of chromosomal aberrations, but the conclusions are weakened by the lack of important experimental information (e.g. number of metaphases scored, mitotic index).

DNA damage, as detected by alkaline comet assay, was shown to occur in tissues, in particular ovary and uterus, of mice and rats orally exposed to arsenite (Dash et al., 2018; Nath Barbhuiya et al., 2020). Arsenite‐induced oxidative stress was suggested to be involved (Dash et al., 2018). Inhibition of DNA repair by arsenic is well documented in cells in culture. Osmond et al. (2010) demonstrated the same effect in mice upon consumption of arsenic in lactational milk or drinking water. In particular, the reduction of BER gene transcript levels was detected in lung tissue harvested from treated adults and altered transcript levels were also reported in neonates feeding from exposed lactating mothers.

Arsenic trioxide is a well‐known toxic metalloid mostly used as an anti‐cancer drug. In cells in culture, it induces more severe genotoxic effects, apoptosis and oxidative stress than sodium arsenite (Jiang et al., 2013). Differences in toxicokinetics between the two arsenical compounds might account for the stronger toxic effects induced by arsenic trioxide (Liu et al., 2021). Three studies of genotoxicity in rodents upon oral administration of arsenic trioxide have been identified and they are reported below. Mice orally exposed to arsenic trioxide for 15 days showed significant increases of chromosomal aberrations in bone marrow cells (Kesari et al., 2012) and of micronuclei in erythrocytes (Khan et al., 2013) when exposed to a range of doses of arsenic (from 0.3 to 30 μg/kg per day). Notwithstanding some experimental limitations (e.g. low number of metaphases scored, lack of cytotoxicity data) the effects are statistically significant and dose‐related. A recent study (Nava‐Rivera et al., 2021) addressed the transgenerational (F1, F2, F3) genotoxic potential of arsenic in a rat model. Rats chronically exposed to arsenic in drinking water (As2O3, 1 mg/L) were mated to produce the arsenic lineage (F1, F2, F3). The induction of DNA breaks was measured by the comet assay in WBC. A significant increase in the level of DNA breaks (measured as percentage of DNA in the tail) in the arsenic‐exposed lineage compared to the control lineage was found in all generations (F0–F3) in both females and males. The genotoxic effects decreased across generations but without loss of statistical significance compared to the control group. Upon chronic exposure to arsenic, transgenerational effects on global DNA methylation and reproductive phenotype were also reported, suggesting that early life exposure to arsenic may affect the health of later generations. An overview of the in vivo genotoxicity studies is provided in Table 9.

In conclusion, iAs does not interact directly with DNA but induces DNA base oxidation and both DNA SSB and DSB. The mutational spectrum of oxidised guanine bases, characterised by G > T transversions, has been detected in animal models. DSB are one of the most deleterious types of DNA lesions. Arsenic induces clastogenic and aneugenic effects both in vitro and in vivo as measured by chromosomal aberrations, micronuclei and aneuploidy with evidence of dose–response effects. In addition, iAs effectively inhibits DNA repair both in vitro and in vivo thus enhancing its own genotoxic potential and acting as a co‐mutagen (EFSA CONTAM Panel, 2009). All these effects occur in vitro at subtoxic concentrations. The methylation of iAs, particularly to trivalent methylated species, should be regarded as an activation process that forms more reactive species, which exert stronger cyto‐ and genotoxic effects (induction of DNA breaks and clastogenic effects), inhibit DNA repair, promote ROS generation and induce cell transformation. Thio‐DMA(V) is emerging as the most cytotoxic arsenic metabolite. The transgenerational genotoxic potential of arsenic has been shown in a rat model upon parental chronic exposure to As2O3 (Nava‐Rivera et al., 2021).

3.1.3.3. Mode of action for genotoxicity

Arsenic species exposure causes oxidative stress which is likely involved in multiple effects by altering signalling pathways and epigenetic modifications or causing direct oxidative damage to molecules.

Arsenic species induce oxidative stress

The mechanisms that have been proposed for arsenic species induced oxidative stress include: (i) As‐induced mitochondrial toxicity, (ii) generation of RONS (reactive oxygen and nitrogen species) during formation of intermediate arsine species (e.g. metabolic processing of DMA), (iii) oxidation of arsenite to arsenate and (iv) interference with cellular antioxidative enzymes such as SOD, CAT and GSH which indirectly result in an increase of RONS levels (reviewed in Hu et al., 2020). Induction of RONS by iAs itself at low doses is questionable, more efficient inducers are the iAs metabolites (see Section on Genotoxicity). Arsenic species induced DNA damage is a mirror of the genotoxic potential of RONS.

Oxidation of DNA guanines occurs and in vivo mutational spectra indicate the typical fingerprint of oxidative DNA damage, i.e. G > T transversions. iAs is an efficient inducer of both single and double DNA strand breaks. Of the many different classes of damage, DNA DSB are the most dangerous since their processing may lead to mutations, loss of heterozygosity and chromosome rearrangements that results in cell death and cancer (Cannan & Pederson, 2016).

Based on the current literature, we can envisage different mechanisms for DSB generation by iAs: (i) during the processing of SSB left unrepaired in the S‐phase (Ying et al., 2009); (ii) during attempted repair of oxidised DNA bases when they occur simultaneously on opposing strands (Cannan & Pederson, 2016); or, (iii) during all cell cycle phases due to in situ production of free radicals which may generate breaks on opposing strands (Litwin et al., 2013). In this case the short‐lived, but highly reactive hydroxyl radicals may react with nearby DNA, producing SSBs. Closely opposed SSBs, created by either route, may spontaneously convert into a DSB. In the simplest version of the SSB to DSB conversion hypothesis, each SSB forms independently (reviewed in da Silva, 2021). In all three scenarios, the inhibition of DNA repair by iAs is the promoting event since the probability of DSB generation increases if SSB persist for a large fraction of the cell cycle.

Arsenic species interfere with the DNA damage response (DDR)

Interference with DDR occurs at two levels: by inhibition of DNA repair and by interference with cell cycle control and the apoptotic pathways. The disturbance of DNA repair systems has been observed at very low concentrations of iAs and its metabolites. Many DNA repair proteins contain redox‐sensitive cysteine residues within zinc finger domains and therefore arsenite‐induced oxidative stress can result in inactivation of their enzymatic function (see Section 3.1.3.1). This has been demonstrated for adenosine diphosphate poly‐ribose polymerase‐1 (PARP1), which could be inhibited by peroxynitrite‐mediated S‐nitrosation of its zinc finger cysteine(s) (Zhou et al., 2019). In the low nanomolar range, arsenite and some of its metabolites ‐MMA(III and DMA(III)‐, are sufficient to interfere with the activity of PARP‐1, thereby increasing SSB and DSB formation in cells in culture. Furthermore, all three trivalent arsenicals inhibited isolated PARP1, indicating a direct interaction with this enzyme (reviewed in Hartwig et al., 2020). Interestingly, one of the three zinc fingers of PARP‐1 (ZnF1) exerts a quite low affinity toward zinc (Bossak et al., 2015) and zinc release due to oxidative stress has been recently shown to regulate PARP1 activity (Wedler et al., 2021). If the first zinc finger of PARP‐1 is not occupied by zinc under normal cellular conditions, this may also explain the particularly high sensitivity of PARP‐1 toward toxic metal ions such as arsenite.

In addition, iAs has been shown to inhibit the main DNA repair mechanisms for both SSB and DSB (i.e. BER, HR, NHEJ) (reviewed in Tam et al., 2020). Arsenite can disrupt DSB repair by multiple mechanisms such as by inhibiting histone epigenetic modifications, which leads to compact chromatin structures unfavourable for DNA DSB repair, or by inactivating the myriad of zinc finger proteins involved in post‐translational modifications of proteins that control DDR and transcription of DNA repair genes (reviewed in Tam et al., 2020). Epigenetic alterations appear also to be relevant at very low concentrations (see Section 3.1.2.3). It is of note that some iAs metabolites have been shown to disturb similar but also different DDR targets (see Section 3.1.3.1). Thus, under mixed exposure conditions, which occur when iAs is metabolised, genomic stability is affected by different pathways.

Arsenic species modulate cellular signalling pathways that affect diverse processes such as cell proliferation, differentiation and apoptosis. Arsenic may influence cell cycle regulators at either the transcriptional or post‐translational levels or affect specific protein–protein interactions. Arsenic has been shown to interfere with protein ubiquitination of cell regulatory proteins (e.g. cyclins, p53), thus playing a key role in cell cycle control. In addition, arsenic may activate the intrinsic and extrinsic caspase pathways, and induce cell apoptosis (reviewed in Tam et al., 2020).

Induction of clastogenic and aneugenic events

iAs is a powerful inducer of CA and MN in both in vitro and in vivo models as well as in humans. In a comparative analysis of occupational and environmental studies reporting MN frequency in lymphocytes of individuals exposed to different genotoxic agents (including pesticides, herbicides, PAHs, formaldehyde, etc.), metals showed the most pronounced fold increase of MN frequency over background with exposure to arsenic (seven studies) being the most effective (6.5‐fold increase) (Nersesyan et al., 2016).

MN are extra‐nuclear bodies originating from acentric chromatid/chromosome fragments or whole chromatids/chromosomes that were not incorporated into the nucleus after cell division. The multiple effects of iAs are compatible with both mechanisms of MN formation. iAs may induce MN containing acentric chromatids/chromosomes fragments by causing DNA breaks and inhibiting their repair. On the other side, disruption of mitotic progression by arsenic species (e.g. through interaction with the mitotic spindle or affecting the methylation pattern of centromere satellite sequences) may generate MN with whole chromatids/chromosomes.

MN as markers of exposure are not very sensitive due to the high baseline level in the healthy population and to the confounding factors (age, sex, diet and lifestyle) that can significantly affect their frequency (Fenech & Bonassi, 2011). In the As‐exposed populations an approximately three‐fold increase of MN frequency is observed when the concentrations of As in drinking water are above 50 μg/L. Conversely, other markers of genotoxicity such as induction of DNA breaks reveal genetic damage at lower As concentrations (positive in a range of concentrations from 10 to 40 μg/L) (see Section 3.1.2.2). This is expected considering that the induction of DNA breaks precedes the formation of MN. Several studies consistently report increased MN frequency in As‐exposed populations revealing the induction of genetic damage that may generate genetic instability. New knowledge obtained over past few years indicates that MN are not only markers of DNA damage and aneuploidy but also inducers of chromosomal hypermutation and sources of pro‐inflammatory DNA leaking from disrupted MN (reviewed in Fenech et al., 2021). The inflammatory consequences of MN formation and their disruption provide a further important explanation why MN are linked prospectively not only with cancer but also with other inflammation‐driven diseases.

Induction of gene mutations

Inorganic arsenic alone is a weak inducer of gene mutations. If diminished DNA repair and elevated oxidative DNA damage constitute the major mechanisms of iAs carcinogenesis, one would expect to observe elevated rates of mutations in iAs‐exposed cell and animal models. Clear evidence indicates that arsenite increases the mutagenicity of other DNA damaging agents such as UV radiation, but it shows a low, if any, mutagenic activity in bacterial as well as in mammalian test systems. However, limitations of the mutation assay systems used, mostly designed to detect point mutations, might be responsible for this failure. Indeed, Hei et al. (1998), by employing a human–hamster hybrid cell assay in which both intragenic and multilocus mutations are detectable, demonstrated that arsenite could induce, in a dose‐dependent manner, mutations (mostly large deletions). They also observed that the mutagenicity of arsenite could be diminished markedly by cotreating cells with a radical scavenger, dimethyl sulfoxide, thus suggesting that RONS species are involved. More recently, by using high‐resolution genomic profiling, Martinez et al. (2010) analysed copy number variations (CNVs) in lung squamous cell carcinomas (SCC) from patients chronically exposed to arsenic with or without smoking history. The results of these specimens were compared with those derived from a panel of lung SCC from an unrelated population without known exposure to As. Multiple CNVs were identified in association with arsenic exposure and were not attributable to smoking status or germline CNVs. Recent advances in next‐generation sequencing, should be applied to unveil how arsenic exposure leads to mutagenesis in animal models or human subjects.

Summary on genotoxicity

Inorganic arsenic and its metabolites, via generation of oxidative stress, induce DNA base oxidation, DNA single and double strand breaks and clastogenic and aneugenic events both in vitro and in vivo. The same events are reported in humans upon chronic iAs exposure.

The Genetic Toxicology Technical Committee (GTCC) of the Health and Environmental Sciences Institute (HESI) has developed an adverse outcome pathway (AOP) that links oxidative DNA damage to two adverse outcomes (AO): mutations and chromosomal aberrations (Cho et al., 2022). Both events have been shown to occur upon exposure to iAs. Because of the lack of direct interaction with DNA, iAs is classified as an indirect genotoxin. However, iAs presents a unique feature, that is its extraordinary ability to inhibit DNA repair at very low concentrations in vitro and at low exposure levels in human studies as inferred from the elevated levels of DNA damage. The inhibition of DNA repair can potentially exacerbate the genotoxic effects not only of exogeneous DNA damage but also of endogenously induced DNA damage that is an important source of genomic instability in cancer (reviewed in Tubbs & Nussenzweig, 2017). This is a detrimental combination that makes of iAs a ‘special’ indirect genotoxin. Even though the interactions with the DNA repair/DDR system are based on protein interactions, and may, therefore, follow non‐linear dose–response relationships, it should be considered that the response of cells under chronic exposure to iAs is similar to that of DNA repair/DDR‐defective cells that present increased genomic instability and increased risk of disease.

3.1.4. Carcinogenicity studies in animals

EFSA CONTAM Panel (2009) noted that most oral carcinogenicity studies on various iAs species gave negative results in mice, rats and dogs, and that an important exception was the studies in which pregnant mice were treated with sodium arsenite in drinking water, and tumours were observed in the adult offspring, demonstrating transplacental carcinogenesis (Liu & Waalkes, 2008; Waalkes et al., 2007). In addition, several studies indicated that iAs is co‐carcinogenic (EFSA CONTAM Panel, 2009). As noted in Section 1.2.3, more recent IARC evaluations have concluded that ‘in view of the overall findings in animals, there is sufficient evidence in experimental animals for the carcinogenicity of iAs compounds’ (IARC, 2012). This conclusion is based on evaluation of studies on a range of arsenic compounds (e.g. dimethylarsenic acid, calcium and sodium arsenate, calcium and sodium arsenite, gallium arsenide, arsenic trioxide and trimethylarsine oxide) performed using different routes of administration (oral, intratracheal and intravenous).

In the early studies of Waalkes and colleagues, noted in the EFSA CONTAM Panel (2009), pregnant CH3 and CD1 mice were treated with drinking water containing sodium arsenite at concentrations of 42,500 or 85,000 μg/L from GD 8 to 18, and the offspring were observed for up to 2 years. The male offspring of C3H mice developed tumours of the liver and adrenal cortex in a dose‐related fashion during adulthood. Female C3H offspring showed dose‐related increases in ovarian and lung tumours. Male CD1 offspring developed tumours of the liver and adrenals while females developed tumours of the urogenital system, ovary, uterus and adrenal.

Since the previous EFSA Opinion, Waalkes and colleagues have published new studies involving ‘whole‐life’ exposure, in which mice were exposed via drinking water prior to breeding, during pregnancy and lactation and the offspring were similarly exposed after weaning. Exposure to sodium arsenite at 0, 6000, 12,000 or 24,000 μg/L in the drinking water resulted in dose‐related increases in lung adenocarcinoma (both sexes), hepatocellular carcinoma (both sexes), gallbladder tumours (males) and uterine carcinomas. Dose‐related increases in ovarian tumours (including carcinomas), and adrenal tumours (both sexes) were increased at all doses (Tokar et al., 2011). In a subsequent study, designed to encompass concentrations of iAs more relevant for human exposure, CD1 mice were exposed to 0, 50, 500 or 5000 μg/L sodium arsenite in drinking water for 3 weeks prior to breeding, during pregnancy and lactation and the offspring were exposed after weaning for up to 2 years (Waalkes et al., 2014). In males, a statistically significant increase in lung adenomas was reported in males at 500 μg/L, and in lung carcinomas at 50 μg/L. In females there was a statistically significant increase in lung adenomas at 50 μg/L. Survival was decreased in this female dose group but not in other groups of females or males. No increase in lung tumours was reported for the other dose groups, and tumours at sites other than the lung are not described. The authors described the dose–response relationship as ‘unusual’ and could not explain it. Cohen et al. (2014) commented on the lack of consistency between the results reported in Waalkes et al. (2014) and Tokar et al. (2011) and suggested that the conflicting results could be due to the high variability in incidence of lung tumours in CD‐1 mice, noting that the incidences were within the range of historical controls reported by Charles River.

Studies of gestational arsenic exposure have been reported by one other research group (Nohara et al., 2012, 2016). In Nohara et al. (2012), pregnant C3H/HeN mice were given drinking water containing 0 or 85,000 μg/L sodium arsenite from day 8 to 18 of gestation. The authors reported that arsenic exposure resulted in a ‘tendency’ toward a higher number of mice with hepatic tumours and a greater multiplicity and size of the tumours compared to the control group. However, statistical analysis was not reported and no tissues other than the liver were examined. In Nohara et al. (2016), pregnant C3H/HeN mice were also given drinking water containing 0 or 85,000 μg/L sodium arsenite from day 8 to 18 of gestation, followed by a reciprocal crossing experiment among the control F1 males and females and arsenite F1 males and females. Male and female F1 mice were mated at 10 weeks of age and the liver tumours were examined in the F2 males at 75–82 weeks. There was a significantly higher incidence of liver tumours in the F2 male offspring of arsenite F1 males than in those of the control F1 males. In contrast, the tumour incidence of the F2 male offspring of arsenite F1 females and the control F1 females were not significantly different. The authors concluded that the tumour‐augmenting effect of gestational arsenite exposure is transmitted to the F2 males via the F1 male offspring, but not via the F1 female offspring.

Garry et al. (2015) reported a detailed analysis of the toxicology literature evaluating the role of in utero arsenic exposure in carcinogenesis. They concluded that the available data did not ‘provide evidence of a causal link between in utero arsenic exposure and cancer or indicated early life‐stage susceptibility to arsenic‐induced cancer, particularly at environmentally relevant doses’.

The CONTAM Panel concluded that although some studies have shown increased tumour incidences following oral (drinking water) administration of iAs, the results of different studies with respect to tumour sites and doses are inconsistent and do not provide a robust basis for use in the risk assessment of iAs.

3.2. Observations in humans

3.2.1. Short‐term effects

In its previous Opinion (EFSA CONTAM Panel, 2009) the CONTAM Panel reported that the acute oral lethal dose of arsenic ranges between 1 and 5 mg arsenic/kg bw and that acute and subacute arsenic poisoning can affect almost all physiological systems of the body including the gastrointestinal, cardiovascular, renal and nervous systems, and to a lesser degree the respiratory, hepatic, haematological and dermal systems. They concluded that while human case reports concerning iAs can provide important information for the clinical course of the poisoning such reports generally do not provide information on dose response which is necessary for risk assessment.

3.2.2. Chronic effects

In its previous Opinion (EFSA CONTAM Panel, 2009) the CONTAM Panel concluded that data from experimental animals cannot be used for risk characterisation because of important species differences in iAs toxicokinetics. The available epidemiological studies related to arsenic in drinking water, or in some instances biomarkers of exposure. Total dietary exposure to iAs was not specifically measured in these studies. The main adverse effects reported to be associated with chronic oral exposure to iAs in humans were skin lesions, cancer, developmental toxicity, neurotoxicity, cardiovascular diseases, abnormal glucose metabolism and diabetes. Neurotoxicity was predominantly reported from acute exposure upon poisoning or suicide, or at high concentrations in drinking water. The evidence of associations of arsenic exposure with cardiovascular diseases (i.e. Blackfoot disease, peripheral vascular disease, coronary heart disease, myocardial infarction and stroke) and diabetes in areas with relatively low levels of iAs exposure was inconclusive. There was also emerging evidence of negative impacts on fetal and infant development, particularly reduced birth weight. The CONTAM Panel concluded that there was sufficient evidence to assume a causal relationship of arsenic exposure with skin lesions and cancers of the urinary bladder, lung and skin.

3.2.2.1. Selection of studies

Although reviews and meta‐analyses were also reviewed, only original cross‐sectional, case–control and cohort studies characterised by estimates on the individual or group level of exposure to iAs were considered. Exposure metrics accepted were estimates of iAs ingestion (usually principally from water), or arsenic drinking water concentrations (assuming 100% iAs), or biomarkers of iAs (which required speciated arsenic metabolites). Only studies that include study subjects with long‐term low to moderate levels of arsenic defined as concentrations of arsenic in water of less than ~ 150 μg/L or biomarker concentrations estimated to results in equivalent doses, were considered. One reason for this was that studies used in previous risk assessments based on arsenic in drinking water showed dose–response relationships with skin lesions and cancer outcomes at arsenic concentrations in water well below 150 μg/L. In addition, studies with only high exposures are much less informative for deriving dose response relationships at low levels. Studies on occupational arsenic exposure were excluded, as well as studies based on total As, unless knowledge of the arsenic sources allowed a reliable estimate of inorganic As. This required reliable information that the relative contribution from recent seafood intake was negligible.

The reliability of all epidemiological studies was assessed in terms of design, exposure assessment, assessment of outcomes, confounding and risk of other sources of bias. For four outcomes, studies exclusion criteria were applied from the start: Studies of lung or urinary bladder cancer were excluded if smoking data were not available. Studies of type 2 diabetes were excluded if data were not available on smoking and BMI. Studies on skin cancer were included only if sun exposure or skin sensitivity to UV light was considered or could be assumed not to affect results. Information on possible use of tanning bed/solarium was not available. Studies on neurodevelopment were excluded if data were not available for key covariates that are important for neurodevelopment. The covariates were (based on Lanphear et al. (2005) that evaluated low‐level exposure to lead and neurodevelopment): (1) sex and age, anthropometry or nutrition, (2) socioeconomic status (SES) or HOME (Home Observation Measurement of the Environment) score and (3) parental education/intelligence quotient (IQ). Because of some overlap of these factors (e.g. between SES and parental education, and between nutrition and SES), the selected studies were required to have adjusted for at least two of these groups of covariates. The CONTAM Panel notes that study populations may have been exposed also to other contaminants than iAs but found no clear indications that there was residual confounding from such exposure.

For multiple studies from the same cohort, we included only the publication with the longest follow‐up time provided that it included appropriate data on exposure, outcomes and covariates. The inclusion and exclusion criteria for epidemiological studies are presented in Table 10 below.

TABLE 10

Criteria for epidemiological studies of long‐term effects in humans.

Study designIn

Cross‐sectional studies

Cohort studies

Case–control studies

Study characteristicsInDuration of exposure > 6 months. For continuous outcomes > 100 subjects and for discrete outcomes > 5, Exposures to arsenic concentrations < 150 μg/L in drinking water or corresponding exposure level for arsenic biomarkers
PopulationInAny country. Any age groups
Exposure estimateIn

iAs intake per day

As in drinking water

iAs in urine and its metabolites

Total urinary iAs (u‐tiAs), i.e. sum of iAs and metabolites MMA and DMA

Other iAs biomarkers that can be transformed into iAs intake

Out

Intake of total As

Total As in urine or blood, unless arsenobetaine could be excluded

Studies on inhalation of arsenic

Studies with occupational exposure

OutcomesInAny long‐term adverse outcomes
CovariatesOut

Lack of age or sex

Lack of smoking data for lung and bladder cancer outcome

Lack of data on sun exposure or skin sensitivity to UV light for skin cancer

Lack of smoking or BMI for T2D outcome

Lack of covariates (child characteristics/parents' education or maternal intelligence/socioeconomic status) for neurodevelopment

LanguageInEnglish
OutNon‐English
Time periodInFrom 1/1/2009 to 18/7/2022 and papers described reviewed by EFSA in 2009
Publication typeInPeer‐reviewed original research, systematic reviews, meta‐analyses a
OutExpert opinions. Editorials. Letters. PhD theses. Conference proceedings. Conference abstracts.

Abbreviations: As, arsenic; BMI, body mass index; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; EFSA, European Food Safety Authority; iAs, inorganic arsenic; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; PhD, Doctor of Philosophy; T2D, type 2 diabetes; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA) ; UV, ultraviolet.

a Such publications have not been used for risk assessment.

3.2.2.2. Cancers

Skin cancer

In 1973, IARC concluded that there was sufficient evidence that arsenic and arsenic compounds caused cancers of the skin (and liver) (IARC, 1973) and assigned it to Group 1 (IARC, 1979). The causal association of arsenic exposure with skin cancer has been confirmed in a number of subsequent re‐evaluations, most recently in 2009 (IARC, 2012; Straif et al., 2009). The CONTAM Opinion of 2009 (EFSA CONTAM Panel, 2009) noted that earlier risk assessments were based on ecological studies from Taiwan (primarily in the southwest endemic arsenic region) that showed strong dose‐related effects of village drinking water arsenic concentrations on skin cancer incidence, prevalence and mortality with extrapolation to lower levels of exposure (Tseng et al., 1968). In 2009, the CONTAM Panel focussed on epidemiological studies on skin cancer with lower levels of exposure (e.g. including drinking water concentrations below 100 μg/L) and derived a Reference Point equivalent to 1–2 μg/L As in drinking water from the study of Karagas et al. (2002).

For the present Opinion, the CONTAM Panel identified 15 studies from a literature search (see Section 2 on Methodology). Out of these, 10 studies did not meet the inclusion criteria (Alshana et al., 2022; Bedaiwi et al., 2022; Choudhury et al., 2018; Engström et al., 2015; Gossai et al., 2017; Kim et al., 2017; Knobeloch et al., 2006; Langston et al., 2022; Wheeler et al., 2013; Zhang et al., 2016). Five studies that fulfilled the inclusion criteria were considered further (see Table 11 below).

TABLE 11

Key epidemiological studies on skin cancer in humans in relation to ingested inorganic arsenic exposure.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic concentration/exposureResultsAdditional information/confounders

Karagas et al. (2001)

USA (New Hampshire)

Case–control study

Histologically confirmed, incident BCC and SCC

BCC/SCC/controls

281/155/263

156/64/136

92/33/73

22/14/26

10/5/11

26/13/15

Concentration in toenail (μg/g)

0.009–0.089

0.090–0.133

0.134–0.211

0.212–0.280

0.281–0.344

0.345–0.81

OR (95% CI) BCC

1.00 (reference)

1.01 (0.76, 1.35)

1.06 (0.74, 1.51)

0.72 (0.40, 1.31)

0.75 (0.31, 1.81)

1.44 (0.74, 2.81)

OR (95% CI) SCC

1.00 (reference)

0.93 (0.64, 1.34)

0.98 (0.61, 1.58)

1.10 (0.55, 2.21)

1.00 (0.33, 3.01)

2.07 (0.92, 4.66)

Adjusted for age and sex. Other covariates, such as smoking and UV light, evaluated but no appreciable effect

Beane Freeman et al. (2004)

USA

Case–control study

Cutaneous melanoma

326/329

52/82

58/83

95/82

121/82

Concentration in toenail (μg/g)

≤ 0.020

0.021–0.039

0.040–0.083

≥ 0.084

OR (95% CI)

1.0

1.0 (0.6, 1.6)

1.7 (1.1, 2.7)

2.1 (1.4, 3.3)

Patients with colorectal cancer as controls. Adjusted for age, gender and education. Effect of history of sunburn considered

Baastrup et al. (2008)

Denmark

Cohort study

Nonmelanoma and melanoma of the skin1010 nonmelanoma/147 melanoma/56,378

Time‐weighted average water concentration (μg/L) median CI (95% CI)

0.7 (0.3–2.1)

IRR (95% CI)

Nonmelanoma

0.99 0.94–1.06

IRR (95% CI)

Melanoma

0.80 0.59–1.08

Adjusted for many factors including UV exposure and smoking

Leonardi et al. (2012)

Hungary, Romania, Slovakia

Case–control study

Histologically confirmed, incident BCC529/540

Lifetime average water As concentration (μg/L)

0.00–0.68

0.68–0.98

0.98–7.00

7.10–19.43

19.54–167.29

Peak daily As dose rate (μg/day)

0.00–0.73

0.73–1.48

1.48–9.09

9.09–32.23

32.23–242.14

Cumulative As dose (g)

0.00–0.01

0.01–0.03

0.03–0.13

0.13–0.55

0.55–4.46

OR (95% CI) BCC

1.00 (reference)

1.39 (0.89, 2.19)

1.20 (0.77, 1.88)

1.73 (0.97, 3.11)

3.03 (1.70, 5.41)

OR (95% CI) BCC

1.00

0.91 (0.59, 1.39)

1.55 (1.00, 2.41)

1.76 (1.01, 3.07)

2.50 (1.39, 4.49)

OR (95% CI) BCC

1.00

1.09 (0.72, 1.67)

1.46 (0.93, 2.27)

1.76 (1.02, 3.04)

2.63 (1.45, 4.78)

Controls were general surgery, orthopaedic and trauma patients. The controls were frequency matched to all potential cancer cases by sex, 5‐year age band and residence in the same county/region of the study area

Adjusted for county, age and sex, education, skin response to 1‐h midday sun and skin complexion

Smoking and BMI data available but did not influence the results and not included in the final models

Gilbert‐Diamond et al. (2013)

USA (New Hampshire)

Case–control study

Histologically confirmed, incident SCC

470/447, Included in the analysis:

323 cases and 319 controls with no seafood intake

u‐tiAs (μg/L)

< 3.36

3.36 to < 5.31

≥ 5.31

Continuous (per ln‐transformed ΣAs μg/L increase)

OR (95% CI) SCC

1 (reference)

0.94 (0.60, 1.45)

1.43 (0.91, 2.27)

1.37 (1.04,1.80)

Population‐based: controls frequency matched to cases on sex and age. Adjusted for sex, age, BMI, education, smoking status, skin reaction to chronic sun exposure and urinary creatinine concentration

Abbreviations: As, arsenic; BCC, basal cell carcinoma; BMI, body mass index; CI, confidence interval; iAs, inorganic arsenic; IRR, incidence rate ratio; n, number; OR, odds ratio; SCC, squamous cell carcinoma; USA, Unites States of America; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA); UV, ultraviolet.

Three of these studies were already reviewed in the 2009 Opinion. In a population‐based case–control study of skin cancers (both basal cell and squamous cell carcinoma) from the U.S., an increased risk of squamous cell carcinomas in the highest exposure category was reported in relation to arsenic measured in toenail clippings (Karagas et al., 2001). In a geographic information system (GIS) analysis of water arsenic and nonmelanoma skin cancer and melanoma in the European Prospective Investigation into Cancer and Nutrition (EPIC) cohort in Denmark, no association was observed after adjustment for region (Baastrup et al., 2008). The CONTAM Panel noted that exposure in this study was based on relatively crude aggregated estimates of water arsenic at low levels (median 0.7 μg/L), with low contrast (only 1% above 5 μg/L). A population‐based case–control study from the U.S. investigated the risk of melanoma skin cancer in relation to toenail arsenic concentration and found a dose‐related increase in risk (Beane Freeman et al., 2004).

Two studies that fulfilled the inclusion criteria were published after the 2009 Opinion. In the Arsenic Health Risk Assessment and Molecular Epidemiology (ASHRAM) study in Hungary, Romania and Slovakia, the relative risk of basal cell carcinoma (BCC) cases showed a dose–response in relation to all three indices of long‐term iAs exposure (lifetime average concentration, cumulative and peak dose), with and without controlling for potential confounders, including indices of UV exposure (Leonardi et al., 2012). An odds ratio of 1.18 (95% CI = 1.08, 1.28) was associated with each 10 μg/L increase in average lifetime drinking water concentration of arsenic (Leonardi et al., 2012). A population‐based case–control study in the U.S. found increased risk of squamous cell carcinomas of the skin with increasing u‐tiAs (OR = 1.37, 95% CI = 1.04, 1.80 per unit increase of ln‐transformed concentrations of arsenic). There was an increasing trend in the odds ratios for squamous cell carcinomas with each urinary arsenic metabolite, with the strongest association for MMA (Gilbert‐Diamond et al., 2013). Exposure to UV radiation is a confounding factor for skin lesions. All studies assessed data on sun exposure or skin sensitivity to UV light in one way or another. However, the studies did not evaluate whether the skin cancer occurred in a UV‐exposed or UV‐unexposed area.

In summary, the epidemiological studies provide sufficient evidence for an association between low to moderate exposure to iAs and both basal cell carcinoma and squamous cell carcinomas of the skin. The data on possible associations with melanoma are not sufficient to reach conclusions.

The results for the key studies are summarised in Table 11.

Bladder cancer

In 2002, the IARC concluded that there was sufficient evidence in humans that arsenic in drinking‐water causes cancer of the urinary bladder (as well as of the lung and skin) and assigned it to Group 1 (IARC, 2004). The assessment was based on ecological studies from Taiwan, Chile and Argentina that were supported by evidence from cohort and case–control studies in Taiwan. The 2009 Opinion (EFSA CONTAM Panel, 2009) focussed on epidemiological studies on bladder cancer in humans informing on dose–response at exposure levels below 100 μg/L in drinking water. The CONTAM Panel noted that some of the studies published since the IARC report supported an excess risk of bladder cancer whereas others did not.

For the present Opinion, the CONTAM Panel identified 26 studies from a literature search. Sixteen of these studies were not considered further as they did either not fulfil the inclusion criteria or were not relevant for this risk assessment (Amin, Stafford, & Guttmann, 2019; Andrew et al., 2009; Chen, Grollman, et al., 2021; Chung et al., 2019; Ferreccio, Smith, et al., 2013; Koutros et al., 2018; Krajewski et al., 2021; Lamm et al., 2013; Liao et al., 2009; Melak et al., 2014; Mendez Jr et al., 2017; Moazed et al., 2021; Saint‐Jacques et al., 2018; Signes‐Pastor, Scot Zens, et al., 2019; Steinmaus et al., 2014a; Tsai, Kuo, et al., 2021). Ten studies met the inclusion criteria (see Section 3.2.2.1).

Three of these studies were already reviewed in the 2009 Opinion: studies in North East Taiwan (Chiou et al., 2001), New Hampshire, USA (Karagas et al., 2004), which were used to calculate reference points in the 2009 Opinion (see Table 12) and in Denmark (Baastrup et al., 2008). The case–control study from Taiwan showed a dose–response in bladder cancer risk, but the number of cases with low‐to‐moderate As concentrations was very low (Chiou et al., 2001). The case–control study from New Hampshire found about a two‐fold risk of bladder cancer in the highest exposure category, but with wide CIs, and the increased risk was found only among smokers (Karagas et al., 2004). In the EPIC cohort in Denmark, no positive association was observed (Baastrup et al., 2008), but as mentioned for skin cancer, exposure in this study was based on relatively crude aggregated estimates of water arsenic at low levels (median 0.7 μg/L), and with low contrast (only 1% above 5 μg/L).

TABLE 12

Key epidemiological studies on bladder cancer and As exposure.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Chiou et al. (2001)

North‐east Taiwan

Cohort study

Area endemic for arseniasis

Cases/person‐years of observation

3/7978

3/6694

2/3013

7/5220

1/7978

1/6694

2/3013

6/5220

As in well water (μg/L)

< 10.0

10.1–50.0

50.1–100.0

> 100.0

< 10.0

10.1–50.0

50.1–100.0

> 100.0

OR (95% CI)

Urinary tract

1.0

1.5 (0.3–8.0)

2.2 (0.4–13.7)

4.8 (1.2–19.4)

Transitional cell carcinoma

1.0

1.9 (0.1–32.5)

8.2 (0.7–99.1)

15.3 (1.7–139.2)

Adjusted for age, sex, smoking and duration of drinking well water

Duration of the study ~ 4 years

Karagas et al. (2004)

USA

Case–control study

Incident transitional cell carcinoma

Cases = 383, controls = 641

Never smoker

15/41

20/56

22/48

11/29

3/14

0/3

0/8

Ever smoker

75/121

99/105

66/109

37/67

18/18

3/10

14/11

Toenail As (μg/g)

0.009–0.059

0.060–0.086

0.087–0.126

0.127–0.193

0.194–0.277

0.278–0.330

0.331–2.484

0.009–0.059

0.060–0.086

0.087–0.126

0.127–0.193

0.194–0.277

0.278–0.330

0.331–2.484

OR (95% CI)

Never smoker

1.00

0.85 (0.38–1.91)

1.18 (0.53–2.66)

1.10 (0.42–2.90)

0.49 (0.12–2.05)

Ever smoker

1.00

1.53 (1.02–2.29)

1.02 (0.66–1.56)

1.00 (0.60–1.67)

1.78 (0.86–3.67)

0.50 (0.1–1.88)

2.17 (0.92–5.11)

ORs are age and sex adjusted

Maximum likelihood estimate change‐point of 0.326 μg/g (95% CI 0.121–0.446), which equates to approximately 50 μg/L, with a 1.1% increase in ORs for 1% increase in toenail

As concentration above change‐point (p = 0.10)

Baastrup et al. (2008)

Denmark

Cohort study

First bladder cancer

56,378

214 Bladder cancer cases

Time‐weighted average exposure (μg/L) median CI (95% CI)

0.7 (0.3–2.1)

Cumulative exposure

(5 mg)

IRR (95% CI)

1.01 (0.93–1.11)

1.0 (0.98–1.04)

Adjusted for smoking status, smoking duration, smoking intensity, education, occupation

Chen, Chiou, Hsu, Hsueh, Wu, Wang, and Chen (2010)Taiwan

Cohort study

Incident urothelial carcinoma

36/8086

3/2288

6/2093

3/907

7/909

10/691

As concentration in well water at enrolment (μg/L)

< 10

10–49.9

50–99.9

100–299.9

≥ 300

RR (95% CI) Urothelial carcinoma

1.00 (reference)

1.85 (0.45–7.61)

2.19 (0.43–11.1)

5.50 (1.39–21.8)

10.8 (2.60–40.3)

Relative to the As concentration < 10 μg/L, those who drank well water with higher (≥ 10 μg/L) concentrations from birth (RR, 4.39; 95% CI, 1.18–16.3), and still drank at enrolment (RR, 4.55; 95% CI, 1.34–15.5), had a significantly increased risk of urinary cancer. Adjusted for age, gender, education, whether started drinking well water from birth, cigarettes smoking status and alcohol consumption status at enrolment

Meliker et al. (2010)

USA

Case–control

Bladder cancer

411/566

188/264

185/263

38/39

Time‐weighted average water As (μg/L)

< 1

1–10

> 10

Continuous (per 5 μg/L increase)

OR (95% CI)

1.00

0.84 (0.63, 1.12)

1.10 (0.65, 1.86)

Ever smoker

0.95 (0.81, 1.11)

Never‐smoker

1.29 (1.03, 1.63)

Controls from the general population. Adjusted for cigarette smoking history, education, history of employment in high‐risk occupation, family history of bladder cancer, age, race and sex

Very similar OR for arsenic intake from water

Steinmaus et al. (2013)

Chile

Case–control

Incidence bladder cancer

232/640

33/202

33/189

71/142

95/107

Lifetime average water As (μg/L)

< 26

26–79

80–197

> 197

OR (95% CI)

1.0 (reference)

0.92 (0.52–1.61)

2.62 (1.53–4.50)

6.00 (3.38–10.64)

Hospital‐based controls. Adjusted for age, sex, smoking, mining work, race, BMI and SES

Wu et al. (2013)

Taiwan

Case–control study

Urothelial carcinoma

261/672

36/224

55/224

170/224

u‐tiAs (μg/L)

< 11.50

11–50‐20.40

≥ 20.40

OR (95% CI)

1.0

1.50 (0.95–2.39)

4.68 (3.06–7.14)

Controls from the hospital having a health examination

Adjusted for age, sex, educational level and alcohol consumption. Evaluation of arsenic‐smoking in interaction analyses

Mostafa and Cherry (2015)

Bangladesh

Case–control study

Urothelial carcinoma (formerly termed transitional cell carcinoma) (histologically confirmed)

1446/1078

238/206

319/190

204/145

278/244

251/143

156/150

Water As μg/L

< 10

10 < 50

50 < 100

100 < 200

200 < 300

300 or more

OR (95% CI)

1.0

1.52 (1.08–2.14)

1.07 (0.73–1.57)

0.99 (0.69–1.41)

1.63 (1.08–2.46)

0.89 (0.55–1.43)

Controls having benign lesions in the ureter, bladder or urethra

Adjusted for sex and smoking

Baris et al. (2016)

USA, New Hampshire

Case–control study

Newly diagnosed histologically confirmed carcinoma of the urinary bladder (including carcinoma in situ)

1213/1418

280/314

260/309

233/304

220/248

26/33

37/29

233/313

269/308

260/311

213/247

34/29

47/29

Average water As concentration, μg/L, average over 40 years

≤ 0.4

> 0.4–0.7

> 0.7–1.6

> 1.6–5.7

> 5.7–8.7

> 8.7

Cumulative As intake, mg, lagged 40 years

≤ 3.5

> 3.5–8.8

> 8.8–22.4

> 22.4–83.5

> 83.5–124.8

> 124.8

OR (95% CI) Bladder Cancer

1.0 (reference)

0.91 (0.71–1.17)

0.93 (0.72–1.20)

1.06 (0.81–1.40)

0.92 (0.51–1.66)

1.49 (0.85–2.61)

OR (95% CI) Bladder Cancer

1.00 (reference)

1.13 (0.87–1.47)

1.21 (0.92–1.58)

1.28 (0.95–1.72)

1.72 (0.96–3.10)

2.24 (1.29–3.89)

Control subjects selected randomly, and frequency matched to cases by state, sex and five‐year age group at diagnosis.

Adjusted for age, sex, Hispanic ethnicity, state of residence, smoking status, high‐risk occupation and exposure to disinfection by‐products

Adjusted for age, sex, Hispanic ethnicity, state of residence, smoking status, high‐risk occupation and exposure to disinfection by‐products

Huang et al. (2018)

Taiwan

Case–control study

Bladder cancer

216/813

72/204

64/203

46/203

34/203

u‐tiAs (μg/L)

≤ 9.78

9.78–17.91

17.91–30.28

> 30.28

OR (95% CI)

1.00 (reference)

1.94 (1.18–3.20)

2.09 (1.18–3.69)

3.52 (1.77–6.96)

Hospital‐based study. Non‐cancer controls were recruited from those receiving health examinations. Adjusted for age, gender, schools, father's educational level, cigarette smoking, alcohol, tea and coffee drinking, pesticide contact, urinary tract calculus, hypertension and diabetes history, and urinary creatinine

Abbreviations: As, arsenic; BMI, body mass index; CI, confidence interval; IRR, incidence rate ratio; n, number; OR, odds ratio; RR, relative risk; SES, socioeconomic status; USA, United States of America; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA).

There were seven new studies considered by the CONTAM Panel. In a cohort study in Taiwan (Chen, Chiou, Hsu, Hsueh, Wu, Wang, & Chen, 2010), an increasing risk of urothelial carcinoma, the main cancer form in the bladder, was found with increasing arsenic concentrations in well water. Two further case–control studies from Taiwan found increased risk of bladder cancer with increased u‐tiAs (Huang et al., 2018; Wu et al., 2013). In Wu et al. (2013) a significantly increased bladder cancer OR of 3.20 was also found for never smokers with a high u‐tiAs (> 15.40 mg/g creatinine). In the study by Huang et al. (2018) the validity might be influenced by the presence of high amounts of DMA in the urine. It should be noted that occupational exposure to bladder carcinogens may be a confounding factor and it was taken into account in most studies.

Two case–control studies with information about life‐time exposure to arsenic were performed in the US. In a population‐based case–control study in South‐eastern Michigan, with low levels of arsenic in drinking water (among cases the median concentration in private wells was 5.3 μg/L and in public supply 0.3 μg/L) no increased risk of bladder cancer was found (Meliker et al., 2010). The effect estimates were similar in never‐smokers and ever smokers. In a larger population‐based case–control study from Northern New England (average arsenic in drinking water between < 0.5 to more than 10 μg/L) (Baris et al., 2016). Koutros et al. (2018) found in the same study population as in Baris et al. (2016), that former smokers and current smokers with the highest cumulative arsenic intake had elevated risks of bladder cancer: OR = 1.4, 95% CI: 0.96–2.0 and OR = 1.6, 0.91–3.0, respectively), whereas the OR among never smokers was 1.1, 0.6–1.9, p‐interaction = 0.49). In a case–control study from Chile, an increased risk of bladder cancer was found in the group with lifetime average water arsenic 80–197 μg/L (Steinmaus et al., 2013). When comparing an average lifetime exposure to arsenic of 11–91 μg/L versus < 11 μg/L, smokers that smoked > 10 cigarettes per day showed increased risk (OR 4.72, 95% CI 1.54–14.49) whereas never smokers showed a weaker increased risk (OR 2.66, 95% CI 0.91–7.83) (Ferreccio, Yuan, et al., 2013).

In a study in Bangladesh no consistent trend was seen for urothelial carcinoma 17 with increasing arsenic concentration (Mostafa & Cherry, 2015). The authors suggested that this lack of association may be due to the fact that the controls were patients with benign lesions in the urinary tract. Arsenic exposure was positively associated with chronic cystitis, the major benign lesion in the controls, meaning that the control group carried a condition that may itself be caused by the exposure.

In summary, the epidemiological studies provide sufficient evidence for an association between low to moderate exposure to iAs and bladder cancer. The results for the key studies are summarised in Table 12.

Lung cancer

Based on studies from Taiwan, Japan, Chile, Argentina and the US, IARC concluded that there was sufficient evidence in humans that arsenic in drinking‐water causes cancer of the lung and assigned it to Group 1 (IARC, 2004). The 2009 Opinion (EFSA CONTAM Panel, 2009) focussed on epidemiological studies on lung cancer in humans informing on dose–response at exposure levels below 100 μg/L in drinking water. The CONTAM Panel noted that some of the studies published since the IARC report supported an excess risk of lung cancer whereas others did not.

The CONTAM Panel identified 19 recent research articles on arsenic exposure and risk for lung cancer. Ten out of these 19 did not meet the inclusion criteria (D'Ippoliti et al., 2015; Fan et al., 2016; Ferdosi et al., 2016; Gamboa‐Loira et al., 2017; Huang et al., 2013; Mannetje et al., 2011; Melak et al., 2014; Mendez Jr et al., 2017; Putila & Guo, 2011; Steinmaus et al., 2014a) and were therefore not considered further.

Nine studies that fulfilled the inclusion criteria were considered further (see Table 13) plus the one study that was used in 2009 for deriving the reference point (Ferreccio et al., 2000). This case–control study from Chile found evidence of an arsenic drinking water exposure‐related increase in lung cancer incidence (Ferreccio et al., 2000). Also, among cases that never smoked, there was an increased risk of lung cancer in the exposure group with 50–199 μg/L arsenic in drinking water (OR = 5.9, 95% CI 1.2–40.2) versus arsenic ≤ 49 μg/L. A re‐analysis of this study performed by Smith and colleagues, which also estimated u‐tiAs, found a similar trend in risk, associated with urinary arsenic and with water As levels (Smith et al., 2009). A more recent case–control study in Chile has shown increased lung cancer risk in relation to As exposure dating back to 40 years ago (Steinmaus et al., 2013; Steinmaus et al., 2014b), of which Steinmaus et al. (2014b) focused on exposure levels below 100 μg/L. This more recent case–control study is larger than the first one presented by Ferreccio et al. (2000) and Smith et al. (2009), and the choice of controls is superior. Results from this study in Chile (Steinmaus et al., 2014b) also indicates that the risk of lung cancer in adult age is affected by exposure during childhood or adolescence, and suggest that early‐life exposures carry a higher risk than exposures during adulthood. A cohort study from Taiwan reported an increased risk of lung cancer with drinking water concentrations of 100 μg/L arsenic, while no excess risk was found in the 10–99 μg/L As concentration range (Chen, Chiou, Hsu, Hsueh, Wu, & Chen, 2010). In never‐smokers, a tendency toward increasing risk was found with higher arsenic in drinking water (OR = 1.22, 95% CI 0.64–2.32 among cases with 10–99.9 μg/L, OR = 1.32, 0.64–2.74 among cases with ≥ 100 μg/L, compared with arsenic < 10 μg/L). In a follow‐up study, a higher risk of lung cancer was found among those individuals that had a low methylation capacity. However, the study was based on only 39 cases of lung cancer in total (Hsu, Tsui, et al., 2017).

TABLE 13

Key epidemiological studies on lung cancer and As exposure.

Reference study population designOutcome definitionPopulation size (n) cases/controlsArsenic exposureResultsAdditional information/confounders

Ferreccio et al. (2000)

Chile

Case–control

Incidence lung cancer

151/419

9/104

5/39

8/23

50/124

79/129

Average water As conc. (μg/L)

0–10

10–29

30–49

50–199

200–400

OR (95% CI)

1.0 (reference)

1.60 (0.50–5.30)

3.90 (1.20–12.30)

5.20 (2.30–11.70)

8.90 (4.00–19.60)

Hospital‐based controls. Adjusted for age, sex, cumulative lifetime cigarette smoking, years of work in copper smelting and socioeconomic status

Heck et al. (2009)

USA, New Hampshire and Vermont

Population‐based case–control

Primary incident lung cancer

223/238

69/65

66/58

44/58

44/57

75/238

17/65

24/58

13/58

21/57

Average toenail As conc. (μg/g)

< 0.05

0.05– < 0.0768

0.0768 – < 0.1137

≥ 0.114

Average toenail As conc. (μg/g)

< 0.05

0.05 – < 0.0768

0.0768 – < 0.1137

≥ 0.114

OR (95% CI)

Lung cancer

1.0

1.34 (0.71–2.53)

1.10 (0.55–2.20)

0.89 (0.46–1.75)

Small cell and squamous cell carcinomas

1.0

2.99 (1.12–7.99)

1.86 (0.62–5.58)

2.75 (1.00–7.57)

Adjusted for sex, age, race/ethnicity, educational attainment, BMI, fish servings per week, smoking (pack‐years) and selenium

Smith et al. (2009)

Chile

Case–control

Incidence lung cancer from national cancer registry

151/419

11/92

7/81

35/87

23/44

11/12

64/103

Average water As/urinary As exposure (μg/L)

< 10/4.9

10–59/34.0

60–199/126.1

200–399/291.0

400–699/533.5

700–999/824.5

OR (95% CI)

1.0 (reference)

0.70 (0.30–1.70)

3.40 (1.80–6.50)

4.70 (2.00–11.00)

5.70 (1.90–16.90)

7.1 (3.4–14.8)

Re‐analysis of the Ferreccio et al. (2000) study and the addition of urine derived from water levels (Smith et al., 2009). Adjusted for age, sex, cumulative lifetime cigarette smoking, years of work in copper smelting and socioeconomic status

Chen, Chiou, Hsu, Hsueh, Wu, and Chen (2010)

Taiwan, arseniasis‐endemic area

Cohort study

Incidence primary lung cancer from national cancer registry

N = 8086, 6888 with Water As, 178 cases

48 cases

51 cases

20 cases

28 cases

31 cases

Water As (μg/L)

< 10

10–49.9

50–99.9

100–299.9

> 300 μg/L

RR (95% CI)

1.0 (reference)

1.10 (0.74–1.63)

0.99 (0.59–1.68)

1.54 (0.97–2.46)

2.25 (1.43–3.55)

Adjusted for age, sex, education, smoking status, alcohol consumption. Significant trends for squamous cell and small cell carcinomas but not for adenocarcinoma. No interaction with smoking

Dauphiné et al. (2013)

Nevada and Kings County, California

Case–control

Incidence primary lung cancer, histological confirmation

196/359

141/241

37/82

18/36

Water As: Highest 5‐year average: 10‐years lag

≤ 10

11–84

≥ 85

OR (95% CI)

1.0 (reference)

0.75 (0.45–1.25)

0.84 (0.41–1.72)

Randoms controls from the area, frequency‐matched by age group. Adjusted for age, sex, education, smoking history and possible exposure to another known lung carcinogen. Also, no association with 40‐year lag or cumulative exposure

García‐Esquinas et al. (2013)

USA, Arizona, Oklahoma and North/South Dakota

Cohort study

Mortality lung cancer from the health register of the state

N = 3932

78 cases /3857 referents

27/1292

20/1296

31/1269

u‐tiAs (μg/g creatinine)

< 6.91

6.91–13.32

> 13.32

HR (95% CI)

1.0 (reference)

0.94 (0.51–1.72)

1.82 (1.00–3.31)

Adjusted for sex, age, education, smoking status, drinking status and BMI

Steinmaus et al. (2013)

Chile

Case–control

Incidence lung cancer

306/640

61/202

61/189

85/142

99/107

Lifetime average water As (μg/L)

< 26

26–79

80–197

> 197

OR (95% CI)

1.0 (reference)

0.98 (0.62–1.53)

1.70 (1.05–2.75)

3.18 (1.90–5.30)

Hospital‐based. Adjusted for age, sex, smoking, mining work, race, BMI and SES

Steinmaus et al. (2014b)

Chile

Case–control

Primary lung cancer

92/288

23/103

32/98

37/87

25/102

31/99

36/87

27/103

29/95

36/90

Mean water As 40 or more years ago (highest single year) (μg/L)

6.5

23.0

58.6

Mean water As 40 or more years ago (highest 5‐year average) (μg/L)

< 10.0

10.0–54.0

> 54.0

Mean water As 40 or more years ago (lifetime average) (μg/L)

< 6.0

6.0–22.1

> 22.1

OR (90% CI)

1.00,

1.43 (0.82, 2.52)

2.01 (1.14, 3.52)

1.00

1.27 (0.73, 2.20)

1.78 (1.02, 3.11)

1.00

1.14 (0.67, 1.95)

1.56 (0.91, 2.67)

Controls without lung, bladder or kidney cancer were randomly selected from the Chilean Electoral Registry and were frequency‐matched by sex and 5‐year age group. Adjusted odds ratios by tertile of the highest known exposure 40 or more years ago. Adjusted for age, sex and smoking behaviour

Argos et al. (2014)

Bangladesh

Cohort

Lung cancer mortality

90 lung cancer deaths/26,043

With baseline water As

12

11

19

Water As (μg/L)

< 14

14.0–82.9

≥ 83.0

HR (95% CI)

1.0

0.85 (0.38–1.93)

1.15 (0.55–2.38)

Adjusted for sex, age, body mass index, education, smoking status and study cohort. Water and urinary As concentrations correlated by 0.7

Hsu, Tsui, et al. (2017)

Taiwan, arseniasis‐endemic area

Cohort

Incident lung cancer39/1300 (numbers in each exposure category not available)

As in water (μg/L)

Each 50 increment

Water As (μg/L)

2–10

10.01–100

100.01–200

> 200

Water As (μg/L)

2–10

10.01–100

100.01–200

> 200

HR (95% CI)

1.03 (1.00–1.07)

Low methylation capacity: Incidence densities/year0.000799

0.002277

0.002646,

0.004674

High methylation capacity: Incidence densities/year

0

0.000410

0

0.001585

Study follow‐up of Chen, Chiou, Hsu, Hsueh, Wu, & Chen (2010) but unclear how the studies are linked.

Adjusted for age, sex, education, occupation, marriage, cigarette smoking and alcohol drinking

Low methylation capacity group defined as those whose PMI or SMI was lower than their respective median values

Abbreviations: As, arsenic; BMI, body mass index; CI, confidence interval; conc., concentration; HR, hazard ratio; n, number; OR, odds ratio; PMI, primary methylation index; RR, relative risk; SES, socioeconomic status; SMI, secondary methylation index, USA, United States of America; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites DMA and MMA).

In a cohort study from Bangladesh with arsenic‐contaminated water, lung cancer mortality was not associated with either concentrations of arsenic in drinking water or urinary As levels (Argos et al., 2014).

A population‐based study from the U.S. found evidence of an increased risk of small cell and squamous cell carcinomas of the lung in the highest tertile of toenail As, but no overall association with lung cancers or interaction effect between arsenic exposure and cigarette smoking in relation to lung cancer risk (Heck et al., 2009). No increased lung cancer risk was found in a case–control study from the U.S. (Dauphiné et al., 2013). However, in a large U.S. cohort study, an association with lung cancer mortality was found in the highest tertile of u‐tiAs (García‐Esquinas et al., 2013).

In summary, the epidemiological studies provide sufficient evidence for an association between low to moderate exposure to iAs and lung cancer.

The results for the key studies are summarised in Table 13.

Breast cancer

Garland et al. (1996) explored the relationship between As, measured in toenails and breast cancer in a nested case–control study of 892 individuals within the Nurses' Health Study cohort. After 5 years of follow‐up, the authors reported no association between arsenic concentrations and breast cancer risk (OR Q5 vs. Q1 = 1.12, 95% CI 0.66–1.91, p‐trend = 0.78). One study used a food frequency questionnaire to quantify As exposure from diet alone and found no association between level of As exposure and breast cancer risk in a Japanese population (Hazard Ratio [HR] = 1.06, 95% CI 0.8–1.41) (Sawada et al., 2013).

In a case–control study from a region in Mexico with elevated As in drinking water, lower total arsenic concentrations in urine were found in women with breast cancer compared with controls (cases median 14.33 μg/L, 90% 4.58–52.24; vs. controls 19.96 μg/L, 90% 6.40–100.04, p < 0.001) (López‐Carrillo et al., 2014). However, a higher risk for breast cancer was found with lower arsenic methylation capacity. Lower total iAs in urine among patients with breast cancer compared with controls, as well as higher risk of breast cancer among women with lower methylation capacity were reported in another case–control study from the same area (López‐Carrillo et al., 2020). The authors hypothesise that the lower concentrations of arsenic in urine among cases is due to less efficient metabolism of arsenic than among the controls. However, it is unclear why this phenomenon would occur for patients with arsenic‐related breast cancer and not for other types of iAs‐related cancer. In a study of young onset breast cancer in discordant sister pairs, there was no association between low‐level arsenic concentrations in toenail clippings (cases sisters' median arsenic 0.049 μg/g, IQR 0.037–0.070; and control sisters 0.048 μg/g IQR 0.034–0.069) and breast cancer risk (O'Brien et al., 2019). In a U.S. cohort study, no increased risk for breast cancer mortality was found with increasing As concentrations in urine (García‐Esquinas et al., 2013).

In further original research articles, arsenic exposure and breast cancer were evaluated, but the studies were not considered further, either because the exposure was based on rice intake as a proxy for arsenic intake (Zhang et al., 2016), or arsenic concentrations in whole blood or in air (Liu et al., 2015; Marciniak et al., 20202021).

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and breast cancer.

Prostate cancer

The 2009 IARC assessment considered the evidence that arsenic causes cancer of the prostate ‘limited’ (Straif et al., 2009).

Earlier ecological studies have found excess of prostate cancer in relation to iAs exposure (Hinwood et al., 1999; Lewis et al., 1999). A report from the southwest of Taiwan, an area with high endemic As concentrations, found a decline in SMRs for prostate cancer since the introduction of tap water with lower As concentrations, suggesting the association may be causal (Yang et al., 2008). As a follow‐up to this study, Hsueh et al. (2017) found in a case–control study from a non‐endemic area in Taiwan that participants with u‐tiAs a concentration > 29.28 μg/L had a significantly higher risk (OR 1.75, 1.06–2.89) of prostate cancer than participants with less than 29.28 μg/L. In a U.S. cohort study, arsenic exposure (median u‐tiAs 9.7, IQR 5.8–15.6, μg/g creatinine), was prospectively associated with prostate cancer mortality (adjusted HRs comparing the 80th vs. 20th percentiles of arsenic 3.30, 95% CI 1.28–8.48; García‐Esquinas et al., 2013). In a case–control study in Cordoba, Argentina, a synergistic effect of having As in drinking water above 10 μg/L and being a rural worker was found for prostate cancer risk. However, no effect estimates were presented for arsenic only (Román et al., 2018).

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and prostate cancer.

Kidney cancer

The 2009 IARC assessment considered the evidence ‘limited’ that iAs causes cancer of the kidney (Straif et al., 2009).

No association between As exposure and kidney cancer was found in a study from Australia (Hinwood et al., 1999), or a study from Utah (Lewis et al., 1999). In an ecological study on two regions in Chile with different concentrations in arsenic in drinking water, a higher mortality from kidney cancer was found in the region with high arsenic drinking water concentrations (Yuan et al., 2010), and with increased risks manifesting 40 years after exposure reduction (Smith et al., 2018). In a case–control study in Chile based on cases (renal cell carcinoma and transitional cell renal pelvis and ureter, and other kidney cancers) recruited 2007–2010, with individual data on exposure and potential confounders, an increased risk for renal pelvis and ureter cancers was found at high exposure level (Ferreccio, Smith, et al., 2013): the adjusted odds ratios by average arsenic intakes of < 400, 400–1000 and > 1000 μg/day (median water concentrations of 60, 300 and 860 μg/L) were 1.00, 5.71 (95% confidence interval: 1.65, 19.82) and 11.09 (3.60, 34.16) (p < 0.001), respectively. Odds ratios were not elevated for renal cell cancer. However, in an ecological study of cases and referents (benign cases) from Bangladesh with low‐level exposure, the risk for renal cell carcinoma increased monotonically with arsenic concentration > 50 μg/L for renal cell carcinoma (Mostafa & Cherry, 2013). Stratification by ‘ever smoked’ confirmed the presence of risk in non‐smokers. In areas with hand‐pumped wells installed more than 15 years ago, the excess risks were observed below 50 μg/L arsenic in drinking water. In a U.S. cohort study, no association was found between increasing u‐tiAs concentrations and kidney cancer mortality (García‐Esquinas et al., 2013). However, an ecological study from the U.S. showed positive association between the highest quartile of exposure (aggregated cumulative county‐level arsenic concentration > 12.89 ppb‐year), compared to the lowest (< 3.83 ppb‐year) for kidney cancer weighted for proportion of population served by community water system adjusted risk ratio: 1.69 (1.37, 2.09) (Krajewski et al., 2021).

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and kidney cancer.

Other cancers

The 2009 IARC assessment considered the evidence ‘limited’ for cancers of the liver and prostate (Straif et al., 2009).

There were few studies on low‐to‐moderate As exposure and risk of cancers of the pancreas, liver and gallbladder and there were few with individual arsenic exposure data taking organic arsenic species into account. Here ecological studies are described where follow‐up case–control studies have been performed.

Arsenic has been linked to liver and pancreatic cancers from ecological observations. Neonatal exposure to arsenic from contaminated milk was associated with increases in liver and pancreatic cancer (Yorifuji et al., 2011). An ecological study from Florida also found that pancreatic cancer patients living within 1 mile of known arsenic‐contaminated wells were significantly more likely to be diagnosed within a cluster of pancreatic cancers relative to cases living more than 3 miles from known sites (OR = 2.1, 95% CI = 1.9, 2.4) (Liu‐Mares et al., 2013). In a Spanish case–control study, where arsenic in toenails was measured before the cancer treatment, an increased risk of pancreatic cancer was found (OR = 2.02, 95% CI 1.08–3.78; p = 0.009) when comparing the highest quartile (> 0.1061 μg/g) versus the lowest quartile (< 0.0518) of concentrations of arsenic on toenails (Amaral et al., 2012). Additional adjustment for the elements found significant in the basic model attenuated the OR estimate, and association with arsenic was no longer statistically significant (highest vs. lowest quartile: OR = 1.72, 95% CI 0.77–3.86; p = 0.201). In a U.S. cohort study, arsenic exposure (median urine iAs metabolite concentrations 9.7, IQR 5.8–15.6, μg/g creatinine), was prospectively associated with pancreatic cancer mortality (adjusted HRs comparing the 80th vs. 20th percentiles of arsenic 2.46, 95% CI 1.09–5.58; García‐Esquinas et al., 2013).

For liver, Wadhwa et al. (2011) reported increased hair and blood levels of arsenic among liver cancer cases from an exposed area in Pakistan, but no risk estimates or dose–response were reported. Further, in a U.S. cohort study, no association was found between increasing total sum of urinary iAs metabolite concentrations and liver cancer mortality (García‐Esquinas et al., 2013).

Head and neck cancers have been linked to arsenic exposure. Still, the data published suffer from several drawbacks which makes it difficult to draw firm conclusions. Either the studies were confounded by high frequency of smokers and alcohol drinkers in the case group, and no analysis was performed among non‐smokers or non‐drinkers (Pal et al., 2017; Pal & Halder, 2019), or only total arsenic was measured in blood or in serum precluding conclusions regarding exposure to iAs (Chen, Qiu, et al., 2021; Khlifi et al., 2014). However, one case–control study on arsenic in non‐smoking tobacco included only non‐smoking and non‐alcohol drinking study participants (Arain et al., 2015). The cases and controls differed in arsenic concentrations in hair and blood, but no risk estimates were provided.

In a large study, 259 patients with gallbladder cancer, 701 patients with gallstones and 851 population‐based controls from China were analysed for total serum arsenic, however, with no speciation or adjustment of seafood intake (Lee et al., 2020). A reduced risk of gallbladder cancer was found with increasing As. For stomach and ovarian cancers, only ecological studies were found (Amin, Ross, et al., 2019; Chen et al., 2015; Núñez et al., 2016). No other cancers have been found to be consistently related to drinking water As exposure.

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and liver, pancreatic and gallbladder cancer.

3.2.2.3. Skin lesions

The potential of iAs to induce skin lesions such as hypo‐ or hyperpigmentation and hyperkeratosis has been well known since the 1960s (Tseng et al., 1968). The 2009 Opinion concluded that As concentrations in drinking water or urine below 100 μg/L were associated with increased incidences of skin lesions (EFSA CONTAM Panel, 2009).

For the present Opinion, the CONTAM Panel identified 18 studies published since 2009 from a literature search (see Section 3.2.2.1). Out of these, three studies met the inclusion criteria which were considered further together with the studies in Bangladesh (Ahsan et al., 2006) and Mongolia (Xia et al., 2009) that were used to calculate reference points in the 2009 Opinion (see Table 14). Fifteen studies did not meet the inclusion criteria (Argos et al., 2015; Das et al., 2016; Guha Mazumder et al., 200920102014; Guo et al., 2006; Hashim et al., 2013; Kazi et al., 2009; Li et al., 2011; Liao et al., 2010; Sy et al., 2017; Wei et al., 2018; Wen et al., 2012; Yajima et al., 2018; Zhang et al., 2014).

TABLE 14

Key epidemiological studies on skin lesions and As exposure.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposure concentration in water (μg/L)Results odds/risk ratio (95% CI)Additional information/confounders

Ahsan et al. (2006)

Bangladesh Cross‐sectional

Dermal lesions

Cases/total

57/2259

90/2122

144/2202

162/2185

242/2183

0.1–8.0

8.1–40.0

40.1–91

91.1–175

175.1–864.0

POR (prevalence odds ratio)

1.00 (reference)

1.91 (1.26, 2.89)

3.03 (2.05, 4.50)

3.71 (2.53, 5.44)

5.39 (3.69, 7.86)

Adjusted for age, BMI, education, cigarette smoking, hukka (waterpipe) smoking, sun exposure in males and land ownership

Xia et al. (2009)

Inner Mongolia, China Cross‐sectional

Dermal lesions

3215

845

1277

3429

1537

1021

92

0–5

5.1–10

10.1–20

20.1–50

50.1–100

100.1–300

> 300

OR

1.0

2.52 (1.47–4.30)

2.83 (1.773–4.525)

3.94 (2.78–5.59)

6.03 (4.05–8.97)

8.83 (5.77–13.51)

7.94 (2.73–23.12)

At the time of interview, they collected water samples at the household's primary source of water

Adjusted for drinking, smoking, education, sex, farm work, income, well type and age

Pierce et al. (2011)

Bangladesh Longitudinal (cohort) study

Dermal lesions

Cases/total

131/2358

125/2118

140/1726

318/2855

100/617

0.1–10

10.1–50

50.1–100

100.1–200

≥ 200.1

HR

1.00

1.17 [0.92, 1.50]

1.70 [1.34, 2.16]

2.22 [1.81, 2.73]

3.76 [2.85, 4.95]

Follow‐up of Ahsan et al. (2006). Age‐ and sex‐adjusted effect estimates. Skin lesion incidence was defined as a new occurrence of skin lesions of any type (as determined at one of the follow‐up interviews) among individuals who previously had no manifestation of any lesion (as determined at baseline and previous follow‐up interviews)

Fatmi et al. (2013)

Pakistan

Nested case‐referent study

Dermal lesions

72/462

44

315

78

97

50–99

100–299

300–399

> 400

Prevalence (%)

4.5 (2.74–6.26)

14.8 (10.88–18.72)

11.7 (13.85–20.23)

12.8 (9.24–14.76)

Nested study was based on household with water arsenic concentrations > 50 μg/L. No adjustments but stratified analysis was performed for age, sex and smoking

Yang et al. (2017)

Inner Mongolia Cross‐sectional

Dermal lesions168/380

< 10

10–100

100–200

200–300

300–400

> 400

Prevalence (%)

F M

19.2 19.4

22.6 25

24.6 37.5

38.8 21.9

47.5 52.9

38.5 61.9

No information about number of case/controls in each arsenic in water concentration group. No adjustments

Abbreviations: As, arsenic; BMI, body mass index; CI, confidence interval; f, female; HR, hazard ratio; m, male; n, number; OR, odds ratio; POR, prevalence odds ratio.

The study by Ahsan et al. (2006) (statistically significant increases in men) and Xia et al. (2009) (statistically significant increases in men and women combined) showed increased risks below < 10 μg/L of As in drinking water. Furthermore, in the study from Inner Mongolia, China, 5% (632) of the 12,334 residents surveyed had skin lesions characteristics of arsenic exposure (Xia et al., 2009).

In a study in Bangladesh from Pierce et al. (2011) increased risks (men and women combined) were found at 50–100 μg/L. The study by Pierce et al. (2011) is a follow‐up study of Ahsan et al. (2006) and is in addition focusing on the role of the diet.

In a new study from Inner Mongolia increasing prevalence of skin lesions were found, particularly in men, in the concentration range 10–100 μg/L (Yang et al., 2017). However, no statistical analysis was performed.

In a study from Pakistan (Fatmi et al., 2013), cases with skin lesions were identified among households with water source concentrations above 50 μg/L. The prevalence of cases with skin lesions was 4.5% in the group with water concentrations 50–99 μg/L, and 14.8 in the group with water concentrations 100–299 μg/L.

Several studies have evaluated modifying factors for the association between arsenic exposure and skin lesions. Higher risks of arsenic‐related skin lesions are for males (Rahman et al., 2006), having low body weight (Guha Mazumder et al., 1998), low levels of amino acids (Wei et al., 2019), impaired one‐carbon metabolism (Pilsner et al., 2009), being anaemic (Kile, Faraj, et al., 2016) and poor arsenic metabolism efficiency (Kile et al., 2011; Yang et al., 2017).

In a large genome‐wide association study in Bangladesh, Pierce et al. (2012) identified the arsenic methylating gene AS3MT to be associated with arsenic metabolism and risk of arsenic skin lesions.

The key studies on skin lesions are presented in Table 14.

In summary, the epidemiological studies provide sufficient evidence that low to moderate arsenic concentrations in drinking water are associated with an increased risk of skin lesions.

Nevertheless, since the major studies are performed in low and medium income regions, and nutrition and health status are modifying factors, it is difficult to translate the risks to populations with more adequate nutrition such as in Europe. The CONTAM Panel is not aware of studies with Western populations showing an association of skin lesions with arsenic exposure.

3.2.2.4. Developmental toxicity

The previous EFSA assessment on arsenic (EFSA CONTAM Panel, 2009) considered the following developmental outcomes in humans: birth weight (BW), preterm birth (PTB), fetal loss/infant death, birth defects and cognitive performance.

Many additional studies on arsenic exposure and developmental effects in humans has been published since the previous EFSA Opinion. In addition to the outcomes previously considered, the CONTAM Panel also reviewed associations between prenatal exposure to iAs and additional outcomes related to fetal growth, namely GA, fetal growth and anthropometric outcomes in childhood.

Birth weight

In the previous EFSA Opinion (EFSA CONTAM Panel, 2009) it was noted that ‘there is emerging evidence of negative impacts on fetal and infant development, particularly reduced birth weight, and there is a need for further evidence regarding the dose‐response relationships and critical exposure times for these outcomes’. Regarding birth weight, only five studies were available at that time (Hopenhayn et al., 2003; Huyck et al., 2007; Myers et al., 2010; Rahman et al., 2009; Yang et al., 2003 [see Table 15]). One of these studies was very small (Huyck et al., 2007), and for two studies exposure assessment was based on aggregated water arsenic.

TABLE 15

Key epidemiological studies relating As exposure with birth weight.

Reference study population designOutcome definitionPopulation size (n)Arsenic exposure a ResultsAdditional information/confounders
Included in 2009 EFSA Opinion

Hopenhayn et al. (2003)

Chile

Cohort

BW844 (442 from Antofagasta and 444 from Valparaiso)Water: Antofagasta: 30–40 μg/L Valparaiso: < 1 μg/L−57 g in Antofagasta (95% CI ‐123, +9)

Aggregated w‐As data

Adjusted for many potential confounders, including GA

Yang et al. (2003)

Taiwan

Cohort

BW18,259 (3872 from As‐exposed townships, and 14,387 from similar reference townships)Water: In As‐exposed townships 30% > 50 μg/L−29 g (95% CI –44, –14) in As‐exposed townshipsAggregated w‐As data. Adjusted for some potential confounders

Huyck et al. (2007)

Rural Dacca, Bangladesh

CS/cohort (samplings early pregnancy and around birth)

BW, LBW49 live births

Water: max Q1: 0.5, Q2: 1.3, Q3: 9.0, Q4: 734 μg/L. Maternal hair 0.14–3.3, toenail 0.2–6.2

(both t‐As μg/g)

−194 g per μg/g As in hair

No association between BW and water‐As or nail‐As

OR for LBW versus hair As around 2 but CI included 1

LBW = < 2750 (=median in these newborns). Poor correlations between w‐As and most biomarkers

Adjusted for a number of potential confounders including GA

Rahman et al. (2009)

Matlab Bangladesh

Cohort

BW1578 singleton live birthsMaternal u‐tiAs: median 95 μg/L

No overall association, but

−1.7 g per μg/L in u‐tiAs at 0–100 μg/L. Mean BW 2.6 kg

Adjusted for many potential confounders. Exposure from water (70% > 10 μg/L, see Rahman et al., 2006)

Myers et al. (2010)

Inner Mongolia, China

CS

BW in term births9890 singleton births 1996–1999 with As measured in the wells of the sub villageWater: 66% < 20, 20% 21–50, 7% 50–100, 7% > 100 μg/L. Data from 1991 to 1997 in well‐water registryNo association between BW and water‐As categoriesw‐As data were averaged by sub village. No potential confounders were detected for BW. Women were generally health and well‐nourished
Not included in the 2009 Opinion

Kwok et al. (2006)

Rural Bangladesh, 261 villages in 3 districts

CS

LBW in term (≥ 37 week) births2006 live singleton births with As measured in the wells of the villagesWater: 2003 samples, mean about 70 μg/L, six categories < 10 μg/L to > 300 μg/L

Adjusted OR 1.00 for LBW (< 2500 g) in full term births

BW not analysed as continuous variable

Water samples (for each household), birth data for 2002, and covariates were collected in 2003 from Community Centres' registers + maternal interviews (2003). Most births at home

Gelmann et al. (2013)

Romania

CS

LBW38 singleton live births with LBW and normal BW from As‐in‐water exposed (19) and unexposed (19) villagesWater: means in exposed 54 μg/L and in unexposed 1 μg/L. u‐iAs 2–90 μg/L in exp and 0–1 μg/L in unexposedAmong exposed those with LBW had higher u‐tiAs than those with normal BWPilot study with too few cases

Chou et al. (2014)

Taiwan

Cohort

BW, LBW299 live births, 21 (7%) LBW (< 2500 g)Maternal u‐tiAs. Median 22 μg/g Cr (90% DMA)Significant adjusted association (multiple linear regression) between u‐MMA and BW. No association with LBWThe association btw u‐MMA and BW disappeared after further adjustment for DNA damage (N7‐MeG)

Laine et al. (2015)

Mexico

CS

BW199 singleton live births, only 4 with LBWWater: < 0.5–236, median 13 μg/L. Maternal u‐tiAs median 23 μg/L

w‐As, u‐tiAs, u‐DMA: null

U‐MMA and U‐%MMA had a significant adjusted negative slope

Mean u‐DMA 43 μg/L in women reporting seafood and 31 μg/L in the others. The association with BW seemed to be at least partly caused by an association with GA

Bloom et al. (2016)

Romania

CS

BW122 live singleton birthsWater: mean 4.1 μg/LNo overall association between As and BWAdjusted for potential confounders. An inverse association was found in the 29 smoking mothers

Gilbert‐Diamond et al. (2016)

USA, New Hampshire

Cohort

BW706 live singleton birthsMaternal u‐tiAs. Median 3.4 μg/LNo association between u‐tiAs, MMA or DMA and BWGeneral additive models adjusted for some potential confounders including gestational age

Kile, Cardenas, et al. (2016)

Rural Bangladesh

Cohort

BW1153 live singleton births

Water: < 0.5–1400, median 2.3 μg/L

Maternal toenail t‐As 0.2–35 μg/g, median 1.5 μg/g

Significant adjusted inverse association (multiple linear regression) between water As and BW, and between nail‐As and BWStructural equation modelling suggested that almost all the association was mediated by an association with GA

Almberg et al. (2017)

USA, Ohio

CS

LBW and VLBW (< 1500 g) in term infants428,804 live singleton births from records 2006–2008. In term births 2.9% LBW and 1.2% VLBWW‐As aggregated by 88 counties and year: 0.5–12 μg/L, median 1.5 μg/L. Based on 2968 measurements in 975 water systemsAdjusted ORs for LBW (1.00) and VLBW (0.99) were normal

ORs adjusted for many potential confounders from the records

ORs were increased in some sub analyses excluding counties with many private wells

Rahman et al. (2017)

Rural Bangladesh

Cohort

BW1180 live singleton births, same study base as Kile, Cardenas, et al. (2016)See Kile, Cardenas, et al. (2016), but median w‐As here 2.2 μg/L and nail‐As 1.2 μg/gAssociations: See Kile, Cardenas, et al. (2016)Mediation analyses suggested that most of the association was mediated by GA, but among the smallest infants there was also a direct effect

Lin et al. (2019)

Rural Bangladesh

Cohort

BW1057 live singleton births, Same study bas as Kile, Cardenas, et al. (2016) and Rahman et al. (2017)Toenail As 0.04–47, median 1.2BW inversely associated with protein, fat and fibre intake, but positively associated with carbohydrate intake. Nail‐As not significantly associated with BW, when adjusted for macronutrients, and nail‐As was not a significant mediator for BWSeems to weaken the conclusions from Kile, Cardenas, et al. (2016) regarding BW versus nail‐As, where macronutrients were not adjusted for. But still association between nail‐As and GA; the latter was a mediator to BW in Kile, Cardenas, et al. (2016)

Bozack et al. (2020)

Rural Bangladesh

Cohort

BWSubset of Kile, Cardenas, et al. (2016): 413 births with % methylation of DNA cord blood at 3 CpG sites in the gene body of DNA methyltransferase 3 alphaSee Kile, Cardenas, et al. (2016)Significant inverse association between nail‐As and BW.See Kile, Cardenas, et al. (2016), Rahman et al. (2017) and Lin et al. (2019). Here structural equation modelling also includes DNA methylation in cord blood.

Howe, Farzan, et al. (2020)

USA, Los Angeles

Cohort

BW

Births with maternal:

116 (hair‐As)

100 (blood As)

167 (u‐iAs)

Medians:

Hair: 0.01 μg/g

Blood: 0.67 μg/g

u‐tiAs: 5.7 μg/L

No association with u‐tiAsIn urine additionally 0.5 μg/L AsB
Howe, Claus Henn, et al. (2020)BW262Median u‐tiAs 5.8 μg/LNo significant association with BW in relation to GANote that then a possible impact of As on BW via GA is not taken into account

Shih et al. (2020)

USA, seven areas

Cohort (NCS)

BW212Total As (median 7.8 and DMA median 3.4 μg/L) in maternal urineNo association btw u‐DMA and BWHigh LODs: u‐tiAs 1.25, DMA 1.7, MMA 0.9. All had u‐tiAs >LOD and 77% had DMA > LOD, but for other species > 60% < LOD

Bulka et al. (2022)

USA nationwide

Cross‐sectional

BW3,305,090 term births in 3105 countiesThe probability of As in water in four categories (≤ 1, > 1, ≤ 5, > 5, ≤ 10, > 10) was estimated in 1 × 1 km grids, based on about 20,000 As measurements in private wells. Estimates were then aggregated on county level

Adjusted beta (grams) per 10% increase in probability of exceeding the following thresholds:

≤ 1, > 1

−3.1 (−4.0, −2.2)

≤ 5, > 5

−2.8 (−4.2, −1.3)

≤ 10, > 10

−3.9 (−6.1, −1.7)

Adjusted for proportion of private wells, maternal age, race/ethnicity, marital status, education, smoking during pregnancy, pre‐pregnancy BMI, rurality/urbanicity, annual concentration of PM < 2.5 μm in the county

Abbreviations: As, arsenic; AsB, arsenobetaine; BMI, body mass index; BW, birth weight; CI, confidence interval; Cr, creatinine; CpG, cytosine‐phosphate‐guanine; CS, cross‐sectional study; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; DNA, deoxyribonucleic acid; EFSA, European Food Safety Authority; GA, gestational age; iAs, inorganic arsenic; km, kilometre(s); LBW, low birth weight; LOD, limit of detection; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; n, number; N7‐MeG, N7‐methylguanine; NCS, National Children's Study; OR, odds ratio; PM, particulate matter; t‐As, total arsenic; u‐DMA, urinary DMA; u‐iAs, urinary inorganic arsenic; u‐MMA, urinary MMA; USA, United States of America; u‐tiAs, total urinary iAs (sum of iAs and methylated its metabolites MMA and DMA); VLBW, very low birth weight; w‐As, water arsenic.

a iAs in urine includes MMA and DMA, but not AsB.

The CONTAM Panel identified 40 additional papers that were not included in the EFSA 2009 Opinion. In 25 of these papers, As exposure and birth weight were reported, but the studies were not considered further either because exposure was assessed by total As in blood, serum or urine and it was not possible to evaluate the contributions of organic vs. inorganic As (Bermúdez et al., 2015; Bloom et al., 2015; Deyssenroth et al., 2018; Govarts et al., 2016; Guan et al., 2012; Hameed et al., 2019; Henn et al., 2016; Lee et al., 2021; Liao et al., 2018; Liu et al., 2018; Mullin et al., 2019; Nyanza et al., 2020, Rahman, Oken, et al., 2021; Remy et al., 2014; Röllin et al., 2017; Wang, Li, Zhang, et al., 2018; Wang, Wang, Wang, et al., 2022; Xu et al., 2011; Xu, Hansen, et al., 2022), and/or the papers did not explicitly report associations between As exposure and birth weight (Fei et al., 2013; Mullin et al., 2019; Muse et al., 2020; Remy et al., 2014; Saha et al., 2012; Thomas et al., 2015; Wai et al., 2020; Wei, Shi, et al., 2017).

In 15 of the studies not previously evaluated exposure assessment was based on water arsenic, urinary inorganic arsenic (iAs species, MMA, or DMA or u‐tiAs), toenail As or hair‐As, and results on associations with birth weight (BW) were reported. These 15 studies, and the five reviewed in the 2009 Opinion are summarised in Table 15.

Three of the five studies reviewed in the 2009 Opinion indicated that infants born to women who drink water with elevated arsenic concentrations during pregnancy have a lower birthweight (Hopenhayn et al., 2003; Rahman et al., 2009; Yang et al., 2003). However, the longitudinal study, carried out in Bangladesh, showed a significant negative association between birthweight or head and chest circumferences and urinary As in the low concentration range (< 100 μg/L in urine), but no further negative effects were shown above about 100 μg/L (Rahman et al., 2009). A study performed in Mongolia did not show adverse birth outcomes (Myers et al., 2010).

In a publication not reported in the 2009 Opinion, Kwok et al. (2006) studied low birth weight (LBW) in 2006 live births in 261 rural Bangladesh villages. No significant association was found between water As and the risk of LBW.

Gelmann et al. (2013) reported ‘a pilot study’ with 19 live births selected from Romanian villages with elevated As in drinking water and 19 from villages with low As in water. Among 19 infants from the As‐exposed villages, u‐tiAs was significantly higher in infants with LBW compared to infants with normal BW.

Chou et al. (2014) studied associations between maternal u‐tiAs and BW in 299 infants in Taiwan and found a significant association between u‐MMA (but not the other species) and BW.

Laine et al. (2015) found no significant association between water As, u‐tiAs, or u‐DMA and BW in 199 Mexican infants. For u‐MMA there was a significant inverse association. It was attenuated when GA (which was inversely associated with u‐MMA) was added to the model.

Bloom et al. (2016) found no overall significant association between maternal As in drinking water (mean 4.1 μg/L) and BW in 122 infants in Romania.

Gilbert‐Diamond et al. (2016) found no significant associations between maternal u‐tiAs, MMA or DMA and BW in infants in New Hampshire, US. However, the median concentration of As in water was only 0.5 μg/L (P75 2.7 μg/L) Therefore, in contrast to the above‐mentioned studies these women had a low As intake from drinking water, and other dietary sources would be important. Results were unchanged if women with recent seafood or rice consumption were excluded.

Kile, Cardenas, et al. (2016) found a statistically significant inverse association between household water As and BW, and between maternal nail‐As and BW in 1153 infants in rural Bangladesh. Structural equation modelling suggested that almost all the association was mediated by GA (which was inversely associated with water As and toenail‐As).

Rahman et al. (2017) used the same study base as in Kile, Cardenas, et al. (2016) and found similar overall associations in 1180 infants but used mediation analysis instead of structural equations. Again, most of the association with BW was mediated by GA (‘indirect effect’), while among the smallest (< p20) infants there was also a direct effect.

Lin et al. (2019) also used this Bangladesh study base (now 1057 infants) but used only toenail As and focused on the possible interaction between As and macronutrients (total energy, fat, carbohydrate and fibre intakes). Maternal intakes of macronutrients were associated with BW, but toenail As did not mediate these associations. Interestingly, in regression models adjusted for energy intake and a number of additional potential confounders compared with the study by Kile, Cardenas, et al. (2016), the inverse relation between toenail As and BW was no longer statistically significant (in contrast to the finding in Kile, Cardenas, et al., 2016).

A fourth paper based on the same Bangladesh study base as in Kile, Cardenas, et al. (2016) was published by Bozack et al. (2020). It included 413 infants with data on the percentage of DNA methylation of CpG sites in cord blood. A significant inverse association was found between toenail As and BW, mediated by GA and DNA methylation, indicating an involvement of epigenetic programming. Analyses were not adjusted for intake of macronutrients.

Almberg et al. (2017) found no statistically significant association between water‐As and LBW in > 400,000 live singleton births in Ohio, US. The median water‐As in community water systems by county was 1.5 μg/L (range 0.5–12 μg/L). There was no overall association between water As and LBW (OR 0.99 (0.98–1.10)) or VLBW, but when the analyses were restricted to counties where use of private wells were uncommon (< 10% of households) 20 counties with about 216,000 births remained. Then the adjusted OR for LBW was 1.06 (95% CI 0.98–1.15) and for VLBW it was 1.14 (1.04–1.24). The presence of private wells is assumed to cause misclassification, since exposure categorisation was only based on water As in community water systems.

Shih et al. (2020) found no significant association between u‐DMA and BW in a mother–child cohort from seven areas in the US. The LODs were high, so only total u‐As and u‐DMA could be quantified.

Howe, Farzan, et al. (2020) found no significant associations between maternal u‐tiAs and BW in 167 infants from Los Angeles US. There was an inverse association between hair‐As and BW, but the levels of hair As were very low (median 0.01 μg/g).

In another population from Los Angeles, Howe, Claus Henn, et al. (2020) found no statistically significant associations between maternal ui‐As and BW in relation to GA in 262 mother‐infant pairs.

Bulka et al. (2022) estimated the probability that As in private well water exceeded certain cut‐offs in the whole of US and compared aggregated estimates on county level with BW in about 3.3 million births in 2016. Estimates were based on machine learning methods taking into account As measurements and a large number of other factors. Associations between the estimated probabilities for As in water and BW were examined, assuming that also As in public water sources was correlated with As in private wells. Significant associations were found as shown in Table 15, though the methods and units make it difficult to evaluate the magnitude of association.

Among the 19 studies using water As, u‐tiAs or toenail As as exposure metrics, the studies by Huyck et al. (2007) and Gelmann et al. (2013) examined very few births. The study by Shih et al. (2020) had high As detection limits. These three studies are therefore less informative in risk assessment.

Overall summary on As and birth weight (BW)

Overall, several studies on exposure to iAs, mainly from As in drinking water show an inverse association with BW. Exposure misclassification (both in Bangladesh and elsewhere), and questionable adjustment for GA may have attenuated some exposure‐outcome estimates. The studies from Bangladesh (Kile, Cardenas, et al., 2016; Rahman et al., 2009) provide strong evidence for reduced BW at low to moderate exposure to iAs. The studies from Chile (Hopenhayn et al., 2003) and Taiwan (Yang et al., 2003) provide moderate evidence for such an association, but there was no association in studies from Mongolia (Myers et al., 2010) and Mexico (Laine et al., 2015). The US studies have low exposure to iAs, low exposure contrast, and there may be an impact of organic As on the As biomarkers, e.g. DMA. This makes these studies less informative. The US study by Bulka et al. (2022) found a significant association despite crude estimates on the individual level, which causes misclassification and therefore may have attenuated a true association. In Bangladesh maternal and fetal undernutrition is common, and low BW and PTB is much more common than in Europe. Therefore, it is uncertain if the findings from Bangladesh are relevant for European populations.

In summary, the epidemiological studies provide sufficient evidence for an association between low to moderate exposure to iAs and BW in Bangladesh. Studies from other countries are not consistent. The relevance for Europe of the results from Bangladesh is unclear.

Preterm birth and gestational age

Many of the studies of birth weight reviewed above also examined associations with preterm birth (PTB) and gestational age (GA). They are summarised in Table 16. Of the studies reviewed in the 2009 Opinion, only one (Yang et al., 2003) met the inclusion criteria for the current evaluation. This study found no statistically significant association between arsenic in water and PTB in Taiwan.

TABLE 16

Key epidemiological studies on associations between As exposure and preterm birth (PTB GA < 37 weeks) or gestational age (GA).

Reference study population designOutcome definitionPopulation size (n)Arsenic exposure a ResultsAdditional information/confounders

Yang et al. (2003)

Taiwan

Cohort

PTB18,259 (3872 from As‐exposed townships, and 14,387 from similar reference townships)Water: In As‐exposed townships 30% > 50 μg/L639 cases (3.5%) of PTB. OR 1.10 (0.91–1.33) for As‐exposed womenAggregated water As data. Adjusted for some potential confounders

Myers et al. (2010)

Inner Mongolia, China

CS

PTB9890 singleton births 1996–1999 with As measured in the wells of the subvillageWater: 66% < 20, 20% 21–50, 7% 50–100, 7% > 100 μg/L. Data from 1991 to 1997 in well‐water registry289 cases (2.9%) of PTB. OR 1.02 (0.72–1.44) for water‐As > 50 versus < 50 μg/LWater‐As data were averaged by subvillage. Adjusted only for antenatal care. No other potential confounders were detected. Women were generally health and well‐nourished

Laine et al. (2015)

Mexico

CS

GA199 singleton live births, only 3 with PTBWater: < 0.5–236, median 13 μg/L. Maternal u‐tiAs median 23 μg/L

Median GA 39 weeks

No association btw w‐As, u‐DMA and GA

u‐tiAs, u‐MMA and u‐%MMA had significant adjusted negative slopes

Mean u‐DMA 43 μg/L in women reporting seafood and 31 μg/L in the others

Gilbert‐Diamond et al. (2016)

USA, New Hampshire

Cohort

GA706 live singleton birthsMaternal u‐tiAs. Median 3.4 μg/LMedian GA 39.5. No association btw u‐tiAs, MMA or DMA and GAGeneral additive models adjusted for some potential confounders

Kile, Cardenas, et al. (2016)

Rural Bangladesh

Cohort

GA1153 live singleton births

Water: < 0.5–1400, median 2.3 μg/L

Maternal toenail t‐As 0.2–35 μg/g, median 1.5 μg/g

254 (22%) with PTB. Significant adjusted inverse association (multiple linear regression) btw water As and GA, and btw nail‐As and GAStructural equation modelling suggested that the impact of As on GA mediated almost all the association btw As and birth weight

Almberg et al. (2017)

USA, Ohio

CS

PTB428,804 live singleton births from records 2006–2008. In term births 2.9% LBW and 1.2% VLBWw‐As aggregated by 88 counties and year: 0.5–12 μg/L, median 1.5 μg/L. Based on 2968 measurements in 975 water systems (only regulated water systems, As in private wells unknown)

11% were PTB. Overall adjusted OR (per mean As in μg/L) for PTB 0.99 (0.98–1.01).

Adjusted OR for women from counties with < 20% private wells: 1.08 (1.02–1.14)

ORs adjusted for many potential confounders from the records.

ORs were increased in subanalyses excluding counties with many private wells

Lin et al. (2019)

Rural Bangladesh

Cohort

GA1057 live singleton births, Same study bas as Kile, Cardenas, et al. (2016) and Rahman et al. (2017)Toenail As 0.04–47, median 1.2 μg/gGA inversely associated with protein, fat and fibre intake, but positively associated with carbohydrate intake. Nail As inversely associated with GA, when adjusted for macronutrientsAs Kile, Cardenas, et al. (2016), but points out the impact of macronutrients, some of which are also associated with As

Bozack et al. (2020)

Rural Bangladesh

Cohort

GASubset of Kile, Cardenas, et al. (2016): 413 births with % methylation of DNA cord blood at 3 CpG sites in the DNA sequence of the gene DNA methyltransferase 3 alphaSee Kile, Cardenas, et al. (2016)Significant inverse association between nail As and GASee Kile, Cardenas, et al. (2016), Rahman et al. (2017) and Lin et al. (2019). Here structural equation modelling also includes DNA methylation in cord blood

Howe, Farzan, et al. (2020)

USA, Los Angeles

Cohort

GA

Births with maternal:

116 (hair‐As)

100 (blood As)

167 (u‐iAs)

Medians:

Hair: 0.01 μg/g

Blood: 0.67 μg/g

u‐tiAs: 5.7 μg/L

No association with u‐iAsIn urine additionally 0.5 μg/L AsB

Shih et al. (2020)

USA, seven areas

Cohort (NCS)

GA212Total As (median 7.8 and DMA median 3.4 μg/L) in maternal urineMean GA 38.8 weeks. No association btw u‐DMA and GAHigh LODs: t‐As 1.25, DMA 1.7, MMA 0.9. All had t‐As > LOD and 77% had DMA > LOD, but for other species > 60% < LOD

Bulka et al. (2022)

USA nationwide

Cross‐sectional

GA, PTB3,580,755 births in 3105 countiesThe probability of As in water in four categories (≤ 1, > 1, ≤ 5, > 5, ≤ 10, > 10) was estimated in 1 × 1 km grids, based on about 20,000 As measurements in private wells. Estimates were then aggregated on county level

Adjusted beta (grams) per 10% increase in probability of exceeding the following thresholds:

≤ 1, > 1

−0.01 (−0.01, −0.01)

≤ 5, > 5

−0.01 (−0.01, −0.00)

≤ 10, > 10

−0.00 (−0.02, −0.00)

Risk differences for PTB zero in all comparisons

Adjusted for proportion of private wells, maternal age, race/ethnicity, marital status, education, smoking during pregnancy, pre‐pregnancy BMI, rurality/urbanicity, annual concentration of PM < 2.5 μm in the county

Abbreviations: As, arsenic; AsB, arsenobetaine; BMI, body mass index; CpG, cytosine‐phosphate‐guanine; CS, cross‐sectional study; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; DNA, deoxyribonucleic acid; GA, gestational age; km, kilometre(s); LOD, limit of detection; LBW, low birth weight; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; n, number; NCS, National Children's Study; OR, odds ratio; PM, particulate matter; PTB, pre‐term birth; t‐As, total arsenic; u‐DMA, urinary DMA; u‐MMA, urinary MMA. u‐tiAs, total urinary inorganic arsenic (sum of inorganic arsenic and its methylated metabolites MMA and DMA); USA, United States of America; VLBW, very low birth weight; w‐As, water‐arsenic.

a iAs in urine includes MMA and DMA, but not AsB.

The CONTAM Panel identified 11 additional papers, meeting the inclusion criteria, that were not included in the EFSA 2009 Opinion.

Some of the studies on GA or PTB that did not fulfil the inclusion criteria are found among those mentioned in the previous Section on BW. Another study not fulfilling the inclusion criteria was a study on PTB by Huang et al. (2021).

Two of the additional studies (Myers et al., 2010 in Mongolia and Almberg et al., 2017 in the US) evaluated the association between iAs exposure and PTB. Myers et al. (2010) found no significant association with PTB, while the study by Almberg et al. (2017) found an association with PTB in a subset of births when the analyses were restricted to counties where use of private wells were uncommon (< 10% of households) and where therefore the exposure assessment is likely to be more valid.

Seven studies that had become available since the 2009 Opinion evaluated the association with GA (see Table 16). The studies from Mexico (Laine et al., 2015), and the US (Gilbert‐Diamond et al., 2016; Howe, Farzan, et al., 2020; Shih et al., 2020) showed no significant associations between u‐tiAs and GA. u‐tiAs levels in the US studies were low. The studies based on a Bangladesh cohort (Bozack et al., 2020; Kile, Cardenas, et al., 2016; Lin et al., 2019) showed inverse associations between u‐tiAs and toenail As and GA. As mentioned in the Section on BW, the inverse association between iAs exposure and BW seemed to be mediated by the inverse association between iAs exposure and GA.

Overall, there is some evidence from the study by Kile, Cardenas, et al. (2016) in Bangladesh that iAs exposure reduces GA (mean 38 weeks in the study). For populations in other countries the evidence of an association is insufficient.

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and gestational age.

Other anthropometric birth outcomes and in utero growth before birth

Among the studies mentioned in the Section on birth weight, five also examined other anthropometric data (Bloom et al., 2016; Gilbert‐Diamond et al., 2016; Laine et al., 2015; Rahman et al., 2009; Shih et al., 2020). For characteristics of the studies, see Table 15 on BW.

Thus, for birth length, head circumference, chest circumference and ponderal index, relatively few studies examined associations with iAs. The results were not consistent.

In addition, there are two studies which assessed fetal growth in utero by ultrasound. Kippler et al. (2012) studied u‐tiAs in pregnant women in rural Bangladesh, 1885 in gestational week (GW) 8 and 1929 at GW 30. Head size, abdominal circumference and femur length were measured by ultrasound at GW 14 and 30. Median maternal u‐tiAs was 79 μg/L at GW 8 and 85 μg/L at GW 30. The authors concluded that u‐tiAs was weakly associated with decreased fetal size in boys.

Another study using ultrasound for assessment of fetal growth was published by Davis et al. (2015) based on 223 pregnant women from New Hampshire, US with a median u‐tiAs of 3.1 μg/L at GW 24–28. In adjusted analyses there was no significant association between maternal u‐tiAs and birth size.

In summary, there is some evidence from studies in Bangladesh that iAs exposure reduces birth size (in addition to birth weight as reviewed above). The relevance for Europe of the results from Bangladesh is unclear. For populations in other countries the evidence of an association is insufficient.

Spontaneous abortion, stillbirth, neonatal mortality

The CONTAM Opinion of 2009 noted that increased risk of spontaneous abortion, stillbirth, preterm birth and/or neonatal death at elevated water arsenic concentrations were indicated in some but not all of the reviewed studies (Ahmad et al., 2001; Cherry et al., 2008; Hopenhayn‐Rich et al., 2000; Kwok et al., 2006; Milton et al., 2005; Rahman et al., 2007; Sen & Chaudhuri, 2008; von Ehrenstein et al., 2006). At that time, the Panel concluded that further evidence was required regarding the dose–response relationships and critical exposure times for these outcomes.

The CONTAM Panel identified 20 publications related to spontaneous abortion, stillbirth and neonatal mortality, of which 8 met the inclusion criteria. Five of these (Cherry et al., 2008; Hopenhayn‐Rich et al., 2000; Kwok et al., 2006; Milton et al., 2005; Rahman et al., 2007) were previously described in EFSA CONTAM Opinion (2009), see Table 17.

TABLE 17

Key epidemiological studies on associations between As exposure and spontaneous abortion, stillbirth, neonatal (1–28 days) and infant (1–365 days) mortality.

Reference study population designOutcome definitionPopulation size and number of casesArsenic exposureResultsAdditional information/confounders
Included in 2009 EFSA Opinion

Hopenhayn‐Rich et al. (2000)

Chile

Ecological longitudinal study

Stillbirth

Infant mortality

Populations of Antofagasta and Valparaiso

w‐As in Antofagasta (μg/L):

1958–70: 860

1971–79: 110

1980–87: 70

1988–96: 40

Water As in Valparaiso < 2 μg/L

Rates of stillbirth/1000

1958–69:

Ant 28, Val 11

1970–73:

Ant 19, Val 8

1974–77:

Ant 9, Val 11

1978–96:

Ant 5, Val 6

Rates of neonatal mort.

1958–61:

Ant 46, Val 22*

1962–65:

Ant 43, Val 25*

1966–69:

Ant 34, Val 23*

1970–73:

Ant 24, Val 23*

1974–77:

Ant 21, Val 21

1978–81:

Ant 17, Val 14*

1982–85:

Ant 13, Val 10*

1986–89:

Ant 11, Val 9.3*

1990–93:

Ant 8.5, Val 8.0*

1994–96:

Ant 7.3, Val 8

Slightly more poverty and low education in Valparaiso at time of the study. Stillbirth was more common in Antofagasta only 1958–1973. The asterisks in the Results column indicate time periods when stillbirth or neonatal mortality were higher in Antofagasta with a statistically significant difference. For neonatal mortality, rates were significantly higher in Valparaiso 1974–77 and 1994–96

Milton et al. (2005)

Rural Bangladesh

Cross‐sectional study

Spontaneous abortion Stillbirth Neonatal mortality

533 first pregnancies

Spontaneous abortion: 90

Stillbirth: 60

Neonatal death: 86

w‐As (μg/L) Median 116

< 50 (33%)

51–100 (10%)

> 100 (57%)

< 50 (33%)

51–100 (10%)

> 100 (57%)

< 50 (33%)

51–100 (10%)

> 100 (57%)

OR spontaneous abortion (cases)

1.0 (19)

2.4 (1.2–5.1) (12)

2.5 (1.5–4.4) (59)

Stillbirth (cases)

1.0 (13)

1.1 (0.3–3.1) (4)

2.5 (1.5–5.9) (45)

Neonatal death (cases)

1.0 (16)

2.7 (1.1–6.7) (12)

1.7 (0.8–3.3) (58)

First model was adjusted for education, age at marriage, age at first pregnancy, history of antenatal care, weight, height, age at first menstruation, duration of menstrual period, history of hypertension, diabetes and vaccination during pregnancy

Final model included only covariates altering the OR by > 10%: participant's height, history of hypertension and diabetes, age at first pregnancy

Kwok et al. (2006)

Rural Bangladesh, 261 villages

Cross‐sectional study

Stillbirth

2006 singleton births with As measured in the wells of the villages

Stillbirth: 53 (2.6%)

w‐As: 2003 samples, median about 150 μg/L, 36% > 200 μg/L.

ORs only presented versus As as continuous variable

Adjusted OR 1.00 (1.00–1.00) per μg/L of water‐As

Water samples (for each household), birth data for 2002, and covariates were collected in 2003 from Community Centres' registers + maternal interviews (2003). Most births at home.

Adjusted for maternal age, household assets, smoking in the household, parity, baseline BMI and duration of tubewell‐use. Strange CI for the OR

Rahman et al. (2007)

Matlab Bangladesh

Cohort study

Neonatal mortality

Infant mortality

29,134 pregnancies

Stillbirth: 829

Neonatal death: 850

Infant death: 1373

w‐As (μg/L), quintiles:

< 10 (median < 1)

10–166 (median 77)

167–276 (median 225)

277–408 (median 340)

≥ 409 (median 515)

< 10 (median < 1)

10–166 (median 77)

167–276 (median 225)

277–408 (median 340)

≥ 409 (median 515)

HR Neonatal death

1.0

1.11 (0.89–1.38)

1.18 (0.95–1.47)

1.17 (0.94–1.46)

1.21 (0.98–1.50)

HR Infant death

1.0

1.13 (0.95–1.35)

1.19 (1.00–1.42)

1.29 (1.08–1.53)

1.19 (1.00–1.41)

Age at pregnancy, order of pregnancy, education, socioeconomic status by asset score, six seasons, calendar year of outcome, location according to service area) were tested, but only calendar was a confounder and adjusted for

p for trend for infant death 0.02

HR for stillbirth not reported

Cherry et al. (2008)

Rural Bangladesh

Semi‐ecological longitudinal study

Stillbirth

30,486 pregnancies in 16 areas with As measured in 7–14 wells in each area

Stillbirth: 971 (3.4%)

w‐As (μg/L)

< 10

10–49

≥ 50

OR

1.0

1.23 (0.87–1.74)

1.80 (1.14–2.86)

As in water averaged per area. Adjusted for individual factors: sex, age, multiple birth, previous pregnancies, previous stillbirth, low SES, maternal and paternal education, maternal high blood pressure and oedema, short gestation, low birth weight, prolonged labour, excess bleeding
Other studies on iAs exposure

Myers et al. (2010)

Inner Mongolia, China

Cohort study

Stillbirth

Neonatal mortality

9890 singleton births 1996–1999 with As measured in the wells of the sub village.

Stillbirth: 51

Neonatal death: 64

Water: 66% < 20, 20% 21–50, 7% 50–100, 7% > 100 μg/L. Data from 1991 to 1997

≤ 50 (median < 20)

> 50 (median about 100)

≤ 50 (median < 20)

> 50 (median about 100)

OR (number of cases)

Stillbirth:

1.0 (43)

1.18 (0.55–2.51) (8)

Neonatal death:

1.0 (49)

2.01 (1.12–3.59) (15)

Maternal age, child sex, previous pregnancies and adequacy of prenatal care utilisation were evaluated, but the only confounder was prenatal care, which was adjusted for

Rahman et al. (2010)

Matlab Bangladesh

Cohort study

Spontaneous abortion

Stillbirth

Infant mortality

2924 pregnancies with data on u‐As in GW8.

Spontaneous abortion 275 (9.4%).

For stillbirth and

1725 pregnancies with data on u‐As in GW8 and GW30.

Stillbirth: 32 (1.9%)

Infant mortality: 36 (2.1%)

u‐tiAs (μg/L), quintiles in GW8, adjusted for SG (median)

< 33 (23)

33–57 (42)

58–121 (80)

122–248 (177)

> 248 (382)

u‐tiAs (mean of GW8 and GW30) (median)

< 38 (30)

39–67 (50)

68–133 (94)

134–267 (189)

> 267 (390)

< 38 (30)

39–67 (50)

68–133 (94)

134–267 (189)

> 267 (390)

OR

Spontaneous abortion:

1.00 (45)

1.28 (0.85–1.93) (57)

1.41 (0.94–2.11) (63)

1.06 (0.69–1.62) (47)

1.44 (0.96–2.15) (63)

Stillbirth:

1.0 (3)

2.06 (0.51–8.38) (6)

2.35 (0.60–9.23) (7)

3.41 (0.92–12.63) (10)

2.02 (0.50–8.24) (6)

Infant mortality:

1.0 (3)

1.78 (0.44–7.16) (6)

1.83 (0.45–7.35) (6)

2.29 (0.58–9.05) (7)

5.01 (1.41–17.84) (14)

Maternal age, previous pregnancies and births, education, economic status, location of residence, season, BMI and GA were evaluated, but none was a confounder for spontaneous abortion.

OR for stillbirth adjusted for economic status and GA

OR for infant mortality adjusted for economic status, location of residence, season and GA

Bloom et al. (2014)

Romania

Case–control study

Spontaneous abortion

150 cases and 150 controls

Water As (μg/L), median 0.4, p90 9.4.

Median (range)

Cases: 0.17 (0–175)

Controls: 1.28 (0–130)

OR 0.98 (0.96–1.01)Adjusted for GA, maternal age smoking in pregnancy, education, vitamin use

Abbreviations: Ant, Antofagasta; As, arsenic; BMI, body mass index; CI, confidence interval; EFSA, European Food Safety Authority; GA, gestational age; GW, gestational week; HR, hazard ratio; OR, odds ratio; SES, socioeconomic status; SG, specific gravity; u‐As, urinary arsenic; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA); Val, Valparaiso; w‐As, water arsenic. *statistically significant difference.

Hopenhayn‐Rich et al. (2000) performed an ecological study in Chile, comparing the rates of stillbirths and infant mortality in the population of Antofagasta, where public water had high As levels with the population of Valparaiso where water As was low (< 2 μg/L). Average water As in Antofagasta was 860 μg/L in 1958–1970, 110 μg/L 1971–1979, 70 μg/L in 1980–1987and 40 μg/L in 1988–1996. Stillbirths declined markedly in Chile over this period, see Table 17. The risk differences in 1958–1969 and 1970–1973 are highly statistically significant. The average socioeconomic status was slightly lower in Valparaiso than in Antofagasta (based on education and % poverty). From 1974 to 1996, the rates were the same in the two cities. The rates of neonatal mortality were also higher in Antofagasta, and the rate difference was significantly higher also in the 1980s when water As was 70 μg/L.

Milton et al. (2005) examined the association between As in drinking water and spontaneous abortion, stillbirth or neonatal death (within 28 days in live births) in 533 first pregnancies in rural Bangladesh and found significant associations with As in water (Table 17).

The study by Kwok et al. (2006) found no association between As in water and stillbirth, based on 53 cases (2.6%) in rural Bangladesh. The study only presents associations with As in water as a continuous variable. The CI is remarkably narrow, considering the low number of cases, so the result is difficult to assess.

Rahman et al. (2007) performed a large register‐based cohort study in Matlab, Bangladesh on associations between As in water and stillbirth, neonatal (within 28 days) death and infant death (within 12 months). Associations with stillbirth were not reported separately from other fetal loss (including induced abortion). For infant mortality there was a significant positive association with increasing water As with similar estimates if divided into death within 28 days or later.

Cherry et al. (2008) performed a large study of water As vs. stillbirth in rural Bangladesh. Based on health registers, 971 stillbirths (3.4%) were found among 30,500 pregnancies in 600 villages. The water As concentrations in 13 subareas were averaged (7–14 tested wells per area) but outcomes and potential confounders were available on the individual level. The authors consider the ORs likely to be underestimated due to (1) the fact that low birth weight and short gestation (which are associated with As exposure and likely mediators) were adjusted for and (2) the averaged exposure estimates of water As, which will cause non‐differential misclassification and attenuation of a true risk. This interpretation is reasonable.

Three studies not included in the EFSA CONTAM Opinion 2009 are shown in Table 17, Rahman et al. (2010) studied the risk of spontaneous abortion, stillbirth and infant mortality versus As exposure (u‐As in pregnancy weeks 8 and 30) in rural Bangladesh (Matlab area). There were significant dose–response associations with stillbirth (p = 0.02 for linear trend) and infant mortality (p = 0.005).

Myers et al. (2010) examined associations between water As and stillbirth and neonatal death in Mongolia. The OR for water As > 50 μg/L versus < 50 μg/L was 1.18 (95% CI 0.55–2.51) for stillbirth (eight cases in the higher category) and 2.01 (1.12–3.59) for neonatal death (15 cases in the higher category).

Bloom et al. (2014) performed a case–control study of As in maternal drinking water and spontaneous abortion before GW 20 and found no association in Romania. The median water As concentrations were, however, low both in cases and controls.

Several studies not fulfilling the inclusion criteria in the present assessment were identified (Ahamed et al., 2006; Ahmad et al., 2001; Aschengrau et al., 1989; Börzsönyi et al., 1992; Buck Louis et al., 2017; Chakraborti et al., 20032004; Guo et al., 2003; Mukherjee et al., 2005; Sen & Chaudhuri, 2008; Shih et al., 2017; von Ehrenstein et al., 2006).

In summary, eight studies fulfilled the inclusion criteria, one from Chile, one from Romania, one from Mongolia and five from Bangladesh.

For spontaneous abortion the studies by Milton et al. (2005), and Rahman et al. (2010) provide sufficient evidence for an association with exposure to iAs in Bangladesh, but the relevance for Europe is unclear.

Regarding stillbirth, there is sufficient evidence for an association with exposure to inorganic As in Bangladesh based on the studies by Milton et al. (2005), Cherry et al. (2008) and Rahman et al. (2010), but the relevance for a Europe is unclear. The study from Chile (Hopenhayn‐Rich et al., 2000) suggested an increased risk at water As of about 100 μg/L, but the risk was not increased in the study from Mongolia at similar water As levels.

For infant mortality (in the neonatal period or later) associations with exposure to inorganic As have been shown in several studies from Bangladesh, as well as in the Chilean study at water As levels of 70 μg/L and in the study from Mongolia at a water As level of about 100 μg/L. Thus, there is sufficient evidence for an association between iAs exposure and infant mortality.

Birth Defects/Congenital malformations

Teratogenicity of arsenic has been shown in animals (see EFSA CONTAM Panel, 2009), especially neural tube defects (NTD), such as anencephaly, meningocele and myelomeningocele (spina bifida). Other relatively common birth defects in humans include congenital heart disease, such as septal defects and orofacial defects (cleft lip and/or palate), which are reviewed separately below.

The previous EFSA Opinion reviewed one study on birth defects in relation to water As (Kwok et al., 2006). Further details on that study are found in the birth weight Section. There was possibly a weak association between water‐As and risk of birth defects, but there were only 11 cases (in 2189 births) and of very different character.

The CONTAM Panel identified 28 additional studies on associations between As exposure and birth defects, 14 of which met the inclusion criteria. The other 14 studies did not fulfil the inclusion criteria because it was either not possible to evaluate the contributions of organic vs. inorganic As (Demir et al., 2019; Jin et al., 2014; Nyanza et al., 2020; Ovayolu et al., 2020; Özel et al., 2019; van Brusselen et al., 2020; Wang, Pi, Yin, et al., 2022), or because of the use of soil As concentrations (Huang et al., 2011; Wu et al., 20112014) or As in placenta (Jin et al., 2013; Pi et al., 2023) as surrogate for As exposure, or because the studies were purely ecological (Engel & Smith, 1994; Sanders et al., 2014; Yu & Zhang, 2011).

Among the 13 additional studies that fulfilled the inclusion criteria, five examined NTD (Brender et al., 2006; Mazumdar et al., 2015; Tauheed et al., 2017; Tindula et al., 2021; Wang, Zhu, et al., 2019), four examined congenital heart disease (Jin et al., 2016; Richter et al., 2022; Rudnai et al., 2014; Zierler et al., 1988) and two (Marie et al., 2018; Suhl et al., 2022) examined many types of birth defects, including NTD and congenital heart disease). One study (Suhl et al., 2018), was focused on OFC.

Neural tube defects

Brender et al. (2006) performed a case–control study of pregnancies with NTD and normal pregnancies from the same area in Texas, US. Total As in urine was analysed in 56 cases and 74 controls 1 year after conception, and participants instructed to refrain from seafood for 2 days. No association was found between u‐As and NTD. However, the levels of total As in urine (median 9 μg/L) were comparable with those found in populations without elevated iAs exposure.

Mazumdar et al. (2015) performed a case–control study in Bangladesh including 57 cases of myelomeningocele and 55 controls. As in drinking water was lower in cases than in controls (75‐percentile 13 μg/L in the cases and 55 μg/L in the controls). Data for a number of potential confounders, including folate supplementation, was collected. No association was found between water As and risk of myelomeningocele, but the interaction between water As and folate supplementation was significant with respect to risk of myelomeningocele, high water As decreasing the protective impact of folate. In a subsequent subgroup study (Tauheed et al., 2017) also investigated epigenetic markers, but the study provides no further data on associations between As exposure and myelomeningocele. The same group performed a new case–control study in Bangladesh with 192 cases of spina bifida (meningocele or myelomeningocele) and 171 hospital‐based controls (Tindula et al., 2021). As was analysed in toenail clippings from mothers and fathers for 155 cases and 123 controls. The GMs of maternal nail As in cases and controls were 0.55 and 0.53 μg/g, and for paternal nail As the GMs were 0.72 and 0.52 μg/g. There was no association between maternal nail‐As and adjusted risk of spina bifida, nor any interaction with maternal serum folate. The OR for paternal toenail As was increased (1.7, 95% CI 1.3–2.4).

Marie et al. (2018) studied the association between several congenital anomalies (including NTD) and water As in a region of France with elevated As in ground water. They included 5263 children with various anomalies and collected data on As in tap water aggregated by commune for each mother/child in the 12 months before birth. Children were categorised in those with water As < 10 μg/L (92.6%, N = 4871) and ≥ 10 (7.4%, N = 392, median 15.5) μg/L. Among cases with water As ≥ 10, only 18 cases (0.03% of 5263) had water As > 30 μg/L. In an adjusted model, also including an interaction term for gender, the OR for any anomaly was 2.2 (95% CI 1.3–3.6). The interaction with gender was significant, with an increased OR only found for girls (OR 2.4, 95% CI 1.4–4.1). In boys the OR was 0.8 (95% CI 0.4–1.5). For girls, overall ORs for anomalies could be separated into ≥ 10–30 μg/L (OR 2.2, 95% CI 1.2–2.8) and ≥ 30 μg/L (OR 10, 95% CI 2.0–41). There was no association between water As and risk of NTD.

In a case–control study from China (511 controls and 263 cases with neural tube defects) with maternal hair arsenic measurements 3 months before and 3 months after conception, a higher hair arsenic concentration in the cases than in the controls was found, median (inter‐quartile range) of 0.093 (0.025–0.387) μg/g hair, and 0.082 (0.030–0.414) μg/g in the controls (Wang, Zhu, et al., 2019). However, in the adjusted logistic regression analysis, maternal hair arsenic concentration above its median of the controls was not associated with a risk of total neural tube defects (OR = 1.32, 95% CI 0.91–1.92), and there was no dose–response relationship.

Suhl et al. (2022) examined non‐cardiac birth defects in a large case–control study in the US. Cases were retrieved from 10 US States in 1997–2011, and controls were selected from hospital logs and birth certificates matched by State and year of the cases. The exposure to iAs was estimated from a food frequency questionnaire, and potential confounders from telephone interviews. The mean iAs intake was estimated at 0.08 μg/kg body weight and day in cases and controls. There were 2130 cases of CNS malformations, about 1600 of which were NTD. There were no significant associations between estimated iAs intake and risk of NTD (aOR for anencephaly 0.8 (0.6–1.0), and for spina bifida 0.8 (0.7–1.0).

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and neural tube defects.

Congenital heart disease

An early study, not described in EFSA CONTAM Panel (2009) was a case–control study of congenital heart disease in Massachusetts, US by Zierler et al. (1988), Table 18. As levels were mostly below the detection limit (LOD 0.8 μg/L), and the 90‐percentile was 0.8 μg/L. It should be noted that at As < 1 μg/L, the contribution of As from drinking water is low compared to intake of iAs from other dietary components. For example, the median lower bound estimated intake for European adults at the time the previous EFSA Opinion (EFSA CONTAM Panel, 2009) was about 0.3 μg/kg corresponding to 20 μg/day for a 70 kg person.

TABLE 18

Key epidemiological studies relating arsenic and congenital heart disease.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Zierler et al. (1988)

Massachusetts, USA

Case–control study

Congenital heart disease in live births, retrieved from birth registry (excluding persistent ductus art. and Downs syndrome)270 cases and 665 controls (random from the same registry)w‐As in public water supplies, P90 was 0.8 μg/L, which was also the LODaOR for detectable vs. no‐detectable w‐As was 0.98 (0.59–1.64) based on 31 cases with As > LODAdjusted for type of water source (ground or surface), other water contaminants and maternal education. OR for coarctation of aorta was 3.4 (1.3–8.9)

Rudnai et al. (2014)

Hungary

Case–control study

Congenital heart disease in live births, retrieved from registry of congenital malformations over 1987–20034889 cases and 4007 controls from the same registry. Controls were children with club foot, Downs syndrome and multiple malformations

w‐As (μg/L)

0–10 (80%)

10–20 (12%)

20–30 (3.5%)

30–40 (2%)

40–50 (1.1%)

> 50 (1.4%)

OR

1.0

1.5 (1.3–1.7)

1.3 (1.0–1.6)

1.4 (1.1–1.9)

1.4 (0.9–2.1)

1.0 (0.7–1.4)

Adjusted for child sex and age of mother

OR for w‐As > 10 μg/L was increased both in urban and rural areas

Jin et al. (2016)

China

Case–control study

Congenital heart disease from hospital records339 cases and 333 controls

Hair As ng/g

Medians 74 (controls and 89 (cases)

< 62

62–86

87–118

> 118

OR

1.0

2.34 (1.46–3.76)

3.43 (2.23–5.83)

5.62 (3.43–9.24)

Adjusted for maternal age, gestational age, previous pregnancy, folic acid suppl., BMI, paternal smoking

These levels are relatively normal also for humans without known exposure to elevated iAs

Marie et al. (2018)

Auvergne, France

Cross‐sectional study

Congenital heart disease from national registry

4891 pregnancies

99 cases

w‐As (μg/L) 12 months before birth

7.4% had ≥ 10 μg/L

< 10

≥ 10 (median 15.5)

< 10

≥ 10

OR

Males (2416)

1

0.73 (0.17–2.13)

Females (2475)

1

3.66 (1.62–7.64)

w‐As aggregated on commune level

ORs adjusted for maternal age, parity, paid employment, size of municipality, year of birth. Interaction with sex was significant

Richter et al. (2022)

Denmark

Cohort study

Congenital heart disease

From national registry

1,042,413 births in 1997–2014

10,600 cases

w‐As (μg/L) (number)

< 0.5 (495970)

0.5–0.9 (235630)

1.0–4.9 (292222)

≥ 5 (18591)

OR (cases)

1.0 (4583)

1.13 (1.08–1.19) (2339)

1.33 (1.27–1.49) (3477)

1.42 (1.24–1.62) (228)

w‐As aggregated by water supplies

Adjusted for year of birth, mother's education and ethnicity. No interaction with sex. Excluding private wells did not change results

Abbreviations: As, arsenic; aOR, adjusted odds ratio; BMI, body mass index; iAs, inorganic arsenic; LOD, limit of detection; n, number; OR, odds ratio; USA, United States of America; w‐As, water arsenic.

Rudnai et al. (2014) performed a very large case–control study, in Hungary, of congenital heart disease. Cases and controls were selected from the same registry, and controls were births with the diagnoses Down syndrome, Club foot and multiple congenital malformations. Data on As in drinking water in 2412 settlements all over Hungary (urban and rural) were collected for each year of the observation period. It should be noted that births with multiple malformations as controls might result in an underestimation of a possible true risk. The exposure response relation was not linear. The OR increased up to water As of 10–20 μg/L, then did not increase further. No increased risk was seen at water As > 50 μg/L (see Table 18).

Jin et al. (2016) studied associations between As in hair and risk of congenital heart disease in a case–control study in China. Increased adjusted ORs were found for quartiles 2–4 of hair‐As compared to the first quartiles, with a dose–response pattern. The median hair‐As was 0.074 μg/g in controls and 0.089 μg/g in the cases. These hair‐As are compatible with other reports in general populations without known increased exposure to iAs (Fowler et al., 2022).

The study by Marie et al. (2018) found an association between As in water and risk of congenital heart defects, but only in girls. The interaction with gender was significant.

Richter et al. (2022) studied the association between As in drinking water (98% from public water works) and congenital heart disease in a national cohort study including all live births in Denmark 1997–2014. Water As in early pregnancy based on residential address and As in water supply areas. There were 3700 cases of congenital heart disease with water‐As > 1 μg/L, and 228 cases with water‐As ≥ 5 μg/L. The adjusted OR increased in all categories of water As above the reference category, which was < 0.5 μg/L with a supralinear exposure‐response relation. Most of the increase in risk was found from the reference category up to a few μg/L. There was no interaction with sex. Results were essentially the same in a large number of sensitivity analyses. As mentioned above, the contribution of As from drinking water at levels < 1 μg/L is low compared to intake of iAs from other dietary components.

In the studies by Zierler et al. (1988) in the US, and by Richter et al. (2022) in Denmark, the water‐As levels (aggregate data linking residences with measurements in public water works) were very low. The size of the study by Zierler et al. (1988), which found no association with congenital heart disease, was modest with 31 cases with estimated As in water > 0.8 μg/L. The Danish cohort study by Richter et al. (2022) had a very impressive size, analysing 10,600 cases of congenital heart disease in all Denmark over an 18‐year period. This study found a very clear association between estimated water As and congenital heart disease. Imprecision in the estimates from the aggregated data, and the varying contribution of iAs in other dietary items might have caused misclassification of iAs exposure, which can be assumed to be non‐differential, and therefore the true association between iAs exposure and the outcome may be even stronger. A caveat is that the exposure‐response found is strange, since the increase in relative risk per increase of water‐As is highest at the very low water‐As levels. This may be less biologically plausible.

In the studies by Rudnai et al. (2014) in Hungary, and Marie et al. (2018) in France there were larger proportions of the populations with high water As as compared to the studies by Zierler et al. (1988) and Richter et al. (2022). The results in these two studies support an association with congenital heart disease at water‐As > 10 μg/L, although Marie et al. (2018) found an increased risk only in girls.

All studies reviewed above find associations between iAs exposure and congenital heart disease. In summary, the epidemiological studies provide sufficient evidence for an association between low to moderate exposure to iAs and congenital heart disease. However, the exposure‐response relations in key studies were atypical.

Table 18 depicts the studies on As and congenital heart defects.

Water As and orofacial cleft

Suhl et al. (2018) performed a case–control study of OFC and As exposure in Iowa, US. 370 cases 1067 controls. There was no association between drinking water As and risk of OFC, but only about 4% had water As > 5 μg/L. Therefore, the impact of water As on total exposure to iAs should be modest. This single study with modest water‐As levels and no increased risk does not permit any conclusion on evidence regarding this birth defect.

In summary there is insufficient evidence of an association between exposure to iAs and OFC.

Water As and any birth defect

The study by Kwok et al. (2006) is too small to permit any conclusions. The study by Marie et al. (2018) found an association between water‐As and ‘any birth defect’ but only in girls.

In summary there is insufficient evidence of an association between exposure to iAs and ‘any’ birth defect.

Growth after birth

Four cohort studies including mother–child pairs investigated the associations between iAs and children's growth, two of which were from Bangladesh and one from the US (Gardner et al., 2013; Igra et al., 2021; Muse et al., 2020; Saha et al., 2012). One of the studies measured concentrations of iAs in urine only in mothers during pregnancy, whereas the other two measured it also in the children. A study from Bangladesh who followed the children until the age of 2 years, found no associations when the quintiles with the highest concentrations of iAs in urine were compared with the quintiles with the lowest concentrations (Saha et al., 2012). Another study from Bangladesh found an association between concentration of iAs in urine and the children's weight at 5 years of age, most apparent among girls (Gardner et al., 2013). However, no association was observed between prenatal exposure (maternal urine at early gestation) and children's weight at 5 years of age. A third study from Bangladesh which followed the children until the age of 10 years did not observe associations between iAs and growth (Igra et al., 2021). A study from the US observed an association between concentrations of iAs in urine and increased length during the first year of life (Muse et al., 2020). However, although the association was statistically significant the magnitude of the effect was very small, each doubling of tAs increased the length among males with 0.12 cm and among females with 0.13 cm. In addition, no associations were observed in the study between concentrations of iAs in urine and weight gain, change in weight‐for‐length or head circumference.

Table 19 depicts the studies on children's growth.

TABLE 19

Key epidemiological studies on children's growth in humans in relation to iAs exposure.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Saha et al. (2012)

Bangladesh

Cohort study

Weight and length from birth to 2 years of age2372 mother–child pairs

Urinary exposure of iAs, μg/L (median)

Mothers at 8 weeks gestation: 80.9

Mothers at 30 weeks gestation: 84.0

Children at 18 months: 34.2

No significant associations at 2 years of age when comparing highest versus lowest quintiles in the adjusted analysesAdjusted for age, sex, maternal BMI and SES

Gardner et al. (2013)

Bangladesh

Cohort study

Children's weight and height at 5 years of age1505 mother–child pairs

Urinary exposure of u‐tiAs μg/L (median)

Mothers at 8 weeks gestation: 82

Children at 5 years: 51

Adjusted differences when comparing the highest (≥ 95th percentile) with the lowest (≤ 5th percentile):

Weight:

−0.33 kg (95% CI; −0.60, −0.06)

Height:

−0.50 cm (95% CI; −1.20, 0.21)

The associations were apparent primarily among girls

Adjusted for child's sex, family SES, season of birth, gestational age, maternal education, maternal height or BMI, maternal tobacco chewing, indoor cooking without ventilation and birth order

The study did also include Cd and Pb

Muse et al. (2020)

New Hampshire, USA

Cohort study

Growth parameters from medical records at different timepoints during the first year760 mother–child pairs

Urinary exposure, μg/L (median)

Maternal in second trimester

iAs 0.36

MMA 0.34

DMA 2.84

u‐tiAs 3.96

Each doubling of urinary u‐tiAs increased the length over the first year of life with ~ 0.12 cm in males and 0.13 cm in females

No associations with weight gain, change in weight‐for‐length and head circumferences

Adjusted for gestational age, sex, maternal BMI, IOM gestational weight gain, education, parity, duration of breast feeding and smoking

Igra et al. (2021)

Bangladesh

Cohort study

Growth until the age of 10 years1530 mother–child pairs

Maternal erythrocytes during pregnancy, μg/kg (median): 4.3

Total urinary in children's urine at age 10 years, μg/L (median): 57

No associations with growth up to 10 yearsAdjusted for gender, age at 10 years, maternal parity, education, socioeconomic status, maternal height and weight

Abbreviations: BMI, body mass index; Cd, cadmium; CI, confidence interval; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; iAs, inorganic arsenic; IOM, Institute of Medicine; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; n, number; Pb, lead; SES, socioeconomic status; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA), USA, United States of America.

In summary, there is insufficient evidence that iAs is associated with children's growth.

Neurodevelopmental effects

The previous EFSA CONTAM Opinion (EFSA CONTAM Panel, 2009), described eight studies investigating associations between iAs exposure and developmental effects (cognitive performance) (Calderon et al., 2001; Rosado et al., 2007; Tofail et al., 2009; Tseng et al., 2006; von Ehrenstein et al., 2007; Wang, Wang, et al., 2007; Wasserman et al., 2004, 2007). Based on those studies, the CONTAM Panel concluded ‘Taken together, these studies provide some evidence for neurobehavioural effects of arsenic during childhood, also at exposure levels occurring in areas with elevated arsenic exposure via drinking water. More longitudinal studies are warranted to evaluate the most critical windows of exposure, the type of effects and dose‐response relationships’.

Since then, many studies have been published but most are cross‐sectional and relatively small. Different tests for cognitive, behavioural or motor/sensory function in children have been applied and at different ages, but the majority of studies have evaluated arsenic exposure vs. cognitive function and in the age span 5–11 years. Further studies have evaluated arsenic exposure and the neurodevelopmental outcomes autism, attention deficit hyperactivity disorder (ADHD).

For the current evaluation, the CONTAM Panel identified 59 studies, of which 20 met the criteria, including five (Rosado et al., 2007; Tofail et al., 2009; von Ehrenstein et al., 2007; Wasserman et al., 20042007) that were already reviewed in the 2009 Opinion.

Of the selected studies, 12 were cross‐sectional, one was case–control and seven were cohort studies (i.e. prospective mother–child cohorts). Thirty‐nine further studies were not selected as they did not meet the inclusion criteria (Calderon et al., 2001; Cusick et al., 2018; de Assis Araujo et al., 2022; de Water et al., 2022; do Nascimento et al., 2015; Forns et al., 2014; Freire et al., 2018; Hsueh et al., 2020; Huang et al., 2011; Jiang et al., 20182022; Khan et al., 20122019; Langley et al., 2015; Levin‐Schwartz et al., 2019; Lin et al., 2013; Lucchini et al., 2019; McDermott et al., 2012; Merced‐Nieves et al., 2022; Nahar, Inaoka, & Fujimura, 2014; Nahar, Inaoka, Fujimura, Watanabe, et al., 2014; Nozadi et al., 2021; Nyanza et al., 2021; Parajuli et al., 2013, 2014, 2015; Rahman et al., 2015; Renzetti et al., 2021; Rocha‐Amador et al., 2007; Rodríguez‐Barranco et al., 2016; Saxena et al., 2022; Valeri et al., 2017; Vibol et al., 2015; Wang, Wang, et al., 2007; Wang, Liu, et al., 2018; Wang, Wang, & Yan, 2022; Wasserman et al., 2011; Wright et al., 2006). Table 20 summarises the selected studies from both the 2009 Opinion and the current evaluation.

TABLE 20

Key epidemiological studies on As exposure and neurodevelopment. a

Reference study population designOutcome definitionPopulation size (n), ageArsenic exposureResultsAdditional information/confounders
Prenatal

von Ehrenstein et al. (2007)

India

Prospective mother–child cohort study

Wechsler Intelligence

Scale for Children (no edition provided), Raven Coloured, Progressive Matrices test, Total Sentence Recall test, Purdue pegboard test

351

5–15 years

w‐As (μg/L)

Mean (SD)

Peak lifetime

147 (322)

During pregnancy

110 (243)

Tertiles

< 10

10–49

50–99

> 100

< 10

10–49

50–99

> 100

B (95% CI)

Full scale IQ

Peak lifetime

Ref

0.006 (−0.031, 0.33)

−0.16 (−0.56, 0.23)

−0.06 (−0.30, 0.18)

During pregnancy

Ref

−0.047 (−0.38, 0.28)

−0.007 (−0.36, 0.34)

−0.002 (−0.24, 0.24)

Adjusted for age, sex, BMI, maternal and paternal education, father's occupation, number of rooms in the house, type of house building material, BMI and mother's age

Rodrigues et al. (2016)

Bangladesh

Prospective mother–child cohort study

Bayley Scales of Infant and Toddler Development, Third Edition (BSID‐III)

525

20–40 months

Sirajdikhan (n = 239)

Pabna (n = 286)

w‐As (μg/L) at 20–40 months

Median (IQR)

Sirajdikhan

1.5 (< 0.75, 21.6)

Pabna

25.7 (4.4, 130)

w‐As at 1st trimester pregnancy

Sirajdikhan

1.3 (< LOD, 1.9)

Pabna

26.5 (5.3, 81)

β (SE, p‐value)

As 20–40 months

Cognitive

Sirajdikhan

−0.002 (0.02, 0.93)

Pabna

−0.06 (0.03, 0.05)

Fine motor

Sirajdikhan

−0.05 (0.03, 0.09)

Pabna

0.02 (0.03, 0.48)

As during pregnancy

Cognitive

Sirajdikhan

0.009 (0.03, 0.78)

Pabna

−0.09 (0.03, 0.006)

Fine motor

Sirajdikhan

0.07 (0.04, 0.10)

Pabna

0.007 (0.03, 0.81)

Different exposure, scores and covariates between clinics and thus were results stratified by area. Adjusted for maternal age and education, child's gender, exposure to second‐hand smoke, HOME score, maternal Raven score and child haematocrit.

Vahter et al. (2020)

Bangladesh

Prospective mother–child cohort study, cross‐sectional at 5 and 10 years

Wechsler Intelligence Scale for Children 4th ed. (WISC‐IV)

1523 mother–child pairs

305

305

304

305

304

296

295

296

295

295

294

293

294

293

293

u‐tiAs (μg/L) children 10 years, Median (range)

22.6 (7.3–29.9)

36.7 (30.0–45.5)

57.3 (45.6–73.1)

104 (73.4–162)

265 (163–940)

u‐tiAs children 5 years

21 (6–27)

34 (28–41)

53 (42–69)

100 (70–149)

236 (150–1020)

u‐tiAs mothers week 8

23.8 (3.2–32.4)

43.5 (32.5–58.8)

83.5 (58.9–122)

174 (122–246)

381 (247–1287)

Β (95% CI)

Full developmental score

Ref

−1.63 (−5.62; 2.36)

−7.23 (−11.3, −3.18)

−6.37 (−10.5, −2.22)

−2.71 (−6.96, 1.54)

Ref

−1.53 (−5.63, 2.56)

−3.30 (−7.41, 0.81)

−2.57 (−6.78, 1.65)

0.59 (−3.67, 4.86)

Ref

−4.52 (−8.61, −0.43)

−5.91 (−10.0, −1.77)

−5.98 (−10.2, −1.77)

−3.41 (−7.60, 0.77)

Adjusted for child gender, age at testing and tester, child BMI‐for‐age, type of school, school grade, number of children in the household, family, HOME score, father's level of education, mother's IQ, home stimulation, child, U‐cadmium and W‐manganese

Soler‐Blasco et al. (2022)

Spain

Prospective mother–child cohort study

McCarthy Scales of Children's Abilities

807

4–5 y

u‐tiAs mothers first trimester (μg/g creatinine)

GM mean (95%CI)

7.78 (7.41–8.17)

Β (95% CI)

General scale

u‐tiAs continuous

−0.49 (−1.53, 0.56)

Adjusted for area of study, creatinine, maternal and paternal educational level, parity, child's sex, attendance at nursery, maternal verbal intelligence quotienteason of sample collection and rice and seafood consumption during the first trimester of pregnancy.

Postnatal

Wasserman et al. (2004)

Bangladesh

Cross‐sectional study

Wechsler Intelligence Scale for Children, version III

201

9.5‐, 0.5 years

w‐As (μg/L)

Mean (SD)

117.8 (145.2)

w‐As (μg/L)

Quartiles

0.1–5.5

5.6–50.0

50.1–176

177–790

u‐As (μg/g creatinine)

296.6 (277.2)

B (p‐value)

Full‐Scale score

w‐As continuous

−1.64 (< 0.01)

w‐As quartiles

Ref

Not provided

−7.8 (< 0.05)

−11.3 (< 0.01)

u‐As continuous

−2.9 (0.09)

Adjusted for maternal education, maternal intelligence, house type, television access, height, head circumference

Speciated arsenic in urine but unclear if arsenobetaine and arsenocholine were subtracted from total urinary arsenic

Wasserman et al. (2007)

Bangladesh

Cross‐sectional study

Wechsler Preschool and Primary Scale of Intelligence, version III

301

6 years

w‐As (μg/L)

Mean (SD)

120.1 (134.4)

w‐As (μg/L)

Quartiles

0.1–20.9

21–77.9

78–184.9

185–864

B (SE, p‐value)

Full‐Scale IQ

w‐As continuous

−1.06 (0.57, < 0.07)

w‐As quartiles

Ref

Not provided

Not provided

−5.70 (p = 0.09)

Adjusted for maternal education and intelligence, home stimulation, school attendance, height, head circumference, W‐manganese

Wasserman et al. (2018)

Bangladesh

Cross‐sectional study

Wechsler Intelligence Scale for Children‐Fourth Edition

694

14–16 years

u‐As (μg/g creatinine)

Mean (SD)

157.79 (208.87)

B (SE, p‐value)

Full‐Scale IQ, U‐As continuous

−5.09 (1.38, < 0.001)

Adjusted for paternal education, maternal intelligence, house construction, head circumference, child education, w‐As of the mother at baseline, B‐selenium, B‐cadmium, B‐lead, B‐manganese.

Speciated arsenic in urine but unclear if arsenobetaine and arsenocholine were subtracted from total urinary arsenic

Rosado et al. (2007)

Mexico

Cross‐sectional study

Weschsler Intelligence Scale for Children Revised Mexican Version (WISC‐RM), a test of number and letter sequencing, Cognitive Abilities Test, Math Achievement Test (MAT), Visual–Spatial Abilities with Figure Design, Peabody Picture Vocabulary Test (PPVT‐Spanish Edition)

557

6–8 years

u‐As (μg/L)

Mean (SD)

58.1 (33.2)

Β (95% CI)

Visual–spatial abilities

−0.024

(−0.045, −0.004)

Peabody picture vocabulary

−0.064

(−0.115, −0.013)

WISC‐RM digit span subscale

−0.014

(−0.025, −0.002)

Visual search

−0.007

(−0.011, −0.002)

OR (95% CI)

Letter sequencing

0.992 (0.987, 0.996)

Adjusted for age, sex, mother's school level, haemoglobin, B‐lead and B‐lead × u‐As interaction when significant.

They had further info on child anthropometry but this was not adjusted for

Parvez et al.

(2011)

Bangladesh

Cross‐sectional study

Bruininks‐Oseretsky

Test version 2. 4 subscales that are summarised into total motor composite score (TMC)

304

8–11 years

w‐As (μg/L)

Mean (SD)

43.3 (73.6)

β (95% CI)

TMC

−1.18 (−2.13, −0.10)

Adjustment for sex, school attendance, head circumference, mother's intelligence, B‐manganese, B‐lead and B‐selenium

Hsieh et al. (2014)

Taiwan

Case–control study

Peabody Developmental Motor Scales, Gross Motor Function Measure, Preschool Language Evaluation Tool, Child Expression Evaluation Tool, Chinese Wechsler Intelligence Scale for Children (3rd ed.), Bayley III Scales of Infant and Toddler Development. Developmental delay defined as performance 2 SD below the mean on age‐appropriate, standardised, norm‐referenced tests

63/35 with and without develop‐mental delays; 5 years

7/12

21/11

35/12

u‐As (μg/g creatinine)

Tertiles of the controls

≤ 13.56

13.57–24.71

> 24.71

OR (95% CI)

Developmental delay

1.0

5.95 (0.64, 55.57)

11.83 (1.52, 91.82)

Adjusted for age, gender, birth weight of children, ethnicity of their mothers, gestational length, B‐lead or B‐mercury. Information about mothers' education but not different between delay groups and not included in the adjusted model

Wasserman et al. (2014)

USA

Cross‐sectional study

The Wechsler Intelligence Scale for Children IV ed

272

8–11 years

141

46

52

33

w‐As (μg/L)

Mean (SD)

9.88 (15.06)

Mean (SD)

1.24 (1.37)

7.37 (1.34)

14.80 (3.06)

42.55 (20.43)

B (SE, p‐value)

Full Scale IQ

Ref

−6.09 (1.98, < 0.01)

−3.15 (1.91, 0.05)

−2.51 (2.29, < 0.10)

Adjustment for maternal IQ, maternal education, HOME score, school district, number of siblings

Desai et al. (2018)

Uruguay

Cross‐sectional study

Woodcock‐Muñoz cognitive battery: 7 subtests from which the general intellectual abilities (GIA) score was derived

328

5–8 years

u‐As (μg/L) children

Median (range)

11.9 (1.4–93.9)

Β (95% CI)

GIA score

−0.05 (−1.91, 1.82)

Adjusted for school clusters at the design stage, child's sex, BMI for age, serum ferritin, B‐lead, haemoglobin, season, test administrator, mother's education, crowding at home, HOME score, parental smoking, source of drinking water, household possessions. No consistent effect modification by folate intake or %MMA

Signes‐Pastor et al. (2019)

Spain

Cross‐sectional study

McCarthy Scales of Children's Abilities (MSCA)

400

4–5 years

u‐As (μg/L) Median (IQR) 4.85 (2.74–7.54)

Β (95% CI)

Motor functions

global scores: −2.29 (−3.95, −0.63)

gross scores: −1.92 (−3.52, −0.31)

fine scores: −1.54 (−3.06, −0.03)

Adjusted for maternal education, child sex, BMI, age at testing, calorie adjusted consumption of rice and fish/seafood. In stratified analyses by sex, negative associations were observed with some scores only in boys

Desai, Barg, Vahter, Queirolo, Peregalli, Mañay, Millen, Yu, Browne, and Kordas (2020)

Uruguay

Cross‐sectional study

Woodcock‐Muñoz Cognitive Battery: 6 subtests from which Broad math score and Broad reading score were derived

239

5–8 years

u‐As (μg/L) children

Median (range) 11.7 (2.6, 50.1)

OR (95% CI)

Broad math score

1.01 (0.98, 1.04)

Broad reading score 1.00 (0.98, 1.03)

Adjusted for season, sex, maternal education, household possessions, HOME score, haemoglobin, B‐lead, H‐manganese. No effect modification by %MMA or B‐vitamins

Desai, Barg, Vahter, Queirolo, Peregalli, Mañay, Millen, Yu, and Kordas (2020)

Uruguay

Cross‐sectional study

Tests from Cambridge Neuropsychological Test Automated Battery (CANTAB): Stockings of Cambridge (SOC), Intra‐dimensional/

extra‐dimensional shift task (IED) and Spatial Span (SSP)

255

5–8 years

u‐As (μg/L) children

Median (range)

9.9 (2.2, 47.7)

w‐As (μg/L)

Median (range) 0.45 (0.1, 18.9)

Β (95% CI)

IED Stages completed: −0.02 (−0.03, −0.002)

IED Pre‐executive shift error: −0.07 (−0.13, −0.01)

SSP Span length: −0.02

(−0.02, −0.01)

Adjusted for age, sex, maternal education, possessions score, HOME score, season, school clusters, B‐lead, haemoglobin, U‐cadmium and H‐manganese. No effect modification of B vitamin intake or %MMA

Abbreviations: As, arsenic; b, blood; BMI, body mass index; BSID‐III, Bayley Scales of Infant and Toddler Development, Third Edition; CANTAB, Cambridge Neuropsychological Test Automated Battery; CI, confidence interval; GIA, general intellectual abilities; GM, geometric mean; HOME, Home Observation Measurement of the Environment score; IED, Intradimensional /extradimensional shift task; IQ, intelligence quotient; IQR, interquartile range; LOD, limit of detection; MAT, Math Achievement Test; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; MSCA, McCarthy Scales of Children's Abilities; n nymber; OR, odds ratio; PPVT, Peabody Picture Vocabulary Test; ref, reference; SD, standard deviation; SE, standard error; SOC, Stockings of Cambridge; SSP, spatial span; TMC, Total Motor Composite score; u, urine; u‐As, urinary arsenic; USA, United States of America; w: water; w‐As, water arsenic; Weschsler Intelligence Scale for Children 4th edition; WISC‐RM, Weschsler Intelligence Scale for Children Revised Mexican Version; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA).

a Due to space limitations, not all outcomes reported in the studies are included in the table. For studies using Wechsler scales, the full score is presented; for studies with many tests, only outcomes that were associated with arsenic are presented.

Prenatal arsenic exposure and cognition

In a study from India on iAs and intellectual function in 5–15 year‐old children assessed both arsenic exposure during pregnancy (mean water arsenic concentrations 110 μg/L) and childhood (mean 147 μg/L) (von Ehrenstein et al., 2007). There were no significant associations with either exposure time window when using arsenic in water as the exposure matrix. Associations were found with child urinary As, but it was not speciated for arsenic metabolites.

Rodrigues et al. (2016) evaluated water As concentrations from pregnancy up to 20–40 months in two regions (Sirajdikhan and Pabna) in Bangladesh and neurodevelopment. No associations were found between concurrent water arsenic (median Sirajdikhan 1.5 μg/L and Pabna 25.7 μg/L) and children's scores for receptive and expressive language, or gross motor domains (not in table). Increasing concurrent water arsenic concentrations were associated with decreasing cognitive scores in Pabna but not in Sirajdikhan. Similar and slightly stronger associations were observed between first trimester water arsenic concentrations and cognitive scores in Pabna.

In a large and well‐characterised mother–child cohort in Matlab in rural Bangladesh, arsenic concentrations were evaluated vs. neurodevelopment at different ages: at 7 months (Tofail et al., 2009), 18 months (Hamadani et al., 2010), 5 years of age (Hamadani et al., 2011), and 10 years of age (Vahter et al., 2020, in table). In Vahter et al. (2020) cognitive abilities of children were assessed in relation to arsenic in maternal urine in early pregnancy (gestational week 8), and in child urine at 5 and 10 years, and in hair at 10 years. Both early maternal urine (82 μg/L) and child arsenic exposure (57 μg/L) at 10 years, were at low levels (around 50 μg/L) inversely associated with cognitive abilities measured as full developmental score at 10 years. Associations were found in the 2nd and 3rd quintiles. Models with children's hair arsenic concentrations showed similar results (not in Table 20).

Soler‐Blasco et al. (2022) evaluated maternal urinary arsenic (geometric mean 7.78, 95% CI 7.41, 8.17 μg/g creatinine), in the first trimester and neurodevelopment (McCarthy Scales of Children's Abilities) in Spanish 4–5 years‐old children. They found inverse associations between MMA concentrations and the scores for the general, verbal, quantitative, memory and working memory scales. However, no associations were found between total iAs and developmental scales in multivariate models.

Postnatal exposure and cognition

In studies from Araihazar in Bangladesh, exposure to arsenic from drinking water (mean appr. 120 μg/L across the studies) was evaluated in relation to intellectual function at different ages. In 9–10‐year‐old children, increasing water arsenic was associated with reduced intellectual function (full scale score), in a dose‐dependent manner (Figure 1 in article, though the effect estimate for the second quartile was not provided; Wasserman et al., 2004). In 6‐year‐old children, increasing water arsenic was negatively associated with full scale scores, and the associations were less strong than in the previously studied 9–10‐year‐olds (Wasserman et al., 2007). In 14–16‐year‐old children, urinary arsenic showed negative associations with all Wechsler test scores, but it is unclear whether the urinary arsenic was corrected for organic arsenic and there are no effect estimates for the dose response analysis (Wasserman et al., 2018).

Rosado et al. (2007) found in 6–7‐year‐old Mexican children inverse associations between urinary arsenic (mean 58.1 μg/L) and cognition measured as visual–spatial abilities with Figure Design, the Peabody Picture Vocabulary Test, the WISC‐RM Digit Span subscale, visual search and letter sequencing tests.

In a study of Taiwanese children with and without developmental delay increased risk (with very wide confidence interval) for developmental delay was found in the highest tertile (> 24.71 μg/creatinine) of child urinary arsenic (Hsieh et al., 2014).

In a study on 3–5 grade school children (Wasserman et al., 2014) in the USA, associations between drinking water As (mean 9.88 μg/L) and IQ were examined. Water arsenic was negatively associated with Full Scale IQ. This association was strongest in the exposure category 5–10 μg/L arsenic in water, but no dose–response was found.

Three studies from Uruguay (Desai et al., 2018; Desai, Barg, Vahter, Queirolo, Peregalli, Mañay, Millen, Yu, Browne, & Kordas, 2020; Desai, Barg, Vahter, Queirolo, Peregalli, Mañay, Millen, Yu, & Kordas, 2020) were based on the same study population of 5–8‐year‐old children (median appr. 10 μg/L). Two of the studies did not find associations between arsenic and cognitive functions (Desai et al., 2018; Desai, Barg, Vahter, Queirolo, Peregalli, Mañay, Millen, Yu, Browne, & Kordas, 2020), whereas one found adverse associations with different measures of executive function (Desai, Barg, Vahter, Queirolo, Peregalli, Mañay, Millen, Yu, & Kordas, 2020).

Postnatal exposure motor/ sensory functions and behaviour

Parvez et al. (2011) investigated the relation between arsenic in water (mean 43.3 μg/L) and motor function in 8–11‐year‐old children and found lower total motor composite scores with increasing arsenic concentrations.

Signes‐Pastor, Vioque, et al. (2019) evaluated arsenic exposure measured in urine and neuropsychological development among Spanish children of 4–5 years of age. Urinary As concentrations (median < 10 μg/L) were negatively associated with the scores of global, gross and fine motor function. They found no associations between iAs and cognitive function.

Emotional and behavioural symptoms at the subclinical level raise the risk of subsequent development of mental disorders and can be assessed during pre‐adolescence through internalising and externalising problems indicators. Roy et al. (2011) found in a study of Mexican 6–7‐year‐old children an association between urinary arsenic (median 55.2 μg/L) in the 2nd quartile (range 36–55.2 μg/L) and teachers' ratings of oppositional behaviour, but there was no dose–response.

Khan et al. (2011) evaluated water arsenic concentrations and classroom behaviour via teacher's report of internalising and externalising problems in 8–11‐year‐old children in Bangladesh, and found no association with As and these outcomes.

In summary, epidemiological studies published since the EFSA Opinion 2009 provide sufficient evidence for a causal association between low to moderate exposure to iAs and impaired cognition. The study by Vahter et al. (2020) is the largest and longitudinal study which reports results both for prenatal exposure and postnatal exposure and there may be different BMD depending on the timing of exposure.

There is insufficient evidence for associations between low to moderate exposure to iAs and motor function and behaviour.

Arsenic and autism spectrum disorder

For the present Opinion, the CONTAM Panel identified 27 studies on associations between As exposure and autism spectrum disorder (ASD) from a literature search (see Section 2 on methodology). Out of these, 16 studies did not meet the inclusion criteria and were thus not considered further (Adams et al., 2013; Amadi et al., 2022; Baj et al., 2021; Elsheshtawy et al., 2011; Fiore et al., 2020; Geier et al., 2012; Li, Li, et al., 2018; Majewska et al., 2010; Rahbar et al., 20122021; Rezaei et al., 2022; Skogheim et al., 2021; Vergani et al., 2011; Wang, Hossain, et al., 2019; Yorbik et al., 2009; Zhao et al., 2022). Eleven studies that fulfilled the inclusion criteria are described in detail in Table 21.

TABLE 21

Key epidemiological studies on autism and As exposure.

Reference study population age designOutcome definitionPopulation size (n) case/controlArsenic concentration/exposureResultsAdditional information/confounders

Al‐Ayadhi (2005)

Arab/up to 14 years

Case–control study

Autism spectrum disorder (autism, attention deficit disorder, Rett's syndrome, Asperger's syndrome)77/80Hair As

Cases versus control mean ppm (SEM):

1.8 (0.16) versus 0.23 (0.05), p < 0.05

Age and sex matched controls. Also results for 12 cases in total with attention deficit disorder, Rett's syndrome, Asperger's syndrome

Kern et al. (2007)

USA

1–6 years,

Case–control study

Autism spectrum disorder, Autism severity (Childhood Autism Rating Scale (CARS))45/45Hair As

Cases versus control mean μg/g (SD):

0.06 (0.06) versus 0.09 (0.08), p < 0.05.

No association between As levels and CARS

Age, sex, race/ethnicity matched controls

Blaurock‐Busch et al. (2011)

Arab

3–9 years,

Case–control study

Autism spectrum disorder (ASD), autism severity (Childhood Autism Rating Scale (CARS))25/25Hair AsCases versus control mean μg/g (SD): 0.2 (0.26) versus 0.06 (0.04), p: 0.01Age and sex matched controls. Significant differences regarding the amount of sea food eaten per month by mothers during pregnancy and by infants in the infancy period were noted

De Palma et al. (2012)

Italy/mean 9 years

Case–control study

Autism spectrum disorder, autism severity (Childhood Autism Rating Scale (CARS)44/61Hair As

Cases versus control median μg/g (IQR):

003 (0.01–0.06) versus 0.04 (0.012–0.07), p: NS

Age matched controls

Obrenovich et al. (2011)

USA (authors)/under 6 years, Case–control study

Autism spectrum disorder26/39Hair AsCases versus control p < 0.001Age matched controls. No details for data

Blaurock‐Busch et al. (2012)

Arab

3–9 years, Case–control study

Autism spectrum disorder, autism severity (Childhood Autism Rating Scale [CARS])44/146Hair As

Cases versus control mean μg/g (SD): 2.94 (4.05) versus 0.7

(95th percentile), p: 0.01

No correlation between total or any subscale of CARS and As (r: −0.2).

Age matched controls. Expanded sample compared with Blaurock‐Busch but only hair As measurement

Fido and Al‐Saad (2016)

Arab

4–7 years,

Case–control study

Autism40/40Hair As

Cases versus control median μg/g (IQR):

013 (0.12–018) versus 0.13 (0.11–0.16) p: NS.

Age and sex matched controls

Skalny et al. (2016)

Russia/2–9 years,

Case–control study

Autism spectrum disorder74/74Hair As

Cases versus control median μg/g (IQR):

0.034 (0.021–0.044) versus 0.031 (0.021–0.058), p: NS

Age and sex matched controls. Additional data for selected age groups. Population of Skalny et al. (2016 and 2017) publications may partly overlap

Skalny et al. (2017)

Russia

3–8 years,

Case–control study

Autism spectrum disorder33/66Hair As

Cases versus control median μg/g (IQR):

0.0343 (0.026–0.0434) versus 0.0426 (0.021–0.0862), p: NS

Age and sex matched controls. Additional data for selected age groups and As correlation with age

Zhai et al. (2019)

China mean 5 years, Case–control study

Autism spectrum disorder79/58Hair As

Cases versus control median (μg/g) (IQR):

0.21 (0.16–0.29) versus 0.09 (0.0–0.16), p < 0.001

Age and site matched. Data also for gut microbiota

Fiłon et al. (2020)

Poland 2–8 years, Case–control study

Autism spectrum disorder30/30Hair As

Cases versus control mean μg/g (SD):

0.216 (0.09) versus 0.061 (0.03), p < 0.001

Controls < 8 years randomly selected

Abbreviations: As, arsenic; ASD, autism spectrum disorder; CARS, Childhood Autism Rating Scale; IQR, interquartile range; n, number; NS, not significant; ppm, parts per million; SD, standard deviation; SEM, standard error of the mean; USA, United States of America.

In a case control study with Arab children, children with ASD had higher concentrations of As in hair compared with controls (Blaurock‐Busch et al., 2011). Also, in an expanded population of the previous study, hair As concentrations were significantly higher in children with ASD compared with controls (Blaurock‐Busch et al., 2012). In two further case control studies of Arab children, one study showed higher hair As among children with ASD compared with controls (Al‐Ayadhi, 2005), but no differences were found in the second study (Fido & Al‐Saad, 2016).

In case–control studies performed in Poland, USA and China, children with ASD had higher hair As concentrations compared with controls (Fiłon et al., 2020; Obrenovich et al., 2011; Zhai et al., 2019).

In a case–control study in Russia, hair As was not significantly different in children with ASD compared with controls (Skalny et al., 2017). In another and larger case control study from the same authors (possibly related to the previous study although there is no statement in the manuscript), hair As concentrations were not significantly different in children with ASD compared with controls (Skalny et al., 2016). Also, in a study in Italy, hair As concentrations did not differ between children with ASD and controls (De Palma et al., 2012), whereas in a US study, hair As concentrations were significantly lower children with ASD than in matched controls (Kern et al., 2007).

Overall, six studies found higher hair As levels in ASD cases, while four did not find any significant differences in hair As levels and one found lower levels of hair As in ASD cases compared to controls. All studies are case–control studies with relatively few cases/controls. This is important as the case–control study is not a relevant study design for a disease likely affecting life and family/child habits. All studies used hair As as exposure metric. It should be noted that hair As in some of the studies was surprisingly high, whereas hair As levels in most of the studies where no association was seen with autism as well as other investigations presented in this Opinion are below 0.1 μg/g.

In summary, there is insufficient evidence for an association between low to moderate exposure to iAs and ASD.

3.2.2.5. Effects on male fertility

Associations between iAs and male fertility were not discussed in EFSA CONTAM Opinion (2009). For the current Opinion, the CONTAM Panel identified eight studies from a literature search, of which one study from China, summarised in Table 22, met the inclusion criteria (Wang et al., 2016). The study did observe a significant association between u‐tiAs and unexplained male infertility. However, very wide confidence intervals clearly indicate the uncertainty in the risk estimates.

TABLE 22

Key epidemiological studies on male fertility in humans in relation to iAs exposure.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Wang et al. (2016)

Nanjing Medical University, China

Case–control

Unexplained male infertility

Cases: 101 infertile men with normal semen whose wives were evaluated to have normal reproductive function

Controls: 61 fertile men with normal semen whose wives had successful pregnancy in last 3 months

Urinary concentration (μg/g creatinine), Medians, Cases – Controls

Asi III 4.10–3.97

Asi V 71.59–0.63

AsB 12.03–7.80

MMAV 4.20–2.93

DMAV 28.23–18.30

u‐tiAs 74.87–5.46

Adjusted OR (95% CI)

4th versus 1st quartile

Asi III 0.59 (0.19–1.83)

Asi V 36.51 (8.25–161.7)

AsB 2.23 (0.70–7.12)

MMAV 1.97 (0.57–6.77)

DMAV 1.27 (0.40–4.03)

u‐tiAs 10.17 (2.72–38.11)

Adjusted for age, BMI, drinking and smoking

Abbreviations: As(III), arsenite; As(V), arsenate; AsB, arsenobetaine; BMI, body mass index; CI, confidence interval; DMA(V), dimethylarsinic acid; iAs, inorganic arsenic; MMA(V), monomethylarsonic acid; n, number; OR, odds ratio; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA).

Six additional studies were also evaluated but did not fulfil the inclusion criteria and were accordingly not considered further (Calogero et al., 2021; Oguri et al., 2016; Sengupta et al., 2013; Sukhn et al., 2018; Tian et al., 2021; Xu et al., 2012).

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and effects on male fertility.

3.2.2.6. Neurotoxicity

EFSA CONTAM Panel (2009) concluded that the available epidemiological studies indicated a relationship between high level oral exposures to iAs and sensitive endpoints for peripheral and central neurotoxicity.

Peripheral neuropathy is observed both at acute and chronic high exposure to arsenic. The clinical features of the arsenic‐linked neuropathy are paresthesias, numbness and pain, particularly in the hands and the soles of the feet. In many cases a symmetrical peripheral neuropathy is one of the earliest symptoms of arsenic poisoning.

The CONTAM Panel identified 21 studies, of which 10 studies met the criteria. The relevant studies are summarised in Table 23. In 11 papers, arsenic and neurotoxicity were reported but the studies were not considered further, as they did not meet the inclusion criteria (Guha Mazumder et al., 2010; Ishii et al., 2017, 2018; Khan et al., 2019; Mathew et al., 2010; Guha Mazumder et al., 2010; OʼBryant et al., 2011; Peters et al., 2021; Shiue, 2015; Szabo et al., 2016; Valappil & Mammen, 2019; Yorifuji et al., 2016).

TABLE 23

Key epidemiological studies on As exposure and neurotoxicity among adults.

Reference study population designOutcome definitionPopulation size (n)Arsenic exposureResultsAdditional information/confounders

Ali et al. (2010)

Bangladesh

Cross‐sectional

Plasma cholinesterase (PChE) activity

141

33

108

w‐As (μg/L)

≤ 50

> 50

PChE (U/L) × 104

1.775 ± 0.371

1.365 ± 0.349

No exposure to pesticides during the last month among the study participants. No significant effects of age, sex and BMI on PChE activity

Edwards, Johnson, et al. (2014)

USA

Cross‐sectional

Neuropsychology core battery for cognition, memory, depression

Full sample:

1390

733 Alzheimer's disease, 127 mild cognitive impairment, 530 with normal cognition

w‐As, mean (SD) (μg/L)

3.97 (3.3)

3.9 (3.1)

3.2 (2.6)

4.0 (3.6)

B coefficient (SE, p‐value)

Full sample:

Confrontation naming:

0.21 (0.08, 0.008)

Immediate verbal memory:

−0.23 (0.09, 0.008),

Delayed verbal memory:

−0.34 (0.10, < 0.001)

Immediate visual memory:

−0.23 (0.10, 0.02)

Delayed visual memory:

−0.58 (0.11, < 0.001)

Adjusted for age, gender, education, obesity, hyperlipidaemia, hypertension, diabetes and selenium level GIS‐estimated

Edwards, Hall, et al. (2014)

USA

Cross‐sectional

Attention, processing speed, verbal fluency, visuospatial abilities, immediate memory and delayed memory; executive functioning

Mini Mental State Examination (MMSE) for global cognition

526

≥ 40 years

w‐As, mean (SD) (μg/L)

6.42 (2.99)

B (SD, p‐value)

Immediate memory raw index

0.15 (0.29, 0.60)

Visuospatial raw index −0.39 (0.19, 0.05)

Language raw index −0.48 (0.15, < 0.05)

Attention raw index −0.66 (0.39, 0.09)

Delayed memory index 0.53 (0.28, 0.06)

Total raw index

−0.82 (0.96, 0.39)

Executive function

0.49 (0.13, < 0.05)

MMSE

−0.14 (0.08, 0.07)

Adjusted for age, gender, education, language of administration, selenium level and APOE ε4 presence

Mukherjee et al. (2014)

India

Cross‐sectional

Neurobehavioral symptoms and depression assessed by subjective symptoms questionnaire and Beck's 21‐point depression inventory‐II; plasma levels of neurotransmitters

654 women

312

342

w‐As (μg/L)

≤ 10 (ref.)

11–50

≤ 10 (ref.)

11–50

OR (95% CI)

transient loss of memory

2.02 (1.17–3.12)

burning sensation in extremities

4.16 (1.89–7.27)

tingling or numbness 3.08 (2.10–7.35)

anxiety

1.49 (1.16–3.46)

fatigue

1.92 (1.49–4.57)

reduced sense of taste

2.29 (1.28–4.62)

reduced sense of smell

1.36 (1.12–2.74)

depression

1.37 (1.11–1.97)

plasma epinephrine and norepinephrine 1.6‐times higher (p < 0.05),

plasma serotonin

1.8‐times higher (p < 0.05);

no difference in plasma dopamine (p > 0.05)

No exposure to pesticides during the last month among the study participants. Logistic regression controlling for age, education, cooking years, menstrual length,

adverse reproductive outcome experienced in past 1 year

Liu et al. (2017)

China

Cross‐sectional

Cognitive impairment by Mini‐Mental State Examination

483 (> 40 years)

148

104

85

146

w‐As (μg/L)

< 10

10–50

50–100

> 100

OR (95% CI)

1.0

1.11 (1.01–1.43)

1.32 (1.21–2.75)

4.01 (2.77–11.03)

Multivariable linear regression model: coefficient WAs = −0.73 (p = 0.02)

Crude odds ratio

Adjusting for sex, age, education and BMI

Karim et al. (2019)

Bangladesh

Cross‐sectional

Cognitive impairment by Mini‐Mental State Examination (MMSE), serum BDNF693 (18–60 years)/sBDNF

w‐As (μg/L)

Range 0.03–1798.60

Interquartile range (IQR): 1.75, 214.0

IQR: 2.81, 207.28

β (95%CI) MMSE

−0.021 (−0.026, −0.015)

Change by IQR (95% CI)

−0.113 (−0.161, −0.064)

β (95%CI) sBDNF

−0.082 (−0.105, −0.059)

Change by IQR (95% CI)

−0.437 (−0.609, −0.265)

Multi‐variable linear regression adjusted for age, sex and education. Also, associations with As in hair and nails showed similar results.

Mochizuki et al. (2019)

Myanmar

Cross‐sectional

Peripheral neuropathy via subjective neurological symptoms and objective neurological examinations of sensory

disturbances

1867 (above 5 years; out of which 456 children 5–15 years)

w‐As (μg/L)

< 10

≥ 10

< 50

≥ 50

Frequencies

Subjective ‘feeling of weakness’: As < 10 μg/L

13% versus As ≥ 10 μg/L 17.6%; and ‘chronic

numbness or pain’: 20.4% versus 24.7%.

Objective pain sensation: As < 50 μg/L 4.7% versus ≥ 50 μg/L 9%); Vibration sensation: 3.8% versus 6.5%; two‐point discrimination: 3.6% versus 7.2%.

In children, no association between symptoms or signs and w‐As

Aggregated water concentrations due to different sources depending on season. No adjustments in any analyses

Wang, Huang, et al. (2021)

China

Cross‐sectional

Cognitive function via Chinese Mini‐Mental State Examination

1556

Cognitive impairment (%)

56 (14.4)

76 (19.5)

87 (22.7)

102 (26.2)

Hair As (mg/kg)

Quartiles:

< 0.01–0.10

0.11–0.21

0.22–0.43

0.44–25.1

OR (95% CI)

1.0

1.480 (0.984, 2.228)

1.517 (1.014, 2.268)

1.919 (1.297, 2.840)

Adjusted for age, sex, education level and location

Rahman, Niemann, and Yusuf (2022)

USA

Cross‐sectional

Sleep disturbance based on questionnaire

1611

(> 20 years)

u‐tiAs (μg/L)

Trouble sleeping

5.40

No trouble sleeping

5.65

OR 95% CI for having arsenous acid in urine above the lower limit of detection

1.0 ref

0.72 (0.51–1.00, p‐value = 0.05)

Unclear whether arsenocbetaine is included in the u‐tiAs concentrations

Adjusted for age, serum

Cotinine, ethnicity and depression

Abbreviations: APOE ε4, apolipoprotein E ε4‐allele; As, arsenic; BDNF, Brain‐Derived Neurotrophic Factor; BMI, body mass index; CI, confidence interval; GIS, geographic information system; IQR, interquartile range; MMSE, Mini‐Mental State Examination; n, number; OR, odds ratio; PChE, plasma cholinesterase; ref, reference; sBDNF, serum Brain‐Derived Neurotrophic Factor; SD, standard deviation; SE, standard error; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA); U, enzyme unit; USA, United States of America; w‐As, water‐arsenic.

Mukherjee et al. (2014) found in pre‐menopausal women of eastern India a higher risk of symptoms from the peripheral nervous system (for example burning sensation in extremities, tingling or numbness, reduced sense of taste and reduced sense of smell) in women who had been drinking water contaminated with As concentrations of 11–50 μg/L compared with women drinking water with As < 10 μg/L. Mochizuki et al. (2019) found in a study on adults and children in Myanmar that more subjective symptoms, possibly due to peripheral neuropathy, were reported at arsenic concentrations in drinking water > 10 μg/L compared with at concentrations < 10 μg/L. Objective peripheral nerve disturbances of both small and large fibres occurred at > 50 μg/L. However, no adjustments were performed in this study and the exposure assessment was crude.

Low chronic exposure to arsenic and associations with impairment of the central nervous system have been evaluated. Mukherjee et al. (2014) found a higher prevalence of depression and other neurobehavioral symptoms in the same pre‐menopausal women mentioned above. Rahman, Niemann, and Yusuf (2022) evaluated in the NHANES survey for years 2015–16, associations between arsenic concentrations in urine and sleep disturbance. Compared with individuals with urinary arsenous acid below the lower level of detection, those with urinary arsenous acid at or above the detection limit had lower odds of sleep disturbance. There was no association for any other metabolites (total arsenic metabolites was not evaluated in the statistical analysis). Liu et al. (2017) reported in a study of Chinese men and women above 40 years, higher risk of cognitive impairment with higher arsenic in drinking water and the impairment was observed already at water arsenic concentrations of 11–50 μg/L compared to concentrations < 10 μg/L. Cognitive impairment with higher arsenic concentrations in drinking water was also found in adults from Bangladesh (Karim et al., 2019) exposed to drinking water with arsenic ranging 5.32–167.94 μg/L. Wang, Huang, et al. (2021) showed in a study from China higher risk of cognitive impairment with higher arsenic concentrations in hair, with a dose–response. The exposure assessment is though difficult in this study as the study participants lived in an area with historically high arsenic concentrations in drinking water polluted from a nearby plant that closed in 2011.

In three studies from Texas, arsenic exposure was estimated by a method based on geographic information system using information on arsenic in ground water (Edwards, Hall, et al., 2014; Edwards, Johnson, et al., 2014; Gong et al., 2011). The authors also reported on a comparison between estimated and measured arsenic values from seven wells and found a reasonable agreement (Edwards, Johnson, et al., 2014). They associated the estimated arsenic levels with different outcomes related to neurophysiological functions. However, the exposure assessment was rather crude. In Edwards, Johnson, et al. (2014), participants with Alzheimer's disease and mild cognitive impairment, were compared with participants with normal cognition. In the whole group, arsenic concentrations were positively associated with language abilities, but also associated with poorer verbal memory, immediate and delayed, as well as poorer visual memory, immediate and delayed. No association was found with depression. In Edwards, Hall, et al. (2014), where rural dwelling adults and elders over 40 years were examined with a battery of neurophysiological tests, estimated higher arsenic concentrations in water were negatively associated with language and executive functioning, and weakly with cognition. Gong et al. (2011) also evaluated estimated arsenic concentrations vs. global cognition in the same study area and same age group (it is not clear if these two latter studies represent independent samples) and found that those with estimated arsenic exposure > 10.0 μg/L had lower MMSE scores than those with arsenic exposure ≤ 10.0 μg/L). However, there were no adjustments in these analyses and no effect estimates presented (not in table).

Several studies have analysed the associations between arsenic and levels of different neurotransmitters or cholinesterase. Ali et al. (2010) investigated the association between arsenic exposure (measured in water) and the activity of plasma cholinesterase in Bangladeshi adults. When dividing the study participants in three exposure groups (low < 129 μg/L arsenic in drinking water; medium 130–264 μg/L and high > 265 μg/L), there was a dose–response with the highest plasma cholinesterase activity in the low exposure group followed by the medium and the high exposure groups (all group comparisons were statistically significant, but no exact values were provided for these comparisons). When dividing study participants according to ≤ 50 and > 50 μg/L As in drinking water, also a lower plasma cholinesterase activity was found in the higher arsenic exposure group. Plasma norepinephrine is used as a measure of sympathetic activity. In the already mentioned Mukherjee et al. (2014) study, levels of plasma neurotransmitters epinephrine, norepinephrine and serotonin were higher among premenopausal women exposed to water arsenic concentrations ranging 11–50 μg/L compared with women exposed to concentrations < 10 μg/L. The authors interpreted these findings as a stress response of the nervous system. In Karim et al. (2019), serum levels of the brain‐derived neurotrophic factor (BDNF), a growth and survival factor for nervous cells, was lower with higher arsenic in water. BDNF and the Mini‐Mental State Examination correlated positively, suggesting that arsenic may mediate its neurotoxicity via lower BDNF levels.

Overall, the available epidemiological studies (particularly Mukherjee et al., 2014; and Liu et al., 2017) indicate a relationship between low level of exposure to iAs and sensitive endpoints for both peripheral and central neurotoxicity. Moreover, studies including biomarkers of effect or mediation support the associations between low‐level arsenic and neurotoxicity. Nevertheless, further epidemiological studies are warranted. It should be noted that in many of the studies, study participants may have been exposed since early in life, and the effects observed may be the result of exposure during developmental periods of the central and the peripheral nervous systems. In Table 23 the studies on neurotoxicity in adults are described.

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and neurotoxicity.

3.2.2.7. Effects on the cardiovascular system

In the previous EFSA Opinion, 13 studies on cardiovascular disease were reviewed. All of them were retrieved from a systematic review by Navas‐Acien et al. (2005). Eight of the 13 studies were performed in Taiwan with very high exposure to As in drinking water. Among the other five studies four (Engel & Smith, 1994; Lewis et al., 1999; Ruiz‐Navarro et al., 1998; Varsanyi et al., 1991) did not fulfil our present inclusion criteria. One of them (Zierold et al., 2004) fulfilled the inclusion criteria, but this study has the risk of information bias since heart disease and hypertension were self‐reported, and many of the 1185 respondents may have known if their water‐As was high.

For the present Opinion, the CONTAM Panel identified 87 additional studies from a literature search and from nine systematic reviews (Abhyankar et al., 2012; Bao et al., 2022; Chowdhury et al., 2018; Karachaliou et al., 2021; Moon et al., 20122017; Tsuji et al., 2014; Xu, Mondal, & Polya, 2020; Zhao et al., 2021). Out of these, 53 studies (Afridi et al., 2011; Ameer et al., 2015; Castiello et al., 2020; Cheng et al., 2021; Dastgiri et al., 2010; Desai et al., 2021; Dong et al., 2022; Gong & O'Bryant, 2012; Guha Mazumder et al., 2012; Gunduz et al., 2015; Guo et al., 2007; Hall et al., 2017; Jovanović et al., 2012; Liao et al., 2012; Lin, Hsu, et al., 2018; Lisabeth et al., 2010; Liu et al., 2022; McLeod et al., 2018; Meliker et al., 2007; Merrill et al., 2017; Nong et al., 2016; Nunes et al., 2022; Pichler et al., 2019; Qu et al., 2022; Rahman et al., 1999, 2014; Rahman, Niemann, & Munson‐McGee, 2022c; Shiue, 2014; Sohel et al., 2009; Soria et al., 2021; Stea et al., 2016; Tang et al., 2022; Wang et al., 2002; Wang et al., 2011; Wang, Hao, et al., 2020; Wang, Karvonen‐Gutierrez, et al., 2021; Wang, Wu, et al., 2007; Wei, Yu, et al., 2017; Wen et al., 2019; Wu et al., 2010; Xu et al., 2021; Xu, Liu, et al., 2022; Xu, Polya, et al., 2020; Yao et al., 2021; Yen et al., 2022; Yildiz et al., 2008; Yu et al., 2017; Yuan et al., 2017; Zhang et al., 2013, 2022; Zhong et al., 2019) did not fulfil the inclusion criteria.

The 34 additional studies that fulfilled the inclusion criteria examined ischemic heart disease (including the diagnoses of myocardial infarction and coronary heart disease), stroke, hypertension/blood pressure, atherosclerosis and overall cardiovascular disease.

Ischemic heart disease (IHD)

The 15 studies are summarised in Table 24. Among those based on As in water the studies performed in Inner Mongolia (Wade et al., 2009, 2015), Bangladesh (Chen, Graziano, et al., 2011; Chen, Wu, Liu, et al., 2013; Wu et al., 2015), Colorado (USA), (James et al., 2015) provide support for an association between water As and risk of IHD. The large study in Denmark (Monrad et al., 2017) provides only limited support for an association (in the Aarhus subcohort). Among the studies based on As in urine, the Strong Heart Study in the USA (Kuo et al., 2022; Moon et al., 2013), and the study of US NHANES (Nigra et al., 2021) also support an association with IHD. Analyses based on As content in toenails (Farzan et al., 2015; Wade et al., 2015) show conflicting results. The ecological (Medrano et al., 2010) and semi‐ecological (D'Ippoliti et al., 2015) studies are considered less reliable, as are the studies with a small/unknown number of self‐reported disease cases (Butts et al., 2015; Zierold et al., 2004).

TABLE 24

Key epidemiological studies on As and cardiovascular disease: Ischemic heart disease (including coronary heart disease, myocardial infarction and ‘heart disease’).

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Zierold et al. (2004)

Study in private well households in Wisconsin, USA

Cross‐sectional

Self‐reported ‘Bypass’, ‘heart attack’

Population 1185

Number of cases not presented

w‐As 0–2389 μg/L, median 2 μg/L,16% > 10 μg/L

< 2

2–10

> 10 μg/L

< 2

2–10

> 10 μg/L

ORs in highest stratum:

Bypass surgery:

1.0

1.77 (0.95–3.30)

2.34 (1.12–4.90)

Heart attack:

1.0

1.31 (0.70–2.30)

2.08 (1.10–4.31)

Adjusted for age, sex, smoking, BMI. Methods and results incompletely described

Wade et al. (2009)

study in Ba Men, Inner Mongolia, China

Retrospective cohort

Heart disease mortality

78,251 person‐years follow‐up 1997–2004

161 deaths in heart disease in those exposed since before 1995 (results in this table)

w‐As (μg/L)

0–5

5.1–20

20.1–100

100.1–300

> 300

Heart disease:

Adj. IRR (cases)

1.0 (44)

1.07 (0.6–1.8) (26)

1.22 (0.8–1.8) (72)

1.55 (0.9–2.7) (17)

2.47 (0.5–12) (2)

Adjusted for age, sex, smoking, education, alcohol, farm work.

Cause of death (blinded to w‐As) from proxy interviews and medical records

Medrano et al. (2010)

study in 651 municipalities in Spain

Ecological

CHD mortality

Population 14.4 million

Number of cases:

88,566

19,709

4725

w‐As (μg/L) (median)

< 1 (0.7)

1–10 (3.9)

> 10 (23.3)

RR

1

1.05 (1.01–1.10)

1.02 (0.96, 1.08)

RR derived from reported % increase. p value trend 0.091

Adjusted for sex, age and covariates at municipal or provincial level (income, hospital beds, prevalence of smoking, hypertension, high serum cholesterol, diabetes, overweight/obesity, and low physical activity, dietary factors, water hardness, magnesium, pH and temperature)

Chen, Chiou, Hsu, Hsueh, Wu, & Chen (2010)

Bangladesh

(HEALS)

Cohort study

IHD Mortality

Cases/Pyrs

14/20,064

16/19,109

15/18,699

26/19,380

w‐As (μg/L) (mean)

0.1–12 (3.7)

12.1–62 (35.9)

62.1–148 (102.5)

148.1–864 (265.7)

u‐tiAs (μg/g creatinine) (mean)

6.6–105.9 (68.5)

106–199 (150.6)

199.1–351.8 (264.9)

352–1100 (641.5)

HR

1

1.22 (0.56, 2.65)

1.49 (0.70, 3.19)

1.94 (0.99, 3.84)

HR

1

1.29 (0.66, 2.51)

1.47 (0.72, 3.01)

1.90 (0.91, 3.98)

p trend 0.03 (water)

p trend 0.06 (u‐As)

Adjusted for sex and baseline age, BMI, smoking, education, changes in u‐As over time

u‐tAs was considered a good measure of iAs (AsB and AsC only 3% in a random speciated subsample, and a high correlation between water‐As and u‐t As)

Moon et al. (2013)

USA (SHS)

Cohort study

CHD Incidence

CHD mortality

Cases/Pyrs

202/13,616

206/13,430

197/12,720

241/12,033

Cases/Pyrs

68 13,616

67 13,430

87 12,720

119 12,033

u‐tiAs (μg/g creatinine) (median)

< 5.8 (4.2)

5.8–9.7 (7.5)

9.8–15.7 (12.4)

> 15.7 (21.8)

< 5.8 (4.2)

5.8–9.7 (7.5)

9.8–15.7 (12.4)

> 15.7 (21.8)

HR

1

1.05 (0.86, 1.28)

0.95 (0.77, 1.19)

1.30 (1.04, 1.62)

1

0.99 (0.70, 1.41)

1.18 (0.83, 1.69)

1.71 (1.19, 2.44)

p trend = 0.006 (incidence)

p trend < 0.001 (mortality)

Adjusted for age, sex, education, smoking status, BMI and LDL cholesterol.

As in water the main source in most participants

Chen, Wu, Liu, et al. (2013)

Bangladesh (HEALS)

Case‐cohort

Heart disease (mainly IHD) incidence.

211 cases of heart disease.

Subcohort 1109.

w‐As (μg/L) (n cases)

0.1–25 (61)

25.1–107 (72)

108–864 (75)

Mean total u‐As in subcohort 277 μg/g creatinine

HR

1.0

1.18 (0.75–1.84)

1.54 (1.02–2.31)

Adjusted for sex, age, smoking, BMI, education, hypertension, diabetes.

Associations with u‐iAs and heart disease not reported, but %MMA (median 13%) was positively associated with risk of heart disease.

Same cohort as in Chen et al. (2011a), but more cases.

Wade et al. (2015)

Case control study in Ba Men, Inner Mongolia, China

IHD

Cases/controls

(recruited 2006–2011)

168/137

105/131

11/4

168/305

105/236

11/26

w‐As (μg/L)

< 10

11–39

> 40

t‐As (μg/g) in toenails

0.11–0.28

0.29–1.37

1.38–34.21

OR

1

1.23 (0.78, 1.93)

4.05 (1.10, 14.99)

1

0.67 (0.33, 1.34)

1.91 (0.73, 4.99)

p trend 0.06.

Adj for diet, BMI, occupation, education, smoking, family history of hypertension, diabetes or heart disease

p trend 0.21

D'Ippoliti et al. (2015)

Cohort study, semi‐ecological

IHD mortality

Population 165,609

Deaths

Men:

380

310

567

Women:

304

263

447

w‐As (μg/L (median)

(municipality level)

< 10 (7.4)

10–20 (12.9)

> 20 (29.7)

< 10 (7.4)

10–20 (12.9)

> 20 (29.7)

HR

1

1.42 (1.15, 1.75)

1.70 (1.33, 2.16)

1

1.36 (1.06, 1.74)

1.23 (0.92, 1.65)

p trend < 0.001

Also significant for cumulative dose. HRs only reported by sex.

Adj for age, calendar period, occupation in the ceramic industry. Area level: socioeconomic status,

smoking sales and radon exposure.

Farzan et al. (2015)

Cohort study in New Hampshire, USA

IHD mortality

Population 3939

IHD deaths 154

As (μg/g) in toenails range (median)

0.01–3.26 (0.09)

w‐As (μg/L) range (median) 0–158 (0.29)

HR per 1 unit ln‐transformed toenail‐As: 0.94 (0.74–1.19)

Cohort based on previous case–control study on skin cancer.

Adjusted for smoking, education, skin cancer (and presumably age and sex, though not mentioned).

James et al. (2015)

Case cohort study in Colorado, USA

(SLVDS)

CHD incidence

96 CHD cases, subcohort 533 (74 cases)

Cases/Pyrs

584,806

181,335

16,534

4 98

w‐As (μg/L) estimated lifetime f (median)

1–20 (5.7)

20–30 (25.3)

30–45 (35.1)

45–88 (50.5)

HR

1

1.23 (0.56, 2.18)

2.18 (1.23, 4.02)

3.10 (1.10, 9.11)

Adjusted for LDL cholesterol and family history of CHD.

Full model adjusted also for sex, smoking, BMI, ethnicity, SES, alcohol, other blood lipids, folate and Se showed very similar HRs.

Butts et al. (2015) Cross‐sectional study in Romania‘Pilot study’ of self‐reported heart disease in pregnant women295 women and 6 cases

w‐As (μg/L)

range 0–175,

median 0.4

aOR per 1 unit ln‐transformed w‐As 1.6 (0.81–3.04)Adjusted for age, smoking, education.

Wu et al. (2015)

Case cohort study in Bangladesh (HEALS)

Incidence of CHD

238 cases of CHD

Subcohort 1375

w‐As (μg/L, (mean; number of cases)

0.1–16 (4.3; 69)

17–85 (47; 86)

86–864 (191; 82)

Adjusted HR

1.0

1.30 (0.83–2.01)

1.40 (0.88–2.23)

Same cohort as Chen, Wu, Liu, et al. (2013), but more cases.

Mean u‐As 119 μg/L (259 μg/g).

Monrad et al. (2017)

Cohort study in Denmark Danish prospective cohort Diet, Cancer and Health (DCH) two cities, Copenhagen and Aarhus.

Myocardial infarction

Cohort 53,856

Incident MI cases 2707, 784 in the Aarhus cohort

w‐As (μg/L 20 years mean). Total cohort median 0.7

0.05–0.57 (0.44)

0.57–0.76 (0.58)

0.76–1.93 (1.18)

1.93–25.3 (2.11)

Aarhus cohort median 2.1

0.08–1.83 (1.30)

1.83–2.11 (2.09)

2.11–2.11 (2.11)

2.21–25.3 (2.11)

Adjusted IRRs

1.0

1.23 (1.11–1.37)

0.98 (0.87–1.10)

1.04 (0.93–1.16)

1.0

0.82 (0.67–1.02)

0.83 (0.68–1.02)

1.44 (1.16–1.78)

Adjusted for age, sex, smoking, BMI, waist, alcohol, physical activity, education, diabetes, hypertension, cholesterol, fruit intake, vegetable intake.

Note low w‐As and low contrast.

Nigra et al. (2021)

Cohort study of NHANES participants 2003–2014

Heart disease mortality

4990 with available u‐As and u‐AsB

77 deaths

u‐tAs (μg/L)

< 2.30

2.31–4.00

4.01–6.50

> 6.50

HR

1.0

1.24 (0.58–2.68)

1.44 (0.65–3.21)

1.21 (0.46–3.14)

Similar results for u‐DMA.

Individuals with u‐AsB ≥ 1.2 μg/L excluded. Overall median (IQR) for u‐tAs 4.42 (2.52–7.20) and for u‐DMA 2.71 (1.35–4.42).

Adjusted for age, sex, ethnicity, u‐creatinine, eGFR, education, BMI, cholesterol and serum cotinine.

Kuo et al. (2022)

Cohort study in USA

(SHS)

CVD mortality (484)

3600

484 deaths

u‐tiAs median 11.2, IQR 12.5 μg/g creatinine

HR per IQR of u‐tiAs:

1.28 (1.08–1.52).

Similar HRs for MMA and DMA (per IQR). Larger HRs when MMA% or DMA% were high.

u‐AsB was low (median 0.68, IQR 0.41–1.54 μg/g creatinine).

Adjusted for age, sex, smoking, BMI, WHR, education, alcohol, u‐creatinine, eGFR, LDL, diabetes, hypertension.

Abbreviations: adj, adjusted; aOR, adjusted odds ratio; As, arsenic; AsB, arsenobetaine; AsC, arsenocholine; BMI, body mass index; CHD, coronary heart disease; CVD, cardiovascular disease; DCH, ‘Diet, Cancer and Health’ study; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; eGFR, estimated glomerular filtration rate; f, female; HEALS, Health Effects of Arsenic Longitudinal Study; HR, hazard ratio; iAs, inorganic arsenic; IHD, ischemic heart disease; IQR, interquartile range; IRR, incidence rate ratio; LDL, low‐density lipoprotein; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; MI, myocardial infarction; n, number; NHANES, National Health and Nutrition Examination Survey; OR, odds ratio; pyr(s), person year(s); RR, risk ratio; Se, selenium; SES, socioeconomic status; SHS, Strong Heart Study; SLVDS, San Luis Valley Diabetes Study; USA, United States of America; u‐As, urinary arsenic; u‐AsB, urinary arsenobetaine; u‐DMA, urinary DMA; u‐tAs, urinary total arsenic; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA); w‐As, water‐arsenic; WHR ratio, waist‐to‐hip ratio.

Two studies were performed in the Ba Men region of Inner Mongolia, China, an agricultural region with elevated levels of As in well water. The first study was a retrospective cohort study in a village with about 12,000 inhabitants (Wade et al., 2009). Cause‐specific mortality was examined during 1997 and 2004. As in the primary water source of each household was analysed. Adjusted rate ratios for mortality in heart disease was analysed by water‐As categories. The adjusted incidence rate was significantly increased when water‐As was treated as a continuous variable. For those exposed from the same water source since 1995, the IRR was 1.19 (95% CI 1.05–1.33) per increase of water As by 50 μg/L, based on 161 deaths. IRRs by categories of water As are shown in Table 24.

The next study from Ba Men was a case–control study (Wade et al., 2015). Cases were hospital patients with sign of ischemic heart disease (myocardial infarction, decreased ejection fraction without valvular disease or ECG changes indicating angina pectoris), and controls were patients from the same hospital with conditions unrelated to As. Cases and controls were recruited 2006–2011. The adjusted OR per 10 μg/L increase of water As was 1.19 (1.03–1.38). The adjusted OR per 1 μg/g increase of toenail As was 1.16 (0.98–1.38). ORs by categories of water As and nail‐As are shown in Table 24.

Three studies were performed in Bangladesh as part of the HEALS project (Chen, Graziano, et al., 2011; Chen, Wu, Liu, et al., 2013; Wu et al., 2015). The first one (Chen, Graziano, et al., 2011) was a prospective cohort study of about 12,000 inhabitants in a defined area, recruited in 2000–2002. Mortality was followed up until 2009 (validated verbal autopsy). As in water was measured in the tube wells (about 6000 wells) used by cohort members, and 96% of participants also delivered a spot urine sample of analysis of total As. The adjusted trend for the HR regarding IHD mortality (71 cases), with water As as a continuous variable, was statistically significant (p = 0.029), and for u‐tiAs as a continuous variable the corresponding p‐value was 0.059. HRs by categories of water As and u‐tiAs are shown in Table 24.

In the second paper (Chen, Wu, Liu, et al., 2013), the incidence of CVD (211 cases, mainly IHD) was studied using a case cohort design. The adjusted HR with water As as a continuous variable was statistically significant. HRs by categories of water As are shown in Table 24. There was also a significant association between MMA% in urine and HR for IHD.

In the third study of the HEALS cohort (Wu et al., 2015), the follow‐up time was longer (2012) and the analysis was based on 237 cases of IHD. There was a significant association between water As as a continuous variable and the adjusted HR for CHD. HRs by categories of water As are shown in Table 24.

A case cohort study by James et al. (2015) in Colorado, US included 96 cases of CHD and showed a strong association between the time‐weighted average lifetime As in water and risk of IHD. The HR was significantly increased already at 30–45 μg/L.

Monrad et al. (2017) performed a very large cohort study in two Danish cities (Copenhagen and Aarhus). There were 2707 incident cases of myocardial infarction. The As levels in drinking water were, however, low. In Copenhagen, about 60% of the population had As in water 0.5 μg/L, and none had levels > 2 μg/L. In Aarhus about 80% had As in water 2 μg/L, about 5% higher than that, and the remaining had As in water < 2 μg/L. Therefore, the analysis of the total cohort will to a large extent be a comparison between two cities. Even if the Aarhus cohort includes a larger range of water As levels, the exposure contrast is still limited. The medians in the third and the fourth quartile of water As is the same.

Two studies using u‐iAs as exposure metric are based on the Strong Heart Study among American Indians in the US. The study by Moon et al. (2013) shows significantly increased risk of CHD incidence and mortality in the upper quartile (Table 24). The study by Kuo et al. (2022) found a significant association between water As and the broader outcome of CVD. The u‐iAs were not very high; the median in upper quartile was 22 μg/g creatinine (Moon et al., 2013). According to the authors, As in water was the main source of iAs exposure in two of the three Strong Heart Study areas (Arizona and the Dakotas). This is consistent with the median inorganic fraction of u‐iAs (8%), and the median MMA of 15% and DMA 78%.

The study by Nigra et al. (2021) based on u‐tiAs after exclusion of those with u‐AsB > 1.2 μg/L provides limited support for an association with heart disease mortality, but the study is relatively small.

Most of the longitudinal studies described above are large, and all of them adjusted their risk estimates for the most important potential confounders. There are no obvious sources of selection bias or information bias.

Several of the studies mentioned above (James et al., 2015; Wade et al., 2015; Wu et al., 2015 ) present results for relatively large data sets with strata of exposure to inorganic As, as concentrations in water. These studies were performed in several different countries (China, US, Bangladesh). The only European study was the Danish one, which also had the lowest exposure levels, but had a narrow range of exposure. Other studies, using As in water, were either based on the same study areas but with fewer cases (Chen, Graziano, et al., 2011; Chen, Wu, Liu, et al., 2013; Wade et al., 2009) or considered having a higher risk of bias (Butts et al., 2015; D'Ippoliti et al., 2015; Medrano et al., 2010; Zierold et al., 2004). Next to Wade et al. (2015), James et al. (2015) and Wu et al. (2015), two studies using u‐tiAs (Moon et al., 2013; Nigra et al., 2021) have also been considered having an appropriate design.

The studies on ischemic heart disease meeting the inclusion criteria are described in Table 24.

In summary, the epidemiological studies provide sufficient evidence for an association between low to moderate exposure to iAs and IHD.

Stroke

The eight studies are summarised in Table 25. Among those based on As in water, the HEALS study in Bangladesh (Wu et al., 2015) showed an association with stroke incidence in the highest stratum (water As 86–864, mean 191 μg/L), while in the stratum of 17–85 μg/L the point estimate was positive (HR 1.14) but with a broad confidence interval (0.65–1.98). The study performed in Inner Mongolia (Wade et al., 2009) showed no association with stroke mortality up to 100 μg/L. The large Danish study (Ersbøll et al., 2018) showed an association in the highest quartile in Aarhus, but the dose–response is difficult to interpret since most cases in the upper quartile had the same water As concentration (2.1 μg/L). The ecological study by Medrano et al. (2010) and the study with an unknown number of self‐reported disease cases (Zierold et al., 2004) are considered less informative. The US studies based on u‐As (Tsinovoi et al., 2018) or As in toenails (Farzan et al., 2015) had very few cases. The studies on stroke are described in Table 25.

TABLE 25

Key epidemiological studies on arsenic and cardiovascular disease: Stroke.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Zierold et al. (2004)

Private well households in Wisconsin, USA

Cross‐sectional

Self‐reported stroke

Population 1185

Number of cases not presented

w‐As 0–2389 μg/L, median 2 μg/L,16% > 10 μg/L.

< 2

2–10

> 10 μg/L

OR

1.0

0.94 (0.40–2.14)

1.53 (0.60–4.07)

Adjusted for age, sex, smoking, BMI. Risk of information bias.

Wade et al. (2009)

Retrospective cohort study in Ba Men, Inner Mongolia, China

Stroke mortality78,251 person‐years during follow‐up 1997–2004. 447 deaths in those exposed since before 1995 (results in this table), 118 of these in stroke.

w‐As (μg/L)

0–5

5.1–20

20.1–100

100.1–300

> 300

Adj. IRR (cases)

1.0 (53)

0.47 (0.3–0.8) (16)

0.51 (0.3–0.8) (41)

0.52 (0.3–1.1) (7)

1.02 (0.2–6.7) (1)

Adjsted for age, sex, smoking, education, alcohol, farm work.

Cause of death (blinded to w‐As) from proxy interviews and medical records.

Medrano et al. (2010)

Ecological study of mortality in 651 municipalities in Spain

Stroke mortality

Population 14.4 million

Number of cases:

81,368

18,327

3895

w‐As (μg/L) (median)

< 1 (0.7)

1–10 (3.9)

> 10 (23.3)

RR

1

1.00 (0.96–1.05)

1.02 (0.95, 1.09)

RR derived from reported % increase. p value trend 0.091

Adjusted for sex, age and covariates at municipal or provincial level (income, hospital beds, prevalence of smoking, hypertension, high serum cholesterol, diabetes, overweight/obesity and low physical activity, dietary factors, water hardness, magnesium, pH and temperature)

Chen, Wu, Liu, et al. (2013)

Case‐cohort study in Bangladesh (HEALS)

Stroke incidence

148 cases of stroke

Subcohort 1109

w‐As (μg/L) (n cases)

0.1–25 (50)

25.1–107 (46)

108–864 (52)

Mean total u‐As in subcohort 277 μg/g creatinine

aHR

1.0

0.86 (0.49–1.51)

1.38 (0.84–2.27)

Adjusted for sex, age, smoking, BMI, education, hypertension and diabetes.

Associations with u‐iAs and stroke not reported. %MMA (median 13%) was not associated with risk of stroke. Same cohort as in Chen et al. (2011a), but more cases

Farzan et al. (2015)

Cohort study in New Hampshire, USA

Stroke mortality

Population 3939

Stroke deaths 43

As (μg/g) in toenails range (median)

0.01–3.26 (0.09)

w‐As (μg/L) range (median) 0–158 (0.29)

HR per 1 unit ln‐transformed toenail‐As: 0.90 (0.61–1.33)Cohort based on previous case–control study on skin cancer. Adjusted for smoking, education, skin cancer (and presumably age and sex, though not mentioned)

Wu et al. (2015)

Case cohort study in Bangladesh (HEALS)

Incidence of stroke

165 cases of stroke

Subcohort 1375

w‐As (μg/L, mean; number of cases)

0.1–16 (4.3; 44)

17–85 (47; 50)

86–864 (191; 71)

Adjusted HR

1.0

1.14 (0.65–1.98)

1.87 (1.06–3.29)

Same cohort as Chen, Wu, Liu, et al. (2013), but more cases.

Mean u‐As 119 μg/L (259 μg/g)

Tsinovoi et al. (2018)

Case cohort study in US (REGARDS)

Incidence of ischemic strokeWith u‐iAs: Population 199, Cases: 41. Total data set 671 cases and subcohort 2486

u‐iAs (μg/g creatinine)

0.1–3.52

3.53–66.8

Adjusted HR

1.0

2.18 (0.64–7.43)

Adjusted for analyses performed versus u‐tAs based on the whole data set showed no association

Ersbøll et al. (2018)

Cohort study in Denmark (Danish prospective cohort Diet, Cancer and Health (DCH), two cities, Copenhagen and Aarhus.

Stroke incidence

Cohort 53,941

Incident stroke cases 2195 (1801 ischemic and 381 haemorrhage, 13 other), 678 in the Aarhus cohort

w‐As (μg/L 20 years mean). Total cohort median 0.7

0.05–0.57 (0.44)

0.57–0.76 (0.58)

0.76–1.93 (1.18)

1.93–25.3 (2.11)

Aarhus cohort median 2.1

0.08–1.83 (1.30)

1.83–2.11 (2.09)

2.11–2.11 (2.11)

2.21–25.3 (2.11)

Adjusted IRRs

1.0

1.21 (1.07–1.36)

1.05 (0.92–1.19)

1.17 (1.04–1.32)

1.0

0.81 (0.62–1.04)

1.09 (0.88–1.34)

1.79 (1.41–2.26)

Adjusted for age, sex, smoking, BMI, waist, alcohol, physical activity, education, fruit intake, vegetable intake and calendar year.

Note low w‐As and low contrast.

Results for ischemic and haemorrhagic stroke relatively similar

Abbreviations: adj, adjusted; aHR, adjusted hazard ratio; BMI, body mass index; DCH, ‘Diet, Cancer and Health’ study; HEALS, Health Effects of Arsenic Longitudinal Study; HR, hazard ratio; IRR, incidence risk ratio; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; n, number; OR, odds ratio; REGARDS, Reasons for Geographic and Racial Differences in Stroke; RR, risk ratio; u‐As, urinary arsenic; u‐iAs, urinary inorganic arsenic; USA, United States of America; u‐tAs, urinary total arsenic; w‐As, water‐arsenic.

In summary, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and stroke.

Atherosclerosis

The eight studies are summarised in Table 26. Among those based on As in water, the Taiwanese studies (the largest one by Hsieh et al., 2011) showed clear associations with carotid artery atherosclerosis (increased intima‐media thickness (IMT and/or plaque). The study in Bangladesh (Chen, Wu, Graziano, et al., 2013) also provides some support, but the association with intima‐media thickness (IMT) was not quite significant on the 5% level.

TABLE 26

Key epidemiological studies on As and cardiovascular disease: Atherosclerosis.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Wu et al. (2006)

Case–control study nested in a Taiwanese cohort

Carotid artery atherosclerosis based on IMT and/or plaque163 cases and 163 controls

w‐As (μg/L)

≤ 50 (25)

50–100 (46)

≥ 100 (89)

OR

1

1.9 (0.9–3.8)

2.6 (1.3–5.0)

Adjusted for age, sex, smoking, cholesterol, hypertension and p‐homocysteine. Median MMA% about 13%, median DMA% about 77%

Hsieh et al. (2008)

Case–control study nested in a Taiwanese cohort

Carotid artery atherosclerosis based on IMT and/or plaque235 cases and 244 controls

w‐As (μg/L) (cases)

≤ 10 (31)

10.1–50 (38)

≥ 50 (175)

OR

1

1.8 (1.0–3.2)

1.9 (1.1–3.1)

Adjusted for age, sex, smoking, diabetes, blood lipids. Same base cohort as Wu et al. (2006)

Hsieh et al. (2011)

Case–control study nested in a Taiwanese cohort

Carotid artery atherosclerosis based on IMT and/or plaque384 cases and 479 controls

w‐As (μg/L) (cases)

≤ 10 (24)

10.1–50 (31)

≥ 50 (325)

OR

1

1.5 (0.7–3.5)

2.0 (1.1–3.9)

Adjusted for age, sex, smoking, blood glucose, hypertension, blood lipids, alcohol, BMI. Same base cohort as Wu et al. (2006) and Hsieh et al. (2008)

Chen, Wu, Graziano, et al. (2013)

Cross‐sectional study in Bangladesh (HEALS)

Carotid artery intima media thickness959

w‐As (μg/L): mean = 41, p10 – p90 = 1–225

u‐As (μg/g creatinine): mean = 171, p10 – p90 = 60–439

w‐As: Beta 5.1

(−0.2–10.3) per 1 SD (102 μg/L)

u‐As: Beta 11.7 (1.8–22) per 1 SD (358 μg/g creatinine)

Adjusted for sex, age, smoking, BMI, education, diabetes and blood pressure.

Significant association with %MMA (median 13%), but not %DMA (median 72%)

Newman et al. (2016)

Cohort study in USA (SHS)

Incident peripheral artery disease based on ABI < 0.9 or > 1.4

Population: 2891

Cases < 0.9280

Cases > 1.4206

u‐tiAs (μg/g creatinine)

(tertiles)

≤ 7.07

7.07–13.3

> 13.3

≤ 7.07

7.07–13.3

> 13.3

MMA% 14

DMA% 78

HR

ABI < 0.9

1.0

1.10 (0.66–1.72)

0.59 (0.33–1.04)

ABI > 1.4

1.0

1.80 (0.93–3.35)

2.40 (1.11–4.95)

Adjusted for sex, age, smoking, education,

BMI, LDL, diabetes, hypertension, estimated glomerular filtration rate and study centre.

Note that ABI < 0.9 is a more accepted measure of atherosclerotic peripheral artery disease than ABI > 1.4

Mateen et al. (2017)

Cohort study in USA (SHS)

Carotid artery atherosclerosis based on IMT and/or plaque

Population 2402

1550 with plaque

u‐tiAs (μg/g creatinine)

≤ 5.64

5.65–9.24

9.25–14.75

14.75–123

≤ 5.64

5.65–9.24

9.25–14.75

14.75–123

RR of plaques

1.0

1.05 (0.97–1.13)

1.04 (0.96–1.13)

1.03 (0.95–1.12)

Mean diff. in IMT

0.00

0.01 (−0.01–0.02)

0.01 (0.00–0.03)

0.02 (0.00–0.04)

Adjusted for sex, age, smoking, education, BMI, LDL, diabetes, hypertension, estimated glomerular filtration rate

Similar tendency for plaque score (no. of segments with plaque)

Sobel et al. (2020)

Cross‐sectional study in USA (MESA)

Carotid artery IMT

Coronary artery calcium (CAC)

ABI

u‐iAs measured in 246 out of total population of 5050 (who had data on self‐reported rice consumption)Overall u‐tiAs (μg/g creatinine) 3.08, IQR 1.96–4.69)

Beta per IQR of u‐tiAs (2.7 μg/g creatinine) for IMT: (CCA) 1.00 (0.96–1.03)

OR for CAC > P75: 1.27 (0.59–2.74).

OR for ABI < 1.0: 1.01 (0.46–2.20)

Adjusted for age, sex, smoking, BMI, exercise, education, eGFR, diabetes, hypertension, high blood lipids, energy intake, site.

The main iAs ‘exposure’ metric was rice intake. There was no association between rice intake and plaque, coronary calcium or ABI, but a positive association between rice intake and u‐tiAs

Grau‐Perez et al. (2022)

Cross‐sectional study in Aragon, Spain (AWHS)

Carotid artery plaque and IMT (CCA, ICA, ECA, bulb)

Carotid artery plaque score (number of segments with plaque)

Femoral artery plaque and IMT

CAC

Population 1873.

Carotid artery plaque 659

Femoral artery plaque 987

CAC positive (> 1) 691

u‐tiAs (μg/g creatinine) median 1.83, IQR 1.25–2.72, P20 1.15 and P80 2.98.

Means: AsB 105, DMA 13.1, MMA 1.38, AsIII0.51, AsV 0.05 μg/L. u‐tiAs was ‘corrected’ possible DMA from seafood by a residual‐based method.

u‐tiAs tertiles:

< 1.42:

1.42–2.35

> 2.35

< 1.42:

1.42–2.35

> 2.35

OR for P80 versus P20:

Carotid artery 1.24 (1.05–1.47)

Femoral artery 1.10 (0.92–1.30)

CAC positive 1.07 (0.91–1.27)

CAC ≥ 10 1.11 (0.92–1.33)

CAC ≥ 100 1.14 (0.88–1.49)

OR for carotid artery plaque score

1.0

1.01 (0.96–1.07)

1.09 (1–02 – 1.16)

OR for CAC score

1.0

0.99 (0.80–1.22)

1.12 (0.90–1.38)

Adjusted for age, sex, smoking, education, eGFR, diabetes, hypertension, high cholesterol

Abbreviations: ABI, Ankle‐Barchial Index; AsB, arsenobetaine; As, arsenic; AsV, arsenate; AWHS, Aragon Workers' Health Study; BMI, body mass index; CAC, coronary artery calcium; CCA, common carotid artery; diff, difference; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; ECA, external carotid artery; eGFR, estimated glomerular filtration rate; HEALS, Health Effects of Arsenic Longitudinal Study; HR, hazard ratio; iAs, inorganic arsenic; ICA, internal carotid artery; IMT, intima‐media thickness; IQR, interquartile range; LDL, low‐density lipoprotein; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; MESA, Multi‐Ethnic Study of Atherosclerosis; n, number; OR, odds ratio; RR, risk ratio; SD, standard deviation; SHS, Strong Heart Study; u‐As, urinary arsenic; u‐iAs, urinary inorganic arsenic; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA); USA, United States of America; w‐As, water‐arsenic.

Among the studies based on As in urine the study by Chen, Wu, Graziano, et al. (2013) found a significant association with IMT, and the study in Spain (Grau‐Perez et al., 2022) found a significant association with IMT and plaque, while in the US SHS study (Mateen et al., 2017) and the US MESA study with very low u‐iAs (Sobel et al., 2020) there was no association with IMT or plaque. The study by Grau‐Perez had very low levels of u‐tiAs but high u‐AsB. Although the authors ‘corrected’ for the potential contribution of DMA from seafood to u‐tiAs, the impact of iAs exposure on the carotid artery findings remain uncertain.

The data on atherosclerosis in the lower limbs (ABI) and the coronary arteries (CAC score) are limited.

In summary, the epidemiological studies based on As in water provide sufficient evidence for a causal association between moderate exposure to iAs and carotid artery atherosclerosis. For other arteries, the evidence is insufficient.

Blood pressure and hypertension

The 11 studies are summarised in Table 27. Eight of them (Chen et al., 2007; Hossain et al., 2017; Islam, Khan, Attia, et al., 2012; Li, Li, Xi, Zheng, Lv, & Sun, 2013; Li, Li, Xi, Zheng, Wang, & Sun, 2013; Mendez et al., 2016; Mordukhovich et al., 2012; Zierold et al., 2004) were cross‐sectional. Two studies (Jiang et al., 2015; Spratlen et al., 2018) had a longitudinal design, and one (Kaufman et al., 2021) presented both cross‐sectional and longitudinal analyses. In most of the studies, the outcome was hypertension, but the longitudinal study by Jiang et al. (2015) and the cross‐sectional studies by Mordukhovich et al. (2012) and Li, Li, Xi, Zheng, Wang, and Sun (2013), examined associations between iAs exposure and blood pressure.

TABLE 27

Key epidemiological studies on As and cardiovascular disease: Blood pressure/hypertension.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Zierold et al. (2004)

Private well households in Wisconsin, USA

Cross‐sectional

Self‐reported ‘high blood pressure’

Population 1185

Number of cases not presented

w‐As 0–2389 μg/L, median 2 μg/L,16% > 10 μg/L.

Results per 3 categories:

< 2, 2–10, > 10 μg/L

ORs in highest stratum:

Hypertension: 1.7 (1.3–2.5)

Adjusted for age, sex, smoking, BMI. Risk of information bias

Chen et al. (2007)

Cross‐sectional study in Bangladesh (HEALS)

HypertensionPopulation 10,910. Mean age about 35 years. Cases 1360 based on SBP and DBP

w‐As quintiles TWA (μg/L) (mean)

0.1–8.0 (2.8)

8.1–40.8 (23.2)

40.9–91 (63.9)

91.1–176 (128)

176.1–864 (283)

OR (cases)

1.0 (289)

1.10 (0.90–1.33) (274)

1.03 (0.85–1.25) (273)

1.01 (0.83–1.22) (259)

1.02 (0.84–1.23) (265)

Adjusted for age, sex, BMI, smoking, education, water consumption. Those taking antihypertensive medications were excluded

Islam, Khan, Attia, et al. (2012)

Cross‐sectional study in rural Bangladesh

Hypertension

Population: 994

Mean age: 45 years

Cases: 66

w‐As (μg/L) (mean)

10–22 (16.5)

23–32 (28.5)

33–261 (141)

≥ 262 (459)

OR (Cases)

1 (22)

1.3 (0.7–2.6) (19)

1.1 (0.5–2.4) (13)

1.0 (0.4–2.2) (12)

Adjusted for age, sex, education, marital status, religion, income and BMI. Smoking was not a confounder. 12 cases were on antihypertensive medication. Pulse pressure was positively associated with w‐As

Guha Mazumder et al. (2012)

Cross‐sectional and case–control study in rural India (West Bengal)

HypertensionVillage with elevated w‐As n = 208, 66 cases (29%). Village with normal w‐As n = 100, 20 cases (20%). Median age about 40 yearsw‐As (μg/L): mean in ‘exposed’ village 50 (range < DL – 326). Total u‐As and hair‐As also measuredAdjusted prevalence OR in ‘exposed’ village 2.9 (1.3–4.8)

Adjusted for age, sex, BMI.

Methods unclear. Within ‘exposed’ village probably no association with cumulative As intake or u‐As but positive association with hair As

Mordukhovich et al. (2012)

Cross‐sectional study of blood pressure in the Normative Ageing Study, USA

Blood pressure639 men with toenail‐As, BP and covariate data collected 1999–2009. Mean age 72 years. 60% on antihypertensive medicationMedian nail‐As 0.08 μg/g, IQR 0.06Beta 0.93 (0.25–1.62) for SBP and 0.17 (−0.20–0.55) for DBP

Adjusted for age, smoking, season, year, education, race and alcohol.

Somewhat higher betas when adjusted for other metals in nail

Li, Li, Xi, Zheng, Wang, and Sun (2013)

Cross‐sectional study in Shanxi, China

Blood pressure and hypertension

Population: 604 Mean age: 46 years

HT cases: 168

u‐tiAs 136 μg/g creatinine, and 91% was iAs, MMA or DMA (69%)

< 94

94–251

> 251

HT aOR for t‐As:

1.0

1.1 (0.6–1.8)

1.6 (1.0–2.7)

Adjusted for sex, age, smoking, BMI and alcohol.

Significant association also with MMA. Significant association between u‐tAs and SBP (partial correlation adjusted for variables mentioned above) but not DBP

Li, Li, Xi, Zheng, Lv, and Sun (2013)

Cross‐sectional study in Inner Mongolia, China

Hypertension

Population: 669 Mean age: 50 years

HT cases: 182

w‐As (μg/L) cases/n

< 10 (24/124)

10–50 (48/207)

> 50 (110/338)

u‐iAs measured but not reported

Hypertension aOR

1.0

1.42 (0.77–2.62)

1.94 (1.02–3.69)

Adjusted for age, gender, smoking, BMI, diabetes, alcohol and Cumulative As Exposure (!). This adjustment should cause underestimation of true OR.

The aOR tended to be higher at high MMA%

Jiang et al. (2015)

Cohort study in Bangladesh (HEALS)

Longitudinal change of blood pressure over 7 years of follow‐up.10,853. Mean age at baseline 37 years. Only 1% on antihypertensive medication

w‐As (μg/L), baseline quartiles

< 12

12–62

62–148

> 148

< 12

12–62

62–148

> 148

Change/year of SBP

Ref

0.43 (0.29–0.56)

0.54 (0.40–0.67)

0.48 (0.35–0.61)

Change/year of DBP

Ref

0.41 (0.31–0.50)

0.41 (0.32–0.51)

0.39 (0.30–0.49)

Adjusted for age, sex, smoking, BMI, education, diabetes, change of U‐iAs.

Similar betas for U‐iAs quartiles (mean u‐tAs in μg/g creatinine):

Q1: 100

Q2: 209

Q3: 317

Q4: 526

Mendez et al. (2016)

Cross‐sectional study in Mexico

Hypertension1160. Age ≥ 18, mean 45 years. 28% on antihypertensive medication. 439 cases of HT

w‐As (μg/L) quartiles (HT cases)

< 25.5 (106)

25.5–47.8 (106)

47.9–78.9 (109)

≥ 79 (118)

Adjusted OR

1.0

1.30 (0.84–2.00)

1.27 (0.82–1.94)

1.41 (0.91–2.17)

Adjusted for age, sex, smoking, alcohol, body weight, high waist circumference, education, recent seafood intake. No association was found with u‐iAs

Hossain et al. (2017)

Cross‐sectional study in rural Bangladesh

HypertensionPopulation 236 from one area with elevated w‐As and one area with normal w‐As. Mean age 36 years. 32 cases of HT

Water: GM 18 μg/L.

Nail: 3.4 μg/g

Hair: 1.3 μg/g

Adj. beta for log‐transformed w‐As vs. SBP: 1.10 (0.26–1.94).

Versus DBP: 0.74 (0.19–1.29)

Probably adjusted for age, sex and smoking, but not clearly described. The focus of the study was the association between w‐As and DNA methylation

Spratlen et al. (2018)

Cohort study in the USA (SHS)

Incident hypertension over 5 years of follow‐up

Population 877. Age at baseline 31 years.

Number of incident cases not reported

u‐tiAs median 6.5, IQR 4.5–10.8 μg/LRR per IQR (6.3 μg/L): 1.03 (0.88–1.20)Adjusted for u‐creatinine, age, sex, region, smoking, alcohol, education, eGFR, BMI. Low u‐AsB: median 0.51, IQR 0.34–1.00

Kaufman et al. (2021)

Cross‐sectional and cohort study in the USA (SHFS)

Hypertension

Cross‐sectional: Population 1910, median age 35 years, 430 cases.

Longitudinal: Population 1453, median age 31, median follow‐up 5 years, 203 incident cases

u‐tiAs (μg/L) quartiles

< 1.34

1.34–1.85

1.86–2.39

≥ 2.40

< 1.34

1.34–1.85

1.86–2.39

≥ 2.40

Adj. prevalence

1.0

1.01 (0.81–1.27)

1.04 (0.82–1.32)

1.19 (1.04–1.82)

Adj RR (incidence)

1.0

1.43 (0.99–2.06)

1.10 (0.75–1.61)

1.10 (0.69–1.74)

Adjusted for age, sex, region, smoking, alcohol, education, eGFR, diabetes, physical activity, dietary factors, u‐creatinine and u‐AsB.

Some overlap between SHS (Spratlen et al., 2018) and SHFS (present study).

Changes in SBP and DBP not significantly associated with u‐iAs

Abbreviations: Adj, adjusted; aOR, adjusted odds ratio; As, arsenic; BMI, body mass index; BP, blood pressure; DBP, diastolic blood pressure; DL, detection level; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; DNA, deoxyribonucleic acid; eGFR, estimated glomerular filtration rate; GM, geometric mean; HEALS, Health Effects of Arsenic Longitudinal Study; HT, hypertension; iAs, inorganic arsenic; IQR, interquartile range; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; n, number; OR, odds ratio; Q, quantile; ref, reference; RR, risk ratio; SBP, systolic blood pressure; SHFS, Strong Heart Family Study; SHS, Strong Heart Study; t‐As, total arsenic; TWA, time‐weighted average; u‐As, urinary arsenic; u‐AsB, urinary arsenobetaine; u‐iAs, urinary inorganic arsenic, USA, United States of America; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA); w‐As, water‐arsenic.

The study by Zierold et al. (2004) which had high risk of information bias, and the study by Hossain et al. (2017) which had few cases were considered less informative.

The longitudinal study of blood pressure by Jiang et al. (2015) provides relatively strong support for the hypothesis that iAs exposure increases blood pressure. Blood pressure was measured with appropriate methods at baseline and on several follow‐up examinations in > 10,000 individuals. Systolic and diastolic blood pressure increased more in individuals with baseline water As in quartiles 2–4 than in the referent quartile (As < 12 μg/L), but there was no monotonic exposure‐response relationship (Table 27). The finding is supported by the smaller cross‐sectional study by Li, Li, Xi, Zheng, Wang, & Sun (2013), which found significant partial correlation coefficients between u‐tiAs (and iAs, MMA, DMA) and SBP, and the study of nail As (Mordukhovich et al., 2012), but not by the study by Kaufman et al. (2021) which found no association between baseline u‐tiAs and change of systolic blood pressure (SBP) or diastolic blood pressure (DBP).

An association between iAs exposure and increased blood pressure is biologically plausible. Vascular toxicity at As exposure is well‐known (causing the so‐called Blackfoot disease). In vitro work has shown that arsenic promotes inflammatory activity, oxidative stress and endothelial dysfunction through several mechanisms, including the activation of stress‐response transcription factors such as activator protein‐1 and nuclear factor‐κB (Abhyankar et al., 2012).

Although there is considerable support for an association, it is based on relatively few studies, one study (Kaufman et al., 2021) showed no association, and the study with the highest quality (Jiang et al., 2015) showed no monotonic relationship. Therefore, the epidemiological studies provide insufficient evidence of a causal association between iAs exposure and blood pressure.

Hypertension

Among the studies of hypertension, two studies from Bangladesh, a very large one by Chen et al. (2007) and a small study by Islam, Khan, Attia, et al. (2012) found no associations with As in water. Two studies from China in areas with known As contamination of drinking water found clear associations between prevalence of hypertension and As in urine (Li, Li, Xi, Zheng, Wang, & Sun, 2013) and water (2013b). A study in Mexico (Mendez et al., 2016) found a somewhat elevated odds ratio for hypertension in the upper quartile of water As, but it was not statistically significant. Spratlen et al. (2018) found no increased incidence of hypertension versus u‐tiAs in a follow‐up of the US Strong Heart Study. Kaufman et al. (2021) analysed an extension of the same cohort (Strong Heart Family Study) and found a significant association between u‐tiAs and prevalent hypertension at baseline, but no increased risk of incident hypertension in the longitudinal analysis. This study had, however, very low u‐iAs concentrations.

The studies of hypertension adjusted for important confounders. The study by Chen et al. (2007) excluded hypertension cases on medication, but such medication was rare (Jiang et al., 2015). The study by Li, Li, Xi, Zheng, Lv, and Sun (2013) adjusted (for unknown reasons) the analysis of hypertension risk vs. As in water by cumulative (years of exposure x As concentration) which likely attenuated the association. The findings in the cross‐sectional studies of hypertension were not consistent. Longitudinal studies are stronger when assessing causality. The longitudinal analyses of incident hypertension in the US SHS and SFHS (Kaufman et al., 2021; Spratlen et al., 2018) showed no association with u‐tiAs.

In summary, the epidemiological studies provide insufficient evidence of an association between iAs exposure and hypertension.

The studies on blood pressure and hypertension are described in Table 27.

3.2.2.8. Respiratory disease

In the previous EFSA Opinion (EFSA CONTAM Panel, 2009) studies on respiratory disease and arsenic exposure were not identified. For the present opinion, the CONTAM Panel identified 26 studies from a literature search. Out of these, eight studies did not meet the inclusion criteria (Bhattacharyya et al., 2014; Liao et al., 2011; Majumdar & Guha Mazumder, 2012; Sanchez et al., 2018; Scannell Bryan et al., 2021; Shih et al., 2019; Smith et al., 2011; Zhou, Wang, et al., 2022). The 19 studies that fulfilled the inclusion criteria (see Section 3.2.2.1) criteria were further considered (Table 28).

TABLE 28

Key epidemiological studies on respiratory diseases and As exposure.

Reference, study population age, designOutcome definitionPopulation size (n) case/controlArsenic concentration/exposureResultsAdditional information/confounders
Parvez et al. (2010), Bangladesh/mean 39 years, cohort (HEALS)Respiratory symptoms (chronic cough, breathing problems, blood in sputum) at follow‐up

11,746

1874 with at least one symptom

w‐As level (μg/L)

Q1: < 7,

Q2: 7–40

Q3: 40–90

Q4: 90–178

Q5: > 178

u‐As (μg/g creatinine):

Q1 < 90,

Q2: 90–160

Q3: 160–246

Q4: 246–406

Q5 > 406

w‐As HR (95% CI):

1.0 (ref),

1.27 (1.09 to 1.48)

1.39 (1.19 to 1.63)

1.43 (1.23 to 1.68)

1.43 (1.22 to 1.68)

u‐As:

1 (ref),

1.10 (0.94 to 1.27)

1.11 (0.95 to 1.29)

1.29 (1.11 to 1.49)

1.35 (1.16 to 1.56)

Adjusted for age, gender, smoking, BMI, education and As‐related skin lesions. The association with w‐As was present also in never‐smokers. Data also available for each of the three questions. High correlation btw w‐As and u‐tAs, very low correlation btw u‐tAs and u‐AsB
Amster et al. (2011), US > 20 years, cross‐sectional (from NHANES 2003–2006)Self‐reported physician diagnosis of asthma, chronic bronchitis and emphysema2676 (asthma: 334, chronic bronchitis: 50, emphysema: 173)u‐tiAs estimated by subtracting organic As from u‐tAs. Median: 5.8 μg/L. u‐tiAs (mean μg/L case/control) Asthma: 8.46/9.36, Chronic bronchitis: 7.74/9.37, Emphysema: 6.51/9.30

OR with u‐tiAs as a continuous variable:

Asthma: 0.92 (0.83, 1.02),

Chronic bronchitis: 1.10 (0.98, 1.19),

Emphysema: 0.93 (0.84, 1.05).

OR comparing P80 with P20:

Asthma: 0.79 (0.43, 1.46),

Chronic bronchitis: 0.76 (0.36, 1.61),

Emphysema: 0.72 (0.13, 3.81)

Adjusted for gender, age, race/ethnicity, education, BMI, serum cotinine and urinary creatinine
Nafees et al. (2011), Pakistan/> 15 years, cross‐sectionalSpirometry and respiratory symptomsAs exposed versus non‐exposed 100/100As exposed > 100 μg/L ‘non‐exposed’ < 10 μg/L in drinking water

Decline in mean spirometry (mL, 95% CI)

In ‘exposed’:

FEV1: −154.3 (−324.7, 16.0; p = 0.076),

FVC: −221.9 (−419.5, −24.3; p = 0.028),

FEV1/ FVC: 2.0 (−25.3, 29.4; p = 0.884).

No significant association between respiratory symptoms and w‐As

Adjusted for age, sex, height and smoking. Analysis include also data for 60 participants with higher w‐As (> 250 μg/L). Levels in the other 40 ‘exposed’ not reported
Pesola et al. (2012), Bangladesh/mean 37 years, cross‐sectional study (HEALS)Dyspnea (interview)11,746, 7568 of which were never‐smokers

w‐As (μg/L)

Q1: < 7

Q2: 7–38

Q3: 39–90

Q4: 91–178

Q5 > 179

u‐As (μg/g creatinine) quintiles (cut‐offs not reported)

OR (95% CI) for never‐smokers:

1.00 (ref)

1.36 (0.97–1.90)

1.96 (1.43–2.70)

2.14 (1.56–2.92)

1.80 (1.31–2.49)

1.00 (ref),

1.37 (0.97–1.92),

1.92 (1.38–2.65),

1.94 (1.41–2.68),

1.87 (1.36–2.58)

Adjusted for age, sex, education, BMI, systolic blood pressure.
Parvez et al. (2013), Bangladesh/mean 42 years, cohort (HEALS)Pre‐ and post‐bronchodilator spirometry for subjects with respiratory symptoms950 with good quality spirometry

w‐As (μg/L) (tertiles, n)

T1: < 19 (312)

T2: 19–97 (315)

T3:> 97 (315)

T1: < 19 (312)

T2: 19–97 (315)

T3:> 97 (315)

u‐tAs (μg/g creatinine, n):

T1 < 125 (298)

T2: 125–285 (321)

T3 > 285 (305)

T1 < 125 (298)

T2: 125–285 (321)

T3 > 285 (305)

w‐As beta (95% CI):

FEV1 (mL):

0 (Ref)

−33.1 (−114.6 to 48.4)

−80.6 (−181.4 to −17.5)

FVC (mL):

0 (Ref)

−13.2 (−97.3 to 71.0)

−97.3 (−181.8 to −12.7)

FEV1 (mL):

0 (Ref)

−67.0 (−148.3 to 14.1)

−90.5 (−173.6 to −7.4)

FVC (mL):

0 (Ref)

−36.4 (−120.4 to 47.0)

−81.0 (−166.7 to 4.8)

Adjusted for age, sex, BMI smoking, betel nut use, education and arsenical skin lesions. Data report post‐bronchodilator findings. FEV1/FVC not significantly associated with w‐As or u‐tAs.

Significant associations with FVC were present also in never‐smokers. For FEV1 p‐values about 0.1.

Das et al. (2014), India, non‐smoking male adults, mean age 35 years

Cross‐sectional study

Respiratory symptoms assessed by questionnaire SpirometryAs exposed versus ‘non‐exposed’ 446/388

As in drinking water (μg/L)

Exposed: 11–50

(mean 22.9)

‘non‐exposed’: ≤ 10 (mean 3.6)

OR (95% CI) for low spirometry values

FEV1 < 80% predicted: 1.22 (1.04–1.72)

FVC < 80% predicted: 1.37 (1.10–1.96)

Spirometry results adjusted for age and height by design, and also for biomass fuel and agricultural activities. Comparisons of respiratory symptoms were unadjusted, but group differences in age, and prevalence of ex‐smokers (about 7%) were minor
George et al. (2015), Bangladesh/1–60 months children (nested in PERCH study), case control studyHospitalisation for severe or very severe pneumonia (physician‐ diagnosed)153/296

Total u‐As (μg/L, median) cases: at hospitalisation 14

30 days later: 16

Controls: 14

At hospitalisation:

Q1 < 5.9

Q2: 6–16.9

Q3: 17–50.9

Q4 ≥ 50

30‐days later:

Q1 < 5.9

Q2: 6.0–14.9,

Q3: 15.0–41.9,

Q4 ≥ 42

OR (95%CI):

1.00

1.75 (0.90–3.40)

2.11 (1.01–4.34)

2.04 (0.92–4.51)

1.00

2.25 (1.23–4.11)

2.29 (1.17–4.47)

2.56 (1.27–5.15)

Adjusted for weight, height, breastfeeding in the prior week, paternal education, age, number of people in the household and urinary creatinine. Sub‐analysis for severe pneumonia with similar findings. Total u‐As considered OK as marker of u‐tiAs exposure in this Matlab area

Steinmaus et al. (2016);

Nardone et al. (2017)

Chile/39–60 years, Cross‐sectional study with retrospective exposure data

Spirometry, respiratory symptoms and self‐reported disease795 individuals from areas with elevated or low As in drinking water

As exposed (long‐term average w‐As (μg/L):

Never‐smokers:

Around 60 (N = 107) versus < 11 (N = 60)

Ever‐smokers:

Around 60 (N = 97) versus < 11 (N = 73)

Steinmaus:

Multiple linear regression of FVC (mL, 90% CI):

−192 (−351, −34)

−62 (−189, 64)

FEV1 not significant

Adjusted for age, gender, height (Steinmaus: also race and smoking). There were also similar associations with highest w‐As any year (so not time‐weighted average) over lifetime or in early age
Ahmed et al. (2017), rural Bangladesh/9 years, cohort of children (from MINIMat)Spirometry data, FeNO, serum CRP and Club cell protein (CC16)540 (total)

u‐As (μg/L, median)

At GW8: 76,

At 4.5 years: 57

At 9 years: 53

Maternal log u‐As association with spirometry (beta, 95% CI) in 9‐year children:

FVC (mL): −12 (−22, −1.5),

FEV1 (mL): −12 (−22, −1.9).

Not significant (NS) for VC, FEV1/FVC, MMF, PEF, CC16;

FeNO: NS for maternal u‐As.

Significant association with log u‐As at 4.5‐years and 9 years in in boys, but not in girls or all children.

Long‐term cumulative As exposure (> 150 μg/L): Significant inverse associations btw spirometry and maternal u‐As and u‐As in children at 4.5 and 9 years

Adjusted for children's age, height‐for‐age z‐score (HAZ), SES, sex, season, mothers' education, plasma concentrations of CRP and maternal micronutrient supplementation groups. Significant associations were restricted to boys, not girls.

Nardone et al. (2017)

(the same study as Steinmaus et al. (2016))

Similar associations as in Steinmaus et al. (2016), but stronger associations in individuals with high BMIAdditional information between correlation of different BMI categories
Powers et al. (2019), USA American Indians/mean 57 years, cohort (Strong Heart Study)Spirometry, respiratory symptoms, self‐reported asthma, emphysema, chronic bronchitis2132, obstructive spirometry in 458, restrictive in 307

u‐tiAs (μg/g creatinine):

T1 ≤ 7,

T2 7.1–13.9,

T3 ≥ 14

OR T3 versus T1

Obstructive 1.17 (0.99–1.40)

Restrictive 1.18 (0.93–1.50

Emphysema 1.66 (1.29, 2.15)

Asthma 0.76 (0.60, 0.96)

Not associated with other symptoms and chronic bronchitis

Adjusted for age, sex, education, site, smoking, eGFR, tuberculosis, BMI, diabetes. Data also for lower limit of normal (LLN), as well as for FEV1, FVC in mL
Prasad et al. (2020), India/mean 41 years, only females, cross sectional studySpirometry281 from three areas with varying w‐As

w‐As level

(μg/L) (n)

> 50 (70) mean 68.5

11–50 (93) mean 22.5

Cont < 10 (118) mean 1.0

High > 50 (70)

Low 11–50 (93)

Controls < 10 (118)

FVC% predicted

73.2 ± 12.9, p < 0.001 versus controls

83.8 ± 19, NS versus controls

88.3 ± 22.4

FEV1% predicted

77.1 ± 17.8, p < 0.001 versus controls

90.5 ± 26.6, NS versus controls

91.1 ± 25.5

Only females and non‐smokers were included. Associations between nail As concentration and As water concentration were significant. Associations btw nail As, and spirometry data showed significant dose–response relations
Rahman, Niemann, and Munson‐McGee (2022a), US > 20 years, cross‐ sectional (NHANES 2011–2016)Chronic bronchitis (self‐reported based on a specific question)

4186

88 cases

u‐tAs, u‐iAs, u‐MMA, u‐DMA, u‐AsB, u‐AsC (not u‐tiAs)No significant association with chronic bronchitisAdjusted for gender, race/ethnicity, education, marital status, age, family income, BMI, cotinine, alcohol and country of birth
Sanchez et al. (2019), USA/mean 63 years, cohort (MESA)Spirometry, CT findings310 (total)

u‐tiAs median (IQR): 3 μg/g (2–5)

increase u‐tiAs per IQR (2.5 μg/g)

mean difference (95% CI):

FEV1 (mL): −46 (−171, 79)

FVC (mL): −15 (−30, 99)

FEV1/FVC%: 0.01 (−0.02, 0.03)

CT findings: non‐significant except for interstitial lung abnormalities: OR (95% CI) 1.95 (0.44, 8.62)

Adjusted for sex, age, race/ethnicity, study site, education, height, weight, smoking. Also data on rice consumption are reported
Siddique et al. (2020), Bangladesh/18–60 years, cross sectionalSpirometry with reversibility test, asthma symptoms, serum IgE

842 Exposed/non‐exposed 653/189

High n: 282, Medium n: 281, Low n: 279

w‐As (μg/L)

(tertiles, n)

0.03–5.30 (279)

5.32–134 (281)

135–1800 (286)

0.03–5.30

5.32–134

135–1800

0.03–5.30

5.32–134

135–1800

0.03–5.30

5.32–134

135–1800

0.03–5.30

5.32–134

135–1800

0.03–5.30

5.32–134

135–1800

β (95% CI)

FEV1:

0 (ref)

−0.123 (−0.219 to −0.028)

−0.203 (−0.334 to −0.071)

FEV6

0 (ref)

−0.122 (−0.211 to −0.033)

−0.169 (−0.309 to −0.028)

FEV1/FEV6

0 (ref)

−0.0.18 (−0.040 to 0.005)

−0.055 (−0.082, −0.028)

Obstruction (OR, 95% CI, cases)

1.0 (ref) (14)

1.94 (1.03 to 3.65) (29)

3.65 (1.84 to 7.24) (54)

Reversible obstruction (OR, 95% CI)

1.0 (ref) (10)

1.76 (1.01 to 3.06) (19)

3.81 (1.85 to 7.85) (41)

Asthma related symptoms (OR, 95% CI)

1.0 (ref)

2.04 (1.06 to 3.92)

3.69 (2.23 to 6.11)

Adjusted for age, sex, BMI, smoking, income, education, occupation and clustering village.

Airway obstruction defined as FEV1/FEV6 < 0.73. Reversibility defined as ≥ 12% increase in FEV1.

There was also a dose–response association btw w‐As and serum IgE.

Data are reported also for As in hair and nail with similar results as for w‐As

Signes‐Pastor, Martinez‐Camblor, et al. (2021), USA, cohort (New Hampshire Birth Cohort Study)At 7.5‐year children Spirometry358u‐tiAs at GW 24–28. Median u‐tiAs: 3.6 μg/L

β (95% CI) for doubling of u‐tiAs:

FEV1: −0.10 (−0.18, −0.02)

FVC: −0.08 (−0.14, −0.01)

FEV1/FVC: 0.002 (−0.003, 0.007)

Adjusted for maternal smoking status, children's age, sex and height
Tsai, Lei, et al. (2021), Taiwan/children 0–14 years, cohort (Taiwan Maternal and Infant Cohort Study)Asthma spirometry, self‐reported allergic rhinitis

261

Allergy in 96

Maternal u‐tiAs at GW 28–38 (Median u‐tiAs: 24.9 μg/L),

Children u‐tiAs until 14 years of age (median u‐tiAs: 17.2–41.1 at various visits)

Maternal u‐tiAs‐OR (95% CI):

Asthma: 2.79 (1.52, 5.14). Allergic rhinitis: Not significant.

Child u‐tiAs: OR (95% CI)

Allergic rhinitis: 1.81 (1.11, 2.93).

Asthma: Not significant

Adjusted for child's sex, breastfeeding, ETS exposure, prenatal AsB, maternal allergy, paternal allergy, child's serum IgE.
Vega‐Millán et al. (2021), Mexico/children (mean 10 years), cross‐sectionalRespiratory infectionsIn three villages: 90/53/73

Mean w‐As (μg/L)

in three villages: 11.8, 23.3, 70.01

u‐tAs: 37.3, 47.04, 132.57

Water or u‐As not significantly associated with respiratory infections.

OR 0.96 (0.87–1.05) with water As as a continuous variable

Adjusted for age, sex, tobacco/smoking, weight, community. Data also for CC16 and MMP9

Abbreviations: As, arsenic; AsB, arsenobetaine; BMI, body mass index; CC16, clara cell secretory protein; CI, confidence interval; cont, control; CRP, plasma c‐reactive protein; CT, computed tomography; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; eGFR, estimated glomerular filtration rate; ETS, environmental tobacco smoke; FeNo, fractional exhaled nitric oxide; FEV(1/6), forced expiratory volume (in 1/6 seconds); FVC, forced vital capacity; GW, gestational week; HAZ, height‐for‐age‐z‐score; HEALS, Health Effects of Arsenic Longitudinal Study; HR, hazard ratio; IgE, immunoglobulin E; IQR, interquartile range; LLN, lower limit of normal; MESA, Multi‐Ethnic Study of Atherosclerosis; MINImat, Maternal and Infant Nutrition Interventions in Matlab; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; MMF, maximum mid‐expiratory flow; MMP9, matrix metalloproteinase 9; n, number; NHANES, National Health and Nutrition Examination Survey; NS, not significant; OR, odds ratio; PEF, peak expiratory flow; PERCH, Pneumonia Etiology Research for Child Health; Q, quantile; RAO, reversible airway obstruction; ref, reference; SES, socioeconomic status; T, tertile; u‐As, urinary arsenic; u‐AsB, urinary arsenobetaine; u‐AsC, urinary arsenocholine; u‐DMA, urinary DMA; u‐MMA, urinary MMA; USA, United States of America; u‐As, urinary arsenic; u‐tAs, total urinary arsenic; u‐tiAs, total urinary inorganic arsenic (iAs and its methylated metabolites DMA and MMA); VC, vital capacity; w‐As, water arsenic.

Prenatal/childhood exposure

In a case control study in children < 5 years in Bangladesh u‐tiAs in the children was a risk factor for pneumonia hospitalisation (George et al., 2015).

In a cohort of children in rural Bangladesh, prenatal arsenic exposure (u‐tiAs in pregnant mothers) or persistently high exposure in childhood was related to impaired lung function with decreases in FEV1 and FVC, mostly in boys (Ahmed et al., 2017).

In a cohort of US children gestational As exposure (assessed by u‐tiAs) at levels relevant to the general US population were associated with reduced children's FVC and FEV1, but not FEV1/FVC. Of note, u‐tiAs levels were low and > 75% of the participants were exposed to low As levels from household water (< 5 μg/L) (Signes‐Pastor, Martinez‐Camblor, et al., 2021).

In a cohort of children 0–14 years in Taiwan, prenatal As exposure (assessed by u‐tiAs) was significantly associated with the occurrence of asthma but not with allergic rhinitis; in contrast childhood exposure (assessed by u‐tiAs) was significantly associated with allergic rhinitis but not asthma (Tsai, Lei, et al., 2021).

In a cross‐sectional study in children in Mexico water or urine As was not significantly associated with respiratory infections (Vega‐Millán et al., 2021).

Adult populations

In a cohort (HEALS) study in adults in Bangladesh a dose–response relationship was observed between iAs exposure (assessed by water As and u‐tiAs) and respiratory symptoms (Parvez et al., 2010).

Based in the same cohort a cross‐sectional study showed that As exposure (assessed by water As and u‐tiAs) was associated with dyspnoea in never‐smokers (Pesola et al., 2012).

In an expansion of this cohort a significant inverse relationship between tiAs exposure (assessed by water As and u‐tiAs) and post‐bronchodilator forced expiratory volume during first second (FEV1) and FVC was reported in those individuals with respiratory symptoms (Parvez et al., 2013).

In a cross‐sectional study in adults there were no associations between u‐tiAs and asthma, emphysema, chronic bronchitis or respiratory symptoms in the general US population (NHANES 2003–2006), but the outcomes of interest were self‐reported and adjustments for organic As were crude (Amster et al., 2011).

In a cross‐sectional study of adults in Pakistan, high (> 100 μg/L and > 250 μg/L) As in drinking water was associated with a decrease in FVC but there was no association with respiratory symptoms (Nafees et al., 2011). However, the association with w‐tiAs 100–250 μg/L is unclear.

In a cross‐sectional study in male adults in India with elevated As in drinking water had higher prevalence of upper and lower respiratory symptoms, asthma and eye irritation, as well as lower lung function of restrictive and obstructive types at spirometry compared with controls (Das et al., 2014).

A study in adults in Chile, two studies showed that As exposure from drinking water was associated with respiratory symptoms. In addition, higher w‐tiAs was associated with lower FVC, but statistically significant only in never smokers (Nardone et al., 2017; Steinmaus et al., 2016).

In a cohort with US adults focused on rice consumption, u‐tiAs was measured in a subgroup but not significantly associated with impaired lung function assessed by spirometry or any CT finding except some that were consistent with interstitial lung disease (Sanchez et al., 2019).

In a cohort of US American Indians u‐tiAs was associated with impaired lung function showing a significant association with restrictive pattern and a trend for obstructive pattern at spirometry. In addition, u‐tiAs was associated with self‐reported emphysema and stopping for breath but not with other symptoms examined (Powers et al., 2019).

In a cross‐sectional study in female non‐smoking adults in India iAs exposure (assessed by water As) was associated with reduction of FEV1 and FVC (Prasad et al., 2020).

In a cross‐sectional study in adults in Bangladesh As exposure (assessed by As in water, hair and nail) was associated with decreased lung volumes and risk of asthma assessed by reversible airway obstruction, asthma‐related symptoms and serum IgE levels (Siddique et al., 2020).

In a cross‐sectional study in adults, u‐As in any form (organic, inorganic or total) was not associated with chronic bronchitis in the general US population from the NHANES 2011–2016 datasets, but the outcome of interest was self‐reported and the number of cases was limited (Rahman, Niemann, & Munson‐McGee, 2022a).

In summary, the epidemiological studies provide sufficient evidence for an association between low to moderate exposure to iAs and impaired lung function (decreased lung volumes). This is also consistent with the conclusion in a recent meta‐analysis (Sanchez et al., 2018). Associations have been reported both in children and in adults. Several biologically plausible mechanisms have been described, cellular injury caused by inflammation, increased ROS generation (Sanchez et al., 2018). There is insufficient evidence for associations between exposure to iAs and respiratory disease (infections and asthma).

3.2.2.9. Effects on diabetes and insulin resistance

In the previous EFSA Opinion (2009) it was concluded that there remained uncertainty whether arsenic contributes to the occurrence of type 2 diabetes and that there was inadequate data from which to inform on dose response at lower level of exposure. In addition to type 2 diabetes, the current Opinion also includes gestational diabetes and type 1 diabetes.

The CONTAM Panel identified 65 studies from a literature search. Out of these, 13 studies met the criteria (Table 29).

TABLE 29

Key epidemiological studies on diabetes and insulin resistance in humans in relation to iAs exposure.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders
Gestational diabetes

Chen, Davis, et al. (2021)

Oklahoma, USA

Case–control

Clinical diagnoses

Cases: 64

Controls: 237

u‐tiAs (μg/L), medians

Cases: 3.02, Controls: 3.42

iSumAs = Sum of inorganic and methylated arsenic

≤ 4.54

4.55–6.75

> 6.76

Adjusted OR (95% CI)

1.00

0.66 (0.29–1.49)

0.73 (0.34–1.61)

No associations with AsIII, AsV, iAs, MMA or DMA

Adjusted for age, race/ethnicity, BMI, smoking, history of GDM

Muñoz et al. (2018)

Arica, Chile

Cross‐sectional

From medical charts (WHO criteria)21 out of 244 had GD

u‐tiAs (μg/L)

Medians were 15.12 among cases and 14.72 among controls

2.05–11.08

11.09–19.90

19.91–69.30

Adjusted OR (95% CI)

1.0

2.98 (0.87–10.18)

1.07 (0.26–4.33)

No association with MMA% or DMA%

Adjusted for age, education, ethnicity and BMI
Type 1 diabetes

Grau‐Pérez et al. (2017)

Five sites in the USA

Cross‐sectional case–control

Clinical diagnoses

Cases: 429

Controls: 174

(in addition 85 with type 2 diabetes)

iAs in plasma (ng/L)

Medians among cases and controls:

ΣAs: 81.7 and 83.1

iAs: 46.6 and 50.2

MMA: 8.9 and 8.1

DMA: 22.8 and 21.0

ΣAs = sum of iAs, MMA and DMA

As metabolism

iAs%: 58.7 and 63.4

MMA%: 11.2 and 10.3

DMA%: 28.9 and 25.2

Adjusted OR (95% CI)

Interquartile range increases

ΣAs (ng/L): 0.90 (0.71–1.13)

iAs (ng/L): 0.85 (0.73–1.00)

MMA (ng/L): 1.21 (0.93–1.57)

DMA (ng/L): 1.15 (0.94–1.42)

iAs%: 0.70 (0.53–0.92)

MMA%: 1.29 (1.01–1.65)

DMA%: 1.27 (1.01–1.58)

Adjusted for age, sex, BMI, educational level and ethnicity
Type 2 diabetes

Steinmaus et al. (2009)

NHANES, USA

Cross‐sectional case–control

Self‐reported diagnoses or fasting glucose serum measurements

Cases: 98

Controls: 697

u‐tiAs (μg/L)

Median and Mean (SD)

All 6.0 and 9.4 (12.9)

T2D 6.2 and 11.1 (16.9)

No T2D 6.0 and 9.2 (12.3)

Adjusted OR (95% CI)

Estimated iAs

≥ 80th versus ≤ 20th percentile

(11.9 vs. 2.7 μg/L)

1.15 (0.53–2.50)

A reanalysis of a former study which observed strong association but did not subtract arsenobetaine from total As

Adjusted for sex, age, ethnicity, education, BMI, cotinine, hypertension medication

Chen, Ahsan, et al. (2010)

Araihazar, Bangladesh

Cross‐sectional

Self‐reported physician diagnosed T2D

HbA1c was analysed for part of the participants

241 out of 11,319 had T2D

‐ Time‐weighted As concentrations (TWA) was estimated as a function of drinking durations and well arsenic concentrations

‐ Total As urine concentrations

‐ As metabolites measured in a random 10% of the populations

Adjusted OR (95% CI)

No significant associations between

TWA and T2D

TWA and glucosuria

Adjusted for age, sex, BMI, smoking, education and creatinine

Li, Li, Xi, Zheng, Lv, and Sun (2013)

Inner Mongolia, China

Cross‐sectional

Fasting blood glucose levels42 out of 669 participants had T2D

w‐As, μg/L (cases/n)

< 10 7/124

10–50 14/207

> 50 21/338

Adjusted OR (95% CI)

< 10 1.00

10–50 1.36 (0.52–3.57)

> 50 1.57 (0.58–4.26)

Adjusted for sex, age, smoking, BMI and alcohol

Nizam et al. (2013)

Faridpur, Bangladesh

Case–control study

Physician‐diagnosed T2D

140 cases

180 controls

Urine As, μg/L, mean

Cases – Controls

u‐tiAs: 252.2–235.3

iAs: 20.0–21.2

MMA: 23.9–22.22

DMA: 208.3–192.0

p‐values case versus controls

u‐tiAs: 0.56

iAs: 0.65

MMA: 0.60

DMA: 0.81

from adjusted models

iAs%: 0.34

(higher among controls)

MMA%: 0.08

(higher among controls)

DMA%: 0.14

(higher among cases)

Adjusted for sex, union, age, water arsenic, income, duration of drinking water, family history of diabetes, smoking and BMI

Kuo et al. (2015)

The Strong Heart Study, USA

Follow‐up study

Baseline 1989–91

Followed through 1998–99.

Fasting plasma glucose level, self‐reported diabetes history or use of anti‐diabetic medicationsOut of 1694 diabetes‐free 396 developed diabetes

Urine As, μg/g (median)

No DM DM

u‐tiAs 8.7 9.1

iAs 0.7 0.7

MMA 1.3 1.2

DMA 6.4 7.0

iAs% 8.4 8.1

MMA% 15.5 14.0

DMA% 75.9 77.4

Adj HR 95% CI

Per doubling increase

u‐tiAs: 0.96 (0.85–1.08)

iAs: 0.98 (0.90–1.06)

MMA: 0.90 (0.81–1.00)

DMA: 0.98 (0.87–1.11)

Methylation –

Conventional approach

(per 5% increase)

Adjusted for age, sex, education, smoking, alcohol, BMI, waist‐to‐hip ratio. Also ‘leave‐one‐out’ approaches were reported

Grau‐Perez et al. (2017)

Strong Heart Family study, USA

Follow‐up

Two outcomes:

– T2D at follow‐up via fasting plasma glucose or self‐reported physician diagnoses or treatments

– HOMA2‐IR, fasting glucose and insulin values

252 out of 1838 developed T2D

Urine As (median)

u‐tiAs 4.4 μg/g creatinine

iAs% 9.5%

MMA% 14.4%

DMA% 75.6%

Adjusted HR (95%CI)

per interquartile range u‐tiAs

Overall: 1.16 (0.94–1.42)

Normal Fasting Glucose at

Baseline (NFG): 1.57 (1.18–2.08)

Impaired Fasting Glucose at

Baseline (IFG): 0.92 (0.67–1.27)

As metabolism was not associated with incident diabetes or insulin resistance

Adjusted for sex, age, education, BMI, waist circumference, smoking, glomerular filtration rate, glucose status (only overall), estimated dietary vitamin B2, vitamin B6 and folate, and AS3MT genotype

Yang et al. (2019)

CARDIA study, USA

Follow‐up study

Baseline 1987–1988

Followed through 2015–16.

Fasting glucose level

599 out of 4102 developed T2D during the 28 years follow‐up

(individuals completed up to 7 follow‐up exams)

Total toenail As level, ppm

Median in respective quintile

Q1: 0.047

Q2: 0.071

Q3: 0.097

Q4: 0.135

Q5: 0.237

Adjusted HR (95% CI)

1.00

0.95 (0.75–1.22)

1.04 (0.81–1.33)

1.00 (0.77–1.30)

0.96 (0.73–1.27)

Adjusted for age, sex, race, study centre, baseline glucose levels, BMI, education, smoking, physical activity and family history for diabetes

Zhang et al. (2020)

Northern China

Cross‐sectional

Fasting plasma glucose levels or self‐reported physician diagnoses or medications49 out of 1003 had T2D

Urine As concentrations,

μg/g Cr

T2D – Non‐T2D (medians)

iAs: 7.6–15.4

MMA: 11.7–17.5

DMA: 95.7–101.7

u‐tiAs: 112.8–138.1

Urine As metabolism

iAs%: 7.0–10.4

MMA%: 10.3–13.6

DMA%: 82.2–74.2

p‐values for U‐As concentrations between

T2D – Non‐T2D

iAs: 0.005

MMA: 0.019

DMA: 0.591

u‐tiAs: 0.196

Adjusted for gender, age, education, job, smoking, drinking, BMI, study area and u‐tiAs in urine.
Insulin resistance

Li et al. (2021)

NHANES (2003–2016), USA

Cross‐sectional

HOMA2‐IR was calculated based on participants fasting glucose and insulin values3730 individuals

Urine As concentrations (μg/g Cr), Medians

u‐tiAs: 5.96

iAs: 0.63

MMA: 1.21

DMA: 3.98

The Geometric Mean Ratio (95% CI) of HOMA2‐IR

Per doubling increase

u‐tiAs: 0.99 (0.97, 1.01)

p‐value = 0.14

Adjusted for age, sex, race/ethnicity, BMI, smoking, alcohol, education, physical activity, fish consumption, B‐Hg

Lin et al. (2014)

Taiwan

Cross‐sectional

Insulin resistance determined by HOMA‐IR303 elementary school students and 319 junior school students

Urine As concentrations (μg/L), Mean

Elementary/Junior

u‐tiAs: 24.54 /25.92

As metabolism

iAs%: 5.06/7.57

MMAV%: 5.00/5.30

DMAV%: 90.52 /87.5

Adjusted Β (95% CI) from linear regression models

u‐tiAs (μg/L)

All students:

0.024 (0.017–0.047)

Elementary:

0.044 (0.017–0.071)

Junior:

0.017 (0.001–0.034)

Adjusted for age, sex, BMI, lipid profiles, liver function, paternal and maternal education, smoking

Abbreviations: As, arsenic; AsIII, arsenite; AsV, arsenate; AsB, arsenobetaine; AS3MT, arsenite methyltransferase; BMI, body mass index; B‐Hg, blood mercury; CAE, cumulative arsenic exposure; CARDIA, Coronary Artery Risk Development in Young Adults; CI, confidence interval; Cr, creatinine; DM, diabetes mellitus; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; GD, gestational diabetes; GDM, gestational diabetes mellitus; GM, geometric mean; HbA1c, glycated haemoglobin A1c; HOMA‐IR, homeostasis model assessment‐insulin resistance; HR, hazard ratio; iAs, inorganic arsenic; IFG, impaired fasting glucose; iSumAs, sum of inorganic and methylated arsenic; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; n, number; NFG, normal fasting glucose; NHANES, National Health and Nutrition Examination Survey; OR, odds ratio; ppm, parts per million; Q, quantile; SD, standard deviation; T2D, type 2 diabetes; t‐As, total arsenic; TWA, time‐weighted average; USA, United States of America; u‐tiAs, total urinary inorganic arsenic (sum of iAs and its methylated metabolites MMA and DMA); w‐As, water‐arsenic; WHO, World Health Organization.

Gestational diabetes

Two studies, one from the U.S. and one from Chile, investigated the association between u‐tiAs and gestational diabetes (Chen, Davis, et al., 2021; Muñoz et al., 2018). The outcome was based on either clinical diagnoses or medical charts. These studies observed no association between iAs exposure measured as u‐tiAs and gestational diabetes (Chen et al., 2021; Muñoz et al., 2018).

Ten additional studies were also evaluated but did not fulfil the inclusion criteria and were accordingly not considered further (Ashley‐Martin et al., 2018; Farzan et al., 2016; McKeating et al., 2021; Peng et al., 2015; Shapiro et al., 2015; Wang, Gao, et al., 2020; Wang, Zhang, et al., 2019; Xia et al., 2018; Zhang, Chen, et al., 2021; Zhang, Zhang, et al., 2021).

Type 1 diabetes

One cross‐sectional study included in this review investigated the association between iAs and type 1 diabetes (Grau‐Pérez et al., 2017). The study, which was from the U.S., observed no association between the total concentrations of inorganic As and its metabolites (iAs + MMA + DMA) in plasma and type 1 diabetes. However, increased concentrations of iAs in serum decreased the risk for type 1 diabetes, whereas concentrations of MMA and DMA, respectively, tended to increase the risk for type 1 diabetes. These associations were somewhat more pronounced when fractions were investigated instead of concentrations.

One study did not fulfil the inclusion criteria and was not considered further (Chafe et al., 2018).

Type 2 diabetes

The five cross‐sectional study populations included in this Opinion are from the U.S., China, Bangladesh and Mongolia (Chen, Ahsan, Slavkovich, Peltier, Gluskin, Parvez, et al., 2010; Li, Li, Xi, Zheng, Lv, & Sun, 2013; Nizam et al., 2013; Steinmaus et al., 2009; Zhang et al., 2020). Four of these studies showed no associations between iAs and type 2 diabetes (Chen, Ahsan, Slavkovich, Peltier, Gluskin, Parvez, et al., 2010; Li, Li, Xi, Zheng, Lv, & Sun, 2013; Nizam et al., 2013; Steinmaus et al., 2009), whereas one did so regarding fractions of arsenic (Zhang et al., 2020). This study, which was from China, did not observe an association between u‐tiAs and type 2 diabetes, whereas the concentrations of iAs and MMA were significantly lower among the individuals with type 2 diabetes. In adjusted analyses, the tertile with highest fraction of MMA had decreased occurrence of type 2 diabetes and the tertile with the highest fraction of DMA had increased occurrence of type 2 diabetes (Zhang et al., 2020).

Three prospective studies from the U.S. investigated the association between iAs and risk of developing type 2 diabetes (Grau‐Perez et al., 2017; Kuo et al., 2015; Yang et al., 2019). Kuo and colleagues found no significant differences in iAs concentrations at baseline between those who developed type 2 diabetes and those who did not (Kuo et al., 2015). However, arsenic metabolism was prospectively associated with type 2 diabetes incidence, higher percentage of MMA being consistently associated with lower risk of diabetes. In a study by Grau‐Perez and colleagues no overall association was indicated between u‐tiAs and type 2 diabetes (Grau‐Perez et al., 2017). However, higher concentrations were associated with an increased risk for type 2 diabetes among those with normal fasting glucose at baseline but not among those with impaired fasting glucose at baseline. A third study by Yang and colleagues with very long follow‐up (up to 28 years) and up to seven follow‐up exams did not find associations between arsenic and type 2 diabetes (Yang et al., 2019). This study analysed arsenic concentrations in toenails and did not perform arsenic specification. However, according to the authors toenails have been suggested to preferentially sequester iAs and its metabolites. In addition, sensitivity analyses with adjustment for fish/seafood consumption did not change the results.

Thirty‐one additional studies were also evaluated but did not fulfil the inclusion criteria and were accordingly not considered further (Arab YarMohammadi et al., 2021; Becker & Azelrad, 2014; Bräuner et al., 2014; Castriota et al., 2018; Currier et al., 2014; Dai et al., 2020; Del Razo et al., 2011; Eick et al., 2019; Feseke et al., 2015; Grau‐Perez et al., 2018 ; Gribble et al., 2012; Hansen et al., 2017; Hsu et al., 2013; Huang et al., 2014; Islam, Khan, Hassan, et al., 2012; Jovanovic et al., 2013; Kim et al., 2013; Kim & Lee, 2011; Konkel, 2020; Liu et al., 2016; Lucio et al., 2020; Makris et al., 2012; McLeod et al., 2019; Pan, Kile, et al., 2013; Pan, Seow, et al., 2013; Rehman et al., 2019; Rhee et al., 2013; Simic et al., 2017; Sripaoraya et al., 2017; Wang, Karvonen‐Gutierrez, et al., 2020; Yuan et al., 2018).

Insulin resistance

A cross‐sectional study in the U.S. did not find an association between u‐tiAs and HOMA2‐IR, i.e. insulin resistance (Li et al., 2021). However, lower MMA% was associated with higher HOMA2‐IR levels among obese participants but not among non‐obese participants. Another cross‐sectional study among students from Taiwan observed that higher concentrations of u‐tiAs was associated with higher HOMA‐IR (Lin et al., 2014).

Ten additional studies were also evaluated but did not fulfil the inclusion criteria and were accordingly not considered further (Fleisch et al., 2022; Lampron‐Goulet et al., 2017; Mondal et al., 2020; Ourshalimian et al., 2019; Park et al., 2016; Paul et al., 2019; Su et al., 2012; Tinkelman et al., 2020; Wang, Mukherjee, et al., 2020; Zhou, Zhao, & Huang, 2022).

In summary, most of the studies were cross‐sectional which might be affected by reverse causality. Although some studies indicate associations, especially in subgroup analyses, the epidemiological studies provide insufficient evidence for an association between low to moderate exposure to iAs and diabetes or glucose metabolism.

3.2.2.10. Metabolic syndrome

The CONTAM Panel identified eight studies from a literature search. Out of these, four studies met the criteria. The relevant studies are summarised in Table 30.

TABLE 30

Key epidemiological studies on metabolic syndrome in humans in relation to iAs exposure.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Chen et al. (2012)

Southwestern Taiwan

Cohort study

Recruited in 1990 and follow‐up in 2002–03

Metabolic syndrome (MetS) based on criteria for fasting plasma glucose, triglycerides, high density lipoprotein, increased systolic of diastolic blood pressure and waist girth247 individuals, 111 with MetS and 136 with no MetS

u‐As (mean) at follow‐up

No Mets – Mets

AsIII (μg/g creatinine) 3.79–4.12

AsV 1.78–1.47

iAs 5.57–5.58

MMA 4.51–3.08

DMA 32.79–35.52

u‐tiAs 42.87–44.19

iAs% 17.2–16.8

AsIII% 10.1–10.4

AsV% 7.3–6.9

MMA% 10.6–9.4

DMA% 72.3–73.5

PMI 0.92–0.75

(MMA/(AsIII+AsV))

SMI 16.12–19.98

(DMA/MMA)

As concentrations in well‐water

(μg/L)

570–684

Cumulative As exposure, CAE

(mg/(L*years))

13.96–17.71

Age‐adjusted p‐values

No Mets versus Mets

AsIII 0.79

AsV 0.29

iAs 0.97

MMA 0.03

DMA 0.42

u‐tiAs 0.66

iAs% 0.98

AsIII% 0.98

AsV% 0.99

MMA% 0.06

DMA% 0.37

PMI 0.06

SMI 0.42

Adjusted OR (95% CI)

Highest versus lowest tertile

In well water 1.24 (0.65–2.37)

CAE 1.73 (0.72–4.19)

In urine:

AsIII% 0.78 (0.41–1.49)

AsV% 0.80 (0.43–1.49)

MMA% 0.35 (0.18–0.66)

DMA% 2.01 (1.05–3.86)

PMI 0.39 (0.20–0.76)

SMI 2.61 (1.35–5.08)

Adjusted for age and betel nut chewing

Kazemifar et al. (2020)

Qazvin province, Iran

Cross‐sectional

Metabolic syndrome using the criteria suggested by the Third National Cholesterol Education Program (NCEP) report45 with metabolic syndrome and 87 without

Urinary concentrations,

Means, (μg/L)

cases – controls

iAsIII 2.32–2.36

iAsV 0.69–0.74

iAs 3.01–3.10

MMA 2.90–3.12

DMA 15.53–14.15

%iAs 14.45–14.46%MMA 13.69–15.33

%DMA 71.85–68.19

MMA/iAs 0.97–0.99

DMA/MMA 5.28–4.59

p‐values for comparisons between cases and controls

iAsIII 0.43

iAsV 0.14

iAs 0.15

MMA 0.06

DMA 0.07

%iAs 0.017%MMA  < 0.001

%DMA  < 0.001

MMA/iAs 0.45

DMA/MMA  < 0.001

Adjusted OR (95% CI) by unit increase

%iAs 0.90 (0.75–1.08)

%MMA 0.76 (0.59–0.97)

%DMA 1.18 (1.07–1.39)

MMA/iAs 0.39 (0.04–3.73)

DMA/MMA 1.86 (1.05–3.31)

Adjusted for age and BMI

Spratlen et al. (2018)

Strong Heart Family Study, USA

Follow‐up study

(Baseline visits 1998–99 and 2001–03 and

Follow‐up visits 2001–03 and 2006–09)

Metabolic syndrome was characterised according to the National Cholesterol Education Program Adult Treatment Panel III guidelines

1047 participants with no MetS at baseline

338 developed MetS during follow‐up

Urinary concentrations at baseline

Medians, No MetS – MetS

u‐tiAs (μg/L) 6.3–7.0

iAs% 10.0–9.9

MMA% 16.0–14.2

DMA% 72.1–75.1

p‐values for comparisons between cases and controls

u‐tiAs (μg/L) 0.21

iAs% 0.01

MMA%  < 0.001

DMA%  < 0.001

Adjusted RR (95% CI)

per IQR increase

u‐tiAs 1.03 (0.90–1.18)

per 5% increase

iAs% 0.94 (0.88–1.01)

MMA% 0.87 (0.79–0.95)

DMA% 1.07 (1.02–1.12)

Adjusted for urinary creatinine concentration, sex, region, education, alcohol, smoking, kidney function and BMI.

Pace et al. (2018)

NHANES, USA

Cross‐sectional

A diagnose was given based on the following variables:

Fasting glucose, systolic or diastolic blood pressure, triglycerides, waist circumferences, HDL‐cholesterol

957 individuals participated whereof

331 had MetS

Urinary As species, mean

(μg/L)

iAsIII: 0.51

MMA III + V: 0.63

DMA III + V: 4.68

%iAs: 9.68%MMA: 11.69

%DMA: 78.63

MMA/iAs: 1.89

DMA/MMA: 10.44

p‐values obtained from adjusted logistic regression

Gender specific analyses

Men ‐ Women

%iAs 0.950–0.977%MMA 0.021–0.153

%DMA 0.150–0.078

MMA/iAs 0.173–0.876

DMA/MMA 0.152–0.037

Adjusted for age, Poverty Income Ratio, ethnicity, smoking

Abbreviations: As, arsenic; AsIII, arsenite; AsV, arsenate; BMI, body mass index; CAE, cumulative arsenic exposure; CI, confidence interval; DMA, sum of dimethylarsinite and dimethylarsinate; DMA(III), dimethylarsinous acid; DMA(V), dimethylarsinic acid; HDL, high‐density lipoprotein; iAs, inorganic arsenic; iAsIII, arsenite; iAsV, arsenate; IQR, interquartile range; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; MMA(III), monomethylarsonous acid; MMA(V), monomethylarsonic acid; MetS, metabolic syndrome; n, number; NCEP, National Cholesterol Education Program; OR, odds ratio; NHANES, National Health and Nutrition Examination Survey; PMI, primary methylation index; RR, risk ratio; SMI, secondary methylation index; u‐As, urinary arsenic; USA, United States of America; u‐tiAs, total urinary iAs (sum of iAs and its methylated metabolites MMA and DMA).

Two cross‐sectional studies and two follow‐up studies investigated the associations between iAs and metabolic syndrome (Chen et al., 2012; Kazemifar et al., 2020; Pace et al., 2018; Spratlen et al., 2018). A cross‐sectional study from Iran did not find associations between urinary concentrations of iAs and metabolic syndromes (Kazemifar et al., 2020). However, increased MMA% was associated with decreased occurrence of metabolic syndrome whereas increased DMA% was associated with increased occurrence of metabolic syndrome. Neither did the second cross‐sectional study, which was performed in the U.S., report an association between urinary concentrations of iAs and metabolic syndrome (Pace et al., 2018). When gender specific analyses were performed in this study, the %MMA was statistically significantly associated with metabolic syndrome among men but not among women. On the other hand, the ratio DMA/MMA gave a significant association among women but not among men. None of two follow‐up studies, one from Taiwan and one the U.S., showed an association between the baseline concentrations of u‐tiAs in urine and the risk of developing metabolic syndrome (Chen et al., 2012; Spratlen et al., 2018). However, both studies showed that increased MMA% was associated with decreased risk of developing metabolic syndrome and that increased DMA% was associated with increased risk of developing metabolic syndrome. Even in longitudinal studies it is difficult to judge whether such associations are causal, since a metabolic syndrome may affect the metabolism of iAs.

Four additional studies were also evaluated but did not fulfil the inclusion criteria and were accordingly not considered further (Asprouli et al., 2019; Bulka et al., 2019; Choi et al., 2014; Martínez‐Barquero et al., 2015).

In summary, the evidence for an association between iAs exposure and metabolic syndrome is insufficient.

3.2.2.11. Renal effects

In the previous EFSA Opinion (EFSA CONTAM Panel, 2009) the only mention of possible effect on the kidney was that a Chinese study had shown a possible interaction between arsenic and cadmium on excretion of biomarkers of renal damage, but the CONTAM Panel concluded that further studies were needed to clarify this.

The CONTAM Panel identified 15 studies on associations between As exposure and renal effects from a literature search. Out of these, 10 studies met the inclusion criteria, while 5 studies did not meet the inclusion criteria (Chen, Chen, et al., 2011; Jiménez‐Córdova et al., 2018; Smith et al., 2012; Tsai et al., 1999; Weidemann et al., 2015).

Chronic kidney disease (CKD)

The most important studies are those that examine the risk of chronic kidney disease (CKD). Five such studies are reviewed below.

Meliker et al. (2007) examined cause‐specific mortality in an ecological study in six counties in Michigan, US with elevated water As concentrations. The population‐weighted water As in the six counties was 7.6 μg/L, based on 9152 water analyses. In the remainder of Michigan, it was 1.3 μg/L, based on 23,691 analyses. The standardised age‐ and race‐specific mortality ratio for kidney disease in the six counties was 1.28 (1.15–1.42) in males and 1.38 (1.25–1.52) in females, based on 614 and 679 deaths. The results were similar when urban/rural counties were considered.

Hsueh et al. (2009) performed a case–control study in Taiwan, selecting 125 patients with CKD (eGFR < 60 mL/min per 1.73 m2 for 3 months) and 229 controls with eGFR ≥ 60, frequency‐matched for age, from a hospital pool. U‐tiAs was significantly (p < 0.001) higher in cases (mean 31.95 μg/g creatinine) than in controls (20.71 μg/g creatinine). The adjusted OR in the upper tertile (based on u‐tiAs in controls) was 4.34 (1.94–9.69).

Zheng et al. (2015) examined incident CKD in 3119 adults from the US Strong Heart Study. The adjusted HR for CKD (502 cases based on low eGFR, dialysis or kidney transplant at follow‐up) was significantly associated with baseline u‐tiAs. In the upper quartile of u‐tiAs the HR was 1.6 (1.2–2.2) in a model adjusted for potential confounders, and 1.3 (0.9–1.8) in a model additionally adjusted for diabetes. The latter model would be over‐adjusted if As associated diabetes is not a confounder but a factor in the causal chain.

Hsu, Hsieh, et al. (2017) examined the incidence of CKD in relation to concentrations of As in drinking water in a cohort of 6093 individuals in Taiwan followed up over 14 years. The adjusted HR for incident CKD (447 cases from National insurance registry) was significantly associated with water As (p for trend 0.01). The HR was 1.36 (1.05–1.76) in the category 50–150 μg/L.

Cheng et al. (2018) performed a nation‐wide cohort study in Taiwan consisting of about 330,000 members of the National Health Insurance, free of CKD, living in 323 townships with measurements of As in drinking water. About one third of the cohort members had been exposed to elevated As (≥ 50 μg/L) in drinking water before the 1990s. Incidence of end‐stage renal disease (ESRD, serious CKD, requiring dialysis treatment) in the cohort was followed up over 12 years and compared with exposure to As in drinking water aggregated on township level. The adjusted data (data on potential confounders available on individual level) hazard ratio of ESRD was 1.12 (1.06–1.19) in individuals with water As concentrations ≥ 50 μg/L, based on 5442 cases. The authors state in the text that the risk at water As concentrations 50–349 μg/L was similar as the risk at ≥ 350 μg/L, without presenting any details.

Other renal outcomes

Several studies examined associations between iAs exposure and biomarkers of renal function or damage. Such results may suffer from reverse causation, the kidney disease causing changes in renal biomarkers, or confounding due to physiological factors affecting both u‐iAs excretion and renal biomarkers.

In a cross‐sectional study in Korea As exposure was not significantly associated with tubular damage in the kidney assessed by biomarkers (Huang et al., 2009).

In a mixed cohort and cross‐sectional analysis in Bangladesh in adults As exposure (assessed in water and urine) was positively associated with proteinuria prevalence at baseline but not associated with development of proteinuria over time (Chen, Parvez, et al., 2011).

In a mixed cohort and cross‐sectional analysis in Bangladesh in adults a positive association of As exposure (assessed in water and urine) with both prevalence and incidence of dipstick haematuria was found (McClintock et al., 2014).

In two cross‐sectional studies of 478 (Peters et al., 2014) and 379 (Peters, Hall, et al., 2015) adults in Bangladesh no significant association was found between As in water or u‐tiAs and estimated GFR (eGFR) as a continuous variable.

Cheng et al. (2017) examined associations between As in water (aggregated on township) and rapid decrease of eGFR in 8854 individuals participating in two health screenings in a period of 12–24 months. The adjusted OR of a decrease of eGFR by > 5 mL/min/1.73 m2 per year was 1.22 (1.05–1.42) in individuals with As in tap water ≥ 50 μg/L compared with < 50 μg/L.

In summary, three longitudinal studies from Taiwan (Cheng et al., 2018; Hsu, Hsieh, et al., 2017; Hsueh et al., 2009) and two from the US (Meliker et al., 2007; Zheng et al., 2015) show associations between As in drinking water or u‐tiAs and risk of CKD. Although one of the studies (Meliker et al., 2007) had an ecological study design, there is sufficient evidence of an association between low to moderate exposure to iAs and risk of CKD. For biomarkers of renal damage, the evidence of an association is insufficient.

The studies meeting the inclusion criteria for renal effects are described in Table 31.

TABLE 31

Key epidemiological studies on renal effects and As exposure.

Reference study population designOutcome definitionPopulation size (n) case/controlArsenic exposureResultsAdditional information/confounders

Meliker et al. (2007)

Population in six counties in Michigan, USA, ecological study

Mortality from kidney disease

Population in the six counties 740,000 and

1293 deaths 1979–1997. Compared to the entire state

Population‐weighted w‐As in the six counties 7.6 μg/L and 1.3 for the rest of MichiganSMR 1.28 (1.15–1.42) in males and 1.38 (1.25–1.52) in femalesAdjusted for age and race by design

Hsueh et al. (2009)

Taiwan

Case control study

CKD (eGFR < 60 over at least 3 months) from hospital files125 cases and 229 controls from hospital pool of patients with eGFR

u‐tiAs, μg/g creatinine (cases/controls)

≤ 11.78 (19/75)

11.78–20.74 (30/78)

> 20.74 (76/76)

OR

1.0

1.41 (0.62–3.19)

4.34 (1.94–9.69)

Adjusted for age, sex, education, ethnicity, smoking, coffee, analgesics, hypertension, diabetes from interviews

Zheng et al. (2015)

USA (Strong Heart Study)

Cohort study

Incident CKD based on eGFR < 60, dialysis or kidney transplant3119 individuals free of CKD at baseline. 502 incident cases

u‐tiAs, μg/g creatinine (cases/non‐cases)

≤ 5.7 (109/663)

5.8–9.7 (110/671)

9.7–15.6 (128/656)

≥ 15.6 (155/627)

HR by quartiles

1

1.1 (0.8–1.4)

1.2 (0.9–1.7)

1.6 (1.2–2.2)

Adjusted for age, gender, location (state), education, smoking status, BMI, hypertension medication, SBP and baseline eGFR. Also, cross‐sectional analyses were performed, showing a positive association (reverse causation)

Hsu, Hsieh, et al. (2017)

Taiwan

Cohort study

Incident CKD from National Health Insurance Registry over 14 years6093 individuals free of CKD at baseline, 447 incident cases

w‐As (μg/L) (individuals/cases)

≤ 10 (2029/132)

10.1–49.9 (1850/140)

50–149.9 (1281/101)

≥ 150 (933/74)

HR

1.0

1.15 (0.91–1.46)

1.36 (1.05–1.76)

1.35 (1.02–1.80)

Adjusted for age, sex, BMI, education, smoking, alcohol, regular analgesic use, hypertension, diabetes, dyslipidaemia.

Cheng et al. (2018)

Cohort study Taiwan

Cohort study

Incident End Stage Renal Disease (ESRD = CKD requiring dialysis) in members of National Health Insurance Registry over 12 years, 1998–2010362,505 individuals, born before 1958 and free of ESRD at baseline. 5442 incident cases of ESRD

w‐As (μg/L) aggregated by 323 townships

< 50

≥ 50

HR

1.0

1.12 (1.06–1.19)

Adjusted for age, sex, income, urbanisation

level, hypertension, hyperlipidaemia, coronary artery disease, diabetes, anaemia, congestive heart failure. Many individuals in the category ≥ 50 μg/L had very high w‐As, but the authors state in the text that the risk was similar in a category 50–349 μg/L and ≥ 350 μg/L without presenting details

Chen, Parvez, et al. (2011), Bangladesh/mean 37 years, cohort for follow‐up, cross‐sectional for baseline proteinuria (HEALS)ProteinuriaProteinuria yes/no 1030/9130

w‐As (μg/L)

Q1: 0.1–7

Q2:8–39

Q3: 40–91

Q4: 92–179

Q5: 180–864

Cohort: HR (95% CI):

1.00 (ref),

0.84 (0.70–1.01),

0.79 (0.65–1.07),

0.85 (0.70–1.04),

0.84 (0.69–1.06),

p: NS.

Cross sectional: OR (95% CI):

1.00 (ref),

1.01 (0.79–1.31),

1.33 (1.04–1.70),

1.54 (1.22–1.96),

1.65 (1.31–2.09),

p < 0.01

Adjusted for urinary creatinine, age, gender, BMI, cigarette smoking status, education length, SBP, DBP, diabetes status. Also, data for u‐As changes over time
Huang et al. (2009), Korea/unknown age, cross sectionalKidney biomarkers of (tubular) toxicity: ß2‐microglobulin (β2 MG), N‐acetyl‐b‐D‐glucosaminidase (NAG) activity in urineTotal 290

u‐tiAs μg/g (50th percentile)

< 3.94 versus > 3.94

High versus low u‐As mean (SD):

β2 MG: 74.3 (152) versus 78.3(171), p: NS;

NAG: 1.82 (1.91) versus 1.66 (2.36), p: NS.

No significant correlation for u‐As and β2‐MG or NAG

No data for adjustments
McClintock et al. (2014), Bangladesh/18–75 years, cross‐sectional (prevalence of hematuria at baseline) and cohort (incidence of hematuria at follow‐up) (HEALS)Haematuria (dipstick test)

Cross‐sectional: 1189/6654;

cohort 5362/949

w‐As (mean 99 μg/L),

0.1–3

3–25

25–66

66–142

142–949.1

per SD increase

Cross sectional, OR (95%):

1.00 (Ref)

1.07 (0.87–1.31)

0.95 (0.76–1.17)

1.30 (1.06–1.59)

1.66 (1.37–2.02)

1.20 (1.13–1.27) p < 0.01

Cohort, HR (95%):

1.00 (Ref)

1.10 (0.88–1.37)

0.96 (0.77–1.20)

1.05 (0.84–1.31)

1.34 (1.09–1.65)

1.10 (1.04–1.16),

p < 0.01

Adjusted for age, BMI, cigarette smoking status, education length, SBP, DBP, occupational dye exposure and change in u‐As before incident visit

Peters et al. (2014), Bangladesh/mean 36 years,

cross‐sectional (HEALS)

Renal function using plasma cystatin C and calculated the estimated glomerular filtration rate (eGFR)Total 478, CKD stages 1–5: 95w‐As (mean 96.5 μg/L)

eGFR prediction and log u‐As B(SE): 22.55 (1.44), p: NS, r‐square: 29.

eGFR prediction and log water‐As B(SE): 20.67 (0.56), p: NS, r‐square: 29.2

Adjusted for log(age), sex, current smoking, log (u‐creatinine), recruitment year. Also data for urinary methylated metabolites
Peters, Hall, et al. (2015), Peters, Liu, et al. (2015), Bangladesh/30–65 years, cross‐sectional (FOX)Renal function using plasma cystatin C and calculated the estimated glomerular filtration rate (eGFR)

Total 374

eGFR< 90 versus > 90: 153/222

w‐As (mean 138 μg/L),

w‐As (mean 138 μg/L)

Nean change (95% CI) in eGFR (mL/min/1.73 m2) for a 10% increase in w‐As: −0.03 (−0.12, 0.07), p: NS; u‐As: −0.16 (−0.37, 0.04), p: NS

GFR < 90 versus > 90: mean (SD)

w‐ As μg/L: 1legend 40.67 (129.4) versus 135.9 (120.1), p: NS;

u‐As μg/L: 248.1 (215.3) versus 219.4 (196.4), p: NS

Adjusted for log (age), sex, log (BMI), ever smoking. Also, data for glutathione

Abbreviations: As, arsenic; BMI, body mass index; CI, confidence interval; CKD, chronic kidney disease; DBP, diastolic blood pressure; eGFR, estimated glomerular filtration rate; ESRD, end stage renal disease; FOX, Folate and Oxidative Stress; GFR, glomerular filtration rate; HEALS, Health Effect of Arsenic Longitudinal Study; HR, hazard ratio; MG, macroglobulin; n, number; NAG, N‐acetyl‐b‐D‐glucosaminidase; NS, not significant; OR, odds ratio; Q, quantile; ref, reference; SBP, systolic blood pressure; SD, standard deviation; SE, standard error; SMR, standardised mortality ratio; tiAs, total inorganic arsenic; USA, United States of America; u‐As, urinary arsenic; u‐tiAs, total urinary inorganic arsenic (sum of iAs and its methylated metabolites MMA and DMA); vs., versus; w‐As, water arsenic.

3.2.2.12. Other effects

For the present Opinion, the CONTAM Panel identified, from a literature search, 26 studies on associations between As exposure and effects other than those discussed above from a literature search (see Section 2 on methodology). Out of these, 12 studies did not meet the inclusion criteria (Akbal et al., 2014; Burchiel et al., 2014; Cárdenas‐González et al., 2016; Jimenez‐Cordova et al., 2018; Jochem et al., 2016; Gong et al., 2015; Linares et al., 2021; Galvez‐Fernandez et al., 2021; Osorio‐Yáñez et al., 2021; Ruan et al., 2022; Rahman, Islam, et al., 2021; Wang, Ding, et al., 2021).

Fourteen studies that fulfilled the eligibility criteria were further considered (Table 32).

TABLE 32

Key epidemiological studies on other effects and As exposure.

Reference, study population age, designOutcome definitionPopulation size (n) case/controlArsenic concentration/exposureResultsAdditional information/confounders
Mortality
Sohel et al. (2009), Bangladesh/> 15 years, cohort studyCancer, cardiovascular mortality, infectious, nonaccidental mortalityDeaths/survived: 9015/106888

w‐As (μg/L) (average in well water)

< 10

10–49

50–149

150–299

> 300

HR (95% CI) for cancer mortality:

1.00 (ref),

1.10 (0.77–1.59),

1.44 (1.06–1.95),

1.75 (1.28–2.40),

1.56 (1.06–2.30), p trend: 0.007.

HR (95% CI) for cardiovascular mortality:

1.00 (ref),

1.03 (0.82–1.29),

1.16 (0.96–1.40),

1.23 (1.01–1.51)

1.37 (1.07–1.77), p trend: 0.026.

HR (95% CI) for infectious mortality:

1.00 (ref),

1.09 (0.92–1.30)

1.30 (1.13–1.49),

1.51 (1.31–1.75),

1.59 (1.33–1.91), p trend: < 0.001.

HR (95% CI) for nonaccidental mortality:

1.00 (ref),

1.16 (1.06–1.26),

1.26 (1.18–1.36),

1.36 (1.27–1.47),

1.35 (1.23–1.48), p trend: < 0.001

Adjusted for age, sex, asset score, education
Wade et al. (2009), China/age data not reported, cross sectional studyAll‐cause mortality, cancer mortality, heart disease mortality, stroke mortality

Deaths/

survived: 572/12600

w‐As,

for 50 μg/L As increase

IRR (95% CI) and all‐cause mortality: 1.02 (0.94, 1.11), p: NS;

cancer mortality:

1.07 (0.89, 1.28), p: NS;

heart disease:

1.12 (1.01, 1.23), p: 0.034;

stroke:

0.82 (0.65, 1.03), p: NS

Adjusted for age, sex, education, smoking, drinking, farm work. Data also for different time of exposure
Argos et al. (2010), Bangladesh/18–75 years, cohort study (HEALS)All‐cause mortality, mortality due to chronic diseaseDeaths/survived: 407/11339

w‐As (μg/L)

0–10,

10.1–50,

50.1–150,

> 150

HR (95% CI) for all‐causemortality:

1.00 (ref),

1.34 (0·99–1·82),

1.09 (0·81–1·47),

1.68 (1·26–2·23).

HR (95%CI) for chronic‐diseasemortality:

1.00 (ref),

1.33 (0·94–1·87),

1.22 (0·87–1·70),

1.68 (1·21–2·33)

Adjusted for age, sex, body‐mass index, systolic blood pressure, education, smoking status. Data also for As dose and relation to change in u‐As over time
Rahman et al. (2013), Bangladesh/5–18 years, cohort (7‐years follow‐up)All‐cause mortality, cancer and cardiovascular mortalityDeaths/survived: 185/58221

Baseline w‐As (μg/L)

< 10

10–50

51–150

151–300

> 300

Cumulative As exposure (μg‐years/L)

< 1000

1000–4000

> 4000

HR (95% CI) for baseline w‐As and all‐cause mortality:

1.00 (ref),

1.13 (0.65–1.96),

0.81 (0.45–1.46),

1.35 (0.92–1.97),

1.51 (1.01–2.23), p trend < 0.05.

HR (95% CI) for cumulative w‐As and all‐cause mortality:

1.00 (ref),

1.17 (0.84–1.65),

1.90 (1.25–2.89), p trend < 0.05.

HR (95%CI) for baseline w‐As and cancer and cardiovascular mortality:

1.00 (ref),

1.53 (0.51–4.57),

1.29 (0.43–3.87),

2.18 (1.15–4.16), p trend < 0.05.

Adjusted for baseline age, educational attainment, socioeconomic status. Data also for time‐weighted lifetime average arsenic in well water.
Rahman et al. (2019), Bangladesh/5–18 years, cohort (13‐years follow‐up)Cancer and cerebrovascular‐cardiovascular‐respiratory mortality

Deaths/survived: cancer 48/58358;

Cerebro

vascular‐cardiovascular‐respiratory: 40/58366

Current w‐ As (μg/L)

< 90.9

90.9–223.1

> 223.1

cumulative w‐As exposure (μg‐years/L)

< 1013.3

1013.3–2711.0

> 2711

average or lifetime average As in well water (μg/L)

< 90.9

90.9–223.1

> 223.1

HR (95% CI) for average w‐As and cancer mortality:

1.00 (ref),

1.2 (0.6–2.3),

0.8 (0.4–1.6), p trend: NS.

HR (95% CI) for cumulative w‐As and cancer mortality:

1.00 (ref),

0.9 (0.5–1.8),

0.7 (0.3–1.5), p trend: NS.

HR (95%CI) for average w‐As and cardio‐cerebro‐respiratory mortality:

1.00 (ref),

2.7 (1.0–7.6),

4.8 (1.8–12.8), p trend: 0.01.

HR (95%CI) for cumulative water As and cardio‐cerebro‐respiratory mortality:

1.00 (ref),

4.0 (1.3–12.0),

5.1 (1.7–15.1), p trend: 0.01

Adjusted for baseline age, sex, educational attainment, socioeconomic status. Data also for time‐weighted lifetime average arsenic in well water.
Endocrine effects
Rahman, Kippler, et al. (2021), Bangladesh/1–15 years, birth cohort study (MINIMat/AsMat)Age at menarcheTotal 809

w‐As, median 80 μg/L (IQR 2–262).

< 10

10–49

50–99

100–199

HR (95% CI):

1.0 (Ref),

0.82 (0.59–1.14),

0.89 (0.58–1.36),

0.82 (0.64–1.05)

Adjusted for maternal socioeconomic status, education, BMI. Data also for As level > 200 μg/L
Viral infections serology

Cardenas, Smit, et al. (2015), USA/6–49 years,

cross‐sectional (NHANES 2003–2004, 2009–2010)

Seroprevalence of VZV IgG antibody

Seropositive/

seronegative

3250/98

u‐tiAs (1) geometric mean 6.57 μg/L (95% CI: 6.26, 7.9).

u‐As minus AsB / AsC (2) geometric mean (total As minus AsB and AsC) 5.64 μg/L (95% CI: 5.20, 6.12).

1‐unit increase in ln‐transformed u‐As (1) (μg/L)

1‐unit increase u‐As (2)

u‐As (1) GM (SE) seropositive versus seronegative: 6.77 (1.02) versus 8.31 (1.08), p: 0.01;

u‐As (2) GM (SE) seropositive versus seronegative: 5.85 (1.04) versus 7.62 (1.12), p: 0.02.

OR (95% CI):

1.87 (1.03, 3.44)

1.40 (1.00, 1.97)

Adjusted for age, sex, race, family poverty–income ratio, BMI classification, ln urinary creatinine, survey cycle.

Cardenas et al. (2016), USA/> 6 years,

cross‐sectional study (NHANES 2003–2012)

Hepatitis A antibodies (total anti‐HAV: IgG and IgM)

Seropositive/

seronegative 5064/6028

u‐tiAs (1) geometric mean 6.34 μg/L (SE 1.01).

u‐tAs minus AsB, AsC (2) geometric mean (total As minus AsB and AsC) 5.06 μg/L (SE 1.02)

1‐unit increase in ln‐transformed u‐As (1) (μg/L)

1‐unit increase u‐As (2)

OR (95% CI):

> = 2 doses vaccination 1·42 (1·11–1·81);

< 2 doses: 1·46 (0·83–2·59;

0 dose: 1·12 (0·98–1·30);

unknown vaccination history: 1·75 (1·22–2·52),

> = 2 doses vaccination: 1·17 (1·04–1·31)

< 2 doses: 1·26 (0·95–1·67)

0 dose: 1·07 (0·99–1·16)

unknown vaccination history: 1·20 (0·97–1·48)

Adjusted for log‐transformed creatinine, age, sex, race, family income/poverty ratio, country of birth, body mass index and survey year.

Cardenas et al. (2018), USA/> 6 years,

cross‐sectional study (NHANES 2003–2014)

Hepatitis B antibodies (total anti‐HAV: IgG and IgM)

Current HBV/

past HBV infection/

Vaccinated/

seronegative 33/636/3491/

8520

u‐As: DMA residual adjusted for arsenobetaine

For 1‐unit increase in ln‐transformed u‐As (DMA residual)

OR (95% CI):

for natural infection versus susceptible: 1.40 (1.15, 1.69);

natural infection versus vaccine induced immunity: 1.65 (1.34, 2.04);

seronegative versus vaccine induced immunity: 1.18 (1.05, 1.33)

Adjusted for log‐transformed creatinine, age, sex, race, family poverty‐income ratio, country of birth, BMI, survey year, log‐transformed serum cotinine, recent seafood consumption, self‐ reported Hepatitis B immunisation. Data also for how many doses of vaccination.

Hepatic effects
Islam et al. (2011), Bangladesh/15–60 years, cross‐sectional studyAST, ALT, ALPQuartiles Q1/Q2/Q3/Q4: 50/50/50/50

w‐As (μg/L):

Q1: 0.11–24.7,

Q2: 34–142,

Q3: 145–242,

Q4:249–546

hair As:

Q1: 0.05–1.43,

Q2: 1.57–2.80,

Q3: 2.81–5.52,

Q4: 5.66–37.24

nail As:

Q1: 0.15–3.17,

Q2: 3.21–6.25,

Q3: 6.26–11.27,

Q4: 11.4–37.42

Coefficient (95% CI) for w‐As and ALP:

1.0 (ref),

0.039 (−0.008, 0.085),

0.087 (0.041, 0.134),

0.165 (0.119, 0.212).

W‐As and AST:

1.0 (ref),

0.025 (−0.020, 0.070),

0.023 (−0.022, 0.067),

0.124 (0.080, 0.169).

w‐As and ALT:

1.0 (ref),

0.060 (−0.011, 0.132),

0.071 (0.00, 0.143)

0.186 (0.115, 0.257).

Hair As and ALP:

1.0 (ref),

0.028 (−0.023, 0.078),

0.044 (−0.005, 0.093),

0.117 (0.066, 0.168).

Hair As and AST:

1.0 (ref),

0.028 (−0.020, 0.075),

0.054 (0.008, 0.099),

0.096 (0.049, 0.144).

Hair As and ALT:

1.0 (ref),

0.064 (−0.010, 0.139),

0.086 (0.014, 0.158),

0.151 (0.076, 0.225).

Nail As and ALP:

1.0 (ref),

0.037 (−0.014, 0.088),

0.057 (0.005, 0.110),

0.109 (0.055, 0.163).

Nail As and AST:

1.0 (ref),

−0.007 (−0.055, 0.040),

0.010 (−0.039, 0.060),

0.062 (0.012, 0.113).

Nail As and ALT:

1.0 (ref),

0.043 (−0.033, 0.118),

0.055 (−0.024, 0.133),

0.084 (0.004, 0.165)

Adjusted for age, sex, BMI, smoking habit, skin lesions
Frediani et al. (2018), USA/> 12 years, cross‐sectional study (NHANES 2005–2014)ALT as a biomarker for NAFLDNAFLD yes/no: 2713/5805

u‐tAs minus AsB/AsC arsenocholine)

μg/L

Q1: < 2.42

Q2: 2.42–4.08

Q3: 4.08–6.99

Q4: > 6.99

1.00 (ref)

1.2 (0.7, 1.9),

1.6 (1.0, 2.8),

2.0 (1.2, 3.4)

Adjusted for age, survey cycle, gender, PIR, BMI Weight status, race/ethnicity.
Hearing loss
Li et al. (2018), Bangladesh/23–45 years, cross‐sectional studyHearing thresholdTotal 145, high versus low toenail As group: 97/48

Toenail As

High > 0.6 μg/g

Low < 0.6 μg/g

High versus low OR (95% CI):

1.28 (0.43–3.83) for 1 kHz,

4.27 (1.51–12.05) for 4 kHz,

3.91 (1.47–10.38) for 8 kHz,

4.15 (1.55–11.09) for 12 kHz

Adjusted for age, sex, smoking history, BMI.
Shokoohi et al. (2021), Iran/10–49 years, cross‐sectionalHearing threshold (Pure Tone Audiometry)Exposed/non‐exposed 120/120

w‐As (μg/L)

non‐exposed: 0.179

exposed: 200 (region A), 76.6 (region B), 74.5 (region C)

Exposed versus non‐exposed OR (95% CI):

2.03 (0.96–4.31) overall,

1.49 (0.24–9.18) for 2 kHz,

2.07 (0.92–4.62) for 4 kHz,

1.85 (0.74–4.66) for 8 kHz

Adjusted for age, smoking.
Allergy
Tsai, Wang, et al. (2021), Taiwan/4 years, birth cohort study (TMICS)Atopic dermatitis (questionnaire)Dermatitis yes/no: 110/260

u‐tiAs (inorganic) 30 trimester of pregnancy

2‐fold increase u‐As

Dermatitis versus control u‐As μg/g creatinine median (IQR):

30.91 (27.51–37.25) versus 28.62 (25.97–33.14), p < 0.001.

OR (95% CI):

2.42 (1.33–4.39)

Adjusted for child's sex, parental allergies, geographic area, exposure to tobacco smoke during pregnancy, exposure to tobacco smoke at age 4 years, maternal educational level. Data also for sensitivity analysis based on the existence of maternal allergy.

Abbreviations: ALP, alkaline phosphatase; ALT, alanine aminotransferase; As, arsenic; AsB, arsenobetaine; AsC, arsenocholine; AsMat, Health Consequences of Arsenic in Matlab; AST, aspartate transaminase; BMI, body mass index; CI, confidence interval; DMA, sum of dimethylarsinous acid and dimethylarsinic acid; GM, geometric mean; HAV, hepatitis A virus; HBV, hepatitis B virus; HEALS, Health Effects of Arsenic Longitudinal Study; HR, hazard ratio; IgG, immunoglobulin G; IgM, immunoglobulin M; IQR, interquartile range; IRR, incidence rate ratio; kHz, kilohertz; MINIMat, Maternal and Infant Nutrition Interventions in Matlab; MMA, sum of monomethylarsonous acid and monomethylarsonic acid; n, number; NAFLD, non‐alcoholic fatty liver disease; NHANES, National Health and Nutrition Examination Survey; NS, not significant; OR, odds ratio; PIR, poverty income ratio; Q, quartile; ref, reference; SE, standard error; tAs, total arsenic; TMICS, Taiwan Maternal and Infant Cohort Study; u‐As, urinary arsenic; u‐tiAs, urinary total inorganic arsenic (sum of iAs and its methylated metabolites MMA and DMA); USA, United States of America; VZV, Varicella‐Zoster Virus; w‐As, water arsenic.

Mortality

In a cohort study in Bangladesh in adults chronic As exposure through drinking water was associated with an increase in the mortality rate (Argos et al., 2010).

In a cohort study with a 7‐years follow‐up in Bangladesh in individuals 5–18 years, water As exposure was associated with increased risk of deaths from all‐causes, cancer and cardiovascular‐related conditions (Rahman et al., 2013).

In the same cohort with a 13‐years follow‐up, higher As concentration in water increased the risk of mortality due to cerebrovascular‐, cardiovascular‐ and respiratory disease but no association was found for cancer related mortality (Rahman et al., 2019).

In a cohort study in Bangladesh in subjects > 15 years of age, As exposure through drinking water was associated with excess mortality after 20–30 years of exposure (Sohel et al., 2009).

In a cross‐sectional study in China heart disease mortality but not all‐cause‐, cancer‐ or stroke related mortality was associated with As exposure in water (Wade et al., 2009).

Endocrine effects

In a birth cohort in Bangladesh in children < 15 years of age increased levels of prenatal As exposure in water were associated with older age at menarche (2.8 months delay), with exposure above Bangladesh's acceptable As level (≥ 50 μg/L) (Rahman, Kippler, et al., 2021).

Viral infections serology

In a cross‐sectional analysis from NHANES 2003–2004, 2009–2010 in US individuals 6–49 years, U‐As was inversely associated with Varicella zoster virus (VZV) IgG seroprevalence (Cardenas, Smit, et al., 2015).

In a cross‐sectional study from NHANES 2003–2012 in US individuals > 6 years old, As exposure was positively associated with the seroprevalence of total anti‐hepatitis A virus (HAV) in participants receiving > 2 doses of HAV vaccine and for individuals unable to recall their immunisation history (Cardenas et al., 2016).

In a cross‐sectional study from NHANES 2003–2014 in US individuals > 6 years, higher u‐As levels were associated with a greater odd of having a serological classification consistent with a past (natural) hepatitis B infection (HBV) (Cardenas et al., 2018).

Hepatic effects

In a cross‐sectional study in Bangladesh in individuals aged 15–60 years, a significant association between As exposure (assessed in water, hair, nail) and serum hepatic enzymes activity (alanine transaminase (ALT), argininosuccinate lyase (ASL), alkaline phosphatase – ALP) was found (Islam et al., 2011).

In a cross‐sectional analysis of NHANES 2005–2014 in US subjects > 12 years of age there was a positive association between U‐As exposure and risk of non‐alcoholic fatty liver disease (NAFLD) estimated by increased levels of ALT (Frediani et al., 2018).

Hearing loss

In a cross‐sectional study in young adults in Bangladesh, As in toenails but not in urine was associated with hearing loss (Li, Ohgami, et al., 2018).

In a cross‐sectional study in adolescents and young adults in Iran, As in water was not significantly associated with hearing loss (Shokoohi et al., 2021).

Allergy

In a birth cohort in Taiwan in children aged < 4 years, prenatal exposure assessed by u‐As was associated with a higher risk of atopic dermatitis in young children, most evident when the mothers had a history of dermatitis (Tsai, Wang, et al., 2021).

In summary, there is insufficient evidence for a causal association between low to moderate exposure to iAs and liver function, endocrine function (not covered in other outcomes mentioned previously), hearing loss, allergy, anaemia and reduced bone mineral density. For these outcomes the number of studies is very limited. Regarding mortality the major disease groups affecting mortality, such as cancer, ischemic heart disease, stroke, chronic kidney disease and infant mortality have already been assessed in previous sections. Therefore, it is not relevant to consider separately the evidence for all‐cause mortality.

3.2.3. Critical effects

In the sections on Observations in humans (Section 3.2.2), the CONTAM Panel assessed a large number of epidemiological studies on different adverse health outcomes. Most of these studies were not available for the previous opinion in 2009. Exposure was based on As in drinking water, urine, toenails or in hair.

The following outcomes were considered as potential critical effects, because for these adverse effects the evidence of association with exposure to low to moderate levels of iAs was considered sufficient to assume causality and because they were considered biologically plausible: lung cancer, bladder cancer, skin cancer, skin lesions, decreased birth weight, spontaneous abortion, stillbirth, infant mortality, congenital heart disease, neurodevelopmental effects, ischemic heart disease and atherosclerosis. Additional information about biologically plausible mechanisms per endpoint is provided in the text below.

3.2.3.1. Skin, bladder and lung cancer

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and increased risk of basal and squamous cell carcinomas of the skin, bladder and lung.

There are several biologically plausible mechanisms for arsenic causing cancer in the tissues where it is metabolised, distributed to or excreted. iAs can induce DNA damage via generation of RONS (reactive oxygen and nitrogen species) and interferes with genome maintenance systems. iAs has been shown to increase the frequency of CA and MN in chronically exposed populations, to cause epigenetic alterations (e.g. changes in DNA‐methylation, histone post‐translational modifications), to contribute to the epithelial‐to‐mesenchymal transition during cell transformation and to target key signalling pathways associated with cancer (see Sections 3.1.2. Biomarkers and 3.1.3. Genotoxicity).

These genotoxic and epigenetic effects have been detected in a variety of in vitro and in vivo model systems including cells relevant for these cancer types, such as primary human keratinocytes and lung cells, and exfoliated urothelial cells derived from exposed subjects where increased frequency of MN has been reported. Moreover, there are characteristic features of arsenic‐induced skin and lung cancer. Bowen's disease is the most common skin cancer induced by arsenic. Cases of Bowen's disease related to arsenic exposure tend to have multi‐focal cancer in sun‐protected skin (Yu et al., 2006), whereas cases not related to arsenic exposure mainly have solitary cancer in sun‐exposed skin (Yu et al., 2006). Recently a compelling study (Speer et al., 2023), has provided evidence that iAs acts as a powerful co‐mutagen and co‐carcinogen alongside UV radiation in a mouse skin carcinogenesis model. By examining a group of human basal cell carcinomas and melanomas, this study has successfully identified, within a subset of tumours, the same distinctive mutational pattern resulting from the combined exposure to these two environmental carcinogens in the mouse model. The carcinogenicity of iAs on lung seems to be cell‐type specific: iAs‐related lung tumours are more often squamous cell carcinomas (Kuo et al., 2017). In addition, there is some preliminary evidence of specific genetic and epigenetic changes in tumours seen after iAs exposure when compared with histologically matched tumours that developed in an iAs‐free environment (reviewed in Hubaux et al., 2013).

The CONTAM Panel considers the association between low to moderate exposure to iAs and cancers of the skin, bladder and lung to be causal.

3.2.3.2. Skin lesions

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and increased risk of skin lesions, i.e. pigmentation changes and hyperkeratosis.

Arsenic tends to accumulate in the skin and there are several biologically plausible mechanisms for arsenic causing skin lesions. Some of the mechanisms are overlapping with skin cancer, since arsenic‐related skin lesions are a risk factor for skin cancer. The CONTAM Panel considers the association between low to moderate exposure to iAs and skin lesions to be causal. It should be noted that because these studies were performed in low and medium income regions in Bangladesh and China and nutrition and health status are important modifying factors, it is difficult to translate these risks to populations with more adequate nutrition such as in Europe.

3.2.3.3. Decreased birth weight

The CONTAM Panel considers the evidence from epidemiological studies performed in Bangladesh (Kile, Cardenas, et al., 2016; Rahman et al., 2009) sufficient for an association between low to moderate exposure to iAs and decreased birth weight, while results from other countries (Chile, Taiwan, Mongolia, Mexico, US) are inconsistent and the evidence therefore insufficient.

Arsenic readily crosses the placenta. Proposed mechanisms for decreased birth weight include disruption of placental vascular function, inflammation, RONS formation and epigenetic effects on the fetus (Bloom et al., 2014). The CONTAM Panel considers this association to be causal. However, the average birth weight in Europe is much higher and undernutrition is less common. Therefore, it is uncertain if an association between iAs exposure and birth weight is present in European populations.

3.2.3.4. Spontaneous abortion and stillbirth

The CONTAM Panel considers the evidence from epidemiological studies performed in Bangladesh sufficient for an association between low to moderate exposure to iAs and increased risk of spontaneous abortion and stillbirth. Embryotoxic effects at high doses of iAs have been shown in animal studies (Wang et al., 2006). Specific mechanisms proposed regarding spontaneous abortion and stillbirth include enzyme inhibition due to thiol reactivity, and cytotoxicity due to altered cell–cell signalling and RONS generation (Milton et al., 2017). The mechanisms mentioned above regarding birth weight are also relevant. The CONTAM Panel considers the association between exposure to arsenic and spontaneous abortion and stillbirth to be causal. However, as discussed above on birth weight, the relevance for Europe is unclear since maternal and fetal undernutrition, low birth weight (BW) and preterm birth (PTB) in Bangladesh is much more common than in Europe. These factors are likely mediators, increasing the risk of spontaneous abortion and stillbirth.

3.2.3.5. Infant mortality

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and increased risk of infant mortality, both in the first 28 days (neonatal period) and later. Associations have been shown in Bangladesh, Mongolia and Chile. Decreased birth weight and preterm birth likely contribute to increased risk of infant mortality. The same is true for effects of iAs on lung function and possible risk of respiratory disease in infancy. Therefore, the CONTAM Panel considers the association of iAs exposure and infant mortality to be causal.

3.2.3.6. Congenital heart disease

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and increased risk of congenital heart disease. The key epidemiological studies by Rudnai et al. (2014) from Hungary and by Richter et al. (2022) from Denmark, are large and of good quality. However, the CONTAM Panel notes that the exposure‐response relation between water As concentrations and risk of congenital heart disease seemed to be supralinear. In the study by Richter et al. (2022) most of the increase in risk was reported from the reference category (< 0.5 μg/L) up to a few μg/L, and in the study by Rudnai et al. (2014), the increased risk was mainly found when mothers' drinking water As was 10–30 μg/L.

The teratogenicity of iAs in experimental animals is well established (EFSA CONTAM Panel, 2009), but the mechanisms are not well characterised. At least two studies in experimental animals have shown heart malformations, mainly septum defects (Lin, Zhuang, et al., 2018; Na et al., 2020). Folate supplementation protected against heart malformations in both these studies. With the support from experimental studies, it seems reasonable to assume that the associations reported in humans are causal.

3.2.3.7. Neurodevelopmental effects

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and effects on cognitive function during childhood.

Several biologically plausible mechanisms for impaired neurodevelopment, including cognition, have been described such as epigenetic alterations (e.g. changes in DNA methylation profiles and histone post‐translational modifications) alterations in neurotransmitter homeostasis, oxidative stress, impaired DNA repair, mitochondrial dysfunction and neuroinflammation (reviewed in Thakur et al., 2021). The CONTAM Panel considers the association between low to moderate exposure to iAs and impaired cognition during childhood to be causal.

3.2.3.8. Ischemic heart disease

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and ischemic heart disease.

Several biologically plausible mechanisms have been described, e.g. inflammatory responses, increased ROS in endothelial cells and signs of compromised endothelial‐dependent vasodilatation (Chowdhury et al., 2018; Ellinsworth, 2015; Karachaliou et al., 2021). The CONTAM Panel considers the association between low to moderate exposure to iAs and ischemic heart disease to be causal.

3.2.3.9. Carotid artery atherosclerosis

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and carotid artery atherosclerosis. As shown in Table 26 of Section 3.2.2, several of the studies show associations between As in water or urine and carotid artery plaque or intima media thickness (IMT). This is biologically plausible, and likely mechanisms are endothelial dysfunction and injury in the arterial vessels, in line with the well‐known risk of so‐called Blackfoot disease at high iAs exposure. Endothelial damage and inflammation increase the risk of plaque formation (Karachaliou et al., 2021). Carotid artery atherosclerosis increases the risk of ischemic stroke and myocardial infarction (Spence et al., 2002). The CONTAM Panel considers the association between low to moderate exposure to iAs and carotid artery atherosclerosis to be causal.

3.2.3.10. Lung function

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and impaired lung function, in children as well as in adults. Several biologically plausible mechanisms have been described, including cellular injury caused by inflammation and increased ROS generation (Sanchez et al., 2018). The CONTAM Panel considers the association between low to moderate exposure to iAs and impaired lung function to be causal.

3.2.3.11. Chronic kidney disease

The CONTAM Panel considers the evidence from epidemiological studies sufficient for an association between low to moderate exposure to iAs and risk of CKD. Several mechanisms for arsenic‐mediated nephrotoxicity have been described including the production of ROS and other free radicals, inflammation and apoptosis (Robles‐Osorio et al., 2015). The CONTAM Panel considers the association with iAs exposure to be causal.

3.2.4. Dose–response analyses

3.2.4.1. Previous dose response analyses

In the previous Opinion on arsenic (EFSA CONTAM Panel, 2009) the data for cancers of the bladder, lung and skin, and skin lesions were considered as possibly providing an appropriate Reference Point.

In the previous Opinion on arsenic, it was noted that the EFSA guidance on the use of benchmark dose modelling (EFSA CONTAM Panel, 2009) provided neither guidance on an appropriate BMR for human data nor on calculation of confounder‐adjusted BMDs and BMDLs. Because the individual data from the studies were not available to the CONTAM Panel, it estimated a 1% extra risk (and its lower 95% CI) based on the unadjusted incidence data reported. The Panel noted that although it would have been possible to estimate a 5% or 10% extra risk, a 1% extra risk would be within the observed data range and was therefore used for dose response modelling.

The CONTAM Panel calculated BMCLs for two studies on dermal lesions considered as potentially relevant (in all of which the exposure metric was iAs concentrations in drinking water) and used BMCLs for lung and bladder cancer calculated by NRC (2001) and for skin cancer identified a so‐called change point for the water level based on Karagas et al. (2002). These BMCLs were then transformed into ranges of BMDLs by applying assumptions for the possible range of daily iAs exposure via food and water consumption, together with body weights relevant to the study populations used in the different studies. Briefly, the CONTAM Panel applied a range of scenarios for estimating total dietary exposure to iAs in the study populations. For rural Asian communities, 50–200 μg iAs/day from food and 3–5 L per day for water consumption including use in cooking, were applied. For North and South American populations, the corresponding ranges were 10–20 μg/day from food and 1–2 L per day. Average body weights were assumed to be 55 kg for rural Asian populations and 70 kg for North and South America (EFSA CONTAM Panel, 2009).

For skin lesions, the studies from Ahsan et al. (2006), and Xia et al. (2009) were used for analysis. With Ahsan et al. (2006), applying the log‐logistic model as the best fitting model a BMC01 of 26.47 μg and a BMCL01 of 22.92 μg iAs/L were obtained. The corresponding BMDL01 values ranged between 2.2 and 5.7 μg iAs/kg bw per day. Modelling the results of the study by Xia et al. (2009) resulted in a BMC01 of 0.56 and a BMCL01 of 0.31 iAs/L, corresponding to a BMDL01 ranging from 0.93 to 3.7 μg iAs/kg bw per day.

Karagas et al. (2002) found an association between skin cancer (basal and squamous cell carcinoma) and toenail iAs in a US population. A regression analysis resulted in a non‐linear curve with a maximum likelihood ‘change point’ (the concentration where the dose–response started to increase, and which might be considered as a NOAEL) of 0.105 μg iAs/g (95% CI 0.093–0.219). This change point was found at a concentration of 1–2 μg iAs/L in drinking water corresponding to a ‘change point’ exposure ranging from 0.16 to 0.31 μg iAs/kg bw per day for skin cancer.

Karagas et al. (2004) found a two‐fold (statistically not significant) increase in bladder cancer in the group with the highest toenail arsenic in a study in New Hampshire (US). A regression analysis showed a non‐linear curve with a maximum likelihood change point of 0.326 μg iAs/g (95% CI 0.121–0.446 corresponding to a ‘change point’ exposure ranging from 0.9 to 1.7 μg iAs/kg bw per day.

In an analysis carried out by the NRC (2001) the data from Chiou et al. (2001) showed a significant trend for urinary tract and transitional cell carcinoma with increasing iAs concentrations in water. The BMC01 18 (and BMCL01) were 129 (42) and 281 (92) μg/L for males and females, respectively from the Chiou data (NRC, 2001), from which the EFSA CONTAM Panel estimated BMDL01 values ranging from 3.2 to 7.5 μg iAs/kg bw per day.

In the study of Ferreccio et al. (2000) increases in lung cancer cases in a Chilean population at 30–49 μg iAs/L and above were observed and the NRC (2001) calculated BMC01 (and BMCL01) values by linear regression of 17 (14) and 27 (21) μg/L for males and females, respectively. The CONTAM Panel transformed the BMCL01 of 14 μg/L (for increased lung cancer incidences in males) to a BMDL01 range of 0.34 to 0.69 μg iAs/kg bw per day.

3.2.4.2. Current dose–response analyses

Overview of the approach

The criteria for inclusion of critical effects and reasoning for inclusion of studies for dose–response analyses are described in detail in Section 3.2.3 on critical effects above. Studies that were considered for dose–response modelling had to meet three criteria: (i) the overall risk of bias was considered low, (ii) the statistical analysis on the association between iAs exposure and the risk of the outcome reported by the authors had to show a statistically significant association with iAs as a continuous variable, a statistically significant trend test and/or a statistically significant increase of risk in the upper exposure category/ies, (iii) results for at least three exposure categories (including the reference category) had to be reported. In the studies selected for dose–response modelling the CONTAM Panel first transformed iAs concentrations in drinking water and urine to daily total iAs exposures per kg/bw for use in BMD calculations. The data analysed are adjusted incidences and resulting number of cases based on the risk ratios reported in these studies and the provided (cohort studies) or provided/estimated (case–control studies) population sizes (see Section ‘Transformations of relative risk estimates to quantal data’ below). The models for quantal data are used to perform BMD analysis. As BMR the CONTAM Panel decided to use a relative increase of 5% of the background incidence after adjustment for confounders. These modelling results are used as the basis for the selection of the Reference Point.

Studies selected for dose–response modelling

Skin cancer

The studies from Leonardi et al. (2012) and Gilbert‐Diamond et al. (2013) meet the above‐mentioned criteria for dose–response modelling.

Bladder cancer

The studies by Chen et al. (2010b) and Steinmaus et al. (2013) meet the above‐mentioned criteria for dose–response modelling. The study by Huang et al. (2018) was not modelled because the study results might be influenced by the presence of high amounts of DMA in the urine.

Lung cancer

The studies from Ferreccio et al. (2000), Smith et al. (2009), Chen et al. (2010a) and Steinmaus et al. (2013), Steinmaus et al. (2014b) meet the above‐mentioned criteria for dose–response modelling. However, since the Chilean case–control studies by Steinmaus et al. (2013) and Steinmaus et al. (2014b) are larger and have better methodological quality than the studies based on the previously used case–control study by Ferreccio et al. (2000) and reanalyzed by Smith et al. (2009) and are from the same Chilean region, these results were not modelled.

Skin lesions

The studies from Ahsan et al. (2006), Xia et al. (2009) and Pierce et al. (2011) meet the above‐mentioned criteria for dose–response modelling.

Decreased birth weight

No studies were modelled in this health outcome category (See section 3.2.3.3 Decreased birth weight). Studies on associations between iAs exposure and birth weight usually present the results from regression analyses with iAs exposure as a continuous variable, rather than analyses based on exposure categories. For example, the study by Rahman et al. (2009) presents the decrease of birth weight per one μg/L of As in water (linear model) and the study by Kile, Cardenas, et al. (2016) presents the decrease of birth weight in gramme per one unit of log‐transformed water As. Therefore, normal dose–response modelling cannot be performed for these studies. In addition, these studies were performed in Bangladesh and the relevance for Europe is unclear since maternal and fetal undernutrition, low birth weight and preterm birth in Bangladesh are much more common than in Europe.

Spontaneous abortion and stillbirth

The studies by Milton et al. (2005) and Rahman et al. (2010) on spontaneous abortion meet the above‐mentioned criteria for dose–response modelling, and the same is true for the studies by Milton et al. (2005), Cherry et al. (2008) and Rahman et al. (2010) on stillbirth. However, as discussed above on birth weight, the relevance for Europe is unclear since maternal and fetal undernutrition, low BW and PTB are more common in Bangladesh than in Europe, and these factors also increase the risk of spontaneous abortion and stillbirth. Studies on birthweight were not included in dose–response modelling.

Infant mortality

The studies by Milton et al. (2005), Rahman et al. (2007) and Rahman et al. (2010) meet the above‐mentioned criteria for dose–response modelling. These studies are from Bangladesh, but associations have also been shown in Mongolia and Chile.

Congenital heart disease

The studies by Rudnai et al. (2014) from Hungary and by Richter et al. (2022) from Denmark meet the above‐mentioned criteria for dose–response modelling.

Neurodevelopmental effects

The studies by Vahter et al. (2020) and Wasserman et al. (2004, 2014) meet the above‐mentioned criteria for dose–response modelling. However, only the study by Vahter et al. (2020) was finally selected for modelling, since it is the largest and it is a longitudinal study with data both for prenatal exposure and postnatal exposure, and the dose–response may depend on the timing of exposure.

Ischemic heart disease

The studies by Moon et al. (2013), Wade et al. (2015), James et al. (2015) and Wu et al. (2015) meet the above‐mentioned criteria for dose–response modelling. The study by Monrad et al. (2017) had the lowest exposure levels, but a very narrow range of exposure. In the total data set, there was no significant dose–response and in the city of Aarhus, where there was a significant association, the median water‐As was the same (2.11 μg/L) in the third and fourth quartiles. This study was therefore not considered appropriate for dose–response modelling. The other studies using As in water, mentioned in Table 24 of this Section, were either based on the same study areas but with fewer cases (Chen, Graziano, et al., 2011; Chen, Wu, Liu, et al., 2013; Wade et al., 2009) or considered to have a high risk of bias (Butts et al., 2015; D'Ippoliti et al., 2015; Medrano et al., 2010; Zierold et al., 2004) and therefore not appropriate for dose–response modelling.

Carotid artery atherosclerosis

No studies were modelled within this health outcome category. Among the studies in a Taiwanese cohort (Table 26), the largest study by Hsieh et al. (2011) meets the above‐mentioned criteria for dose–response modelling. Since carotid artery atherosclerosis is a subclinical outcome, and there was only one study of carotid artery plaque (but several studies on atherosclerotic cardiovascular disease), it was decided not to use the study by Hsieh et al. (2011) for BMD modelling. The study of IMT by Chen, Wu, Graziano, et al. (2013) only presented data on associations with water As as a continuous variable.

Lung function

Among the studies in Table 28, the studies by Parvez et al. (2013), Powers et al. (2019) and Siddique et al. (2020) meet the above‐mentioned criteria for dose–response modelling.

Chronic kidney disease

The studies by Hsueh et al. (2009) and Zheng et al. (2015) meet the above‐mentioned criteria for dose–response modelling.

Risk of bias analysis

The risk of bias was considered already as part of the review of studies in Section 3.2.2, as described in 3.2.2.1, albeit not being carried out formally. A formal extensive comparative risk‐of‐bias assessment, however, was conducted for the studies selected for dose–response modelling (Table 33). The rationale behind this additional level of scrutiny was that a comparative assessment of the evidence providing the modelling inputs across different endpoints and health outcome categories would enhance the informed deliberations related to the derivation of reference points. For these studies, which had been already assessed as of low risk of bias, the risk of bias was classified as either low (L) or moderate (M) separately for selection bias (‘Sel’), information bias (‘Inf’) and confounding (‘Con’). In addition, the assumed direction (Dir) of the potential bias on the reported risk estimate is commented on. No specific tool was used for the risk of bias assessment. However, the general structure is in agreement with the draft SC guidance on appraisal of epi studies (EFSA Scientific Committee, 2020). An explanation of the basis for the assessment of risk of bias is provided in Annex C. Risk of bias for the studies listed in Table 33 was assessed independently by two experts, the assessments were compared, discussed and consensus was reached on all points.

TABLE 33

Summary of benchmark analysis and potential reference points for inorganic arsenic using a BMR of a 5% relative increase of the background incidence after adjustment for confounders (prevalence for the cross‐sectional studies).

Critical effectReferencePopulationStudy designMatrixInput parameters for exposure estimate 1 , 2 , 3 , 4 , 5 , 6 , 7 , 8 , 9 , 10 Results in μg t‐iAs/kg bw per day 11 Risk of bias analysis
BMDLBMDBMDUSel $ Inf & ConDir
Skin cancer Leonardi et al. (2012)Hungary, Slovakia, RomaniaLow‐tiAs10.010.050.08L/ML/LLU
Gilbert‐Diamond et al. (2013)USALou‐tiAs60.060.150.21L/LL/LLU
Bladder cancer Chen, Chiou, Hsu, Hsueh, Wu, Wang, and Chen (2010)TaiwanLow‐tiAs40.151.335.46L/LL/LMU
Steinmaus et al. (2013)ChileLow‐tiAs30.571.091.64L/LL/LMU
Lung cancer Chen, Chiou, Hsu, Hsueh, Wu, and Chen (2010))TaiwanLow‐tiAs41.214.9710.80LL/LMU
Steinmaus et al. (2013)ChileLow‐tiAs32.072.793.79L/LL/LLU
Steinmaus et al. (2014b)ChileLow‐tiAs30.170.761.71L/LL/LLU
Skin lesions Ahsan et al. (2006)BangladeshCSw‐tiAs50.080.310.60LL/LLU
Xia et al. (2009)ChinaCSw‐tiAs40.110.180.25LL/LLU
Pierce et al. (2011)BangladeshLow‐tiAs50.801.612.86LL/LLU
Spontaneous abortion and stillbirth Milton et al. (2005), stillbirthBangladeshCSw‐tiAs50.874.086.71LL/MLU
Milton et al. (2005), spontaneous abortionBangladeshCSw‐tiAs50.582.665.54LL/MLU
Cherry et al. (2008), stillbirthBangladeshLow‐tiAs51.372.022.64LM/LM
Infant mortality Rahman et al. (2007)BangladeshLow‐tiAs50.985.3412.06LL/LM
Neuro develop‐mental effects 12 Vahter et al. (2020), full developmental score, children u‐tiAs 10 yearsBangladeshCSu‐tiAs100.541.532.05LL/LLU
Vahter et al. (2020), full developmental score, mothers u‐tiAs at GW 8BangladeshLou‐tiAs71.361.812.12LL/LLU
Ischemic heart disease Wade et al. (2015)ChinaLow‐tiAs41.231.672.28LL/LLU
James et al. (2015)USALow‐tiAs20.170.480.92LL/LLU
Wu et al. (2015)BangladeshLow‐tiAs50.813.5811.51LL/LLU
Moon et al. (2013)USALou‐tiAs80.150.310.40LL/LLU
Lung function Powers et al. (2019), airflow obstructionUSA (American Indians)Lou‐tiAs80.070.170.26LL/MLU
Powers et al. (2019), restrictive patternUSA (American Indians)Lou‐tiAs80.100.230.28LL/MLU
Powers et al. (2019), emphysemaUSA (American Indians)Lou‐tiAs80.040.150.25LL/MLU
Chronic kidney disease Hsueh et al. (2009)TaiwanLou‐tiAs90.120.280.40L/MM/LL
Zheng et al. (2015)USALou‐tiAs80.060.150.24L/MM/MLU

Abbreviations: BMD, benchmark dose; BMDL, benchmark dose lower confidence limit; BMDU, benchmark dose upper confidence limit; BMR, benchmark response; bw, body weight; CS, cross‐sectional study; GW, gestational week; Con, Confounding bias; Dir, Assumed direction; L, low risk of bias; H; high risk of bias; Inf, Information bias; Lo, longitudinal (cohort, case–control, case‐cohort) study; M, medium risk of bias; Sel, Selection bias; U, unclear; USA, United States of America; u‐tiAs, urinary total inorganic arsenic; w‐tiAs, water total inorganic arsenic; ↓ underestimation of true risk (so overestimation of BMD).

$ In the case of case–control studies the ranking was carried out for cases (left side of ‘/’) and controls (right side of ‘/’) separately.
& For all studies, this was carried out separately for exposure misclassification (left side of ‘/’) and exposure misclassification/case diagnosis (right side of ‘/’). The exposure misclassification due to estimating iAs from food in studies based on As in drinking water will be more pronounced in the reference category with low water As (see section Risk of bias analysis).
1 Total daily exposures (μg/kg bw per day) were calculated assuming a default water consumption of 1.5 L per day, a default exposure via food of 7.7 μg iAs/day and a default body weight of 70 kg.
2 Total daily exposures (μg/kg bw per day) were calculated assuming a default water consumption of 1.5 L per day, a default exposure via food of 15 μg iAs/day and a default body weight of 70 kg.
3 Total daily exposures (μg/kg bw per day) were calculated based on daily water arsenic intakes, assuming a default exposure via food of 20 μg iAs/day and a default body weight of 70 kg.
4 Total daily exposures (μg/kg bw per day) were calculated assuming a default water consumption of 3 L per day, a default exposure via food of 36 μg iAs/day and a default body weight of 55 kg.
5 Total daily exposures (μg/kg bw per day) were calculated assuming a default water consumption of 4 L per day, a default exposure via food of 60 μg iAs/day and a default body weight of 55 kg.
6 Total daily exposures (μg/kg bw per day) were calculated using reported urinary concentrations and assuming, a urinary output of 2L per day and using bodyweights of 70 kg.
7 Total daily exposures (μg/kg bw per day) were calculated using reported urinary concentrations and assuming, a urinary output of 2L per day and using bodyweights of 55 kg.
8 Total daily exposures (μg/kg bw per day) were calculated by multiplying the concentration in μg creatinine/g reported by the normal creatinine excretion in 24 h in order to transform the concentration (in μg/g) to μg/24 h. The multiplication factor (normal creatinine excretion) was 1.2 for women and 1.8 for men.
9 Total daily exposures (μg/kg bw per day) were calculated by multiplying the concentration in μg creatinine/g reported by the normal creatinine excretion in 24 h in order to transform the concentration (in μg/g) to μg/24 h. The multiplication factor (normal creatinine excretion) was 1.0 for women and 1.5 for men.
10 Total daily exposures (μg/kg bw per day) were calculated using reported urinary concentrations and assuming, a urinary output of 1 L per day and using bodyweights of 23 kg (as reported in the publication).
11 A BMR of 5% (relative increase of the background incidence after adjustment for confounders) was applied to all calculations.
12 For these studies continuous data was modelled, applying 5% extra risk.

One type of information bias is misclassification of exposure, which deserves a special comment. The ideal situation for an epidemiological study is that the exposure for each study participant is known for each day over a long‐time period. The studies reviewed in this Opinion are based either on As in drinking water or on iAs in urine.

Arsenic in drinking water has generally been measured once or a couple of times in the drinking water of the study participants. The mean arsenic concentrations over time are usually relatively stable, but they can vary from day to day. In addition, the water intake may vary somewhat within individuals (between days) and between individuals. It may also have changed over the years preceding the collection of water samples. Therefore, there will always be some misclassification of long‐term intake of iAs from drinking water. For studies using As in water as exposure metric, it is also necessary to take into account the exposure from other dietary sources. This contribution from other food is known only on a group level. So when estimating the total daily dose of iAs, a fixed estimate of the intake of iAs from other food (different for different countries) must be added. In reality, the iAs intake in other food will vary between individuals. In summary, the iAs exposure in all studies based on As in water plus assumed intake from other food will be subject to misclassification. In general, non‐differential misclassification leads to a bias toward the null, so a reported increased risk can be assumed to be underestimated.

The studies using iAs in urine (including their metabolites; u‐tiAs) have the advantage that the excretion of u‐tiAs mirrors the total intake of iAs (water + other food) on the day of sampling and the day before. This is an advantage compared with the above‐mentioned studies based on As in water and an assumed intake of iAs from other food. A disadvantage of using u‐tiAs is that it mirrors exposure to iAs only in the past 1 or 2 days. This is not important when exposure is stable, but if exposure (from drinking water or other food) changed over the years preceding urine collection, this is not captured by u‐tiAs. The within‐individual variability of u‐tiAs in urine (between days) is usually small, as shown by studies using repeated sampling (Steinmaus et al., 2005). This is in contrast to total As in urine, which has a higher variability due to the impact of variable intake of organic As species (Barregard et al., 2021). The concentration of u‐tiAs in a spot sample must be transformed into an estimated 24 h u‐tiAs to estimate the daily intake of iAs. This is another source of misclassification, since a fixed estimate of the 24 h urine volume (2 L used in this Opinion for adults) is used, while in real life the average 24 h urine volume differs between individuals. In summary, as for the studies based on As in water plus an assumed intake from other food, studies based on u‐tiAs will also be subject to misclassification. It is likely that when the iAs from water is low, misclassification in studies based on u‐tiAs is somewhat smaller than in the studies based on As in drinking water. Also, for studies based on iAs in urine, the misclassification will almost always be non‐differential (not associated with the outcome) and attenuate a true association. An exception is when u‐tiAs is used in studies of CKD since then the disease can be assumed to decrease the excretion of u‐tiAs, which will partly mask a true positive association between iAs exposure and CKD, meaning that the underestimation of risk may be higher than in the case of non‐differential misclassification alone.

Conversion of water and urine iAs concentrations to inorganic arsenic exposures

The results of the epidemiological studies are based upon associations of adverse outcomes with either concentrations of arsenic measured in drinking water (w‐tiAs, which is inorganic As) or measured in urine (u‐tiAs). Unlike in the 2009 Opinion, where BMCLs were first derived and then transformed to BMDLs after adding exposure via food (EFSA CONTAM Panel, 2009), for the present Opinion the CONTAM Panel decided to follow an approach applied by JECFA (FAO/WHO, 2011) and to first transform iAs concentrations in drinking water and urine to daily total iAs exposures per kg/bw for use in BMD calculations. The CONTAM Panel considered this approach to be the most appropriate method to derive BMDs for iAs. For the derivation of default dietary exposures for studies where water concentrations were reported for the present Opinion, the EFSA CONTAM Panel refined some of the values used in the previous Opinion (EFSA CONTAM Panel, 2009), taking into account additional information. For studies with populations from Europe, North and South America an average bodyweight of 70 kg was assumed while for Asian populations (China, Bangladesh) the corresponding value was 55 kg. These values are the same as those applied by the EFSA CONTAM Panel (2009) and JECFA (FAO/WHO, 2011).

For studies, where iAs concentrations in water were reported, dietary exposures were used as follows.

For European populations, for average iAs exposure via food, a value of 7.7 μg iAs/day was assumed. This value is based on the UB median mean exposure of European adults of 0.11 μg iAs/kg bw per day estimated by EFSA in the recent report on iAs dietary exposure (EFSA, 2021; see also Table 34 of Section 3.5.1) and a body weight of 70 kg.

TABLE 34

Summary statistics of the dietary chronic mean and P95 exposure assessment (μg/kg bw per day) to iAs across European dietary surveys. Estimates were rounded to two decimal places.

Mean dietary exposure (μg/kg bw per day)
Lower bound (LB)Upper bound (UB)
N MinMedianMaxMinMedianMax
Infants 130.090.150.220.260.420.61
Toddlers 160.120.170.300.340.440.61
Other children 190.070.110.170.190.300.37
Adolescents 200.040.060.110.100.160.23
Adults 220.030.040.070.080.110.15
Elderly 200.030.030.060.060.100.14
Very elderly 150.030.030.050.080.100.14
Pregnant women 50.040.060.070.100.130.14
Lactating women 20.030.060.090.14
95th percentile dietary exposure (μg/kg bw per day)
Lower bound (LB) Upper bound (UB)
N Min Median Max Min Median Max
Infants 130.210.360.520.760.841.20
Toddlers 160.240.370.580.620.750.99
Other children 190.170.260.370.410.540.67
Adolescents 200.100.140.260.210.300.44
Adults 220.070.100.190.160.210.33
Elderly 200.060.080.140.130.180.25
Very elderly 150.070.080.140.140.170.23
Pregnant women 50.090.120.190.170.220.28
Lactating women 20.080.140.160.25

Abbreviations: bw, body weight; LB, lower bound; N, number; UB, upper bound.

For U.S. populations (only the study of James et al., 2015), a value of 15 μg iAs/day for average exposure via food was assumed based on information provided in a study also dealing with a population from Colorado from the same authors (James et al., 2013). In that paper, estimating exposures based on drinking water As concentrations, the group with the lowest w‐As concentrations (≤ 2 μg/L) had a mean u‐tiAs of 11.3 μg/g creatinine. Assuming a water concentration of 1 μg/L iAs, and taking into account 24 h creatinine excretion, the CONTAM Panel estimated exposure from diet of about 15 μg/day.

For Chilean populations, a value of 20 μg iAs/day has been applied for average exposure via food based on the estimates from Sancha (2000) and Muñoz et al. (2005). Sancha (2000) estimated an iAs dietary exposure of 13 μg/day for the inhabitants of urban populations of Northern Chile while Muñoz et al. (2005) estimated a substantially higher exposure of 39 μg iAs/day for the inhabitants of Santiago de Chile. In addition, Díaz et al. (2004) studied the dietary intake of iAs in a rural village in Northern Chile and ended up with a dietary exposure estimate ranging from 38 to 58 μg iAs/day. The CONTAM Panel concluded that a lower estimate of 20 μg iAs/day was most appropriate for the Chilean studies considered for risk characterisation. This was mainly based on two publications. In the publication of Ferreccio and Sancha (2006) it was noted that the relatively high iAs levels estimated in food in the studies by Díaz et al. (2004) was only relevant for a certain population of indigenous people in the Andes. In the second publication Muñoz et al. (2005) transformed the measured total As concentrations to iAs levels with a single conversion factor of 50%, which was assumed to overestimate the true dietary exposure of the Chilean population.

For populations from Taiwan/China, an average dietary exposure of 36 μg iAs/day was assumed. This estimate is based on a dietary intake estimate of 0.65 μg iAs/kg per day from a survey of Taiwanese eating habits carried out by the Taiwan Department of Health (TDOH, 2007) reported in the publications by Mendez et al. (2020) and Allen et al. (2020), and assuming a body weight of 55 kg.

Finally, for populations from Bangladesh, a value of 60 μg iAs/day as average exposure via food was used based on an estimate published by Vahter et al. (2006). There, the authors found a significant association between arsenic in urine and drinking water for pregnant women in gestational weeks 6–8 (linear regression: ln UAs = 3.0 + 0.70 WAs0.2, R 2 = 0.39, p < 0.001). The ratio U‐As to water As was 76. The average dietary exposure of the inhabitants of Bangladesh was then estimated from this linear regression, assuming a urine volume of 2 L and a water intake of 4 L. In support of this estimate, Lindberg et al. (2008) calculated that the contribution to urinary arsenic from arsenic exposure from food and other water sources was about 45 μg/L for populations from Bangladesh, including both men and women of wide age ranges. Overall, the estimate of 60 μg/day is uncertain.

An average water consumption of 1.5 L per day was assumed for populations in Europe, North and South America (i.e. the mid‐point of the range assumed in the EFSA CONTAM Panel, 2009). The EFSA guidance on default values (EFSA Scientific Committee, 2012) recommends a 2 L default value for chronic daily total liquid intake (including milk, tap water and other beverages). In the epidemiological studies selected, only iAs concentrations in drinking water were reported. The CONTAM Panel considered it justified to apply a consumption value of 1.5 L per day for European, US and Chilean populations, unless other data were provided by the authors. The corresponding values for China/Taiwan and Bangladesh were 3 and 4 L respectively (FAO/WHO, 2011). The JECFA noted that water consumption can vary greatly based on for instance, region, temperature and food consumption. In particular, when the habitual diet includes foods like rice that take up large quantities of water, the total water consumption can increase from 1.5 L up to 5 L per day (FAO/WHO, 2011).

For studies where concentrations of u‐tiAs were provided in μg/L, a urinary output of 2 L per day was assumed for all populations (Sieniawska et al., 2012), and total daily exposure was estimated using average body weights of 70 kg and 55 kg, as noted above. Note that for the study by Vahter et al. (2020), a urinary output of 1 L per day and a body weight of 23 kg (reported) were used for children of 10 years.

If u‐tiAs was given in μg/g creatinine, then the concentration in μg/g was multiplied by the normal 24 h creatinine excretion in order to transform the concentration (in μg/g) to μg/24 h. For women in Europe and the Americas the multiplication factor was 1.2 (Pouillot et al., 2022; Sallsten & Barregard, 2021) and 1.0 for Asian populations (due to lower body weight and muscle mass). For men in Europe and the Americas this factor was 1.8 (Pouillot et al., 2022; Sallsten & Barregard, 2021 ) and 1.5 for Asians (due to lower body weight and muscle mass).

The exposure estimates have been derived as described above for each individual study and are presented in detail in Annex D.

Calculation of benchmark doses

The benchmark dose (BMD) analyses were performed using the EFSA ‘Bayesian BMD’ webtool, an online application that uses the R‐package BMABMDR to perform data preparation, model fitting, model averaging and plotting. The guidance of the Scientific Committee on BMD modelling (EFSA Scientific Committee, 2022) was taken into account. The CONTAM Panel recognised that the guidance did not specifically address modelling of epidemiological data.

Transformations of relative risk estimates to quantal data

The results of the epidemiological studies were reported as crude and adjusted risk estimates in the form of incidence rate (IR), incidence rate ratio (IRR), hazard ratio (HR), odds ratio (OR) and prevalence ratios (PRs). In some instances, these indices were named relative risk (RR). The adjusted risk estimate reflects the risk estimate adjusted for potential confounders, such as age, sex, smoking, socioeconomic status etc. and is therefore a better estimate of the true risk from exposure than the crude risk estimates.

Since the current BMD approach is not designed to model relative risk estimates such as IRR, HR or OR, it was necessary to transform the relative risks to natural numbers/integers. This was based on the approach used by JECFA (FAO/WHO, 2011).

For cohort studies, the incidence rate or the cumulative incidence in the reference category was calculated. This incidence was multiplied by the adjusted risk estimate to obtain an adjusted incidence estimate, which was then used to calculate the ‘adjusted number of cases’ (as integers). Having obtained the ‘adjusted number of cases’ and the population size (provided in the papers or calculated from number of cases and incidence rates) in each exposure category, these data could be used as input into the EFSA BMD webtool.

For case–control studies, the approach was similar. A case–control study usually uses the OR as the estimate of the RR. When the outcome (e.g. a disease) has a low incidence (e.g. < 1% of a population), the OR can be considered a good approximation of the RR.

In order to make inferences about the source population, its distribution of exposure had to be derived. In a case–control study the number of controls in each exposure category reflects the distribution of exposure in the source population from which the controls were selected, and this was used to distribute the source population into exposure categories. The ‘adjusted number of cases’ was calculated in a similar way as for the cohort studies. To obtain the ‘adjusted number of cases’ in a specific exposure category (e.g. ‘A’), first the number of cases in the reference category was multiplied by the adjusted OR in ‘A’. It was then multiplied by the ratio of the number of controls in ‘A’ and the reference category (‘R’). This is because the ratio of controls in A and R reflects the relative distribution of exposure categories in the source population. Finally, the adjusted case numbers were ‘normalised’ so that their sum was identical to the initial sum of cases.

If the outcome was not one with a low incidence (< 1%), the OR can be used to obtain the RR using the following formula: RR = OR/(1 − p0 + (p0 × OR)) where p0 is the baseline risk (Grant, 2014).

The cross‐sectional studies usually used the adjusted OR (the prevalence odds ratio) as the risk estimate, and therefore the approach was in line with that used for case–control studies.

In most studies also the number of outcome cases (e.g. cancer) by exposure category (e.g. water As concentration) was reported. For cohort studies and case cohort studies the size of the cohort (or the subcohort) was reported (as the number of individuals or the number of person‐years) and in most studies these data were also reported by exposure category. For most of the case–control studies, the number of cases and controls in total and by exposure category were reported, as well as the approximate size of the source population from which the controls were selected. The source populations in the respective dose categories were set to have the same relative sizes as the number of controls. If the source populations were not presented in the papers, they were retrieved from national statistics for the areas from which the cases were recruited. If there was an age inclusion of cases, this was taken into account when estimating the source populations. For cross‐sectional studies, the number of cases, the population sizes and or the prevalence (proportion) were presented by exposure category.

The transformations have been performed (including the number of cases and the sizes of the source populations) as described above for each individual study and are presented in detail in Annex D.

Selection of the benchmark response for critical effects

The EFSA guidance on benchmark dose modelling (EFSA Scientific Committee, 2022) does not provide formal guidance on modelling of epidemiological data, including the selection of the appropriate BMR for human data. The default BMR for quantal experimental animal data is 10% (extra risk). EFSA (2005) considers that such a value is the lowest statistically significant increased incidence that can be measured in most experimental animal studies. Previous risk assessments based on human data, which generally have larger numbers of subjects than in animal studies, have considered appropriate BMRs on a case‐by‐case basis, and arrived at lower response values (see EFSA CONTAM Panel, 2009). In the arsenic Opinion in 2009, the CONTAM Panel decided to use 1% extra risk when modelling the risk of skin lesions (see Section 3.2.4.1). In addition, BMCLs, for 1% extra risk for lung and bladder cancer derived by NRC (2001) were taken into account. For lung cancer, the NRC (2001) calculations were based on the relative risks in one of the studies. The JECFA (FAO/WHO, 2011) used a BMR of 0.5% extra risk considering that the low end of the observed data range was 0.5% increased (cancer) incidence. Also, EPA (US EPA, 2012) and Haber et al. (2018) noted that for epidemiological data response rates of 10% extra risk may involve upward extrapolation, in which case a lower BMR value needs to be applied.

In the subsection on Endpoints used for calculation of benchmark doses the ‘adjusted number of cases’ was calculated in each exposure category for human studies selected for modelling. The source population for the cases was provided in cohort studies or estimated in case–control studies. This allowed for the use of quantal data and the EFSA software. But the observation time during which the cases occurred varied between studies and for most of them it was short. For the cancer outcomes the cumulative incidence (the ratio between the number of cases and the size of the source population over the observation time) in the case–control studies was only around 0.02%. The CONTAM Panel considers it not to be appropriate to model the dose at which the incidence of an outcome increases from – for example – 0.02% to 1.02%, since this would correspond to a very large (50‐fold) relative increase of the background incidence after adjustment for confounders. The fact that the observation time and the time period for recruiting cases in control studies differs a lot between studies makes it inappropriate to add an extra (absolute) incidence to the background incidence. Therefore, the BMR was instead expressed in terms of relative risk, i.e. a relative increase of the background incidence after adjustment for confounders. This response definition better accounts for the variation in ‘observation times’ which is regarded to increase the comparability of BMD results across studies.

More specifically, the CONTAM Panel decided to use a BMR of 5%, expressed as relative increase of the background incidence after adjustment for confounders by 5% as the BMR and use the modelling results for the derivation of an appropriate Reference Point. For considered endpoints a BMR of 1%–5% (relative increase of the background incidence after adjustment for confounders) is regarded to be relevant for public health. Depending on the background response such BMRs can represent very low absolute increases, which may have an impact on the BMD estimation. However, model averaging used in the BMD modelling process improves the estimation precision. It allows to estimate more accurately the BMD associated with smaller BMRs compared to using a single model only. The Bayesian model averaging allows to incorporate additional information in the modelling process, which also influence the estimation precision. In particular, ensuring that the background incidence is centred around the observed incidences for the lowest exposure group (as expected based on expert knowledge) contributes as well to improved precision in the BMD estimation for lower BMR values. The inferences used to estimate corrected number of cases (after adjustment for confounders) to build the associated incidence, entailing large sample sizes, allows in addition to improve estimation precision. Results from the analyses fulfilled criteria in the EFSA guidance (2022) related to BMD uncertainty (see next Section). The uncertainty in BMDs across different BMRs (1%–10%, relative increase of the background incidence after adjustment for confounders) was also assessed in more detail, promoting a value of 5% (Section 3.7). The consequence of using a BMR that is substantially higher than 5% is also discussed in Section 3.7. However, this is not regarded as a better option than the modelling approach taken.

In addition, the CONTAM Panel used the estimated incidence in the reference category as an informative prior for the background parameter in the dose–response modelling. As a general approach, it is regarded to be more appropriate, as this is additional information that can be incorporated in the modelling process guiding the model rather than allowing the model to estimate a theoretical background response. The latter would imply extrapolations below the observed exposure ranges that would be dependent on the models used in the analysis.

When the midpoint of the highest exposure category was not reported, both the lower limit (in most cases the most conservative approach) and twice this value were used. For some outcomes the incidence or prevalence of the outcome was high (in some cases > 10%), and an increase of extra (absolute) risk was meaningful, but for consistency, the relative increase of background incidence was modelled also for these studies.

Results of dose–response analyses

Table 33 shows an overview of the results of the benchmark dose analyses and the potential RP for the studies selected for dose–response modelling.

The EFSA guidance (2022) includes recommendations for assessing when the BMD is too uncertain for the BMDL to be used as a Reference Point (RP). The CONTAM Panel considers that these criteria are also applicable when modelling epidemiological studies. According to the criteria, the BMDL was not recommended as an RP when (i) none of the candidate models fit the data sufficiently well, (ii) BMD/BMDL > 20, (iii) the BMD is 10 times lower than the lowest non‐zero dose or (iv) BMDU/BMDL > 50. If one or more of these criteria were not met the study was not considered further.

The following studies were excluded from further consideration since none of the candidate models adequately fitted the data: Richter et al. (2022) and Rudnai et al. (2014) on congenital heart disease; Parvez et al. (2013) on FEV1 and FVC; Powers et al. (2019) on asthma‐related symptoms; Siddique et al. (2020) on obstruction, reversible obstruction asthma‐related symptoms, FEV1 and FEV6, and Milton et al. (2005) on infant mortality. Additionally, the following endpoints did not comply with the BMD/BMDL and/or BMDU/BMDL criteria: Rahman et al. (2010) on infant mortality and stillbirth; Siddique et al. (2020) on obstruction and asthma‐related symptoms and were therefore not considered further. Rahman et al. (2010) on spontaneous abortion was rejected following visual screening due to the non‐monotonic dose response seen.

BMDLs are only listed in Table 33 if all recommendations are fulfilled. Further information and BMD reports are listed in Annex E. In addition, the table shows the evaluation of the risk of bias (selection bias, information bias and confounding) and its assumed direction.

3.2.5. Identification of a reference point for hazard assessment

The lowest BMDLs (< 0.1 μg/kg bw per day) in Table 33 (Section 3.2.4.2) based on a BMR of 5% relative increase of the background incidence after adjustment for confounders were calculated for skin cancer, skin lesions, lung outcomes and chronic kidney disease. Low BMDLs (≤ 0.17 μg/kg bw per day) were also calculated for lung cancer, bladder cancer and ischemic heart disease.

The CONTAM Panel decided to use the BMDL05 (0.06 μg/kg bw per day) for the case–control study on skin cancer (type: squamous cell carcinoma) in the US by Gilbert‐Diamond et al. (2013) as a Reference Point (RP). The association between iAs exposure and skin cancer is well established. The study was considered to have a low risk of bias. Exposure categories were based on u‐tiAs. The confounder‐adjusted risk of skin cancer was positively associated with u‐tiAs (untransformed as well as log‐transformed) with statistical significance, although the adjusted OR in the upper tertile was not statistically significant (OR 1.43, 95% CI 0.91–2.27), Table 11.

The BMDL05 of 0.01 μg/kg bw per day derived for the other case–control study on skin cancer (type basal cell carcinoma), performed in Hungary, Romania and Slovakia by Leonardi et al. (2012) was also considered as RP. This study too showed a statistically significant association between iAs exposure, as measured by As in drinking water, Table 11. The BMDL05 derived from this was much lower than that from study by Gilbert‐Diamond et al. (2013) (0.06 μg/kg bw per day). Both these studies were considered as being of good quality. The study by Leonardi et al. (2012) had the advantage of five doses compared to three for the Gilbert‐Diamond study.

However, the study by Leonardi et al. (2012) used hospital controls and was therefore considered having a slightly higher risk of bias than the study by Gilbert‐Diamond et al. (2013), which used population controls.

Moreover, when modelling the study by Gilbert‐Diamond et al. (2013), which was based on u‐tiAs, there was no need to use a fixed default additional intake of iAs from other food than drinking water, which was necessary for the study by Leonardi et al. (2012). The US population used for the study of skin cancer was considered comparable with the European population in terms of nutrition and socioeconomic factors.

The results when modelling the Gilbert‐Diamond study using a BMR of 1% or 10% (relative increase of the background incidence after adjustment for confounders) showed lower and higher BMDs and BMDLs, respectively, but differences were, however, not very large, which further strengthens the validity of the RP. Finally, squamous cell carcinoma is a more serious type of skin cancer than basal cell carcinoma. In addition, the BMDL05 when modelling the study by Leonardi et al. (2012) was about 10 times lower than the median dose in the lowest dose category, and the BMD was below the dose in the reference category, which makes the estimate uncertain. However, the results from the study by Leonardi et al. (2012) support the choice of skin cancer as a RP for hazard assessment and the relatively low BMDL05 calculated when modelling the study by Gilbert‐Diamond et al. (2013).

The BMDL05 values for the lung outcomes in the cross‐sectional study in the US by Powers et al. (2019) were also low, one of them as low as 0.04 μg/kg bw per day. This outcome was based on self‐reporting (if a medical person ever told them they had emphysema). The BMDLs for impaired lung function showed BMDLs of 0.07 and 0.1 μg/kg bw per day. Overall, an RP of 0.06 μg/kg bw per day should therefore be also considered to cover lung outcomes other than lung cancer which is is described separately below.

The BMDLs for some of the studies on lung cancer (Steinmaus et al., 2014b), bladder cancer (Chen, Chiou, Hsu, Hsueh, Wu, Wang, & Chen, 2010), ischemic heart disease (Moon et al., 2013) and chronic kidney disease (Hsueh et al., 2009) were in the range 0.12–0.17 μg/kg bw per day. An RP of 0.06 μg/kg bw per day should therefore also to cover those outcomes. For lung cancer, a common and very serious disease, the CONTAM Panel discussed whether a 5% relative increase in incidence was too high to use as a BMR. However, the CONTAM Panel notes that the BMDL when modelling the study by Steinmaus et al. (2014b) with a BMR of 1% relative increase in incidence is 0.06 μg/kg bw per day (see Annex E).

Some of the other studies listed in Table 33 also showed low BMDLs. This was the case for one of the studies on skin lesions (Ahsan et al., 2006), a cross‐sectional study in Bangladesh. However, the study by Pierce et al. (2011) on the same population used a longitudinal design and was therefore considered more valid. In addition, the BMD for the study by Ahsan et al. (2006) was lower than the dose in the reference group. Another example is the longitudinal study on CKD by Zheng et al. (2015). This outcome was based on an estimated GFR of < 60 mL/min per 1.73 m2. The study by Hsueh et al. (2009) had stronger criteria for CKD (reduced eGFR for at least 3 months) and low eGFR (mean 28 mL/min per 1.73 m2) and is likely to be more valid.

For other outcomes listed in Table 33, the BMDLs were considerably higher and a RP of 0.06 μg/kg bw per day is therefore considered to cover these outcomes as well.

3.3. Consideration of the approach to risk characterisation

iAs is a human carcinogen, being causally associated with cancers of the skin, lung and bladder, with lesser evidence for various other types of cancer. iAs does not interact directly with DNA but induces DNA base oxidation and both DNA single and double strand breaks. Clastogenic and aneugenic effects have been demonstrated both in vitro and in vivo as measured by a dose‐dependent increase of chromosomal aberrations, micronuclei and aneuploidy. In addition, iAs effectively inhibits DNA repair both in vitro and in vivo thus enhancing its own genotoxic potential and acting as a co‐mutagen. iAs induces clastogenic events in humans in a range of low to moderate exposure levels. These effects have also been reported when exposure occurs in the pre‐natal and early stages of life. Moreover, cross‐generational effects of iAs on genotoxicity, DNA methylation and reproduction (Nava‐Rivera et al., 2021; Yin et al., 2022) have been described in rodent models. Based on the evidence that iAs exerts its genotoxic effects, at least in part, by inducing oxidative DNA base modifications and DNA strand breaks, it has been proposed that iAs has an indirect genotoxic mode of action via production of oxidative stress, which could be thresholded.

However, there is clear evidence that iAs induces DNA double strand breaks, both in vitro and in vivo (including humans) at low levels of exposure. These lesions have been shown to arise by both replication‐dependent and replication‐independent mechanisms, suggesting that genotoxic clustered DNA lesions may also arise due to the presence of radical species in the close vicinity of DNA. On the other side, the aneugenic effects of iAs are likely to be thresholded, probably involving proteins of the spindle apparatus. Determining whether a threshold dose‐effect relationship exists for the clastogenic effects remains challenging (Elhajouji et al., 2011) No threshold has been reported for chromosomal damage after exposure of human cells to low doses of ionising radiation (Boei et al., 2012) likely due to unrepaired DSB. Since DSB have been observed after iAs exposure it remains possible that iAs‐induced clastogenic effects are not thresholded.

In addition, iAs is a potent inhibitor of DNA repair, which is the underlying mechanism for its co‐mutagenic effects. The interactions with the DNA repair/DDR system are based on protein interactions, again indicating the possibility of thresholded mechanisms. However, the inhibition of DNA repair impacts on the main self‐defence mechanisms that play a crucial role in genotoxicity thus potentially affecting the shape of the dose–response curves at low doses. iAs is also involved in multiple effects by altering signalling pathways and the epigenetic landscape. Changes in DNA methylation are reported in populations exposed to low to moderate levels of iAs. Major epigenetic alterations, DNA methylation and histone modifications, are regulated by various enzymes and thus are likely to be thresholded. The epigenome may act as a modifier of exposure effects at low doses and of risk for long‐lasting effects.

All the effects described above are associated with carcinogenesis. Thus, it can be envisaged that both thresholded and non‐thresholded mechanisms could apply to the genotoxic effects of iAs. In addition, the methylation of iAs, particularly to trivalent methylated as well as pentavalent methylated thiolated species, should be regarded as an activation process that forms reactive species, which might exert even stronger genotoxic effects, inhibit DNA damage response including DNA repair pathways, promote ROS generation and induce cell transformation. It has been shown in vitro that inorganic arsenic metabolites are genotoxic and disturb various targets of the cellular DNA damage response. Genomic stability is likely to be affected by iAs and its metabolites in parallel by different pathways. Given the combination of different actions, there are considerable uncertainties regarding the shape of the dose–response curves for carcinogenicity at low levels of exposure.

Therefore, it is not appropriate to establish a health‐based guidance value (HBGV) and a margin of exposure (MOE) approach has been applied in the risk characterisation (see Section 3.6). There are no precedents in EFSA for identification of an MOE of low concern, when using a BMDL derived from human cancer data. Therefore, the Panel decided not to determine a value for an MOE of low concern.

3.4. Occurrence data

3.4.1. Occurrence data used in the present assessment

Following the ToR, the present risk assessment should take into account the updated dietary exposure assessment endorsed by the CONTAM Panel in November 2020 (EFSA, 2021). This Section provides a summary of the occurrence data used for that exposure assessment; detailed information can be found in the corresponding EFSA scientific report (EFSA, 2021).

After data cleaning and analysis of the submitted occurrence data, the 2020 EFSA scientific report initially considered a total of 13,608 analytical results on iAs from samples collected between 2013 and 2018 in 23 different European countries. Out of them 7623 analytical results corresponded to drinking water 19 and 5985 to different types of food. Overall, most of the samples were detected using ICP–MS, although for the samples of drinking water the preferred detection method was Atomic Emission Spectrometry (AES). Among the samples of drinking water, the highest mean levels of arsenic were reported for unspecified drinking water (mean = 2.0–2.4 μg/kg, LB–UB) while the lowest levels were for carbonated mineral water (mean = 0.8–1.7 μg/kg, LB–UB). A total of 356 samples of different types of drinking water were reported with iAs levels above 10 μg/L; 20 they were excluded to avoid introducing a bias when estimating dietary exposure to iAs.

The most represented food category was ‘Grains and grain‐based products’ with a total of 2928 analytical data submitted on iAs; within this food category close to 1800 samples of different types of rice were included together with rice‐based commodities known to contain relatively high levels of iAs. Rice samples and rice‐based products were reported with the highest levels of iAs, with mean values of 233 μg/kg in red rice (LB = UB) and 128–131 μg/kg (LB–UB) in brown rice among the rice samples. It is also important to note the relatively high levels of iAs reported for few samples codified as ‘Rice cakes/Rice waffles/Rice crackers’, with mean levels of 146–148 μg/kg (LB‐UB). The relatively high presence of iAs in this type of product was already identified in the 2014 EFSA scientific report (EFSA, 2014) and is also profusely described in the literature (Dominguez‐Gonzalez et al., 2020; Islam et al., 2017).

Another food category with a high number of samples was ‘Fish and other seafood’ (n = 938), with almost half of the samples codified as ‘Fish meat’ (n = 451). The reported levels of iAs for ‘Fish meat’ were relatively low (mean LB = 4 μg/kg) while higher levels were reported for crustaceans (mean LB = 15 μg/kg) and for molluscs, in particular in clams (mean LB = 108 μg/kg). The food group ‘Vegetables and vegetable products (including fungi)’ was also well represented with 589 samples. Particular attention was given to mushrooms as they are well known to accumulate different arsenic species, including iAs, depending on the substrate and the fungal species. Overall, iAs was more often quantified in samples of wildly growing mushrooms as compared to those cultivated; mean levels up to 16.9 μg/kg (LB) were reported for samples of Cantharelle (Cantharellus cibarius) and up to 17.2 μg /kg (mean LB) for few samples of the genus Boletus. Seaweed samples accounted for almost half of the samples (n = 279) reported as ‘Vegetables and vegetable products (including fungi)’. Although arsenic in seaweeds is primarily found in the form of arsenosugars, some brown alga in particular the seaweed Hiziki or Hijiki (Sargassum fusiforme, syn. Hizikia fusiformis) but also Kombu (Laminaria spp.) reported levels of iAs close to 100 mg/kg and high iAs/tAs ratios (75%–85%).

For the assessment of the exposure in the young population, a total of 482 samples of ‘Food for infants and young children’ with data on iAs were available. Among the samples codified as ‘Cereal‐based food for infants and young children’ (n = 224), the highest levels of iAs were reported for samples containing rice as ingredient (mean levels = 77–79 μg/kg, LB‐UB). To mention a group of samples codified as ‘Biscuits, rusks and cookies for children’ with relatively high levels of iAs (mean levels = 83–94 μg/kg, LB‐UB); under this category are included typical snacks (rice cakes, rice crackers, rice biscuits) that are widely used to feed infants and young children. On the other hand, very low levels of iAs were reported for both ‘Follow‐on formula, milk‐based’ and ‘Infant formula, milk‐based’, with mean values of 0.08–2.1 μg/kg (LB–UB) and 0.02–4.0 μg/kg (LB–UB), respectively. This is in line with data reported in the literature, although the contribution of infant and follow‐on formulae might drastically increase in certain areas due to the use of drinking water with relatively high levels of iAs to reconstitute the formulae. Additionally, for infants and children who suffer from milk intolerance (e.g. lactose) and/or milk allergies (e.g. proteins) milk‐based infant and follow‐on formulae are replaced by the so‐called ‘Foods for special medical purposes’ (FSMPs). Among them, rice‐based formulae are gaining acceptance (Bocquet et al., 2019) and although no occurrence data were available for this particular food group, a dedicated exposure scenario was conducted in infants based on its consumption using occurrence data from the literature (see EFSA, 2021 for further details). During the preparation of the 2021 EFSA scientific report several uncertainties were identified linked to the occurrence data used in dietary exposure assessment. A more detailed analysis of these uncertainties is provided in the Uncertainty analysis.

3.5. Exposure assessment

3.5.1. Exposure assessment used for the present opinion

Following the ToR, the present risk assessment should take into account the updated dietary exposure assessment endorsed by the CONTAM Panel in November 2020 (EFSA, 2021). This Section provides an overview of that assessment, including the exposure estimates across different age classes in Europe and the main foods contributing to the dietary exposure to iAs; detailed information can be found in the corresponding EFSA scientific report (EFSA, 2021).

The 2021 EFSA scientific report considered consumption data from 23 different European countries and a total of 44 different dietary surveys (87,945 subjects) to estimate the chronic dietary exposure to iAs. In addition to the different age classes considered (Infants, Toddlers, Other children, Adolescents, Adults, Elderly and Very elderly), seven surveys were also used providing consumption data on specific population groups: ‘Pregnant women’ (five dietary surveys) and ‘Lactating women’ (two dietary surveys). The highest dietary exposure to iAs 21 was estimated in the young population (infants, toddlers and other children). The highest mean dietary exposure estimates at the lower bound (LB) were in toddlers (0.30 μg/kg bw per day), and in both infants and toddlers (0.61 μg/kg bw per day) at the upper bound (UB). At the 95th percentile, the highest exposure estimates (LB–UB) were 0.58 and 1.20 μg/kg bw per day for toddlers and infants, respectively. In general, UB estimates were two to three times higher than LB estimates (EFSA, 2021). Table 34 shows a summary of the chronic dietary exposure estimates to iAs across 44 different dietary surveys carried out in 23 different European countries.

The main contributors to these differences between LB and UB exposure were some food categories (e.g. ‘Milk and dairy products’, ‘Grains and grain‐based products (no rice)’ and ‘Fruit and vegetable juices’, among others) with low LB values, a relatively high number of left‐censored data and relatively high consumption in different age classes. Across the different age classes, the main contributors to the dietary exposure to iAs (LB) were ‘Rice’, ‘Rice‐based products’, ‘Grains and grain‐based products (no rice)’ and ‘Drinking water’. Particular foodstuffs indicated for the young population (e.g. ‘Cereal‐based food for infants and young children’ and ‘Biscuits, rusks and cookies for children’) made a relevant contribution in the dietary exposure to iAs in this age group. Overall, the contribution of milk‐based infant and follow‐on formulae to the intake of iAs was rather low due to its relatively low iAs levels, although the final levels could be strongly affected by the iAs present in the drinking water used for reconstitution. In the adult population, food groups such as ‘Vegetables and vegetable products’ and ‘Fish and other seafood’ were also apparent sources of iAs in certain countries.

Different ad hoc dietary exposure scenarios were conducted to complement the general exposure scenario (EFSA, 2021). As an example, in infants, exposure to iAs was estimated considering the consumption of rice‐based formula. A mean level of iAs of 12 μg/kg (n = 7, range 9–20 μg /kg) as reported by Meyer et al. (2018) for this type of samples, and consumption values of 200 and 260 mL/kg bw per day (mean and high‐level consumption) as recommended by the EFSA Scientific Committee for infants below 16 weeks of age (EFSA Scientific Committee, 2017) were used. Dietary exposure to iAs was estimated as 0.30 and 0.39 μg/kg bw per day in mean and high consumers, respectively. Another ad hoc scenario was carried out using the reported consumption of ‘Cereal‐based food for infants and young children’, and the occurrence data from the 93 samples codified under this group identified as having rice as an ingredient. The consumption of this type of food with rice as ingredient is becoming more and more popular among young children affected by either celiac disease or gluten intolerance that must follow a gluten‐free diet. Dietary exposure estimates to iAs at the P95 as high as 0.62 μg/kg bw day and 0.70 μg/kg bw day were calculated for toddlers and infants, respectively. These values correspond to exposure estimates considering high consumption of cereal‐based food of 45 g/day and 50 g/day for infants and toddlers respectively with relatively high levels of iAs (P95 occurrence (LB) of 130 μg/kg). As compared to the previous 2014 EFSA scientific report on dietary exposure to iAs, the estimates were noticeably lower, with the maximum means and 95th percentile estimates across the different age classes around 1.5–3 times lower. These differences were explained by the different occurrence/consumption data and/or the different methodology used in both reports; dietary exposure estimates in the 2021 EFSA scientific report were in good agreement with recently published scientific literature that also made use of measured iAs to estimate dietary exposure to iAs.

Several recommendations regarding the exposure assessment for iAs which are still valid were made in the 2021 EFSA scientific report. They are therefore not described in the present Opinion.

3.6. Risk characterisation

There are no precedents in EFSA for identification of an MOE of low concern, when using a BMDL derived from human cancer data. Therefore, the Panel decided not to determine a value for an MOE of low concern.

The Reference Point (0.06 μg/kg bw per day) identified by the CONTAM Panel for skin cancer is within the range of the dietary exposure estimates for iAs for average (0.03–0.15 μg/kg bw per day) and slightly below the range of high‐level adult consumers (0.07–0.33 μg/kg bw per day) in Europe (see Table 34). Therefore, in adults, the MOEs range between 2 and 0.4 for mean consumers and between 0.9 and 0.2 at the 95th percentile exposure, respectively. An MOE of 1 describes the exposure level that could be associated with a 5% increase relative to the background incidence for skin cancer, based on the available data. Despite the uncertainties, the CONTAM Panel concludes that these MOEs raise a health concern for skin cancer. The CONTAM Panel noted that the BMDLs for some of the studies on lung cancer, bladder cancer, ischemic heart disease and chronic kidney disease were in the range 0.12–0.17 μg/kg bw per day, which is also within the range of dietary exposure estimates. Therefore, there is a possible concern also for these endpoints.

The CONTAM Panel notes that dietary exposure is higher in the younger age groups and therefore the respective MOEs are smaller. However, this does not necessarily indicate that children are at greater risk, because the effects are due to long‐term exposure and most of the epidemiological studies are conducted in adults who would also have had higher dietary exposure during early life. Therefore, the CONTAM Panel concludes that children are covered by this risk characterisation.

Arsenic is known to impair DNA damage response and DNA repair and individuals with genetic variants in these pathways have increased cancer susceptibility (see Section 3.3). Although risk characterisation is based on the results of relatively large epidemiological studies, susceptible individuals of higher genetic risk may not be adequately represented in these studies. Therefore, dietary exposure to arsenic may be of greater concern for such individuals than for the general population.

3.7. Uncertainty analysis

The evaluation of the uncertainties in the present assessment was performed following the principles laid down in the Guidance on uncertainty analysis in scientific assessments (EFSA Scientific Committee, 2018). The purpose of the uncertainty analysis is to identify and quantify the major uncertainties of the specific risk assessment and combine them to assess the overall certainty of the final conclusion.

To harmonise uncertainty analyses, the CONTAM Panel has developed a road map that structures the risk assessment process in broader groupings, as well as subgroupings. Sets of questions have also been defined to help the collection of uncertainties. There are four overarching elements of the road map: (i) chemical characterisation and analytical methods, (ii) exposure assessment, (iii) hazard identification and characterisation and (iv) risk characterisation. Based on the available time and resources, the CONTAM Panel decided to mainly focus on (i), (iii) and (iv), rather than (ii). The exposure assessment was carried out earlier and it has also been published as a standalone document (EFSA, 2021). The uncertainties identified for (i) and (iii) are described in Table 35. As an extension of (iii) a quantitative assessment of BMD uncertainty depending on the BMR is also performed separately.

TABLE 35

Elements of the CONTAM road map and relevance for the uncertainty analysis ‐ hazard identification and characterisation and risk characterisation.

Main groupSub‐groupOverarching questionsDescription of uncertaintyUncertainty a Priority ranking b
Chemical characterisation and analytical methods Chemical characterisation Is there uncertainty in the chemical composition of the compound administered to experimental animals and in in vitro studies?Uncertainty in the applied dose No
Exact composition of the tested compounds No
Analytical methods Is there uncertainty in the analytical process?Lack of certified reference materials and proficiency tests No
Hazard identification and characterisation Epigenetics Is there uncertainty in the growing body of evidence regarding epigenetic effects?Relevance of the data to human health effects Yes 1
Genotoxicity Is there uncertainty on the genotoxicity of the substanceUncertainty regarding mode of action for both iAs and its human metabolites Yes 1
ADME

Is there uncertainty in any aspect of human ADME?

This may include insufficient information on absorption, distribution, metabolism or excretion of iAs

Absorption Yes 1
Distribution Yes 1
Metabolism Yes 1
Excretion Yes 1
Biomarkers of exposure Is there uncertainty in the studies of biomarkers of exposure?Variability of urinary As excretion. Yes 1
Epidemiological studies Is there U in the validity of the studies causing a risk of bias?Study design Yes. Differs between studies, for details, see risk of bias assessment. The overall risk of bias for modelled studies was considered to be low

1–2.

A few of the studies were cross‐sectional

Selection bi

1–2

Some case–control studies used hospital controls

Exposure assessment2. Exposure assessment is uncertain in all the studies. For example, arsenic in drinking water may vary, and water intake and intake of iAs from other food vary between individuals. The untertainty is assumed to be less in studies using u‐tiAs, but u‐tiAs mirrors variability in exposure. In most cases the misclassification will be non‐differential causing an underestimation of a true risk
Outcome assessment1–2. A few studies used self‐reported outcomes.
Confounding2. Residual confounding is always possible. Some studies have probably over‐adjusted for covariates in the causal chain
Dose–response analysis of critical end points Is there uncertainty regarding the dose response analysis e.g. trend occurrence, large data variation, possible covariates?Small number of cases in some of the studies Yes 2
Geographical representativeness and consistency and human variability due to genetic differences Yes 1
Human variability due to genetic differences Yes 2
Selection of RP Are there uncertainties in the selection of the RP that are not covered by the BMD confidence interval e.g. is model uncertainty covered?May not cover all uncertainties related to epidemiological studies, and dose–response analysis of critical endpoint(s) Variation Yes 2
Is there uncertainty on the relevance of the selected BMR and (how) will this affect the results from BMD analysis?Selection of a relatively low BMR may increase uncertainty of the BMD and BMDL at the BMR Yes 2
Risk characterisation Risk metric Uncertainty due to uncertainty in exposure estimates and RP/BMDs Yes 2

Abbreviations: ADME, absorption, distribution, metabolism, excretion; As, arsenic; bi, bias; BMD, benchmark dose, BMDL, benchmark dose lower confidence limit; BMR, benchmark response, CONTAM, Panel on Contaminants in the Food Chain; iAs, inorganic arsenic; RP, reference point; U, uncertainty, u‐tiAs, total urinary inorganic arsenic (sum of iAs and its methylated metabolites MMA and DMA).

a Yes, No or NA (not applicable).
b 0 ‐ negligible, 1 – low, 2 – medium, 3 – high.

At the level of the risk characterisation (iv), a quantification of overall uncertainty in the margin of exposure is performed. This data‐driven analysis (section 3.7.3) is based on information of uncertainty described by the exposure and BMD estimates in the Opinion, i.e. LB and UB scenarios for the exposure and estimated BMD credible intervals. This assessment is therefore conditional on the assumption that identified uncertainties below (Section 3.7.1) that may not be covered by the exposure and BMD estimates used, or the sensitivity analyses performed (Sections 3.7.3.3 and 3.7.3.4), is relatively low. The overall conclusion in section 3.7.4 is therefore based on the quantitative analysis (3.7.3) in combination with qualitative consideration of remaining uncertainty.

3.7.1. Identification and prioritisation of uncertainties

No major uncertainties were identified with respect to chemical characterisation and analytical methods, but several uncertainties were identified related to hazard and risk characterisation (Table 35). At the level of the risk characterisation metric (the margin of exposure, MOE) the impact of identified uncertainties related to epigenetics, genotoxicity, ADME, as well as biomarkers of exposure were regarded to be of low priority (see Table 35). Also, uncertainties related to the validity of the epidemiological studies used for dose–response analysis were of low priority, considering the assessment of risk of bias (Section 3.2.4.2 and Table 33). However, other uncertainties related to the epidemiological studies, dose–response analysis of critical endpoints and selection of an RP were generally of medium priority. This includes uncertainties related to the exposure assessments in all the studies; e.g. the uncertainty in the intake (L/day) of As‐contaminated water and the assumed dietary intake of iAs. Default values for water intake have been used as described in Section 3.2.4.2, i.e. they ranged from 1.5 to 4 L/day across critical studies (see Table 33). This intake was rarely quantified by interviews (except in Steinmaus et al., 2014b). The intake of iAs from other food had to be estimated from the literature, i.e. assumed intakes ranged from 7.7 to 60 μg/kg bw per day across critical studies (see Table 33). For the studies using u‐tiAs (e.g. the critical study, Gilbert‐Diamond et al., 2013), this source of uncertainty was not present, but there is uncertainty in the transformation from urinary iAs to dietary iAs. Also, related to the epidemiological studies residual confounding is always possible. Some studies have probably over‐adjusted for covariates in the causal chain. With respect to the dose–response analysis of critical endpoints, the small sample sizes of some of the individual studies were regarded to be of medium priority. In some cases, despite significant dose–response associations reported in the studies, the odds ratios (ORs) in separate exposure categories were not always statistically significant.

All these uncertainties may not be covered by the estimated BMD credible interval. Also, while covered by the BMD credible interval, it was considered that the relatively low BMR selected may have an impact on the uncertainty in the RP. Therefore, a more detailed quantitative analysis of BMD uncertainty was performed (see Section 3.7.2). Also, a broad quantification of the uncertainty in the margin of exposure across critical endpoints was accomplished (see Section 3.7.3). To further address identified uncertainties related to the epidemiological studies this quantification of uncertainty included sensitivity analyses related to the selected exposure categories and the midpoints in the exposure categories (see Section 3.7.3.3), and the estimated sizes of the source populations for case–control studies (see Section 3.7.3.4). Also, studies selected for the quantitative analysis in Section 3.7.3 cover most of the assumptions on daily water and dietary intakes noted in Table 33.

Uncertainties related to the exposure assessment are presented in EFSA (2021). They concerned both the occurrence and consumption data, as well as the linkage of that data and use of a factor for preparation of foods as consumed. As for uncertainties in Table 35 the uncertainties related to the exposure were regarded to have the potential to cause both over‐ and underestimation, but they were not prioritised.

3.7.2. Assessment of BMD uncertainty

Estimating BMDs associated with low response levels, regarded to be relevant for public health, may introduce extra uncertainty compared to using higher BMRs. This includes the impact of the estimated exposure from food. While the use of a BMR of 5% (relative increase) was compatible with the BMD uncertainty criteria in EFSA (EFSA Scientific Committee, 2022) this issue is addressed in more detail by evaluating how the BMD credible interval depends on the BMR and affects the BMDLs. Also, the approach taken to modelling total exposure compared to iAs water concentrations is discussed in this section.

3.7.2.1. Comparison of relative BMRs of 1%–10%

As one part, BMD uncertainty quantified by the ratio between the BMDU and BMDL was assessed across the relevant response region, i.e. at BMRs of 1, 5 and 10% (see Table 36). As expected, the BMDU to BMDL ratio increases as the BMR becomes lower. When reducing the BMR from 10% to 5%, the BMDU to BMDL ratio increases by less than a factor of 2, in all data sets. However, when further reducing the BMR from 5% to 1% the BMDU to BMDL ratio increases by more than a factor of 2 for several data sets. Also, in two instances the uncertainty at a BMR of 5% is the lowest, but in one instance it is the highest.

TABLE 36

Probability of exceeding the BMD (MOE < 1) associated with a BMR of 5% for adults, and information of BMD uncertainty for BMRs of 1%, 5% and 10% (relative increase of the background incidence after adjustment for confounders).

Critical effectReferenceBMDU:BMDL ratioBMD:BMDL ratioProbability of exceeding the BMD (BMR of 5%) b
BMR 1%BMR 5%BMR 10%BMR 1%BMR 5%BMR 10%Mean exposure95th percentile
Skin cancer Gilbert‐Diamond et al. (2013)6.43.42.63.72.41.80.17Unlikely0.69Likely
Leonardi et al. (2012) a 7.249 a 4.33.20.86Likely1.0Very likely
Bladder cancer Chen, Chiou, Hsu, Hsueh, Wu, Wang, and Chen (2010) a 3723 a 9.05.90.01Very unlikely0.08Very unlikely
Steinmaus et al. (2013) c 3.72.42.02.31.71.5< 0.01Very unlikely< 0.01Very unlikely
Steinmaus et al. (2013) d 4.62.92.32.61.91.7< 0.01Very unlikely< 0.01Very unlikely
Steinmaus et al. (2013) e 207.75.25.62.72.30.01Very unlikely0.10Unlikely
Steinmaus et al. (2013) f 136.04.35.53.22.5< 0.01Very unlikely0.01Very unlikely
Lung cancer Chen, Chiou, Hsu, Hsueh, Wu, and Chen (2010)238.95.38.24.12.8< 0.01Very unlikely< 0.01Very unlikely
Steinmaus et al. (2013) c 3.83.72.12.22.51.6< 0.01Very unlikely< 0.01Very unlikely
Steinmaus et al. (2013) d 2.71.81.71.61.31.3< 0.01Very unlikely< 0.01Very unlikely
Steinmaus et al. (2013) e 477 a 6.66.35.73.42.6< 0.01Very unlikely< 0.01Very unlikely
Steinmaus et al. (2013) f 3.52.31.92.21.71.5< 0.01Very unlikely< 0.01Very unlikely
Steinmaus et al. (2014b) g 177.95.28.24.13.10.01Very unlikely0.07Very unlikely
Steinmaus et al. (2014b) h 29106.6124.63.30.01Very unlikely0.06Very unlikely
Steinmaus et al. (2014b) i 247.95.28.23.92.80.02Very unlikely0.16Unlikely

Abbreviations: BMD, benchmark dose; BMDL, benchmark dose lower confidence limit, BMDU, benchmark dose upper confidence limit; BMR, benchmark response; MOE, margin of exposure.

a Results not compatible with criteria in EFSA guidance (EFSA Scientific Committee, 2022) related to BMD uncertainty (Best fitting model fits sufficiently well, BMDU/BMDL ≤ 50, The lowest non‐zero dose/BMD ≤ 10 and BMD/BMDL ≤ 20).
b Estimated probabilities based on combination of uncertainty distributions for the BMD and the exposure. For Gilbert‐Diamond et al. (2013), Chen et al. (2010b) and Steinmaus et al. (2014b) the posterior BMD distribution is used as part of these calculations, while the generalised extreme value distribution (parametric approach) is used for the other studies. In the latter case the BMD credible interval values were used to estimate the best parameter set for the extreme value distribution recovering the reported percentiles, and they were used to generate a parametric distribution resembling the BMD posterior distribution. In both cases a uniform uncertainty distribution is used for the exposure. Considering the mean exposure the parametric approach gives a probability of 0.14 (i.e. 0.03 units lower) for Gilbert‐Diamond et al. (2013) and for Chen, Chiou, et al. (2010b) and Steinmaus et al. (2014b) results differ by less than 0.01 units. Considering the 95th percentile the parametric approach gives probabilities that are 0.01–0.02 units lower than the table values for Gilbert‐Diamond et al. (2013), Chen et al. (2010b) and Steinmaus et al. (2014b). Estimated probabilities are classified (by terms) according to the approximate probability scale recommended for harmonised use in EFSA Scientific Committee (2018). The approximate scale has been simplified at the extremes so that all probabilities below 0.10 and above 0.90 classify as ‘very unlikely’ and ‘very likely’, respectively. Estimates below 1% are reported as < 0.01.
c Lifetime average, all years, based on concentrations of arsenic in water, and water intake of 1.9 L per person per day.
d Lifetime average, all years, based on arsenic daily intakes. The preferred exposure estimate for the study.
e Lifetime average before 1971, based on concentrations of arsenic in water, and water intake of 1.9 L per person per day.
f Lifetime average before 1971, based on arsenic daily intakes.
g Lifetime average, based on arsenic daily intakes.
h The highest 5‐year average, based on arsenic daily intakes. The preferred exposure estimate for the study.
i The highest single year, based on arsenic daily intakes.

Considering the BMDU to BMDL ratios in combination with the BMD to BMDL ratios presented in Table 36, it can be concluded that the BMD distribution is mostly left skewed (negatively skewed) so that the BMD to BMDL ratio is larger than the corresponding BMDU to BMD ratio. Generally, this is also more pronounced as the BMR becomes lower. Overall, using a BMR of 5% is compatible with respect to uncertainty criteria in the EFSA BMD guidance (EFSA Scientific Committee, 2022). In Table 36, these criteria are violated for three data sets in the case of a BMR of 1%, considering all critical data sets adequate for modelling.

Results are herein presented and discussed for the cancer endpoints. Consideration of all critical effects (data not shown) provided a similar picture to that described above.

3.7.2.2. Comparison of relative BMRs of 5% and 50%

As an extension, a substantially higher BMR of 50% (relative increase of the background incidence after adjustment for confounders) was applied for comparative purposes. This was done for bladder cancer (Chen, Chiou, Hsu, Hsueh, Wu, Wang, & Chen, 2010; Steinmaus et al., 2013) and lung cancer (Steinmaus et al., 2014b). These studies were deemed eligible for BMD modelling as discussed above and allowed the use of a BMR of 50%. For Gilbert‐Diamond et al. (2013) a BMR of 50% is outside the response range making the BMD estimate highly unstable. Chen et al. (2010) was selected since this data set shows the highest uncertainty in terms of the BMDU to BMDL ratio. In the case of Steinmaus et al. (2013) exposure estimates corresponding to lifetime average before 1971 were considered, and for Steinmaus et al. (2014b) exposure estimates corresponding to the 5 years of highest exposure were used. In Northern Chile the iAs levels in the drinking water decreased over the years and in the study by Steinmaus et al. (2013) the increased cancer incidences are, according to the authors, caused by the formerly much higher exposure, rather than the lower arsenic levels in water in recent decades. The background exposure from food is 0.65 and 0.3 μg/kg bw per day for the studies by Chen et al. (2010), and Steinmaus et al. (2013, 2014), respectively.

For Chen et al. (2010) the BMDU50 to BMDL50 ratio was 6.9 (7.31/1.06), which is about a factor 5 smaller than at a BMR of 5% (BMDU05:BMDL05 = 5.46/0.15 = 36, Table 36). For Steinmaus et al. (2013) the BMDU50 to BMDL50 ratio was 2.1 (3.44/1.67). This is about three times lower than at a BMR of 5% (BMDU05:BMDL05 = 1.81/0.30 = 6.0, Table 36). For Steinmaus et al. (2014b) the BMDU50 to BMDL50 ratio was 1.9 (1.97/1.06). This is about five times smaller than at a BMR of 5% (Table 36, BMDU05:BMDL05 = 1.71/0.17 = 10.1, Table 36). The actual BMDL50 for Chen et al. (2010), Steinmaus et al. (2013) and Steinmaus et al. (2014b) is about a factor 7, a factor 6 and a factor 6 higher than the BMDL05, respectively. All BMD reports can be found in Annex E.

In conclusion, using the target BMR relevant for public health (BMR of 5%, relative increase of the background incidence after adjustment for confounders) is regarded as an approach which does not create undue uncertainty around the BMD.

3.7.2.3. Modelling total exposure versus water concentrations

The BMD analysis was performed based on estimates of total exposure. The observed incidences relate to this exposure metric. An alternative is to use the water iAs concentrations as a basis for modelling. However, this will provide a dose–response shape that differs (to some degree) due to differences in relative dose spacing between the two approaches (see Appendix to Annex E5). Consequently, this may affect both the point estimate of the BMD and its uncertainty. Based on analyses of a sub‐set of data for modelled outcomes, the point estimate of the BMD was generally not very different between the two approaches. However, the BMD uncertainty (BMDU to BMDL ratio) became higher (up to a factor 6) under the approach taken (total exposure) compared to first modelling the As concentrations and then adding exposure from food. The BMDLs were then also lower for the approach taken. The CONTAM panel considered that it is most appropriate to model the actual (total) dose–response relationship. The benchmark concentration (BMC) reports for the alternative modelling approach are shown in Annex E5.

3.7.3. Quantification of uncertainty in the margin of exposure

Monte Carlo simulations were conducted to support the (data‐driven) quantification of overall uncertainty. The uncertainties in estimated BMDs and exposures were characterised by probability distributions that were combined to a distribution for the margin for exposure. The probability of exceeding the BMD (i.e. probability of a MOE < 1) for each selected critical effect and exposure scenario (mean or 95th percentile of exposure for adults) was then estimated. Results of this analysis are shown in Table 36, where estimated probabilities are also classified according to the approximate probability scale recommended for harmonised use in the EFSA guidance on uncertainty analysis (EFSA Scientific Committee, 2018).

3.7.3.1. Technical description of the approach

A generalised extreme value distribution was used to approximate the non‐parametric BMD uncertainty distribution generated by the EFSA BMD software. The extreme value distribution was selected due to its flexibility to describe various forms of the BMD distribution, i.e. it can assume asymmetrical shapes in either direction (left or right skew), and it can also approximate a symmetrical case. The 5th percentile, the mode and 95th percentile of this distribution was first defined by the BMDL, the BMD and the BMDU, respectively. The three parameters of the generalised extreme value distribution were then estimated by using a non‐linear system solver (fsolve) in Matlab. N = 30,000 BMD values were randomly generated from the estimated uncertainty distribution for each endpoint. For a number of selected critical effects (Chen, Chiou, Hsu, Hsueh, Wu, Wang, & Chen, 2010; Gilbert‐Diamond et al., 2013 and Steinmaus et al., 2014b) a more accurate approach was applied that uses the distribution generated by the BMD software (posterior BMD distribution) instead of a fitted generalised extreme value distribution.

The uncertainty distribution for the exposure was assumed to be uniformly distributed. The lower and upper limits of the distribution were estimated by consideration of MIN and MAX results across LB and UB scenarios for adults in Table 34. More specifically, a randomly generated value between MIN and MAX for the LB exposure scenario was combined with a randomly generated value between the MIN and MAX for the associated UB exposure scenario. From this resulting LB‐UB interval, a new value was randomly generated representing one alternative exposure. As for the BMD, N = 30,000 exposure values were generated in this way (i.e. one value generated from 30,000 LB‐UB intervals) with respect to the mean and the 95th percentile of exposure, respectively. The approach applied assumes that the MIN and MAX values for LB scenarios are correlated to the MIN and MAX values for associated UB scenarios. This ensures that the LB is smaller than the UB across simulated LB‐UB intervals.

MOEs were estimated by Monte Carlo simulations, i.e. by randomly combining the N = 30,000 generated BMD and exposure values for each endpoint and exposure scenario. Probabilities in Table 36 correspond to the frequency of an MOE below 1.

3.7.3.2. General results

Estimated probabilities of exceeding the BMD in Table 36 for the different critical effects are summarised below by consulting the approximate probability scale recommended for harmonised use in EFSA (EFSA Scientific Committee, 2018). Also, as noted in the previous section, the distribution generated by the BMD software (posterior BMD distribution) rather than a fitted extreme value distribution was used for Gilbert‐Diamond et al. (2013), Chen, Chiou, et al. (2010) and Steinmaus et al. (2014b) as part of the analysis. Based on these datasets, the difference in result depending on BMD distribution was small, i.e. probabilities differed by 0 to 0.03 units (see footnote in Table 36 for details).

Based on the most critical study on skin cancer it is unlikely (probability = 0.17) that the mean exposure exceeds the estimated BMD, while it is likely (probability = 0.7) that an exposure at the 95th percentile does exceed it. Considering the BMD from the other skin cancer study it is likely/very likely that both exposure scenarios exceed the estimated BMD. For both the mean and 95th percentile of exposure it is unlikely (probability = 0.16 or lower) that any of the estimated BMDs from the studies on bladder and lung cancer are exceeded. For lung cancer (Steinmaus et al., 2014b) this also applies if considering a BMD associated with a BMR of 1% instead of 5% (relative increase of the background incidence after adjustment for confounders, data not shown).

3.7.3.3. Sensitivity analyses related to exposure categories and midpoints used for dose–response modelling

Some of the studies listed in Table 33 reported increased risk estimates for more than one way of characterising the exposure. The study by Steinmaus et al. (2014b) reported risk estimates for time‐weighted average over the lifetime as well as the average for the 5 years with highest exposure and the year with the highest exposure. All three alternatives only considered exposure > 40 years back in time, so exposure at young age. The CONTAM Panel considered the average for the 5 years with highest exposure being the most relevant metric, but due to the uncertainty on this choice, sensitivity analyses were performed using also the other two metrics. As can be seen in Table 36 the results for Steinmaus et al. (2014b) for these metrics are similar. Also, it can be noted in Table 36 that probability results for the different exposure metrics in Steinmaus et al. (2013) are similar.

In some of the studies the reference category was reported as ‘< X μg/L’ As in water (or similar when expressed for u‐tiAs) and the highest exposure category was reported as ‘> Y μg/L’. In such cases the calculated doses used half the value of X for the reference category and the value of Y for the highest dose, but a sensitivity analysis 2 × Y was also tested for the highest dose. See Annex F for more details. For intermediate categories, the mean of the cut‐offs was used as midpoint. For some key studies, such as Gilbert‐Diamond et al. (2013) and Steinmaus et al. (2014b), the exact midpoints were obtained from the authors (personal communication Diane Gilbert‐Diamond and Craig M. Steinmaus). Results from the sensitivity analysis are presented in Table 37 for Steinmaus et al. (2013) and Chen et al. (2010). It is shown that doubling of Y has an effect on the BMDL, BMD and BMDU estimates. For the considered data this has no practical impact on the probability of exceeding the BMD for the mean exposure scenario. For the 95th percentile of exposure the probability changes classification from ‘very unlikely’ to ‘unlikely’ in two of the three cases.

TABLE 37

Sensitivity analysis related to estimation of population size with respect to the BMD associated with a relative BMR of 5%, and the probability of exceeding this BMD for adults.

ReferenceScenarioBMDLBMDBMDUProbability of exceeding the BMD b
Mean exposure95th percentile
Gilbert‐Diamond et al. (2013), skin cancerReference scenario0.0620.150.210.14Unlikely0.70Likely
10% decrease0.0560.140.210.16Unlikely0.72Likely
20% decrease0.0610.150.210.14Unlikely0.70Likely
10% increase0.0610.150.210.14Unlikely0.70Likely
20% increase0.0600.150.210.15Unlikely0.71Likely
Steinmaus et al. (2014b), lung cancerReference scenario a 0.190.761.5< 0.01Very unlikely0.05Very unlikely
10% decrease0.180.781.40.01Very unlikely0.05Very unlikely
20% decrease0.180.781.50.01Very unlikely0.05Very unlikely
10% increase0.150.721.40.01Very unlikely0.07Very unlikely
20% increase0.160.701.40.01Very unlikely0.06Very unlikely
Steinmaus et al. (2013), bladder cancerReference scenario a 0.571.11.6< 0.01Very unlikely< 0.01Very unlikely
Highest exposure point estimate doubled0.120.260.570.01Very unlikely0.21Unlikely
Chen, Chiou, Hsu, Hsueh, Wu, Wang, and Chen (2010), bladder cancerReference scenario0.151.35.50.02Very unlikely0.06Very unlikely
Highest exposure point estimate doubled0.120.362.80.02Very unlikely0.14Unlikely
Steinmaus et al. (2013), lung cancerReference scenario a 2.12.83.8< 0.01Very unlikely< 0.01Very unlikely
Highest exposure point estimate doubled1.13.17.8< 0.01Very unlikely< 0.01Very unlikely

Abbreviations: BMD, benchmark dose; BMDL, benchmark dose lower confidence limit, BMDU, benchmark dose upper confidence limit.

a Lifetime average, based on arsenic daily intakes.
b Estimated probabilities based on combination of a generalised extreme value distribution for the BMD, and a uniform distribution for the exposure. For comparability the generalised extreme value distribution is used across all datasets in Table 37 while probabilities in Table 36 for datasets corresponding to reference scenarios for Gilbert‐Diamond et al. (2013), Chen, Chiou, et al. (2010b) and Steinmaus et al. (2014b) are based on using the posterior BMD distribution as part of the analysis. Estimated probabilities are classified (by terms) according to the approximate probability scale recommended for harmonised use in EFSA (EFSA Scientific Committee, 2018). The approximate scale has been simplified at the extremes so that all probabilities below 0.10 and above 0.90 classify as ‘very unlikely’ and ‘very likely’, respectively. Estimates below 1% are reported as < 0.01.

3.7.3.4. Sensitivity analysis related to estimation of population size

As mentioned in Section 3.2.4.2 (Transformations of relative risk estimates to quantal data), the source population size is one of model inputs. This parameter was occasionally not provided in the respective publications and relevant estimates were then calculated based on national statistics. Although national statistics provide the most valid source population estimates, some loss of accuracy may occur. Hence, sensitivity analyses were performed repeating the BMD modelling after increasing or decreasing the estimated source population sizes by 10 or 20% for the critical study (Gilbert‐Diamond et al., 2013) and for the study by Steinmaus et al., 2014b).

When interpreting the results of this analysis, the following points are relevant:

  1. For case–control studies, an estimation of the source population size must be performed since incidence rates cannot be calculated based on the cases and (sample of) controls alone.
  2. Incidence rates would decrease across all exposure groups as the source population size increases.
  3. Using a relative increase of the background incidence (after adjustment for confounders) as the model input, the impact on the BMD point estimate can be expected to be negligible.
  4. It is the precision of the BMD estimate (and thereby the BMDL and BMDU) that is expected to be more affected by variation in the source population size.
  5. No sensitivity analysis was deemed necessary regarding the case estimates since the case ascertainment and capture in the critical study was considered of low risk of bias.

Results in Table 37 show that for both studies (Gilbert‐Diamond et al., 2013; Steinmaus et al., 2014b) the assessed decreases or increases in population size have negligible effects on BMD estimates and the probability of exceeding these BMDs. It should also be noted that since the case–control study uses a sample of controls from the source population to assess the distribution of exposure, the risk estimate is less precise than it is when the whole source population is used. However, the loss of precision is small if the number of controls is large (Rothman et al., 2008). For the case–control study by Gilbert‐Diamond et al. (2013), the use of a sample of controls instead of the entire population decreases the lower confidence limit in the risk estimate by about 20%.

3.7.4. Summary on uncertainties

It is likely that the 95th percentile of exposure exceeds the estimated BMD based on Gilbert‐Diamond et al. (2013). Considering the mean exposure, the quantitative analysis suggests that it is unlikely that the estimated BMD is exceeded. The other study on skin cancer suggests the BMD is likely to be exceeded (Leonardi et al., 2012). Accounting for sensitivity analyses and qualitative consideration of the impact of remaining uncertainty, the CONTAM Panel considers that conclusions based on the approximate probability scale are relevant with respect to the general population.

4. CONCLUSIONS

4.1. General information

The metalloid arsenic is widely present as an environmental contaminant both from natural occurrence and anthropogenic activity. Arsenic occurs in various organic and inorganic forms. In food and feed iAs species are predominantly in the +3 or + 5 oxidation state, present as thio complexes or, as the oxo anions arsenite and arsenate. During sample preparation and analysis iAs bound to thio groups in peptides or proteins in food is converted to arsenite and arsenate and hence data on occurrence are nearly exclusively recorded as these two species. The sum of the quantified arsenite and arsenate in food (in mg As/kg wet weight) is often referred to as iAs. Determination of iAs consists of three main aspects: extraction, separation and element‐specific detection.

4.2. Toxicokinetics

  • In humans, iAs is rapidly and to a large extent (45%–80%) absorbed after ingestion. It is widely distributed in the body in almost all organs and readily crosses the placental barrier. Little is transferred to human milk. It is metabolised in humans by reduction, oxidative methylation, thiolation and glutathiolation. The degree of methylation is crucial for its toxic effects. It is eliminated via the urine in the form of iAs and its methylated metabolites, with a half‐life of around 2–3 days.
  • The toxicokinetics of laboratory animals differ considerably from those of humans in particular with regard to their methylation capacity and thus results from animal experiments are not a suitable basis for human health risk assessment.

4.3. Biomarkers of exposure

  • There is no universally accepted biomarker for chronic iAs exposure.
  • Measurements of total arsenic in blood or urine represent not only iAs but also organic arsenic species (e.g. originating from seafood consumption) and are thus of limited value.
  • Specific measurements of iAs, and its methylated metabolites, or the sum of iAs and its methylated metabolites, also named total urinary iAs (u‐tiAs), provide more appropriate estimates of iAs exposure. If continuous iAs exposure is stable over time, urinary arsenic may be used as a biomarker provided that arsenic species or u‐tiAs are analysed.
  • Inorganic arsenic accumulates in hair and nails. Thus, arsenic in hair and nails may also be used as biomarker for chronic iAs exposure.

4.4. Markers of genotoxicity

  • Chronic exposure to iAs via contaminated water in adulthood as well as in early life is associated with increased DNA damage (DNA breaks and oxidatively induced DNA base modifications) and clastogenic events in somatic cells from exposed individuals.

4.5. Markers of epigenetic effects

  • In utero or adult life chronic exposure to low levels of arsenic in drinking water is significantly associated with epigenetic alteration of DNA methylation levels in blood cells.

4.6. In vitro and in vivo genotoxicity

  • Although iAs does not directly interact with DNA, it induces oxidative stress, which can play a role in the formation of DNA base oxidation as well as both DNA single and double strand breaks. The production of oxidative clustered DNA lesions is likely responsible for the occurrence of double strand breaks.
  • Arsenic interferes with DNA damage response at two levels: by inhibition of DNA repair and by interference with cell cycle control and apoptotic pathways. This is likely to account for the co‐mutagenic effects of iAs when combined with exposure to DNA‐damaging agents (e.g. UV radiation or benzo(a)pyrene).
  • Inorganic arsenic is a weak mutagen but effectively induces clastogenic and aneugenic effects both in vitro and in vivo as measured by the occurrence of chromosomal aberrations, micronuclei and aneuploidy.

The methylation of iAs, particularly to trivalent methylated species, should be regarded as an activation process that forms more reactive species, which exert stronger cyto‐ and genotoxic effects.

4.7. Carcinogenicity in studies with animals

  • Although some studies have shown increased tumour incidence following oral (drinking water) administration of iAs, the results of different studies with respect to tumour sites and doses are inconsistent and do not provide a robust basis for use in risk assessment.

4.8. Observations in humans and selection of critical studies

  • The CONTAM Panel only considered studies including study subjects with exposure to long‐term low to moderate levels of arsenic, defined as concentrations of arsenic in water of less than ~ 150 μg/L, or biomarker concentrations estimated to result from equivalent doses.
  • The CONTAM Panel considers the association between low to moderate exposure to iAs and cancers of the skin, bladder and lung to be sufficient and causal.
  • The CONTAM Panel considers the association between low to moderate exposure to iAs and an increased risk for spontaneous abortion, stillbirth, infant mortality, congenital heart disease, neurodevelopmental effects, ischemic heart disease, respiratory disease, chronic kidney disease and atherosclerosis to be sufficient and causal.
  • The CONTAM Panel considers the evidence from epidemiological studies sufficient and causal for an association between low to moderate exposure to iAs and increased risk of skin lesions, i.e. pigmentation changes and hyperkeratosis. However, since the respective epidemiological studies were performed in low and medium income regions (Bangladesh, China) and nutrition and health status are important modifying factors, it is difficult to translate these risks to populations with more adequate nutrition, such as in Europe.
  • The evidence from epidemiological studies performed in Bangladesh is sufficient to assume a causal association between low to moderate exposure to iAs and decreased birth weight. Results from other countries (Chile, Taiwan, Mongolia, Mexico, US) are mixed and thus the overall evidence is therefore insufficient. The average birth weight in Europe is much higher and undernutrition is less common. Therefore, the relevance of the evidence from Bangladesh in a European setting is unclear.
  • There is insufficient evidence for an association between low to moderate exposure to iAs and breast, prostate, kidney, liver, pancreatic and gallbladder cancer, male fertility, neurotoxicity, stroke and hypertension, glucose metabolism, diabetes and metabolic syndrome.

4.9. Dose response analysis approach

  • Data from studies that were considered for dose–response modelling had to meet three criteria: (i) the overall risk of bias was considered low, (ii) the statistical analysis on the association between iAs exposure and the risk of the outcome reported by the authors had to show a statistically significant association with iAs as a continuous variable, a statistically significant trend test and/or a statistically significant increase of risk in the upper exposure category, (iii) results for at least three exposure categories (including the reference category) had to be reported.
  • In the studies selected for dose–response modelling the CONTAM Panel first transformed iAs concentrations in drinking water (applying region specific exposure estimates from food) or urine (u‐tiAs) to daily total iAs dietary exposures per kg bw per day for use in BMD calculations.
  • The data analysed are adjusted incidences and resulting number of cases based on the risk ratios reported in these studies and the provided (cohort studies) or estimated (case–control studies) source population sizes.
  • As BMR, a relative increase of the background incidence after adjustment for confounders by 5% was used.
  • Overall, 20 of the identified potentially critical studies fulfilled all the above criteria and the respective BMD modellings that met the criteria specified in the EFSA BMD guidance (EFSA Scientific Committee, 2022) were used as the basis for the selection of an appropriate RP.

4.10. Identification of a reference point

  • The lowest BMDLs (< 0.1 μg/kg bw per day) based on a BMR of 5% (relative increase of the background incidence after adjustment for confounders) were calculated for skin cancer, skin lesions, respiratory disease and chronic kidney disease.
  • Low BMDLs (≤ 0.17 μg/kg per day) were also calculated for lung cancer, bladder cancer and ischemic heart disease.
  • The CONTAM Panel decided to use the BMDL05 (0.06 μg/kg bw per day) from a case–control study on skin cancer (type: squamous cell carcinoma) in the US, as a Reference Point (RP). The results from a second skin cancer study (type: basal cell carcinomas) performed in Hungary, Romania and Slovakia support the choice for an RP for skin cancer for hazard assessment.
  • The CONTAM Panel concluded that an RP of 0.06 μg iAs/kg bw per day should be considered also to cover lung cancer, bladder cancer, skin lesions, ischemic heart disease, chronic kidney disease, respiratory disease, spontaneous abortion, stillbirth, infant mortality and neurodevelopmental effects.

4.11. Consideration of the approach to risk characterisation

  • Inorganic arsenic is a genotoxic carcinogen. Both thresholded and non‐thresholded mechanisms could apply to the different genotoxic effects of iAs and its trivalent and pentavalent methylated metabolites. Therefore, the CONTAM Panel concluded that it is not appropriate to establish a health‐based guidance value (HBGV) and a MOE approach has been applied in the risk characterisation.

4.12. Risk characterisation

  • There are no precedents in EFSA for identification of an MOE of low concern, when using a BMDL derived from human cancer data. Therefore, the Panel decided not to determine a value for an MOE of low concern.
  • The Reference Point (0.06 μg iAs/kg bw per day) identified by the CONTAM Panel for skin cancer is within the range of mean dietary exposure estimates for iAs in adults (0.03–0.15 μg/kg bw per day) and below any of the 95th percentile exposure estimates in adults (range = 0.07–0.33 μg iAs/kg bw per day). in Europe. Therefore, in adults, the MOEs range between 2 and 0.4 for mean consumers and between 0.9 and 0.2 at the 95th percentile exposure, respectively. An MOE of 1 describes the exposure level that could be associated with a 5% increase relative to the background incidence for skin cancer, based on the available data. Despite the uncertainties, the CONTAM Panel concludes that these MOEs raise a health concern.
  • The CONTAM Panel notes that dietary exposure is higher in the younger age groups and therefore the respective MOEs are smaller. However, this does not necessarily indicate that children are at greater risk, because the effects are due to long‐term exposure and most of the epidemiological studies are conducted in adults who would also have had higher dietary exposure during early life. Therefore, the CONTAM Panel concludes that children are covered by this risk characterisation.
  • Although risk characterisation is based on the results of relatively large epidemiological studies, susceptible individuals of higher genetic risk may not be adequately represented in these studies. Therefore, dietary exposure to arsenic may be is of greater concern for such individuals than for the general population.

4.13. Overall uncertainty in the risk characterisation

  • Based on the conditional uncertainty analysis, and considering both studies on skin cancer the probability that mean exposure scenario exceeds the associated BMDs range from unlikely (likelihood ≈ 0.17) to likely (likelihood ≈ 0.86).

5. RECOMMENDATIONS

  • The CONTAM Panel notes that an EFSA guidance on the use of human data for risk assessments is needed, and in particular on BMD modelling of epidemiological data and for a quantitative risk assessment for genotoxic carcinogens based on epidemiological data is needed.
  • The relevance of arsenic‐induced epigenetic alterations to disease risk in exposed populations needs to be investigated.
  • The interplay between epigenetic and genetic alterations induced by iAs deserves to be further investigated.
  • The mechanisms of induction of DNA double strand breaks by arsenic should be investigated to clarify its mode of interaction with DNA.
  • Further investigation is needed to address the mechanisms responsible for the induction of genomic instability by iAs. The health effects of pre‐ and perinatal exposure to arsenic and how arsenic‐induced alterations occurring during early life can impact the risk of disease in adult life should be further investigated.
  • The role of inter‐individual variations in the susceptibility to arsenic‐related health conditions should be investigated with a focus on arsenic biotransformation and differences in DNA repair.
  • Several recommendations regarding the dietary exposure assessment for iAs were made in the 2021 EFSA scientific report, which are still valid.

ABBREVIATIONS

γH2AX
serine139‐phosphorylated histone H2AX
8‐OHdG
8‐Hydroxy‐2’‐Deoxyguanosine
8‐oxodG
8‐hydroxydeoxyguanosine
A
adenine
ABI
Ankle‐Barchial Index
ADHD
attention deficit hyperactivity disorder
Adj
adjusted
ADME
absorption, distribution, metabolism and excretion
ADP
adenosine diphosphate
AES
atomic emission spectrometry
aHR
adjusted hazard ratio
Akt
protein kinase B
ALP
alkaline phosphatase
ALT
alanine aminotransferase
AO
adverse outcome
AOP
adverse outcome pathway
aOR
adjusted odds ratio
AP
apurinic/apyrimidinic
APE1
apurinic/apyrimidinic endonuclease 1
APOE ε4
apolipoprotein E ε4‐allele
As
arsenic
As(III)
arsenite
As(V)
arsenate
AsB
arsenobetaine
AsMat
Health Consequences of Arsenic in Matlab
As2O3
arsenic trioxide
AS3MT
arsenic(III)‐methyltransferase
ASCVD
atherosclerotic cardiovascular disease
ASD
autism spectrum disorder
ASHRAM
Arsenic Health Risk Assessment and Molecular Epidemiology
AST
aspartate transaminase
ATG
arsenic triglutathione
ATPase
adenosine triphosphatase
AWHS
Aragon Workers' Health Study
BCC
basal cell carcinoma
BDNF
brain‐derived neurotrophic factor
BEAS‐2B
bronchial epithelial airway sensitive‐2B (normal human bronchial epithelial cell line)
BER
base excision repair
BfR
German Bundesinstitut für Risikobewertung
B‐Hg
blood mercury
BMD
benchmark dose
BMDL
benchmark dose lower confidence limit
BMDLx
benchmark dose lower confidence limit for an extra/relative risk of X% (specified in the context)
BMDU
benchmark dose upper confidence limit
BMDUx
benchmark dose upper confidence limit for an extra/relative risk of X% (specified in the context)
BMI
body mass index
BMR
benchmark response
BP
blood pressure
BSID‐III
Bayley Scales of Infant and Toddler Development, Third Edition
bw
body weight
BW
birthweight
CA
chromosomal aberration
CAC
coronary artery calcium
CAE
cumulative arsenic exposure
CAK
CDK‐activating kinase
CANTAB
Tests from Cambridge Neuropsychological Test Automated Battery
CARDIA
Coronary Artery Risk Development in Young Adults
CARS
Childhood Autism Rating Scale
CAT
catalase
CC16
clara cell secretory protein
CCA
common carotid artery
Cd
cadmium
CFA
colony‐forming ability
CD4/CD8
types of T helper cells
CHD
coronary heart disease
CI
confidence interval
CKD
chronic kidney disease
CM‐H2DCFDA
chloromethyl derivative of H2DCFDA
CNS
central nervous system
CNV
copy number variation
Codex
Codex Alimentarius Commission
conc.
concentration
cont
control
CONTAM
the Panel on Contaminants in the Food Chain
COT
UK Committee on Toxicity of Chemicals in Food, Consumer Products and the Environment
CPD
cyclobutane pyrimidine dimer
CpG
cytosine‐phosphate‐guanine
Cr
creatinine
CRP
C‐reactive protein
CT
computed tomography
Cu
copper
CVD
cardiovascular disease
DATA Unit
EFSA Evidence Management Unit
DBP
diastolic blood pressure
DCFH‐DA
2′,7′ dichloro‐dihydrofluorescein diacetate
DDR
DNA damage response
DL
detection level
DM
diabetes mellitus
DMA
sum of DMA(III) and DMA(V)
DMA(III)
dimethylarsinite, dimethylarsinous acid
DMA(V)
dimethylarsinate, dimethylarsinic acid
DMAG
dimethylarsinic glutathione
DMMTA
dimethylmonothioarsenate
DN
double negative
DNMT
deoxyribonucleic acid methyltransferase
DP
double positive
DSB
double strand break
ECA
external carotid artery
ECG
electrocardiogram
EC‐HPLC
high‐performance liquid chromatography with electrochemical detection
EEC
European Economic Community
eGFR
estimated glomerular filtration rate
EGR1
early growth response factor 1
ELISA
enzyme linked immunosorbent assay
EMT
epithelial‐to‐mesenchymal transition
EPIC
European Prospective Investigation into Cancer and Nutrition
ERCC2
excision repair cross‐complementation group 2
ESRD
end stage renal disease
ETS
environmental tobacco smoke
EWAS
epigenome‐wide association study
F0,1,2,3
filial generation 0, 1, 2, 3
FANCD2
Fanconi anaemia complementation group D2
FANCL
Fanconi anaemia complementation group L
FAO
Food and Agriculture Organization
FD
fluorescence detection
FDA
Food and Drug Administration (United States)
FeNo
fractional exhaled nitric oxide
FEV
forced expiratory volume
FOX
Folate and Oxidative Stress
FPG
formamidopyrimidine DNA glycosylase
FSMP
food for special medical purpose
FVC
forced vital capacity
GA
gestational age
GC
gas chromatography
GCLC
glutamate‐cysteine ligase catalytic subunit
GD
gestational diabetes
GDM
gestational diabetes mellitus
gH2AX
serine139‐phosphorylated histone H2AX
GI
gastrointestinal
GIA
general intellectual abilities
GIS
geographic information system
GM
geometric mean
GSH
glutathione
GST
glutathione S‐transferase
GSTO1
glutathione S‐transferase omega 1
GTCC
Genetic Toxicology Technical Committee
Gy
Grey
GW
gestational week
H2AX
H2A histone family member X
H2O2
hydrogen peroxide
HaCaT
human epidermal keratinocyte line
HAV
hepatitis A virus
HAZ
height‐for‐age‐z‐score
HbA1c
glycolated haemoglobin A1c
HBGV
health‐based guidance value
HBV
hepatitis B virus
HDL
high‐density lipoprotein
HEALS
Health Effects of Arsenic Longitudinal Study
HeLa cells
human cell line named after Henrietta Lacks
HepG2
human hepatoblastoma cells
HESI
Health and Environmental Sciences Institute
HG‐AAS
hydride generation atomic absorption spectrometry
hOGG1
human 8‐oxoguanine DNA glycosylase 1
HOMA‐IR
Homeostatic Model Assessment of Insulin Resistance
HOME
Home Observation Measurement of the Environment
HPLC
high‐performance liquid chromatography
HPLC‐ED
high‐performance liquid chromatography with electrochemical detection
HR
hazard ratio
HR
homologous recombination
HT
hypertension
iAs
inorganic arsenic
iAs(III)
arsenite
iAs(V)
arsenate
IED
intra‐dimensional/extra‐dimensional shift task
IgE
immunoglobulin E
IFG
impaired fasting glucose
IgG
immunoglobulin G
IARC
International Agency for Research on Cancer
IC50
inhibitory concentration 50%
ICA
internal carotid artery
ICL
inter‐strand DNA cross‐link
ICP–MS
inductively coupled plasma mass spectrometry
IHD
ischemic heart disease
IMR‐90 cells
human lung fibroblasts
IMT
intima‐media thickness
IOM
Institute of Medicine
IQR
interquartile range
IQ
intelligence quotient
IRR
incidence rate ratio
iSumAs
sum of inorganic and methylated arsenic
JECFA
Joint FAO/WHO Expert Committee on Food Additives
LB
lower bound
LBW
low birth weight
LC–FD
liquid chromatography coupled to fluorescence detector
LC–MS
liquid chromatography coupled to mass spectrometry
LC–MS/MS
liquid chromatography–tandem mass spectrometry
LD50
lethal dose killing 50% of the animals
LDL
low‐density lipoprotein
LIG
DNA ligase
LINE‐1
long interspersed nucleotide element‐1
LLN
lower limit of normal
ln
natural logarithm
LOAEC
lowest‐observed‐adverse‐effect‐concentration
LOAEL
lowest‐observed‐adverse‐effect‐level
LOD
limit of detection
LOEL
lowest‐observed‐effect‐level
log
logarithmic
LOQ
limit of quantification
LS‐174T
human epithelial colorectal adenocarcinoma cells
MAT
Math Achievement Test
MEF
mouse embryonic fibroblasts
MESA
Multi‐Ethnic Study of Atherosclerosis
MeSH
Medical Subject Headings
MetS
metabolic syndrome
MG
macroglobulin
MI
myocardial infarction
MINIMat
Maternal and Infant Nutrition Interventions in Matlab
miRNA
micro ribonucleic acid
ML
maximum level
MMA
sum of MMA(III) and MMA(V)
MMA(III)
methylarsonite, monomethylarsonous acid
MMA(V)
methylarsonate, monomethylarsonic acid
MMF
maximum mid‐expiratory flow
MMAG
monomethylarsonic diglutathione
MMP9
matrix metalloproteinase 9
MMSE
Mini‐Mental State Examination
MN
micronucleus
MoA
mode of action
MOE
margin of exposure
MSCA
McCarthy Scales of Children's Abilities
mRNA
messenger ribonucleic acid
MS
mass spectrometry
MS/MS
tandem mass spectrometry
MTS
3‐(4,5‐dimethylthiazol‐2‐yl)‐5‐(3‐carboxymethoxyphenyl)‐2‐(4‐sulfophenyl)‐2H‐tetrazolium
MTT
3‐(4,5‐dimethylthiazol‐2‐yl)‐2,5‐diphenyl‐2H‐tetrazolium bromide
N7‐MeG
N7‐methylguanine
NA
not applicable
NAC
N‐acetyl‐l‐cysteine
NAFLD
non‐alcoholic fatty liver disease
NAG
N‐acetyl‐b‐d‐glucosaminidase
NCEP
National Cholesterol Education Program
NER
nucleotide excision repair
NFG
normal fasting glucose
NHANES
National Health and Nutrition Examination Survey
NHBE
normal human bronchial epithelial cells
NHBF
normal human bronchial fibroblast cells
NHEJ
nonhomologous end‐joining
NLM
the U.S. National Library of Medicine
NOAEL
no‐observed‐adverse‐effect‐level
NOEL
no‐observed‐effect‐level
NS
not significant
NTD
neural tube defect
OECD
Organisation for Economic Co‐operation and Development
OFC
orofacial cleft
OGG1
8‐oxoguanine DNA glycosylase 1
OR
odds ratio
P95
95th percentile
PAH
polycyclic aromatic hydrocarbon
PARPzf
the first zinc finger of PARP‐1
PARP‐1
poly (adenosine diphosphate‐ribose) polymerase 1
Pb
lead
PChE
plasma cholinesterase
PEF
peak expiratory flow rate
PERCH
Pneumonia Etiology Research for Child Health
PFGE
pulse field gel electrophoresis
PIR
Poverty Income Ratio
PLK1
polo‐like kinase 1
PM
particulate matter
PMI
primary methylation index
PCR
polymerase chain reaction
Pol β/POLB
DNA polymerase beta
PP
photoproduct
ppb
parts per billion
ppm
parts per million
PPVT
Peabody Picture Vocabulary Test
PTB
preterm birth
PTM
post translational modification
PTWI
provisional tolerable weekly intake
Q
quantile
RA
risk assessment
RBC
red blood cells
RC
risk characterisation
RCC
relative cell count
REGARDS
the REasons for Geographic and Racial Differences in Stroke
RNS
reactive nitrogen species
RONS
reactive oxygen and nitrogen species
ROS
reactive oxygen species
RP
reference point
RR
relative risk or risk ratio
RT‐PCR
reverse transcriptase‐polymerase chain reaction
RWPE‐1
immortalised normal prostate epithelial cells
SAM
S‐adenosylmethionine
sBDNF
serum brain‐derived neurotrophic factor
SBP
systolic blood pressure
SCC
squamous cell carcinoma
SD
standard deviation
Se
selenium
SE
standard error
SEM
standard error of the mean
SES
socioeconomic status
SG
specific gravity
SHFS
Strong Heart Family Study
SHS
Strong Heart Study
SLVDS
San Luis Valley Diabetes Study
SMI
secondary methylation index
SMR
standardised mortality ratio
SOC
Stockings of Cambridge
SOCS3
suppressor of cytokine signalling 3
SOD
superoxide dismutase
SSB
single strand break
SSP
Spatial Span
T
tertile
T2D
type 2 diabetes
tAs
total arsenic
TDI
tolerable daily intake
thio‐DMA(V)
thiodimethylarsinic acid
TK
toxicokinetics
TMC
total motor composite score
TMICS
Taiwan Maternal and Infant Cohort Study
ToR
terms of reference
TWA
time‐weighted average
TWI
tolerable weekly intake
U
uncertainty or enzyme unit
u‐As
urinary arsenic
u‐AsB
urinary arsenobetaine
UB
upper bound
u‐creatinine
urinary creatinine
u‐DMA
urinary DMA
UF
uncertainty factor
u‐iAs
urinary inorganic arsenic
u‐tiAs
total urinary inorganic arsenic (sum of iAs and its methylated metabolites MMA and DMA)
u‐MMA
urinary MMA
UROtsa
immortalised human urothelial cells
UV
ultraviolet light
UVC
ultraviolet light C
UVR
ultraviolet radiation
VC
vital capacity
VH10hTert
tert‐immortalised human diploid skin fibroblasts
VLBW
very low birth weight
w‐As
water‐arsenic
WG
working group
WHO
World Health Organization
WHR ratio
waist‐to‐hip ratio
WISC‐IV
Wechsler Intelligence Scale for Children 4th edition
WISC‐RM
Wechsler Intelligence Scale for Children Revised Mexican Version
WOSL
without skin lesions
WSL
with skin lesions
wt
wild‐type
XPC
Xeroderma Pigmentosum Complementation Group C
XPE
Xeroderma Pigmentosum Complementation Group E
XRCC1
X‐ray repair cross‐complementing protein 1
Zn
zinc

CONFLICT OF INTEREST

If you wish to access the declaration of interests of any expert contributing to an EFSA scientific assessment, please contact ue.aporue.asfe@tnemeganamtseretni.

REQUESTOR

European Commission

QUESTION NUMBER

EFSA‐Q‐2021‐00250

COPYRIGHT FOR NON‐EFSA CONTENT

EFSA may include images or other content for which is does not hold copyright. In such cases, EFSA indicates the copyright holder and users should seek permission to reproduce the content from the original source.

PANEL MEMBERS

Margherita Bignami, Laurent Bodin, James Kevin Chipman, Jesús del Mazo, Bettina Grasl‐Kraupp, Christer Hogstrand, Laurentius (Ron) Hoogenboom, Jean‐Charles Leblanc, Carlo Stefano Nebbia, Elsa Nielsen, Evangelia Ntzani, Annette Petersen, Salomon Sand, Dieter Schrenk, Tanja Schwerdtle, Christiane Vleminckx, and Heather Wallace.

Supporting information

Protocol for risk assessment

Literature searches

Benchmark dose modelling reports

Benchmark dose modelling reports

Benchmark dose modelling reports

Benchmark dose modelling reports

Benchmark concentration modelling reports

Outcome of the public consultation

ACKNOWLEDGEMENTS

The CONTAM Panel wishes to thank Federico Cruciani, Angelos Rizos and Katja Schirmer for the support provided to this scientific output.

Annexes A. AnnexProtocol for risk assessment

A.1. 

Annex A is provided as a separate pdf file containing the risk assessment protocol selected by the CONTAM Panel to update the previous risk assessments of inorganic arsenic.

Annex B. Literature searches

B.1. 

Annex B is provided as a separate pdf file, and it contains the details of the comprehensive literature searches conducted for the risk assessment of inorganic arsenic.

Annex C. Risk of bias analysis

C.1. 

Annex C is provided as a separate Excel file containing the harmonised outcome of the two independent analyses and is available on the EFSA Knowledge Junction community on Zenodo at: https://doi.org/10.5281/zenodo.10459723.

Annex D. Exposure estimates and transformations

D.1. 

Annex D is provided in form of two separate Excel files, Annex D1 and Annex D2. It includes conversion of water and urine iAs concentrations to iAs exposures, as well as transformations of relative risk estimates to quantal data. The Annexes D1 and D2 are available on the EFSA Knowledge Junction community on Zenodo at the following links: https://doi.org/10.5281/zenodo.10459767. (D1).

Annex E. BMD reports

E.1. 

Annex E is provided as five separate pdf files for BMD reports (Annex E1: BMR of 5%, Annex E2: BMR of 1%, Annex E3: BMR of 10% and Annex E4: BMR of 50% and BMR (relative increase of the background incidence after adjustment for confounders). In addition, the BMC reports for the alternative modelling approach (used only for the uncertainty analysis) using BMR of 5% (relative increase of the background incidence after adjustment for confounders) are provided in Annex E5.

Annex F. BMD summary table

F.1. 

Annex F is provided as a separate Excel file that presents a summary of the BMD analyses (BMRs of 1%, 5%, 10% and 50% as relative increase of the background incidence after adjustment for confounders). It includes important modelling outcomes such as BMD, BMDL and BMDU values, and compares these results to the criteria specified in the EFSA BMD guidance (EFSA Scientific Committee, 2022). Study‐specific details and BMD input parameters are also provided. The Annex is available on the EFSA Knowledge Junction community on Zenodo at: https://doi.org/10.5281/zenodo.10460959.

Annex G. Outcome of the public consultation

G.1. 

Annex G provides responses to the public consultation comments along with the original comments, all within a separate pdf file.

Annexes A, B, E and G can be found in the online version of this output (in the ‘Supporting information’ section).

Notes

EFSA CONTAM Panel (EFSA Panel on Contaminants in the Food Chain) , Schrenk, D. , Bignami, M. , Bodin, L. , Chipman, J. K. , del Mazo, J. , Grasl‐Kraupp, B. , Hogstrand, C. , Hoogenboom, L. R. , Leblanc, J.‐C. , Nebbia, C. S. , Nielsen, E. , Ntzani, E. , Petersen, A. , Sand, S. , Vleminckx, C. , Wallace, H. , Barregård, L. , Benford, D. , … Schwerdtle, T. (2024). Update of the risk assessment of inorganic arsenic in food. EFSA Journal, 22(1), e8488. 10.2903/j.efsa.2023.8488 [CrossRef] [Google Scholar]

Adopted: 18 December 2023

Note: A plain language summary of this scientific opinion is available at https://efsa.onlinelibrary.wiley.com/doi/10.2903/j.efsa.2024.P220101

Notes

1 EFSA (European Food Safety Authority), 2014. Scientific report of on the dietary exposure to inorganic arsenic in the European population. EFSA Journal, 12(3), 3597.

2 Commission Recommendation (EU) 2015/1381 of 10 August 2015 on the monitoring of arsenic in food. OJ L 213, 12.8.2015, p. 213.

3 Council Regulation (EEC) No 315/93 of February 1993 laying down Community procedures for contaminants in food. OJ L 37, 13.2.1993, p. 1–5.

4 Commission Regulation (EU) 2023/915 of 25 April 2023 on maximum levels for certain contaminants in food and repealing Regulation (EC) No 1881/2006 OJ L 119/103, 5.5.2023, p. 103–107.

5 Council Directive 98/83/EC of 3 November 1998 on the quality of water intended for human consumption. OJ L 330, 5.12.1998, p. 32.

6 Directive (EU) 2020/2184 of the European Parliament and of the Council on the quality of water for human consumption of 16 December 2020. OJ 435, 23.12.2020, p. 1–62.

7 Commission Directive 2003/40/EC establishing the list, concentration limits and labelling requirements for the constituents of natural mineral waters and the conditions for using ozone‐enriched air for the treatment of natural mineral waters and spring waters of 16 May 2003. OJ L 126, 22.5.2003, p. 34.

8 https://www.fao.org/news/story/it/item/238558/icode/

9 https://www.fao.org/news/story/en/item/419100/icode/

10 Web of Science (WoS), formally ISI Web of Knowledge, Thomson Reuters. https://thomsonreuters.com/thomson‐reuters‐webof‐science/

11 https://www.ncbi.nlm.nih.gov/

12 EndNote X6, Thomson Reuters. https://endnote.com/

13 Identifying articles that have been cited in articles found in a search (see Jalali & Wohlin, 2012).

14 A chronic health condition arising from prolonged ingestion (not less than 6 months) of arsenic above a safe dose, usually manifested by characteristic skin lesions, with or without involvement of internal organs.

15 A condition characterised by an increased thickness of the stratum corneum, the outer layer of the skin.

16 Cells are exposed to a DNA damaging agent in vitro to investigate how they respond.

17 Formerly termed transitional cell carcinoma.

18 The NRC in their publication used the term ‘ED01’ as the dose corresponding to a theoretical 1% excess risk of cancer mortality in a US population. The CONTAM Panel considered such a value as equivalent to a BMC01 since the NRC ED01 actually related to concentrations of arsenic in water and not doses.

19 Almost all samples reported as drinking water were analysed for total arsenic (7560 out of 7623). It was assumed that the reported levels on tAs correspond to iAs as it is well established that almost all arsenic in drinking water is inorganic, either As(III) or As(V) (US EPA, 2001; FAO/WHO, 2011).

20 Parametric value established by the Council Directive 98/83/EC for water intended for human consumption and ML established for natural mineral waters by Commission Directive 2003/40/EC.

21 As indicated in Section ‘3.4. Occurrence data’, the analytical data used in this exposure assessment are on iAs. For the data on ‘Drinking water’ the inmense majority of the data were reported as total arsenic but they are considered as iAs (US EPA, 2001; FAO/WHO, 2011).

REFERENCES

  • Abhyankar, L. N. , Jones, M. R. , Guallar, E. , & Navas‐Acien, A. (2012). Arsenic exposure and hypertension: A systematic review. Environmental Health Perspectives, 120, 494–500. 10.1289/ehp.1103988 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Adams, J. B. , Audhya, T. , McDonough‐Means, S. , Rubin, R. A. , Quig, D. , Geis, E. , Gehn, E. , Loresto, M. , Mitchell, J. , Atwood, S. , Barnhouse, S. , & Lee, W. (2013). Toxicological status of children with autism vs. neurotypical children and the association with autism severity. Biological Trace Element Research, 151, 171–180. 10.1007/s12011-012-9551-1 [PubMed] [CrossRef] [Google Scholar]
  • Afridi, H. I. , Kazi, T. G. , Kazi, N. , Kandhro, G. A. , Baig, J. A. , Jamali, M. K. , Arain, M. B. , Shah, A. Q. , Shah, F. , Khan, S. , & Kolachi, N. F. (2011). Association of Environmental Toxic Elements in biological samples of myocardial infarction patients at different stages. Biological Trace Element Research, 141, 26–40. 10.1007/s12011-010-8713-2 [PubMed] [CrossRef] [Google Scholar]
  • Ahamed, S. , Kumar Sengupta, M. , Mukherjee, A. , Amir Hossain, M. , Das, B. , Nayak, B. , Pal, A. , Chandra Mukherjee, S. , Pati, S. , Nath Dutta, R. , Chatterjee, G. , Mukherjee, A. , Srivastava, R. , & Chakraborti, D. (2006). Arsenic groundwater contamination and its health effects in the state of Uttar Pradesh (UP) in upper and middle ganga plain, India: A severe danger. Science of the Total Environment, 370, 310–322. 10.1016/j.scitotenv.2006.06.015 [PubMed] [CrossRef] [Google Scholar]
  • Ahmad, S. A. , Sayed, S. U. , Barua, S. , Khan, M. H. , Jalil, A. , Hadi, S. A. , & Talukder, H. K. (2001). Arsenic in drinking water and pregnancy outcomes. Environmental Health Perspectives, 109(6), 629–631. [PMC free article] [PubMed] [Google Scholar]
  • Ahmed, S. , Akhtar, E. , Roy, A. , von Ehrenstein, O. S. , Vahter, M. , Wagatsuma, Y. , & Raqib, R. (2017). Arsenic exposure alters lung function and airway inflammation in children: A cohort study in rural Bangladesh. Environment International, 101, 108–116. 10.1016/j.envint.2017.01.014 [PubMed] [CrossRef] [Google Scholar]
  • Ahsan, H. , Chen, Y. , Parvez, F. , Zablotska, L. , Argos, M. , Hussain, I. , Momotaj, H. , Levy, D. , Cheng, Z. , Slavkovich, V. , van Geen, A. , Howe, G. R. , & Graziano, J. H. (2006). Arsenic exposure from drinking water and risk of premalignant skin lesions in Bangladesh: Baseline results from the health effects of arsenic longitudinal study. American Journal of Epidemiology, 163(12), 1138–1148. 10.1093/aje/kwj154 [PubMed] [CrossRef] [Google Scholar]
  • Akbal, A. , Yılmaz, H. , & Tutkun, E. (2014). Arsenic exposure associated with decreased bone mineralization in male. The Aging Male, 17, 256–258. 10.3109/13685538.2013.819326 [PubMed] [CrossRef] [Google Scholar]
  • Al‐Ayadhi, L. Y. (2005). Heavy metals and trace elements in hair samples of autistic children in Central Saudi Arabia. Neurosciences (Riyadh), 10, 213–218. [PubMed] [Google Scholar]
  • Ali, N. , Hoque, M. A. , Haque, A. , Salam, K. A. , Karim, M. R. , Rahman, A. , Islam, K. , Saud, Z. A. , Khalek, M. A. , Akhand, A. A. , Hossain, M. , Mandal, A. , Karim, M. R. , Miyataka, H. , Himeno, S. , & Hossain, K. (2010). Association between arsenic exposure and plasma cholinesterase activity: A population based study in Bangladesh. Environmental Health, 9, 36. 10.1186/1476-069x-9-36 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Allen, B. , Shao, K. , Hobbie, K. , Mendez, W., Jr. , Lee, J. S. , Cote, I. , Druwe, I. , Gift, J. , & Davis, J. A. (2020). Systematic dose‐response of environmental epidemiologic studies: Dose and response pre‐analysis. Environment International, 142, 105810. 10.1016/j.envint.2020.105810 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Almberg, K. S. , Turyk, M. E. , Jones, R. M. , Rankin, K. , Freels, S. , Graber, J. M. , & Stayner, L. T. (2017). Arsenic in drinking water and adverse birth outcomes in Ohio. Environmental Research, 157, 52–59. 10.1016/j.envres.2017.05.010 [PubMed] [CrossRef] [Google Scholar]
  • Alshana, U. , Altun, B. , Ertaş, N. , Çakmak, G. , Kadioglu, E. , Hisarlı, D. , Aşık, E. , Atabey, E. , Çelebi, C. R. , Bilir, N. , Serçe, H. , Tuncer, A. M. , & Burgaz, S. (2022). Evaluation of low‐to‐moderate arsenic exposure, metabolism and skin lesions in a Turkish rural population exposed through drinking water. Chemosphere, 304, 135277. [PubMed] [Google Scholar]
  • Amadi, C. N. , Orish, C. N. , Frazzoli, C. , & Orisakwe, O. E. (2022). Association of autism with toxic metals: A systematic review of case‐control studies. Pharmacology, Biochemistry, and Behavior, 212, 173313. 10.1016/j.pbb.2021.173313 [PubMed] [CrossRef] [Google Scholar]
  • Amaral, A. F. , Porta, M. , Silverman, D. T. , Milne, R. L. , Kogevinas, M. , Rothman, N. , Cantor, K. P. , Jackson, B. P. , Pumarega, J. A. , López, T. , Carrato, A. , Guarner, L. , Real, F. X. , & Malats, N. (2012). Pancreatic cancer risk and levels of trace elements. Gut, 61(11), 1583–1588. 10.1136/gutjnl-2011-301086 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ameer, S. S. , Engström, K. , Harari, F. , Concha, G. , Vahter, M. , & Broberg, K. (2015). The effects of arsenic exposure on blood pressure and early risk markers of cardiovascular disease: Evidence for population differences. Environmental Research, 140, 32–36. 10.1016/j.envres.2015.03.010 [PubMed] [CrossRef] [Google Scholar]
  • Ameer, S. S. , Engström, K. , Hossain, M. B. , Concha, G. , Vahter, M. , & Broberg, K. (2017). Arsenic exposure from drinking water is associated with decreased gene expression and increased DNA methylation in peripheral blood. Toxicology and Applied Pharmacology, 321, 57–66. 10.1016/j.taap.2017.02.019 [PubMed] [CrossRef] [Google Scholar]
  • Amin, R. W. , Ross, A. M. , Lee, J. , Guy, J. , & Stafford, B. (2019). Patterns of ovarian cancer and uterine cancer mortality and incidence in the contiguous USA. Science of the Total Environment, 697, 134128. 10.1016/j.scitotenv.2019.134128 [PubMed] [CrossRef] [Google Scholar]
  • Amin, R. W. , Stafford, B. , & Guttmann, R. P. (2019). A spatial study of bladder cancer mortality and incidence in the contiguous US: 2000–2014. Science of the Total Environment, 670, 806–813. 10.1016/j.scitotenv.2019.03.290 [PubMed] [CrossRef] [Google Scholar]
  • Amster, E. D. , Cho, J. I. , & Christiani, D. (2011). Urine arsenic concentration and obstructive pulmonary disease in the US population. Journal of Toxicology and Environmental Health, Part A, 74(11), 716–727. [PMC free article] [PubMed] [Google Scholar]
  • Andrew, A. S. , Mason, R. A. , Kelsey, K. T. , Schned, A. R. , Marsit, C. J. , Nelson, H. H. , & Karagas, M. R. (2009). DNA repair genotype interacts with arsenic exposure to increase bladder cancer risk. Toxicology Letters, 187, 10–14. 10.1016/j.toxlet.2009.01.013 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Annangi, B. , Bonassi, S. , Marcos, R. , & Hernández, A. (2016). Biomonitoring of humans exposed to arsenic, chromium, nickel, vanadium, and complex mixtures of metals by using the micronucleus test in lymphocytes. Mutation Research, 770, 140–161. 10.1016/j.mrrev.2016.03.003 [PubMed] [CrossRef] [Google Scholar]
  • Antonelli, R. , Shao, K. , Thomas, D. J. , Sams, R. , & Cowden, J. (2014). AS3MT, GSTO, and PNP polymorphisms: Impact on arsenic methylation and implications for disease susceptibility. Environmental Research, 132, 156–167. 10.1016/j.envres.2014.03.012 [PubMed] [CrossRef] [Google Scholar]
  • Aposhian, V. H. , Zakharyan, R. A. , Avram, M. D. , Sampayo‐Reyes, A. , & Wollenberg, M. L. (2004). A review of the enzymology of arsenic metabolism and a new potential role of hydrogen peroxide in the detoxication of the trivalent arsenic species. Toxicology and Applied Pharmacology, 198(3), 327–335. [PubMed] [Google Scholar]
  • Arab YarMohammadi, A. , Arbabi Bidgoli, S. , & Ziarati, P. (2021). Increased urinary arsenic concentration in newly diagnosed type 2 diabetes mellitus: A gender‐independent, smoking‐dependent exposure biomarker in older adults in Tehran. Environmental Science and Pollution Research International, 28, 27769–27777. 10.1007/s11356-020-10261-w [PubMed] [CrossRef] [Google Scholar]
  • Arain, S. S. , Kazi, T. G. , Afridi, H. I. , Talpur, F. N. , Kazi, A. G. , Brahman, K. D. , Naeemullah, P. A. H. , & Kamboh, M. A. (2015). Correlation of arsenic levels in smokeless tobacco products and biological samples of Oral cancer patients and control consumers. Biological Trace Element Research, 168(2), 287–295. 10.1007/s12011-015-0355-y [PubMed] [CrossRef] [Google Scholar]
  • Argos, M. , Chen, L. , Jasmine, F. , Tong, L. , Pierce, B. L. , Roy, S. , Paul‐Brutus, R. , Gamble, M. V. , Harper, K. N. , Parvez, F. , Rahman, M. , Rakibuz‐Zaman, M. , Slavkovich, V. , Baron, J. A. , Graziano, J. H. , Kibriya, M. G. , & Ahsan, H. (2015). Gene‐specific differential DNA methylation and chronic arsenic exposure in an epigenome‐wide association study of adults in Bangladesh. Environmental Health Perspectives, 123, 64–71. 10.1289/ehp.1307884 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Argos, M. , Kalra, T. , Rathouz, P. J. , Chen, Y. , Pierce, B. , Parvez, F. , Islam, T. , Ahmed, A. , Rakibuz‐Zaman, M. , Hasan, R. , Sarwar, G. , Slavkovich, V. , van Geen, A. , Graziano, J. , & Ahsan, H. (2010). Arsenic exposure from drinking water, and all‐cause and chronic‐disease mortalities in Bangladesh (HEALS): A prospective cohort study. Lancet, 376, 252–258. 10.1016/s0140-6736(10)60481-3 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Argos, M. , Parvez, F. , Rahman, M. , Rakibuz‐Zaman, M. , Ahmed, A. , Hore, S. K. , Islam, T. , Chen, Y. , Pierce, B. L. , Slavkovich, V. , Olopade, C. , Yunus, M. , Baron, J. A. , Graziano, J. H. , & Ahsan, H. (2014). Arsenic and lung disease mortality in Bangladeshi adults. Epidemiology, 25, 536–543. 10.1097/ede.0000000000000106 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Arikan, I. , Namdar, N. D. , Kahraman, C. , Dagci, M. , & Ece, E. (2015). Assessment of arsenic levels in body samples and chronic exposure in people using water with a high concentration of arsenic: A field study in Kutahya. Asian Pacific Journal of Cancer Prevention, 16, 3183–3188. 10.7314/apjcp.2015.16.8.3183 [PubMed] [CrossRef] [Google Scholar]
  • Aschengrau, A. , Zierler, S. , & Cohen, A. (1989). Quality of community drinking water and the occurrence of spontaneous abortion. Archives of Environmental Health, 44, 283–290. 10.1080/00039896.1989.9935895 [PubMed] [CrossRef] [Google Scholar]
  • Ashley‐Martin, J. , Dodds, L. , Arbuckle, T. E. , Bouchard, M. F. , Shapiro, G. D. , Fisher, M. , Monnier, P. , Morisset, A. S. , & Ettinger, A. S. (2018). Association between maternal urinary speciated arsenic concentrations and gestational diabetes in a cohort of Canadian women. Environment International, 121, 714–720. 10.1016/j.envint.2018.10.008 [PubMed] [CrossRef] [Google Scholar]
  • Asprouli, E. , Kalafati, I. P. , Sakellari, A. , Karavoltsos, S. , Vlachogiannakos, J. , Revenas, K. , Kokkinos, A. , Dassenakis, M. , Dedoussis, G. V. , & Kalogeropoulos, N. (2019). Evaluation of plasma trace elements in different stages of nonalcoholic fatty liver disease. Biological Trace Element Research, 188, 326–333. 10.1007/s12011-018-1432-9 [PubMed] [CrossRef] [Google Scholar]
  • Baastrup, R. , Sørensen, M. , Balstrøm, T. , Frederiksen, K. , Larsen, C. L. , Tjønneland, A. , Overvad, K. , & Raaschou‐Nielsen, O. (2008). Arsenic in drinking‐water and risk for cancer in Denmark. Environmental Health Perspectives, 116, 231–237. 10.1289/ehp.10623 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bach, J. , Peremartí, J. , Annangi, B. , Marcos, R. , & Hernández, A. (2015). Reduced cellular DNA repair capacity after environmentally relevant arsenic exposure. Influence of Ogg1 deficiency. Mutation Research, 779, 144–151. 10.1016/j.mrfmmm.2015.07.004 [PubMed] [CrossRef] [Google Scholar]
  • Bach, J. , Sampayo‐Reyes, A. , Marcos, R. , & Hernández, A. (2014). Ogg1 genetic background determines the genotoxic potential of environmentally relevant arsenic exposures. Archives of Toxicology, 88, 585–596. 10.1007/s00204-013-1151-0 [PubMed] [CrossRef] [Google Scholar]
  • Bailey, K. A. , & Fry, R. C. (2012). In Sahu S. C. (Ed.), Arsenic‐induced changes to the Epigenome, in toxicology and epigenetics. Chichester. [Google Scholar]
  • Baj, J. , Flieger, W. , Flieger, M. , Forma, A. , Sitarz, E. , Skorzynska‐Dziduszko, K. , Grochowski, C. , Maciejewski, R. , & Karakula‐Juchnowicz, H. (2021). Autism spectrum disorder: Trace elements imbalances and the pathogenesis and severity of autistic symptoms. Neuroscience and Biobehavioral Reviews, 129, 117–132. 10.1016/j.neubiorev.2021.07.029 [PubMed] [CrossRef] [Google Scholar]
  • Bandyopadhyay, A. K. , Paul, S. , Adak, S. , & Giri, A. K. (2016). Reduced LINE‐1 methylation is associated with arsenic‐induced genotoxic stress in children. Biometals, 29, 731–741. 10.1007/s10534-016-9950-4 [PubMed] [CrossRef] [Google Scholar]
  • Banerjee, M. , Banerjee, N. , Bhattacharjee, P. , Mondal, D. , Lythgoe, P. R. , Martínez, M. , Pan, J. , Polya, D. A. , & Giri, A. K. (2013). High arsenic in rice is associated with elevated genotoxic effects in humans. Scientific Reports, 3, 2195. 10.1038/srep02195 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Banerjee, M. , Banerjee, N. , Ghosh, P. , Das, J. K. , Basu, S. , Sarkar, A. K. , States, J. C. , & Giri, A. K. (2010). Evaluation of the serum catalase and myeloperoxidase activities in chronic arsenic‐exposed individuals and concomitant cytogenetic damage. Toxicology and Applied Pharmacology, 249, 47–54. 10.1016/j.taap.2010.08.013 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Banerjee, M. , Sarma, N. , Biswas, R. , Roy, J. , Mukherjee, A. , & Giri, A. K. (2008). DNA repair deficiency leads to susceptibility to develop arsenic‐induced premalignant skin lesions. International Journal of Cancer, 123, 283–287. 10.1002/ijc.23478 [PubMed] [CrossRef] [Google Scholar]
  • Banerjee, N. , Bandyopadhyay, A. K. , Dutta, S. , Das, J. K. , Roy Chowdhury, T. , Bandyopadhyay, A. , & Giri, A. K. (2017). Increased microRNA 21 expression contributes to arsenic induced skin lesions, skin cancers and respiratory distress in chronically exposed individuals. Toxicology, 378, 10–16. 10.1016/j.tox.2017.01.006 [PubMed] [CrossRef] [Google Scholar]
  • Banerjee, N. , Das, S. , Tripathy, S. , Bandyopadhyay, A. K. , Sarma, N. , Bandyopadhyay, A. , & Giri, A. K. (2019). MicroRNAs play an important role in contributing to arsenic susceptibility in the chronically exposed individuals of West Bengal, India. Environmental Science and Pollution Research International, 26, 28052–28061. 10.1007/s11356-019-05980-8 [PubMed] [CrossRef] [Google Scholar]
  • Bao, Q.‐J. , Zhao, K. , Guo, Y. , Wu, X.‐T. , Yang, J.‐C. , & Yang, M.‐F. (2022). Environmental toxic metal contaminants and risk of stroke: A systematic review and meta‐analysis. Environmental Science and Pollution Research, 29, 32545–32565. 10.1007/s11356-022-18866-z [PubMed] [CrossRef] [Google Scholar]
  • Baris, D. , Waddell, R. , Beane Freeman, L. E. , Schwenn, M. , Colt, J. S. , Ayotte, J. D. , Ward, M. H. , Nuckols, J. , Schned, A. , Jackson, B. , Clerkin, C. , Rothman, N. , Moore, L. E. , Taylor, A. , Robinson, G. , Hosain, G. M. , Armenti, K. R. , McCoy, R. , Samanic, C. , … Silverman, D. T. (2016). Elevated bladder cancer in northern New England: The role of drinking Water and arsenic. Journal of the National Cancer Institute, 108. 10.1093/jnci/djw099 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Barregard, L. , Ellingsen, D. G. , Berlinger, B. , Weinbruch, S. , Harari, F. , & Sallsten, G. (2021). Normal variability of 22 elements in 24‐hour urine samples – Results from a biobank from healthy non‐smoking adults. International Journal of Hygiene and Environmental Health, 233, 113693. 10.1016/j.ijheh.2021.113693 [PubMed] [CrossRef] [Google Scholar]
  • Bartel, M. , Ebert, F. , Leffers, L. , Karst, U. , & Schwerdtle, T. (2011). Toxicological characterization of the inorganic and organic arsenic metabolite Thio‐DMA in cultured human Lung cells. Journal of Toxicology, 2011, 373141. 10.1155/2011/373141 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bartolotta, S. A. , Pacskowski, M. G. , Hick, A. , & Carballo, M. A. (2011). Micronuclei assay in exfoliated buccal cells from individuals exposed to arsenic in Argentina. Archives of Environmental Contamination and Toxicology, 61, 337–343. 10.1007/s00244-010-9607-1 [PubMed] [CrossRef] [Google Scholar]
  • Beane Freeman, L. E. , Dennis, L. K. , Lynch, C. F. , Thorne, P. S. , & Just, C. L. (2004). Toenail arsenic content and cutaneous melanoma in Iowa. American Journal of Epidemiology, 160(7), 679–687. [PubMed] [Google Scholar]
  • Becker, A. , & Azelrad, D. (2014). Arsenic and type 2 diabetes: Commentary on association of inorganic arsenic exposure with type 2 diabetes mellitus: A meta‐analysis by Wang et al. Journal of Epidemiology and Community Health, 68, 393–395. 10.1136/jech-2013-203463 [PubMed] [CrossRef] [Google Scholar]
  • Bedaiwi, A. , Wysong, A. , Rogan, E. G. , Clarey, D. , & Arcari, C. M. (2022). Arsenic exposure and melanoma among US adults aged 20 or older, 2003–2016. Public Health Reports, 137(3), 548–556. [PMC free article] [PubMed] [Google Scholar]
  • Benhusein, G. , Mutch, E. , Almezogi, J. , & Williams, F. (2016). In vitro genotoxic effect of arsenical compounds in HepG2 liver cells. British Journal of Pharmaceutical Research, 13, 1–10. 10.9734/BJPR/2016/28796 [CrossRef] [Google Scholar]
  • Bermúdez, L. , García‐Vicent, C. , López, J. , Torró, M. I. , & Lurbe, E. (2015). Assessment of ten trace elements in umbilical cord blood and maternal blood: Association with birth weight. Journal of Translational Medicine, 13, 291. 10.1186/s12967-015-0654-2 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • BfR (Bundesinstitut für Risikobewertung) . (2014). Arsenic in rice and rice products. BfR Opinion No. 018/2015 of 24 June 2014. https://mobil.bfr.bund.de/cm/349/arsenic‐in‐rice‐and‐rice‐products.pdf [Google Scholar]
  • Bhattacharjee, P. , Paul, S. , & Bhattacharjee, P. (2020). Understanding the mechanistic insight of arsenic exposure and decoding the histone cipher. Toxicology, 430, 152340. 10.1016/j.tox.2019.152340 [PubMed] [CrossRef] [Google Scholar]
  • Bhattacharjee, P. , Sanyal, T. , Bhattacharjee, S. , & Bhattacharjee, P. (2018). Epigenetic alteration of mismatch repair genes in the population chronically exposed to arsenic in West Bengal, India. Environmental Research, 163, 289–296. 10.1016/j.envres.2018.01.002 [PubMed] [CrossRef] [Google Scholar]
  • Bhattacharyya, P. , Sen, P. , Ghosh, A. , Saha, C. , Bhattacharya, P. P. , Das, A. , Majumdar, K. , & Mazumder, G. (2014). Chronic lung disease and detection of pulmonary artery dilatation in high resolution computerized tomography of chest in chronic arsenic exposure. Journal of Environmental Science and Health. Part A, Toxic/Hazardous Substances & Environmental Engineering, 49, 1453–1461. 10.1080/10934529.2014.937157 [PubMed] [CrossRef] [Google Scholar]
  • Björklund, K. L. , Vahter, M. , Palm, B. , Grandér, M. , Lignell, S. , & Berglund, M. (2012). Metals and trace element concentrations in breast milk of first time healthy mothers: A biological monitoring study. Environmental Health, 11, 92. 10.1186/1476-069x-11-92 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Blaurock‐Busch, E. , Amin, O. R. , & Rabah, T. (2011). Heavy metals and trace elements in hair and urine of a sample of Arab children with autistic spectrum disorder. Maedica, 6:247‐57. [PMC free article] [PubMed] [Google Scholar]
  • Blaurock‐Busch, E. , Amin, O. R. , Dessoki, H. H. & Rabah, T. (2012). Toxic metals and essential elements in hair and severity of symptoms among children with autism. Maedica (Bucur), 7, 38–48. [PMC free article] [PubMed] [Google Scholar]
  • Bloom, M. S. , Buck Louis, G. M. , Sundaram, R. , Maisog, J. M. , Steuerwald, A. J. , & Parsons, P. J. (2015). Birth outcomes and background exposures to select elements, the longitudinal investigation of fertility and the environment (LIFE). Environmental Research, 138, 118–129. 10.1016/j.envres.2015.01.008 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bloom, M. S. , Neamtiu, I. A. , Surdu, S. , Pop, C. , Anastasiu, D. , Appleton, A. A. , Fitzgerald, E. F. , & Gurzau, E. S. (2016). Low level arsenic contaminated water consumption and birth outcomes in Romania‐an exploratory study. Reproductive Toxicology, 59, 8–16. 10.1016/j.reprotox.2015.10.012 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bloom, M. S. , Surdu, S. , Neamtiu, I. A. , & Gurzau, E. S. (2014). Maternal arsenic exposure and birth outcomes: A comprehensive review of the epidemiologic literature focused on drinking water. International Journal of Hygiene and Environmental Health, 217, 709–719. 10.1016/j.ijheh.2014.03.004 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bocquet, A. , Dupont, C. , Chouraqui, J. P. , Darmaun, D. , Feillet, F. , Frelut, M. L. , Girardet, J. P. , Hankard, R. , Lapillonne, A. , Rozé, J. C. , Simeoni, U. , Turck, D. , & Briend, A. (2019). Efficacy and safety of hydrolyzed rice‐protein formulas for the treatment of cow's milk protein allergy. Archives de Pédiatrie, 26, 238–246. 10.1016/j.arcped.2019.03.001 [PubMed] [CrossRef] [Google Scholar]
  • Boei, J. J. W. A. , Vermeulen, S. , Skubakova, M. M. , Meijers, M. , Loenen, W. A. M. , Wolterbeek, R. , Mullenders, L. H. F. , Vrieling, H. , & Giphart‐Gassler, M. (2012). No threshold for the induction of chromosomal damage at clinically relevant low doses of X rays. Radiation Research, 177, 602–613. [PubMed] [Google Scholar]
  • Bonassi, S. , Norppa, H. , Ceppi, M. , Strömberg, U. , Vermeulen, R. , Znaor, A. , Cebulska‐Wasilewska, A. , Fabianova, E. , Fucic, A. , Gundy, S. , Hansteen, I. L. , Knudsen, L. E. , Lazutka, J. , Rossner, P. , Sram, R. J. , & Boffetta, P. (2008). Chromosomal aberration frequency in lymphocytes predicts the risk of cancer: Results from a pooled cohort study of 22 358 subjects in 11 countries. Carcinogenesis, 29, 1178–1183. 10.1093/carcin/bgn075 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Börzsönyi, M. , Bereczky, A. , Rudnai, P. , Csanady, M. , & Horvath, A. (1992). Epidemiological studies on human subjects exposed to arsenic in drinking water in Southeast Hungary. Archives of Toxicology, 66, 77–78. 10.1007/bf02307274 [PubMed] [CrossRef] [Google Scholar]
  • Bossak, K. , Goch, W. , Piątek, K. , Frączyk, T. , Poznański, J. , Bonna, A. , Keil, C. , Hartwig, A. , & Bal, W. (2015). Unusual Zn(II) affinities of zinc fingers of poly(ADP‐ribose)polymerase 1 (PARP‐1) nuclear protein. Chemical Research in Toxicology, 28, 191–201. 10.1021/tx500320f [PubMed] [CrossRef] [Google Scholar]
  • Bozack, A. K. , Cardenas, A. , Geldhof, J. , Quamruzzaman, Q. , Rahman, M. , Mostofa, G. , Christiani, D. C. , & Kile, M. L. (2020). Cord blood DNA methylation of DNMT3A mediates the association between in utero arsenic exposure and birth outcomes: Results from a prospective birth cohort in Bangladesh. Environmental Research, 183, 109134. 10.1016/j.envres.2020.109134 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bozack, A. K. , Saxena, R. , & Gamble, M. V. (2018). Nutritional influences on one‐carbon metabolism: Effects on arsenic methylation and toxicity. Annual Review of Nutrition, 38, 401–429. 10.1146/annurev-nutr-082117-051757 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bräuner, E. V. , Nordsborg, R. B. , Andersen, Z. J. , Tjønneland, A. , Loft, S. , & Raaschou‐Nielsen, O. (2014). Long‐term exposure to low‐level arsenic in drinking water and diabetes incidence: A prospective study of the diet, cancer and health cohort. Environmental Health Perspectives, 122, 1059–1065. 10.1289/ehp.1408198 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bredfeldt, T. G. , Jagadish, B. , Eblin, K. E. , Mash, E. A. , & Gandolfi, A. J. (2006). Monomethylarsonous acid induces transformation of human bladder cells. Toxicology and Applied Pharmacology, 216, 69–79. 10.1016/j.taap.2006.04.011 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Brender, J. D. , Suarez, L. , Felkner, M. , Gilani, Z. , Stinchcomb, D. , Moody, K. , Henry, J. , & Hendricks, K. (2006). Maternal exposure to arsenic, cadmium, lead, and mercury and neural tube defects in offspring. Environmental Research, 101(1), 132–139. [PubMed] [Google Scholar]
  • Broberg, K. , Ahmed, S. , Engström, K. , Hossain, M. B. , Jurkovic Mlakar, S. , Bottai, M. , Grandér, M. , Raqib, R. , & Vahter, M. (2014). Arsenic exposure in early pregnancy alters genome‐wide DNA methylation in cord blood, particularly in boys. Journal of Developmental Origins of Health and Disease, 5, 288–298. 10.1017/s2040174414000221 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Buchet, J. P. , Lauwerys, R. , & Roels, H. (1981a). Comparison of the urinary‐excretion of arsenic metabolites after a single oral dose of sodium arsenite, monomethylarsonate, or dimethylarsinate in man. International Archives of Occupational and Environmental Health, 48(1), 71–79. [PubMed] [Google Scholar]
  • Buchet, J. P. , Lauwerys, R. , & Roels, H. (1981b). Urinary excretion of inorganic arsenic and its metabolites after repeated ingestion of sodium metaarsenite by volunteers. International Archives of Occupational and Environmental Health, 48(2), 111–118. 10.1007/BF00378431 [PubMed] [CrossRef] [Google Scholar]
  • Buck Louis, G. M. , Smarr, M. M. , Sundaram, R. , Steuerwald, A. J. , Sapra, K. J. , Lu, Z. , & Parsons, P. J. (2017). Low‐level environmental metals and metalloids and incident pregnancy loss. Reproductive Toxicology, 69, 68–74. 10.1016/j.reprotox.2017.01.011 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bulka, C. M. , Persky, V. W. , Daviglus, M. L. , Durazo‐Arvizu, R. A. , & Argos, M. (2019). Multiple metal exposures and metabolic syndrome: A cross‐sectional analysis of the National Health and nutrition examination survey 2011‐2014. Environmental Research, 168, 397–405. 10.1016/j.envres.2018.10.022 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bulka, C. M. , Scannell Bryan, M. , Lombard, M. A. , Bartell, S. M. , Jones, D. K. , Bradley, P. M. , Vieira, V. M. , Silverman, D. T. , Focazio, M. , Toccalino, P. L. , Daniel, J. , Backer, L. C. , Ayotte, J. D. , Gribble, M. O. , & Argos, M. (2022). Arsenic in private well water and birth outcomes in the United States. Environment International, 163, 107176. 10.1016/j.envint.2022.107176 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Burchiel, S. W. , Lauer, F. T. , Beswick, E. J. , Gandolfi, A. J. , Parvez, F. , Liu, K. J. , & Hudson, L. G. (2014). Differential susceptibility of human peripheral blood T cells to suppression by environmental levels of sodium arsenite and monomethylarsonous acid. PLoS One, 9, e109192. 10.1371/journal.pone.0109192 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bustaffa, E. , Stoccoro, A. , Bianchi, F. , & Migliore, L. (2014). Genotoxic and epigenetic mechanisms in arsenic carcinogenicity. Archives of Toxicology, 88, 1043–1067. 10.1007/s00204-014-1233-7 [PubMed] [CrossRef] [Google Scholar]
  • Butts, C. D. , Bloom, M. S. , Neamtiu, I. A. , Surdu, S. , Pop, C. , Anastasiu, D. , Fitzgerald, E. F. , & Gurzau, E. S. (2015). A pilot study of low‐moderate drinking water arsenic contamination and chronic diseases among reproductive age women in Timiş County, Romania. Environmental Toxicology and Pharmacology, 40, 1001–1004. 10.1016/j.etap.2015.11.003 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Caceres, D. D. , Werlinger, F. , Orellana, M. , Jara, M. , Rocha, R. , Alvarado, S. A. , & Luis, Q. (2010). Polymorphism of glutathione S‐transferase (GST) variants and its effect on distribution of urinary arsenic species in people exposed to low inorganic arsenic in tap water: An exploratory study. Archives of Environmental & Occupational Health, 65, 140–147. 10.1080/19338240903390354 [PubMed] [CrossRef] [Google Scholar]
  • Calderon, J. , Navarro, M. E. , Jimenez‐Capdeville, M. E. , Santos‐Diaz, M. A. , Golden, A. , Rodriguez‐Levya, I. , Borja‐Aburto, V. , & Diaz‐Barriga, F. (2001). Exposure to arsenic and lead and neuropsychological development in Mexican children. Environmental Research, 85(2), 69–76. [PubMed] [Google Scholar]
  • Calogero, A. E. , Fiore, M. , Giacone, F. , Altomare, M. , Asero, P. , Ledda, C. , Romeo, G. , Mongioì, L. M. , Copat, C. , Giuffrida, M. , Vicari, E. , Sciacca, S. , & Ferrante, M. (2021). Exposure to multiple metals/metalloids and human semen quality: A cross‐sectional study. Ecotoxicology and Environmental Safety, 215, 112165. 10.1016/j.ecoenv.2021.112165 [PubMed] [CrossRef] [Google Scholar]
  • Cannan, W. J. , & Pederson, D. S. (2016). Mechanisms and consequences of double‐Strand DNA break formation in chromatin. Journal of Cellular Physiology, 231, 3–14. 10.1002/jcp.25048 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cardenas, A. , Houseman, E. A. , Baccarelli, A. A. , Quamruzzaman, Q. , Rahman, M. , Mostofa, G. , Wright, R. O. , Christiani, D. C. , & Kile, M. L. (2015). In utero arsenic exposure and epigenome‐wide associations in placenta, umbilical artery, and human umbilical vein endothelial cells. Epigenetics, 10, 1054–1063. 10.1080/15592294.2015.1105424 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cardenas, A. , Smit, E. , Bethel, J. W. , Houseman, E. A. , & Kile, M. L. (2016). Arsenic exposure and the seroprevalence of total hepatitis a antibodies in the US population: NHANES, 2003–2012. Epidemiology and Infection, 144, 1641–1651. 10.1017/s0950268815003088 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cardenas, A. , Smit, E. , Houseman, E. A. , Kerkvliet, N. I. , Bethel, J. W. , & Kile, M. L. (2015). Arsenic exposure and prevalence of the varicella zoster virus in the United States: NHANES (2003–2004 and 2009–2010). Environmental Health Perspectives, 123, 590–596. 10.1289/ehp.1408731 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cardenas, A. , Smit, E. , Welch, B. M. , Bethel, J. , & Kile, M. L. (2018). Cross sectional association of arsenic and seroprevalence of hepatitis B infection in the United States (NHANES 2003–2014). Environmental Research, 166, 570–576. 10.1016/j.envres.2018.06.023 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cárdenas‐González, M. , Osorio‐Yáñez, C. , Gaspar‐Ramírez, O. , Pavković, M. , Ochoa‐Martínez, A. , López‐Ventura, D. , Medeiros, M. , Barbier, O. C. , Pérez‐Maldonado, I. N. , Sabbisetti, V. S. , Bonventre, J. V. , & Vaidya, V. S. (2016). Environmental exposure to arsenic and chromium in children is associated with kidney injury molecule‐1. Environmental Research, 150, 653–662. 10.1016/j.envres.2016.06.032 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cardoso, A. F. P. , Udoh, K. T. , & States, J. C. (2020). Arsenic‐induced changes in miRNA expression in cancer and other diseases. Toxicology and Applied Pharmacology, 409, 115306. 10.1016/j.taap.2020.115306 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cardoso, A. P. F. , Al‐Eryani, L. , & States, J. C. (2018). Arsenic‐induced carcinogenesis: The impact of miRNA dysregulation. Toxicological Sciences, 165(2), 284–290. 10.1093/toxsci/kfy128 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Carignan, C. C. , Cottingham, K. L. , Jackson, B. P. , Farzan, S. F. , Gandolfi, A. J. , Punshon, T. , Folt, C. L. , & Karagas, M. R. (2015). Estimated exposure to arsenic in breastfed and formula‐fed infants in a United States cohort. Environmental Health Perspectives, 123, 500–506. 10.1289/ehp.1408789 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Castiello, F. , Olmedo, P. , Gil, F. , Molina, M. , Mundo, A. , Romero, R. R. , Ruíz, C. , Gómez‐Vida, J. , Vela‐Soria, F. , & Freire, C. (2020). Association of urinary metal concentrations with blood pressure and serum hormones in Spanish male adolescents. Environmental Research, 182, 108958. 10.1016/j.envres.2019.108958 [PubMed] [CrossRef] [Google Scholar]
  • Castriota, F. , Acevedo, J. , Ferreccio, C. , Smith, A. H. , Liaw, J. , Smith, M. T. , & Steinmaus, C. (2018). Obesity and increased susceptibility to arsenic‐related type 2 diabetes in northern Chile. Environmental Research, 167, 248–254. 10.1016/j.envres.2018.07.022 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Catanzaro, I. , Schiera, G. , Sciandrello, G. , Barbata, G. , Caradonna, F. , Proia, P. , & Di Liegro, I. (2010). Biological effects of inorganic arsenic on primary cultures of rat astrocytes. International Journal of Molecular Medicine, 26, 457–462. 10.3892/ijmm_00000485 [PubMed] [CrossRef] [Google Scholar]
  • Chafe, R. , Aslanov, R. , Sarkar, A. , Gregory, P. , Comeau, A. , & Newhook, L. A. (2018). Association of type 1 diabetes and concentrations of drinking water components in Newfoundland and Labrador, Canada. BMJ Open Diabetes Research & Care, 6, 7. 10.1136/bmjdrc-2017-000466 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chakraborti, D. , Mukherjee, S. C. , Pati, S. , Sengupta, M. K. , Rahman, M. M. , Chowdhury, U. K. , Lodh, D. , Chanda, C. R. , Chakraborti, A. K. , & Basu, G. K. (2003). Arsenic groundwater contamination in middle ganga plain, Bihar, India: A future danger? Environmental Health Perspectives, 111, 1194–1201. 10.1289/ehp.5966 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chakraborti, D. , Sengupta, M. K. , Rahman, M. M. , Ahamed, S. , Chowdhury, U. K. , Hossain, M. A. , Mukherjee, S. C. , Pati, S. , Saha, K. C. , Dutta, R. N. , & Quamruzzaman, Q. (2004). Groundwater arsenic contamination and its health effects in the ganga‐Meghna‐Brahmaputra plain. Journal of Environmental Monitoring, 6, 74n–83n. [PubMed] [Google Scholar]
  • Chang, Y. W. , & Singh, K. P. (2019). Arsenic‐induced neoplastic transformation involves epithelial–mesenchymal transition and activation of the β‐catenin/c‐Myc pathway in human kidney epithelial cells. Chemical Research in Toxicology, 32(6), 1299–1309. [PubMed] [Google Scholar]
  • Chatterjee, D. , Bandyopadhyay, A. , Sarma, N. , Basu, S. , Roychowdhury, T. , Roy, S. S. , & Giri, A. K. (2018). Role of microRNAs in senescence and its contribution to peripheral neuropathy in the arsenic exposed population of West Bengal, India. Environmental Pollution, 233, 596–603. 10.1016/j.envpol.2017.09.063 [PubMed] [CrossRef] [Google Scholar]
  • Chen, C. H. , Grollman, A. P. , Huang, C. Y. , Shun, C. T. , Sidorenko, V. S. , Hashimoto, K. , Moriya, M. , Turesky, R. J. , Yun, B. H. , Tsai, K. , Wu, S. , Chuang, P. Y. , Tang, C. H. , Yang, W. H. , Tzai, T. S. , Tsai, Y. S. , Dickman, K. G. , & Pu, Y. S. (2021). Additive effects of arsenic and Aristolochic acid in chemical carcinogenesis of upper urinary tract Urothelium. Cancer Epidemiology, Biomarkers & Prevention, 30, 317–325. 10.1158/1055-9965.Epi-20-1090 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chen, C. L. , Chiou, H. Y. , Hsu, L. I. , Hsueh, Y. M. , Wu, M. M. , & Chen, C. J. (2010a). Ingested arsenic, characteristics of well water consumption and risk of different histological types of lung cancer in northeastern Taiwan. Environmental Research, 110, 455–462. 10.1016/j.envres.2009.08.010 [PubMed] [CrossRef] [Google Scholar]
  • Chen, C. L. , Chiou, H. Y. , Hsu, L. I. , Hsueh, Y. M. , Wu, M. M. , Wang, Y. H. , & Chen, C. J. (2010b). Arsenic in drinking water and risk of urinary tract cancer: A follow‐up study from northeastern Taiwan. Cancer Epidemiology, Biomarkers & Prevention, 19, 101–110. 10.1158/1055-9965.Epi-09-0333 [PubMed] [CrossRef] [Google Scholar]
  • Chen, J.‐W. , Chen, H.‐Y. , Li, W.‐F. , Liou, S.‐H. , Chen, C.‐J. , Wu, J.‐H. , & Wang, S.‐L. (2011). The association between total urinary arsenic concentration and renal dysfunction in a community‐based population from Central Taiwan. Chemosphere, 84, 17–24. 10.1016/j.chemosphere.2011.02.091 [PubMed] [CrossRef] [Google Scholar]
  • Chen, J. W. , Wang, S. L. , Wang, Y. H. , Sun, C. W. , Huang, Y. L. , Chen, C. J. , & Li, W. F. (2012). Arsenic methylation, GSTO1 polymorphisms, and metabolic syndrome in an arseniasis endemic area of southwestern Taiwan. Chemosphere, 88, 432–438. 10.1016/j.chemosphere.2012.02.059 [PubMed] [CrossRef] [Google Scholar]
  • Chen, K. , Liao, Q. L. , Ma, Z. W. , Jin, Y. , Hua, M. , Bi, J. , & Huang, L. (2015). Association of soil arsenic and nickel exposure with cancer mortality rates, a town‐scale ecological study in Suzhou, China. Environmental Science and Pollution Research International, 22, 5395–5404. 10.1007/s11356-014-3790-y [PubMed] [CrossRef] [Google Scholar]
  • Chen, Q. , Qiu, Y. , Chen, L. , Lin, J. , Yan, L. J. , Bao, X. D. , Lin, L. S. , Pan, L. Z. , Shi, B. , Zheng, X. Y. , Chen, F. , He, B. C. , Wang, J. , & Liu, F. Q. (2021). Association between serum arsenic and oral cancer risk: A case‐control study in Southeast China. Community Dentistry and Oral Epidemiology, 50, 83–90. 10.1111/cdoe.12633 [PubMed] [CrossRef] [Google Scholar]
  • Chen, W. J. , Davis, E. M. , Stoner, J. A. , Robledo, C. , Goodman, J. R. , Garwe, T. , Janitz, A. E. , Xu, C. , Hwang, J. , & Peck, J. D. (2021). Urinary total arsenic and arsenic methylation capacity in pregnancy and gestational diabetes mellitus: A case‐control study. Chemosphere, 271, 129828. 10.1016/j.chemosphere.2021.129828 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chen, Y. , Ahsan, H. , Slavkovich, V. , Peltier, G. L. , Gluskin, R. T. , Parvez, F. , Liu, X. , & Graziano, J. H. (2010). No association between arsenic exposure from drinking water and diabetes mellitus: A cross‐sectional study in Bangladesh. Environmental Health Perspectives, 118, 1299–1305. 10.1289/ehp.0901559 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chen, Y. , Factor‐Litvak, P. , Howe, G. R. , Graziano, J. H. , Brandt‐Rauf, P. , Parvez, F. , van Geen, A. , & Ahsan, H. (2007). Arsenic exposure from drinking water, dietary intakes of B vitamins and folate, and risk of high blood pressure in Bangladesh: A population‐based, cross‐sectional study. American Journal of Epidemiology, 165, 541–552. 10.1093/aje/kwk037 [PubMed] [CrossRef] [Google Scholar]
  • Chen, Y. , Graziano, J. H. , Parvez, F. , Liu, M. , Slavkovich, V. , Kalra, T. , Argos, M. , Islam, T. , Ahmed, A. , Rakibuz‐Zaman, M. , Hasan, R. , Sarwar, G. , Levy, D. , van Geen, A. , & Ahsan, H. (2011). Arsenic exposure from drinking water and mortality from cardiovascular disease in Bangladesh: Prospective cohort study. BMJ, 342, d2431. 10.1136/bmj.d2431 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chen, Y. , Parvez, F. , Liu, M. , Pesola, G. R. , Gamble, M. V. , Slavkovich, V. , Islam, T. , Ahmed, A. , Hasan, R. , Graziano, J. H. , & Ahsan, H. (2011). Association between arsenic exposure from drinking water and proteinuria: Results from the health effects of arsenic longitudinal study. International Journal of Epidemiology, 40, 828–835. 10.1093/ije/dyr022 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chen, Y. , Wu, F. , Graziano, J. H. , Parvez, F. , Liu, M. , Paul, R. R. , Shaheen, I. , Sarwar, G. , Ahmed, A. , Islam, T. , Slavkovich, V. , Rundek, T. , Demmer, R. T. , Desvarieux, M. , & Ahsan, H. (2013). Arsenic exposure from drinking Water, arsenic methylation capacity, and carotid intima‐media thickness in Bangladesh. American Journal of Epidemiology, 178, 372–381. 10.1093/aje/kwt001 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chen, Y. , Wu, F. , Liu, M. , Parvez, F. , Slavkovich, V. , Eunus, M. , Ahmed, A. , Argos, M. , Islam, T. , Rakibuz‐Zaman, M. , Hasan, R. , Sarwar, G. , Levy, D. , Graziano, J. , & Ahsan, H. (2013). A prospective study of arsenic exposure, arsenic methylation capacity, and risk of cardiovascular disease in Bangladesh. Environmental Health Perspectives, 121, 832–838. 10.1289/ehp.1205797 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cheng, J. , Li, Y. , He, Q. , Luo, L. , Zhang, Y. , Gao, Y. , Feng, H. , Zhao, L. , Wei, W. , Fu, S. , & Sun, D. (2021). Essential hypertension in patients exposed to high‐arsenic exposed areas in western China: Genetic susceptibility and urinary arsenic metabolism characteristics. Journal of Trace Elements in Medicine and Biology, 67, 126778. 10.1016/j.jtemb.2021.126778 [PubMed] [CrossRef] [Google Scholar]
  • Cheng, Y. Y. , Chang, Y. T. , Cheng, H. L. , Shen, K. H. , Sung, J. M. , & Guo, H. R. (2018). Associations between arsenic in drinking water and occurrence of end‐stage renal disease with modifications by comorbidities: A nationwide population‐based study in Taiwan. Sci Total Environ, 626, 581–591. 10.1016/j.scitotenv.2018.01.043 [PubMed] [CrossRef] [Google Scholar]
  • Cheng, Y. Y. , Huang, N. C. , Chang, Y. T. , Sung, J. M. , Shen, K. H. , Tsai, C. C. , & Guo, H. R. (2017). Associations between arsenic in drinking water and the progression of chronic kidney disease: A nationwide study in Taiwan. Journal of Hazardous Materials, 321, 432–439. 10.1016/j.jhazmat.2016.09.032 [PubMed] [CrossRef] [Google Scholar]
  • Cherry, N. , Shaikh, K. , McDonald, C. , & Chowdhury, Z. (2008). Stillbirth in rural Bangladesh: Arsenic exposure and other etiological factors: A report from Gonoshasthaya Kendra. Bulletin of the World Health Organization, 86, 172–177. [PMC free article] [PubMed] [Google Scholar]
  • Chervona, Y. , Hall, M. N. , Arita, A. , Wu, F. , Sun, H. , Tseng, H. C. , Ali, E. , Uddin, M. N. , Liu, X. , Zoroddu, M. A. , Gamble, M. V. , & Costa, M. (2012). Associations between arsenic exposure and global posttranslational histone modifications among adults in Bangladesh. Cancer Epidemiology, Biomarkers & Prevention, 21, 2252–2260. 10.1158/1055-9965.Epi-12-0833 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chiou, H. Y. , Chiou, S. T. , Hsu, Y. H. , Chou, Y. L. , Tseng, C. H. , Wei, M. L. , & Chen, C. J. (2001). Incidence of transitional cell carcinoma and arsenic in drinking water: A follow‐up study of 8,102 residents in an arseniasis‐endemic area in northeastern Taiwan. American Journal of Epidemiology, 153(5), 411–418. [PubMed] [Google Scholar]
  • Cho, E. , Allemang, A. , Audebert, M. , Chauhan, V. , Dertinger, S. , Hendriks, G. , Luijten, M. , Marchetti, F. , Minocherhomji, S. , Pfuhler, S. , Roberts, D. J. , Trenz, K. , & Yauk, C. L. (2022). AOP report: Development of an adverse outcome pathway for oxidative DNA damage leading to mutations and chromosomal aberrations. Environmental and Molecular Mutagenesis, 63, 118–134. 10.1002/em.22479 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Choi, W. S. , Kim, S. H. , & Chung, J. H. (2014). Relationships of hair mineral concentrations with insulin resistance in metabolic syndrome. Biological Trace Element Research, 158, 323–329. 10.1007/s12011-014-9946-2 [PubMed] [CrossRef] [Google Scholar]
  • Chou, W.‐C. , Chung, Y.‐T. , Chen, H.‐Y. , Wang, C.‐J. , Ying, T.‐H. , Chuang, C.‐Y. , Tseng, Y.‐C. , & Wang, S.‐L. (2014). Maternal arsenic exposure and DNA damage biomarkers, and the associations with birth outcomes in a general population from Taiwan. PLoS One, 9, e86398. 10.1371/journal.pone.0086398 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Choudhury, M. I. M. , Shabnam, N. , Ahsan, T. , Ahsan, S. M. A. , Kabir, M. S. , Khan, R. M. , Miah, M. A. , Uddin, M. K. , & Liton, M. A. R. (2018). Cutaneous malignancy due to Arsenicosis in Bangladesh: 12‐year study in tertiary level hospital. BioMed Research International, 2018, 4678362. 10.1155/2018/4678362 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chowdhury, R. , Ramond, A. , O'Keeffe, L. M. , Shahzad, S. , Kunutsor, S. K. , Muka, T. , Gregson, J. , Willeit, P. , Warnakula, S. , Khan, H. , Chowdhury, S. , Gobin, R. , Franco, O. H. , & Di Angelantonio, E. (2018). Environmental toxic metal contaminants and risk of cardiovascular disease: Systematic review and meta‐analysis. BMJ, 362, k3310. 10.1136/bmj.k3310 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Chung, C. J. , Lee, H. L. , Chang, C. H. , Chang, H. , Liu, C. S. , Jung, W. T. , Liu, H. J. , Liou, S. H. , Chung, M. C. , & Hsueh, Y. M. (2019). Measurement of urinary arsenic profiles and DNA hypomethylation in a case‐control study of urothelial carcinoma. Archives of Toxicology, 93, 2155–2164. 10.1007/s00204-019-02500-y [PubMed] [CrossRef] [Google Scholar]
  • Cohen, S. M. , Arnold, L. L. , Klaunig, J. E. , & Goodman, J. I. (2014). Re: Waalkes et al.: Lung tumors in mice induced by "whole‐life" inorganic arsenic exposure at human‐relevant doses. Archives of Toxicology, 88(11), 2061–2062. 10.1007/s00204-014-1328-1 [PubMed] [CrossRef] [Google Scholar]
  • Colín‐Torres, C. G. , Murillo‐Jiménez, J. M. , Del Razo, L. M. , Sánchez‐Peña, L. C. , Becerra‐Rueda, O. F. , & Marmolejo‐Rodríguez, A. J. (2014). Urinary arsenic levels influenced by abandoned mine tailings in the southernmost Baja California peninsula, Mexico. Environmental Geochemistry and Health, 36, 845–854. 10.1007/s10653-014-9603-x [PubMed] [CrossRef] [Google Scholar]
  • Concha, G. , Vogler, G. , Lezcano, D. , Nermell, B. , & Vahter, M. (1998). Exposure to inorganic arsenic metabolites during early human development. Toxicological Sciences, 44, 185–190. 10.1006/toxs.1998.2486 [PubMed] [CrossRef] [Google Scholar]
  • Concha, G. , Vogler, G. , Nermell, B. , & Vahter, M. (1998). Low‐level arsenic excretion in breast milk of native Andean women exposed to high levels of arsenic in the drinking water. International Archives of Occupational and Environmental Health, 71, 42–46. 10.1007/s004200050248 [PubMed] [CrossRef] [Google Scholar]
  • Coryell, M. , Roggenbeck, B. A. , & Walk, S. T. (2019). The human gut Microbiome's influence on arsenic toxicity. Current Pharmacology Reports, 5, 491–504. 10.1007/s40495-019-00206-4 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • COT (Committee on toxicity of chemicals in food, consumer products and the environment) . (2016). Statement on potential risks from arsenic in the diet of infants aged 0 to 12 months and children aged 1 to 5 years. https://cot.food.gov.uk/sites/default/files/finalstatementonarsenic_0.pdf
  • Cubadda, F. , Aureli, F. , D'Amato, M. , Raggi, A. , Turco, A. C. , & Mantovani, A. (2012). Speciated urinary arsenic as a biomarker of dietary exposure to inorganic arsenic in residents living in high‐arsenic areas in Latium, Italy. Pure and Applied Chemistry, 84, 203–214. 10.1351/pac-con-11-09-29 [CrossRef] [Google Scholar]
  • Currier, J. M. , Ishida, M. C. , González‐Horta, C. , Sánchez‐Ramírez, B. , Ballinas‐Casarrubias, L. , Gutiérrez‐Torres, D. S. , Cerón, R. H. , Morales, D. V. , Terrazas, F. A. , Del Razo, L. M. , García‐Vargas, G. G. , Saunders, R. J. , Drobná, Z. , Fry, R. C. , Matoušek, T. , Buse, J. B. , Mendez, M. A. , Loomis, D. , & Stýblo, M. (2014). Associations between arsenic species in exfoliated urothelial cells and prevalence of diabetes among residents of Chihuahua, Mexico. Environmental Health Perspectives, 122, 1088–1094. 10.1289/ehp.1307756 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Cusick, S. E. , Jaramillo, E. G. , Moody, E. C. , Ssemata, A. S. , Bitwayi, D. , Lund, T. C. , & Mupere, E. (2018). Assessment of blood levels of heavy metals including lead and manganese in healthy children living in the Katanga settlement of Kampala, Uganda. BMC Public Health, 18, 717. 10.1186/s12889-018-5589-0 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • da Silva, M. S. (2021). DNA double‐strand breaks: A double‐edged sword for trypanosomatids. Frontiers in Cell and Development Biology, 9, 669041. 10.3389/fcell.2021.669041 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Dai, L. , Lv, X. , Chen, Z. , Huang, Z. , Li, B. , Xie, Y. , Duan, Y. , Zhao, H. , Wang, Y. , Yu, Q. , Li, S. , Zhou, Y. , & Shen, X. (2020). Elevated whole blood arsenic level is associated with type 2 diabetes in coal‐burning areas in Guizhou. Toxicology and Applied Pharmacology, 403, 115135. 10.1016/j.taap.2020.115135 [PubMed] [CrossRef] [Google Scholar]
  • Das, D. , Bindhani, B. , Mukherjee, B. , Saha, H. , Biswas, P. , Dutta, K. , Prasad, P. , Sinha, D. , & Ray, M. R. (2014). Chronic low‐level arsenic exposure reduces lung function in male population without skin lesions. International Journal of Public Health, 59(4), 655–663. [PubMed] [Google Scholar]
  • Das, N. , Giri, A. , Chakraborty, S. , & Bhattacharjee, P. (2016). Association of single nucleotide polymorphism with arsenic‐induced skin lesions and genetic damage in exposed population of West Bengal, India. Mutation Research, 809, 50–56. 10.1016/j.mrgentox.2016.09.006 [PubMed] [CrossRef] [Google Scholar]
  • Dash, M. , Maity, M. , Dey, A. , Perveen, H. , Khatun, S. , Jana, L. , & Chattopadhyay, S. (2018). The consequence of NAC on sodium arsenite‐induced uterine oxidative stress. Toxicology Reports, 5, 278–287. 10.1016/j.toxrep.2018.02.003 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Dastgiri, S. , Mosaferi, M. , Fizi, M. A. , Olfati, N. , Zolali, S. , Pouladi, N. , & Azarfam, P. (2010). Arsenic exposure, dermatological lesions, hypertension, and chromosomal abnormalities among people in a rural community of Northwest Iran. Journal of Health, Population, and Nutrition, 28, 14–22. 10.3329/jhpn.v28i1.4519 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Dauphiné, D. C. , Smith, A. H. , Yuan, Y. , Balmes, J. R. , Bates, M. N. , & Steinmaus, C. (2013). Case‐control study of arsenic in drinking water and lung cancer in California and Nevada. International Journal of Environmental Research and Public Health, 10, 3310–3324. 10.3390/ijerph10083310 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Davis, M. A. , Higgins, J. , Li, Z. , Gilbert‐Diamond, D. , Baker, E. R. , Das, A. , & Karagas, M. R. (2015). Preliminary analysis of in utero low‐level arsenic exposure and fetal growth using biometric measurements extracted from fetal ultrasound reports. Environmental Health, 14, 12. 10.1186/1476-069x-14-12 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • de Assis Araujo, M. S. , Froes‐Asmus, C. I. R. , de Figueiredo, N. D. , Camara, V. M. , Luiz, R. R. , Prata‐Barbosa, A. , Martins, M. M. , Jacob, S. D. C. , Santos, L. , Vicentini Neto, S. A. , Rezende Filho, J. F. , & Amim Junior, J. (2022). Prenatal exposure to metals and neurodevelopment in infants at six months: Rio birth cohort study of environmental exposure and childhood development (PIPA project). International Journal of Environmental Research and Public Health, 19. 10.3390/ijerph19074295 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • De Loma, J. , Skröder, H. , Raqib, R. , Vahter, M. , & Broberg, K. (2018). Arsenite methyltransferase (AS3MT) polymorphisms and arsenic methylation in children in rural Bangladesh. Toxicology and Applied Pharmacology, 357, 80–87. 10.1016/j.taap.2018.08.020 [PubMed] [CrossRef] [Google Scholar]
  • De Palma, G. , Catalani, S. , Franco, A. , Brighenti, M. , & Apostoli, P. (2012). Lack of correlation between metallic elements analyzed in hair by ICP‐MS and autism. Journal of Autism and Developmental Disorders, 42, 342–353. 10.1007/s10803-011-1245-6 [PubMed] [CrossRef] [Google Scholar]
  • de Water, E. , Curtin, P. , Gennings, C. , Chelonis, J. J. , Paule, M. , Bixby, M. , McRae, N. , Svensson, K. , Schnaas, L. , Pantic, I. , Téllez‐Rojo, M. M. , Wright, R. O. , & Horton, M. K. (2022). Prenatal metal mixture concentrations and reward motivation in children. Neurotoxicology, 88, 124–133. 10.1016/j.neuro.2021.11.008 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Del Razo, L. M. , García‐Vargas, G. G. , Valenzuela, O. L. , Castellanos, E. H. , Sánchez‐Peña, L. C. , Currier, J. M. , Drobná, Z. , Loomis, D. , & Stýblo, M. (2011). Exposure to arsenic in drinking water is associated with increased prevalence of diabetes: A cross‐sectional study in the Zimapán and Lagunera regions in Mexico. Environmental Health, 10, 73. 10.1186/1476-069x-10-73 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Demanelis, K. , Argos, M. , Tong, L. , Shinkle, J. , Sabarinathan, M. , Rakibuz‐Zaman, M. , Sarwar, G. , Shahriar, H. , Islam, T. , Rahman, M. , Yunus, M. , Graziano, J. H. , Broberg, K. , Engström, K. , Jasmine, F. , Ahsan, H. , & Pierce, B. L. (2019). Association of arsenic exposure with whole blood DNA methylation: An epigenome‐wide study of Bangladeshi adults. Environmental Health Perspectives, 127, 57011. 10.1289/ehp3849 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Demir, N. , Başaranoğlu, M. , Huyut, Z. , Değer, İ. , Karaman, K. , Şekeroğlu, M. R. , & Tuncer, O. (2019). The relationship between mother and infant plasma trace element and heavy metal levels and the risk of neural tube defect in infants. The Journal of Maternal‐Fetal & Neonatal Medicine, 32, 1433–1440. 10.1080/14767058.2017.1408064 [PubMed] [CrossRef] [Google Scholar]
  • Desai, G. , Barg, G. , Queirolo, E. I. , Vahter, M. , Peregalli, F. , Mañay, N. , & Kordas, K. (2018). A cross‐sectional study of general cognitive abilities among Uruguayan school children with low‐level arsenic exposure, potential effect modification by methylation capacity and dietary folate. Environmental Research, 164, 124–131. 10.1016/j.envres.2018.02.021 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Desai, G. , Barg, G. , Vahter, M. , Queirolo, E. I. , Peregalli, F. , Mañay, N. , Millen, A. E. , Yu, J. , Browne, R. W. , & Kordas, K. (2020a). Low level arsenic exposure, B‐vitamins, and achievement among Uruguayan school children. International Journal of Hygiene and Environmental Health, 223, 124–131. 10.1016/j.ijheh.2019.09.011 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Desai, G. , Barg, G. , Vahter, M. , Queirolo, E. I. , Peregalli, F. , Mañay, N. , Millen, A. E. , Yu, J. , & Kordas, K. (2020b). Executive functions in school children from Montevideo, Uruguay and their associations with concurrent low‐level arsenic exposure. Environment International, 142, 105883. 10.1016/j.envint.2020.105883 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Desai, G. , Niu, Z. , Luo, W. , Frndak, S. , Shaver, A. L. , & Kordas, K. (2021). Low‐level exposure to lead, mercury, arsenic, and cadmium, and blood pressure among 8‐17‐year‐old participants of the 2009–2016 National Health and nutrition examination survey. Environmental Research, 197, 111086. 10.1016/j.envres.2021.111086 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Deyssenroth, M. A. , Gennings, C. , Liu, S. H. , Peng, S. , Hao, K. , Lambertini, L. , Jackson, B. P. , Karagas, M. R. , Marsit, C. J. , & Chen, J. (2018). Intrauterine multi‐metal exposure is associated with reduced fetal growth through modulation of the placental gene network. Environment International, 120, 373–381. 10.1016/j.envint.2018.08.010 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Díaz, O. P. , Leyton, I. , Muñoz, O. , Núñez, N. , Devesa, V. , Súñer, M. A. , Vélez, D. , & Montoro, R. (2004). Contribution of water, bread, and vegetables (raw and cooked) to dietary intake of inorganic arsenic in a rural village of northern Chile. Journal of Agricultural and Food Chemistry, 52, 1773–1779. 10.1021/jf035168t [PubMed] [CrossRef] [Google Scholar]
  • Ding, W. , Liu, W. , Cooper, K. L. , Qin, X. J. , de Souza Bergo, P. L. , Hudson, L. G. , & Liu, K. J. (2009). Inhibition of poly(ADP‐ribose) polymerase‐1 by arsenite interferes with repair of oxidative DNA damage. The Journal of Biological Chemistry, 284, 6809–6817. 10.1074/jbc.M805566200 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • D'Ippoliti, D. , Santelli, E. , De Sario, M. , Scortichini, M. , Davoli, M. , & Michelozzi, P. (2015). Arsenic in drinking Water and mortality for cancer and chronic diseases in Central Italy, 1990–2010. PLoS One, 10, e0138182. 10.1371/journal.pone.0138182 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • do Nascimento, S. N. , Barth, A. , Göethel, G. , Baierle, M. , Charão, M. F. , Brucker, N. , Moro, A. M. , Bubols, G. B. , Sobreira, J. S. , Sauer, E. , Rocha, R. , Gioda, A. , Dias, A. C. , Salles, J. F. , & Garcia, S. C. (2015). Cognitive deficits and ALA‐D‐inhibition in children exposed to multiple metals. Environmental Research, 136, 387–395. 10.1016/j.envres.2014.10.003 [PubMed] [CrossRef] [Google Scholar]
  • Dominguez‐Gonzalez, M. R. , Barciela‐Alonso, M. C. , Calvo‐Millan, V. G. , Herbello‐Hermelo, P. , & Bermejo‐Barrera, P. (2020). The bioavailability of arsenic species in rice. Analytical and Bioanalytical Chemistry, 412, 3253–3259. [PubMed] [Google Scholar]
  • Dong, J. , Wang, J. Q. , Qian, Q. , Li, G. C. , Yang, D. Q. , & Jiang, C. (2019). Micronucleus assay for monitoring the genotoxic effects of arsenic in human populations: A systematic review of the literature and meta‐analysis. Mutation Research, 780, 1–10. 10.1016/j.mrrev.2019.02.002 [PubMed] [CrossRef] [Google Scholar]
  • Dong, Y. , Zhou, M. , Zhang, M. , Wang, H. , Chen, Y. , Liu, X. , Yang, S. , Lang, H. , Sun, G. , & Li, X. (2022). The association between arsenic metabolism and hypertension among population with varying arsenic exposure levels in China. Exposure and Health, 14, 411–430. 10.1007/s12403-022-00462-8 [CrossRef] [Google Scholar]
  • Dopp, E. , von Recklinghausen, U. , Diaz‐Bone, R. , Hirner, A. V. , & Rettenmeier, A. W. (2010). Cellular uptake, subcellular distribution and toxicity of arsenic compounds in methylating and non‐methylating cells. Environmental Research, 110(5), 435–442. 10.1016/j.envres.2009.08.012 [PubMed] [CrossRef] [Google Scholar]
  • Dutta, K. , Prasad, P. , & Sinha, D. (2015). Chronic low level arsenic exposure evokes inflammatory responses and DNA damage. International Journal of Hygiene and Environmental Health, 218, 564–574. 10.1016/j.ijheh.2015.06.003 [PubMed] [CrossRef] [Google Scholar]
  • Ebert, F. , Leffers, L. , Weber, T. , Berndt, S. , Mangerich, A. , Beneke, S. , Bürkle, A. , & Schwerdtle, T. (2014). Toxicological properties of the thiolated inorganic arsenic and arsenosugar metabolite thio‐dimethylarsinic acid in human bladder cells. Journal of Trace Elements in Medicine and Biology, 28(2), 138–146. 10.1016/j.jtemb.2013.06.004 [PubMed] [CrossRef] [Google Scholar]
  • Ebert, F. , Weiss, A. , Bültemeyer, M. , Hamann, I. , Hartwig, A. , & Schwerdtle, T. (2011). Arsenicals affect base excision repair by several mechanisms. Mutation Research, 715, 32–41. 10.1016/j.mrfmmm.2011.07.004 [PubMed] [CrossRef] [Google Scholar]
  • Edwards, M. , Hall, J. , Gong, G. , & O'Bryant, S. E. (2014). Arsenic exposure, AS3MT polymorphism, and neuropsychological functioning among rural dwelling adults and elders: A cross‐sectional study. Environmental Health, 13, 15. 10.1186/1476-069x-13-15 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Edwards, M. , Johnson, L. , Mauer, C. , Barber, R. , Hall, J. , & O'Bryant, S. (2014). Regional specific groundwater arsenic levels and neuropsychological functioning: A cross‐sectional study. International Journal of Environmental Health Research, 24, 546–557. 10.1080/09603123.2014.883591 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • EFSA (European Food Safety Authority) . (2005). Opinion of the scientific committee on a request from EFSA related to a harmonised approach for risk assessment of substances which are both genotoxic and carcinogenic. EFSA Journal, 3(10), 282. 10.2903/j.efsa.2005.282 [CrossRef] [Google Scholar]
  • EFSA (European Food Safety Authority) . (2014). Scientific report of on the dietary exposure to inorganic arsenic in the European population. EFSA Journal, 12(3), 3597. 10.2903/sp.efsa.2014.3579 [CrossRef] [Google Scholar]
  • EFSA (European Food Safety Authority) , Arcella, D. , Cascio, C. , & Gómez Ruiz, J. Á. (2021). Scientific report on the chronic dietary exposure to inorganic arsenic. EFSA Journal, 19(1), 6380. 10.2903/j.efsa.2021.6380 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • EFSA CONTAM Panel (EFSA Panel on Contaminants in the Food Chain) . (2009). Scientific opinion on arsenic in food. EFSA Journal, 7(10), 1351. 10.2903/j.efsa.2009.1351 [CrossRef] [Google Scholar]
  • EFSA Scientific Committee . (2012). Guidance on selected default values to be used by the EFSA scientific committee, scientific panels and units in the absence of actual measured data. EFSA Journal, 10(3), 2579. 10.2903/j.efsa.2012.2579 [CrossRef] [Google Scholar]
  • EFSA Scientific Committee , Benford, D. , Halldorsson, T. , Jeger, M. J. , Knutsen, H. K. , More, S. , Naegeli, H. , Noteborn, H. , Ockleford, C. , Ricci, A. , Rychen, G. , Schlatter, J. R. , Silano, V. , Solecki, R. , Turck, D. , Younes, M. , Craig, P. , Hart, A. , Von Goetz, N. , … Hardy, A. (2018). Guidance on uncertainty analysis in scientific assessments. EFSA Journal, 16(1), 5123. 10.2903/j.efsa.2018.5123 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • EFSA Scientific Committee , Hardy, A. , Benford, D. , Halldorsson, T. , Jeger, M. J. , Knutsen, H. K. , More, S. , Naegeli, H. , Noteborn, H. , Ockleford, C. , Ricci, A. , Rychen, G. , Schlatter, J. R. , Silano, V. , Solecki, R. , Turck, D. , Bresson, J.‐L. , Dusemund, B. , Gundert‐Remy, U. , … Mortensen, A. (2017). Guidance on the risk assessment of substances present in food intended for infants below 16 weeks of age. EFSA Journal, 15(5), 4849. 10.2903/j.efsa.2017.4849 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • EFSA Scientific Committee , More, S. , Bambidis, V. , Benford, D. , Bragard, C. , Hernandez‐Jerez, A. , Bennekou, S. H. , Koutsoumanis, K. , Machera, K. , Naegeli, H. , Nielsen, S. S. , Schlatter, J. R. , Schrenk, D. , Silano, V. , Turck, D. , Younes, M. , Fletcher, T. , Greiner, M. , Ntzani, E. , … Halldorsson, T. I. (2020). Draft for internal testing scientific committee guidance on appraising and integrating evidence from epidemiological studies for use in EFSA's scientific assessments. EFSA Journal, 18(8), 6221. 10.2903/j.efsa.2020.6221 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • EFSA Scientific Committee , More, S. J. , Bampidis, V. , Benford, D. , Bragard, C. , Halldorsson, T. I. , Hern ´andez‐Jerez, A. F. , Bennekou, S. H. , Koutsoumanis, K. , Lambr ´e, C. , Machera, K. , Mennes, W. , Mullins, E. , Nielsen, S. S. , Schrenk, D. , Turck, D. , Younes, M. , Aerts, M. , Edler, L. , … Schlatter, J. (2022). Guidance on the use of the benchmark dose approach in risk assessment. EFSA Journal, 20(10), 7584. 10.2903/j.efsa.2022.7584 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Eick, S. M. , Ferreccio, C. , Acevedo, J. , Castriota, F. , Cordero, J. F. , Roh, T. , Smith, A. H. , Smith, M. T. , & Steinmaus, C. (2019). Socioeconomic status and the association between arsenic exposure and type 2 diabetes. Environmental Research, 172, 578–585. 10.1016/j.envres.2019.03.013 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Elhajouji, A. , Lukamowicz, M. , Cammerer, Z. , & Kirsch‐Volders, M. (2011). Potential thresholds for genotoxic effects by micronucleus scoring. Mutagenesis, 26, 199–204. 10.1093/mutage/geq089 [PubMed] [CrossRef] [Google Scholar]
  • Ellinsworth, D. C. (2015). Arsenic, reactive oxygen, and endothelial dysfunction. The Journal of Pharmacology and Experimental Therapeutics, 353, 458–464. 10.1124/jpet.115.223289 [PubMed] [CrossRef] [Google Scholar]
  • Elsheshtawy, E. , Tobar, S. , Sherra, K. , Atallah, S. , & Elkasaby, R. (2011). Study of some biomarkers in hair of children with autism. Middle East Current Psychiatry, 18, 6–10. 10.1097/01.Xme.0000392842.64112.64 [CrossRef] [Google Scholar]
  • Engel, R. R. , & Smith, A. H. (1994). Arsenic in drinking water and mortality from vascular disease: An ecologic analysis in 30 counties in the United States. Archives of Environmental Health, 49, 418–427. 10.1080/00039896.1994.9954996 [PubMed] [CrossRef] [Google Scholar]
  • Engström, K. , Vahter, M. , Mlakar, S. J. , Concha, G. , Nermell, B. , Raqib, R. , Cardozo, A. , & Broberg, K. (2011). Polymorphisms in arsenic(+III oxidation state) methyltransferase (AS3MT) predict gene expression of AS3MT as well as arsenic metabolism. Environmental Health Perspectives, 119, 182–188. 10.1289/ehp.1002471 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Engström, K. S. , Vahter, M. , Fletcher, T. , Leonardi, G. , Goessler, W. , Gurzau, E. , Koppova, K. , Rudnai, P. , Kumar, R. , & Broberg, K. (2015). Genetic variation in arsenic (+3 oxidation state) methyltransferase (AS3MT), arsenic metabolism and risk of basal cell carcinoma in a European population. Environmental and Molecular Mutagenesis, 56, 60–69. 10.1002/em.21896 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ersbøll, A. K. , Monrad, M. , Sørensen, M. , Baastrup, R. , Hansen, B. , Bach, F. W. , Tjønneland, A. , Overvad, K. , & Raaschou‐Nielsen, O. (2018). Low‐level exposure to arsenic in drinking water and incidence rate of stroke: A cohort study in Denmark. Environment International, 120, 72–80. 10.1016/j.envint.2018.07.040 [PubMed] [CrossRef] [Google Scholar]
  • Evans, M. D. , Mistry, V. , Singh, R. , Gackowski, D. , Różalski, R. , Siomek‐Gorecka, A. , Phillips, D. H. , Zuo, J. , Mullenders, L. , Pines, A. , Nakabeppu, Y. , Sakumi, K. , Sekiguchi, M. , Tsuzuki, T. , Bignami, M. , Oliński, R. , & Cooke, M. S. (2016). Nucleotide excision repair of oxidised genomic DNA is not a source of urinary 8‐oxo‐7,8‐dihydro‐2′‐deoxyguanosine. Free Radical Biology & Medicine, 99, 385–391. 10.1016/j.freeradbiomed.2016.08.018 [PubMed] [CrossRef] [Google Scholar]
  • Fan, Y. , Jiang, Y. , Hu, P. , Chang, R. , Yao, S. , Wang, B. , Li, X. , Zhou, Q. , & Qiao, Y. (2016). Modification of association between prior lung disease and lung cancer by inhaled arsenic: A prospective occupational‐based cohort study in Yunnan, China. Journal of Exposure Science & Environmental Epidemiology, 26, 464–470. 10.1038/jes.2016.22 [PubMed] [CrossRef] [Google Scholar]
  • FAO/WHO (Food and Agriculture Organization/ World Health Organization) . (1983). Evaluation of certain food additives and contaminants. WHO Food Additive Report Series, No. 18. International Programme on Chemical Safety, World Health Organization, Geneva.
  • FAO/WHO (Food and Agriculture Organization/World Health Organization) . (2011). Safety evaluation of certain contaminants in food. WHO Food Additives Series, No. 63/FAO JECFA Monographs 8. Geneva, Switzerland: World Health Organization and Rome, Italy: Food and Agriculture Organization of the United Nations. https://whqlibdoc.who.int/publications/2011/9789241660631_eng.pdf
  • Farzan, S. F. , Chen, Y. , Rees, J. R. , Zens, M. S. , & Karagas, M. R. (2015). Risk of death from cardiovascular disease associated with low‐level arsenic exposure among long‐term smokers in a US population‐based study. Toxicology and Applied Pharmacology, 287, 93–97. 10.1016/j.taap.2015.05.013 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Farzan, S. F. , Gossai, A. , Chen, Y. , Chasan‐Taber, L. , Baker, E. , & Karagas, M. (2016). Maternal arsenic exposure and gestational diabetes and glucose intolerance in the New Hampshire birth cohort study. Environmental Health, 15, 106. 10.1186/s12940-016-0194-0 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Fatmi, Z. , Abbasi, I. N. , Ahmed, M. , Kazi, A. , & Kayama, F. (2013). Burden of skin lesions of arsenicosis at higher exposure through groundwater of taluka Gambat district Khairpur, Pakistan: A cross‐sectional survey. Environmental Geochemistry and Health, 35, 341–346. 10.1007/s10653-012-9498-3 [PubMed] [CrossRef] [Google Scholar]
  • Fei, D. L. , Koestler, D. C. , Li, Z. , Giambelli, C. , Sanchez‐Mejias, A. , Gosse, J. A. , Marsit, C. J. , Karagas, M. R. , & Robbins, D. J. (2013). Association between in utero arsenic exposure, placental gene expression, and infant birth weight: A US birth cohort study. Environmental Health, 12, 58. 10.1186/1476-069x-12-58 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Fenech, M. , & Bonassi, S. (2011). The effect of age, gender, diet and lifestyle on DNA damage measured using micronucleus frequency in human peripheral blood lymphocytes. Mutagenesis, 26, 43–49. 10.1093/mutage/geq050 [PubMed] [CrossRef] [Google Scholar]
  • Fenech, M. , Knasmueller, S. , Bolognesi, C. , Holland, N. , Bonassi, S. , & Kirsch‐Volders, M. (2020). Micronuclei as biomarkers of DNA damage, aneuploidy, inducers of chromosomal hypermutation and as sources of pro‐inflammatory DNA in humans. Mutation Research, Reviews in Mutation Research, 786, 108342. 10.1016/j.mrrev.2020.108342 [PubMed] [CrossRef] [Google Scholar]
  • Fenech, M. , Knasmueller, S. , Knudsen, L. E. , Kirsch‐Volders, M. , Deo, P. , Franzke, B. , Stopper, H. , Andreassi, M.‐G. , Bolognesi, C. , Dhillon, V. S. , Laffon, B. , Wagner, K.‐H. , & Bonassi, S. (2021). “Micronuclei and disease” special issue: Aims, scope, and synthesis of outcomes. Mutation Research/Reviews in Mutation Research, 788, 108384. 10.1016/j.mrrev.2021.108384 [PubMed] [CrossRef] [Google Scholar]
  • Ferdosi, H. , Dissen, E. K. , Afari‐Dwamena, N. A. , Li, J. , Chen, R. , Feinleib, M. , & Lamm, S. H. (2016). Arsenic in drinking Water and Lung cancer mortality in the United States: An analysis based on US counties and 30 years of observation (1950–1979). Journal of Environmental and Public Health, 2016, 1602929. 10.1155/2016/1602929 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ferreccio, C. , González, C. , Milosavjlevic, V. , Marshall, G. , Sancha, A. M. , & Smith, A. H. (2000). Lung cancer and arsenic concentrations in drinking water in Chile. Epidemiology, 11, 673–679. 10.1097/00001648-200011000-00010 [PubMed] [CrossRef] [Google Scholar]
  • Ferreccio, C. , & Sancha, A. M. (2006). Arsenic exposure and its impact on health in Chile. Journal of Health, Population, and Nutrition, 24, 164–175. [PubMed] [Google Scholar]
  • Ferreccio, C. , Smith, A. H. , Durán, V. , Barlaro, T. , Benítez, H. , Valdés, R. , Aguirre, J. J. , Moore, L. E. , Acevedo, J. , Vásquez, M. I. , Pérez, L. , Yuan, Y. , Liaw, J. , Cantor, K. P. , & Steinmaus, C. (2013). Case‐control study of arsenic in drinking water and kidney cancer in uniquely exposed northern Chile. American Journal of Epidemiology, 178, 813–818. 10.1093/aje/kwt059 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ferreccio, C. , Yuan, Y. , Calle, J. , Benítez, H. , Parra, R. L. , Acevedo, J. , Smith, A. H. , Liaw, J. , & Steinmaus, C. (2013). Arsenic, tobacco smoke, and occupation: Associations of multiple agents with lung and bladder cancer. Epidemiology, 24(6), 898–905. 10.1097/EDE.0b013e31829e3e03 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Feseke, S. K. , St‐Laurent, J. , Anassour‐Sidi, E. , Ayotte, P. , Bouchard, M. , & Levallois, P. (2015). Arsenic exposure and type 2 diabetes: Results from the 2007–2009 Canadian health measures survey. Health Promotion and Chronic Disease Prevention in Canada, 35, 63–72. 10.24095/hpcdp.35.4.01 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Fido, A. , & Al‐Saad, S. (2016). Toxic trace elements in the hair of children with autism. Autism, 9, 290–298. 10.1177/1362361305053255 [PubMed] [CrossRef] [Google Scholar]
  • Fillol, C. , Dor, F. , Labat, L. , Boltz, P. , Le Bouard, J. , Mantey, K. , Mannschott, C. , Puskarczyk, E. , Viller, F. , Momas, I. , & Seta, N. (2010). Urinary arsenic concentrations and speciation in residents living in an area with naturally contaminated soils. Science of the Total Environment, 408, 1190–1194. 10.1016/j.scitotenv.2009.11.046 [PubMed] [CrossRef] [Google Scholar]
  • Fiłon, J. , Ustymowicz‐Farbiszewska, J. , & Krajewska‐Kułak, E. (2020). Analysis of lead, arsenic and calcium content in the hair of children with autism spectrum disorder. BMC Public Health, 20, 383. 10.1186/s12889-020-08496-w [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Fiore, M. , Barone, R. , Copat, C. , Grasso, A. , Cristaldi, A. , Rizzo, R. , & Ferrante, M. (2020). Metal and essential element levels in hair and association with autism severity. Journal of Trace Elements in Medicine and Biology, 57, 126409. 10.1016/j.jtemb.2019.126409 [PubMed] [CrossRef] [Google Scholar]
  • Fleisch, A. F. , Mukherjee, S. K. , Biswas, S. K. , Obrycki, J. F. , Ekramullah, S. M. , Arman, D. M. , Islam, J. , Christiani, D. C. , & Mazumdar, M. (2022). Arsenic exposure during pregnancy and postpartum maternal glucose tolerance: Evidence from Bangladesh. Environmental Health, 21, 13. 10.1186/s12940-021-00811-1 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Forns, J. , Fort, M. , Casas, M. , Cáceres, A. , Guxens, M. , Gascon, M. , Garcia‐Esteban, R. , Julvez, J. , Grimalt, J. O. , & Sunyer, J. (2014). Exposure to metals during pregnancy and neuropsychological development at the age of 4 years. Neurotoxicology, 40, 16–22. 10.1016/j.neuro.2013.10.006 [PubMed] [CrossRef] [Google Scholar]
  • Fowler, B. A. , Chou, C. H. S. J. , Jones, R. L. , Costa, M. , & Chen, C.‐J. (2022). Chapter 3 ‐ Arsenic. In Nordberg G. F. & Costa M. (Eds.), Handbook on the toxicology of metals (fifth edition) (pp. 41–89). Academic Press. [Google Scholar]
  • Frediani, J. K. , Naioti, E. A. , Vos, M. B. , Figueroa, J. , Marsit, C. J. , & Welsh, J. A. (2018). Arsenic exposure and risk of nonalcoholic fatty liver disease (NAFLD) among U.S. adolescents and adults: An association modified by race/ethnicity, NHANES 2005–2014. Environmental Health, 17, 6. 10.1186/s12940-017-0350-1 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Freire, C. , Amaya, E. , Gil, F. , Fernández, M. F. , Murcia, M. , Llop, S. , Andiarena, A. , Aurrekoetxea, J. , Bustamante, M. , Guxens, M. , Ezama, E. , Fernández‐Tardón, G. , & Olea, N. (2018). Prenatal co‐exposure to neurotoxic metals and neurodevelopment in preschool children: The environment and childhood (INMA) project. Sci Total Environ, 621, 340–351. 10.1016/j.scitotenv.2017.11.273 [PubMed] [CrossRef] [Google Scholar]
  • Fu, S. , Wu, J. , Li, Y. , Liu, Y. , Gao, Y. , Yao, F. , Qiu, C. , Song, L. , Wu, Y. , Liao, Y. , & Sun, D. (2014). Urinary arsenic metabolism in a Western Chinese population exposed to high‐dose inorganic arsenic in drinking water: Influence of ethnicity and genetic polymorphisms. Toxicology and Applied Pharmacology, 274, 117–123. 10.1016/j.taap.2013.11.004 [PubMed] [CrossRef] [Google Scholar]
  • Fujioka, M. , Gi, M. , Kawachi, S. , Tatsumi, K. , Ishii, N. , Doi, K. , Kakehashi, A. , & Wanibuchi, H. (2016). Examination of in vivo mutagenicity of sodium arsenite and dimethylarsinic acid in gpt delta rats. Journal of Environmental Sciences (China), 49, 125–130. 10.1016/j.jes.2016.07.005 [PubMed] [CrossRef] [Google Scholar]
  • Galvez‐Fernandez, M. , Grau‐Perez, M. , Garcia‐Barrera, T. , Ramirez‐Acosta, S. , Gomez‐Ariza, J. L. , Perez‐Gomez, B. , Galan‐Labaca, I. , Navas‐Acien, A. , Redon, J. , Briongos‐Figuero, L. S. , Dueñas‐Laita, A. , Perez‐Castrillon, J. L. , Tellez‐Plaza, M. , & Martin‐Escudero, J. C. (2021). Arsenic, cadmium, and selenium exposures and bone mineral density‐related endpoints: The HORTEGA study. Free Radical Biology & Medicine, 162, 392–400. 10.1016/j.freeradbiomed.2020.10.318 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gamboa‐Loira, B. , Cebrián, M. E. , Franco‐Marina, F. , & López‐Carrillo, L. (2017). Arsenic metabolism and cancer risk: A meta‐analysis. Environmental Research, 156, 551–558. 10.1016/j.envres.2017.04.016 [PubMed] [CrossRef] [Google Scholar]
  • Ganapathy, S. , Liu, J. , Xiong, R. , Yu, T. , Makriyannis, A. , & Chen, C. (2019). Chronic low dose arsenic exposure preferentially perturbs mitotic phase of the cell cycle. Genes & Cancer, 10, 39–51. 10.18632/genesandcancer.185 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gao, J. , Tong, L. , Argos, M. , Scannell Bryan, M. , Ahmed, A. , Rakibuz‐Zaman, M. , Kibriya, M. G. , Jasmine, F. , Slavkovich, V. , Graziano, J. H. , Ahsan, H. , & Pierce, B. L. (2015). The genetic architecture of arsenic metabolism efficiency:A SNP‐based heritability study of Bangladeshi adults. Environmental Health Perspectives, 123, 985–992. 10.1289/ehp.1408909 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • García‐Esquinas, E. , Pollán, M. , Umans, J. G. , Francesconi, K. A. , Goessler, W. , Guallar, E. , Howard, B. , Farley, J. , Best, L. G. , & Navas‐Acien, A. (2013). Arsenic exposure and cancer mortality in a US‐based prospective cohort: The strong heart study. Cancer Epidemiology, Biomarkers & Prevention, 22, 1944–1953. 10.1158/1055-9965.Epi-13-0234-t [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gardner, R. M. , Kippler, M. , Tofail, F. , Bottai, M. , Hamadani, J. , Grandér, M. , Nermell, B. , Palm, B. , Rasmussen, K. M. , & Vahter, M. (2013). Environmental exposure to metals and children's growth to age 5 years: A prospective cohort study. American Journal of Epidemiology, 177, 1356–1367. 10.1093/aje/kws437 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Garland, M. , Morris, J. S. , Colditz, G. A. , Stampfer, M. J. , Spate, V. L. , Baskett, C. K. , Rosner, B. , Speizer, F. E. , Willett, W. C. , & Hunter, D. J. (1996). Toenail trace element levels and breast cancer: A prospective study. American Journal of Epidemiology, 144, 653–660. 10.1093/oxfordjournals.aje.a008977 [PubMed] [CrossRef] [Google Scholar]
  • Garry, M. R. , Santamaria, A. B. , Williams, A. L. , & DeSesso, J. M. (2015). In utero arsenic exposure in mice and early life susceptibility to cancer. Regulatory Toxicology and Pharmacology, 73, 378–390. 10.1016/j.yrtph.2015.07.023 [PubMed] [CrossRef] [Google Scholar]
  • Geier, D. , Kern, J. , King, P. , Sykes, L. , & Geier, M. (2012). Hair toxic metal concentrations and autism Spectrum disorder severity in young children. International Journal of Environmental Research and Public Health, 9, 4486–4497. 10.3390/ijerph9124486 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gelmann, E. R. , Gurzau, E. , Gurzau, A. , Goessler, W. , Kunrath, J. , Yeckel, C. W. , & McCarty, K. M. (2013). A pilot study: The importance of inter‐individual differences in inorganic arsenic metabolism for birth weight outcome. Environmental Toxicology and Pharmacology, 36, 1266–1275. 10.1016/j.etap.2013.10.006 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • George, C. M. , Brooks, W. A. , Graziano, J. H. , Nonyane, B. A. , Hossain, L. , Goswami, D. , Zaman, K. , Yunus, M. , Khan, A. F. , Jahan, Y. , & Ahmed, D. (2015). Arsenic exposure is associated with pediatric pneumonia in rural Bangladesh: A case control study. Environmental Health, 14(1), 10. [PMC free article] [PubMed] [Google Scholar]
  • Gilbert‐Diamond, D. , Emond, J. A. , Baker, E. R. , Korrick, S. A. , & Karagas, M. R. (2016). Relation between in utero arsenic exposure and birth outcomes in a cohort of mothers and their newborns from New Hampshire. Environmental Health Perspectives, 124, 1299–1307. 10.1289/ehp.1510065 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gilbert‐Diamond, D. , Li, Z. , Perry, A. E. , Spencer, S. K. , Gandolfi, A. J. , & Karagas, M. R. (2013). A population‐based case–control study of urinary arsenic species and squamous cell carcinoma in New Hampshire, USA. Environmental Health Perspectives, 121(10), 1154–1160. [PMC free article] [PubMed] [Google Scholar]
  • Gliga, A. R. , Engström, K. , Kippler, M. , Skröder, H. , Ahmed, S. , Vahter, M. , Raqib, R. , & Broberg, K. (2018). Prenatal arsenic exposure is associated with increased plasma IGFBP3 concentrations in 9‐year‐old children partly via changes in DNA methylation. Archives of Toxicology, 92, 2487–2500. 10.1007/s00204-018-2239-3 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gong, G. , Basom, J. , Mattevada, S. , & Onger, F. (2015). Association of hypothyroidism with low‐level arsenic exposure in rural West Texas. Environmental Research, 138, 154–160. 10.1016/j.envres.2015.02.001 [PubMed] [CrossRef] [Google Scholar]
  • Gong, G. , Hargrave, K. A. , Hobson, V. , Spallholz, J. , Boylan, M. , Lefforge, D. , & O'Bryant, S. E. (2011). Low‐level groundwater arsenic exposure impacts cognition: A project FRONTIER study. Journal of Environmental Health, 74, 16–22. [PubMed] [Google Scholar]
  • Gong, G. , & O'Bryant, S. E. (2012). Low‐level arsenic exposure, AS3MT gene polymorphism and cardiovascular diseases in rural Texas counties. Environmental Research, 113, 52–57. 10.1016/j.envres.2012.01.003 [PubMed] [CrossRef] [Google Scholar]
  • González‐Martínez, F. , Sánchez‐Rodas, D. , Cáceres, D. D. , Martínez, M. F. , Quiñones, L. A. , & Johnson‐Restrepo, B. (2018). Arsenic exposure, profiles of urinary arsenic species, and polymorphism effects of glutathione‐s‐transferase and metallothioneins. Chemosphere, 212, 927–936. 10.1016/j.chemosphere.2018.08.139 [PubMed] [CrossRef] [Google Scholar]
  • Gossai, A. , Zens, M. S. , Punshon, T. , Jackson, B. P. , Perry, A. E. , & Karagas, M. R. (2017). Rice consumption and squamous cell carcinoma of the skin in a United States population. Environmental Health Perspectives, 125, 097005. 10.1289/ehp1065 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Govarts, E. , Remy, S. , Bruckers, L. , Den Hond, E. , Sioen, I. , Nelen, V. , Baeyens, W. , Nawrot, T. S. , Loots, I. , Van Larebeke, N. , & Schoeters, G. (2016). Combined effects of prenatal exposures to environmental chemicals on birth weight. International Journal of Environmental Research and Public Health, 13. 10.3390/ijerph13050495 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Grant, R. L. (2014). Converting an odds ratio to a range of plausible relative risks for better communication of research findings. BMJ, 348, f7450. 10.1136/bmj.f7450 Erratum in: BMJ 2014;348:g2124. PMID: 24464277. [PubMed] [CrossRef] [Google Scholar]
  • Grau‐Perez, M. , Caballero‐Mateos, M. J. , Domingo‐Relloso, A. , Navas‐Acien, A. , Gomez‐Ariza, J. L. , Garcia‐Barrera, T. , Leon‐Latre, M. , Soriano‐Gil, Z. , Jarauta, E. , Cenarro, A. , Moreno‐Franco, B. , Laclaustra, M. , Civeira, F. , Casasnovas, J. A. , Guallar, E. , & Tellez‐Plaza, M. (2022). Toxic metals and subclinical atherosclerosis in carotid, femoral, and coronary vascular territories: The Aragon workers health study. Arteriosclerosis, Thrombosis, and Vascular Biology, 42, 87–99. 10.1161/ATVBAHA.121.316358 [PubMed] [CrossRef] [Google Scholar]
  • Grau‐Perez, M. , Kuo, C. C. , Spratlen, M. , Thayer, K. A. , Mendez, M. A. , Hamman, R. F. , Dabelea, D. , Adgate, J. L. , Knowler, W. C. , Bell, R. A. , Miller, F. W. , Liese, A. D. , Zhang, C. , Douillet, C. , Drobná, Z. , Mayer‐Davis, E. J. , Styblo, M. , & Navas‐Acien, A. (2017). The Association of Arsenic Exposure and Metabolism with type 1 and type 2 diabetes in youth: The SEARCH case‐control study. Diabetes Care, 40, 46–53. 10.2337/dc16-0810 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Grau‐Perez, M. , Navas‐Acien, A. , Galan‐Chilet, I. , Briongos‐Figuero, L. S. , Morchon‐Simon, D. , Bermudez, J. D. , Crainiceanu, C. M. , de Marco, G. , Rentero‐Garrido, P. , Garcia‐Barrera, T. , Gomez‐Ariza, J. L. , Casasnovas, J. A. , Martin‐Escudero, J. C. , Redon, J. , Chaves, F. J. , & Tellez‐Plaza, M. (2018). Arsenic exposure, diabetes‐related genes and diabetes prevalence in a general population from Spain. Environmental Pollution, 235, 948–955. 10.1016/j.envpol.2018.01.008 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Green, B. B. , Karagas, M. R. , Punshon, T. , Jackson, B. P. , Robbins, D. J. , Houseman, E. A. , & Marsit, C. J. (2016). Epigenome‐wide assessment of DNA methylation in the placenta and arsenic exposure in the New Hampshire birth cohort study (USA). Environmental Health Perspectives, 124, 1253–1260. 10.1289/ehp.1510437 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Gribble, M. O. , Howard, B. V. , Umans, J. G. , Shara, N. M. , Francesconi, K. A. , Goessler, W. , Crainiceanu, C. M. , Silbergeld, E. K. , Guallar, E. , & Navas‐Acien, A. (2012). Arsenic exposure, diabetes prevalence, and diabetes control in the strong heart study. American Journal of Epidemiology, 176, 865–874. 10.1093/aje/kws153 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Guan, H. , Piao, F. , Zhang, X. , Li, X. , Li, Q. , Xu, L. , Kitamura, F. , & Yokoyama, K. (2012). Prenatal exposure to arsenic and its effects on fetal development in the general population of Dalian. Biological Trace Element Research, 149, 10–15. 10.1007/s12011-012-9396-7 [PubMed] [CrossRef] [Google Scholar]
  • Guha Mazumder, D. N. , Deb, D. , Biswas, A. , Saha, C. , Nandy, A. , Das, A. , Ghose, A. , Bhattacharya, K. , & Mazumdar, K. K. (2014). Dietary arsenic exposure with low level of arsenic in drinking water and biomarker: A study in West Bengal. Journal of Environmental Science and Health. Part A, Toxic/Hazardous Substances & Environmental Engineering, 49, 555–564. 10.1080/10934529.2014.859042 [PubMed] [CrossRef] [Google Scholar]
  • Guha Mazumder, D. N. , Ghosh, A. , Majumdar, K. K. , Ghosh, N. , Saha, C. , & Mazumder, R. N. (2010). Arsenic contamination of ground water and its health impact on population of district of nadia, West Bengal, India. Indian Journal of Community Medicine, 35, 331–338. 10.4103/0970-0218.66897 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Guha Mazumder, D. N. , Haque, R. , Ghosh, N. , De, B. K. , Santra, A. , Chakraborty, D. , & Smith, A. H. (1998). Arsenic levels in drinking water and the prevalence of skin lesions in West Bengal, India. International Journal of Epidemiology, 27, 871–877. 10.1093/ije/27.5.871 [PubMed] [CrossRef] [Google Scholar]
  • Guha Mazumder, D. N. , Majumdar, K. K. , Santra, S. C. , Kol, H. , & Vicheth, C. (2009). Occurrence of arsenicosis in a rural village of Cambodia. Journal of Environmental Science and Health. Part A, Toxic/Hazardous Substances & Environmental Engineering, 44(5), 480–487. 10.1080/10934520902719886 [PubMed] [CrossRef] [Google Scholar]
  • Guha Mazumder, D. N. , Purkayastha, I. , Ghose, A. , Mistry, G. , Saha, C. , Nandy, A. K. , Das, A. , & Majumdar, K. K. (2012). Hypertension in chronic arsenic exposure: A case control study in West Bengal. Journal of Environmental Science and Health. Part A, Toxic/Hazardous Substances & Environmental Engineering, 47, 1514–1520. 10.1080/10934529.2012.680329 [PubMed] [CrossRef] [Google Scholar]
  • Gunduz, O. , Bakar, C. , Simsek, C. , Baba, A. , Elci, A. , Gurleyuk, H. , Mutlu, M. , & Cakir, A. (2015). Statistical analysis of causes of death (2005–2010) in villages of Simav plain, Turkey, with high arsenic levels in drinking water supplies. Archives of Environmental & Occupational Health, 70, 35–46. 10.1080/19338244.2013.872076 [PubMed] [CrossRef] [Google Scholar]
  • Guo, J. , Lv, N. , Tang, J. , Zhang, X. , Peng, L. , Du, X. , Li, S. , Luo, Q. , Zhang, D. , & Chen, G. (2018). Associations of blood metal exposure with thyroid hormones in Chinese pregnant women: A cross‐sectional study. Environment International, 121, 1185–1192. 10.1016/j.envint.2018.10.038 [PubMed] [CrossRef] [Google Scholar]
  • Guo, J. X. , Hu, L. , Yand, P. Z. , Tanabe, K. , Miyatalre, M. , & Chen, Y. (2007). Chronic arsenic poisoning in drinking water in Inner Mongolia and its associated health effects. Journal of Environmental Science and Health. Part A, Toxic/Hazardous Substances & Environmental Engineering, 42, 1853–1858. 10.1080/10934520701566918 [PubMed] [CrossRef] [Google Scholar]
  • Guo, X. , Fujino, Y. , Chai, J. , Wu, K. , Xia, Y. , Li, Y. , Lv, J. , Sun, Z. , & Yoshimura, T. (2003). The prevalence of subjective symptoms after exposure to arsenic in drinking water in Inner Mongolia, China. Journal of Epidemiology, 13, 211–215. 10.2188/jea.13.211 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Guo, X. , Liu, Z. , Huang, C. , & You, L. (2006). Levels of arsenic in drinking‐water and cutaneous lesions in Inner Mongolia. Journal of Health, Population, and Nutrition, 24, 214–220. [PubMed] [Google Scholar]
  • Gurr, J. R. , Yih, L. H. , Samikkannu, T. , Bau, D. T. , Lin, S. Y. , & Jan, K. Y. (2003). Nitric oxide production by arsenite. Mutation Research, 533, 173–182. 10.1016/j.mrfmmm.2003.08.024 [PubMed] [CrossRef] [Google Scholar]
  • Haber, L. T. , Dourson, M. L. , Allen, B. C. , Hertzberg, R. C. , Parker, A. , Vincent, M. J. , Maier, A. , & Boobis, A. R. (2018). Benchmark dose (BMD) modeling: Current practice, issues, and challenges. Critical Reviews in Toxicology, 48, 387–415. 10.1080/10408444.2018.1430121 [PubMed] [CrossRef] [Google Scholar]
  • Hall, E. M. , Acevedo, J. , López, F. G. , Cortés, S. , Ferreccio, C. , Smith, A. H. , & Steinmaus, C. M. (2017). Hypertension among adults exposed to drinking water arsenic in northern Chile. Environmental Research, 153, 99–105. 10.1016/j.envres.2016.11.016 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hamadani, J. D. , Grantham‐McGregor, S. M. , Tofail, F. , Nermell, B. , Fängström, B. , Huda, S. N. , Yesmin, S. , Rahman, M. , Vera‐Hernández, M. , Arifeen, S. E. , & Vahter, M. (2010). Pre‐ and postnatal arsenic exposure and child development at 18 months of age: A cohort study in rural Bangladesh. International Journal of Epidemiology, 39, 1206–1216. 10.1093/ije/dyp369 [PubMed] [CrossRef] [Google Scholar]
  • Hamadani, J. D. , Tofail, F. , Nermell, B. , Gardner, R. , Shiraji, S. , Bottai, M. , Arifeen, S. E. , Huda, S. N. , & Vahter, M. (2011). Critical windows of exposure for arsenic‐associated impairment of cognitive function in pre‐school girls and boys: A population‐based cohort study. International Journal of Epidemiology, 40, 1593–1604. 10.1093/ije/dyr176 [PubMed] [CrossRef] [Google Scholar]
  • Hameed, E. R. A. , Shehata, M. A. , Ahmed, H. H. , Sherif, L. S. , Elnady, H. G. , & Waheed, H. (2019). Relation of heavy metals in cord and maternal Blood to neonatal anthropometric indices. Journal of Clinical and Diagnostic Research, 13, SC01–SC05. 10.7860/jcdr/2019/39929.12653 [CrossRef] [Google Scholar]
  • Hansen, A. F. , Simić, A. , Åsvold, B. O. , Romundstad, P. R. , Midthjell, K. , Syversen, T. , & Flaten, T. P. (2017). Trace elements in early phase type 2 diabetes mellitus‐a population‐based study. The HUNT study in Norway. Journal of Trace Elements in Medicine and Biology, 40, 46–53. 10.1016/j.jtemb.2016.12.008 [PubMed] [CrossRef] [Google Scholar]
  • Hansen, H. R. , Raab, A. , Jaspars, M. , Milne, B. F. , & Feldmann, J. (2004). Sulfur‐containing arsenical mistaken for dimethylarsinous acid [DMA(III)] and identified as a natural metabolite in urine: Major implications for studies on arsenic metabolism and toxicity. Chemical Research in Toxicology, 17, 1086–1091. 10.1021/tx049978q [PubMed] [CrossRef] [Google Scholar]
  • Harper, K. N. , Liu, X. , Hall, M. N. , Ilievski, V. , Oka, J. , Calancie, L. , Slavkovich, V. , Levy, D. , Siddique, A. , Alam, S. , Mey, J. L. , van Geen, A. , Graziano, J. H. , & Gamble, M. V. (2014). A dose‐response study of arsenic exposure and markers of oxidative damage in Bangladesh. Journal of Occupational and Environmental Medicine, 56, 652–658. 10.1097/jom.0000000000000166 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hartwig, A. , Arand, M. , Epe, B. , Guth, S. , Jahnke, G. , Lampen, A. , Martus, H. J. , Monien, B. , Rietjens, I. , Schmitz‐Spanke, S. , Schriever‐Schwemmer, G. , Steinberg, P. , & Eisenbrand, G. (2020). Mode of action‐based risk assessment of genotoxic carcinogens. Archives of Toxicology, 94, 1787–1877. 10.1007/s00204-020-02733-2 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hartwig, A. , Pelzer, A. , Asmuss, M. , & Bürkle, A. (2003). Very low concentrations of arsenite suppress poly(ADP‐ribosyl)ation in mammalian cells. International Journal of Cancer, 104, 1–6. 10.1002/ijc.10911 [PubMed] [CrossRef] [Google Scholar]
  • Hartwig, A. , & Schwerdtle, T. (2009). Arsenic‐induced carcinogenicity: New insights in molecular mechanism. Metal complex–DNA interactions, 491–510. [Google Scholar]
  • Hashim, J. H. , Radzi, R. S. , Aljunid, S. M. , Nur, A. M. , Ismail, A. , Baguma, D. , Sthiannopkao, S. , Phan, K. , Wong, M. H. , Sao, V. , & Yasin, M. S. (2013). Hair arsenic levels and prevalence of arsenicosis in three Cambodian provinces. Science of the Total Environment, 463‐464, 1210–1216. 10.1016/j.scitotenv.2013.04.084 [PubMed] [CrossRef] [Google Scholar]
  • Heck, J. E. , Andrew, A. S. , Onega, T. , Rigas, J. R. , Jackson, B. P. , Karagas, M. R. , & Duell, E. J. (2009). Lung cancer in a US population with low to moderate arsenic exposure. Environmental Health Perspectives, 117, 1718–1723. 10.1289/ehp.0900566 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hei, T. K. , Liu, S. X. , & Waldren, C. (1998). Mutagenicity of arsenic in mammalian cells: Role of reactive oxygen species. Proceedings of the National Academy of Sciences of the United States of America, 95, 8103–8107. 10.1073/pnas.95.14.8103 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Henn, B. C. , Ettinger, A. S. , Hopkins, M. R. , Jim, R. , Amarasiriwardena, C. , Christiani, D. C. , Coull, B. A. , Bellinger, D. C. , & Wright, R. O. (2016). Prenatal arsenic exposure and birth outcomes among a population residing near a mining‐related superfund site. Environmental Health Perspectives, 124, 1308–1315. 10.1289/ehp.1510070 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hinhumpatch, P. , Navasumrit, P. , Chaisatra, K. , Promvijit, J. , Mahidol, C. , & Ruchirawat, M. (2013). Oxidative DNA damage and repair in children exposed to low levels of arsenic in utero and during early childhood: Application of salivary and urinary biomarkers. Toxicology and Applied Pharmacology, 273, 569–579. 10.1016/j.taap.2013.10.002 [PubMed] [CrossRef] [Google Scholar]
  • Hinwood, A. L. , Jolley, D. J. , & Sim, M. R. (1999). Cancer incidence and high environmental arsenic concentrations in rural populations: Results of an ecological study. International Journal of Environmental Health Research, 9, 131–141. 10.1080/09603129973272 [CrossRef] [Google Scholar]
  • Holcomb, N. , Goswami, M. , Han, S. G. , Scott, T. , D'Orazio, J. , Orren, D. K. , Gairola, C. G. , & Mellon, I. (2017). Inorganic arsenic inhibits the nucleotide excision repair pathway and reduces the expression of XPC. DNA Repair (Amst), 52, 70–80. 10.1016/j.dnarep.2017.02.009 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hopenhayn, C. , Ferreccio, C. , Browning, S. R. , Huang, B. , Peralta, C. , Gibb, H. , & Hertz‐Picciotto, I. (2003). Arsenic exposure from drinking water and birth weight. Epidemiology, 14(5), 593–602. [PubMed] [Google Scholar]
  • Hopenhayn‐Rich, C. , Browning, S. R. , Hertz‐Picciotto, I. , Ferreccio, C. , Peralta, C. , & Gibb, H. (2000). Chronic arsenic exposure and risk of infant mortality in two areas of Chile. Environmental Health Perspectives, 108(7), 667–673. [PMC free article] [PubMed] [Google Scholar]
  • Hossain, K. , Suzuki, T. , Hasibuzzaman, M. M. , Islam, M. S. , Rahman, A. , Paul, S. K. , Tanu, T. , Hossain, S. , Saud, Z. A. , Rahman, M. , Nikkon, F. , Miyataka, H. , Himeno, S. , & Nohara, K. (2017). Chronic exposure to arsenic, LINE‐1 hypomethylation, and blood pressure: A cross‐sectional study in Bangladesh. Environmental Health, 16, 20. 10.1186/s12940-017-0231-7 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hossain, M. B. , Vahter, M. , Concha, G. , & Broberg, K. (2012). Environmental arsenic exposure and DNA methylation of the tumor suppressor gene p16 and the DNA repair gene MLH1: Effect of arsenic metabolism and genotype. Metallomics, 4, 1167–1175. 10.1039/c2mt20120h [PubMed] [CrossRef] [Google Scholar]
  • Howe, C. G. , Claus Henn, B. , Eckel, S. P. , Farzan, S. F. , Grubbs, B. H. , Chavez, T. A. , Hodes, T. L. , Faham, D. , Al‐Marayati, L. , Lerner, D. , Quimby, A. , Twogood, S. , Richards, M. J. , Meeker, J. D. , Bastain, T. M. , & Breton, C. V. (2020). Prenatal metal mixtures and birth weight for gestational age in a predominately lower‐income Hispanic pregnancy cohort in Los Angeles. Environmental Health Perspectives, 128, 117001. 10.1289/ehp7201 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Howe, C. G. , Farzan, S. F. , Garcia, E. , Jursa, T. , Iyer, R. , Berhane, K. , Chavez, T. A. , Hodes, T. L. , Grubbs, B. H. , Funk, W. E. , Smith, D. R. , Bastain, T. M. , & Breton, C. V. (2020). Arsenic and birth outcomes in a predominately lower income Hispanic pregnancy cohort in Los Angeles. Environmental Research, 184, 109294. 10.1016/j.envres.2020.109294 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Howe, C. G. , Liu, X. H. , Hall, M. N. , Slavkovich, V. , Ilievski, V. , Parvez, F. , Siddique, A. B. , Shahriar, H. , Uddin, M. N. , Islam, T. , Graziano, J. H. , Costa, M. , & Gamble, M. V. (2016). Associations between Blood and urine arsenic concentrations and global levels of post‐translational histone modifications in Bangladeshi men and women. Environmental Health Perspectives, 124, 1234–1240. 10.1289/ehp.1510412 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hsieh, R. L. , Huang, Y. L. , Shiue, H. S. , Huang, S. R. , Lin, M. I. , Mu, S. C. , Chung, C. J. , & Hsueh, Y. M. (2014). Arsenic methylation capacity and developmental delay in preschool children in Taiwan. International Journal of Hygiene and Environmental Health, 217, 678–686. 10.1016/j.ijheh.2014.02.004 [PubMed] [CrossRef] [Google Scholar]
  • Hsieh, Y. C. , Hsieh, F. I. , Lien, L. M. , Chou, Y. L. , Chiou, H. Y. , & Chen, C. J. (2008). Risk of carotid atherosclerosis associated with genetic polymorphisms of apolipoprotein E and inflammatory genes among arsenic exposed residents in Taiwan. Toxicology and Applied Pharmacology, 227, 1–7. 10.1016/j.taap.2007.10.013 [PubMed] [CrossRef] [Google Scholar]
  • Hsieh, Y. C. , Lien, L. M. , Chung, W. T. , Hsieh, F. I. , Hsieh, P. F. , Wu, M. M. , Tseng, H. P. , Chiou, H. Y. , & Chen, C. J. (2011). Significantly increased risk of carotid atherosclerosis with arsenic exposure and polymorphisms in arsenic metabolism genes. Environmental Research, 111, 804–810. 10.1016/j.envres.2011.05.003 [PubMed] [CrossRef] [Google Scholar]
  • Hsu, K. H. , Tsui, K. H. , Hsu, L. I. , Chiou, H. Y. , & Chen, C. J. (2017). Dose‐response relationship between inorganic arsenic exposure and Lung cancer among Arseniasis residents with low methylation capacity. Cancer Epidemiology, Biomarkers & Prevention, 26, 756–761. 10.1158/1055-9965.Epi-16-0281 [PubMed] [CrossRef] [Google Scholar]
  • Hsu, L. I. , Hsieh, F. I. , Wang, Y. H. , Lai, T. S. , Wu, M. M. , Chen, C. J. , Chiou, H. Y. , & Hsu, K. H. (2017). Arsenic exposure from drinking Water and the incidence of CKD in low to moderate exposed areas of Taiwan: A 14‐year prospective study. American Journal of Kidney Diseases, 70, 787–797. 10.1053/j.ajkd.2017.06.012 [PubMed] [CrossRef] [Google Scholar]
  • Hsu, L. I. , Wang, Y. H. , Chiou, H. Y. , Wu, M. M. , Yang, T. Y. , Chen, Y. H. , Tseng, C. H. , & Chen, C. J. (2013). The association of diabetes mellitus with subsequent internal cancers in the arsenic‐exposed area of Taiwan. Journal of Asian Earth Sciences, 73, 452–459. 10.1016/j.jseaes.2013.04.048 [CrossRef] [Google Scholar]
  • Hsueh, Y. M. , Chung, C. J. , Shiue, H. S. , Chen, J. B. , Chiang, S. S. , Yang, M. H. , Tai, C. W. , & Su, C. T. (2009). Urinary arsenic species and CKD in a Taiwanese population: A case‐control study. American Journal of Kidney Diseases, 54, 859–870. 10.1053/j.ajkd.2009.06.016 [PubMed] [CrossRef] [Google Scholar]
  • Hsueh, Y. M. , Lin, Y. C. , Chung, C. J. , Huang, Y. L. , Hsieh, R. L. , Huang, P. T. , Wu, M. Y. , Shiue, H. S. , Chien, S. N. , Lee, C. Y. , Lin, M. I. , Mu, S. C. , & Su, C. T. (2020). Combined effect of polymorphisms of MTHFR and MTR and arsenic methylation capacity on developmental delay in preschool children in Taiwan. Archives of Toxicology, 94, 2027–2038. 10.1007/s00204-020-02745-y [PubMed] [CrossRef] [Google Scholar]
  • Hsueh, Y. M. , Su, C. T. , Shiue, H. S. , Chen, W. J. , Pu, Y. S. , Lin, Y. C. , Tsai, C. S. , & Huang, C. Y. (2017). Levels of plasma selenium and urinary total arsenic interact to affect the risk for prostate cancer. Food and Chemical Toxicology, 107, 167–175. 10.1016/j.fct.2017.06.031 [PubMed] [CrossRef] [Google Scholar]
  • Hu, Y. , Li, J. , Lou, B. , Wu, R. , Wang, G. , Lu, C. , Wang, H. , Pi, J. , & Xu, Y. (2020). The role of reactive oxygen species in arsenic toxicity. Biomolecules, 10. 10.3390/biom10020240 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Huang, C. Y. , Lin, Y. C. , Shiue, H. S. , Chen, W. J. , Su, C. T. , Pu, Y. S. , Ao, P. L. , & Hsueh, Y. M. (2018). Comparison of arsenic methylation capacity and polymorphisms of arsenic methylation genes between bladder cancer and upper tract urothelial carcinoma. Toxicology Letters, 295, 64–73. 10.1016/j.toxlet.2018.05.035 [PubMed] [CrossRef] [Google Scholar]
  • Huang, H. , Wei, Y. , Xia, Y. , Wei, L. , Chen, X. , Zhang, R. , Su, L. , Rahman, M. L. , Rahman, M. , Qamruzzaman, Q. , Guo, W. , Shen, H. , Hu, Z. , Christiani, D. C. , & Chen, F. (2021). Child marriage, maternal serum metal exposure, and risk of preterm birth in rural Bangladesh: Evidence from mediation analysis. Journal of Exposure Science & Environmental Epidemiology, 31, 571–580. 10.1038/s41370-021-00319-3 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Huang, H. H. , Huang, J. Y. , Lung, C. C. , Wu, C. L. , Ho, C. C. , Sun, Y. H. , Ko, P. C. , Su, S. Y. , Chen, S. C. , & Liaw, Y. P. (2013). Cell‐type specificity of lung cancer associated with low‐dose soil heavy metal contamination in Taiwan: An ecological study. BMC Public Health, 13, 330. 10.1186/1471-2458-13-330 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Huang, J. , Wu, J. , Li, T. , Song, X. , Zhang, B. , Zhang, P. , & Zheng, X. (2011). Effect of exposure to trace elements in the soil on the prevalence of neural tube defects in a high‐risk area of China. Biomedical and Environmental Sciences, 24, 94–101. 10.3967/0895-3988.2011.02.002 [PubMed] [CrossRef] [Google Scholar]
  • Huang, J. W. , Cheng, Y. Y. , Sung, T. C. , Guo, H. R. , & Sthiannopkao, S. (2014). Association between arsenic exposure and diabetes mellitus in Cambodia. BioMed Research International, 2014, 683124. 10.1155/2014/683124 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Huang, M. , Choi, S. J. , Kim, D. W. , Kim, N. Y. , Park, C. H. , Yu, S. D. , Kim, D. S. , Park, K. S. , Song, J. S. , Kim, H. , Choi, B. S. , Yu, I. J. , & Park, J. D. (2009). Risk assessment of low‐level cadmium and arsenic on the kidney. Journal of Toxicology and Environmental Health. Part A, 72, 1493–1498. 10.1080/15287390903213095 [PubMed] [CrossRef] [Google Scholar]
  • Hubaux, R. , Becker‐Santos, D. D. , Enfield, K. S. , Rowbotham, D. , Lam, S. , Lam, W. L. , & Martinez, V. D. (2013). Molecular features in arsenic‐induced lung tumors. Molecular Cancer, 12, 20. 10.1186/1476-4598-12-20 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Huyck, K. L. , Kile, M. L. , Mahiuddin, G. , Quamruzzaman, Q. , Rahman, M. , Breton, C. V. , Dobson, C. B. , Frelich, J. , Hoffman, E. , Yousuf, J. , Afroz, S. , Islam, S. , & Christiani, D. C. (2007). Maternal arsenic exposure associated with low birth weight in Bangladesh. Journal of Occupational and Environmental Medicine, 49(10), 1097–1104. [PubMed] [Google Scholar]
  • IARC (International Agency for Research on Cancer) . (1973). Some inorganic and organometallic compounds. IARC Monographs on the Evaluation of Carcinogenic Risk of Chemicals to Man, 2, 1–181. [Google Scholar]
  • IARC (International Agency for Research on Cancer) . (1979). Criteria to Select Chemicals for IARC Monographs (IARC Intern Tech Rep No. 79/003).
  • IARC (International Agency for Research on Cancer) . (2004). Some drinking‐water disinfectants and contaminants, including arsenic. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, 84, 1–477. [PMC free article] [PubMed] [Google Scholar]
  • IARC (International Agency for Research on Cancer) . (2012). Arsenic and arsenic compounds. IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, 100C, 41–93. [Google Scholar]
  • Igra, A. M. , Warnqvist, A. , Rahman, S. M. , Ekström, E. C. , Rahman, A. , Vahter, M. , & Kippler, M. (2021). Environmental metal exposure and growth to 10 years of age in a longitudinal mother‐child cohort in rural Bangladesh. Environment International, 156, 106738. 10.1016/j.envint.2021.106738 [PubMed] [CrossRef] [Google Scholar]
  • Intarasunanont, P. , Navasumrit, P. , Waraprasit, S. , Chaisatra, K. , Suk, W. A. , Mahidol, C. , & Ruchirawat, M. (2012). Effects of arsenic exposure on DNA methylation in cord blood samples from newborn babies and in a human lymphoblast cell line. Environmental Health, 11, 31. 10.1186/1476-069x-11-31 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ishii, N. , Mochizuki, H. , Ebihara, Y. , Shiomi, K. , & Nakazato, M. (2017). Incidence of clinical symptoms and neurological signs in patients with chronic arsenic exposure in Miyazaki, Japan: A 40‐year retrospective descriptive study. Journal of the Neurological Sciences, 381, 499. 10.1016/j.jns.2017.08.3613 [CrossRef] [Google Scholar]
  • Ishii, N. , Mochizuki, H. , Ebihara, Y. , Shiomi, K. , & Nakazato, M. (2018). Clinical symptoms, neurological signs, and electrophysiological findings in surviving residents with probable arsenic exposure in Toroku, Japan. Archives of Environmental Contamination and Toxicology, 75, 521–529. 10.1007/s00244-018-0544-8 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Islam, K. , Haque, A. , Karim, R. , Fajol, A. , Hossain, E. , Salam, K. A. , Ali, N. , Saud, Z. A. , Rahman, M. , Rahman, M. , Karim, R. , Sultana, P. , Hossain, M. , Akhand, A. A. , Mandal, A. , Miyataka, H. , Himeno, S. , & Hossain, K. (2011). Dose‐response relationship between arsenic exposure and the serum enzymes for liver function tests in the individuals exposed to arsenic: A cross sectional study in Bangladesh. Environmental Health, 10, 64. 10.1186/1476-069x-10-64 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Islam, M. R. , Khan, I. , Attia, J. , Hassan, S. M. , McEvoy, M. , D'Este, C. , Azim, S. , Akhter, A. , Akter, S. , Shahidullah, S. M. , & Milton, A. H. (2012). Association between hypertension and chronic arsenic exposure in drinking water: A cross‐sectional study in Bangladesh. International Journal of Environmental Research and Public Health, 9, 4522–4536. 10.3390/ijerph9124522 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Islam, R. , Khan, I. , Hassan, S. N. , McEvoy, M. , D'Este, C. , Attia, J. , Peel, R. , Sultana, M. , Akter, S. , & Milton, A. H. (2012). Association between type 2 diabetes and chronic arsenic exposure in drinking water: A cross sectional study in Bangladesh. Environmental Health, 11, 38. 10.1186/1476-069x-11-38 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Islam, S. , Rahman, M. M. , Rahman, M. A. , & Naidu, R. (2017). Inorganic arsenic in rice and rice‐based diets: Health risk assessment. Food Control, 82, 196–202. [Google Scholar]
  • Jalali, S. , & Wohlin, C. (2012). Systematic literature studies: Database searches vs. In Backward snowballing. 6th ACM‐IEEE international symposium on empirical software engineering and measurement. https://www.diva‐portal.org/smash/get/diva2:834640/FULLTEXT01.pdf [Google Scholar]
  • James, K. A. , Byers, T. , Hokanson, J. E. , Meliker, J. R. , Zerbe, G. O. , & Marshall, J. A. (2015). Association between lifetime exposure to inorganic arsenic in drinking water and coronary heart disease in Colorado residents. Environmental Health Perspectives, 123, 128–134. 10.1289/ehp.1307839 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • James, K. A. , Meliker, J. R. , Marshall, J. A. , Hokanson, J. E. , Zerbe, G. O. , & Byers, T. E. (2013). Validation of estimates of past exposure to arsenic in drinking water using historical urinary arsenic concentrations. Journal of Exposure Science & Environmental Epidemiology, 23, 450–454. 10.1038/jes.2013.8 [PubMed] [CrossRef] [Google Scholar]
  • Jiang, C. B. , Hsueh, Y. M. , Kuo, G. L. , Hsu, C. H. , Chang, J. H. , & Chien, L. C. (2018). Preliminary study of urinary arsenic concentration and arsenic methylation capacity effects on neurodevelopment in very low birth weight preterm children under 24 months of corrected age. Medicine (Baltimore), 97, e12800. 10.1097/md.0000000000012800 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Jiang, C.‐B. , Kao, C.‐S. , Chien, L.‐C. , Chen, Y.‐J. , & Liao, K.‐W. (2022). Associations among prenatal and postnatal arsenic, lead, and cadmium exposures and motor development in 3‐year‐old children: A longitudinal birth cohort study in Taiwan. Environmental Science and Pollution Research, 29, 43191–43200. 10.1007/s11356-021-18321-5 [PubMed] [CrossRef] [Google Scholar]
  • Jiang, J. , Bellani, M. , Li, L. , Wang, P. , Seidman, M. M. , & Wang, Y. (2017). Arsenite binds to the RING finger domain of FANCL E3 ubiquitin ligase and inhibits DNA Interstrand crosslink repair. ACS Chemical Biology, 12, 1858–1866. 10.1021/acschembio.6b01135 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Jiang, J. , Liu, M. , Parvez, F. , Wang, B. , Wu, F. , Eunus, M. , Bangalore, S. , Newman, J. D. , Ahmed, A. , Islam, T. , Rakibuz‐Zaman, M. , Hasan, R. , Sarwar, G. , Levy, D. , Slavkovich, V. , Argos, M. , Scannell Bryan, M. , Farzan, S. F. , Hayes, R. B. , … Chen, Y. (2015). Association between arsenic exposure from drinking Water and longitudinal change in Blood pressure among HEALS cohort participants. Environmental Health Perspectives, 123, 806–812. 10.1289/ehp.1409004 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Jiang, X. , Chen, C. , Zhao, W. , & Zhang, Z. (2013). Sodium arsenite and arsenic trioxide differently affect the oxidative stress, genotoxicity and apoptosis in A549 cells: An implication for the paradoxical mechanism. Environmental Toxicology and Pharmacology, 36, 891–902. 10.1016/j.etap.2013.08.002 [PubMed] [CrossRef] [Google Scholar]
  • Jiménez‐Córdova, M. I. , Cárdenas‐González, M. , Aguilar‐Madrid, G. , Sanchez‐Peña, L. C. , Barrera‐Hernández, Á. , Domínguez‐Guerrero, I. A. , González‐Horta, C. , Barbier, O. C. , & Del Razo, L. M. (2018). Evaluation of kidney injury biomarkers in an adult Mexican population environmentally exposed to fluoride and low arsenic levels. Toxicology and Applied Pharmacology, 352, 97–106. 10.1016/j.taap.2018.05.027 [PubMed] [CrossRef] [Google Scholar]
  • Jimenez‐Villarreal, J. , Rivas‐Armendariz, D. I. , Pineda‐Belmontes, C. P. , Betancourt‐Martínez, N. D. , Macías‐Corral, M. A. , Guerra‐Alanis, A. J. , Niño‐Castañeda, M. S. , & Morán‐Martínez, J. (2017). Detection of damage on single‐ or double‐stranded DNA in a population exposed to arsenic in drinking water. Genetics and Molecular Research, 16. 10.4238/gmr16029241 [PubMed] [CrossRef] [Google Scholar]
  • Jin, L. , Liu, J. , Ye, B. , & Ren, A. (2014). Concentrations of selected heavy metals in maternal blood and associated factors in rural areas in Shanxi Province, China. Environment International, 66, 157–164. 10.1016/j.envint.2014.01.016 [PubMed] [CrossRef] [Google Scholar]
  • Jin, L. , Zhang, L. , Li, Z. , Liu, J. M. , Ye, R. , & Ren, A. (2013). Placental concentrations of mercury, lead, cadmium, and arsenic and the risk of neural tube defects in a Chinese population. Reproductive Toxicology, 35, 25–31. 10.1016/j.reprotox.2012.10.015 [PubMed] [CrossRef] [Google Scholar]
  • Jin, X. , Tian, X. , Liu, Z. , Hu, H. , Li, X. , Deng, Y. , Li, N. , & Zhu, J. (2016). Maternal exposure to arsenic and cadmium and the risk of congenital heart defects in offspring. Reproductive Toxicology, 59, 109–116. 10.1016/j.reprotox.2015.12.007 [PubMed] [CrossRef] [Google Scholar]
  • Jochem, W. , Razzaque, A. , & Root, E. (2016). Effects of health intervention programs and arsenic exposure on child mortality from acute lower respiratory infections in rural Bangladesh. International Journal of Health Geographics, 15, 32. 10.1186/s12942-016-0061-9 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Jovanovic, D. , Rasic‐Milutinovic, Z. , Paunovic, K. , Jakovljevic, B. , Plavsic, S. , & Milosevic, J. (2013). Low levels of arsenic in drinking water and type 2 diabetes in middle Banat region, Serbia. International Journal of Hygiene and Environmental Health, 216, 50–55. 10.1016/j.ijheh.2012.01.001 [PubMed] [CrossRef] [Google Scholar]
  • Jovanović, D. D. , Paunović, K. , Manojlović, D. D. , Jakovljević, B. , Rasic‐Milutinović, Z. , & Dojcinović, B. P. (2012). Arsenic in drinking water and acute coronary syndrome in Zrenjanin municipality, Serbia. Environmental Research, 117, 75–82. 10.1016/j.envres.2012.04.016 [PubMed] [CrossRef] [Google Scholar]
  • Karachaliou, C. , Sgourou, A. , Kakkos, S. , & Kalavrouziotis, I. (2021). Arsenic exposure promotes the emergence of cardiovascular diseases. Reviews on Environmental Health, 37, 467–486. 10.1515/reveh-2021-0004 [PubMed] [CrossRef] [Google Scholar]
  • Karagas, M. R. , Le, X. C. , Morris, S. , Blum, J. , Lu, X. , Spate, V. , Carey, M. , Stannard, V. , Klaue, B. , & Tosteson, T. D. (2001). Markers of low level arsenic exposure for evaluating human cancer risks in a US population. International Journal of Occupational Medicine and Environmental Health, 14, 171–175. [PubMed] [Google Scholar]
  • Karagas, M. R. , Stukel, T. A. , & Tosteson, T. D. (2002). Assessment of cancer risk and environmental levels of arsenic in New Hampshire. International Journal of Hygiene and Environmental Health, 205, 85–94. [PubMed] [Google Scholar]
  • Karagas, M. R. , Tosteson, T. D. , Morris, J. S. , Demidenko, E. , Mott, L. A. , Heaney, J. , & Schned, A. (2004). Incidence of transitional cell carcinoma of the bladder and arsenic exposure in New Hampshire. Cancer Causes & Control, 15, 465–472. [PubMed] [Google Scholar]
  • Karakaya, A. , Jaruga, P. , Bohr, V. A. , Grollman, A. P. , & Dizdaroglu, M. (1997). Kinetics of excision of purine lesions from DNA by Escherichia coli Fpg protein. Nucleic Acids Research, 25, 474–479. 10.1093/nar/25.3.474 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Karim, Y. , Siddique, A. E. , Hossen, F. , Rahman, M. , Mondal, V. , Banna, H. U. , Hasibuzzaman, M. M. , Hosen, Z. , Islam, M. S. , Sarker, M. K. , Nikkon, F. , Saud, Z. A. , Xin, L. , Himeno, S. , & Hossain, K. (2019). Dose‐dependent relationships between chronic arsenic exposure and cognitive impairment and serum brain‐derived neurotrophic factor. Environment International, 131, 105029. 10.1016/j.envint.2019.105029 [PubMed] [CrossRef] [Google Scholar]
  • Kaufman, J. A. , Mattison, C. , Fretts, A. M. , Umans, J. G. , Cole, S. A. , Voruganti, V. S. , Goessler, W. , Best, L. G. , Zhang, Y. , Tellez‐Plaza, M. , Navas‐Acien, A. , & Gribble, M. O. (2021). Arsenic, blood pressure, and hypertension in the strong heart family study. Environmental Research, 195, 110864. 10.1016/j.envres.2021.110864 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kaushal, A. , Zhang, H. , Karmaus, W. J. J. , Everson, T. M. , Marsit, C. J. , Karagas, M. R. , Tsai, S. F. , Wen, H. J. , & Wang, S. L. (2017). Genome‐wide DNA methylation at birth in relation to in utero arsenic exposure and the associated health in later life. Environmental Health, 16, 50. 10.1186/s12940-017-0262-0 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kazemifar, A. M. , Shafikhani, A. A. , Mozhdehipanah, H. , Khamesi, S. , & Arami, M. (2020). Evaluation of different types of arsenic methylation and its relationship with metabolic syndrome in an area chronically exposed to arsenic. Environmental Analysis Health and Toxicology, 35, e2020006. 10.5620/eaht.e2020006 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kazi, T. G. , Arain, M. B. , Baig, J. A. , Jamali, M. K. , Afridi, H. I. , Jalbani, N. , Sarfraz, R. A. , Shah, A. Q. , & Niaz, A. (2009). The correlation of arsenic levels in drinking water with the biological samples of skin disorders. Science of the Total Environment, 407, 1019–1026. 10.1016/j.scitotenv.2008.10.013 [PubMed] [CrossRef] [Google Scholar]
  • Kern, J. K. , Grannemann, B. D. , Trivedi, M. H. , & Adams, J. B. (2007). Sulfhydryl‐reactive metals in autism. Journal of Toxicology and Environmental Health, Part A, 70, 715–721. 10.1080/15287390601188060 [PubMed] [CrossRef] [Google Scholar]
  • Kesari, V. P. , Kumar, A. , & Khan, P. K. (2012). Genotoxic potential of arsenic at its reference dose. Ecotoxicology and Environmental Safety, 80, 126–131. 10.1016/j.ecoenv.2012.02.018 [PubMed] [CrossRef] [Google Scholar]
  • Khan, K. , Factor‐Litvak, P. , Wasserman, G. A. , Liu, X. , Ahmed, E. , Parvez, F. , Slavkovich, V. , Levy, D. , Mey, J. , van Geen, A. , & Graziano, J. H. (2011). Manganese exposure from drinking Water and Children's classroom behavior in Bangladesh. Environmental Health Perspectives, 119, 1501–1506. 10.1289/ehp.1003397 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Khan, K. , Wasserman, G. A. , Liu, X. , Ahmed, E. , Parvez, F. , Slavkovich, V. , Levy, D. , Mey, J. , van Geen, A. , Graziano, J. H. , & Factor‐Litvak, P. (2012). Manganese exposure from drinking water and children's academic achievement. Neurotoxicology, 33, 91–97. 10.1016/j.neuro.2011.12.002 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Khan, K. M. , Parvez, F. , Zoeller, R. T. , Hocevar, B. A. , Kamendulis, L. M. , Rohlman, D. , Eunus, M. , & Graziano, J. (2019). Thyroid hormones and neurobehavioral functions among adolescents chronically exposed to groundwater with geogenic arsenic in Bangladesh. Science of the Total Environment, 678, 278–287. 10.1016/j.scitotenv.2019.04.426 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Khan, P. K. , Kesari, V. P. , & Kumar, A. (2013). Mouse micronucleus assay as a surrogate to assess genotoxic potential of arsenic at its human reference dose. Chemosphere, 90, 993–997. 10.1016/j.chemosphere.2012.07.021 [PubMed] [CrossRef] [Google Scholar]
  • Khlifi, R. , Olmedo, P. , Gil, F. , Molka, F. T. , Hammami, B. , Ahmed, R. , & Amel, H. C. (2014). Risk of laryngeal and nasopharyngeal cancer associated with arsenic and cadmium in the Tunisian population. Environmental Science and Pollution Research International, 21, 2032–2042. 10.1007/s11356-013-2105-z [PubMed] [CrossRef] [Google Scholar]
  • Kile, M. L. , Baccarelli, A. , Hoffman, E. , Tarantini, L. , Quamruzzaman, Q. , Rahman, M. , Mahiuddin, G. , Mostofa, G. , Hsueh, Y. M. , Wright, R. O. , & Christiani, D. C. (2012). Prenatal arsenic exposure and DNA methylation in maternal and umbilical cord blood leukocytes. Environmental Health Perspectives, 120, 1061–1066. 10.1289/ehp.1104173 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kile, M. L. , Cardenas, A. , Rodrigues, E. , Mazumdar, M. , Dobson, C. , Golam, M. , Quamruzzaman, Q. , Rahman, M. , & Christiani, D. C. (2016). Estimating effects of arsenic exposure during pregnancy on perinatal outcomes in a Bangladeshi cohort. Epidemiology, 27, 173–181. 10.1097/ede.0000000000000416 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kile, M. L. , Faraj, J. M. , Ronnenberg, A. G. , Quamruzzaman, Q. , Rahman, M. , Mostofa, G. , Afroz, S. , & Christiani, D. C. (2016). A cross sectional study of anemia and iron deficiency as risk factors for arsenic‐induced skin lesions in Bangladeshi women. BMC Public Health, 16, 158. 10.1186/s12889-016-2824-4 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kile, M. L. , Hoffman, E. , Rodrigues, E. G. , Breton, C. V. , Quamruzzaman, Q. , Rahman, M. , Mahiuddin, G. , Hsueh, Y. M. , & Christiani, D. C. (2011). A pathway‐based analysis of urinary arsenic metabolites and skin lesions. American Journal of Epidemiology, 173, 778–786. 10.1093/aje/kwq427 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kile, M. L. , Rodrigues, E. G. , Mazumdar, M. , Dobson, C. B. , Diao, N. , Golam, M. , Quamruzzaman, Q. , Rahman, M. , & Christiani, D. C. (2014). A prospective cohort study of the association between drinking water arsenic exposure and self‐reported maternal health symptoms during pregnancy in Bangladesh. Environmental Health, 13, 29. 10.1186/1476-069x-13-29 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kim, N. H. , Mason, C. C. , Nelson, R. G. , Afton, S. E. , Essader, A. S. , Medlin, J. E. , Levine, K. E. , Hoppin, J. A. , Lin, C. , Knowler, W. C. , & Sandler, D. P. (2013). Arsenic exposure and incidence of type 2 diabetes in southwestern American Indians. American Journal of Epidemiology, 177, 962–969. 10.1093/aje/kws329 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kim, T. H. , Seo, J. W. , Hong, Y. S. , & Song, K. H. (2017). Case‐control study of chronic low‐level exposure of inorganic arsenic species and non‐melanoma skin cancer. The Journal of Dermatology, 44, 1374–1379. 10.1111/1346-8138.13993 [PubMed] [CrossRef] [Google Scholar]
  • Kim, Y. , & Lee, B. K. (2011). Association between urinary arsenic and diabetes mellitus in the Korean general population according to KNHANES 2008. Science of the Total Environment, 409, 4054–4062. 10.1016/j.scitotenv.2011.06.003 [PubMed] [CrossRef] [Google Scholar]
  • Kippler, M. , Wagatsuma, Y. , Rahman, A. , Nermell, B. , Persson, L. , Raqib, R. , & Vahter, M. (2012). Environmental exposure to arsenic and cadmium during pregnancy and fetal size: A longitudinal study in rural Bangladesh. Reproductive Toxicology, 34, 504–511. 10.1016/j.reprotox.2012.08.002 [PubMed] [CrossRef] [Google Scholar]
  • Kligerman, A. D. , Malik, S. I. , & Campbell, J. A. (2010). Cytogenetic insights into DNA damage and repair of lesions induced by a monomethylated trivalent arsenical. Mutation Research, 695, 2–8. 10.1016/j.mrgentox.2009.09.007 [PubMed] [CrossRef] [Google Scholar]
  • Knobeloch, L. M. , Zierold, K. M. , & Anderson, H. A. (2006). Association of arsenic‐contaminated drinking‐water with prevalence of skin cancer in Wisconsin's Fox River valley. Journal of Health, Population, and Nutrition, 24, 206–213. [PubMed] [Google Scholar]
  • Koestler, D. C. , Avissar‐Whiting, M. , Houseman, E. A. , Karagas, M. R. , & Marsit, C. J. (2013). Differential DNA methylation in umbilical cord blood of infants exposed to low levels of arsenic in utero. Environmental Health Perspectives, 121, 971–977. 10.1289/ehp.1205925 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kojima, C. , Ramirez, D. C. , Tokar, E. J. , Himeno, S. , Drobná, Z. , Stýblo, M. , Mason, R. P. , & Waalkes, M. P. (2009). Requirement of arsenic biomethylation for oxidative DNA damage. Journal of the National Cancer Institute, 101, 1670–1681. 10.1093/jnci/djp414 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Konkel, L. (2020). Arsenic exposure and glucose metabolism: Experimental studies suggest implications for type 2 diabetes. Environmental Health Perspectives, 128, 94003. 10.1289/ehp7160 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kopp, B. , Zalko, D. , & Audebert, M. (2018). Genotoxicity of 11 heavy metals detected as food contaminants in two human cell lines. Environmental and Molecular Mutagenesis, 59, 202–210. 10.1002/em.22157 [PubMed] [CrossRef] [Google Scholar]
  • Koutros, S. , Baris, D. , Waddell, R. , Beane Freeman, L. E. , Colt, J. S. , Schwenn, M. , Johnson, A. , Ward, M. H. , Hosain, G. M. , Moore, L. E. , Stolzenberg‐Solomon, R. , Rothman, N. , Karagas, M. R. , & Silverman, D. T. (2018). Potential effect modifiers of the arsenic‐bladder cancer risk relationship. International Journal of Cancer, 143, 2640–2646. 10.1002/ijc.31720 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Krajewski, A. K. , Jimenez, M. P. , Rappazzo, K. M. , Lobdell, D. T. , & Jagai, J. S. (2021). Aggregated cumulative county arsenic in drinking water and associations with bladder, colorectal, and kidney cancers, accounting for population served. Journal of Exposure Science & Environmental Epidemiology, 31, 979–989. 10.1038/s41370-021-00314-8 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kuo, C.‐C. , Balakrishnan, P. , Gribble, M. O. , Best, L. G. , Goessler, W. , Umans, J. G. , & Navas‐Acien, A. (2022). The association of arsenic exposure and arsenic metabolism with all‐cause, cardiovascular and cancer mortality in the strong heart study. Environment International, 159, 107029. 10.1016/j.envint.2021.107029 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kuo, C. C. , Howard, B. V. , Umans, J. G. , Gribble, M. O. , Best, L. G. , Francesconi, K. A. , Goessler, W. , Lee, E. , Guallar, E. , & Navas‐Acien, A. (2015). Arsenic exposure, arsenic metabolism, and incident diabetes in the strong heart study. Diabetes Care, 38, 620–627. 10.2337/dc14-1641 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kuo, Y. C. , Lo, Y. S. , & Guo, H. R. (2017). Lung cancer associated with arsenic ingestion: Cell‐type specificity and dose response. Epidemiology, 28, S106–S112. 10.1097/EDE.0000000000000743 [PubMed] [CrossRef] [Google Scholar]
  • Kurzius‐Spencer, M. , da Silva, V. , Thomson, C. A. , Hartz, V. , Hsu, C. H. , Burgess, J. L. , O'Rourke, M. K. , & Harris, R. B. (2017). Nutrients in one‐carbon metabolism and urinary arsenic methylation in the National Health and nutrition examination survey (NHANES) 2003–2004. Science of the Total Environment, 607‐608, 381–390. 10.1016/j.scitotenv.2017.07.019 [PubMed] [CrossRef] [Google Scholar]
  • Kwok, R. K. , Kaufmann, R. B. , & Jakariya, M. (2006). Arsenic in drinking‐water and reproductive health outcomes: A study of participants in the Bangladesh integrated nutrition programme. Journal of Health Population and Nutrition, 24, 190–205. [PubMed] [Google Scholar]
  • Lai, Y. , Zhao, W. , Chen, C. , Wu, M. , & Zhang, Z. (2011). Role of DNA polymerase beta in the genotoxicity of arsenic. Environmental and Molecular Mutagenesis, 52, 460–468. 10.1002/em.20643 [PubMed] [CrossRef] [Google Scholar]
  • Laine, J. E. , Bailey, K. A. , Rubio‐Andrade, M. , Olshan, A. F. , Smeester, L. , Drobná, Z. , Herring, A. H. , Stýblo, M. , García‐Vargas, G. G. , & Fry, R. C. (2015). Maternal arsenic exposure, arsenic methylation efficiency, and birth outcomes in the biomarkers of exposure to ARsenic (BEAR) pregnancy cohort in Mexico. Environmental Health Perspectives, 123, 186–192. 10.1289/ehp.1307476 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lambrou, A. , Baccarelli, A. , Wright, R. O. , Weisskopf, M. , Bollati, V. , Amarasiriwardena, C. , Vokonas, P. , & Schwartz, J. (2012). Arsenic exposure and DNA methylation among elderly men. Epidemiology, 23, 668–676. 10.1097/EDE.0b013e31825afb0b [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lamm, S. H. , Robbins, S. A. , Zhou, C. , Lu, J. , Chen, R. , & Feinleib, M. (2013). Bladder/lung cancer mortality in Blackfoot‐disease (BFD)‐endemic area villages with low (< 150 μg/L) well water arsenic levels–an exploration of the dose‐response Poisson analysis. Regulatory Toxicology and Pharmacology, 65, 147–156. 10.1016/j.yrtph.2012.10.012 [PubMed] [CrossRef] [Google Scholar]
  • Lampron‐Goulet, É. , Gagnon, F. , & Langlois, M. F. (2017). Association between consumption of private well water contaminated by low levels of arsenic and dysglycemia in a rural region of Quebec, Canada. Environmental Research, 159, 232–238. 10.1016/j.envres.2017.07.049 [PubMed] [CrossRef] [Google Scholar]
  • Langley, R. L. , Kao, Y. , Mort, S. A. , Bateman, A. , Simpson, B. D. , & Reich, B. J. (2015). Adverse neurodevelopmental effects and hearing loss in children associated with manganese in well water, North Carolina, USA. Journal of Environmental and Occupational Science, 4, 62–69. 10.5455/jeos.20150403060427 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Langston, M. E. , Brown, H. E. , Lynch, C. F. , Roe, D. J. , & Dennis, L. K. (2022). Ambient UVR and environmental arsenic exposure in relation to cutaneous melanoma in Iowa. International Journal of Environmental Research and Public Health, 19(3), 1742. [PMC free article] [PubMed] [Google Scholar]
  • Lanphear, B. P. , Hornung, R. , Khoury, J. , Yolton, K. , Baghurst, P. , Bellinger, D. C. , Canfield, R. L. , Dietrich, K. N. , Bornschein, R. , Greene, T. , Rothenberg, S. J. , Needleman, H. L. , Schnaas, L. , Wasserman, G. , Graziano, J. , & Roberts, R. (2005). Low‐level environmental lead exposure and children's intellectual function: An international pooled analysis. Environmental Health Perspectives, 113, 894–899. 10.1289/ehp.7688 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lee, M. H. , Gao, Y. T. , Huang, Y. H. , McGee, E. E. , Lam, T. , Wang, B. , Shen, M. C. , Rashid, A. , Pfeiffer, R. M. , Hsing, A. W. , & Koshiol, J. (2020). A Metallomic approach to assess associations of serum metal levels with gallstones and gallbladder cancer. Hepatology, 71, 917–928. 10.1002/hep.30861 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lee, M.‐S. , Eum, K.‐D. , Golam, M. , Quamruzzaman, Q. , Kile, M. L. , Mazumdar, M. , & Christiani, D. C. (2021). Umbilical cord Blood metal mixtures and birth size in Bangladeshi children. Environmental Health Perspectives, 129, 57006. 10.1289/ehp7502 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Leffers, L. , Ebert, F. , Taleshi, M. S. , Francesconi, K. A. , & Schwerdtle, T. (2013). In vitro toxicological characterization of two arsenosugars and their metabolites. Molecular Nutrition & Food Research, 57, 1270–1282. 10.1002/mnfr.201200821 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Leonardi, G. , Vahter, M. , Clemens, F. , Goessler, W. , Gurzau, E. , Hemminki, K. , Hough, R. , Koppova, K. , Kumar, R. , Rudnai, P. , Surdu, S. , & Fletcher, T. (2012). Inorganic arsenic and basal cell carcinoma in areas of Hungary, Romania, and Slovakia: A case‐control study. Environmental Health Perspectives, 120, 721–726. 10.1289/ehp.1103534 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Levin‐Schwartz, Y. , Gennings, C. , Schnaas, L. , Del Carmen Hernández Chávez, M. , Bellinger, D. C. , Téllez‐Rojo, M. M. , Baccarelli, A. A. , & Wright, R. O. (2019). Time‐varying associations between prenatal metal mixtures and rapid visual processing in children. Environmental Health, 18, 92. 10.1186/s12940-019-0526-y [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lewis, D. R. , Southwick, J. W. , Ouellet‐Hellstrom, R. , Rench, J. , & Calderon, R. L. (1999). Drinking water arsenic in Utah: A cohort mortality study. Environmental Health Perspectives, 107, 359–365. 10.1289/ehp.99107359 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Li, H. , Engström, K. , Vahter, M. , & Broberg, K. (2012). Arsenic exposure through drinking water is associated with longer telomeres in peripheral blood. Chemical Research in Toxicology, 25, 2333–2339. 10.1021/tx300222t [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Li, H. , Li, H. , Li, Y. , Liu, Y. , & Zhao, Z. (2018). Blood mercury, arsenic, cadmium, and Lead in children with autism Spectrum disorder. Biological Trace Element Research, 181, 31–37. 10.1007/s12011-017-1002-6 [PubMed] [CrossRef] [Google Scholar]
  • Li, X. , Li, B. , Xi, S. , Zheng, Q. , Lv, X. , & Sun, G. (2013a). Prolonged environmental exposure of arsenic through drinking water on the risk of hypertension and type 2 diabetes. Environmental Science and Pollution Research International, 20, 8151–8161. 10.1007/s11356-013-1768-9 [PubMed] [CrossRef] [Google Scholar]
  • Li, X. , Li, B. , Xi, S. , Zheng, Q. , Wang, D. , & Sun, G. (2013b). Association of urinary monomethylated arsenic concentration and risk of hypertension: A cross‐sectional study from arsenic contaminated areas in northwestern China. Environmental Health, 12, 37. 10.1186/1476-069x-12-37 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Li, X. , Li, B. , Xu, Y. , Wang, Y. , Jin, Y. , Itoh, T. , Yoshida, T. , & Sun, G. (2011). Arsenic methylation capacity and its correlation with skin lesions induced by contaminated drinking water consumption in residents of chronic arsenicosis area. Environmental Toxicology, 26, 118–123. 10.1002/tox.20535 [PubMed] [CrossRef] [Google Scholar]
  • Li, X. , Ohgami, N. , Yajima, I. , Xu, H. , Iida, M. , Oshino, R. , Ninomiya, H. , Shen, D. , Ahsan, N. , Akhand, A. A. , & Kato, M. (2018). Arsenic level in toenails is associated with hearing loss in humans. PLoS One, 13, e0198743. 10.1371/journal.pone.0198743 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Li, X. , Wang, X. , & Park, S. K. (2021). Associations between rice consumption, arsenic metabolism, and insulin resistance in adults without diabetes. International Journal of Hygiene and Environmental Health, 237, 113834. 10.1016/j.ijheh.2021.113834 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Li, Z. , Lei, Z. , Wang, Y. , Wang, C. , Xu, J. , & Wang, Z. (2014). Studying cytotoxicity of low concentration arsenic on PC 12 cell line. Analytical Methods, 6, 1709. [Google Scholar]
  • Liao, C. M. , Chio, C. P. , Cheng, Y. H. , Hsieh, N. H. , Chen, W. Y. , & Chen, S. C. (2011). Quantitative links between arsenic exposure and influenza a (H1N1) infection‐associated lung function exacerbations risk. Risk Analysis, 31, 1281–1294. 10.1111/j.1539-6924.2010.01575.x [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liao, C. M. , Lin, T. L. , Hsieh, N. H. , & Chen, W. Y. (2010). Assessing the arsenic‐contaminated rice (Oryza sativa) associated children skin lesions. Journal of Hazardous Materials, 176, 239–251. 10.1016/j.jhazmat.2009.11.019 [PubMed] [CrossRef] [Google Scholar]
  • Liao, C. M. , Shen, H. H. , Chen, C. L. , Hsu, L. I. , Lin, T. L. , Chen, S. C. , & Chen, C. J. (2009). Risk assessment of arsenic‐induced internal cancer at long‐term low dose exposure. Journal of Hazardous Materials, 165, 652–663. 10.1016/j.jhazmat.2008.10.095 [PubMed] [CrossRef] [Google Scholar]
  • Liao, K. W. , Chang, C. H. , Tsai, M. S. , Chien, L. C. , Chung, M. Y. , Mao, I. F. , Tsai, Y. A. , & Chen, M. L. (2018). Associations between urinary total arsenic levels, fetal development, and neonatal birth outcomes: A cohort study in Taiwan. Science of the Total Environment, 612, 1373–1379. 10.1016/j.scitotenv.2017.08.312 [PubMed] [CrossRef] [Google Scholar]
  • Liao, Y. T. , Chen, C. J. , Li, W. F. , Hsu, L. I. , Tsai, L. Y. , Huang, Y. L. , Sun, C. W. , Chen, W. J. , & Wang, S. L. (2012). Elevated lactate dehydrogenase activity and increased cardiovascular mortality in the arsenic‐endemic areas of southwestern Taiwan. Toxicology and Applied Pharmacology, 262, 232–237. 10.1016/j.taap.2012.04.028 [PubMed] [CrossRef] [Google Scholar]
  • Lin, C. C. , Chen, Y. C. , Su, F. C. , Lin, C. M. , Liao, H. F. , Hwang, Y. H. , Hsieh, W. S. , Jeng, S. F. , Su, Y. N. , & Chen, P. C. (2013). In utero exposure to environmental lead and manganese and neurodevelopment at 2 years of age. Environmental Research, 123, 52–57. 10.1016/j.envres.2013.03.003 [PubMed] [CrossRef] [Google Scholar]
  • Lin, C. H. , Hsu, Y. T. , Yen, C. C. , Chen, H. H. , Tseng, C. J. , Lo, Y. K. , & Chan, J. Y. H. (2018). Association between heavy metal levels and acute ischemic stroke. Journal of Biomedical Science, 25, 49. 10.1186/s12929-018-0446-0 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lin, H. C. , Huang, Y. K. , Shiue, H. S. , Chen, L. S. , Choy, C. S. , Huang, S. R. , Han, B. C. , & Hsueh, Y. M. (2014). Arsenic methylation capacity and obesity are associated with insulin resistance in obese children and adolescents. Food and Chemical Toxicology, 74, 60–67. 10.1016/j.fct.2014.08.018 [PubMed] [CrossRef] [Google Scholar]
  • Lin, P. D. , Bromage, S. , Mostofa, M. G. , Rahman, M. , Allen, J. , Oken, E. , Kile, M. L. , & Christiani, D. C. (2019). Mediating role of arsenic in the relationship between diet and pregnancy outcomes: Prospective birth cohort in Bangladesh. Environmental Health, 18, 10. 10.1186/s12940-019-0450-1 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lin, Y. , Zhuang, L. , Yi, H. , Xu, L. , Huang, H. , He, D. , Zhao, X. , Ma, H. , & Wu, L. (2018). Embryonic protective role of folate in arsenic‐induced cardiac malformations in rats. International Journal of Clinical and Experimental Pathology, 11, 1946–1955. [PMC free article] [PubMed] [Google Scholar]
  • Linares, A. M. , Unrine, J. M. , Thaxton Wigging, A. , Tantalean, J. C. , & Radulescu, V. C. (2021). Blood's concentration of Lead and arsenic associated with anemia in Peruvian children. Journal of Environmental and Public Health, 2021, 1–8. 10.1155/2021/7283514 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lindberg, A. L. , Ekström, E. C. , Nermell, B. , Rahman, M. , Lönnerdal, B. , Persson, L. A. , & Vahter, M. (2008). Gender and age differences in the metabolism of inorganic arsenic in a highly exposed population in Bangladesh. Environmental Research, 106, 110–120. 10.1016/j.envres.2007.08.011 [PubMed] [CrossRef] [Google Scholar]
  • Lisabeth, L. D. , Ahn, H. J. , Chen, J. J. , Sealy‐Jefferson, S. , Burke, J. F. , & Meliker, J. R. (2010). Arsenic in drinking water and stroke hospitalizations in Michigan. Stroke, 41, 2499–2504. 10.1161/strokeaha.110.585281 [PubMed] [CrossRef] [Google Scholar]
  • Litwin, I. , Bocer, T. , Dziadkowiec, D. , & Wysocki, R. (2013). Oxidative stress and replication‐independent DNA breakage induced by arsenic in Saccharomyces cerevisiae. PLoS Genetics, 9, e1003640. 10.1371/journal.pgen.1003640 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu, B. , Feng, W. , Wang, J. , Li, Y. , Han, X. , Hu, H. , Guo, H. , Zhang, X. , & He, M. (2016). Association of urinary metals levels with type 2 diabetes risk in coke oven workers. Environmental Pollution, 210, 1–8. 10.1016/j.envpol.2015.11.046 [PubMed] [CrossRef] [Google Scholar]
  • Liu, H. , Lu, S. , Zhang, B. , Xia, W. , Liu, W. , Peng, Y. , Zhang, H. , Wu, K. , Xu, S. , & Li, Y. (2018). Maternal arsenic exposure and birth outcomes: A birth cohort study in Wuhan, China. Environmental Pollution, 236, 817–823. 10.1016/j.envpol.2018.02.012 [PubMed] [CrossRef] [Google Scholar]
  • Liu, J. , Gao, Y. , Liu, H. , Sun, J. , Liu, Y. , Wu, J. , Li, D. , & Sun, D. (2017). Assessment of relationship on excess arsenic intake from drinking water and cognitive impairment in adults and elders in arsenicosis areas. International Journal of Hygiene and Environmental Health, 220, 424–430. 10.1016/j.ijheh.2016.12.004 [PubMed] [CrossRef] [Google Scholar]
  • Liu, J. , Song, W. , Li, Y. , Wang, Y. , Cui, Y. , Huang, J. , Wang, Q. , & Wei, S. (2022). Burden of coronary heart disease and cancer from dietary exposure to inorganic arsenic in adults in China, 2016. Annals of Global Health, 88, 28. 10.5334/aogh.3620 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu, J. , & Waalkes, M. P. (2008). Liver is a target of arsenic carcinogenesis. Toxicological Sciences, 105, 24–32. 10.1093/toxsci/kfn120 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu, R. , Nelson, D. O. , Hurley, S. , Hertz, A. , & Reynolds, P. (2015). Residential exposure to estrogen disrupting hazardous air pollutants and breast cancer risk: The California teachers study. Epidemiology, 26, 365–373. 10.1097/ede.0000000000000277 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu, W. , Wang, B. , Zhao, Y. , Wu, Z. , Dong, A. , Chen, H. , Lin, L. , Lu, J. , & Hai, X. (2021). Pharmacokinetic characteristics, tissue bioaccumulation and toxicity profiles of Oral arsenic trioxide in rats: Implications for the treatment and risk assessment of acute Promyelocytic leukemia. Frontiers in Pharmacology, 12. 10.3389/fphar.2021.647687 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu, X. , Zheng, Y. , Zhang, W. , Zhang, X. , Lioyd‐Jones, D. M. , Baccarelli, A. A. , Ning, H. , Fornage, M. , He, K. , Liu, K. , & Hou, L. (2014). Blood methylomics in response to arsenic exposure in a low‐exposed US population. Journal of Exposure Science & Environmental Epidemiology, 24, 145–149. 10.1038/jes.2013.89 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu‐Mares, W. , Mackinnon, J. A. , Sherman, R. , Fleming, L. E. , Rocha‐Lima, C. , Hu, J. J. , & Lee, D. J. (2013). Pancreatic cancer clusters and arsenic‐contaminated drinking water wells in Florida. BMC Cancer, 13, 111. 10.1186/1471-2407-13-111 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • López‐Carrillo, L. , Gamboa‐Loira, B. , Becerra, W. , Hernández‐Alcaraz, C. , Hernández‐Ramírez, R. U. , Gandolfi, A. J. , Franco‐Marina, F. , & Cebrián, M. E. (2016). Dietary micronutrient intake and its relationship with arsenic metabolism in Mexican women. Environmental Research, 151, 445–450. 10.1016/j.envres.2016.08.015 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • López‐Carrillo, L. , Gamboa‐Loira, B. , Gandolfi, A. J. , & Cebrián, M. E. (2020). Inorganic arsenic methylation capacity and breast cancer by immunohistochemical subtypes in northern Mexican women. Environmental Research, 184, 109361. 10.1016/j.envres.2020.109361 [PubMed] [CrossRef] [Google Scholar]
  • López‐Carrillo, L. , Hernández‐Ramírez, R. U. , Gandolfi, A. J. , Ornelas‐Aguirre, J. M. , Torres‐Sánchez, L. , & Cebrian, M. E. (2014). Arsenic methylation capacity is associated with breast cancer in northern Mexico. Toxicology and Applied Pharmacology, 280, 53–59. 10.1016/j.taap.2014.07.013 [PubMed] [CrossRef] [Google Scholar]
  • Lucchini, R. G. , Guazzetti, S. , Renzetti, S. , Conversano, M. , Cagna, G. , Fedrighi, C. , Giorgino, A. , Peli, M. , Placidi, D. , Zoni, S. , Forte, G. , Majorani, C. , Pino, A. , Senofonte, O. , Petrucci, F. , & Alimonti, A. (2019). Neurocognitive impact of metal exposure and social stressors among schoolchildren in Taranto, Italy. Environmental Health, 18, 67. 10.1186/s12940-019-0505-3 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lucio, M. , Barbir, R. , Vučić Lovrenčić, M. , Canecki Varžić, S. , Ljubić, S. , Smirčić Duvnjak, L. , Šerić, V. , Milić, M. , Tariba Lovaković, B. , Krivohlavek, A. , Vinković Vrček, I. , & Michalke, B. (2020). Association between arsenic exposure and biomarkers of type 2 diabetes mellitus in a Croatian population: A comparative observational pilot study. Science of the Total Environment, 720, 137575. 10.1016/j.scitotenv.2020.137575 [PubMed] [CrossRef] [Google Scholar]
  • Majewska, M. D. , Urbanowicz, E. , Rok‐Bujko, P. , Namyslowska, I. , & Mierzejewski, P. (2010). Age‐dependent lower or higher levels of hair mercury in autistic children than in healthy controls. Acta Neurobiologiae Experimentalis (Wars), 70, 196–208. [PubMed] [Google Scholar]
  • Majumdar, K. K. , & Guha Mazumder, D. N. (2012). Effect of drinking arsenic‐contaminated water in children. Indian Journal of Public Health, 56, 223–226. 10.4103/0019-557x.104250 [PubMed] [CrossRef] [Google Scholar]
  • Majumdar, S. , Chanda, S. , Ganguli, B. , Guha Mazumder, D. N. , Lahiri, S. , & Dasgupta, U. B. (2010). Arsenic exposure induces genomic hypermethylation. Environmental Toxicology, 25, 315–318. 10.1002/tox.20497 [PubMed] [CrossRef] [Google Scholar]
  • Makris, K. C. , Christophi, C. A. , Paisi, M. , & Ettinger, A. S. (2012). A preliminary assessment of low level arsenic exposure and diabetes mellitus in Cyprus. BMC Public Health, 12, 334. 10.1186/1471-2458-12-334 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mannetje, A. , Bencko, V. , Brennan, P. , Zaridze, D. , Szeszenia‐Dabrowska, N. , Rudnai, P. , Lissowska, J. , Fabiánová, E. , Cassidy, A. , Mates, D. , Foretova, L. , Janout, V. , Fevotte, J. , Fletcher, T. , & Boffetta, P. (2011). Occupational exposure to metal compounds and lung cancer. Results from a multi‐center case‐control study in central/Eastern Europe and UK. Cancer Causes & Control, 22, 1669–1680. 10.1007/s10552-011-9843-3 [PubMed] [CrossRef] [Google Scholar]
  • Mar Wai, K. , Umezaki, M. , Mar, O. , Umemura, M. , & Watanabe, C. (2019). Arsenic exposure through drinking Water and oxidative stress status: A cross‐sectional study in the Ayeyarwady region, Myanmar. Journal of Trace Elements in Medicine and Biology, 54, 103–109. 10.1016/j.jtemb.2019.04.009 [PubMed] [CrossRef] [Google Scholar]
  • Marciniak, W. , Derkacz, R. , Muszyńska, M. , Baszuk, P. , Gronwald, J. , Huzarski, T. , Cybulski, C. , Jakubowska, A. , Falco, M. , Dębniak, T. , Lener, M. , Oszurek, O. , Pullella, K. , Kotsopoulos, J. , Sun, P. , Narod, S. A. , & Lubiński, J. (2020). Blood arsenic levels and the risk of familial breast cancer in Poland. International Journal of Cancer, 146, 2721–2727. 10.1002/ijc.32595 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Marciniak, W. , Matoušek, T. , Domchek, S. , Paradiso, A. , Patruno, M. , Irmejs, A. , Roderte, I. , Derkacz, R. , Baszuk, P. , Kuświk, M. , Cybulski, C. , Huzarski, T. , Gronwald, J. , Dębniak, T. , Falco, M. , Lener, M. R. , Jakubowska, A. , Pullella, K. , Kotsopoulos, J. , … Lubiński, J. (2021). Blood arsenic levels as a marker of breast cancer risk among BRCA1 carriers. Cancers (Basel), 13. 10.3390/cancers13133345 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Marie, C. , Léger, S. , Guttmann, A. , Marchiset, N. , Rivière, O. , Perthus, I. , Lémery, D. , Vendittelli, F. , & Sauvant‐Rochat, M. P. (2018). In utero exposure to arsenic in tap water and congenital anomalies: A French semi‐ecological study. International Journal of Hygiene and Environmental Health, 221, 1116–1123. 10.1016/j.ijheh.2018.07.012 [PubMed] [CrossRef] [Google Scholar]
  • Martinez, V. D. , Buys, T. P. , Adonis, M. , Benítez, H. , Gallegos, I. , Lam, S. , Lam, W. L. , & Gil, L. (2010). Arsenic‐related DNA copy‐number alterations in lung squamous cell carcinomas. British Journal of Cancer, 103, 1277–1283. 10.1038/sj.bjc.6605879 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Martínez‐Barquero, V. , de Marco, G. , Martínez‐Hervas, S. , Rentero, P. , Galan‐Chilet, I. , Blesa, S. , Morchon, D. , Morcillo, S. , Rojo, G. , Ascaso, J. F. , Real, J. T. , Martín‐Escudero, J. C. , & Chaves, F. J. (2015). Polymorphisms in endothelin system genes, arsenic levels and obesity risk. PLoS One, 10, e0118471. 10.1371/journal.pone.0118471 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mateen, F. J. , Grau‐Perez, M. , Pollak, J. S. , Moon, K. A. , Howard, B. V. , Umans, J. G. , Best, L. G. , Francesconi, K. A. , Goessler, W. , Crainiceanu, C. , Guallar, E. , Devereux, R. B. , Roman, M. J. , & Navas‐Acien, A. (2017). Chronic arsenic exposure and risk of carotid artery disease: The strong heart study. Environmental Research, 157, 127–134. 10.1016/j.envres.2017.05.020 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mathew, L. , Vale, A. , & Adcock, J. E. (2010). Arsenical peripheral neuropathy. Practical Neurology, 10, 34–38. 10.1136/jnnp.2009.201830 [PubMed] [CrossRef] [Google Scholar]
  • Mazumdar, M. , Ibne Hasan, M. O. , Hamid, R. , Valeri, L. , Paul, L. , Selhub, J. , Rodrigues, E. G. , Silva, F. , Mia, S. , Mostofa, M. G. , Quamruzzaman, Q. , Rahman, M. , & Christiani, D. C. (2015). Arsenic is associated with reduced effect of folic acid in myelomeningocele prevention: A case control study in Bangladesh. Environmental Health, 14, 34. 10.1186/s12940-015-0020-0 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • McClintock, T. R. , Chen, Y. , Parvez, F. , Makarov, D. V. , Ge, W. , Islam, T. , Ahmed, A. , Rakibuz‐Zaman, M. , Hasan, R. , Sarwar, G. , Slavkovich, V. , Bjurlin, M. A. , Graziano, J. H. , & Ahsan, H. (2014). Association between arsenic exposure from drinking water and hematuria: Results from the health effects of arsenic longitudinal study. Toxicology and Applied Pharmacology, 276, 21–27. 10.1016/j.taap.2014.01.015 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • McDermott, S. , Bao, W. , Marjorie Aelion, C. , Cai, B. , & Lawson, A. (2012). When are fetuses and young children most susceptible to soil metal concentrations of arsenic, lead and mercury? Spatial and Spatiotemporal Epidemiology, 3(3), 265–272. 10.1016/j.sste.2012.05.001 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • McKeating, D. R. , Fisher, J. J. , MacDonald, T. , Walker, S. , Tong, S. , Bennett, W. W. , Kaitu'u‐Lino, T. J. , & Perkins, A. V. (2021). Circulating trace elements for the prediction of preeclampsia and small for gestational age babies. Metabolomics, 17(10), 90. 10.1007/s11306-021-01840-0 [PubMed] [CrossRef] [Google Scholar]
  • McLeod, L. , Bharadwaj, L. , Epp, T. Y. , & Waldner, C. L. (2018). Ecological analysis of associations between groundwater quality and hypertension and cardiovascular disease in rural Saskatchewan, Canada using Bayesian hierarchical models and administrative health data. Environmental Research, 167, 329–340. 10.1016/j.envres.2018.07.038 [PubMed] [CrossRef] [Google Scholar]
  • McLeod, L. , Bharadwaj, L. , Epp, T. Y. , & Waldner, C. L. (2019). Bayesian hierarchical models as tools to evaluate the association between groundwater quality and the occurrence of type 2 diabetes in rural Saskatchewan, Canada. Archives of Environmental Contamination and Toxicology, 76, 375–393. 10.1007/s00244-018-00588-4 [PubMed] [CrossRef] [Google Scholar]
  • McNally, E. J. , Luncsford, P. J. , & Armanios, M. (2019). Long telomeres and cancer risk: The price of cellular immortality. The Journal of Clinical Investigation, 129(9), 3474–3481. 10.1172/JCI120851 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Medrano, M. A. , Boix, R. , Pastor‐Barriuso, R. , Palau, M. , Damián, J. , Ramis, R. , Del Barrio, J. L. , & Navas‐Acien, A. (2010). Arsenic in public water supplies and cardiovascular mortality in Spain. Environmental Research, 110, 448–454. 10.1016/j.envres.2009.10.002 [PubMed] [CrossRef] [Google Scholar]
  • Meharg, A. A. , Williams, P. N. , Deacon, C. M. , Norton, G. J. , Hossain, M. , Louhing, D. , Marwa, E. , Lawgalwi, Y. , Taggart, M. , Cascio, C. , & Haris, P. (2014). Urinary excretion of arsenic following rice consumption. Environmental Pollution, 194, 181–187. 10.1016/j.envpol.2014.07.031 [PubMed] [CrossRef] [Google Scholar]
  • Mehta, M. , & Hundal, S. S. (2014). Assessment of genotoxic potential of arsenic in female albino rats at permissible dose levels. Toxicology International, 21(1), 24–28. 10.4103/0971-6580.128787 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Melak, D. , Ferreccio, C. , Kalman, D. , Parra, R. , Acevedo, J. , Pérez, L. , Cortés, S. , Smith, A. H. , Yuan, Y. , Liaw, J. , & Steinmaus, C. (2014). Arsenic methylation and lung and bladder cancer in a case‐control study in northern Chile. Toxicology and Applied Pharmacology, 274, 225–231. 10.1016/j.taap.2013.11.014 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Meliker, J. R. , Slotnick, M. J. , AvRuskin, G. A. , Schottenfeld, D. , Jacquez, G. M. , Wilson, M. L. , Goovaerts, P. , Franzblau, A. , & Nriagu, J. O. (2010). Lifetime exposure to arsenic in drinking water and bladder cancer: A population‐based case‐control study in Michigan, USA. Cancer Causes & Control, 21, 745–757. 10.1007/s10552-010-9503-z [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Meliker, J. R. , Wahl, R. L. , Cameron, L. L. , & Nriagu, J. O. (2007). Arsenic in drinking water and cerebrovascular disease, diabetes mellitus, and kidney disease in Michigan: A standardized mortality ratio analysis. Environmental Health, 6(1), 1–11. [PMC free article] [PubMed] [Google Scholar]
  • Mendez, W., Jr. , Shao, K. , Lee, J. S. , Cote, I. , Druwe, I. L. , Davis, A. , & Gift, J. S. (2020). Model averaging methods for the evaluation of dose‐response model uncertainty when assessing the suitability of studies for estimating risk. Environment International, 143, 105857. 10.1016/j.envint.2020.105857 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mendez, W. M., Jr. , Eftim, S. , Cohen, J. , Warren, I. , Cowden, J. , Lee, J. S. , & Sams, R. (2017). Relationships between arsenic concentrations in drinking water and lung and bladder cancer incidence in U.S. counties. Journal of Exposure Science & Environmental Epidemiology, 27, 235–243. 10.1038/jes.2016.58 [PubMed] [CrossRef] [Google Scholar]
  • Mendez, M. A. , González‐Horta, C. , Sánchez‐Ramírez, B. , Ballinas‐Casarrubias, L. , Cerón, R. H. , Morales, D. V. , Terrazas, F. A. , Ishida, M. C. , Gutiérrez‐Torres, D. S. , Saunders, R. J. , Drobná, Z. , Fry, R. C. , Buse, J. B. , Loomis, D. , García‐Vargas, G. G. , Del Razo, L. M. , & Stýblo, M. (2016). Chronic exposure to arsenic and markers of Cardiometabolic risk: A cross‐sectional study in Chihuahua, Mexico. Environmental Health Perspectives, 124, 104–111. 10.1289/ehp.1408742 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Merced‐Nieves, F. M. , Chelonis, J. , Pantic, I. , Schnass, L. , Tellez‐Rojo, M. M. , Braun, J. M. , Paule, M. G. , Wright, R. J. , Wright, R. O. , & Curtin, P. (2022). Prenatal trace elements mixture is associated with learning deficits on a behavioral acquisition task among young children. New Directions for Child and Adolescent Development., 2022, 53–66. 10.1002/cad.20458 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Merrill, P. D. , Ampah, S. B. , He, K. , Rembert, N. J. , Brockman, J. , Kleindorfer, D. , & McClure, L. A. (2017). Association between trace elements in the environment and stroke risk: The reasons for geographic and racial differences in stroke (REGARDS) study. Journal of Trace Elements in Medicine and Biology, 42, 45–49. 10.1016/j.jtemb.2017.04.003 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Meyer, R. , Carey, M. P. , Turner, P. J. , & Meharg, A. A. (2018). Low inorganic arsenic in hydrolysed rice formula used for cow's milk protein allergy. Pediatric Allergy and Immunology, 29, 561–563. 10.1111/pai.12913.29701313 [PubMed] [CrossRef] [Google Scholar]
  • Meyer, S. , Raber, G. , Ebert, F. , Leffers, L. , Müller, S. M. , Taleshi, M. S. , Francesconi, K. A. , & Schwerdtle, T. (2015). In vitro toxicological characterisation of arsenic‐containing fatty acids and three of their metabolites. Toxicology Research (Camb), 4, 1289–1296. 10.1039/c5tx00122f [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Middleton, D. R. S. , Watts, M. J. , Hamilton, E. M. , Ander, E. L. , Close, R. M. , Exley, K. S. , Crabbe, H. , Leonardi, G. S. , Fletcher, T. , & Polya, D. A. (2016). Urinary arsenic profiles reveal exposures to inorganic arsenic from private drinking water supplies in Cornwall, UK. Scientific Reports, 6, 25656. 10.1038/srep25656 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Miklavcic, A. , Casetta, A. , Snoj Tratnik, J. , Mazej, D. , Krsnik, M. , Mariuz, M. , Sofianou, K. , Spirić, Z. , Barbone, F. , & Horvat, M. (2013). Mercury, arsenic and selenium exposure levels in relation to fish consumption in the Mediterranean area. Environmental Research, 120, 7–17. 10.1016/j.envres.2012.08.010 [PubMed] [CrossRef] [Google Scholar]
  • Milton, A. H. , Hussain, S. , Akter, S. , Rahman, M. , Mouly, T. A. , & Mitchell, K. (2017). A review of the effects of chronic arsenic exposure on adverse pregnancy outcomes. International Journal of Environmental Research and Public Health, 14. 10.3390/ijerph14060556 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Milton, A. H. , Smith, W. , Rahman, B. , Hasan, Z. , Kulsum, U. , Dear, K. , Rakibuddin, M. , & Ali, A. (2005). Chronic arsenic exposure and adverse pregnancy outcomes in Bangladesh. Epidemiology, 16(1), 82–86. [PubMed] [Google Scholar]
  • Moazed, V. , Jafari, E. , Ebadzadeh, M. R. , Pourzare, A. , Gharehchahi, H. R. , Mangeli, F. , & Jamali, E. (2021). Association between serum cadmium and arsenic levels with bladder cancer: A case‐control study. International Journal of Cancer Management, 14(7). [Google Scholar]
  • Mochizuki, H. , Phyu, K. P. , Aung, M. N. , Zin, P. W. , Yano, Y. , Myint, M. Z. , Thit, W. M. , Yamamoto, Y. , Hishikawa, Y. , Thant, K. Z. , Maruyama, M. , & Kuroda, Y. (2019). Peripheral neuropathy induced by drinking water contaminated with low‐dose arsenic in Myanmar. Environmental Health and Preventive Medicine, 24, 23. 10.1186/s12199-019-0781-0 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Moe, B. , Peng, H. , Lu, X. , Chen, B. , Chen, L. W. L. , Gabos, S. , Li, X. F. , & Le, X. C. (2016). Comparative cytotoxicity of fourteen trivalent and pentavalent arsenic species determined using real‐time cell sensing. Journal of Environmental Sciences (China), 49, 113–124. [PubMed] [Google Scholar]
  • Mondal, V. , Hosen, Z. , Hossen, F. , Siddique, A. E. , Tony, S. R. , Islam, Z. , Islam, M. S. , Hossain, S. , Islam, K. , Sarker, M. K. , Hasibuzzaman, M. M. , Liu, L. Z. , Jiang, B. H. , Hoque, M. M. , Saud, Z. A. , Xin, L. , Himeno, S. , & Hossain, K. (2020). Arsenic exposure‐related hyperglycemia is linked to insulin resistance with concomitant reduction of skeletal muscle mass. Environment International, 143, 105890. 10.1016/j.envint.2020.105890 [PubMed] [CrossRef] [Google Scholar]
  • Monrad, M. , Ersbøll, A. K. , Sørensen, M. , Baastrup, R. , Hansen, B. , Gammelmark, A. , Tjønneland, A. , Overvad, K. , & Raaschou‐Nielsen, O. (2017). Low‐level arsenic in drinking water and risk of incident myocardial infarction: A cohort study. Environmental Research, 154, 318–324. 10.1016/j.envres.2017.01.028 [PubMed] [CrossRef] [Google Scholar]
  • Moon, K. , Guallar, E. , & Navas‐Acien, A. (2012). Arsenic exposure and cardiovascular disease: An updated systematic review. Current Atherosclerosis Reports, 14, 542–555. 10.1007/s11883-012-0280-x [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Moon, K. A. , Guallar, E. , Umans, J. G. , Devereux, R. B. , Best, L. G. , Francesconi, K. A. , Goessler, W. , Pollak, J. , Silbergeld, E. K. , Howard, B. V. , & Navas‐Acien, A. (2013). Association between exposure to low to moderate arsenic levels and incident cardiovascular disease. A prospective cohort study. Annals of Internal Medicine, 159, 649–659. 10.7326/0003-4819-159-10-201311190-00719 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Moon, K. A. , Oberoi, S. , Barchowsky, A. , Chen, Y. , Guallar, E. , Nachman, K. E. , Rahman, M. , Sohel, N. , D'Ippoliti, D. , Wade, T. J. , James, K. A. , Farzan, S. F. , Karagas, M. R. , Ahsan, H. , & Navas‐Acien, A. (2017). A dose‐response meta‐analysis of chronic arsenic exposure and incident cardiovascular disease. International Journal of Epidemiology, 46, 1924–1939. 10.1093/ije/dyx202 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mordukhovich, I. , Wright, R. O. , Hu, H. , Amarasiriwardena, C. , Baccarelli, A. , Litonjua, A. , Sparrow, D. , Vokonas, P. , & Schwartz, J. (2012). Associations of toenail arsenic, cadmium, mercury, manganese, and lead with blood pressure in the normative aging study. Environmental Health Perspectives, 120, 98–104. 10.1289/ehp.1002805 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Moriya, M. , & Grollman, A. P. (1993). Mutations in the mutY gene of Escherichia coli enhance the frequency of targeted G:C → T:A transversions induced by a single 8‐oxoguanine residue in single‐stranded DNA. Molecular and General Genetics MGG, 239, 72–76. [PubMed] [Google Scholar]
  • Mostafa, M. G. , & Cherry, N. (2013). Arsenic in drinking water and renal cancers in rural Bangladesh. Occupational and Environmental Medicine, 70(11), 768–773. 10.1136/oemed-2013-101443 [PubMed] [CrossRef] [Google Scholar]
  • Mostafa, M. G. , & Cherry, N. (2015). Arsenic in drinking Water, transition cell cancer and chronic cystitis in rural Bangladesh. International Journal of Environmental Research and Public Health, 12, 13739–13749. 10.3390/ijerph121113739 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Muenyi, C. S. , Ljungman, M. , & States, J. C. (2015). Arsenic disruption of DNA damage responses‐potential role in carcinogenesis and chemotherapy. Biomolecules, 5(4), 2184–2193. 10.3390/biom5042184 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mukherjee, B. , Bindhani, B. , Saha, H. , Sinha, D. , & Ray, M. R. (2014). Platelet hyperactivity, neurobehavioral symptoms and depression among Indian women chronically exposed to low level of arsenic. Neurotoxicology, 45, 159–167. 10.1016/j.neuro.2014.10.011 [PubMed] [CrossRef] [Google Scholar]
  • Mukherjee, S. C. , Saha, K. C. , Pati, S. , Dutta, R. N. , Rahman, M. M. , Sengupta, M. K. , Ahamed, S. , Lodh, D. , Das, B. , Hossain, M. A. , Nayak, B. , Mukherjee, A. , Chakraborti, D. , Dulta, S. K. , Palit, S. K. , Kaies, I. , Barua, A. K. , & Asad, K. A. (2005). Murshidabad–one of the nine groundwater arsenic‐affected districts of West Bengal, India. Part II: Dermatological, neurological, and obstetric findings. Clinical Toxicology (Philadelphia, Pa.), 43, 835–848. 10.1080/15563650500357495 [PubMed] [CrossRef] [Google Scholar]
  • Mullin, A. M. , Amarasiriwardena, C. , Cantoral‐Preciado, A. , Claus Henn, B. , Leon Hsu, H. H. , Sanders, A. P. , Svensson, K. , Tamayo‐Ortiz, M. , Téllez‐Rojo, M. M. , Wright, R. O. , & Burris, H. H. (2019). Maternal blood arsenic levels and associations with birth weight‐for‐gestational age. Environmental Research, 177, 108603. 10.1016/j.envres.2019.108603 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Muñoz, M. P. , Valdés, M. , Muñoz‐Quezada, M. T. , Lucero, B. , Rubilar, P. , Pino, P. , & Iglesias, V. (2018). Urinary inorganic arsenic concentration and gestational diabetes mellitus in pregnant women from Arica, Chile. International Journal of Environmental Research and Public Health, 15. 10.3390/ijerph15071418 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Muñoz, O. , Bastias, J. M. , Araya, M. , Morales, A. , Orellana, C. , Rebolledo, R. , & Velez, D. (2005). Estimation of the dietary intake of cadmium, lead, mercury, and arsenic by the population of Santiago (Chile) using a Total diet study. Food and Chemical Toxicology, 43, 1647–1655. 10.1016/j.fct.2005.05.006 [PubMed] [CrossRef] [Google Scholar]
  • Muse, M. E. , Li, Z. , Baker, E. R. , Cottingham, K. L. , Korrick, S. A. , Karagas, M. R. , & Gilbert‐Diamond, D. (2020). Relation between in utero arsenic exposure and growth during the first year of life in a New Hampshire pregnancy cohort. Environmental Research, 180, 108604. 10.1016/j.envres.2019.108604 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Myers, S. L. , Lobdell, D. T. , Liu, Z. , Xia, Y. , Ren, H. , Li, Y. , Kwok, R. K. , Mumford, J. L. , & Mendola, P. (2010). Maternal drinking water arsenic exposure and perinatal outcomes in Inner Mongolia, China. Journal of Epidemiology and Community Health, 64, 325–329. 10.1136/jech.2008.084392 [PubMed] [CrossRef] [Google Scholar]
  • Na, L. , Xiumei, Z. , Lingzi, Z. , Deqin, H. , Xuanxuan, Z. , Huanhuan, G. , Yuan, L. , & Xiujuan, C. (2020). Research into the intervention effect of folic acid on arsenic‐induced heart abnormalities in fetal rats during the periconception period. BMC Cardiovascular Disorders, 20, 139. 10.1186/s12872-020-01418-z [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nafees, A. A. , Kazi, A. , Fatmi, Z. , Irfan, M. , Ali, A. , & Kayama, F. (2011). Lung function decrement with arsenic exposure to drinking groundwater along river Indus: A comparative cross‐sectional study. Environmental Geochemistry and Health, 33(2), 203–216. [PubMed] [Google Scholar]
  • Nahar, M. N. , Inaoka, T. , & Fujimura, M. (2014). A consecutive study on arsenic exposure and intelligence quotient (IQ) of children in Bangladesh. Environmental Health and Preventive Medicine, 19, 194–199. 10.1007/s12199-013-0374-2 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nahar, M. N. , Inaoka, T. , Fujimura, M. , Watanabe, C. , Shimizu, H. , Tasmin, S. , & Sultana, N. (2014). Arsenic contamination in groundwater and its effects on adolescent intelligence and social competence in Bangladesh with special reference to daily drinking/cooking water intake. Environmental Health and Preventive Medicine, 19, 151–158. 10.1007/s12199-013-0369-z [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Naranmandura, H. , Carew, M. W. , Xu, S. , Lee, J. , Leslie, E. M. , Weinfeld, M. , & Le, X. C. (2011). Comparative toxicity of arsenic metabolites in human bladder cancer EJ‐1 cells. Chemical Research in Toxicology, 24(9), 1586–1596. 10.1021/tx200291p [PubMed] [CrossRef] [Google Scholar]
  • Naranmandura, H. , Xu, S. , Sawata, T. , Hao, W. H. , Liu, H. , Bu, N. , Ogra, Y. , Lou, Y. J. , & Suzuki, N. (2011). Mitochondria are the main target organelle for trivalent monomethylarsonous acid (MMA(III))‐induced cytotoxicity. Chemical Research in Toxicology, 24(7), 1094–1103. 10.1021/tx200156k [PubMed] [CrossRef] [Google Scholar]
  • Nardone, A. , Ferreccio, C. , Acevedo, J. , Enanoria, W. , Blair, A. , Smith, A. H. , Balmes, J. , & Steinmaus, C. (2017). The impact of BMI on non‐malignant respiratory symptoms and lung function in arsenic exposed adults of northern Chile. Environmental Research, 158, 710–719. [PMC free article] [PubMed] [Google Scholar]
  • Nath Barbhuiya, S. , Barhoi, D. , Giri, A. , & Giri, S. (2020). Arsenic and smokeless tobacco exposure induces DNA damage and oxidative stress in reproductive organs of female Swiss albino mice. Journal of Environmental Science and Health, Part C: Toxicology and Carcinogenesis, 38(4), 384–408. 10.1080/26896583.2020.1860400 [PubMed] [CrossRef] [Google Scholar]
  • Nava‐Rivera, L. E. , Betancourt‐Martínez, N. D. , Lozoya‐Martínez, R. , Carranza‐Rosales, P. , Guzmán‐Delgado, N. E. , Carranza‐Torres, I. E. , Delgado‐Aguirre, H. , Zambrano‐Ortíz, J. O. , & Morán‐Martínez, J. (2021). Transgenerational effects in DNA methylation, genotoxicity and reproductive phenotype by chronic arsenic exposure. Scientific Reports, 11(1), 1–16. [PMC free article] [PubMed] [Google Scholar]
  • Navas‐Acien, A. , Sharrett, A. R. , Silbergeld, E. K. , Schwartz, B. S. , Nachman, K. E. , Burke, T. A. , & Guallar, E. (2005). Arsenic exposure and cardiovascular disease: A systematic review of the epidemiologic evidence. American Journal of Epidemiology, 162(11), 1037–1049.vars. [PubMed] [Google Scholar]
  • Navasumrit, P. , Chaisatra, K. , Promvijit, J. , Parnlob, V. , Waraprasit, S. , Chompoobut, C. , Binh, T. T. , Hai, D. N. , Bao, N. D. , Hai, N. K. , Kim, K. W. , Samson, L. D. , Graziano, J. H. , Mahidol, C. , & Ruchirawat, M. (2019). Exposure to arsenic in utero is associated with various types of DNA damage and micronuclei in newborns: A birth cohort study. Environmental Health, 18, 51. 10.1186/s12940-019-0481-7 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nermell, B. , Lindberg, A. L. , Rahman, M. , Berglund, M. , Persson, L. A. , El Arifeen, S. , & Vahter, M. (2007). Urinary arsenic concentration adjustment factors and malnutrition. Environmental research, 106, 212–218. 10.1016/j.envres.2007.08.005 [PubMed] [CrossRef] [Google Scholar]
  • Nersesyan, A. , Fenech, M. , Bolognesi, C. , Mišík, M. , Setayesh, T. , Wultsch, G. , Bonassi, S. , Thomas, P. , & Knasmüller, S. (2016). Use of the lymphocyte cytokinesis‐block micronucleus assay in occupational biomonitoring of genome damage caused by in vivo exposure to chemical genotoxins: Past, present and future. Mutation Research, Reviews in Mutation Research, 770, 1–11. 10.1016/j.mrrev.2016.05.003 [PubMed] [CrossRef] [Google Scholar]
  • Newman, J. D. , Navas‐Acien, A. , Kuo, C. C. , Guallar, E. , Howard, B. V. , Fabsitz, R. R. , Devereux, R. B. , Umans, J. G. , Francesconi, K. A. , Goessler, W. , Best, L. T. , & Tellez‐Plaza, M. (2016). Peripheral arterial disease and its association with arsenic exposure and metabolism in the strong heart study. American Journal of Epidemiology, 184, 806–817. 10.1093/aje/kww002 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Niedzwiecki, M. M. , Liu, X. , Hall, M. N. , Thomas, T. , Slavkovich, V. , Ilievski, V. , Levy, D. , Alam, S. , Siddique, A. B. , Parvez, F. , Graziano, J. H. , & Gamble, M. V. (2015). Sex‐specific associations of arsenic exposure with global DNA methylation and hydroxymethylation in leukocytes: Results from two studies in Bangladesh. Cancer Epidemiology, Biomarkers & Prevention, 24, 1748–1757. 10.1158/1055-9965.Epi-15-0432 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nigra, A. E. , Moon, K. A. , Jones, M. R. , Sanchez, T. R. , & Navas‐Acien, A. (2021). Urinary arsenic and heart disease mortality in NHANES 2003–2014. Environmental Research, 200, 111387. 10.1016/j.envres.2021.111387 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nizam, S. , Kato, M. , Yatsuya, H. , Khalequzzaman, M. , Ohnuma, S. , Naito, H. , & Nakajima, T. (2013). Differences in urinary arsenic metabolites between diabetic and non‐diabetic subjects in Bangladesh. International Journal of Environmental Research and Public Health, 10, 1006–1019. 10.3390/ijerph10031006 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nohara, K. , Okamura, K. , Suzuki, T. , Murai, H. , Ito, T. , Shinjo, K. , Takumi, S. , Michikawa, T. , Kondo, Y. , & Hata, K. (2016). Augmenting effects of gestational arsenite exposure of C3H mice on the hepatic tumors of the F₂ male offspring via the F₁ male offspring. Journal of Applied Toxicology, 36(1), 105–112. 10.1002/jat.3149 [PubMed] [CrossRef] [Google Scholar]
  • Nohara, K. , Tateishi, Y. , Suzuki, T. , Okamura, K. , Murai, H. , Takumi, S. , Maekawa, F. , Nishimura, N. , Kobori, M. , & Ito, T. (2012). Late‐onset increases in oxidative stress and other tumorigenic activities and tumors with a ha‐ras mutation in the liver of adult male C3H mice gestationally exposed to arsenic. Toxicological Sciences, 129(2), 293–304. 10.1093/toxsci/kfs203 [PubMed] [CrossRef] [Google Scholar]
  • Nollen, M. , Ebert, F. , Moser, J. , Mullenders, L. H. , Hartwig, A. , & Schwerdtle, T. (2009). Impact of arsenic on nucleotide excision repair: XPC function, protein level, and gene expression. Molecular Nutrition & Food Research, 53(5), 572–582. 10.1002/mnfr.200800480 [PubMed] [CrossRef] [Google Scholar]
  • Nong, Q. , Zhang, Y. , Guallar, E. , & Zhong, Q. (2016). Arsenic exposure and predicted 10‐year atherosclerotic cardiovascular risk using the pooled cohort equations in U.S. hypertensive adults. International Journal of Environmental Research and Public Health, 13. 10.3390/ijerph13111093 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nozadi, S. S. , Li, L. , Luo, L. , MacKenzie, D. , Erdei, E. , Du, R. , Roman, C. W. , Hoover, J. , O'Donald, E. , Burnette, C. , & Lewis, J. (2021). Prenatal metal exposures and Infants' developmental outcomes in a Navajo population. International Journal of Environmental Research and Public Health, 19. 10.3390/ijerph19010425 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • NRC (National Research Council (US) Subcommittee to Update the 1999 Arsenic in Drinking Water Report) . (2001). Arsenic in drinking Water: 2001 Update. National Academies Press (US). https://www.ncbi.nlm.nih.gov/books/NBK223678/, 10.17226/10194 [PubMed] [CrossRef] [Google Scholar]
  • Nunes, F. L. S. , Lima, S. C. V. C. , Lyra, C. O. , Marchioni, D. M. , Pedrosa, L. F. C. , Barbosa Junior, F. , & Sena‐Evangelista, K. C. M. (2022). The impact of essential and toxic elements on cardiometabolic risk factors in adults and older people. Journal of Trace Elements in Medicine and Biology: Organ of the Society for Minerals and Trace Elements (GMS), 72, 126991. 10.1016/j.jtemb.2022.126991 [PubMed] [CrossRef] [Google Scholar]
  • Núñez, O. , Fernández‐Navarro, P. , Martín‐Méndez, I. , Bel‐Lan, A. , Locutura, J. F. , & López‐Abente, G. (2016). Arsenic and chromium topsoil levels and cancer mortality in Spain. Environmental Science and Pollution Research International, 17, 17664–17675. 10.1007/s11356-016-6806-y [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nyanza, E. C. , Bernier, F. P. , Martin, J. W. , Manyama, M. , Hatfield, J. , & Dewey, D. (2021). Effects of prenatal exposure and co‐exposure to metallic or metalloid elements on early infant neurodevelopmental outcomes in areas with small‐scale gold mining activities in northern Tanzania. Environment International, 149, 106104. 10.1016/j.envint.2020.106104 [PubMed] [CrossRef] [Google Scholar]
  • Nyanza, E. C. , Dewey, D. , Manyama, M. , Martin, J. W. , Hatfield, J. , & Bernier, F. P. (2020). Maternal exposure to arsenic and mercury and associated risk of adverse birth outcomes in small‐scale gold mining communities in northern Tanzania. Environment International, 137, 105450. 10.1016/j.envint.2019.105450 [PubMed] [CrossRef] [Google Scholar]
  • Obrenovich, M. E. , Shamberger, R. J. , & Lonsdale, D. (2011). Altered heavy metals and Transketolase found in autistic Spectrum disorder. Biological Trace Element Research, 144, 475–486. 10.1007/s12011-011-9146-2 [PubMed] [CrossRef] [Google Scholar]
  • O'Brien, K. M. , White, A. J. , Jackson, B. P. , Karagas, M. R. , Sandler, D. P. , & Weinberg, C. R. (2019). Toenail‐based metal concentrations and young‐onset breast cancer. American Journal of Epidemiology, 188(4), 34–43. 10.1093/aje/kwaa035 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • O'Bryant, S. E. , Edwards, M. , Menon, C. V. , Gong, G. , & Barber, R. (2011). Long‐term low‐level arsenic exposure is associated with poorer neuropsychological functioning: A project FRONTIER study. International Journal of Environmental Research and Public Health, 8(3), 861–874. [PMC free article] [PubMed] [Google Scholar]
  • Oguri, T. , Yoshinaga, J. , Toshima, H. , Mizumoto, Y. , Hatakeyama, S. , & Tokuoka, S. (2016). Urinary inorganic arsenic concentrations and semen quality of male partners of subfertile couples in Tokyo. Journal of Environmental Science and Health, Part A, 51(6), 463–466. [PubMed] [Google Scholar]
  • Okamura, K. , & Nohara, K. (2016). Long‐term arsenite exposure induces premature senescence in B cell lymphoma A20 cells. Archives of Toxicology, 90(4), 793–803. 10.1007/s00204-015-1500-2 [PubMed] [CrossRef] [Google Scholar]
  • Osmond, M. J. , Kunz, B. A. , & Snow, E. T. (2010). Age and exposure to arsenic alter base excision repair transcript levels in mice. Mutagenesis, 25(5), 517–522. 10.1093/mutage/geq037 [PubMed] [CrossRef] [Google Scholar]
  • Osorio‐Yáñez, C. , Sanchez‐Guerra, M. , Solano, M. , Baccarelli, A. , Wright, R. , Sanders, A. P. , Tellez‐Rojo, M. M. , & Tamayo‐Ortiz, M. (2021). Metal exposure and bone remodeling during pregnancy: Results from the PROGRESS cohort study. Environmental Pollution, 282, 116962. 10.1016/j.envpol.2021.116962 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ourshalimian, S. , Naser, A. M. , Rahman, M. , Doza, S. , Stowell, J. , Narayan, K. M. V. , Shamsudduha, M. , & Gribble, M. O. (2019). Arsenic and fasting blood glucose in the context of other drinking water chemicals: A cross‐sectional study in Bangladesh. Environmental Research, 172, 249–257. 10.1016/j.envres.2018.12.049 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ovayolu, A. , Ovayolu, G. , Karaman, E. , Yuce, T. , Ozek, M. A. , & Turksoy, V. A. (2020). Amniotic fluid levels of selected trace elements and heavy metals in pregnancies complicated with neural tube defects. Congenital Anomalies, 60(5), 136–141. [PubMed] [Google Scholar]
  • Özel, Ş. , Ozyer, S. , Aykut, O. , Çinar, M. , Yılmaz, O. H. , Caglar, A. , & Engin‐Ustun, Y. (2019). Maternal second trimester blood levels of selected heavy metals in pregnancies complicated with neural tube defects. The Journal of Maternal‐Fetal & Neonatal Medicine, 32, 2547–2553. 10.1080/14767058.2018.1441280 [PubMed] [CrossRef] [Google Scholar]
  • Pace, C. , Smith‐Gagen, J. , & Angermann, J. (2018). Arsenic methylation capacity and metabolic syndrome in the 2013–2014 U.S. National Health and nutrition examination survey (NHANES). International Journal of Environmental Research and Public Health, 15. 10.3390/ijerph15010168 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Pal, P. , & Halder, A. (2019). Is there any role of arsenic toxicity in HPV related Oral squamous cell carcinoma? Biological Trace Element Research, 188(2), 274–283. 10.1007/s12011-018-1419-6 [PubMed] [CrossRef] [Google Scholar]
  • Pal, P. , Raychowdhury, R. , Dolai, T. K. , Roy, S. , Dastidar, R. , & Halder, A. (2017). Study of arsenic exposure in oral/oropharyngeal carcinoma in West Bengal. International Journal of Occupational Medicine and Environmental Health, 30(2), 271–279. 10.13075/ijomeh.1896.00806 [PubMed] [CrossRef] [Google Scholar]
  • Pan, W. C. , Kile, M. L. , Seow, W. J. , Lin, X. , Quamruzzaman, Q. , Rahman, M. , Mahiuddin, G. , Mostofa, G. , Lu, Q. , & Christiani, D. C. (2013). Genetic susceptible locus in NOTCH2 interacts with arsenic in drinking water on risk of type 2 diabetes. PLoS One, 8, e70792. 10.1371/journal.pone.0070792 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Pan, W. C. , Seow, W. J. , Kile, M. L. , Hoffman, E. B. , Quamruzzaman, Q. , Rahman, M. , Mahiuddin, G. , Mostofa, G. , Lu, Q. , & Christiani, D. C. (2013). Association of low to moderate levels of arsenic exposure with risk of type 2 diabetes in Bangladesh. American Journal of Epidemiology, 178, 1563–1570. 10.1093/aje/kwt195 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Parajuli, R. P. , Fujiwara, T. , Umezaki, M. , Furusawa, H. , & Watanabe, C. (2014). Home environment and prenatal exposure to lead, arsenic and zinc on the neurodevelopment of six‐month‐old infants living in Chitwan Valley, Nepal. Neurotoxicology and Teratology, 41, 89–95. 10.1016/j.ntt.2013.12.006 [PubMed] [CrossRef] [Google Scholar]
  • Parajuli, R. P. , Fujiwara, T. , Umezaki, M. , & Watanabe, C. (2013). Association of cord blood levels of lead, arsenic, and zinc with neurodevelopmental indicators in newborns: A birth cohort study in Chitwan Valley. Nepal. Environ Res, 121, 45–51. 10.1016/j.envres.2012.10.010 [PubMed] [CrossRef] [Google Scholar]
  • Parajuli, R. P. , Umezaki, M. , Fujiwara, T. , & Watanabe, C. (2015). Association of cord blood levels of lead, arsenic, and zinc and home environment with children neurodevelopment at 36 months living in Chitwan Valley, Nepal. PLoS One, 10, e0120992. 10.1371/journal.pone.0120992 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Park, S. K. , Peng, Q. , Bielak, L. F. , Silver, K. D. , Peyser, P. A. , & Mitchell, B. D. (2016). Arsenic exposure is associated with diminished insulin sensitivity in non‐diabetic Amish adults. Diabetes/Metabolism Research and Reviews, 32, 565–571. 10.1002/dmrr.2769 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Parvez, F. , Chen, Y. , Brandt‐Rauf, P. W. , Slavkovich, V. , Islam, T. , Ahmed, A. , Argos, M. , Hassan, R. , Yunus, M. , Haque, S. E. , & Balac, O. (2010). A prospective study of respiratory symptoms associated with chronic arsenic exposure in Bangladesh: Findings from the health effects of arsenic longitudinal study (HEALS). Thorax, 65(6), 528–533. [PMC free article] [PubMed] [Google Scholar]
  • Parvez, F. , Chen, Y. , Yunus, M. , Olopade, C. , Segers, S. , Slavkovich, V. , Argos, M. , Hasan, R. , Ahmed, A. , Islam, T. , Akter, M. M. , Graziano, J. H. , & Ahsan, H. (2013). Arsenic exposure and impaired lung function. Findings from a large population‐based prospective cohort study. American Journal of Respiratory and Critical Care Medicine, 188, 813–819. 10.1164/rccm.201212-2282OC [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Parvez, F. , Wasserman, G. A. , Factor‐Litvak, P. , Liu, X. , Slavkovich, V. , Siddique, A. B. , Sultana, R. , Sultana, R. , Islam, T. , Levy, D. , Mey, J. L. , van Geen, A. , Khan, K. , Kline, J. , Ahsan, H. , & Graziano, J. H. (2011). Arsenic exposure and motor function among children in Bangladesh. Environmental Health Perspectives, 119(11), 1665–1670. 10.1289/ehp.1103548 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Paul, S. , Banerjee, N. , Chatterjee, A. , Sau, T. J. , Das, J. K. , Mishra, P. K. , Chakrabarti, P. , Bandyopadhyay, A. , & Giri, A. K. (2014). Arsenic‐induced promoter hypomethylation and over‐expression of ERCC2 reduces DNA repair capacity in humans by non‐disjunction of the ERCC2–Cdk7 complex. Metallomics, 6(4), 864–873. [PubMed] [Google Scholar]
  • Paul, S. , Bhattacharjee, P. , Mishra, P. K. , Chatterjee, D. , Biswas, A. , Deb, D. , Ghosh, A. , Mazumder, G. , & Giri, A. K. (2013). Human urothelial micronucleus assay to assess genotoxic recovery by reduction of arsenic in drinking water: A cohort study in West Bengal, India. Biometals, 26, 855–862. 10.1007/s10534-013-9652-0 [PubMed] [CrossRef] [Google Scholar]
  • Paul, S. , Chakraborty, T. , Halder, A. , Bandopadhyay, D. , Chaudhuri, U. , & De, M. (2011). Association of genotoxic effects of arsenic with haematological malignancy in West Bengal. Human & Experimental Toxicology, 30, 165–170. 10.1177/0960327110368694 [PubMed] [CrossRef] [Google Scholar]
  • Paul, S. , Das, N. , Bhattacharjee, P. , Banerjee, M. , Das, J. K. , Sarma, N. , Sarkar, A. , Bandyopadhyay, A. K. , Sau, T. J. , Basu, S. , Banerjee, S. , Majumder, P. , & Giri, A. K. (2013). Arsenic‐induced toxicity and carcinogenicity: A two‐wave cross‐sectional study in arsenicosis individuals in West Bengal, India. Journal of Exposure Science & Environmental Epidemiology, 23, 156–162. 10.1038/jes.2012.91 [PubMed] [CrossRef] [Google Scholar]
  • Paul, S. K. , Islam, M. S. , Hasibuzzaman, M. M. , Hossain, F. , Anjum, A. , Saud, Z. A. , Haque, M. M. , Sultana, P. , Haque, A. , Andric, K. B. , Rahman, A. , Karim, M. R. , Siddique, A. E. , Karim, Y. , Rahman, M. , Miyataka, H. , Xin, L. , Himeno, S. , & Hossain, K. (2019). Higher risk of hyperglycemia with greater susceptibility in females in chronic arsenic‐exposed individuals in Bangladesh. Science of the Total Environment, 668, 1004–1012. 10.1016/j.scitotenv.2019.03.029 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Pei, Q. , Ma, N. , Zhang, J. , Xu, W. , Li, Y. , Ma, Z. , Li, Y. , Tian, F. , Zhang, W. , Mu, J. , Li, Y. , Wang, D. , Liu, H. , Yang, M. , Ma, C. , & Yun, F. (2013). Oxidative DNA damage of peripheral blood polymorphonuclear leukocytes, selectively induced by chronic arsenic exposure, is associated with extent of arsenic‐related skin lesions. Toxicology and Applied Pharmacology, 266, 143–149. 10.1016/j.taap.2012.10.031 [PubMed] [CrossRef] [Google Scholar]
  • Peng, S. , Liu, L. , Zhang, X. , Heinrich, J. , Zhang, J. , Schramm, K. W. , Huang, Q. , Tian, M. , Eqani, S. A. , & Shen, H. (2015). A nested case‐control study indicating heavy metal residues in meconium associate with maternal gestational diabetes mellitus risk. Environmental Health, 14, 19. 10.1186/s12940-015-0004-0 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Pérez‐Vázquez, M. S. , Ochoa‐Martínez, Á. C. , RuÍz‐Vera, T. , Araiza‐Gamboa, Y. , & Pérez‐Maldonado, I. N. (2017). Evaluation of epigenetic alterations (mir‐126 and mir‐155 expression levels) in Mexican children exposed to inorganic arsenic via drinking water. Environmental Science and Pollution Research International, 24, 28036–28045. 10.1007/s11356-017-0367-6 [PubMed] [CrossRef] [Google Scholar]
  • Pesola, G. R. , Parvez, F. , Chen, Y. , Ahmed, A. , Hasan, R. , & Ahsan, H. (2012). Arsenic exposure from drinking water and dyspnoea risk in Araihazar, Bangladesh: A population‐based study. European Respiratory Journal, 39(5), 1076–1083. [PMC free article] [PubMed] [Google Scholar]
  • Peters, B. A. , Hall, M. N. , Liu, X. , Neugut, Y. D. , Pilsner, J. R. , Levy, D. , Ilievski, V. , Slavkovich, V. , Islam, T. , Factor‐Litvak, P. , Graziano, J. H. , & Gamble, M. V. (2014). Creatinine, arsenic metabolism, and renal function in an arsenic‐exposed population in Bangladesh. PLoS One, 9, e113760. 10.1371/journal.pone.0113760 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Peters, B. A. , Hall, M. N. , Liu, X. , Slavkovich, V. , Ilievski, V. , Alam, S. , Siddique, A. B. , Islam, T. , Graziano, J. H. , & Gamble, M. V. (2015). Renal function is associated with indicators of arsenic methylation capacity in Bangladeshi adults. Environmental Research, 143, 123–130. 10.1016/j.envres.2015.10.001 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Peters, B. A. , Liu, X. , Hall, M. N. , Ilievski, V. , Slavkovich, V. , Siddique, A. B. , Alam, S. , Islam, T. , Graziano, J. H. , & Gamble, M. V. (2015). Arsenic exposure, inflammation, and renal function in Bangladeshi adults: Effect modification by plasma glutathione redox potential. Free Radical Biology & Medicine, 85, 174–182. 10.1016/j.freeradbiomed.2015.04.020 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Peters, S. , Broberg, K. , Gallo, V. , Levi, M. , Kippler, M. , Vineis, P. , Veldink, J. , van den Berg, L. , Middleton, L. , Travis, R. C. , Bergmann, M. M. , Palli, D. , Grioni, S. , Tumino, R. , Elbaz, A. , Vlaar, T. , Mancini, F. , Kühn, T. , Katzke, V. , … Vermeulen, R. (2021). Blood metal levels and amyotrophic lateral sclerosis risk: A prospective cohort. Annals of Neurology, 89, 125–133. 10.1002/ana.25932 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Petursdottir, A. H. , Sloth, J. J. , & Feldmann, J. (2015). Introduction of regulations for arsenic in feed and food with emphasis on inorganic arsenic, and implications for analytical chemistry. Analytical and Bioanalytical Chemistry, 407(28), 8385–8396. 10.1007/s00216-015-9019-1 [PubMed] [CrossRef] [Google Scholar]
  • Phookphan, P. , Navasumrit, P. , Waraprasit, S. , Promvijit, J. , Chaisatra, K. , Ngaotepprutaram, T. , & Ruchirawat, M. (2017). Hypomethylation of inflammatory genes (COX2, EGR1, and SOCS3) and increased urinary 8‐nitroguanine in arsenic‐exposed newborns and children. Toxicology and Applied Pharmacology, 316, 36–47. 10.1016/j.taap.2016.12.015 [PubMed] [CrossRef] [Google Scholar]
  • Pi, X. , Wang, C. , Yin, S. , Jin, L. , Li, Z. , Wang, L. , Liu, J. , Zhang, Y. , & Ren, A. (2023). Arsenic exposure, periconceptional folic acid supplementation, and the risk for neural tube defects: A case–control study. Exposure and Health, 15, 245–254. 10.1007/s12403-022-00487-z [CrossRef] [Google Scholar]
  • Pichler, G. , Grau‐Perez, M. , Tellez‐Plaza, M. , Umans, J. , Best, L. , Cole, S. , Goessler, W. , Francesconi, K. , Newman, J. , Redon, J. , Devereux, R. , & Navas‐Acien, A. (2019). Association of arsenic exposure with cardiac geometry and left ventricular function in young adults. Circulation. Cardiovascular Imaging, 12, e009018. 10.1161/circimaging.119.009018 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Pierce, B. L. , Argos, M. , Chen, Y. , Melkonian, S. , Parvez, F. , Islam, T. , Ahmed, A. , Hasan, R. , Rathouz, P. J. , & Ahsan, H. (2011). Arsenic exposure, dietary patterns, and skin lesion risk in Bangladesh: A prospective study. American Journal of Epidemiology, 173(3), 345–354. 10.1093/aje/kwq366 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Pierce, B. L. , Kibriya, M. G. , Tong, L. , Jasmine, F. , Argos, M. , Roy, S. , Paul‐Brutus, R. , Rahaman, R. , Rakibuz‐Zaman, M. , Parvez, F. , Ahmed, A. , Quasem, I. , Hore, S. K. , Alam, S. , Islam, T. , Slavkovich, V. , Gamble, M. V. , Yunus, M. , Rahman, M. , … Ahsan, H. (2012). Genome‐wide association study identifies chromosome 10q24.32 variants associated with arsenic metabolism and toxicity phenotypes in Bangladesh. PLoS Genetics, 8(2), e1002522. 10.1371/journal.pgen.1002522 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Pilsner, J. R. , Hall, M. N. , Liu, X. , Ilievski, V. , Slavkovich, V. , Levy, D. , Factor‐Litvak, P. , Yunus, M. , Rahman, M. , Graziano, J. H. , & Gamble, M. V. (2012). Influence of prenatal arsenic exposure and newborn sex on global methylation of cord blood DNA. PLoS One, 7(5), e37147. [PMC free article] [PubMed] [Google Scholar]
  • Pilsner, J. R. , Liu, X. , Ahsan, H. , Ilievski, V. , Slavkovich, V. , Levy, D. , Factor‐Litvak, P. , Graziano, J. H. , & Gamble, M. V. (2009). Folate deficiency, hyperhomocysteinemia, low urinary creatinine, and hypomethylation of leukocyte DNA are risk factors for arsenic‐induced skin lesions. Environmental Health Perspectives, 117(2), 254–260. 10.1289/ehp.11872 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Pouillot, R. , Farakos, S. S. , Spungen, J. , Schaefer, H. R. , Flannery, B. M. , & Van Doren, J. M. (2022). Cadmium physiologically based pharmacokinetic (PBPK) models for forward and reverse dosimetry: Review, evaluation, and adaptation to the U.S. population. Toxicology Letters, 367, 67–75. 10.1016/j.toxlet.2022.07.812 [PubMed] [CrossRef] [Google Scholar]
  • Powers, M. , Sanchez, T. R. , Grau‐Perez, M. , Yeh, F. , Francesconi, K. A. , Goessler, W. , George, C. M. , Heaney, C. , Best, L. G. , Umans, J. G. , Brown, R. H. , & Navas‐Acien, A. (2019). Low‐moderate arsenic exposure and respiratory in American Indian communities in the strong heart study. Environmental Health, 18, 104. 10.1186/s12940-019-0539-6 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Prasad, P. , Sarkar, N. , & Sinha, D. (2020). Effect of low‐ and high‐level groundwater arsenic on peripheral blood and lung function of exposed rural women. Regulatory Toxicology and Pharmacology, 115, 104684. 10.1016/j.yrtph.2020.104684 [PubMed] [CrossRef] [Google Scholar]
  • Putila, J. J. , & Guo, N. L. (2011). Association of arsenic exposure with lung cancer incidence rates in the United States. PLoS One, 6, e25886. 10.1371/journal.pone.0025886 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Qu, Y. , Lv, Y. , Ji, S. , Ding, L. , Zhao, F. , Zhu, Y. , Zhang, W. , Hu, X. , Lu, Y. , Li, Y. , Zhang, X. , Zhang, M. , Yang, Y. , Li, C. , Zhang, M. , Li, Z. , Chen, C. , Zheng, L. , Gu, H. , … Shi, X. (2022). Effect of exposures to mixtures of lead and various metals on hypertension, pre‐hypertension, and blood pressure: A cross‐sectional study from the China National Human Biomonitoring. Environmental Pollution, 299, 118864. 10.1016/j.envpol.2022.118864 [PubMed] [CrossRef] [Google Scholar]
  • Rager, J. E. , Bailey, K. A. , Smeester, L. , Miller, S. K. , Parker, J. S. , Laine, J. E. , Drobná, Z. , Currier, J. , Douillet, C. , Olshan, A. F. , Rubio‐Andrade, M. , Stýblo, M. , García‐Vargas, G. , & Fry, R. C. (2014). Prenatal arsenic exposure and the epigenome: Altered microRNAs associated with innate and adaptive immune signaling in newborn cord blood. Environmental and Molecular Mutagenesis, 55(3), 196–208. [PMC free article] [PubMed] [Google Scholar]
  • Rahbar, M. H. , Ibrahim, S. H. , Azam, S. I. , Hessabi, M. , Karim, F. , Kim, S. , Zhang, J. , Gulzar Ali, N. , & Loveland, K. A. (2021). Concentrations of Lead, mercury, arsenic, cadmium, manganese, and aluminum in the Blood of Pakistani children with and without autism Spectrum disorder and their associated factors. International Journal of Environmental Research and Public Health, 18. 10.3390/ijerph18168625 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rahbar, M. H. , Samms‐Vaughan, M. , Ardjomand‐Hessabi, M. , Loveland, K. A. , Dickerson, A. S. , Chen, Z. , Bressler, J. , Shakespeare‐Pellington, S. , Grove, M. L. , Bloom, K. , Wirth, J. , Pearson, D. A. , & Boerwinkle, E. (2012). The role of drinking water sources, consumption of vegetables and seafood in relation to blood arsenic concentrations of Jamaican children with and without autism Spectrum disorders. Science of the Total Environment, 433, 362–370. 10.1016/j.scitotenv.2012.06.085 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rahman, A. , Islam, M. S. , Tony, S. R. , Siddique, A. E. , Mondal, V. , Hosen, Z. , Islam, Z. , Hossain, M. I. , Rahman, M. , Anjum, A. , Paul, S. K. , Hossen, F. , Sarker, M. K. , Hossain, S. , Salam, K. A. , Haque, A. , Hoque, M. A. , Saud, Z. A. , Xin, L. , … Hossain, K. (2021). T helper 2‐driven immune dysfunction in chronic arsenic‐exposed individuals and its link to the features of allergic asthma. Toxicology and Applied Pharmacology, 420, 115532. 10.1016/j.taap.2021.115532 [PubMed] [CrossRef] [Google Scholar]
  • Rahman, A. , Kippler, M. , Pervin, J. , Tarafder, C. , Lucy, I. J. , Svefors, P. , El Arifeen, S. , & Persson, L. A. (2021). A cohort study of the association between prenatal arsenic exposure and age at menarche in a rural area, Bangladesh. Environment International, 154. 10.1016/j.envint.2021.106562 [PubMed] [CrossRef] [Google Scholar]
  • Rahman, A. , Persson, L. , Nermell, B. , El Arifeen, S. , Ekström, E. C. , Smith, A. H. , & Vahter, M. (2010). Arsenic exposure and risk of spontaneous abortion, stillbirth, and infant mortality. Epidemiology, 21, 797–804. 10.1097/EDE.0b013e3181f56a0d [PubMed] [CrossRef] [Google Scholar]
  • Rahman, A. , Vahter, M. , Ekstrom, E. C. , Rahman, M. , Mustafa, A. M. G. , Wahed, M. A. , Yunus, M. , & Persson, L. A. (2007). Association of arsenic exposure during pregnancy with foetal loss and infant death: A cohort study in Bangladesh. American Journal of Epidemiology, 165(12), 1389–1396. [PubMed] [Google Scholar]
  • Rahman, A. , Vahter, M. , Smith, A. H. , Nermell, B. , Yunus, M. , El Arifeen, S. , Persson, L. A. , & Ekstrom, E. C. (2009). Arsenic exposure during pregnancy and size at birth: A prospective cohort study in Bangladesh. American Journal of Epidemiology, 169(3), 304–312. [PubMed] [Google Scholar]
  • Rahman, H. H. , Niemann, D. , & Munson‐McGee, S. H. (2022a). Urinary metals, arsenic, and polycyclic aromatic hydrocarbon exposure and risk of chronic bronchitis in the US adult population. Environmental Science and Pollution Research, 27, 1–2. [PubMed] [Google Scholar]
  • Rahman, H. H. , Niemann, D. , & Munson‐McGee, S. H. (2022c). Environmental exposure to metals and the risk of high blood pressure: A cross‐sectional study from NHANES 2015–2016. Environmental Science and Pollution Research, 29, 531–542. 10.1007/s11356-021-15726-0 [PubMed] [CrossRef] [Google Scholar]
  • Rahman, H. H. , Niemann, D. , & Yusuf, K. K. (2022). Association of urinary arsenic and sleep disorder in the US population: NHANES 2015–2016. Environmental Science and Pollution Research International, 29(4), 5496–5504. [PubMed] [Google Scholar]
  • Rahman, M. , Sohel, N. , Hore, S. K. , Yunus, M. , Bhuiya, A. , & Streatfield, P. K. (2015). Prenatal arsenic exposure and drowning among children in Bangladesh. Global Health Action, 8, 28702. 10.3402/gha.v8.28702 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rahman, M. , Sohel, N. , Yunus, F. M. , Alam, N. , Nahar, Q. , Streatfield, P. K. , & Yunus, M. (2019). Arsenic exposure and young adult's mortality risk: A 13‐year follow‐up study in Matlab, Bangladesh. Environment International, 123, 358–367. 10.1016/j.envint.2018.12.006 [PubMed] [CrossRef] [Google Scholar]
  • Rahman, M. , Sohel, N. , Yunus, M. , Chowdhury, M. E. , Hore, S. K. , Zaman, K. , Bhuiya, A. , & Streatfield, P. K. (2013). Increased childhood mortality and arsenic in drinking water in Matlab, Bangladesh: A population‐based cohort study. PLoS One, 8, e55014. 10.1371/journal.pone.0055014 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rahman, M. , Sohel, N. , Yunus, M. , Chowdhury, M. E. , Hore, S. K. , Zaman, K. , Bhuiya, A. , & Streatfield, P. K. (2014). A prospective cohort study of stroke mortality and arsenic in drinking water in Bangladeshi adults. BMC Public Health, 14, 174. 10.1186/1471-2458-14-174 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rahman, M. , Tondel, M. , Ahmad, S. A. , Chowdhury, I. A. , Faruquee, M. H. , & Axelson, O. (1999). Hypertension and arsenic exposure in Bangladesh. Hypertension, 33, 74–78. [PubMed] [Google Scholar]
  • Rahman, M. , Vahter, M. , Sohel, N. , Yunus, M. , Wahed, M. A. , Streatfield, P. K. , Ekström, E. C. , & Persson, L. A. (2006). Arsenic exposure and age and sex‐specific risk for skin lesions: A population‐based case‐referent study in Bangladesh. Environmental Health Perspectives, 114(12), 1847–1852. 10.1289/ehp.9207 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rahman, M. L. , Oken, E. , Hivert, M.‐F. , Rifas‐Shiman, S. , Lin, P.‐I. D. , Colicino, E. , Wright, R. O. , Amarasiriwardena, C. , Henn, B. G. C. , Gold, D. R. , Coull, B. A. , & Cardenas, A. (2021). Early pregnancy exposure to metal mixture and birth outcomes‐a prospective study in project viva. Environment International, 156, 106714. 10.1016/j.envint.2021.106714 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rahman, M. L. , Valeri, L. , Kile, M. L. , Mazumdar, M. , Mostofa, G. , Qamruzzaman, Q. , Rahman, M. , Baccarelli, A. , Liang, L. , Hauser, R. , & Christiani, D. C. (2017). Investigating causal relation between prenatal arsenic exposure and birthweight: Are smaller infants more susceptible? Environment International, 108, 32–40. 10.1016/j.envint.2017.07.026 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Raml, R. , Rumpler, A. , Goessler, W. , Vahter, M. , Li, L. , Ochi, T. , & Francesconi, K. A. (2007). Thio‐dimethylarsinate is a common metabolite in urine samples from arsenic‐exposed women in Bangladesh. Toxicology and Applied Pharmacology, 222(3), 374–380. 10.1016/j.taap.2006.12.014 [PubMed] [CrossRef] [Google Scholar]
  • Ran, S. , Liu, J. , & Li, S. (2020). A systematic review of the various effect of arsenic on glutathione synthesis in vitro and in vivo . BioMed Research International, 2020, 9414196. 10.1155/2020/9414196 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rehman, K. , Fatima, F. , & Akash, M. S. H. (2019). Biochemical investigation of association of arsenic exposure with risk factors of diabetes mellitus in Pakistani population and its validation in animal model. Environmental Monitoring and Assessment, 191, 511. 10.1007/s10661-019-7670-2 [PubMed] [CrossRef] [Google Scholar]
  • Rehman, K. , Fu, Y. J. , Zhang, Y. F. , Wang, Q. Q. , Wu, B. , Wu, Y. , Zhou, X. Y. , Sun, W. H. , Sun, T. F. , & Naranmandura, H. (2014). Trivalent methylated arsenic metabolites induce apoptosis in human myeloid leukemic HL‐60 cells through generation of reactive oxygen species. Metallomics, 6(8), 1502–1512. 10.1039/c4mt00119b [PubMed] [CrossRef] [Google Scholar]
  • Rehman, K. , & Naranmandura, H. (2012). Arsenic metabolism and thioarsenicals. Metallomics, 4(9), 881–892. [PubMed] [Google Scholar]
  • Reichard, J. F. , & Puga, A. (2010). Effects of arsenic exposure on DNA methylation and epigenetic gene regulation. Epigenomics, 2(1), 87–104. [PMC free article] [PubMed] [Google Scholar]
  • Remy, S. , Govarts, E. , Bruckers, L. , Paulussen, M. , Wens, B. , Hond, E. D. , Nelen, V. , Baeyens, W. , van Larebeke, N. , Loots, I. , Sioen, I. , & Schoeters, G. (2014). Expression of the sFLT1 gene in cord blood cells is associated to maternal arsenic exposure and decreased birth weight. PLoS One, 9, e92677. 10.1371/journall.pone.0092677 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Renzetti, S. , Cagna, G. , Calza, S. , Conversano, M. , Fedrighi, C. , Forte, G. , Giorgino, A. , Guazzetti, S. , Majorani, C. , Oppini, M. , Peli, M. , Petrucci, F. , Pino, A. , Placidi, D. , Senofonte, O. , Zoni, S. , Alimonti, A. , & Lucchini, R. G. (2021). The effects of the exposure to neurotoxic elements on Italian schoolchildren behavior. Scientific Reports, 11, 9898. 10.1038/s41598-021-88969-z [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rezaei, M. , Rezaei, A. , Esmaeili, A. , Nakhaee, S. , Azadi, N. A. , & Mansouri, B. (2022). A case‐control study on the relationship between urine trace element levels and autism spectrum disorder among Iranian children. Environmental Science and Pollution Research., 29, 57287–57295. 10.1007/s11356-022-19933-1 [PubMed] [CrossRef] [Google Scholar]
  • Rhee, S. Y. , Hwang, Y. C. , Woo, J. T. , Chin, S. O. , Chon, S. , & Kim, Y. S. (2013). Arsenic exposure and prevalence of diabetes mellitus in Korean adults. Journal of Korean Medical Science, 28, 861–868. 10.3346/jkms.2013.28.6.861 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Richter, F. , Kloster, S. , Wodschow, K. , Hansen, B. , Schullehner, J. , Kristiansen, S. M. , Petersen, M. M. , Strandberg‐Larsen, K. , & Ersbøll, A. K. (2022). Maternal exposure to arsenic in drinking water and risk of congenital heart disease in the offspring. Environment International, 160, 107051. 10.1016/j.envint.2021.107051 [PubMed] [CrossRef] [Google Scholar]
  • Robles‐Osorio, M. L. , Sabath‐Silva, E. , & Sabath, E. (2015). Arsenic‐mediated nephrotoxicity. Renal Failure, 37, 542–547. 10.3109/0886022X.2015.1013419 [PubMed] [CrossRef] [Google Scholar]
  • Rocha‐Amador, D. , Navarro, M. E. , Carrizales, L. , Morales, R. , & Calderón, J. (2007). Decreased intelligence in children and exposure to fluoride and arsenic in drinking water. Cadernos de Saúde Pública, 23(Suppl 4), S579–S587. 10.1590/s0102-311x2007001600018 [PubMed] [CrossRef] [Google Scholar]
  • Rodrigues, E. G. , Bellinger, D. C. , Valeri, L. , Hasan, M. O. , Quamruzzaman, Q. , Golam, M. , Kile, M. L. , Christiani, D. C. , Wright, R. O. , & Mazumdar, M. (2016). Neurodevelopmental outcomes among 2‐ to 3‐year‐old children in Bangladesh with elevated blood lead and exposure to arsenic and manganese in drinking water. Environmental Health, 15, 44. 10.1186/s12940-016-0127-y [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rodrigues, E. G. , Kile, M. , Dobson, C. , Amarasiriwardena, C. , Quamruzzaman, Q. , Rahman, M. , Golam, M. , & Christiani, D. C. (2015). Maternal‐infant biomarkers of prenatal exposure to arsenic and manganese. Journal of Exposure Science & Environmental Epidemiology, 25(6), 639–648. [PMC free article] [PubMed] [Google Scholar]
  • Rodríguez‐Barranco, M. , Gil, F. , Hernández, A. F. , Alguacil, J. , Lorca, A. , Mendoza, R. , Gómez, I. , Molina‐Villalba, I. , González‐Alzaga, B. , Aguilar‐Garduño, C. , Rohlman, D. S. , & Lacasaña, M. (2016). Postnatal arsenic exposure and attention impairment in school children. Cortex, 74, 370–382. 10.1016/j.cortex.2014.12.018 [PubMed] [CrossRef] [Google Scholar]
  • Rojas, D. , Rager, J. E. , Smeester, L. , Bailey, K. A. , Drobná, Z. , Rubio‐Andrade, M. , Stýblo, M. , García‐Vargas, G. , & Fry, R. C. (2015). Prenatal arsenic exposure and the epigenome: Identifying sites of 5‐methylcytosine alterations that predict functional changes in gene expression in newborn cord blood and subsequent birth outcomes. Toxicological Sciences, 143, 97–106. 10.1093/toxsci/kfu210 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Röllin, H. B. , Channa, K. , Olutola, B. G. , & Odland, J. (2017). Evaluation of in utero exposure to arsenic in South Africa. Science of the Total Environment, 575, 338–346. 10.1016/j.scitotenv.2016.10.044 [PubMed] [CrossRef] [Google Scholar]
  • Román, M. D. , Niclis, C. , Aballay, L. R. , Lantieri, M. J. , Díaz, M. D. P. , & Muñoz, S. E. (2018). Do exposure to arsenic, occupation and diet have synergistic effects on prostate cancer risk? Asian Pacific Journal of Cancer Prevention, 19(6), 1495–1501. 10.22034/APJCP.2018.19.6.1495 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rosado, J. L. , Ronquillo, D. , Kordas, K. , Rojas, O. , Alatorre, J. , Lopez, P. , Garcia‐Vargas, G. , Del Carmen, C. M. , Cebrián, M. E. , & Stoltzfus, R. J. (2007). Arsenic exposure and cognitive performance in Mexican schoolchildren. Environmental Health Perspectives, 115(9), 1371–1375. 10.1289/ehp.9961 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Rothman, K. J. , Greenland, S. , & Lash, T. L. (2008). Modern epidemiology. Wolters Kluwer Health/Lippincott Williams & Wilkins Philadelphia. [Google Scholar]
  • Roy, A. , Kordas, K. , Lopez, P. , Rosado, J. L. , Cebrian, M. E. , Vargas, G. G. , Ronquillo, D. , & Stoltzfus, R. J. (2011). Association between arsenic exposure and behavior among first‐graders from Torreón, Mexico. Environmental Research, 111(5), 670–676. 10.1016/j.envres.2011.03.003 [PubMed] [CrossRef] [Google Scholar]
  • Roy, N. K. , Murphy, A. , & Costa, M. (2020). Arsenic Methyltransferase and methylation of inorganic arsenic. Biomolecules, 10(9), 1351. 10.3390/biom10091351 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Roy, P. , Mukherjee, A. , & Giri, S. (2016). Evaluation of genetic damage in tobacco and arsenic exposed population of southern Assam, India using buccal cytome assay and comet assay. Ecotoxicology and Environmental Safety, 124, 169–176. 10.1016/j.ecoenv.2015.10.019 [PubMed] [CrossRef] [Google Scholar]
  • Ruan, F. , Zhang, J. , Liu, J. , Sun, X. , Li, Y. , Xu, S. , & Xia, W. (2022). Association between prenatal exposure to metal mixtures and early childhood allergic diseases. Environmental Research, 206, 112615. 10.1016/j.envres.2021.112615 [PubMed] [CrossRef] [Google Scholar]
  • Rudnai, T. , Sandor, J. , Kadar, M. , Borsanyi, M. , Beres, J. , Metneki, J. , Maraczi, G. , & Rudnai, P. (2014). Arsenic in drinking water and congenital heart anomalies in Hungary. International Journal of Hygiene and Environmental Health, 217, 813–818. 10.1016/j.ijheh.2014.05.002 [PubMed] [CrossRef] [Google Scholar]
  • Ruiz‐Navarro, M. L. , Navarro‐Alarcón, M. , González‐de, L. , la Serrana, H. , Pérez‐Valero, V. , & López‐Martinez, M. C. (1998). Urine arsenic concentrations in healthy adults as indicators of environmental contamination: Relation with some pathologies. Sci Total Environ, 216, 55–61. [PubMed] [Google Scholar]
  • Saha, K. K. , Engström, A. , Hamadani, J. D. , Tofail, F. , Rasmussen, K. M. , & Vahter, M. (2012). Pre‐ and postnatal arsenic exposure and body size to 2 years of age: A cohort study in rural Bangladesh. Environmental Health Perspectives, 120, 1208–1214. 10.1289/ehp.1003378 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Saint‐Jacques, N. , Brown, P. , Nauta, L. , Boxall, J. , Parker, L. , & Dummer, T. J. B. (2018). Estimating the risk of bladder and kidney cancer from exposure to low‐levels of arsenic in drinking water, Nova Scotia, Canada. Environment International, 110, 95–104. 10.1016/j.envint.2017.10.014 [PubMed] [CrossRef] [Google Scholar]
  • Sallsten, G. , & Barregard, L. (2021). Variability of urinary creatinine in healthy individuals. International Journal of Environmental Research and Public Health, 18. 10.3390/ijerph18063166 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sampayo‐Reyes, A. , Hernández, A. , El‐Yamani, N. , López‐Campos, C. , Mayet‐Machado, E. , Rincón‐Castañeda, C. B. , Limones‐Aguilar Mde, L. , López‐Campos, J. E. , de León, M. B. , González‐Hernández, S. , Hinojosa‐Garza, D. , & Marcos, R. (2010). Arsenic induces DNA damage in environmentally exposed Mexican children and adults. Influence of GSTO1 and AS3MT polymorphisms. Toxicological Sciences, 117(1), 63–71. [PubMed] [Google Scholar]
  • Sancha, A. M. (2000). Estimate of the current exposure of the urban population of northern Chile to arsenic. IAHS‐AISH Publication: 3–8.
  • Sanchez, T. R. , Oelsner, E. C. , Lederer, D. J. , Lo Cascio, C. M. , Jones, M. R. , Grau‐Perez, M. , Francesconi, K. A. , Goessler, W. , Perzanowski, M. S. , Barr, R. G. , & Navas‐Acien, A. (2019). Rice consumption and subclinical lung disease in US adults: Observational evidence from the multi‐ethnic study of atherosclerosis. American Journal of Epidemiology, 188(9), 1655–1665. [PMC free article] [PubMed] [Google Scholar]
  • Sanchez, T. R. , Powers, M. , Perzanowski, M. , George, C. M. , Graziano, J. H. , & Navas‐Acien, A. (2018). A meta‐analysis of arsenic exposure and Lung function: Is there evidence of restrictive or obstructive Lung disease? Current Environmental Health Reports, 5, 244–254. 10.1007/s40572-018-0192-1 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sanders, A. , Desrosiers, T. , Warren, J. , Herring, A. , Enright, D. , Olshan, A. , Meyer, R. , & Fry, R. (2014). Association between arsenic, cadmium, manganese, and lead levels in private wells and birth defects prevalence in North Carolina: A semi‐ecologic study. BMC Public Health, 14, 955. 10.1186/1471-2458-14-955 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sawada, N. , Iwasaki, M. , Inoue, M. , Takachi, R. , Sasazuki, S. , Yamaji, T. , Shimazu, T. , & Tsugane, S. (2013). Dietary arsenic intake and subsequent risk of cancer: The Japan public health center‐based (JPHC) prospective study. Cancer Causes & Control, 24, 1403–1415. 10.1007/s10552-013-0220-2 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Saxena, R. , Gamble, M. , Wasserman, G. A. , Liu, X. , Parvez, F. , Navas‐Acien, A. , Islam, T. , Factor‐Litvak, P. , Uddin, M. N. , Kioumourtzoglou, M.‐A. , Gibson, E. A. , Shahriar, H. , Slavkovich, V. , Ilievski, V. , LoIacono, N. , Balac, O. , & Graziano, J. H. (2022). Mixed metals exposure and cognitive function in Bangladeshi adolescents. Ecotoxicology and Environmental Safety, 232, 113229. 10.1016/j.ecoenv.2022.113229 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Scannell Bryan, M. , Sofer, T. , Afshar, M. , Mossavar‐Rahmani, Y. , Hosgood, H. D. , Punjabi, N. M. , Zeng, D. , Daviglus, M. L. , & Argos, M. (2021). Mendelian randomization analysis of arsenic metabolism and pulmonary function within the Hispanic community health study/study of Latinos. Scientific Reports, 11, 13470. 10.1038/s41598-021-92911-8 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Schuhmacher‐Wolz, U. , Dieter, H. H. , Klein, D. , & Schneider, K. (2009). Oral exposure to inorganic arsenic: Evaluation of its carcinogenic and non‐carcinogenic effects. Critical Reviews in Toxicology, 39(4), 271–298. [PubMed] [Google Scholar]
  • Sen, J. , & Chaudhuri, A. B. (2008). Arsenic exposure through drinking water and its effect on pregnancy outcome in Bengali women. Arhiv Za Higijenu Rada Toksikologiju, 59, 271–275. [PubMed] [Google Scholar]
  • Sengupta, M. , Deb, I. , Sharma, G. D. , & Kar, K. K. (2013). Human sperm and other seminal constituents in male infertile patients from arsenic and cadmium rich areas of southern Assam. Systems Biology in Reproductive Medicine, 59, 199–209. 10.3109/19396368.2013.783143 [PubMed] [CrossRef] [Google Scholar]
  • Shapiro, G. D. , Dodds, L. , Arbuckle, T. E. , Ashley‐Martin, J. , Fraser, W. , Fisher, M. , Taback, S. , Keely, E. , Bouchard, M. F. , Monnier, P. , Dallaire, R. , Morisset, A. , & Ettinger, A. S. (2015). Exposure to phthalates, bisphenol a and metals in pregnancy and the association with impaired glucose tolerance and gestational diabetes mellitus: The MIREC study. Environment International, 83, 63–71. 10.1016/j.envint.2015.05.016 [PubMed] [CrossRef] [Google Scholar]
  • Shen, H. , Xu, W. , Zhang, J. , Chen, M. , Martin, F. L. , Xia, Y. , Liu, L. , Dong, S. , & Zhu, Y. G. (2013). Urinary metabolic biomarkers link oxidative stress indicators associated with general arsenic exposure to male infertility in a han chinese population. Environmental Science & Technology, 47, 8843–8851. 10.1021/es402025n [PubMed] [CrossRef] [Google Scholar]
  • Shih, Y. H. , Argos, M. , & Turyk, M. E. (2019). Urinary arsenic concentration, airway inflammation, and lung function in the US adult population. Environmental Research, 175, 308–315. [PubMed] [Google Scholar]
  • Shih, Y. H. , Islam, T. , Hore, S. K. , Sarwar, G. , Shahriar, M. H. , Yunus, M. , Graziano, J. H. , Harjes, J. , Baron, J. A. , Parvez, F. , Ahsan, H. , & Argos, M. (2017). Associations between prenatal arsenic exposure with adverse pregnancy outcome and child mortality. Environmental Research, 158, 456–461. 10.1016/j.envres.2017.07.004 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Shih, Y. H. , Scannell Bryan, M. , & Argos, M. (2020). Association between prenatal arsenic exposure, birth outcomes, and pregnancy complications: An observational study within the National Children's study cohort. Environmental Research, 183, 109182. 10.1016/j.envres.2020.109182 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Shiue, I. (2014). Higher urinary heavy metal, arsenic, and phthalate concentrations in people with high blood pressure: US NHANES, 2009–2010. Blood Pressure, 23, 363–369. 10.3109/08037051.2014.925228 [PubMed] [CrossRef] [Google Scholar]
  • Shiue, I. (2015). Urinary heavy metals, phthalates and polyaromatic hydrocarbons independent of health events are associated with adult depression: USA NHANES, 2011–2012. Environmental Science and Pollution Research International, 22, 17095–17103. 10.1007/s11356-015-4944-2 [PubMed] [CrossRef] [Google Scholar]
  • Shokoohi, R. , Khazaei, M. , Karami, M. , Seidmohammadi, A. , Berijani, N. , Khotanlou, H. , & Torkshavand, Z. (2021). The relationship between chronic exposure to arsenic through drinking water and hearing function in exposed population aged 10‐49 years: A cross‐sectional study. Ecotoxicology and Environmental Safety, 211, 111939. 10.1016/j.ecoenv.2021.111939 [PubMed] [CrossRef] [Google Scholar]
  • Siddique, A. E. , Rahman, M. , Hossain, M. I. , Karim, Y. , Hasibuzzaman, M. M. , Biswas, S. , Islam, M. S. , Rahman, A. , Hossen, F. , Mondal, V. , & Banna, H. U. (2020). Association between chronic arsenic exposure and the characteristic features of asthma. Chemosphere, 246, 125790. [PubMed] [Google Scholar]
  • Sieniawska, C. E. , Jung, L. C. , Olufadi, R. , & Walker, V. (2012). Twenty‐four‐hour urinary trace element excretion: Reference intervals and interpretive issues. Annals of Clinical Biochemistry, 49(Pt 4), 341–351. 10.1258/acb.2011.011179 [PubMed] [CrossRef] [Google Scholar]
  • Signes‐Pastor, A. J. , Cottingham, K. L. , Carey, M. , Sayarath, V. , Palys, T. , Meharg, A. A. , Folt, C. L. , & Karagas, M. R. (2018). Infants' dietary arsenic exposure during transition to solid food. Scientific Reports, 8, 7114. 10.1038/s41598-018-25372-1 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Signes‐Pastor, A. J. , Gutiérrez‐González, E. , García‐Villarino, M. , Rodríguez‐Cabrera, F. D. , López‐Moreno, J. J. , Varea‐Jiménez, E. , Pastor‐Barriuso, R. , Pollán, M. , Navas‐Acien, A. , Pérez‐Gómez, B. , & Karagas, M. R. (2021). Toenails as a biomarker of exposure to arsenic: A review. Environmental Research, 195, 110286. 10.1016/j.envres.2020.110286 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Signes‐Pastor, A. J. , Martinez‐Camblor, P. , Baker, E. , Madan, J. , Guill, M. F. , & Karagas, M. R. (2021). Prenatal exposure to arsenic and lung function in children from the New Hampshire birth cohort study. Environment International, 155, 106673. [PMC free article] [PubMed] [Google Scholar]
  • Signes‐Pastor, A. J. , Scot Zens, M. , Seigne, J. , Schned, A. , & Karagas, M. R. (2019). Rice consumption and incidence of bladder cancer in the United States population. Epidemiology, 30, e4–e5. 10.1097/ede.0000000000000955 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Signes‐Pastor, A. J. , Vioque, J. , Navarrete‐Muñoz, E. M. , Carey, M. , García‐Villarino, M. , Fernández‐Somoano, A. , Tardón, A. , Santa‐Marina, L. , Irizar, A. , Casas, M. , Guxens, M. , Llop, S. , Soler‐Blasco, R. , García‐de‐la‐Hera, M. , Karagas, M. R. , & Meharg, A. A. (2019). Inorganic arsenic exposure and neuropsychological development of children of 4–5 years of age living in Spain. Environmental Research, 174, 135–142. 10.1016/j.envres.2019.04.028 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Simic, A. , Hansen, A. F. , Åsvold, B. O. , Romundstad, P. R. , Midthjell, K. , Syversen, T. , & Flaten, T. P. (2017). Trace element status in patients with type 2 diabetes in Norway: The HUNT3 survey. Journal of Trace Elements in Medicine and Biology, 41, 91–98. 10.1016/j.jtemb.2017.03.001 [PubMed] [CrossRef] [Google Scholar]
  • Singh, K. P. , Kumari, R. , Treas, J. , & DuMond, J. W. (2011). Chronic exposure to arsenic causes increased cell survival, DNA damage, and increased expression of mitochondrial transcription factor a (mtTFA) in human prostate epithelial cells. Chemical Research in Toxicology, 24(3), 340–349. 10.1021/tx1003112 [PubMed] [CrossRef] [Google Scholar]
  • Skalny, A. V. , Simashkova, N. V. , Klyushnik, T. P. , Grabeklis, A. R. , Bjørklund, G. , Skalnaya, M. G. , Nikonorov, A. A. , & Tinkov, A. A. (2017). Hair toxic and essential trace elements in children with autism spectrum disorder. Metabolic Brain Disease, 32, 195–202. 10.1007/s11011-016-9899-6 [PubMed] [CrossRef] [Google Scholar]
  • Skalny, A. V. , Simashkova, N. V. , Klyushnik, T. P. , Grabeklis, A. R. , Radysh, I. V. , Skalnaya, M. G. , & Tinkov, A. A. (2016). Analysis of hair trace elements in children with autism Spectrum disorders and communication disorders. Biological Trace Element Research, 177, 215–223. 10.1007/s12011-016-0878-x [PubMed] [CrossRef] [Google Scholar]
  • Skogheim, T. S. , Weyde, K. V. F. , Engel, S. M. , Aase, H. , Surén, P. , Øie, M. G. , Biele, G. , Reichborn‐Kjennerud, T. , Caspersen, I. H. , Hornig, M. , Haug, L. S. , & Villanger, G. D. (2021). Metal and essential element concentrations during pregnancy and associations with autism spectrum disorder and attention‐deficit/hyperactivity disorder in children. Environment International, 152, 106468. 10.1016/j.envint.2021.106468 [PubMed] [CrossRef] [Google Scholar]
  • Smeester, L. , Rager, J. E. , Bailey, K. A. , Guan, X. , Smith, N. , García‐Vargas, G. , Del Razo, L. M. , Drobná, Z. , Kelkar, H. , Stýblo, M. , & Fry, R. C. (2011). Epigenetic changes in individuals with arsenicosis. Chemical Research in Toxicology, 24(2), 165–167. [PMC free article] [PubMed] [Google Scholar]
  • Smith, A. H. , Ercumen, A. , Yuan, Y. , & Steinmaus, C. M. (2009). Increased lung cancer risks are similar whether arsenic is ingested or inhaled. Journal of Exposure Science & Environmental Epidemiology, 19(4), 343–348. 10.1038/jes.2008.73 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Smith, A. H. , Marshall, G. , Liaw, J. , Yuan, Y. , Ferreccio, C. , & Steinmaus, C. (2012). Mortality in young adults following in utero and childhood exposure to arsenic in drinking water. Environmental Health Perspectives, 120, 1527–1531. 10.1289/ehp.1104867 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Smith, A. H. , Marshall, G. , Roh, T. , Ferreccio, C. , Liaw, J. , & Steinmaus, C. (2018). Lung, bladder, and kidney cancer mortality 40 years after arsenic exposure reduction. Journal of the National Cancer Institute, 110, 241–249. 10.1093/jnci/djx201 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Smith, A. H. , Marshall, G. , Yuan, Y. , Liaw, J. , Ferreccio, C. , & Steinmaus, C. (2011). Evidence from Chile that arsenic in drinking water may increase mortality from pulmonary tuberculosis. American Journal of Epidemiology, 173, 414–420. 10.1093/aje/kwq383 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Snoj Tratnik, J. , Falnoga, I. , Mazej, D. , Kocman, D. , Fajon, V. , Jagodic, M. , Stajnko, A. , Trdin, A. , Šlejkovec, Z. , Jeran, Z. , Osredkar, J. , Sešek‐Briški, A. , Krsnik, M. , Kobal, A. B. , Kononenko, L. , & Horvat, M. (2019). Results of the first national human biomonitoring in Slovenia: Trace elements in men and lactating women, predictors of exposure and reference values. International Journal of Hygiene and Environmental Health, 222, 563–582. 10.1016/j.ijheh.2019.02.008 [PubMed] [CrossRef] [Google Scholar]
  • Sobel, M. H. , Sanchez, T. R. , Jones, M. R. , Kaufman, J. D. , Francesconi, K. A. , Blaha, M. J. , Vaidya, D. , Shimbo, D. , Gossler, W. , Gamble, M. V. , Genkinger, J. M. , & Navas‐Acien, A. (2020). Rice intake, arsenic exposure, and subclinical cardiovascular disease among US adults in MESA. Journal of the American Heart Association, 9, e015658. 10.1161/jaha.119.015658 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sohel, N. , Persson, L. Å. , Rahman, M. , Streatfield, P. K. , Yunus, M. , Ekström, E.‐C. , & Vahter, M. (2009). Arsenic in drinking Water and adult mortality. Epidemiology, 20, 824–830. 10.1097/EDE.0b013e3181bb56ec [PubMed] [CrossRef] [Google Scholar]
  • Soler‐Blasco, R. , Murcia, M. , Lozano, M. , Sarzo, B. , Esplugues, A. , Riutort‐Mayol, G. , Vioque, J. , Lertxundi, N. , Santa Marina, L. , Lertxundi, A. , Irizar, A. , Braeuer, S. , Ballester, F. , & Llop, S. (2022). Prenatal arsenic exposure, arsenic methylation efficiency, and neuropsychological development among preschool children in a Spanish birth cohort. Environmental Research, 207, 112208. 10.1016/j.envres.2021.112208 [PubMed] [CrossRef] [Google Scholar]
  • Soria, A. G. , Guber, R. S. , Tefaha, L. M. , Aragón, F. F. , Romero, C. J. , Del Valle, T. R. , Sandoval, N. G. , & Áleman, M. N. (2021). Prevalence of arterial hypertension and cardiovascular risk factors in a rural population exposed to arsenic in Argentina. Revista Peruana de Medicina Experimental y Salud Pública, 38, 530–536. 10.17843/rpmesp.2021.384.9402 [PubMed] [CrossRef] [Google Scholar]
  • Speer, R. M. , Nandi, S. P. , Cooper, K. L. , Zhou, X. , Yu, H. , Guo, Y. , Hudson, L. G. , Alexandrov, L. B. , & Liu, K. J. (2023). Arsenic is a potent co‐mutagen of ultraviolet light. bioRxiv [Preprint]. 10.1101/2023.02.22.529578 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Spence, J. D. , Eliasziw, M. , DiCicco, M. , Hackam, D. G. , Galil, R. , & Lohmann, T. (2002). Carotid plaque area: A tool for targeting and evaluating vascular preventive therapy. Stroke, 33, 2916–2922. 10.1161/01.str.0000042207.16156.b9 [PubMed] [CrossRef] [Google Scholar]
  • Spratlen, M. J. , Grau‐Perez, M. , Best, L. G. , Yracheta, J. , Lazo, M. , Vaidya, D. , Balakrishnan, P. , Gamble, M. V. , Francesconi, K. A. , Goessler, W. , Cole, S. A. , Umans, J. G. , Howard, B. V. , & Navas‐Acien, A. (2018). The Association of Arsenic Exposure and Arsenic Metabolism with the metabolic syndrome and its individual components: Prospective evidence from the strong heart family study. American Journal of Epidemiology, 187, 1598–1612. 10.1093/aje/kwy048 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sripaoraya, K. , Siriwong, W. , Pavittranon, S. , & Chapman, R. S. (2017). Environmental arsenic exposure and risk of diabetes type 2 in Ron Phibun subdistrict, Nakhon Si Thammarat Province, Thailand: Unmatched and matched case‐control studies. Risk Management and Healthcare Policy, 10, 41–48. 10.2147/rmhp.S128277 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Stea, F. , Faita, F. , Borghini, A. , Faita, F. , Bianchi, F. , Bustaffa, E. , Minichilli, F. , Andreassi, M. G. , & Sicari, R. (2016). Arsenic and subclinical vascular damage in a sample of Italian young adults: A cross‐sectional analysis. Environmental Science and Pollution Research International, 23, 20307–20314. 10.1007/s11356-016-7260-6 [PubMed] [CrossRef] [Google Scholar]
  • Steinmaus, C. , Ferreccio, C. , Acevedo, J. , Balmes, J. R. , Liaw, J. , Troncoso, P. , Dauphiné, D. C. , Nardone, A. , & Smith, A. H. (2016). High risks of lung disease associated with early‐life and moderate lifetime arsenic exposure in northern Chile. Toxicology and Applied Pharmacology, 313, 10–15. [PMC free article] [PubMed] [Google Scholar]
  • Steinmaus, C. , Ferreccio, C. , Acevedo, J. , Yuan, Y. , Liaw, J. , Durán, V. , Cuevas, S. , García, J. , Meza, R. , Valdés, R. , Valdés, G. , Benítez, H. , VanderLinde, V. , Villagra, V. , Cantor, K. P. , Moore, L. E. , Perez, S. G. , Steinmaus, S. , & Smith, A. H. (2014a). Increased lung and bladder cancer incidence in adults after in utero and early‐life arsenic exposure. Cancer Epidemiology, Biomarkers & Prevention, 23(8), 1529–1538. 10.1158/1055-9965.EPI-14-0059 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Steinmaus, C. , Ferreccio, C. , Yuan, Y. , Acevedo, J. , González, F. , Perez, L. , Cortés, S. , Balmes, J. R. , Liaw, J. , & Smith, A. H. (2014b). Elevated lung cancer in younger adults and low concentrations of arsenic in water. American Journal of Epidemiology, 180(11), 1082–1087. 10.1093/aje/kwu238 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Steinmaus, C. , Yuan, Y. , Kalman, D. , Atallah, R. , & Smith, A. H. (2005). Intraindividual variability in arsenic methylation in a U.S. population. Cancer Epidemiology, Biomarkers & Prevention, 14(4), 919–924. 10.1158/1055-9965.EPI-04-0277 [PubMed] [CrossRef] [Google Scholar]
  • Steinmaus, C. , Yuan, Y. , Liaw, J. , & Smith, A. H. (2009). Low‐level population exposure to inorganic arsenic in the United States and diabetes mellitus: A reanalysis. Epidemiology, 20(6), 807–815. 10.1097/EDE.0b013e3181b0fd29 [PubMed] [CrossRef] [Google Scholar]
  • Steinmaus, C. M. , Ferreccio, C. , Romo, J. A. , Yuan, Y. , Cortes, S. , Marshall, G. , Moore, L. E. , Balmes, J. R. , Liaw, J. , Golden, T. , & Smith, A. H. (2013). Drinking water arsenic in northern Chile: High cancer risks 40 years after exposure cessation. Cancer Epidemiology, Biomarkers & Prevention, 22(4), 623–630. [PMC free article] [PubMed] [Google Scholar]
  • Stetson, S. J. , Lawrence, C. , Whitcomb, S. , & Kanagy, C. (2020). Determination of four arsenic species in environmental water samples by liquid chromatography‐ inductively coupled plasma ‐ tandem mass spectrometry. MethodsX, 8, 101183. 10.1016/j.mex.2020.101183 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Stiboller, M. , Raber, G. , Lenters, V. , Gjengedal, E. L. F. , Eggesbø, M. , & Francesconi, K. A. (2017). Arsenolipids detected in the Milk of nursing mothers. Environmental Science & Technology Letters, 4(7), 273–279. [Google Scholar]
  • Straif, K. , Benbrahim‐Tallaa, L. , Baan, R. , Grosse, Y. , Secretan, B. , El Ghissassi, F. , Bouvard, V. , Guha, N. , Freeman, C. , Galichet, L. , Cogliano, V. , & the Working Group WHO/IARC . (2009). A review of human carcinogens‐part C: Metals, arsenic, dusts, and fibres. Lancet Oncology, 10(5), 453–454. [PubMed] [Google Scholar]
  • Styblo, M. , Del Razo, L. M. , Vega, L. , Germolec, D. R. , LeCluyse, E. L. , Hamilton, G. A. , Reed, W. , Wang, C. , Cullen, W. R. , & Thomas, D. J. (2000). Comparative toxicity of trivalent and pentavalent inorganic and methylated arsenicals in rat and human cells. Archives of Toxicology, 74(6), 289–299. [PubMed] [Google Scholar]
  • Su, C. T. , Lin, H. C. , Choy, C. S. , Huang, Y. K. , Huang, S. R. , & Hsueh, Y. M. (2012). The relationship between obesity, insulin and arsenic methylation capability in Taiwan adolescents. Sci Total Environ, 414, 152–158. 10.1016/j.scitotenv.2011.10.023 [PubMed] [CrossRef] [Google Scholar]
  • Suhl, J. , Conway, K. M. , Rhoads, A. , Langlois, P. H. , Feldkamp, M. L. , Michalski, A. M. , Oleson, J. J. , Sidhu, A. , Kancherla, V. , Obrycki, J. , Mazumdar, M. , Romitti, P. A. , & Natl Birth Defects Prevention . (2022). Pre‐pregnancy exposure to arsenic in diet and non‐cardiac birth defects. Public Health Nutrition, 26, 620–632. 10.1017/s1368980022001318 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Suhl, J. , Leonard, S. , Weyer, P. , Rhoads, A. , Siega‐Riz, A. M. , Renée Anthony, T. , Burns, T. L. , Conway, K. M. , Langlois, P. H. , & Romitti, P. A. (2018). Maternal arsenic exposure and nonsyndromic orofacial clefts. Birth Defects Research, 110, 1455–1467. 10.1002/bdr2.1386 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sukhn, C. , Awwad, J. , Ghantous, A. , & Zaatari, G. (2018). Associations of semen quality with non‐essential heavy metals in blood and seminal fluid: Data from the environment and male infertility (EMI) study in Lebanon. Journal of Assisted Reproduction and Genetics, 35(9), 1691–1701. 10.1007/s10815-018-1236-z [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sy, S. M. T. , Salud‐Gnilo, C. M. , Yap‐Silva, C. , & Tababa, E. J. L. (2017). A retrospective review of the dermatologic manifestations of chronic arsenic poisoning in The Philippines. International Journal of Dermatology, 56, 721–725. 10.1111/ijd.13615 [PubMed] [CrossRef] [Google Scholar]
  • Szabo, S. T. , Harry, G. J. , Hayden, K. M. , Szabo, D. T. , & Birnbaum, L. (2016). Comparison of metal levels between postmortem brain and ventricular fluid in Alzheimer's disease and nondemented elderly controls. Toxicological Sciences, 150, 292–300. 10.1093/toxsci/kfv325 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Takumi, S. , Aoki, Y. , Sano, T. , Suzuki, T. , Nohmi, T. , & Nohara, K. (2014). In vivo mutagenicity of arsenite in the livers of gpt delta transgenic mice. Mutation Research, Genetic Toxicology and Environmental Mutagenesis, 760, 42–47. 10.1016/j.mrgentox.2013.12.001 [PubMed] [CrossRef] [Google Scholar]
  • Tam, G. K. , Charbonneau, S. M. , Bryce, F. , Pomroy, C. , & Sandi, E. (1979). Metabolism of inorganic arsenic (74As) in humans following oral ingestion. Toxicology and Applied Pharmacology, 50(2), 319–322. [PubMed] [Google Scholar]
  • Tam, L. M. , Price, N. E. , & Wang, Y. (2020). Molecular mechanisms of arsenic‐induced disruption of DNA repair. Chemical Research in Toxicology, 33(3), 709–726. 10.1021/acs.chemrestox.9b00464 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tang, J. , Zhu, Q. , Xu, Y. , Zhou, Y. , Zhu, L. , Jin, L. , Wang, W. , Gao, L. , Chen, G. , & Zhao, H. (2022). Total arsenic, dimethylarsinic acid, lead, cadmium, total mercury, methylmercury and hypertension among Asian populations in the United States: NHANES 2011–2018. Ecotoxicology and Environmental Safety, 241, 113776. 10.1016/j.ecoenv.2022.113776 [PubMed] [CrossRef] [Google Scholar]
  • Tauheed, J. , Sanchez‐Guerra, M. , Lee, J. J. , Paul, L. , Ibne Hasan, M. O. S. , Quamruzzaman, Q. , Selhub, J. , Wright, R. O. , Christiani, D. C. , Coull, B. A. , Baccarelli, A. A. , & Mazumdar, M. (2017). Associations between post translational histone modifications, myelomeningocele risk, environmental arsenic exposure, and folate deficiency among participants in a case control study in Bangladesh. Epigenetics, 12, 484–491. 10.1080/15592294.2017.1312238 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Taylor, V. , Goodale, B. , Raab, A. , Schwerdtle, T. , Reimer, K. , Conklin, S. , Karagas, M. R. , & Francesconi, K. A. (2017). Human exposure to organic arsenic species from seafood. Science of the Total Environment, 580, 266–282. 10.1016/j.scitotenv.2016.12.113 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Taylor, V. F. , Li, Z. , Sayarath, V. , Palys, T. J. , Morse, K. R. , Scholz‐Bright, R. A. , & Karagas, M. R. (2017). Distinct arsenic metabolites following seaweed consumption in humans. Scientific Reports, 7(1), 3920. 10.1038/s41598-017-03883-7 Erratum in: Scientific Reports 2018 Mar 2;8(1):4145. PMID: 28634348; PMCID: PMC5478658. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • TDOH (Taiwan Department of Health) . (2007). Compilation of exposure factors. DOH96‐HP‐1801. Taiwan Department of Health. [Google Scholar]
  • Thakur, M. , Rachamalla, M. , Niyogi, S. , Datusalia, A. K. , & Flora, S. J. S. (2021). Molecular mechanism of arsenic‐induced neurotoxicity including neuronal dysfunctions. International Journal of Molecular Sciences, 22(18), 10077. 10.3390/ijms221810077 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Thomas, D. J. (2021). Arsenic methylation ‐ lessons from three decades of research. Toxicology, 457, 152800. 10.1016/j.tox.2021.152800 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Thomas, D. J. , Styblo, M. , & Lin, S. (2001). The cellular metabolism and systemic toxicity of arsenic. Toxicology and Applied Pharmacology, 176(2), 127–144. 10.1006/taap.2001.9258 PMID: 11601889. [PubMed] [CrossRef] [Google Scholar]
  • Thomas, S. , Arbuckle, T. E. , Fisher, M. , Fraser, W. D. , Ettinger, A. , & King, W. (2015). Metals exposure and risk of small‐for‐gestational age birth in a Canadian birth cohort: The MIREC study. Environmental Research, 140, 430–439. 10.1016/j.envres.2015.04.018 [PubMed] [CrossRef] [Google Scholar]
  • Tian, M. , Wang, Y. X. , Wang, X. , Wang, H. , Liu, L. , Zhang, J. , Nan, B. , Shen, H. , & Huang, Q. (2021). Environmental doses of arsenic exposure are associated with increased reproductive‐age male urinary hormone excretion and in vitro Leydig cell steroidogenesis. Journal of Hazardous Materials, 408, 124904. 10.1016/j.jhazmat.2020.124904 [PubMed] [CrossRef] [Google Scholar]
  • Tindula, G. , Mukherjee, S. K. , Ekramullah, S. M. , Arman, D. M. , Biswas, S. K. , Islam, J. , Obrycki, J. F. , Christiani, D. C. , Liang, L. , Warf, B. C. , & Mazumdar, M. (2021). Parental metal exposures as potential risk factors for spina bifida in Bangladesh. Environment International, 157, 106800. 10.1016/j.envint.2021.106800 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tinkelman, N. E. , Spratlen, M. J. , Domingo‐Relloso, A. , Tellez‐Plaza, M. , Grau‐Perez, M. , Francesconi, K. A. , Goessler, W. , Howard, B. V. , MacCluer, J. , North, K. E. , Umans, J. G. , Factor‐Litvak, P. , Cole, S. A. , & Navas‐Acien, A. (2020). Associations of maternal arsenic exposure with adult fasting glucose and insulin resistance in the strong heart study and strong heart family study. Environment International, 137, 105531. 10.1016/j.envint.2020.105531 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tofail, F. , Vahter, M. , Hamadani, J. D. , Nermell, B. , Huda, S. N. , Yunus, M. , Rahman, M. , & Grantham‐McGregor, S. M. (2009). Effect of arsenic exposure during pregnancy on infant development at 7 months in rural Matlab, Bangladesh. Environmental Health Perspectives, 117(2), 288–293. 10.1289/ehp.11670 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tokar, E. J. , Diwan, B. A. , Ward, J. M. , Delker, D. A. , & Waalkes, M. P. (2011). Carcinogenic effects of “whole‐life” exposure to inorganic arsenic in CD1 mice. Toxicology Science, 119(1), 73–83. 10.1093/toxsci/kfq315 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tokar, E. J. , Person, R. J. , Sun, Y. , Perantoni, A. O. , & Waalkes, M. P. (2013). Chronic exposure of renal stem cells to inorganic arsenic induces a cancer phenotype. Chemical Research in Toxicology, 26(1), 96–105. 10.1021/tx3004054 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tovar‐Sanchez, E. , Mussali‐Galante, P. , Martinez‐Pacheco, M. , Ortiz‐Hernandez, M. L. , Sanchez‐Salinas, E. , & Olvera‐Velona, A. (2016). Relationship BETWEEN genotoxic damage and arsenic BLOOD concentrations in individuals residing in an arsenic contaminated area in MORELOS, Mexico. Revista Internacional De Contaminacion Ambiental, 32, 101–117. [Google Scholar]
  • Tsai, S. M. , Wang, T. N. , & Ko, Y. C. (1999). Mortality for certain diseases in areas with high levels of arsenic in drinking water. Archives of Environmental Health, 54, 186–193. 10.1080/00039899909602258 [PubMed] [CrossRef] [Google Scholar]
  • Tsai, T. L. , Kuo, C. C. , Hsu, L. I. , Tsai, S. F. , Chiou, H. Y. , Chen, C. J. , Hsu, K. H. , & Wang, S. L. (2021). Association between arsenic exposure, DNA damage, and urological cancers incidence: A long‐term follow‐up study of residents in an arseniasis endemic area of northeastern Taiwan. Chemosphere, 266, 129094. 10.1016/j.chemosphere.2020.129094 [PubMed] [CrossRef] [Google Scholar]
  • Tsai, T. L. , Lei, W. T. , Kuo, C. C. , Sun, H. L. , Su, P. H. , & Wang, S. L. (2021). Maternal and childhood exposure to inorganic arsenic and airway allergy–a 15‐year birth cohort follow‐up study. Environment International, 146, 106243. [PubMed] [Google Scholar]
  • Tsai, T. L. , Wang, S.‐L. , Hsieh, C.‐J. , Wen, H.‐J. , Kuo, C.‐C. , Liu, H.‐J. , Sun, C.‐W. , Chen, M.‐L. , Wu, M.‐T. , & Grp, T. S. (2021). Association between prenatal exposure to metals and atopic dermatitis among children aged 4 years in Taiwan. JAMA Network Open, 4, e2131327. 10.1001/jamanetworkopen.2021.31327 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tseng, H. P. , Wang, Y. H. , Wu, M. M. , The, H. W. , Chiou, H. Y. , & Chen, C. J. (2006). Association between chronic exposure to arsenic and slow nerve conduction velocity among adolescents in Taiwan. Journal of Health, Population, and Nutrition, 24, 182–189. [PubMed] [Google Scholar]
  • Tseng, W. P. , Chu, H. M. , How, S. W. , Fong, J. M. , Lin, C. S. , & Yeh, S. (1968). Prevalence of skin cancer in an endemic area of chronic arsenicism in Taiwan. Journal of the National Cancer Institute, 40(3), 453–463. [PubMed] [Google Scholar]
  • Tsinovoi, C. L. , Xun, P. , McClure, L. A. , Carioni, V. M. O. , Brockman, J. D. , Cai, J. , Guallar, E. , Cushman, M. , Unverzagt, F. W. , Howard, V. J. , & He, K. (2018). Arsenic exposure in relation to ischemic stroke: The reasons for geographic and racial differences in stroke study. Stroke, 49, 19–26. 10.1161/strokeaha.117.018891 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tsuji, J. S. , Perez, V. , Garry, M. R. , & Alexander, D. D. (2014). Association of low‐level arsenic exposure in drinking water with cardiovascular disease: A systematic review and risk assessment. Toxicology, 323, 78–94. 10.1016/j.tox.2014.06.008 [PubMed] [CrossRef] [Google Scholar]
  • Tubbs, A. , & Nussenzweig, A. (2017). Endogenous DNA damage as a source of genomic instability in cancer. Cell, 168(4), 644–656. 10.1016/j.cell.2017.01.002 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Unterberg, M. , Leffers, L. , Hubner, F. , Humpf, H. U. , Lepikhov, K. , Walter, J. , Ebert, F. , & Schwerdtle, T. (2014). Toxicity of arsenite and thio‐DMA(V) after long‐term (21 days) incubation of human urothelial cells: Cytotoxicity, genotoxicity and epigenetics. Toxicology Research, 3, 456–464. 10.1039/c4tx00036f [CrossRef] [Google Scholar]
  • US EPA (United States Environmental Protection Agency) . (2001). National primary drinking water regulations; arsenic and clarifications to compliance and new source contaminants monitoring. Federal Register, 66, 6976–7066. [Google Scholar]
  • US EPA (United States Environmental Protection Agency) . (2012). Benchmark dose technical guidance [website]. Office of the Science Advisor, U.S. Environmental Protection Agency. https://www.epa.gov/risk/benchmark‐dose‐technical‐guidance [Google Scholar]
  • US FDA (U.S. Food and Drug Administration) , 2016. Arsenic in Rice and Rice products risk assessment report. https://www.fda.gov/files/food/published/Arsenic‐in‐Rice‐and‐Rice‐Products‐Risk‐Assessment‐Report‐PDF.pdf [Google Scholar]
  • Vahter, M. (1999). Methylation of inorganic arsenic in different mammalian species and population groups. Science Progress, 82(1), 69–88. [PMC free article] [PubMed] [Google Scholar]
  • Vahter, M. , Skröder, H. , Rahman, S. M. , Levi, M. , Derakhshani Hamadani, J. , & Kippler, M. (2020). Prenatal and childhood arsenic exposure through drinking water and food and cognitive abilities at 10 years of age: A prospective cohort study. Environment International, 139, 105723. 10.1016/j.envint.2020.105723 [PubMed] [CrossRef] [Google Scholar]
  • Vahter, M. E. , Li, L. , Nermell, B. , Rahman, A. , El Arifeen, S. , Rahman, M. , Persson, L. A. , & Ekström, E. C. (2006). Arsenic exposure in pregnancy: A population‐based study in Matlab, Bangladesh. Journal of Health, Population, and Nutrition, 24, 236–245. [PubMed] [Google Scholar]
  • Valappil, A. V. , & Mammen, A. (2019). Subacute arsenic neuropathy: Clinical and electrophysiological observations. Journal of Neuroscience in Rural Practice, 10, 529–532. 10.1055/s-0039-1695693 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Valeri, L. , Mazumdar, M. M. , Bobb, J. F. , Claus Henn, B. , Rodrigues, E. , Sharif, O. I. A. , Kile, M. L. , Quamruzzaman, Q. , Afroz, S. , Golam, M. , Amarasiriwardena, C. , Bellinger, D. C. , Christiani, D. C. , Coull, B. A. , & Wright, R. O. (2017). The joint effect of prenatal exposure to metal mixtures on neurodevelopmental outcomes at 20–40 months of age: Evidence from rural Bangladesh. Environmental Health Perspectives, 125(6), 067015. 10.1289/EHP614 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Van Brusselen, D. , Kayembe‐Kitenge, T. , Mbuyi‐Musanzayi, S. , Lubala Kasole, T. , Kabamba Ngombe, L. , Musa Obadia, P. , Kyanika Wa Mukoma, D. , Van Herck, K. , Avonts, D. , Devriendt, K. , Smolders, E. , Nkulu, C. B. L. , & Nemery, B. (2020). Metal mining and birth defects: A case‐control study in Lubumbashi, Democratic Republic of the Congo. The Lancet Planetary Health, 4(4), e158–e167. [PubMed] [Google Scholar]
  • Varsanyi, I. , Fodre, Z. , & Bartha, A. (1991). Arsenic in drinking‐water and mortality in the southern great plain, Hungary. Environmental Geochemistry and Health, 13, 14–22. [PubMed] [Google Scholar]
  • Vega‐Millán, C. B. , Dévora‐Figueroa, A. G. , Burgess, J. L. , Beamer, P. I. , Furlong, M. , Lantz, R. C. , Meza‐Figueroa, D. , García‐Rico, L. , Meza‐Escalante, E. R. , Balderas‐Cortés, J. J. , & Meza‐Montenegro, M. M. (2021). Inflammation biomarkers associated with arsenic exposure by drinking water and respiratory outcomes in indigenous children from three Yaqui villages in southern Sonora, México. Environmental Science and Pollution Research, 28(26), 34355–34366. [PMC free article] [PubMed] [Google Scholar]
  • Vergani, L. , Cristina, L. , Rivaro, P. , Luisa, A. , Shyti, G. , Edvige, V. , Minniti, G. , Grasselli, E. , Canesi, L. , & Voci, A. (2011). Metals, metallothioneins and oxidative stress in blood of autistic children. Research in Autism Spectrum Disorders, 5, 286–293. 10.1016/j.rasd.2010.04.010 [CrossRef] [Google Scholar]
  • Vibol, S. , Hashim, J. H. , & Sarmani, S. (2015). Neurobehavioral effects of arsenic exposure among secondary school children in the Kandal Province, Cambodia. Environmental Research, 137, 329–337. 10.1016/j.envres.2014.12.001 [PubMed] [CrossRef] [Google Scholar]
  • Villarreal, J. J. , Ortiz, B. M. , Garza, S. M. , Armendariz, D. I. R. , Villa, V. D. B. , Rosales, P. C. , Martinez, N. D. B. , Aguirre, H. D. , & Martinez, J. M. (2019). Telomere length analysis in residents of a community exposed to arsenic. Journal of Biochemical and Molecular Toxicology, 33, 4. 10.1002/jbt.22230 [PubMed] [CrossRef] [Google Scholar]
  • von Ehrenstein, O. S. , Guha Mazumder, D. N. , Hira‐Smith, M. , Ghosh, N. , Yuan, Y. , Windham, G. , Ghosh, A. , Haque, R. , Lahiri, S. , Kalman, D. , Das, S. , & Smith, A. H. (2006). Pregnancy outcomes, infant mortality, and arsenic in drinking water in West Bengal. India. American Journal of Epidemiology, 163(7), 662–669. [PubMed] [Google Scholar]
  • von Ehrenstein, O. S. , Poddar, S. , Yuan, Y. , Mazumder, G. , Eskenazi, B. , Basu, A. , Hira‐Smith, M. , Ghosh, N. , Lahiri, S. , Haque, R. , Ghosh, A. , Kalman, D. , Das, S. , & Smith, A. H. (2007). Children's intellectual function in relation to arsenic exposure. Epidemiology, 18(1), 44–51. 10.1097/01.ede.0000248900.65613.a9 [PubMed] [CrossRef] [Google Scholar]
  • Waalkes, M. P. , Liu, J. , & Diwan, B. A. (2007). Transplacental arsenic carcinogenesis in mice. Toxicology Applied in Pharmacology, 223(3), 271–280. 10.1016/j.taap.2006.12.034 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Waalkes, M. P. , Qu, W. , Tokar, E. J. , Kissling, G. E. , & Dixon, D. (2014). Lung tumors in mice induced by "whole‐life" inorganic arsenic exposure at human‐relevant doses. Archives of Toxicology, 88(8), 1619–1629. 10.1007/s00204-014-1305-8 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wade, T. , Xia, Y. , Wu, K. , Li, Y. , Ning, Z. , Le, X. C. , Lu, X. , Feng, Y. , He, X. , & Mumford, J. (2009). Increased mortality associated with well‐Water arsenic exposure in Inner Mongolia, China. International Journal of Environmental Research and Public Health, 6, 1107–1123. 10.3390/ijerph6031107 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wade, T. J. , Xia, Y. , Mumford, J. , Wu, K. , Le, X. C. , Sams, E. , & Sanders, W. E. (2015). Cardiovascular disease and arsenic exposure in Inner Mongolia, China: A case control study. Environmental Health, 14, 35. 10.1186/s12940-015-0022-y [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wadhwa, S. K. , Kazi, T. G. , Chandio, A. A. , Afridi, H. I. , Kolachi, N. F. , Khan, S. , Kandhro, G. A. , Nasreen, S. , Shah, A. Q. , & Baig, J. A. (2011). Comparative study of liver cancer patients in arsenic exposed and non‐exposed areas of Pakistan. Biological Trace Element Research, 144(1–3), 86–96. 10.1007/s12011-011-9036-7 [PubMed] [CrossRef] [Google Scholar]
  • Wai, K. M. , Ser, P. H. , Ahmad, S. A. , Yasmin, R. , Ito, Y. , Konishi, S. , Umezaki, M. , & Watanabe, C. (2020). In‐utero arsenic exposure and growth of infants from birth to 6 months of age: A prospective cohort study in rural Bangladesh. International Journal of Environmental Health Research, 30, 421–434. 10.1080/09603123.2019.1597835 [PubMed] [CrossRef] [Google Scholar]
  • Wang, A. , Holladay, S. D. , Wolf, D. C. , Ahmed, S. A. , & Robertson, J. L. (2006). Reproductive and developmental toxicity of arsenic in rodents: A review. International Journal of Toxicology, 25, 319–331. 10.1080/10915810600840776 [PubMed] [CrossRef] [Google Scholar]
  • Wang, B. , Liu, J. , Liu, B. , Liu, X. , & Yu, X. (2018). Prenatal exposure to arsenic and neurobehavioral development of newborns in China. Environment International, 121(Pt 1), 421–427. 10.1016/j.envint.2018.09.031 [PubMed] [CrossRef] [Google Scholar]
  • Wang, B. , Zhu, Y. , Yan, L. , Zhang, J. , Wang, X. , Cheng, H. , Li, Z. , Ye, R. , & Ren, A. (2019). Association of maternal chronic arsenic exposure with the risk of neural tube defects in northern China. Environment International, 126, 222–227. 10.1016/j.envint.2019.02.016 [PubMed] [CrossRef] [Google Scholar]
  • Wang, C. , Hao, L. , Liu, C. , Chen, R. , Wang, W. , Chen, Y. , Yang, Y. , Meng, X. , Fu, Q. , Ying, Z. , & Kan, H. (2020). Associations between fine particulate matter constituents and daily cardiovascular mortality in Shanghai. China. Ecotoxicol Environ Saf, 191, 110154. 10.1016/j.ecoenv.2019.110154 [PubMed] [CrossRef] [Google Scholar]
  • Wang, C. , Pi, X. , Yin, S. , Liu, M. , Tian, T. , Jin, L. , Liu, J. , Li, Z. , Wang, L. , Yuan, Z. , Wang, Y. , & Ren, A. (2022). Maternal exposure to heavy metals and risk for severe congenital heart defects in offspring. Environmental Research, 212, 113432. 10.1016/j.envres.2022.113432 [PubMed] [CrossRef] [Google Scholar]
  • Wang, C. H. , Jeng, J. S. , Yip, P. K. , Chen, C. L. , Hsu, L. I. , Hsueh, Y. M. , Chiou, H. Y. , Wu, M. M. , & Chen, C. J. (2002). Biological gradient between long‐term arsenic exposure and carotid atherosclerosis. Circulation, 105(15), 1804–1809. [PubMed] [Google Scholar]
  • Wang, M. , Hossain, F. , Sulaiman, R. , & Ren, X. (2019). Exposure to inorganic arsenic and Lead and autism Spectrum disorder in children: A systematic review and meta‐analysis. Chemical Research in Toxicology, 32, 1904–1919. 10.1021/acs.chemrestox.9b00134 [PubMed] [CrossRef] [Google Scholar]
  • Wang, S. L. , Li, W. F. , Chen, C. J. , Huang, Y. L. , Chen, J. W. , Chang, K. H. , Tsai, L. Y. , & Chou, K. M. (2011). Hypertension incidence after tap‐water implementation: A 13‐year follow‐up study in the arseniasis‐endemic area of southwestern Taiwan. Sci Total Environ, 409, 4528–4535. 10.1016/j.scitotenv.2011.07.058 [PubMed] [CrossRef] [Google Scholar]
  • Wang, S. X. , Wang, Z. H. , Cheng, X. T. , Li, J. , Sang, Z. P. , Zhang, X. D. , Han, L. L. , Qiao, X. Y. , Wu, Z. M. , & Wang, Z. Q. (2007). Arsenic and fluoride exposure in drinking water: children's IQ and growth in Shanyin county, Shanxi province, China. Environmental Health Perspectives, 115(4), 643–647. 10.1289/ehp.9270 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wang, X. , Ding, N. , Harlow, S. D., Jr. , Mukherjee, B. , Gold, E. B. , & Park, S. K. (2021). Urinary metals and metal mixtures and timing of natural menopause in midlife women: The study of Women's health across the nation. Environment International, 157, 106781. 10.1016/j.envint.2021.106781 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wang, X. , Gao, D. , Zhang, G. , Zhang, X. , Li, Q. , Gao, Q. , Chen, R. , Xu, S. , Huang, L. , Zhang, Y. , Lin, L. , Zhong, C. , Chen, X. , Sun, G. , Song, Y. , Yang, X. , Hao, L. , Yang, H. , Yang, L. , & Yang, N. (2020). Exposure to multiple metals in early pregnancy and gestational diabetes mellitus: A prospective cohort study. Environment International, 135, 105370. 10.1016/j.envint.2019.105370 [PubMed] [CrossRef] [Google Scholar]
  • Wang, X. , Huang, X. , Zhou, L. , Chen, J. , Zhang, X. , Xu, K. , Huang, Z. , He, M. , Shen, M. , Chen, X. , Tang, B. , Shen, L. , & Zhou, Y. (2021). Association of arsenic exposure and cognitive impairment: A population‐based cross‐sectional study in China. Neurotoxicology, 82, 100–107. 10.1016/j.neuro.2020.11.009 [PubMed] [CrossRef] [Google Scholar]
  • Wang, X. , Karvonen‐Gutierrez, C. A. , Herman, W. H. , Mukherjee, B. , Harlow, S. D. , & Park, S. K. (2020). Urinary metals and incident diabetes in midlife women: Study of Women's health across the nation (SWAN). BMJ Open Diabetes Research & Care, 8, e001233. 10.1136/bmjdrc-2020-001233 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wang, X. , Karvonen‐Gutierrez, C. A. , Herman, W. H. , Mukherjee, B. , Harlow, S. D. , & Park, S. K. (2021). Urinary heavy metals and longitudinal changes in Blood pressure in midlife women: The study of Women's health across the nation. Hypertension, 78(2), 543–551. 10.1161/HYPERTENSIONAHA.121.17295 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wang, X. , Mukherjee, B. , Karvonen‐Gutierrez, C. A. , Herman, W. H. , Batterman, S. , Harlow, S. D. , & Park, S. K. (2020). Urinary metal mixtures and longitudinal changes in glucose homeostasis: The study of Women's health across the nation (SWAN). Environment International, 145, 106109. 10.1016/j.envint.2020.106109 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wang, X. , Zhang, J. , Xu, W. , Huang, Q. , Liu, L. , Tian, M. , Xia, Y. , Zhang, W. , & Shen, H. (2016). Low‐level environmental arsenic exposure correlates with unexplained male infertility risk. Science of the Total Environment, 571, 307–313. [PubMed] [Google Scholar]
  • Wang, Y. , Wang, S. , Wang, Y. , Lu, A. , Cao, L. , Wang, J. , Gao, Z. , & Yan, C. (2022). Effects of prenatal exposure to arsenic on neonatal birth size in Wujiang. China. Chemosphere, 299. 10.1016/j.chemosphere.2022.134441 [PubMed] [CrossRef] [Google Scholar]
  • Wang, Y. , Wang, Y. , & Yan, C. (2022). Gender differences in trace element exposures with cognitive abilities of school‐aged children: A cohort study in Wujiang city, China. Environmental Science and Pollution Research International, 29, 64807–64821. 10.1007/s11356-022-20353-4 [PubMed] [CrossRef] [Google Scholar]
  • Wang, Y. , Zhang, P. , Chen, X. , Wu, W. , Feng, Y. , Yang, H. , Li, M. , Xie, B. , Guo, P. , Warren, J. L. , Shi, X. , Wang, S. , & Zhang, Y. (2019). Multiple metal concentrations and gestational diabetes mellitus in Taiyuan. China. Chemosphere, 237, 124412. 10.1016/j.chemosphere.2019.124412 [PubMed] [CrossRef] [Google Scholar]
  • Wang, Y. H. , Wu, M. M. , Hong, C. T. , Lien, L. M. , Hsieh, Y. C. , Tseng, H. P. , Chang, S. F. , Su, C. L. , Chiou, H. Y. , & Chen, C. J. (2007). Effects of arsenic exposure and genetic polymorphisms of p53, glutathione S‐transferase M1, T1, and P1 on the risk of carotid atherosclerosis in Taiwan. Atherosclerosis, 192(2), 305–312. [PubMed] [Google Scholar]
  • Wang, Z. , Humphries, B. , Xiao, H. , Jiang, Y. , & Yang, C. (2013). Epithelial to mesenchymal transition in arsenic‐transformed cells promotes angiogenesis through activating β‐catenin‐vascular endothelial growth factor pathway. Toxicology and Applied Pharmacology, 271(1), 20–29. [PMC free article] [PubMed] [Google Scholar]
  • Wasserman, G. A. , Liu, X. , Loiacono, N. J. , Kline, J. , Factor‐Litvak, P. , van Geen, A. , Mey, J. L. , Levy, D. , Abramson, R. , Schwartz, A. , & Graziano, J. H. (2014). A cross‐sectional study of well water arsenic and child IQ in Maine schoolchildren. Environmental Health, 13(1), 23. 10.1186/1476-069X-13-23 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wasserman, G. A. , Liu, X. , Parvez, F. , Ahsan, H. , Factor‐Litvak, P. , Kline, J. , van Geen, A. , Slavkovich, V. , Loiacono, N. J. , Levy, D. , Cheng, Z. , & Graziano, J. H. (2007). Water arsenic exposure and intellectual function in 6‐year‐old children in Araihazar, Bangladesh. Environmental Health Perspectives, 115(2), 285–289. 10.1289/ehp.9501 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wasserman, G. A. , Liu, X. , Parvez, F. , Chen, Y. , Factor‐Litvak, P. , LoIacono, N. J. , Levy, D. , Shahriar, H. , Uddin, M. N. , Islam, T. , Lomax, A. , Saxena, R. , Gibson, E. A. , Kioumourtzoglou, M. A. , Balac, O. , Sanchez, T. , Kline, J. K. , Santiago, D. , Ellis, T. , … Graziano, J. H. (2018). A cross‐sectional study of water arsenic exposure and intellectual function in adolescence in Araihazar, Bangladesh. Environment International, 118, 304–313. 10.1016/j.envint.2018.05.037 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wasserman, G. A. , Liu, X. , Parvez, F. , Factor‐Litvak, P. , Ahsan, H. , Levy, D. , Kline, J. , van Geen, A. , Mey, J. , Slavkovich, V. , Siddique, A. B. , Islam, T. , & Graziano, J. H. (2011). Arsenic and manganese exposure and children's intellectual function. Neurotoxicology, 32, 450–457. 10.1016/j.neuro.2011.03.009 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wasserman, G. A. , Liu, X. H. , Parvez, F. , Ahsan, H. , Factor‐Litvak, P. , van Geen, A. , Slavkovich, V. , Lolacono, N. J. , Cheng, Z. Q. , Hussain, L. , Momotaj, H. , & Graziano, J. H. (2004). Water arsenic exposure and children's intellectual function in Araihazar. Bangladesh. Environmental Health Perspectives, 112(13), 1329–1333. [PMC free article] [PubMed] [Google Scholar]
  • Watters, G. P. , Smart, D. J. , Harvey, J. S. , & Austin, C. A. (2009). H2AX phosphorylation as a genotoxicity endpoint. Mutation Research, 679(1–2), 50–58. 10.1016/j.mrgentox.2009.07.007 [PubMed] [CrossRef] [Google Scholar]
  • Wedler, N. , Matthäus, T. , Strauch, B. , Dilger, E. , Waterstraat, M. , Mangerich, A. , & Hartwig, A. (2021). Impact of the cellular zinc status on PARP‐1 activity and genomic stability in HeLa S3 cells. Chemical Research in Toxicology, 34(3), 839–848. [PubMed] [Google Scholar]
  • Wei, B. , Yu, J. , Kong, C. , Li, H. , Yang, L. , Xia, Y. , & Wu, K. (2018). A follow‐up study of the development of skin lesions associated with arsenic exposure duration. Environmental Geochemistry and Health, 40, 2729–2738. 10.1007/s10653-018-0136-6 [PubMed] [CrossRef] [Google Scholar]
  • Wei, B. , Yu, J. , Wang, J. , Yang, L. , Li, H. , Kong, C. , Xia, Y. , & Wu, K. (2017). The relationships between arsenic methylation and both skin lesions and hypertension caused by chronic exposure to arsenic in drinking water. Environmental Toxicology and Pharmacology, 53, 89–94. 10.1016/j.etap.2017.05.009 [PubMed] [CrossRef] [Google Scholar]
  • Wei, Y. , Jia, C. , Lan, Y. , Hou, X. , Zuo, J. , Li, J. , Wang, T. , & Mao, G. (2019). The association of tryptophan and phenylalanine are associated with arsenic‐induced skin lesions in a Chinese population chronically exposed to arsenic via drinking water: A case‐control study. BMJ Open, 9(10), e025336. 10.1136/bmjopen-2018-025336 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wei, Y. , Shi, Q. , Wang, Z. , Zhang, R. , Su, L. , Quamruzzaman, Q. , Rahman, M. , Chen, F. , & Christiani, D. C. (2017). Maternal/fetal metabolomes appear to mediate the impact of arsenic exposure on birth weight: A pilot study. Journal of Exposure Science & Environmental Epidemiology, 27, 313–319. 10.1038/jes.2016.74 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Weidemann, D. , Kuo, C. C. , Navas‐Acien, A. , Abraham, A. G. , Weaver, V. , & Fadrowski, J. (2015). Association of arsenic with kidney function in adolescents and young adults: Results from the National Health and nutrition examination survey 2009–2012. Environmental Research, 140, 317–324. 10.1016/j.envres.2015.03.030 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wen, J. , Wen, W. , Li, L. , & Liu, H. (2012). Methylation capacity of arsenic and skin lesions in smelter plant workers. Environmental Toxicology and Pharmacology, 34, 624–630. 10.1016/j.etap.2012.07.003 [PubMed] [CrossRef] [Google Scholar]
  • Wen, Y. , Huang, S. , Zhang, Y. , Zhang, H. , Zhou, L. , Li, D. , Xie, C. , Lv, Z. , Guo, Y. , Ke, Y. , Wu, T. , & Cheng, J. (2019). Associations of multiple plasma metals with the risk of ischemic stroke: A case‐control study. Environment International, 125, 125–134. 10.1016/j.envint.2018.12.037 [PubMed] [CrossRef] [Google Scholar]
  • Wheeler, B. W. , Kothencz, G. , & Pollard, A. S. (2013). Geography of non‐melanoma skin cancer and ecological associations with environmental risk factors in England. British Journal of Cancer, 109, 235–241. 10.1038/bjc.2013.288 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wright, R. O. , Amarasiriwardena, C. , Woolf, A. D. , Jim, R. , & Bellinger, D. C. (2006). Neuropsychological correlates of hair arsenic, manganese, and cadmium levels in school‐age children residing near a hazardous waste site. Neurotoxicology, 27(2), 210–216. 10.1016/j.neuro.2005.10.001 [PubMed] [CrossRef] [Google Scholar]
  • Wu, C. C. , Chen, M. C. , Huang, Y. K. , Huang, C. Y. , Lai, L. A. , Chung, C. J. , Shiue, H. S. , Pu, Y. S. , Lin, Y. C. , Han, B. C. , Wang, Y. H. , & Hsueh, Y. M. (2013). Environmental tobacco smoke and arsenic methylation capacity are associated with urothelial carcinoma. Journal of the Formosan Medical Association, 112, 554–560. 10.1016/j.jfma.2013.05.012 [PubMed] [CrossRef] [Google Scholar]
  • Wu, F. , Jasmine, F. , Kibriya, M. G. , Liu, M. , Cheng, X. , Parvez, F. , Islam, T. , Ahmed, A. , Rakibuz‐Zaman, M. , Jiang, J. , Roy, S. , Paul‐Brutus, R. , Slavkovich, V. , Islam, T. , Levy, D. , VanderWeele, T. J. , Pierce, B. L. , Graziano, J. H. , Ahsan, H. , & Chen, Y. (2015). Interaction between arsenic exposure from drinking water and genetic polymorphisms on cardiovascular disease in Bangladesh: A prospective case‐cohort study. Environmental Health Perspectives, 123, 451–457. 10.1289/ehp.1307883 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wu, J. , Chen, G. , Liao, Y. , Song, X. , Pei, L. , Wang, J. , & Zheng, X. (2011). Arsenic levels in the soil and risk of birth defects: A population‐based case‐control study using GIS technology. Journal of Environmental Health, 74, 20–25. [PubMed] [Google Scholar]
  • Wu, J. , Zhang, C. , Pei, L. , Chen, G. , & Zheng, X. (2014). Association between risk of birth defects occurring level and arsenic concentrations in soils of Lvliang, Shanxi province of China. Environmental Pollution, 191, 1–7. 10.1016/j.envpol.2014.04.004 [PubMed] [CrossRef] [Google Scholar]
  • Wu, M. M. , Chiou, H. Y. , Chen, C. L. , Wang, Y. H. , Hsieh, Y. C. , Lien, L. M. , Lee, T. C. , & Chen, C. J. (2010). GT‐repeat polymorphism in the heme oxygenase‐1 gene promoter is associated with cardiovascular mortality risk in an arsenic‐exposed population in northeastern Taiwan. Toxicology and Applied Pharmacology, 248, 226–233. 10.1016/j.taap.2010.08.005 [PubMed] [CrossRef] [Google Scholar]
  • Wu, M. M. , Chiou, H. Y. , Hsueh, Y. M. , Hong, C. T. , Su, C. L. , Chang, S. F. , Huang, W. L. , Wang, H. T. , Wang, Y. H. , Hsieh, Y. C. , & Chen, C. J. (2006). Effect of plasma homocysteine level and urinary monomethylarsonic acid on the risk of arsenic‐associated carotid atherosclerosis. Toxicology and Applied Pharmacology, 216(1), 168–175. 10.1016/j.taap.2006.05.005 [PubMed] [CrossRef] [Google Scholar]
  • Xia, X. , Liang, C. , Sheng, J. , Yan, S. , Huang, K. , Li, Z. , Pan, W. , Tao, R. , Hao, J. , Zhu, B. , Tong, S. , & Tao, F. (2018). Association between serum arsenic levels and gestational diabetes mellitus: A population‐based birth cohort study. Environmental Pollution, 235, 850–856. 10.1016/j.envpol.2018.01.016 [PubMed] [CrossRef] [Google Scholar]
  • Xia, Y. , Wade, T. J. , Wu, K. , Li, Y. , Ning, Z. , Le, X. C. , He, X. , Chen, B. , Feng, Y. , & Mumford, J. L. (2009). Well water arsenic exposure, arsenic induced skin‐lesions and self‐reported morbidity in Inner Mongolia. International Journal of Environmental Research and Public Health, 6(3), 1010–1025. 10.3390/ijerph6031010 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Xie, H. , Huang, S. , Martin, S. , & Wise, J. P., Sr. (2014). Arsenic is cytotoxic and genotoxic to primary human lung cells. Mutation Research, Genetic Toxicology and Environmental Mutagenesis, 760, 33–41. 10.1016/j.mrgentox.2013.11.001 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Xiong, C. , Stiboller, M. , Glabonjat, R. A. , Rieger, J. , Paton, L. , & Francesconi, K. A. (2020). Transport of arsenolipids to the milk of a nursing mother after consuming salmon fish. Journal of Trace Elements in Medicine and Biology, 61, 126502. 10.1016/j.jtemb.2020.126502 [PubMed] [CrossRef] [Google Scholar]
  • Xu, H. , McClain, S. , Medina, S. , Lauer, F. T. , Douillet, C. , Liu, K. J. , Hudson, L. G. , Stýblo, M. , & Burchiel, S. W. (2016). Differential sensitivities of bone marrow, spleen and thymus to genotoxicity induced by environmentally relevant concentrations of arsenite. Toxicology Letters, 262, 55–61. 10.1016/j.toxlet.2016.09.008 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Xu, H. , Medina, S. , Lauer, F. T. , Douillet, C. , Liu, K. J. , Stýblo, M. , & Burchiel, S. W. (2017). Genotoxicity induced by monomethylarsonous acid (MMA+3) in mouse thymic developing T cells. Toxicology Letters, 279, 60–66. 10.1016/j.toxlet.2017.07.897 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Xu, L. , Liu, Y. , Zhao, Q. , Du, H. , Gao, Y. , Bai, M. , Lv, J. , Guo, Y. , Li, L. , Sun, L. , Lin, X. , Chen, Z. , Chen, Y. , & Zong, G. (2022). Urinary element profiles and associations with cardiometabolic diseases: A cross‐sectional study across ten areas in China. Environmental Research, 205, 112535. 10.1016/j.envres.2021.112535 [PubMed] [CrossRef] [Google Scholar]
  • Xu, L. , Mondal, D. , & Polya, D. A. (2020). Positive Association of Cardiovascular Disease (CVD) with chronic exposure to drinking Water arsenic (As) at concentrations below the WHO provisional guideline value: A systematic review and meta‐analysis. International Journal of Environmental Research and Public Health, 17. 10.3390/ijerph17072536 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Xu, L. , Polya, D. A. , Li, Q. , & Mondal, D. (2020). Association of low‐level inorganic arsenic exposure from rice with age‐standardized mortality risk of cardiovascular disease (CVD) in England and Wales. Sci Total Environ, 743, 140534. 10.1016/j.scitotenv.2020.140534 [PubMed] [CrossRef] [Google Scholar]
  • Xu, L. , Suman, S. , Sharma, P. , Kumar, R. , Singh, S. K. , Bose, N. , Ghosh, A. , Rahman, M. M. , Polya, D. A. , & Mondal, D. (2021). Assessment of hypertension association with arsenic exposure from food and drinking water in Bihar. India. Ecotoxicology and Environmental Safety, 223. 10.1016/j.ecoenv.2021.112572 [PubMed] [CrossRef] [Google Scholar]
  • Xu, L. , Yokoyama, K. , Tian, Y. , Piao, F. Y. , Kitamura, F. , Kida, H. , & Wang, P. (2011). Decrease in birth weight and gestational age by arsenic among the newborn in Shanghai. China. Nihon Koshu Eisei Zasshi (JAPANESE JOURNAL OF PUBLIC HEALTH), 58(2), 89–95. [PubMed] [Google Scholar]
  • Xu, S. , Hansen, S. , Sripada, K. , Aarsland, T. , Horvat, M. , Mazej, D. , Alvarez, M. V. , & Odland, J. O. (2022). Maternal Blood levels of toxic and essential elements and birth outcomes in Argentina: The EMASAR study. International Journal of Environmental Research and Public Health, 19. 10.3390/ijerph19063643 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Xu, W. , Bao, H. , Liu, F. , Liu, L. , Zhu, Y. G. , She, J. , Dong, S. , Cai, M. , Li, L. , Li, C. , & Shen, H. (2012). Environmental exposure to arsenic may reduce human semen quality: Associations derived from a Chinese cross‐sectional study. Environmental Health, 11(1), 1–9. [PMC free article] [PubMed] [Google Scholar]
  • Xu, X. , Drobná, Z. , Voruganti, V. S. , Barron, K. , González‐Horta, C. , Sánchez‐Ramírez, B. , Ballinas‐Casarrubias, L. , Cerón, R. H. , Morales, D. V. , Terrazas, F. A. , Ishida, M. C. , Gutiérrez‐Torres, D. S. , Saunders, R. J. , Crandell, J. , Fry, R. C. , Loomis, D. , García‐Vargas, G. G. , Del Razo, L. M. , Stýblo, M. , & Mendez, M. A. (2016). Association between variants in arsenic (+3 oxidation state) Methyltranserase (AS3MT) and urinary metabolites of inorganic arsenic: Role of exposure level. Toxicological Sciences, 153, 112–123. 10.1093/toxsci/kfw112 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yajima, I. , Ahsan, N. , Akhand, A. A. , Al Hossain, M. A. , Yoshinaga, M. , Ohgami, N. , Iida, M. , Oshino, R. , Naito, M. , Wakai, K. , & Kato, M. (2018). Arsenic levels in cutaneous appendicular organs are correlated with digitally evaluated hyperpigmented skin of the forehead but not the sole in Bangladesh residents. Journal of Exposure Science & Environmental Epidemiology, 28(1), 64–68. 10.1038/jes.2016.70 [PubMed] [CrossRef] [Google Scholar]
  • Yang, C. Y. , Chang, C. C. , & Chiu, H. F. (2008). Does arsenic exposure Iincrease the risk for prostate cancer? Journal of Toxicology and Environmental Health‐Part A‐Current Issues, 71(23), 1559–1563. [PubMed] [Google Scholar]
  • Yang, C. Y. , Chang, C. C. , Tsai, S. S. , Chuang, H. Y. , Ho, C. K. , & Wu, T. N. (2003). Arsenic in drinking water and adverse pregnancy outcome in an arseniasis‐endemic area in northeastern Taiwan. Environmental Research, 91(1), 29–34. [PubMed] [Google Scholar]
  • Yang, K. , Xun, P. , Carnethon, M. , Carson, A. P. , Lu, L. , Zhu, J. , & He, K. (2019). Low to moderate toenail arsenic levels in young adulthood and incidence of diabetes later in life: Findings from the CARDIA trace element study. Environmental Research, 171, 321–327. 10.1016/j.envres.2019.01.035 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yang, L. , Chai, Y. , Yu, J. , Wei, B. , Xia, Y. , Wu, K. , Gao, J. , Guo, Z. , & Cui, N. (2017). Associations of arsenic metabolites, methylation capacity, and skin lesions caused by chronic exposure to high arsenic in tube well water. Environmental Toxicology, 32(1), 28–36. 10.1002/tox.22209 [PubMed] [CrossRef] [Google Scholar]
  • Yao, X. , Steven, X. X. , Yang, Y. , Zhu, Z. , Zhu, Z. , Tao, F. , & Yuan, M. (2021). Stratification of population in NHANES 2009–2014 based on exposure pattern of lead, cadmium, mercury, and arsenic and their association with cardiovascular, renal and respiratory outcomes. Environment International, 149, 106410. 10.1016/j.envint.2021.106410 [PubMed] [CrossRef] [Google Scholar]
  • Yen, C.‐C. , Chen, H.‐H. , Hsu, Y.‐T. , Tseng, C.‐J. , & Lin, C.‐H. (2022). Effects of heavy metals in acute ischemic stroke patients a cross‐sectional study. Medicine, 101, e28973. 10.1097/md.0000000000028973 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yildiz, A. , Karaca, M. , Biceroglu, S. , Nalbantcilar, M. T. , Coskun, U. , Arik, F. , Aliyev, F. , Yiginer, O. , & Turkoglu, C. (2008). Effect of chronic arsenic exposure from drinking waters on the QT interval and transmural disper‐Sion of repolarization. The Journal of International Medical Research, 36, 471–478. [PubMed] [Google Scholar]
  • Yin, G. , Xia, L. , Hou, Y. , Li, Y. , Cao, D. , Liu, Y. , Chen, J. , Liu, J. , Zhang, L. , Yang, Q. , Zhang, Q. , & Tang, N. (2022). Transgenerational male reproductive effect of prenatal arsenic exposure: Abnormal spermatogenesis with Igf2/H19 epigenetic alteration in CD1 mouse. International Journal of Environmental Health Research, 32, 1248–1260. 10.1080/09603123.2020.1870668 [PubMed] [CrossRef] [Google Scholar]
  • Ying, S. , Myers, K. , Bottomley, S. , Helleday, T. , & Bryant, H. E. (2009). BRCA2‐dependent homologous recombination is required for repair of Arsenite‐induced replication lesions in mammalian cells. Nucleic Acids Research, 37(15), 5105–5113. 10.1093/nar/gkp538 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yorbik, Ö. , Kurt, İ. , Haşimi, A. , & Öztürk, Ö. (2009). Chromium, cadmium, and Lead levels in urine of children with autism and typically developing controls. Biological Trace Element Research, 135, 10–15. 10.1007/s12011-009-8494-7 [PubMed] [CrossRef] [Google Scholar]
  • Yorifuji, T. , Kato, T. , Ohta, H. , Bellinger, D. C. , Matsuoka, K. , & Grandjean, P. (2016). Neurological and neuropsychological functions in adults with a history of developmental arsenic poisoning from contaminated milk powder. Neurotoxicology and Teratology, 53, 75–80. 10.1016/j.ntt.2015.12.001 [PubMed] [CrossRef] [Google Scholar]
  • Yorifuji, T. , Tsuda, T. , Doi, H. , & Grandjean, P. (2011). Cancer excess after arsenic exposure from contaminated milk powder. Environmental Health Preventive Medicine, 16(3), 164–170. 10.1007/s12199-010-0182-x [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yoshinaga‐Sakurai, K. , Shinde, R. , Rodriguez, M. , Rosen, B. P. , & El‐Hage, N. (2020). Comparative cytotoxicity of inorganic Arsenite and Methylarsenite in human brain cells. ACS Chemical Neuroscience, 11(5), 743–751. 10.1021/acschemneuro.9b00653 [PubMed] [CrossRef] [Google Scholar]
  • Yu, H. S. , Liao, W. T. , & Chai, C. Y. (2006). Arsenic carcinogenesis in the skin. Journal of Biomedical Science, 13(5), 657–666. 10.1007/s11373-006-9092-8 [PubMed] [CrossRef] [Google Scholar]
  • Yu, H. Y. , & Zhang, K. L. (2011). Links between environmental geochemistry and rate of birth defects: Shanxi Province, China. Sci Total Environ, 409, 447–451. 10.1016/j.scitotenv.2010.10.019 [PubMed] [CrossRef] [Google Scholar]
  • Yu, Y. , Guo, Y. , Zhang, J. , Xie, J. , Zhu, Y. , Yan, J. , Wang, B. , & Li, Z. (2017). A perspective of chronic low exposure of arsenic on non‐working women: Risk of hypertension. Sci Total Environ, 580, 69–73. 10.1016/j.scitotenv.2016.11.204 [PubMed] [CrossRef] [Google Scholar]
  • Yuan, Y. , Marshall, G. , Ferreccio, C. , Steinmaus, C. , Liaw, J. , Bates, M. , & Smith, A. H. (2010). Kidney cancer mortality: Fifty‐year latency patterns related to arsenic exposure. Epidemiology, 21(1), 103–108. 10.1097/EDE.0b013e3181c21e46 [PubMed] [CrossRef] [Google Scholar]
  • Yuan, Y. , Xiao, Y. , Feng, W. , Liu, Y. , Yu, Y. , Zhou, L. , Qiu, G. , Wang, H. , Liu, B. , Liu, K. , Yang, H. , Li, X. , Min, X. , Zhang, C. , Xu, C. , Zhang, X. , He, M. , Hu, F. B. , Pan, A. , & Wu, T. (2017). Plasma metal concentrations and incident coronary heart disease in Chinese adults: The Dongfeng‐Tongji cohort. Environmental Health Perspectives, 125, 107007. 10.1289/ehp1521 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yuan, Y. , Xiao, Y. , Yu, Y. , Liu, Y. , Feng, W. , Qiu, G. , Wang, H. , Liu, B. , Wang, J. , Zhou, L. , Liu, K. , Xu, X. , Yang, H. , Li, X. , Qi, L. , Zhang, X. , He, M. , Hu, F. B. , Pan, A. , & Wu, T. (2018). Associations of multiple plasma metals with incident type 2 diabetes in Chinese adults: The Dongfeng‐Tongji cohort. Environmental Pollution, 237, 917–925. 10.1016/j.envpol.2018.01.046 [PubMed] [CrossRef] [Google Scholar]
  • Zeng, Q. , Zou, Z. , Wang, Q. , Sun, B. , Liu, Y. , Liang, B. , Liu, Q. , & Zhang, A. (2019). Association and risk of five miRNAs with arsenic‐induced multiorgan damage. Science of the Total Environment, 680, 1–9. 10.1016/j.scitotenv.2019.05.042 [PubMed] [CrossRef] [Google Scholar]
  • Zhai, Q. , Cen, S. , Jiang, J. , Zhao, J. , Zhang, H. , & Chen, W. (2019). Disturbance of trace element and gut microbiota profiles as indicators of autism spectrum disorder: A pilot study of Chinese children. Environmental Research, 171, 501–509. 10.1016/j.envres.2019.01.060 [PubMed] [CrossRef] [Google Scholar]
  • Zhang, C. , Kibriya, M. G. , Jasmine, F. , Roy, S. , Gao, J. , Sabarinathan, M. , Shinkle, J. , Delgado, D. , Ahmed, A. , Islam, T. , Eunus, M. , Islam, M. T. , Hasan, R. , Graziano, J. H. , Ahsan, H. , & Pierce, B. L. (2018). A study of telomere length, arsenic exposure, and arsenic toxicity in a Bangladeshi cohort. Environmental Research, 164, 346–355. 10.1016/j.envres.2018.03.005 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhang, C. , Mao, G. , He, S. , Yang, Z. , Yang, W. , Zhang, X. , Qiu, W. , Ta, N. , Cao, L. , Yang, H. , & Guo, X. (2013). Relationship between long‐term exposure to low‐level arsenic in drinking water and the prevalence of abnormal blood pressure. Journal of Hazardous Materials, 262, 1154–1158. 10.1016/j.jhazmat.2012.09.045 [PubMed] [CrossRef] [Google Scholar]
  • Zhang, Q. , Hou, Y. , Wang, D. , Xu, Y. , Wang, H. , Liu, J. , Xia, L. , Li, Y. , Tang, N. , Zheng, Q. , & Sun, G. (2020). Interactions of arsenic metabolism with arsenic exposure and individual factors on diabetes occurrence: Baseline findings from arsenic and non‐communicable disease cohort (AsNCD) in China. Environmental Pollution, 265, 114968. 10.1016/j.envpol.2020.114968 [PubMed] [CrossRef] [Google Scholar]
  • Zhang, Q. , Wang, D. , Zheng, Q. , Zheng, Y. , Wang, H. , Xu, Y. , Li, X. , & Sun, G. (2014). Joint effects of urinary arsenic methylation capacity with potential modifiers on arsenicosis: A cross‐sectional study from an endemic arsenism area in Huhhot Basin, northern China. Environmental Research, 132, 281–289. 10.1016/j.envres.2014.04.036 [PubMed] [CrossRef] [Google Scholar]
  • Zhang, Q. , Zhang, X. , Li, S. , Liu, H. , Liu, L. , Huang, Q. , Hou, Y. , Liang, X. , Cui, B. , Zhang, M. , Xia, L. , Zhang, L. , Li, C. , Li, J. , Sun, G. , & Tang, N. (2021). Joint effect of urinary arsenic species and serum one‐carbon metabolism nutrients on gestational diabetes mellitus: A cross‐sectional study of Chinese pregnant women. Environment International, 156, 106741. 10.1016/j.envint.2021.106741 [PubMed] [CrossRef] [Google Scholar]
  • Zhang, R. , Zhang, X. , Wu, K. , Wu, H. , Sun, Q. , Hu, F. B. , Han, J. , Willett, W. C. , & Giovannucci, E. L. (2016). Rice consumption and cancer incidence in US men and women. International Journal of Cancer, 138, 555–564. 10.1002/ijc.29704 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhang, Y. , Chen, T. , Zhang, Y. , Hu, Q. , Wang, X. , Chang, H. , Mao, J. H. , Snijders, A. M. , & Xia, Y. (2021). Contribution of trace element exposure to gestational diabetes mellitus through disturbing the gut microbiome. Environment International, 153, 106520. 10.1016/j.envint.2021.106520 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhang, Z. , Zhao, S. , Wu, H. , Qin, W. , Zhang, T. , Wang, Y. , Tang, Y. , Qi, S. , Cao, Y. , & Gao, X. (2022). Cross‐sectional study: Relationship between serum trace elements and hypertension. Journal of Trace Elements in Medicine and Biology, 69, 126893. 10.1016/j.jtemb.2021.126893 [PubMed] [CrossRef] [Google Scholar]
  • Zhao, G. , Liu, S. J. , Gan, X. Y. , Li, J. R. , Wu, X. X. , Liu, S. Y. , Jin, Y. S. , Zhang, K. R. , & Wu, H. M. (2022). Analysis of whole Blood and urine trace elements in children with autism Spectrum disorders and autistic behaviors. Biological Trace Element Research, 201, 627–635. 10.1007/s12011-022-03197-4 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhao, Y. N. , Li, M. , Tian, X. L. , Xie, J. X. , Liu, P. H. , Ying, X. D. , Wang, M. , Yuan, J. Y. , Gao, Y. , Tian, F. J. , & Yan, X. Y. (2021). Effects of arsenic exposure on lipid metabolism: A systematic review and meta‐analysis. Toxicology Mechanisms and Methods, 31, 188–196. 10.1080/15376516.2020.1864537 [PubMed] [CrossRef] [Google Scholar]
  • Zheng, L. Y. , Umans, J. G. , Yeh, F. , Francesconi, K. A. , Goessler, W. , Silbergeld, E. K. , Bandeen‐Roche, K. , Guallar, E. , Howard, B. V. , Weaver, V. M. , & Navas‐Acien, A. (2015). The association of urine arsenic with prevalent and incident chronic kidney disease: Evidence from the strong heart study. Epidemiology, 26, 601–612. 10.1097/ede.0000000000000313 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhong, Q. , Zhang, C. , Zhang, Q. , Jiang, C. X. , Qin, Q. R. , Chen, J. , Liang, L. , & Huang, F. (2019). Arsenic exposure and incident hypertension of adult residents living in rural areas along the Yangtze River, Anhui, China. Journal of Occupational and Environmental Medicine, 61, 271–277. 10.1097/jom.0000000000001511 [PubMed] [CrossRef] [Google Scholar]
  • Zhou, M. , Wang, X. , Yang, S. , Wang, B. , Ma, J. , Wang, D. , Guo, Y. , Shi, T. , & Chen, W. (2022). Cross‐sectional and longitudinal associations between urinary arsenic and lung function among urban Chinese adults. Science of the Total Environment, 844, 157028. [PubMed] [Google Scholar]
  • Zhou, M. , Zhao, E. , & Huang, R. (2022). Association of urinary arsenic with insulin resistance: Cross‐sectional analysis of the National Health and nutrition examination survey, 2015–2016. Ecotoxicology and Environmental Safety, 231, 113218. 10.1016/j.ecoenv.2022.113218 [PubMed] [CrossRef] [Google Scholar]
  • Zhou, X. , Ding, X. , Shen, J. , Yang, D. , Hudson, L. G. , & Liu, K. J. (2019). Peroxynitrite contributes to arsenic‐induced PARP‐1 inhibition through ROS/RNS generation. Toxicology and Applied Pharmacology, 378, 114602. 10.1016/j.taap.2019.114602 [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zierler, S. , Theodore, M. , Cohen, A. , & Rothman, K. J. (1988). Chemical quality of maternal drinking water and congenital heart disease. International Journal of Epidemiology, 17(3), 589–594. [PubMed] [Google Scholar]
  • Zierold, K. M. , Knobeloch, L. , & Anderson, H. (2004). Prevalence of chronic diseases in adults exposed to arsenic‐contaminated drinking water. American Journal of Public Health, 94, 1936–1937. [PMC free article] [PubMed] [Google Scholar]

Articles from EFSA Journal are provided here courtesy of Wiley

-