Arsenic: Handbook On The Toxicology of Metals 4E

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 44

C H A P T E R

28

Arsenic
BRUCE A. FOWLER, C.-H. SELENE J. CHOU, ROBERT L. JONES, DEXTER W. SULLIVAN JR,
AND C.-J. CHEN

ABSTRACT Biotransformation of inorganic arsenic has been


shown to occur in both animals and humans. Meth-
Arsenic in the environment occurs in both organic ylated compounds, such as methylarsonic acid and
and inorganic compounds in its trivalent or pentava- dimethylarsinic acid, have been detected in the urine
lent state. Certain fish and crustaceans contain very after ingestion or inhalation of inorganic arsenic.
high levels of organic arsenic, often as arsenobetaine. Reduction of arsenate and oxidation of arsenite in vivo
In most other foodstuffs, levels of arsenic are low have been demonstrated in experimental animals.
and the form is not known. The total daily intake of Recently, a human arsenic methyltransferase has been
arsenic in the general population is reported to be identified.
approximately a few tenths of a milligram, but var- Medications, contaminated food, beverages, and
ies to a great extent depending on the amount of fish drinking water have given rise to a number of episodes
consumed. of arsenic poisoning.
Both organic arsenic in seafood and inorganic arse- Inorganic arsenic-induced skin lesions such as der-
nic in water, beverages, and drugs have been shown matoses, which may include eruption, pigmentation,
to be readily absorbed (70-90%) by the gastrointestinal or leukodermal hyperkeratosis, may ultimately lead
tract. Some reports also indicate a fairly high degree of to the development of skin cancer and Bowen dis-
absorption after the inhalation of arsenic. ease. Effects on the nervous system (e.g. peripheral
Absorbed arsenic, irrespective of form, is widely nervous disturbance), as well as on the heart and cir-
distributed in the body. After exposure to inorganic culatory system (e.g. abnormal electrocardiograms,
arsenic, clearance of arsenic from the skin, upper gas- peripheral vascular disturbances with gangrene
trointestinal tract, epididymis, thyroid, and skeleton of the extremities, ischemic heart disease, cerebral
is slower than from other organs. The highest levels infarction, and erectile dysfunction), have also been
of arsenic in humans are normally found in the hair, reported after chronic exposure to inorganic arsenic.
nails, and skin. The main route of excretion is through Hematological changes after inorganic arsenic expo-
the kidneys. After ingestion of arsenite or arsenate, sure are characterized by anemia and leukopenia.
approximately 35% of the dose is excreted within 2 Chronic oral ingestion of inorganic arsenic in drink-
days. From animal experiments it seems that insoluble ing water has also been reported to cause internal
inorganic arsenic inhaled through the airway is depos- cancers (of the lung, bladder, kidney, and liver), dia-
ited and retained in lung tissue for a relatively long betes, hypertension, cataract, pterygium, and devel-
time. Animal data indicate arsenobetaine accumula- opmental retardation.
tion in cartilage, testes, epididymis, and muscle. Of Arsenic poisoning among industrial workers is
ingested arsenobetaine, 50-80% is excreted in the urine characterized by perforation of the nasal septum, skin
within 2 days. changes, and peripheral neuritis. There is substantial

Handbook on the Toxicology of Metals 4E


http://dx.doi.org/10.1016/B978-0-444-59453-2.00028-7 581 Copyright © 2015 Elsevier B.V. All rights reserved.
582 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

epidemiological evidence of an excessive risk of lung CAS Nos for the major arsenic compounds are:
cancer among workers exposed to arsenic. arsine, 7784-42-1; arsenic trioxide, 1327-53-3; sodium
Arsine gas is a powerful hemolytic poison encoun- arsenite, 7784-46-5; sodium arsenate, 7631-89-2; arsenic
tered under some industrial conditions. Arsine poison- pentoxide, 1303-28-2; monomethylarsenic acid, 124-58-
ing is characterized by nausea, vomiting, headache, 3; and dimethylarsinic acid, 75-60-5.
shortness of breath, and hemoglobinuria. Literature on
the toxicology and environmental aspects of arsenicals
has been reviewed by the World Health Organization 2  METHODS AND PROBLEMS OF
(WHO, 1981, 2001), in monographs by the Interna- ANALYSIS
tional Agency for Research on Cancer (IARC, 1982,
2004, 2006, 2012), Fowler (1983), and by the National Until a few decades ago, the only available meth-
Research Council (NRC) (1999). ods for analyzing arsenic, such as Reinsch’s method,
Marsh’s test, and Gutzeit’s test, were qualitative rather
than quantitative in nature. Because of this limitation,
1  PHYSICAL AND CHEMICAL PROPERTIES the results of studies based on these methods must be
evaluated with caution.
Arsenic [As; Chemical Abstracts Service (CAS) No. The molybdenum blue method and the silver dieth-
7440-38-2 for metallic arsenic]: atomic weight, 74.9; yldithiocarbamate method are two reasonably good
atomic number, 33; metalloid. Allotropes: crystal- quantitative colorimetric methods that have limits
line (hexagonal-rhombic) gray arsenic (stable), den- of detection in the range of 1-50 μg/L in a 5-mL solu-
sity 5.727 g/cm3 (25°C/4°C), melting point, 818°C tion (Sandell, 1959; Vasak and Sedivec, 1952). Atomic
(36 atm), boiling point 615°C (sublimate), vapor pres- absorption spectrophotometric (AAS) measure-
sure 1 mmHg (372°C); yellow arsenic (metastable), ment of generated arsine gas (Holak, 1969) has been
density 1.97 g/cm3, melting point 815°C (under addi- widely used for the determination of total arsenic and
tional pressure); and black arsenic, valence states has a detection limit of 0.04 μg. Atomic fluorescence
−3, 0, +3, and +5. When elemental arsenic is heated spectrometry (AFS) with online hydride generation
at atmospheric pressure without oxygen, it sublimes (Karthiheyan and Hirata, 2003) has the advantage
without melting and forms a yellow gas. Heating of less scattering and matrix effects and has a detec-
arsenic in air will yield a white smoke consisting of tion limit of 0.03 μg/L. Neutron activation analysis
arsenic trioxide. (NAA) has a detection limit of 0.1 ng for total arsenic
From both the biological and the toxicological (Liebscher and Smith, 1968). Proton-induced X-ray
points of view, arsenic compounds may be classi- emission analysis (PIXEA), with a detection limit of
fied into three major groups: (1) inorganic arsenic 0.1 mg/kg, has been used for the simultaneous deter-
compounds; (2) organic arsenic compounds; and (3) mination of arsenic and a number of other elements
arsine gas. The toxicology of this last compound, in biological samples (Fowler et al., 1975; Walter et al.,
which has special properties, will be dealt with sepa- 1974). Advancements in inductively coupled plasma
rately in Section 10 of this chapter. The most common spectroscopy (ICP) have resulted in the general use
inorganic trivalent arsenic compounds are arsenic tri- of either ICP-coupled optical emission spectroscopy
oxide, sodium arsenite, and arsenic trichloride. Pen- (ICP-OES) or mass spectroscopy (ICP-MS) detection
tavalent inorganic compounds are arsenic pentoxide, methods. Although the ICP-OES method showed
arsenic acid, and arsenate (e.g. lead arsenate and cal- promise (Hueber and Winefordner, 1995), it has been
cium arsenate). Common organic arsenic compounds largely replaced with the ICP-MS method, with its
are arsanilic acid, methylarsonic acid, dimethylar- higher sensitivity, less interferences than ICP-OES,
sinic acid (cacodylic acid), and arsenobetaine (NRC, and detection limits of 0.002 μg/L (Marawi et al., 1994).
1999). The primary drawback of ICP-MS is the generation of
Arsenic trioxide is only slightly soluble in water; Ar-Cl interference in the plasma, which can be over-
in sodium hydroxide it forms arsenite, and with come by either high-resolution ICP-MS (Townsend,
concentrated hydrochloric acid it forms arsenic tri- 1999) or the use of a dynamic reaction cell (Nixon et al.,
chloride. Sodium arsenite and sodium arsenate are 2004; Tanner et al., 2000; Jarrett et al., 2007) or collision
highly soluble in water. Interchanges in valence state cell (B’Hymer and Caruso, 2004; Nakazato et al., 2002)
may occur in water solutions, depending on the pH technology. ICP-MS is the most widely used method
of the solution, as well as the presence of other sub- for the determination of total arsenic (Hung et al.,
stances that can be reduced or oxidized (Reay and 2004). All of these methods are suitable for the mea-
Asher, 1977). surement of total arsenic.
28 Arsenic 583

Several methods for the quantitative determination 3  PRODUCTION AND USES


of the various valence states and chemical forms of
arsenic have also been developed. Pulse polarography 3.1 Production
(detection limit of 0.3 μg As/L water) is capable of dis-
tinguishing polarographically active As(III) from inac- Arsenic is widely distributed in the Earth’s crust,
tive As(V) (Myers and Osteryoung, 1973). A method to which contains about 3.4 ppm arsenic (Wedepohl,
collect generated arsines in a cold trap, evaporate them 1991). It usually exists in nature in sulfide ores.
at different temperatures, and measure them by atomic Although there are more than 150 arsenic-bearing
emission spectroscopy has been found to have a detec- minerals (Budavari et al., 2001; Carapella, 1992),
tion limit of the order of 1 ng (Braman and Foreback, arsenopyrite is by far the most common. Arsenic
1973). With this method, it is possible to distinguish trioxide (white arsenic) is principally obtained as a by-
As(III), As(V), methylarsonic acid, and dimethylarsinic product of smelting copper, lead, or gold ores. When
acid (DMA). these ores are smelted, the arsenic becomes gaseous
The speciation of arsenic in water and other matri- and is trapped by electrostatic precipitators as a crude
ces such as urine has seen major advances because dust that is then roasted, whereby arsenic trioxide is
of coupled techniques like ion chromatography (IC) driven off. Purified arsenic trioxide is collected in a
coupled to ICP (Sheppard et al., 1992). In this tech- cooling chamber, and the principal impurity is Sb2O3
nique, IC separates the species of arsenic before their (Pinto and McGill, 1953). Most commercially avail-
introduction into the ICP-MS detector (or other detec- able arsenic compounds are produced from arsenic
tion method). This enables the automation of specia- trioxide. The average annual world production of
tion method(s) for the different matrices. The most arsenic on the basis of limited data (1975-1977) was
commonly used method for speciation of the various approximately 60,000 tons, and this level of produc-
arsenic species is high-performance liquid chroma- tion seems to be stable (WHO, 1981). In 2003, China
tography (HPLC) in its various forms. Arsenic(III), was the world leader in the production of com-
As(V), monomethylarsonic acid (MMA), DMA, and mercial-grade arsenic, followed by Chile and Peru
arsenobetaine are separated by HPLC and determined (Brooks, 2003). The United States is the world’s lead-
online by ICP-MS. Two forms of HPLC may be used, ing consumer of arsenic; however, the United States
ion pairing and ion exchange, with absolute detec- ceased production in 1985.
tion limits for arsenic ranging between 50 and 300 pg
(Beauchemin et al., 1989). An ICP-MS detector in com-
3.2 Uses
bination with HPLC has been used for the analysis of
six arsenic compounds, with detection limits for arse- Arsenic was known as a therapeutic agent as early
nic species in the range of 10-30 pg. (Demesmay et al., as 400 bc and has been widely used as such since then.
1994). A more recent analytical method has been devel- From the nineteenth century onward, an inorganic
oped using HPLC-ICP-DRC (i.e Dynamic Reaction arsenic compound known as Fowler’s solution (liquor
Cell)-MS for biomonitoring the U.S. population that arsenicalis B.P. 1963; a potassium arsenite solution con-
enables the quantification of seven species of arsenic: taining 7.6 g As/L) has been used for the treatment of
As(III), As(V), MMA, DMA, arsenobetaine, and tri- leukemia, psoriasis, and chronic bronchial asthma,
methylarsine oxide (TMAO), with limits of detection and organic arsenic compounds have been extensively
for the various species ranging from 0.4 to 1.7 μg/L used in the treatment of spirochetal and protozoal dis-
(Verdon et al., 2009). ease (Martindale, 1977; NRC, 1999). However, many
More recent publications on the speciation of countries have now banned the use of Fowler’s solu-
arsenicals have taken advantage of advances in tion. Recently, arsenic trioxide has reportedly been
sample preparation methods for ICP-MS technolo- used in the treatment of acute promyelocytic leukemia
gies. These advances have permitted measurements (Gallagher, 1998; Wang, 2001). Recent studies have
of speciated arsenicals in air (Lewis et al., 2012), sea- reported that As3+ produces anticancer effects in leu-
food (Moreda-Piñeiro et al., 2010, 2011), urine (Davis kemic cells (Goussetis et al., 2010; Zheng et al., 2005)
et al., 2010; Chen et al., 2012), and blood (Nunes et al., and human glioblastoma cells (Pucer et al., 2010), and
2010). induces autophagy and apoptosis in human fibrosar-
Methods and problems of arsenic analysis, includ- coma (Chiu et al., 2010) and ovarian cancer cells (Smith
ing speciation in various matrices, have been reviewed et al., 2010). Altered gene regulation patterns are associ-
by Braman (1977, 1983), Hung et al. (2004), Muñoz ated with decreased cell growth in As3+-exposed human
and Palmero (2005), Gong et al. (2002), and Rose et al. liver cancer (Yoo et al., 2009), bladder cancer (Jutooru
(2001). et al., 2010), and breast cancer (Zhang et al., 2011) cells.
584 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

The major current uses of arsenic are in pesticides infant foods (Burlo et al, 2012; Carbonell-Barrachina
(e.g. lead arsenate, calcium arsenate, and sodium arse- et al., 2012), particularly for infants with celiac disease
nite), herbicides [e.g. monosodium arsenate and caco- who must consume gluten-free foodstuffs. For the gen-
dylic acid (i.e. DMA)]; cotton desiccants (e.g. arsenic eral population, a shift in a rice-based diet to one with
acid), and wood preservatives (e.g. copper chromium a lower arsenic content based on grains such as maize,
arsenate). Arsenic is also used as a bronzing or decol- sorghum, or millet (Adomako et al., 2011) may reduce
orizing agent in the manufacture of glass and in the dietary exposure to this element. Please see Chapter 6 for
fabrication of opal glass and enamels. It has also been a more complete discussion of arsenic in food.
used in the manufacture of dyestuffs and chemical Arsenic concentrations in marine organisms and
warfare gases, and is still used in the purification of seaweed are, in general, considerably higher than
industrial gases (sulfur removal). Elemental arsenic is those in other foods. Many species of bony fish con-
used as an additive in the production of several alloys tain between 1 and 10 mg/kg, whereas certain bottom-
to increase hardness and heat resistance. In the live- feeding fish, crustaceans, and seaweed may contain
stock industry, organic arsenical is sometimes added more than 100 mg As/kg (Lunde, 1973). Both lipid-
to swine and poultry feed as an antimicrobial medi- soluble and water-soluble organic arsenic compounds
cine. In 1999-2000, approximately 70% of the broiler have been isolated from marine organisms (Kurosawa
industry added roxarsone to broiler poultry feed et al., 1980; Lunde, 1975; Penrose et al., 1977). Certain
(Garbarino et al., 2003). Recently, artificial gallium algae grown in the presence of arsenate may contain
arsenide and indium arsenide crystals have become arsenophospholipids (Cooney et al., 1978; Irgolic et al.,
important materials in semiconductors, solar cells, and 1977), whereas other researchers have shown that
materials used for space research (Brooks, 2005; IARC, brown kelp contains mainly arsenosugars (Edmonds
2006; Yamauchi et al., 1992). and Francesconi, 1981a). Arsenosugars are also found
in mussels, oysters, and clams (Le et al. 2004). Anaero-
bic decomposition of the brown kelp produces dimeth-
yloxarsylethanol, a possible precursor of arsenobetaine
4  ENVIRONMENTAL LEVELS AND
(Edmonds et al., 1982). Arsenobetaine, the arsenic ana-
EXPOSURES
log of betaine, seems to be the most commonly occur-
ring water-soluble arsenical in fish and Crustacea
4.1  Food and Daily Intake
(Cannon et al., 1981; Edmonds and Francesconi, 1981b;
With the exception of some kinds of fish, crusta- Edmonds et al., 1977; Kurosawa et al., 1980; Luten
ceans, and seaweed, most food contains low levels et al., 1982). Indications of the presence of arsenocho-
of arsenic, normally below 0.25 mg/kg (Jelinek and line in some species have also been reported (Norin
Corneliussen, 1977). The degree of arsenic uptake and Christakopoulos, 1982). Arsenobetaine does not
in plants appears to be related to the concentration seem to be harmful to humans and is excreted rapidly
of soluble arsenic in the soil, the chemical composi- and unchanged in urine (Cullen, 1998).
tion of the soil, and the plant species (Walsh et al., The amount of arsenic ingested daily by humans
1977). Surface contamination with insecticides may through food is greatly influenced by the amount of
increase the concentration of arsenic in plants. Exten- seafood in the diet. Jelinek and Corneliussen (1977)
sive lists of arsenic concentrations in foodstuffs have noted that foods belonging to the meat, fish, and poul-
been published by the National Academy of Sciences try groups contributed most of the arsenic ingested.
National Research Council (NRC, 1999). However, all They estimated the average daily intake of arsenic
of the chemical forms of arsenic present in food have from food in the United States in the early seventies to
not yet been established. Adair et al. (2006) reported be 0.01-0.02 mg. A duplicate dietary study from Japan
two chemical and enzymatic extraction methods fol- (Mohri et al., 1990) reported daily dietary intake of
lowed by IC-ICP-MS analysis for the analysis of DMA arsenic in four volunteers to be between 27 to 376 μg,
and inorganic arsenic from rice cooked in arsenic- with a mean of 182 μg/day primarily as the methylate
contaminated water. Measured concentrations of arsenic species.
DMA ranged from 22 to 270 ngAs/g rice and of inor- Schoof and colleagues (1999b) reported the analy-
ganic arsenic ranged from 31 to 108 ngAs/g rice. sis of 40 commodities accounting for 90% of dietary
There is growing concern about the uptake of inorganic arsenic intake. Average inorganic arsenic in
­arsenic into certain plants (such as rice) from ­arsenic- seafood ranged from < 1 to 2 ng/g. The highest inor-
contaminated irrigation water, and this topic is consid- ganic arsenic concentrations were found in raw rice
ered in more detail in Chapter 6, “Toxic Metals in Food.” (74 ng/g), followed by flour (11 ng/g), grape juice
This has been noted as a special problem for rice-based (9 ng/g), and cooked spinach (6 ng/g). Schoof et al.
28 Arsenic 585

(1999a) estimated that intake of inorganic arsenic in the DMA—with the methylated forms generally in lower
U.S. diet ranges from 1 to 20 μg/day, with a mean of concentrations than the inorganic ones. Andreae (1978)
3.2 μg/day. Nriagu and Lin (1995) analyzed 26 brands reported that, in general, arsenate is the dominant form
of wild rice sold in the United States and found arsenic in seawater. Clement and Faust (1973) found that only
levels ranging from 0.006 to 0.142 μg/g dry weight. At approximately 8% of the total arsenic in well-aerated
a mean level of chicken consumption of 60 g/­person/ stream water was in the form of As(III), whereas all
day, Laskey et al. (2004) estimated that people may of the arsenic in anaerobic reservoirs seemed to be in
ingest 1.38-5.24  μg/day of inorganic arsenic from the form of As(III). The chemical form of arsenic in dif-
chicken. ferent groundwaters is largely unknown. Clement and
Wine made from grapes sprayed with arsenic-­ Faust (1973) found that 25-50% of the total arsenic in a
containing insecticides may contain high levels of few groundwater samples was in the form of As(III).
­arsenic. Levels of up to 0.5 mg/L have been reported, The average daily intake of arsenic through drink-
with most of the arsenic being in the trivalent inorganic ing water can vary widely, depending on the source of
form (Crecelius, 1977a). Zoeteman and Brinkmann the water. McCabe et al. (1970) reported that less than
(1976) reported a mean concentration of 0.02 mg/L 1% of more than 18,000 community water supplies in
(range 0.001-0.19 mg/L) in bottled mineral water. The the United States had concentrations exceeding 0.01 mg
arsenic in mineral water is probably of natural origin. As/L. Engel and Smith (1994) investigated the levels of
arsenic in drinking water throughout the United States
between 1968 and 1984. They found that 30 counties
4.2 Water
in 11 states had mean arsenic levels > 5 μg/L, 15 coun-
Arsenic is widely distributed in surface water, ties had mean levels of 5-10 μg/L, 10 counties had
groundwater, and finished drinking water. Seawa- mean levels of 10-20 μg/L, and 5 counties had levels
ter generally contains 0.001-0.005 mg As/L (Ferguson > 20 μg/L. The highest levels were found in Churchill
and Gavis, 1972). The arsenic concentration of rivers County, Nevada, where 89% of the population was
and lakes varies considerably: most levels are well exposed to a mean arsenic concentration of 100 μg/L
below 0.01 mg/L, but in some instances they may even and 11% to a mean of 27 μg/L. More recently, inves-
be as high as about 1 mg/L (Andreae, 1978; Durum tigators at the U.S. Geological Survey (Focazio et al.,
et al., 1971; Sagner, 1973). A survey of 293 stations in 2000) have developed an extensive map of the occur-
two nationwide sampling networks on major U.S. rence of arsenic in groundwater in the United States on
rivers found median arsenic levels to be 1 μg/L; the the basis of 18,850 sample locations; 2262 of these were
­75th-percentile level was 3 μg/L (Smith et al., 1987). identified as public water supplies. The results of these
The natural concentration of total arsenic in ground- studies indicate a number of states in specific regional
water depends on the arsenic content of the bedrock. areas (e.g. New England, the Midwest, the Rocky
Arsenic levels in groundwater average about 1-2 μg/L, Mountain states, and the Pacific Coast) with elevated
except in some western states of the USA with vol- concentrations (> 10.0 μg/L) in the groundwater sup-
canic rock and sulfide mineral deposits high in arse- plies; therefore, the issue of elevated arsenic concentra-
nic, where arsenic levels up to 3.4 mg/L have been tions in groundwater is national in scope. In January
observed (Robertson, 1989; Welch et al., 1988). Approx- 2001, the U.S. Environmental Protection Agency pro-
imately 13% of groundwater samples from 800 wells posed lowering the Maximum Contaminant Level for
in an area in Nova Scotia, Canada, where the arsenic arsenic from 50 to 10 μg/L (EPA, 2001), with a compli-
content of the bedrock is high, had concentrations ance date of 23 January, 2006 (http://water.epa.gov/
exceeding 0.05 mg As/L (Grantham and Jones, 1977). lawsregs/rulesregs/sdwa/arsenic/index.cfm).
Arsenic was detected in 1298 out of 3452 surface water With an assumed daily intake of 1.5 L drinking
samples recorded in the STORET database for 2004 at water, a concentration of 0.01 mg/L will result in the
concentrations ranging from 0.138 to 1700 μg/L (EPA, daily ingestion of 0.015 mg arsenic.
2005a). In Japan, concentrations of up to 1.7 mg As/L Some developing countries, such as Mexico, Bangla-
have been recorded in hot-spring water (Kawakami, desh, India, and Vietnam, have highly elevated levels
1967). In Cordoba, Argentina, groundwater levels of of arsenic in drinking water in some regions (Bagla
up to 3.4 mg As/L have been reported (Arguello et al., and Kaiser, 1996; Berg et al., 2001; Tondel et al., 1999;
1938). In Taiwan, artesian well water has been shown Wyatt et al., 1998a,b). In Bangladesh and West Bengal,
to contain up to 1.8 mg/L (Kuo, 1968). India, it is estimated that more than 1 million people
Braman and Foreback (1973) and Crecelius (1977a,b) are drinking arsenic-contaminated water and tens of
noted several different forms of arsenic in natural millions more could be at risk in areas that have not
waters—arsenate, arsenite, methylarsonic acid, and been tested for contamination. Analysis of 20,000
586 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

tube-well waters indicated that 62% have arsenic lev- from 14 to 17 μg/g throughout the study area, which is
els above the WHO permissible exposure limit (PEL) within the range of natural background levels.
of 10 μg/L, with some as high as 3700 μg/L (Bagla and
Kaiser, 1996). Arsenic concentrations in water samples
4.4 Air
from private small-scale tube-wells in Hanoi, Vietnam,
averaged 159 μg/L, ranging from 1 to 3050 μg/L (Berg Arsenic naturally occurs in soil and is present in
et al., 2001). the atmosphere as airborne dust. It is also emitted
from volcanoes and is found in areas of dormant vol-
canism. Arsenic naturally occurs in seawater and veg-
4.3 Soil
etation, and is released into the atmosphere through
Arsenic is widely distributed in the Earth’s crust, sea salt spray and forest fires (ATSDR, 2007). Smelting
which contains about 3.4 ppm (Wedepohl, 1991). It of nonferrous metals, burning of coal, oil and wood
is mostly found in nature as minerals, and only to combustion, and the use of arsenicals as pesticides are
a small extent in its elemental form. Typical arsenic the major anthropogenic sources of airborne arsenic.
concentrations for uncontaminated soils range from Chilvers and Peterson (1987) estimated global natu-
1 to 40 ppm (μg/g), with the lowest concentrations ral and anthropogenic arsenic emissions to the atmo-
found in sandy soils and soils derived from granites. sphere to be 73,500 and 28,100 metric tons per year,
Higher arsenic concentrations are found in alluvial respectively. Copper smelting and coal combustion
soils and soils with a high organic content (Mandal accounted for 65% of anthropogenic emissions. There
and Suzuki, 2002). is evidence that anthropogenic emissions from smelt-
Soils in mining areas or near smelters may con- ers are lower now than they were in the 1980s. The
tain high levels of arsenic. Arsenic concentrations up National Air Toxics Assessment reported the total
to 27,000 mg/kg were reported in soils contaminated anthropogenic emissions of arsenic compounds in
with mine or smelter wastes (EPA, 1982). Soils at an the United States in 1996 to be 355 tons/year (EPA,
abandoned mining site in the Tamar Valley in south- 2005b).
west England have arsenic concentrations that may Arsenic in ambient air is usually a mixture of
exceed 50,000 mg/kg (Erry et al., 1999). particular arsenite and arsenate (EPA, 2012). Mean
Soil on agricultural lands treated with arsenical levels in ambient air in the United States have been
pesticides may retain substantial amounts of arsenic. reported to range from < 1 to 3 ng/m3 in remote areas
A study reported an arsenic concentration of 22 mg/ and from 10 to 20 ng/m3 in urban areas. The 133 sta-
kg in treated soil compared with 2 ppm in nearby tions of the U.S. National Air Sampling Network
untreated soil (EPA, 1982). Soil samples from 13-year- reported in 1964 that the annual average concentra-
old fruit orchards in New York State, where lead arse- tions of arsenic in air ranged from nondetectable
nate had been used for pest control for many years, (< 0.01 μg/m3) to 0.75 μg/m3 (Sullivan, 1969). The
had arsenic concentrations of 1.6-141 mg/kg (Merwin overall average was approximately 0.02 μg/m3. The
et al., 1994). highest value (1.4 μg/m3) was a quarterly average
Natural concentrations of arsenic in sediments are in El Paso, Texas, where a large copper smelter is
usually < 10 μg/g dry weight, but can vary widely located. Daily mean concentrations of up to 1.6 μg/
around the world (Mandal and Suzuki, 2002). Contam- m3 have been found near a smelter in Romania
ination by heavy metals is a serious problem in some (Gabor and Coldea, 1977).
developing countries. Sepetiba Bay, a semi-enclosed Dousova and colleagues (2007) reported arse-
coastal lagoon in Brazil, had sediment arsenic concen- nic concentrations in ice and snow samples from the
trations up to 80,000 μg/g in an area adjacent to a plant Czech-Polish border taken for the two periods, 1984-
that produced zinc and cadmium (Moreira, 1996). The 1986 and 2003-2005. They showed declines from
literature regarding the cycling and effects of arsenic 50 μg/L to 3 μg/L for ice and from 15 μg/L to < 2 μg/L
in coastal marine environments has been reviewed by for snow. Inorganic arsenic, primarily as arsenic triox-
Sanders et al. (1994). ide, has been reported from fossil fuel and industrial
It has recently been suggested that the wood pre- processes (Pacyna, 1987). Arsenic trisulfide has also
servative, chromated copper arsenate, commonly used been reported from coal combustion, organic arsines
in dock pilings and bulkheads, can be toxic to estua- from oil combustion, and arsenic trichloride from
rine organisms. Wendt et al. (1996) measured arsenic refuse incineration.
in surface sediments from creeks with high densities of Measurements of arsenic concentrations in cases
docks and from nearby reference creeks with no docks. of occupational exposure have been rare. Patty (1962)
The average concentrations in the sediments ranged reported that the highest exposure in insecticide
28 Arsenic 587

manufacturing was usually found in the mixing, 2006a). For arsine, the OSHA PEL is 0.05 mgAs/m3
screening, drying, bagging, and drum-filling opera- (TWA). The ACGIH TLV for arsine is 0.05 mg/m3
tions. He reported concentrations of arsenic in air (NIOSH, 2006b).
ranging from 0.5 to 45 mg/m3 during these operations.
High levels of exposure to arsenic fumes and dusts 4.5 Tobacco
may occur in the smelting industries. Lundgren (1954)
reported an average concentration ranging between Arsenic in tobacco originates from insecticides,
0.06 and 2 mg/m3 in a Swedish copper smelter. In a especially lead arsenate. In the past, the arsenic con-
Japanese copper refinery, arsenic concentrations in tent of various brands of American cigarettes averaged
the air of nine workshops were measured at 0.006- 12.6 μg/cigarette in 1932-1933 and increased to 42 μg/
0.011 mg/m3 under normally ventilated conditions cigarette in the early 1950s (Satterlee, 1956). After the
and 0.08-0.19 mg/m3 under unventilated conditions. ban of arsenical pesticides, maximum arsenic levels
Average arsenic concentrations in the air of 0.001- were reduced to 3 μg/g (Kraus et al., 2000). An inter-
0.012 mg/m3 were found around furnaces in copper national literature survey reports arsenic emissions of
and ferronickel smelters (Kodama et al., 1976). 0-1.4 μg/cigarette from mainstream (inhaled) cigarette
When airborne arsenic in a U.S. copper smelter was smoke (Smith et al., 1997). Arsenic emissions of 0.015-
measured during 1943-1965, 8-h average levels in the 0.023 μg/cigarette have been measured for sidestream
arsenic roaster, arsenic refinery, and main flue ranged smoke from a burning cigarette (Landsberger and Wu,
between 6.9 and 20 mg/m3 (Welch et al., 1982). Enter- 1995). In Japan, cigarettes have been reported to con-
line and Marsh (1982) summarized some reports of tain less than 1 μg/g (Maruyama et al., 1970).
airborne arsenic concentrations in the area around an
arsenic plant in the United States for the period 1938-
1957. They found that levels varied but were very high, 5 METABOLISM
particularly before 1951 (e.g. 0.8-62.4 mg/m3 in 1938).
This section on arsenical metabolism is focused on
All levels were very high in relation to the current PEL
historical descriptive in vivo information related to
of 0.01 mg/m3.
biological monitoring of arsenic in humans and ani-
Substantial exposure to airborne arsenic may also
mal models. Section 5.6.1 reviews more mechanistic
take place in individuals working with arsenic-con-
studies of arsenical metabolism and the relationships
taining insecticides and wood preservatives. Han-
between the formation of methylated arsenic species
dling, including burning, of preserved wood could
and mechanisms of toxicity at the cellular and molecu-
also constitute a risk for the general population
lar levels of biological organization.
(Peters et al., 1984). Nygren et al. (1992) investigated
occupational exposure to airborne dust, chromium,
copper, and arsenic in six joinery shops in Sweden, 5.1 Absorption
where impregnated wood was used for production.
5.1.1 Inhalation
The mean airborne concentration of arsenic around
various types of joinery machines ranged from 0.54 to Airborne arsenic is usually in the form of As2O3.
3.1 μg/m3. More than 23% of the particles (by particle count) in
Workers at three microelectronics factories where samples of arsenic-polluted air were reported to be
gallium arsenide was used have been reported to larger than 5.5 μm (Pinto and McGill, 1953). Particles of
have exposure at or above the Occupational Safety this nature will undergo a high rate of deposition in the
and Health Administration (OSHA) action level for upper respiratory tract. As a result of deposition in the
arsenic of 5 μg/m3, with a maximum exposure of nasopharyngeal region and mucociliary clearance in
8.2 μg/m3 (Sheehy and Jones, 1993). The toxicologi- the airways, much of the inhaled arsenic may be swal-
cal literature and carcinogenic potential of gallium lowed and then absorbed from the gastrointestinal
arsenide (GaAs) have been recently reviewed by the tract. Analytical data on fly ash emitted from a coal-
International Agency for Research on Cancer (IARC, fired power plant have indicated a pronounced con-
2006), and GaAs was identified as a Class I human centration of arsenic in particles of 1.1-2.1-μm diameter
carcinogen on the basis of the inorganic arsenic (Davison et al., 1974).
component. Very few data are available on the deposition and
The current OSHA PEL is 0.010 mgAs/m3 [time- absorption of inhaled arsenic in humans. In eight ter-
weighted average (TWA)]. The American Conference of minal lung cancer patients, Holland et al. (1959) esti-
Governmental Industrial Hygienists (ACGIH) thresh- mated deposition to be approximately 40%, of which
old limit value (TLV) is 0.01 mgAs/m3 (TWA) (NIOSH, 75-85% was absorbed; thus, the overall absorption
588 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

(expressed as a percentage of inhaled arsenic) was dose-dependent effects of methylated arsenic species
approximately 30-34%. However, great caution must be in humans. Organic arsenic compounds in seafoods
exercised when relating these data to healthy humans. are also readily absorbed (< 80%) from the gastrointes-
In workers exposed to arsenic trioxide dusts in smelt- tinal tract in both animals and humans (Charbonneau
ers, the amount of arsenic excreted in the urine (the et al., 1978; Marafante et al., 1984; Munro, 1976; Tam
main route of excretion) was approximately 40-60% of et al., 1982; Vahter et al., 1983). On the basis of uri-
the estimated inhaled dose (Pinto et al., 1976; Vahter nary excretion studies in volunteers, it seems that both
et al., 1986). Absorption of arsenic trioxide dusts and MMA and DMA are well absorbed (by at least 75-85%)
fumes (assessed by the measurement of urinary metab- across the gastrointestinal tract (Buchet et al., 1981a).
olites) correlated with TWA arsenic air concentrations Animal experiments indicate that growth promoters,
from personal breathing zone air samplers (Offergelt such as arsanilic acid, are absorbed to only 15-40%
et al., 1992). In contrast, arsenic sulfide and lead arse- (Calvert, 1975).
nate were cleared slowly (Marafante and Vahter, 1987), In the case of arsenic trioxide, which is only slightly
indicating that the rate of absorption may be lower if soluble in water, gastrointestinal absorption depends
the inhaled arsenic is in a highly insoluble form. on factors such as particle size and the pH of the gas-
tric juice.
5.1.2 Ingestion
5.1.3  Skin Absorption
Both human and animal data indicate that more
than 90% of an ingested dose of dissolved inorganic Systemic toxic effects have resulted from occupa-
trivalent or pentavalent arsenic is absorbed from the tional accidents in which arsenic acid or arsenic tri-
gastrointestinal tract (Bettley and O’Shea, 1975; Char- chloride has been splashed onto workers, indicating
bonneau et al., 1978; Crecelius, 1977b; Marafante et al., that the skin is a possible route for arsenic absorption
1981; Pomroy et al., 1980; Vahter and Norin, 1980). The (Garb and Hine, 1977). However, no quantitative stud-
most direct evidence is from a study that evaluated the ies have been located on the absorption of inorganic
6-day elimination of arsenic in healthy humans given arsenicals in humans after dermal exposure. Percu-
water from a high-arsenic sampling site; it reported taneous absorption of 73As as arsenic acid has been
approximately 95% absorption (Zheng et al., 2002). In measured in skin from cadavers (Wester et al., 1993).
contrast, ingestion of arsenic triselenide did not lead Labeled arsenic was applied to skin in diffusion cells,
to a measurable increase in urinary excretion (Mappes, and transit through the skin into receptor fluid mea-
1977), indicating that gastrointestinal absorption may sured. After 24 h, 0.93% of the dose passed through the
be much lower when highly insoluble forms of arse- skin, and 0.98% remained in the skin after washing.
nic are ingested. Recent studies (Calatayud et al., 2012) Dermal absorption of arsenic has been measured in
have implicated intestinal transporters in the absorp- Rhesus monkeys (Wester et al., 1993). After 24 h, 6.4%
tion of inorganic arsenic species, and help to explain of 73 As as arsenic acid was absorbed systemically.
on a mechanistic level the relatively high absorption
of inorganic arsenicals from the gastrointestinal tract
5.2  Transport and Distribution
noted above. Other studies (Drobna et al., 2010) of arse-
nic membrane transporters in the human liver found After absorption by the lungs or the gastrointestinal
that the retention of inorganic arsenic and methylated tract, arsenic is transported by the blood to other parts
metabolites in human hepatocytes exposed to submi- of the body. After exposure to arsenite or arsenate,
cromolar concentrations of As3+ was negatively associ- most arsenic is cleared from the blood (Charbonneau
ated with expression of the Canalicular multispecific et al., 1978; Marafante et al., 1981; Mealey et al., 1959).
organic anion transporter 1/multidrug resistance- In rats, however, a substantial part of the absorbed
associated protein 2 (MRP2). Expression of MRP2 was arsenic accumulates in red blood cells, where it is
positively associated with the production and release firmly bound to hemoglobin (Hisanaga, 1982; Mara-
of DMA species into the cell culture medium. At ele- fante et al., 1982). Data from a number of investiga-
vated micromolar exposures of As3+, which inhibited tors indicate that the main form of arsenic bound to
the formation of MMA and DMA species, there was a rat hemoglobin is DMA (Lerman and Clarkson, 1983;
positive association between expression of solute car- Rowland and Davies, 1982; Vahter et al., 1984). As a
rier family 2, facilitated glucose transporter member 2 consequence of the efficient biotransformation of both
(GLUT2) and hepatocyte retention of inorganic arsenic trivalent and pentavalent inorganic arsenic in vivo
and MMA species. The dose-dependent aspects of these (see later), tissue partitioning is somewhat similar for
findings may be important for explaining the in vivo the different forms of exposure. In humans as well as
28 Arsenic 589

in most animal species, exposure to either arsenite or 1966). Arsenate, on the other hand, may interfere
arsenate leads to an initial accumulation in the liver, with metabolic phosphorylation reactions, probably
kidneys, and lungs (Bertolero et al., 1981). Tissue anal- because of its chemical similarity to phosphate (Fowler
ysis of organs taken from an individual after death et al., 1975; Mitchell et al., 1971). Such chemical simi-
from ingestion of 3 g arsenic (in the form of arsenic tri- larities can also explain the accumulation of arsenate
oxide) showed a much higher concentration of arsenic in the skeleton (Lindgren et al., 1982, 1984).
in the liver (147 μg/g) than in the kidney (27 μg/g) or When particles of inorganic arsenic compounds
muscle, heart, spleen, pancreas, lungs, or cerebellum such as arsenic trioxide, calcium arsenate, and arsenic
(11-12 μg/g) (Benramdane et al., 1999). Small amounts trisulfide were endotracheally instilled into the lung
were also found in other parts of the brain (8 μg/g), in of rats or hamsters, arsenic trioxide and arsenic trisul-
the skin (3 μg/g), and in hemolyzed blood (0.4 μg/g). fide rapidly disappeared from the lung, decreasing to
Clearance from these tissues is, however, rather rapid; a level of approximately 0.1-1% of the given dose over
in contrast, long-term retention of arsenic is seen in the a period of 24 h to 1 week. However, half of the dose of
hair, skin, squamous epithelium of the upper gastroin- calcium arsenate given to Syrian Golden hamsters was
testinal tract, epididymis, thyroid, lens, and skeleton retained in the lung for 1-5 weeks (Inamasu et al., 1982;
(Lindgren et al., 1982). Human autopsy data have also Pershagen et al., 1984).
shown high arsenic levels in the hair, nails, teeth, bone, In hamsters, MMA and DMA formed in vivo by the
and skin (Kadowaki, 1960; Liebscher and Smith, 1968; methylation of inorganic arsenic seem to be distrib-
Sumino et al., 1975; Yukawa et al., 1980). uted to all tissues (Takahashi et al., 1988; Yamauchi
Arsenic passes through the placenta in hamsters and Yamamura, 1985). This is supported by studies
after intravenous injections of high doses of sodium in animals in which MMA and DMA were found in
arsenate (Ferm, 1977). Kadowaki (1960) reported all tissues after acute oral doses (Yamauchi and Yama-
that arsenic levels in the human fetus increased as mura 1984; Yamauchi et al., 1988). Studies on the tissue
pregnancy progressed. Arsenic levels in bone, liver, distribution of arsenobetaine in mice and rabbits have
skin, and brain were 2-4 times higher in newborn shown that this arsenical is rapidly cleared from most
babies than in 7-month-old fetuses. In a population of tissues (Vahter et al., 1983). The longest retention was
Andean women exposed to high concentrations (about observed in cartilage, testes, epididymis, and muscles.
200 ppb) of inorganic arsenic in drinking water, arsenic Animal data indicate that arsenocholine, like choline,
concentrations in breast milk ranged from about 0.0008 may be incorporated into phospholipids (Marafante
to 0.008 ppm (Concha et al., 1998a,b). et al., 1984).
Although the distribution patterns seen after expo-
sure to different chemical forms of inorganic arsenic
5.3 Biotransformation
are similar, recent studies in mice and rabbits have
shown that arsenite gives rise to higher concentrations The biotransformation of inorganic arsenic is com-
than arsenate in most tissues (Lindgren et al., 1982; plicated by the different metabolites induced and by
Vahter and Marafante, 1983; Vahter and Norin, 1980). differences among species. Two basic processes are
Only the kidneys and the skeleton had higher levels involved: (1) reduction/oxidation reactions that inter-
in arsenate-treated than in arsenite-treated animals. convert As(III) and As(V), and (2) methylation reac-
In mice, radiolabel from orally administered 74As was tions, which convert arsenite to MMA and DMA. The
widely distributed to all tissues, with the highest lev- resulting series of reactions result in the reduction of
els in the skin, kidney, and liver (Hughes et al., 2003). inorganic arsenate to arsenite (if necessary), methyla-
The major portion of injected radioactive arsenic accu- tion to MMA(V), reduction to MMA(III), and methyla-
mulated in the mammalian tissues has been found in tion to DMA(V). These processes seem to be similar
the protein fraction (Marafante et al., 1981). Trivalent whether exposure is by the inhalation, oral, or paren-
arsenicals can chemically combine with sulfhydryl teral route.
groups, which may explain their inhibition of enzy- That trivalent inorganic arsenic is oxidized in vivo
matic reactions (Webb, 1966), as well as their accumu- is indicated by the identification of pentavalent arse-
lation in keratin-rich tissues such as hair, skin, and the nic in the urine of both animals and humans exposed
epithelium of the upper gastrointestinal tract. to arsenite (Bencko et al., 1976; Mealey et al., 1959;
It has also been found that the toxic effects of arsenic Vahter and Envall, 1983). The opposite reaction (i.e. the
can be reversed by parenteral administration of sulfhy- reduction of arsenate to arsenite) has also been demon-
dryl/thiol (SH) compounds of the reduced type, e.g. strated in mice and rabbits (Vahter and Envall, 1983).
glutathione, cysteine, and dimercaprol/British anti- Both arsenite and arsenate, after reduction to arsenite
Lewisite (BAL) (Peters, 1955; Vallee et al., 1960; Webb, (McBride et al., 1978), are methylated in vivo. The
590 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

major metabolite present in the urine of experimental mice. Polymorphic forms of the AS3MT enzyme sys-
animals exposed to inorganic arsenic is DMA (Ber- tem, either alone or in combination with polymorphic
tolero et al., 1981; Charbonneau et al., 1979; Inamasu, forms of enzymes such as glutathione S-transferase
1983; Tam et al., 1978; Vahter, 1981). Guinea pigs, mar- (GST), in humans may play a role in explaining dif-
mosets, and tamarin monkeys do not methylate inor- ferences in susceptibility to the various clinical mani-
ganic arsenic (Healy et al., 1998; Vahter and Marafante, festations of arsenical toxicity (Fujihara et al., 2010;
1985; Vahter et al., 1982; Zakharyan et al., 1996). Expo- Gomez-Rubio et al., 2010; Song et al., 2011; Paiva et al.,
sure of humans to either arsenites or arsenates results 2010). AS3MT and low-level arsenic exposure has been
in increased levels of inorganic As(III), As(V), MMA, recently linked to cardiovascular diseases in several
and DMA in the urine (Aposhian et al., 2000a,b). In rural counties in Texas (Gong and O’Bryant, 2012).
humans, urinary excretion at low dose levels consists Other studies (Sampayo-Reyes et al., 2010) that exam-
of approximately 20-25% inorganic arsenic, 15-25% ined arsenic-induced genotoxicity in Mexican chil-
methylarsonic acid, and 40-75% DMA (Buchet et al., dren environmentally exposed to arsenic in drinking
1981a; Crecelius, 1977b; Hopenhayn et al., 2003; Lof- water reported that DNA damage was modulated by
fredo et al., 2003; Mandal et al., 2001; Smith et al., 1977; both AS3MT and GSTO1 alleles. Wide variations in the
Tam et al., 1979; Yamauchi and Yamamura, 1979a,b). relative abundance of genetic variants of the AS3MT
Methylation efficiency decreases with increasing dose system have been reported by Fujihara et al. (2011)
levels (Mahieu et al., 1981; Vahter, 1981). The sub- who studied Mexican and German populations. Stud-
strate for methylation is As(III); As(V) is not methyl- ies into arsenic metabolism by human gastrointestinal
ated unless it is first reduced to As(III) (Buchet and flora following incubation with arsenic-contaminated
Lauwerys 1985, 1988; Lerman et al., 1983). Reduction soils reported extensive formation of MMA species
of arsenate to arsenite can be mediated by glutathi- and suggested that future arsenic toxicokinetic stud-
one (Menzel et al., 1994). The main site of methylation ies should take presystemic metabolism of arsenical by
seems to be the liver, where the methylation process gastrointestinal flora into account in order to improve
is mediated by methyltransferases that use S-adeno- aspects of risk characterization. These data suggest
sylmethionine as a cosubstrate (Buchet and Lauwerys, that epidemiological studies in arsenic-exposed popu-
1985, 1988). The relative proportions of As(III), As(V), lations should consider the interactive effects of other
MMA, and DMA in urine can vary, depending on the relevant enzyme systems that may be involved in cell
chemical administered, time after exposure, dose level, injury processes. Studies in Taiwan (Hsu et al., 2011)
and exposed species. Arsenic derived from exposure to did not find a close correlation between various GST
particles of GaAs or InAs is methylated in manner sim- alleles and urothelial carcinoma except for GSTO1 and
ilar to that of As(III) (Yamauchi et al., 1986, 1992).With GSTT1 at high arsenic exposure concentrations. Per-
relatively constant exposure levels, these metabolic sons living in the Red River Delta area of Vietnam have
proportions remain similar over time (Concha et al., also been recently reported (Agusa et al., 2012) to show
2002) and seem to be similar among family members variations in arsenic metabolism in relation to GST and
(Chung et al., 2002). It is expected that measurements AS3MT polymorphic forms.
of MMA and DMA in urine will provide useful infor- Arsenobetaine appears not to be biotransformed
mation for risk assessment purposes once the relation- in vivo, but is excreted unchanged mainly in urine
ships between the in vivo formation of these major (Cannon et al., 1981; Vahter et al., 1983). Arsenocholine
metabolites of inorganic arsenic and risk of toxicity or is, to a great extent, oxidized to arsenobetaine (Mara-
cancer are more fully delineated. fante et al., 1984).
In this regard, a number of recent studies have DMA and arsanilic acid are not converted to inor-
focused on arsenic methyltransferase and polymorphic ganic arsenic in vivo (Buchet et al., 1981a; Marafante
forms of this enzyme (AS3MT) in particular as being et al., 1987; Stevens et al., 1977; Vahter et al., 1984;
of central importance in the generation of methylated Yoshida et al., 2001).
arsenical metabolites in animals (Drobna et al., 2009;
Hughes et al., 2010), and hence in tissue dosimetry of
5.4 Excretion
methylated arsenical species. Other recent studies in
mice (García-Montalvo et al., 2011) reported the dose- The major route of excretion after exposure to inor-
dependent formation of methylated arsenic species in ganic arsenic is through the kidneys. Only a low per-
urine, indicating that the methylation process is subject centage is excreted in the feces (Apostoli et al., 1999;
to saturation and/or inhibition at higher doses of arse- Bertolero et al., 1981; Hunter et al., 1942; Mealey et al.,
nite. This information is potentially useful in the devel- 1959). The rate of urinary excretion varies, depend-
opment of a biologically based dose-response model in ing on the chemical form of arsenic and the species
28 Arsenic 591

exposed, as seen in a national survey of U.S. residents 5.5  Biological Half-Time


(Caldwell et al., 2009; CDC, 2013). The primary forms
Animals exposed to arsenic through inhalation or
of arsenic found in the urine of inhalation-exposed
drinking water will have increased tissue levels dur-
humans are DMA and MMA, with inorganic arsenic
ing the first weeks or months, but the levels will then
making up < 25% of the total urinary arsenic (Apostoli
decrease even if the exposure is prolonged (Bencko and
et al., 1999). In humans exposed to a single low dose of
Symon, 1969, 1970; Hisanaga, 1982; Katsura, 1958). It
arsenite, approximately 35% was excreted in the urine
has been suggested that the decrease of arsenic concen-
over a period of 48 h (Buchet et al., 1980, 1981a). At
trations in internal organs, such as the liver, after long-
low exposure levels, urinary arsenic levels generally
term exposure may be due to a higher rate of excretion.
increase linearly with increasing arsenic intake (Calde-
Changes in tissue retention in the case of chronic expo-
ron et al., 1999). Rabbits exposed to a similar low dose
sure should therefore be considered when evaluating
excrete approximately 80% (Bertolero et al., 1981), mice
biological half-times for arsenic compounds.
as much as 90% (Vahter, 1981), and marmoset monkeys
The biological half-time of arsenic in rats after
as little as 15% within the same period of time (Vahter
a single exposure is long (approximately 60 days)
et al., 1982). In the case of continuous human intake
because of the accumulation of arsenic in the blood.
over a few days, 60-70% of the daily dose is excreted
In humans and other animals, the major proportion of
in the urine (Buchet et al., 1981b; Mappes, 1977). After
arsenic is eliminated at a much higher rate. Generally,
exposure to arsenate, the limited human data available
whole body clearance is fairly rapid, with half-times
indicate a rate of excretion similar to that for arsenite
of 40-60 h in humans (Buchet et al., 1981b; Mappes,
(Pomroy et al., 1980; Yamauchi and Yamamura, 1979b).
1977). Three different phases of urinary excretion have
Animal data indicate a somewhat faster excretion rate
been demonstrated in humans after a single intrave-
for arsenate than for arsenite (Charbonneau et al., 1978;
nous injection of radiolabeled arsenite, with half-times
Hollins et al., 1979; Vahter, 1981).
of approximately 2 h, 8 h, and 8 days, respectively
During lactation, a very small percentage of ingested
(Mealey et al., 1959). After oral intake of radiolabeled
arsenic may also be excreted in breast milk (Concha
pentavalent arsenic, 66% was excreted with a half-time
et al., 1998a). Other routes of elimination of inorganic
of 2.1 days, 30% with a half-time of 9.5 days, and 3.7%
arsenic, although of less importance, include the skin,
with a half-time of 38 days in the three phases (Pomroy
hair, nails, and sweat (Molin and Wester, 1976).
et al., 1980).
Studies in humans indicate that ingested MMA
As stated previously, the major proportion of arse-
and DMA excreted are mainly in the urine (75-85%),
nic in ingested seafood is rapidly eliminated through
and that this mostly occurs within 1 day (Buchet
the kidneys. The biological half-time for these organic
et al., 1981a). After ingestion, by humans, of organic
compounds is estimated to be less than 20 h. Studies
arsenic compounds present in seafoods, 50-80% of
in humans indicate that ingested MMA and DMA are
the dose was eliminated in the urine within 2 days
mainly excreted in the urine (75-85%), and this occurs
(Tam et al., 1982). A similar rate of excretion was
mostly within 1 day (Buchet et al., 1981a).
seen in rabbits exposed to arsenobetaine (Vahter
The WHO International Programme on Chemical
et al., 1983).
Safety (WHO/IPCS, 2001) has examined a number of
The elimination of organic arsenic compounds used
issues related to the roles of elemental speciation in
as drugs was noted by Hogan and Eagle (1944). They
human health risk assessment.
found that trivalent organic arsenicals were elimi-
nated much more slowly than pentavalent arsenicals
in the urine of rabbits. More recent studies by Lin et al. 5.6  Mechanisms of Arsenical Toxicity
(2005) reported that the tissue-specific distribution and
5.6.1  Mechanisms of Arsenical Metabolism and
elimination rates for inorganic and methylated arsenic
Toxicity
species in rabbits after a single acute administration
of arsenic trioxide (AsIII) over a dose range of 0.2- Arsenical inhibition of cellular respiration as a pri-
1.5 mgAs/kg for up to 30 and 60 days. The elimination mary cause of cell death has been appreciated for a
pattern was nonlinear; the liver and lung showed the number of decades (ATSDR, 2007; NRC, 1999, 2001),
highest concentrations of DMA, which was the major and the methylation of inorganic arsenic to form meth-
metabolite measured, on day 30. Similar organ-specific ylated species such as MMA and DMA has also been
differences were observed in B6C3F1 mice after acute studied for a number of decades (Braman and Fore-
oral administration of arsenate [As(V)]. As for the rab- back, 1973; Challenger, 1945, 1951). In recent years, sci-
bits, DMA was the predominant methylated species at entific attention has focused on trying to understand
later time points (Kenyon et al., 2005). the relationships that must exist between the in vivo
592 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

methylation of inorganic arsenic and mechanisms of They found that selenite acts as a noncompetitive
arsenical toxicity. This issue is of great practical impor- inhibitor of this enzyme. The tissue dosimetry of pen-
tance because the methylation of inorganic arsenic tavalent and trivalent MMA was studied by Hughes
was originally thought to be a detoxification pathway; et al. (2005), who found that a much greater percentage
however, more recent studies (NRC, 1999) have sug- of the administered dose of MMA(III) was converted
gested that highly toxic reactive oxygen species (ROS) to DMA than after administration of MMA (V).
generated by MMA(III) and DMA(III) mitochondrial
toxicity may also play an important role in both the 5.6.2.2  Arsenical Methylation
cellular toxicity and carcinogenicity of this element. The main characteristics of the pathway for inor-
The increased presence of MMA(III) in the urine of a ganic arsenic methylation have been established since
Mexican population exposed to inorganic arsenic in the pioneering work of Challenger (1945, 1951). The
drinking water on a chronic basis was found to form pathway involves a series of reductions and oxidation
the basis for identifying subpopulations at a greater steps which interconvert As5+ to As3+ to As5+ during
risk of arsenic-induced toxicity and cancer (Steinmaus the methylation steps, ultimately resulting in the for-
et al., 2005a,b; Valenzuela et al., 2005). These studies mation of MMA(V) and DMA(V). The further metabo-
also reported a strong link between dietary intakes of lism and excretion of DMA and trimethylarsine oxide
protein and other nutrients and the ability to meth- has been extensively studied in rodents by Yamau-
ylate arsenicals such that persons with low dietary chi and coworkers (Yamauchi and Yamamura, 1984;
intakes of these nutrients would be more susceptible Yamauchi et al., 1989, 1990). The enzyme responsible
than others to arsenic-induced cancers. The central role for the reduction of As(V) to As(III) has not yet been
of oxidative stress induced by arsenic in cell death via identified, but a GSH- and NAD-dependent As(V)
apoptosis or necrosis and carcinogenicity via oxidative reductase activity linked to glycolysis was identified
damage to cellular DNA cannot be underestimated in rat liver cytosol and human erythrocyte lysate by
because it may permit attenuation of these deleterious Nemeti and Gregus (2005). During this metabolism,
cellular effects through nutritional interventions and MMA(III) and DMA(III) are also formed, and MMA(III)
stimulation of the cellular antioxidant systems. and DMA(III) in particular have been demonstrated to
be highly toxic and reactive chemical species (Mass
5.6.2 Metabolism et al., 2001). The kinetics of in vitro arsenic methyla-
tion in mouse hepatocytes were reported by Kedderis
The focus in this section on arsenical metabolism et al. (2006), who found a concentration-dependent
is to review mechanistic linkages between the mecha- formation of MMA(III) up to a concentration of about
nisms of inorganic arsenical methylation and mecha- 2 μmol/L As(III), followed by a decline at higher con-
nisms of toxicity at the cellular and molecular levels of centrations. They also suggested a role for a glutathi-
biological organization. one complex-based mechanism in this methylation
pathway. This hypothesis is consistent with obser-
5.6.2.1  Arsenical Reduction/Oxidation vations from a number of other laboratories (Chang
A key step in the methylation pathway for inorganic et al., 1991; Csanaky et al., 2003; Hayakawa et al., 2005;
arsenicals is the reduction and oxidation of arsenic Hirano et al., 2004; Kala et al., 2004; Kobayashi et al.,
between the −3, +3, and +5 oxidation states during the 2005; Waters et al., 2004). The formation of arsenic-glu-
various steps in the methylation pathway. Styblo and tathione complexes has also been reported (Cui et al.,
Thomas (1995) reported the inhibition of glutathione 2004; Suzuki et al., 2001) to play important roles in the
reductase by arsenoglutathione in vitro and demon- biliary and urinary excretion of arsenic in rats.
strated that reduced glutathione (GSH) was required In recent years, an arsenic methyltransferase
for the in vitro methylation of arsenic. (AS3MT) has been identified (Drobna et al., 2005; Li
More recent studies by Nemeti and Gregus (2005) et al., 2005; Wood et al., 2006) and demonstrated to
demonstrated the reduction of As5+ to As3+ in human exist in a number of polymorphic forms (Wood et al.,
erythrocyte lysates and rat liver cytosol through an 2006). Zakharyan et al. (2005) studied the interactions
arsenic reductase, which required glycolytic substrates, of arsenic species with selenite, glutathione, and the
GSH, and nicotinamide diamide (NAD) for its enzyme omega class of human glutathione transferase. They
activity. Zakharyan et al. (2005) studied the interaction found that human MMA(V) reductase and human
of selenite with the omega class human glutathione GSTO-1 were identical proteins, and that selenite inhi-
S-transferase [GSTO-1; identical to monomethylar- bition of MMA(V) formation was reversed by dithioth-
sonic acid reductase (MMA(V) reductase)], which cata- reitol but not by GSH, which is the required substrate
lyzes the reduction of As(V), MMA(V), and DMA(V). of this enzyme. They found that three selenium atoms
28 Arsenic 593

and three GSH molecules were bound to the mono- factor-kappa-B (NF-κB) and activator protein-1 (AP-1)
meric form of the enzyme. This enzyme and its dem- (Felix et al., 2005). These various cellular responses to
onstrated polymorphic forms may well explain the arsenical-induced oxidative stress are discussed later
differences in methylation patterns observed in both in greater detail.
animals and humans (NRC, 1999), as well as differ-
ences in susceptibility to a variety of toxic effects 5.6.3.3  Antioxidant Cellular Defense Systems
including various cancers (Smith et al., 2006), hepato- 5.6.3.3.1  Glutathione  A major intracellular antioxi-
megaly (Guha Mazumder et al., 2005), and heme and dant is GSH, which exists in many cell types at mil-
porphyrin metabolism (Fowler et al., 2005; Garcia- limolar concentrations. Increased production of this
Vargas et al., 1994; Ng et al., 2005; Woods and Fowler, small SH-rich molecule may occur by induction of
1978; Yamauchi et al., 1998). glutathione synthetase in response to ROS-mediated
oxidative stress. Glutathione depletion studies using
5.6.3  Mechanisms of Arsenical Toxicity 1-butathionine sulfoximine have produced increased
sensitivity among cells exposed to arsenic(III) in vitro
5.6.3.1  Mitochondrial Inhibition and Oxidative Stress
(Maiti and Chatterjee, 2001; Shimizu et al., 1998), indi-
Arsenical inhibition of mitochondrial respiration cating the importance of this intracellular antioxidant
supported by NAD-linked substrates that use the molecule in mediating arsenical toxicity in target cell
lipoic acid cofactor for the pyruvate dehydrogenase populations.
complex is regarded to be a primary mechanism by
which arsenicals produce cell injury/cell death and 5.6.3.4  Stress Proteins/Heat Shock Proteins (HSPs 25/27,
cancer (Fowler and Woods, 1979; Fowler et al., 1979; 32, 60, 70, and 90)
Chen et al., 1986; NRC, 1999). Inorganic arsenicals, Arsenicals are known to induce a number of the
MMA, and DMA have each been shown to produce major stress protein families, presumably as a conse-
oxidative stress by means of inhibiting mitochondrial quence of ROS formation with the attendant oxidation
respiration, resulting in the formation of ROS, which of protein side chains and the onset of proteotoxicity.
may cause DNA mutations and ultimately play a role Cells of a similar nature vary in their susceptibility to
in the development of cancer (Liu et al., 2005). In addi- arsenicals, partly because of their ability to produce
tion, either acute or chronic exposure to arsenicals stress proteins. This was demonstrated by Madden
was shown to produce a characteristic porphyrinuria et al. (2002), who showed that rat and human kidney
pattern in experimental animals (Conner et al., 1993, cell lines varied in their sensitivity as a function of 60-,
1995; Fowler et al., 2005; Woods and Fowler, 1978) and 70-, and 90-kDa stress protein expression patterns. For
humans (Garcia-Vargas et al., 1994; Garcia-Vargas and both cell lines, toxicity occurred once the capacity to
Hernandez-Zavala, 1996; Ng et al., 2002, 2005; Yamau- produce the stress proteins was exceeded at elevated
chi et al., 1998), dominated by elevated levels of uro- dose levels. Similar results have been reported for
porphyrins with lesser amounts of coproporphyrin. HSp70 in lung cells (Han et al., 2005; Lau et al., 2004)
This pattern is, hence, conserved across species and and in human epidermal cells (Lee et al., 2005). Similar
may serve as a useful biomarker for early arsenical responses have been observed in female rat urothelial
toxicity to target organs such as the liver and hemato- cells after in vivo exposure to DMA for 28 days (Sen
poietic system. It should also be noted that porphyrins et al., 2005) and the human UROtsa cell line exposed to
are also toxic and capable of catalyzing the formation arsenite (Rossi et al., 2002). Arsenite induction of heat
of ROS, thus exacerbating direct arsenical toxicity. shock 70 kDa protein (HSp70) has been consistently
observed by a number of investigators in a variety
5.6.3.2  Reactive Oxygen Species of cell types, including hepatoma cell lines (Farzaneh
The ROS generated by inhibition of mitochondrial et al., 2005; Gottschalg et al., 2006), human MCF-7
respiratory function and ATP depletion (Chen et al., breast cancer cells (Barnes et al., 2002), human lung
1986) produce oxidative stress in a number of major fibroblasts (HEL cells), human leukemia cells (THP-1
organ systems (Flora et al., 2005; Li et al., 2002; Ramos cells) (Rossner et al., 2003), rat oligodendrocytes
et al., 1995) and elicit a number of cellular responses, (Goldbaum and Richter-Landsberg, 2001), and chick
including upregulation of glutathione synthesis (Flora embryos (Papaconstantinou et al., 2003).
et al., 2005), induction of the major stress protein fam- In addition to the major stress family proteins, the
ilies (Lee et al., 2005; Madden et al., 2002; Sen et al., formation of ROS also induces the SH-rich protein
2005), and altered regulation of signaling pathways metallothionein (MT), which also has antioxidant prop-
involving nitric oxide synthetase/endothelial nitric erties (Gottschalg et al., 2006; Liu et al., 2001). It should
oxide synthase (eNOS) (Tsou et al., 2005), and nuclear also be noted that studies by Shimizu et al. (1998)
594 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

reported that zinc induction of MT exerted no discern- In agreement with the role of ROS in mediating arseni-
ible protective effects on arsenical toxicity; therefore, cal induction of apoptosis, administration of antioxi-
the potential mechanisms of protection against arseni- dants such as ascorbic acid and α-tocopherol have been
cal toxicity may be complex and system dependent. shown (Ramanathan et al., 2005) to reduce tumor necro-
The lower molecular mass HSP25(rat)/HSP27(human) sis factor (TNF-α; encoded by TNF) levels and activate
have also been reported to be induced by arsenite the caspase cascade system. The value of arsenicals in
exposure in mouse kidney podocyte cultures (Eichler mediating apoptosis has been explored in relation to
et al., 2005, 2006) and also seem to play major roles in the treatment of various cancers such as acute mega-
the regulation of apoptosis via the extrinsic caspase-8 karyocytic leukemia (Lam et al., 2005) or promyelo-
pathway (Eichler et al., 2006). It is interesting to note cytic leukemia (Zheng et al., 2005), where As2O3 has
that arsenic(III) induction of the stress protein response been shown to activate a number of central factors in
for HSP27 and HSP70 has been shown to be regulated the apoptotic pathway. Similar delays in the recovery
by reducing agents such as dithiothreitol (Kato et al., of leukocytopenia and neutropenia during acute pro-
1997) and by reduced levels of GSH in arsenic-exposed myelocytic leukemia have been reported after treat-
cells (Ito et al., 1998), indicating the strong link between ment of patients with As2O3 relative to treatment with
arsenic-induced oxidative stress and stress protein all-trans retinoic acid (Shinjo et al., 2005). On the other
induction patterns. In addition, arsenite exposure of hand, long-term, low-level exposure of cultured human
cells has also been well documented to induce HSP32/ keratinocytes to arsenite has been shown (Pi et al., 2005)
heme oxygenase 1 (HO-1), which is the rate-limiting to produce a generalized resistance to the induction of
enzyme in the heme degradative pathway (Gong et al., apoptosis, which may provide a role for arsenic as a
2002; Lee et al., 2005) and plays a major role in the recy- cocarcinogen with ultraviolet (UV) light in skin cancer,
cling of heme iron. as discussed later. A number of other investigators have
reported similar arsenic-induced alterations in apopto-
5.6.3.5  Altered Cell Signaling Pathways sis regulation in other cell types such as neuronal (Jou
Arsenic-induced alterations of the blood vasculature et al., 2004; Namgung and Xia, 2000) and lymphoma
leading to the development of “Blackfoot disease” are cell lines (Muscarella and Bloom, 2002) and in devel-
common findings among persons exposed to arsenic in oping mouse embryos (Liu et al., 2003). Interestingly,
drinking water. Studies by Tsou et al. (2005) reported HSP70 (discussed earlier) has been reported to pro-
arsenic-induced downregulation of eNOS, supporting tect against arsenic-induced apoptosis (Mosser et al.,
the hypothesis that altered eNOS function may play an 2000) at a step in apoptotic pathway after cytochrome
important role in mediating this clinically important c release and before caspase-3 activation (Li et al., 2000).
outcome. Studies by others (Wu et al., 1999, 2002) dem-
onstrated a number of alterations in cell signaling path- 5.6.3.7 Necrosis
ways, including the mitogen-activated protein kinase As with many toxic metals, cellular necrosis is usu-
(MAPKs), extracellular signal-regulated kinase (ERK), ally observed after the administration of high-dose
and Ras via the epidermal growth factor receptor (EGFR) arsenicals (Shen et al., 2006), and there is strong dose
pathway in a human airway epithelial cell line (BEAS- dependency as to whether apoptosis or necrosis is
2B). Similar alterations in the c-jun/AP-1 and NF-κB sig- observed after exposure to this agent.
nal transduction systems, with concomitant alterations
in the expression of a number of the major stress protein 5.6.3.8 Cancer
families (HSP32 and HSP72), were observed by Wijew- In recent years, there have been many detailed
eera et al. (2001) in precision-cut rat lung slices. reports on the diagnosis, molecular markers, and possi-
ble underlying mechanisms of arsenic-induced cancers
5.6.3.6  Mechanisms of Cell Death in various organ systems. As with the mechanisms of
5.6.3.6.1  Apoptosis Arsenite at low dose levels cell injury and apoptosis discussed previously, the gen-
in vitro has been demonstrated to produce apoptosis eration of oxidative stress from the mitochondrial pro-
in rat thymocytes (Bustamente et al., 1997). Subsequent duction of ROS seems to be a central feature of cancer
studies (Tian et al., 2005) demonstrated that As2O3 initiation (NRC, 1999). Yu et al. (2006) reviewed arse-
induces opening of the mitochondrial membrane per- nic-induced skin cancers and the probable combined
meability transition pore, with attendant release of roles of arsenic exposure and UVB radiation in produc-
cytochrome c and intramitochondrial calcium stores. ing skin lesions. Kuo et al. (1997) examined the possible
Other investigators (Zhang et al., 2003; Zheng et al., roles p53 gene overexpression in skin cancer and con-
2005) have shown that As2O3 also induces Bax activa- cluded that this was not a prerequisite for the devel-
tion in hematopoietic cells secondary to ROS formation. opment of skin cancer. Dong (2002) noted increased
28 Arsenic 595

phosphorylation of ERKs in cell lines and observed 6.1 Organs


that arsenic did not induce p53 transactivation. Waal-
Median arsenic concentrations in the organs of
kes et al. (2004) reported altered estrogen signaling in
healthy people from Scotland who died in accidents
male mice with hepatocellular carcinomas produced
ranged from 0.012 mg/kg in the brain to 0.46 mg/
by in utero exposure to arsenic. They concluded that
kg in hair (both dry weight) (Liebscher and Smith,
overexpression of estrogen receptor alpha (ERα), prob-
1968). Median concentrations in organs of people
ably through hypomethylation of the promoter region
from Japan who had generally consumed relatively
of this gene, may play a role in hepatocarcinogenesis
high amounts of seafood ranged from 0.02 mg/kg
in these mice. Subsequent studies by Cui et al. (2006)
wet weight in the pancreas to 0.89 mg/kg in the nails
demonstrated that arsenic trioxide does inhibit DNA
(Kadowaki, 1960). The first study used NAA on vac-
methyltransferase and activates methylation-inhibited
uum-dried samples and the second used polarogra-
genes in human HepG2 liver cancer cells. Hour et al.
phy. Values for all organs analyzed in the two studies
(2006) reported the differential expression of various
are given in Table 1.
molecular markers in surgical samples of uroepithelial
Tissue analysis of organs taken from an individ-
cells from arsenic-exposed and nonarsenic-exposed
ual after death from ingestion of 8 g arsenic trioxide
patients. They found a decreased GSH content in
(approximately 3 g arsenic) showed a much higher con-
cells from the arsenic-exposed patients and increased
centration of arsenic in the liver (147 μg/g) than in the
expression of apoptosis regulator Bcl-2 and proto-
kidneys (27 μg/g) or muscle, heart, spleen, pancreas,
oncogene c-Fos. Shen et al. (2006) examined the effects
lungs, or cerebellum (11-12 μg/g) (Benramdane, et al.
of three methylated arsenicals [MMA(V), DMA(V),
1999). Small amounts were also found in other parts of
and TMAO] on the urinary bladder epithelium of both
male and female rats exposed to these substances for
13 weeks. They noted metabolic differences between TABLE 1  Arsenic Concentrations in Human Organs and
males and females with regard to further metabolism Tissues in Scotland and Japana
and concluded that DMA(V) is more toxic to the blad-
Arsenic Concentration (mg/kg)
der epithelium of female rats, which are also more sus-
ceptible to bladder carcinogenesis from this agent. Scotland Japan
Mizoi et al. (2005) reported that oxidative stress (dry wt.) (wet wt.)
caused by DMA(III) generated as a reduction prod-
Tissue/organ Median Range Median
uct of DMA(V) seem to play a central role in lung and
skin tumors in mice exposed to DMA(V), as judged Adrenal 0.029 0.002-0.293 –
by the increased formation of 8-oxo-2ʹdeoxyguanosine Aorta 0.031 0.003-0.570 –
(8-OHdG) DNA adducts. It is also important to note Whole blood 0.038 0.001-0.920 –
Bone 0.057 0.010-0.240 0.118 (femur)
that an increased presence of 8-OHdG adducts has
Bone – – 0.074 (rib)
been reported in urine samples of persons living in Brain 0.013 0.001-0.036 0.034
Inner Mongolia (Fujino et al., 2005) and Cambodia Hair 0.460 0.020-8.17 0.174
(Kubota et al., 2006) exposed to arsenic in contami- Heart 0.024 0.002-0.078 0.041
nated drinking water, suggesting that these adducts Intestine, large – – 0.025
Intestine, small – – 0.022
may be potentially useful biomarkers for the long-
Kidney 0.033 0.002-0.363 0.041
term risk of arsenic-induced cancers such as Bowen Liver 0.028 0.005-0.246 0.042
disease (Matsui et al., 1999). It is important to note that Lung 0.082 0.006-0.514 0.047
the stress protein response discussed previously may Muscle 0.063 0.012-0.431 0.029
play an important regulatory role in mediating the car- Nail 0.300 0.020-2.90 0.892
Ovary 0.037 0.013-0.260 –
cinogenic or cocarcinogenic effects of arsenic through
Pancreas 0.045 0.005-0.410 0.020
interaction with a number of proto-oncogenes and the Prostate 0.046 0.010-0.090 –
apoptotic caspase system (Khalil et al., 2006; Stanhill Skin 0.090 0.009-0.590 0.064
et al., 2006; Yaglom et al., 2003). Spleen 0.020 0.001-0.132 0.021
Stomach 0.037 0.003-0.104 0.022
Teeth 0.050 0.003-0.635 0.078
Thymus 0.015 0.003-0.332 –
6  BIOLOGICAL MONITORING Thyroid 0.042 0.001-0.314 –
Uterus 0.031 0.010-0.188 0.036
A review by Mandal and Suzuki (2002) document
many cases of human arsenic exposures around the aCompiled from Liebscher and Smith (1968) and Kadowaki (1960),
world, as well as sources of arsenic in the environment. respectively
596 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

the brain (8 μg/g), in skin (3 μg/g), and in hemolyzed arsphenamines from arsanilic acid. The mean arsenic
blood (0.4 μg/g). level was 0.44 mg/L, with a range of 0.04-3.8 mg/L.
Inorganic arsenic can easily pass through the pla- Calderon et al. (1999) found a correlation between
centa. High levels of arsenic were found in the liver, the log of the mean total urinary arsenic concentra-
kidney, and brain after autopsy of an infant prema- tion/creatinine (TAs/c; μg/mg) in people living in
turely born to a young mother who had ingested inor- areas with arsenic-contaminated drinking water
ganic arsenic at week 30 of gestation (Lugo et al., 1969). sources and the log of the inorganic arsenic concentra-
Arsenic was detected in human breast milk at con- tion in the drinking water (lnAs; μg/L).
centrations of 0.00013-0.00082 ppm (Somogyi and Beck, Arsenic concentration in urine may be used as an
1993). In a population of Andean women exposed to index of exposure, but a number of factors such as a
high concentrations (approximately 200 ppb) of inor- diet containing mainly seafood and the time between
ganic arsenic in drinking water, concentrations of arse- exposure and urine sampling have to be considered.
nic in breast milk ranged from about 0.0008-0.008 ppm Because “fish arsenic” (i.e. arsenobetaine) is essentially
(Concha et al., 1998a). nontoxic, total urinary arsenic content may overesti-
It is not known to what extent arsenic concentra- mate exposures to arsenic species that are of concern
tions in human organs represent inorganic arsenic or for health. Experience has shown that total urinary
organic arsenic originating in the diet. Concentrations arsenic may lead to the overestimation of exposure to
of 0.014-0.21 mg As/kg wet weight have been observed inorganic arsenic even if the subjects are requested to
in the lungs of smelter workers who had retired 2-19 refrain from eating seafood (Pinto et al., 1976). Dietary
years before death; in contrast, unexposed controls had habits should be examined thoroughly when the total
0.001-0.018 mg/kg, indicating a long retention time of urinary arsenic concentration is to be used as an index
arsenic in the lungs after inhalation, which may occur of exposure. Analytical differentiation of the different
in certain occupations (Brune et al., 1980). forms of arsenic in the urine is an appropriate way of
determining how much and what form of arsenic has
been absorbed (Buchet et al., 1980; Norin and Vahter,
6.2 Urine
1981). The sum of urinary excretion of inorganic arse-
Most arsenic absorbed from the lungs or the gas- nic and its monomethylated and dimethylated metabo-
trointestinal tract is excreted in the urine within 1-2 lites is a good indicator of exposure to inorganic arsenic
days. Therefore, the urinary arsenic level is generally when there is some fish consumption. Buchet et al.
accepted to be the most reliable indicator of recent (2003) determined such values by means of hydride
arsenic exposure, and this approach has proved useful generation combined with AAS or AAF and reported
in identifying above-average exposures in populations a mean value of approximately 7 μg/g creatinine and
living near industrial point sources of arsenic (Polis- a 95% cutoff of 18 μg/g creatinine in a group from the
sar et al., 1990). Normal levels of arsenic in the urine general population of Belgium. In a population group
of people with no known high exposure appear to be from China not known to be exposed to inorganic arse-
in the range of 5-50 μg As/L (Baker et al., 1977; Bencko nic, there was a mean of approximately 25 μg/g cre-
and Symon, 1977; Braman and Foreback, 1973; Buchet atinine and a 95% cutoff of 50 μg/g creatinine (Buchet
et al., 1980; Smith et al., 1977; Caldwell et al., 2009; et al., 2003). Recently, Basu et al. (2011) noted that urine
CDC, 2013). The ingestion of seafood may, however, creatinine concentration was the strongest biological
increase the concentration to more than 1 mg As/L marker of arsenic methylation efficiency and therefore
(Buchet et al., 1980; Norin and Vahter, 1981; Pinto et al., should not be used to adjust for urine concentration in
1976; Schrenk and Schreibeis, 1958). arsenic studies.
In a study of As2O3 exposure in a smelter, Pinto et al.
(1976) found an average urinary arsenic concentration
6.3 Blood
of 0.053 mg/L among 200 workers who had not been
exposed to arsenic. The average arsenic concentra- Using NAA, Brune et al. (1966) found a mean con-
tion in the urine of 24 men who had been exposed to centration of 0.004 mg/kg in the whole blood of normal
a mean air concentration of 53 μg/m3 (range 3-295 μg/ people and 0.035 mg/kg in uremic patients. Heydorn
m3) in the same factory was 0.038-0.539 mg/L, with an (1970) reported a mean arsenic concentration in whole
overall average of 0.174 mg/L. They found a correla- blood of 0.022 mg/L in Taiwanese subjects, also using
tion between airborne arsenic concentrations and arse- NAA.
nic in urine, but the values showed a wide scatter. In three studies of As-exposed populations, Vahter
Watrous and McCaughey (1945) determined urinary et al. (1995) and Concha et al. (1998a,b) found the aver-
levels of arsenic among workers in a factory producing age total blood As to be 7.6, 9, and 10 μg/L, respectively.
28 Arsenic 597

In one of these studies, the range of blood As concen- extract As from the hair. The average percentage recov-
tration of an unexposed population was 1-2 μg/L (Con- ery of the species versus the total As analysis value
cha et al., 1998b). Using HPLC-ICP-MS, Suzuki et al. was 57%. This is common in these types of analysis, in
(2002) and Mandal et al. (2004) could separate and which the laboratory is extracting analytes from solid
quantify the As species in the blood of an As-exposed matrices. Raab and Feldmann (2005) speciated As in
population. They found arsenobetaine, DMA, MMA, the hair of an exposed population and found As(III),
and inorganic As in the blood samples by speciating As(V), MMA(V), and DMA(V). The study also found
the blood. They also found a total As concentration of that the method of extraction varied with the abun-
10.1 μg/L As in blood from an As-exposed individual. dance of different species and the extraction efficiency
Mandal et al. (2004) also separated the blood into red of the specific species relative to the total As concen-
blood cells and plasma, and determined the fraction of tration. In all of these As speciation studies, the pres-
the various species in these blood components. ence of arsenobetaine was not detected; therefore, the
As stated in Section 5.2, arsenic clearance from the assumption is that it does not accumulate in hair.
blood is very rapid. The period between exposure and The mean concentration of arsenic in hair has been
sampling will, therefore, be of importance if the blood found to reflect the degree of arsenic pollution in com-
levels are being related to the exposure level. Fur- munity air (Bencko and Symon, 1977; Bencko et al.,
thermore, the intake of seafood may greatly influence 1971; Hammer et al., 1971). Concentrations of arsenic
blood arsenic levels. For these reasons, blood arsenic in hair were generally found to increase with the arse-
cannot be regarded as a useful indicator of exposure. nic concentration in air. Hinwood et al. (2003) looked
A review by Taylor et al. (2004) tabulates both the at the general types of environmental and personal
technologies used for the determination of As in blood, As exposures, classified the population into exposure
hair, nails, and urine, and summarizes the basic find- categories, and determined the hair As concentration
ings for the different biological matrices. of these different exposures. They classified the expo-
sures into high water, high soil, and personal exposure.
The greatest effect was for the high-water exposures,
6.4 Hair
in which they found a mean of 5.5 μg/g As in the hair
Attempts have been made to correlate normal with a range of 1.2-20 μg/g.
concentrations of arsenic in hair to exposure to inor- Evaluating hair As from unexposed populations
ganic arsenic. Smith (1964) found that 80% of 1000 such as the U.S. National Human Exposure Assessment
people tested had a concentration below 1 mg As/kg Survey, showed there to be no correlation between hair
in hair, with an average of 0.81 mg/kg and a median As and food, drinking water, dust intake, or exposure
of 0.51 mg/kg. Liebscher and Smith (1968) gave 0.02- (Pellizzari and Clayton, 2006). The average hair As in
8.17 mg/kg as the range of arsenic concentrations in this population was 0.17 μg/g As, with a range of 0.06-
hair, based on an examination of 1250 people without 0.33 μg/g As (10th to 90th percentiles).
known exposure to high levels of this element. Yang Arsenic trioxide (As2O3) has been used as a chemo-
et al. (2002), Mandal et al. (2003), and Uchino et al. therapeutic agent for some leukemia treatments. Shen
(2006) examined correlations between hair As and As et al. (1997) looked at the incorporation of As in the
intake from water. They all found a correlation, but the hair of several patients during a period when arsenic
results differed by a factor of 2-3 for the level of As in trioxide was being administered. They found that As
the hair versus the level of As in the water. This could was incorporated into the hair to a level of 2.5-2.7 μg/g
be due to many factors, including the mode of washing (about five to seven times greater than the baseline lev-
the hair before analysis (to remove external contami- els) shortly after arsenic trioxide administration, and
nation), analytical methods (both used ICP-MS), and that hair As levels decreased after drug administration
digestion techniques. Yang et al. (2002) looked at the was stopped. The literature on the medicinal uses of
average hair As as a function of the extent of arseni- arsenical has been reviewed by Ratnaike (2003).
cism in the patients and found a range of 2-4.5 μg/g As, It should be noted that there are several caveats
depending on the extent of hyperkeratosis, depigmen- related to the analysis of hair As that must be taken
tation, and pigmentation. Mandal et al. (2003) found into consideration when evaluating an individual or
an average concentration of 4.5 μg/g As in hair, with population suspected to have undergone chronic or
a range of 0.7-16.2 μg/g. Mandal et al. (2003) also spe- acute As exposure or poisoning. It may not be possible
ciated hair samples using HPLC-ICP-MS and found to distinguish between arsenic adsorbed onto the hair
As(III), As(V), MMA(V), and DMA(V). The main diffi- from external contamination and arsenic incorporated
culty in interpreting these results is the variation in the into hair from the internal body burden. Hindmarsh
speciation results depending on the technique used to (2002) has shown that one can measure As in hair well
598 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

before the As could have been incorporated through TABLE 2  Acute and Subacute Toxicity of Arsenic
hair growth. This could be due to As in the sweat or
Organ System Symptoms and Sign
other mechanisms contaminating the hair. He has also
shown that hair can take up As directly from water Gastrointestinal Nausea, vomiting, thirst, anorexia, heartburn,
containing As, and that As is not removed by the nor- abdominal pain, diarrhea with bloody stool
mal washing procedures used in analytical methods. Dermal Dermatitis, vesticulation, melanosis
Neural Encephalopathy (hyperpyrexia, convulsion,
Therefore, one must be cautious when using hair As
tremor, coma, disorientation), neuritis,
as an indicator of absorbed arsenic, if the external con- peripheral neuropathy (primarily sensory
tamination cannot be controlled in some way (Vahter type, paresthesia, hyperesthesia, numbness
et al., 1983). of extremities, neuralgia, muscular cramp,
and weakness)
Renal Cortical necrosis, leukocyturia, glycosuria,
hematuria, oliguria, uremia
7 EFFECTS Hepatic Congestion, fatty infiltration, central necrosis,
acute yellow atrophy, cholangitis,
Inorganic arsenic has been recognized as a human cholecystitis
poison since ancient times. Arsenic-related human tox- Hematological Anemia, thrombocytopenia, leukopenia, bone
icity is systemic, involving a number of organ systems. marrow suppression
Cardiovascular Cardiac abnormality (ventricular fibrillation
The acute, subacute, and chronic toxic effects of inor-
and atypical tachycardia), prolonged Q-T
ganic arsenic exposure through inhalation and inges- interval, abnormal T wave; congestive heart
tion have been reviewed periodically (ATSDR, 2007; failure, hypotension
Chen, 2011a,b; Chen et al., 1997a,b; IARC, 2004, 2006; Respiratory Irritation of nasal mucosa, pharynx, larynx
Jean et al., 2010; NRC, 1999; WHO, 1981, 2001). and bronchi, pulmonary edema,
tracheobronchitis, bronchial pneumonia,
nasal septum performation
7.1 Lethality Ophthalmic Conjunctivitis

It has been estimated that the acute lethal dose of Sources: WHO, 1981, 2001; ATSDR, 2007; Gorby, 1994; Morton and
ingested inorganic arsenic in humans is about 1-3 mg/ Dunnette, 1994; Chen et al., 1995; NRC, 1999.
kg/day, whereas inhalation and dermal exposures to
inorganic arsenic have not been associated with acute
lethality (ATSDR, 2007). Animals are not as sensitive to damage in the kidney include capillaries, tubules,
inorganic arsenic as humans, and this difference may and glomeruli. Neuropathy is mainly produced by
be due to differences in gastrointestinal absorption and axonal degeneration, although myelin disruption
methylation capability (Vahter, 1994). also presents. Most studies on the acute and subacute
toxicity of inorganic arsenic do not include details
of exposure dose and/or adequacy of sample size to
7.2  Acute and Subacute Effects allow the assessment of dose-response relationships
Both acute and subacute toxicity of arsenic are (ATSDR, 2007; Chen et al., 1997a; Gorby, 1994; IARC,
shown in Table 2. They involve many organ sys- 2004; Morton and Dunnette, 1994; NRC, 1999, 2001;
tems, including the gastrointestinal, dermal, ner- WHO, 1981, 2001).
vous, renal, hepatic, hematological, cardiovascular,
respiratory, and ophthalmic systems. The earli-
7.3  Chronic Noncardiovascular Effects
est and most common presentation of ingestion of
a large dose of arsenic is the acute gastrointestinal Chronic noncardiovascular effects of inorganic
syndrome, which starts with a metallic or garlic- arsenic also involve multiorgan systems, as shown
like taste associated with a dry mouth, burning lips, in Table 3. The clinical appearance of these clinical
and dysphagia. Gastrointestinal syndrome, which manifestations of arsenic intoxication in humans is
is caused by the paralysis of capillary control in the insidious in onset and depends on the magnitude of
digestive tract, may lead to a decrease in blood vol- the dose and the time course of exposure. A multitude
ume and blood pressure, electrolyte imbalance, and of effects may ensue from interference by arsenic on
shock. In acute arsenic poisoning, the fundamental the action of enzymes, essential cations, and tran-
lesion of endothelial cellular toxicity accounts for scriptional events in cells throughout the human body
the predominant clinical features. Capillary damage (ATSDR, 2007; Chen, 2011b; Chen et al., 1997a; Gorby,
leads to generalized vasodilation, transudation of 1994; IARC, 2004; Morton and Dunnette, 1994; NRC,
plasma, and multiorgan failure. The sites of arsenic 1999; WHO, 1981, 2001).
28 Arsenic 599

TABLE 3  Chronic Noncardiovascular Toxicity of Arsenic dose-related increases in characteristic skin lesions in
human populations consuming arsenic-contaminated
Organ System Symptoms and Signs
drinking water. More recent studies by Dastgiri et al.
Dermal Hyperpigmentation with depigmentation, (2010) in Iran have reported marked elevations of
facial edema, palmoplantar hyperkeratosis, arsenical-induced skin lesions among persons living
desquamation in a village with elevated concentrations of arsenic in
Gastrointestinal Esophagitis, gastritis, colitis, abdominal
the drinking water relative to those in another village
discomfort, anorexia, malabsorption, weight
loss with no such exposure. Other studies in Bangladesh
Neural Hearing loss, mental retardation, encephalopathy, (Lindberg et al., 2010), Nepal (Maden et al., 2011),
symmetrical peripheral polyneuropathy and Inner Mongolia (Mao et al., 2010) have reported
(sensimotor type, resembling Landry- similar skin lesions related to arsenic exposures from
Guillain-Barré syndrome), electromyographic
drinking water. Chronic arsenic exposure from drink-
abnormalities
Hepatic Cirrhosis, hepatomegaly, portal hypertension ing water was associated with an increased incidence
without cirrhosis, fatty degeneration of skin lesions even at low levels of arsenic exposure
Hematological Bone marrow hypoplasis, aplastic anemia, (50.1-100 μg/L) in Bangladesh (Argos et al. 2011).
anemia, leukopenia, thrombocytopenia, Overall, these findings indicate the increasing recog-
impaired folate metabolism, karyorrhexis
nition of the global nature of public health problems
Respiratory Rhinopharyngolaryngitis, tracheobronchitis,
pulmonary insufficiency (emphysematous related to arsenical contamination of drinking water,
lesions), chronic restrictive/obstructive as documented by the classic clinical manifestations
diseases of arsenical toxicity.
Metabolic Diabetes mellitus
Immunological Decreased progression of lymphocytes from S
7.3.2  Gastrointestinal Effects
phase to M phase of the cell cycle
Ophthalmic Lens opacity Long-term, low-dose exposure to ingested arsenic was
reported to induce various gastrointestinal symptoms,
Sources: WHO, 1981, 2001; ATSDR, 2007; Gorby, 1994; Morton and including gastroenteritis, dyspepsia, nausea, diarrhea,
Dunnette, 1994; Chen et al., 1997a; NRC, 1999; Chen et al., 1995. anorexia, and abdominal discomfort in Japan, Chile,
China, India, and Mexico. However, the arsenic dose and
7.3.1  Dermal Effects exposure duration required to induce the gastrointestinal
effects were not well characterized (IARC, 2004).
The most distinct characteristics of arsenic toxic-
ity are the classical cutaneous manifestations, includ-
7.3.3  Neural Effects
ing hyperpigmentation with depigmentation and
palmoplantar hyperkeratosis. These have been con- Peripheral polyneuropathy is often among the
sistently observed among those with occupational, sequelae of acute oral arsenic poisoning. However,
environmental, and medicinal exposures to arsenic the occurrence of the polyneuropathy is inconsistently
through inhalation and ingestion. Dermal effects seen in individuals chronically exposed to arsenic at
were most commonly reported after the ingestion low concentrations. Abnormal electromyographic
of arsenic-containing water in various regions of findings mostly suggestive of sensory neuropathy
the world, including Argentina, Bangladesh, Chile, were reported in Canada, India, Japan, and China.
China, Japan, Mexico, Inner Mongolia, and Taiwan However, neuropathy was not reported among resi-
(Chen, 2011a,b; IARC, 2004; NRC, 1999; WHO, 1981, dents drinking arsenic-containing water in Argentina
2001). The magnitude of arsenic dose and exposure and Chile (IARC, 2004; NRC, 1999). There was no sig-
duration needed to induce hyperpigmentation and nificant association between nerve conduction veloc-
hyperkeratosis have only been investigated to a lim- ity measurements and arsenic exposure in subjects
ited extent. A dose-response relationship between the in Alaska (Kreiss et al., 1983) and Utah (Southwick
arsenic concentration in drinking water and the prev- et al., 1983). In contrast, there was a significant asso-
alence of hyperpigmentation and hyperkeratosis was ciation between chronic exposure to arsenic and slow
observed in a survey of 40,421 inhabitants in coastal nerve conduction velocity among adolescents in Tai-
areas of southwestern Taiwan (Tseng et al., 1968) and wan (Tseng et al., 2006). Peripheral neuritis and cog-
in a survey of 7683 residents of West Bengal, India nitive and memory impairment have been reported
(Guha Mazumder et al., 1998b). More recent studies in arsenic-exposed residents in Texas (Kilburn, 1997),
in Inner Mongolia (Guo et al., 2001, 2006), Bangladesh whereas reduced intellectual function has been found
(Ahsan et al., 2006), and West Bengal (Haque et al., to be associated with arsenic exposure from drink-
2003; Mukherjee et al., 2005) have reported similar ing water in Thailand (Siripitayakunkit et al., 1999)
600 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

and Bangladesh (Wasserman et al., 2004). A cross- 7.3.6  Respiratory Effects


sectional study on neurobehavioral development in
Chronic respiratory effects of arsenic have been
adolescents in northeastern Taiwan found a signifi-
primarily reported as a result of occupational expo-
cant association with cumulative arsenic exposure
sure through inhalation of arsenic-containing dust or
for pattern memory and switching attention but not
fumes (Morton and Dunnette, 1994), as well as envi-
for continuous performance and symbol digit (Tsai
ronmental exposure through the ingestion of arsenic-
et al., 2003). Significantly decreased motor function
containing drinking water (IARC, 2004; NRC, 1999).
in neurobehavioral tests has been reported in chil-
Respiratory effects included rhinitis, pharyngitis, lar-
dren exposed to arsenic in drinking water (Parvez
yngitis, tracheobronchitis, chronic cough, crepitation,
et al. 2011). Hamadani et al.(2011) found adverse
shortness of breath, and chronic restrictive and/or
effects of early life arsenic exposure on the intelli-
obstructive lung diseases. More recent studies in West
gence quotient of girls, but not of boys at 5 years of
Bengal (Guha Mazumder et al., 2005; von Ehrenstein
age. O’Bryant et al. (2011) also reported an associa-
et al., 2005) and Chile (Adonis et al., 2005; Smith et al.,
tion between long-term, low-level arsenic exposure
2006) reported increased rates of both obstructive lung
and poorer neuropsychological functioning in adults
lesions, including bronchiectasis and lung cancer in
and seniors.
persons exposed to arsenic through drinking water
that were exacerbated by smoking. Adonis et al. (2005)
7.3.4  Hepatic Effects
reported that in men, the estimated risk of lung cancer
Chronic exposure to arsenic in medicines and correlated with the CYP 1A1*2A (MspI) polymorphism
drinking water was reported to induce liver cir- but not with the GSTM1 null genotype. Other studies
rhosis, portal hypertension without cirrhosis, have found dose-response relationships between arse-
and fatty degeneration (IARC, 2004; Morton and nic drinking water exposures and clinical symptoms
Dunnette, 1994; NRC, 1999; WHO, 1981, 2001). Vine- of respiratory diseases in Banglandesh (Parvez et al.,
yard workers who ingested an arsenic-contaminated 2010) and decrements of lung function in arsenic-
wine were found to have a high prevalence of cir- exposed populations living along the Indus River in
rhosis (Lüchtrath, 1983). The simultaneous use of Pakistan (Nafees et al., 2011). Arsenic exposure during
excessive ethanol may also contribute to the occur- pregnancy increased the risk of lower respiratory tract
rence of liver cirrhosis. Increased mortality from infection during infancy (Rahman et al., 2011).
liver cirrhosis was also observed in two cohorts of
arsenic-exposed copper smelter workers (Axelson 7.3.7  Metabolic Effects
et al., 1978; Welch et al., 1982). Hepatomegaly has
An increased prevalence of diabetes mellitus was
also been reported in cases of chronic arsenic toxic-
observed among residents in an endemic area of arse-
ity caused by drinking arsenic-contaminated water
niasis in southwestern Taiwan, and there was a dose-
in India and China (Guha Mazumder et al., 2005;
response relationship between cumulative arsenic
IARC, 2004; Guha Mazumder and Dasgupta , 2011).
exposure and the prevalence of diabetes mellitus (Lai
More recent studies on arsenic-exposed populations
et al., 1994; Tseng et al., 2000). There was excess mor-
in Bangladesh (Islam et al., 2011) reported a clear
tality from diabetes mellitus among residents who
dose-response relationship between arsenic drink-
lived in an arseniasis-endemic area of southwestern
ing water exposures and both (1) arsenic in hair
Taiwan (Tsai et al., 1999). Arsenic-exposed workers in
and nails and (2) elevated activities of liver serum
both copper smelter and glass-producing plants were
enzyme such as alkaline phosphatase and alanine
reported to have an increased risk of diabetes melli-
and aspartate aminotransferases.
tus (Rahman and Axelson, 1995; Rahman et al., 1996).
A dose-response relationship was reported between
7.3.5  Hematological Effects
the prevalence of diabetes mellitus and that of arse-
Long-term exposure to arsenic has a general depres- nic in drinking water in Bangladesh (Rahman et al.,
sant effect on the hematopoietic system. Hematological 1998, 1999a), and between the incidence of diabetes
changes include normochromatic normocytic anemia, mellitus and the presence of arsenic in drinking water
megaloblastic anemia, erythrocyte karyorrhexis, gran- in southwestern Taiwan (Tseng et al., 2000, 2002). Sig-
ulocytopenia, thrombocytopenia, aplastic anemia, and nificantly higher levels of glycosylated hemoglobin
myelodysplasia. However, no hematological abnor- were seen in arsenic-exposed workers than in unex-
mality was found to be associated with chronic arsenic posed ones in Denmark (Jensen and Hansen, 1998).
exposure through ingestion in populations in Alaska, More recent epidemiological studies on this impor-
Arizona, Michigan, and Utah (Morton and Dunnette, tant public health area have shown differences among
1994; NRC, 1999). arsenic-exposed populations in various countries.
28 Arsenic 601

Cross-sectional studies by Chen et al. (2010b) found stimulation of the diabetic response is that, even at
no association between drinking water arsenic expo- low dose levels, arsenic inhibition of mitochondrial
sure and the prevalence of diabetes mellitus in a respiration will tax the finite capacity of hormonal/
population cohort. In contrast, studies by Del Razo intracellular carbohydrate regulatory systems to
et al. (2011) showed a positive association in a cohort compensate for this basic derangement of cellular
living in regions of endemic arsenicosis in Mexico, metabolism. The ability of these regulatory systems
which was linked to production of the dimethylar- to regulate carbohydrate metabolism would eventu-
sinite (DMAsIII) urinary metabolite. Using data from ally be overwhelmed, resulting in the development
the Korean NHANES study, which is representative and/or exacerbation of an arsenic-induced insulin-
of the general Korean population, Kim and Lee (2011) resistant diabetic state in susceptible individuals.
also reported a positive association between total This would be of particular concern under chronic
urinary arsenic concentrations and the prevalence of exposure conditions.
diabetes mellitus. Differences among the findings of
these studies may be explained in part by genetic dif- 7.3.8  Immunological Effects
ferences in the production of the DMAsIII metabolite
produced by polymorphisms in the AS3MT, and or The peripheral blood lymphocyte count of arsenic-
a combined effect of variations in the GST or HO-1 exposed subjects was slightly increased relative to the
enzyme systems, as discussed above. Previous epide- unexposed controls, and the progression of lympho-
miological studies are consistent with earlier experi- cytes from the S phase to M phase of cell cycle after
mental animal studies (Ghafghazi et al., 1980; Schiller phytohemagglutinin incubation was decreased (Gon-
et al., 1981), which demonstrated marked exacer- sebatt et al., 1994). The retardation in cell replication
bating interactive effects between arsenic exposure, of lymphocytes was much more striking in arsenic-
altered insulin responsiveness, and urinary markers induced skin cancer patients than in matched controls
of diabetes in an alloxan-induced diabetes rodent (Hsu et al., 1997).
model. Other studies (Diaz-Villaseñor et al., 2006)
have reported impaired insulin secretion and tran- 7.3.9  Ophthalmic Effects
scription in rat pancreatic cells treated in vitro with Long-term exposure to arsenic was reported to be
arsenite (AsIII) over a concentration range of 0.5- associated with an increased prevalence of lens opac-
10 μmol/L As for 72 or 144 h. They observed both ity. There was a dose-response relationship between
altered cellular insulin secretion at 144 h in response cumulative arsenic exposure and the prevalence of
to glucose and decreased insulin mRNA expression at posterior subcapsular opacity, but not with the prev-
72 h at the 5 μmol/L As concentration. Possible under- alence of nuclear opacity and cortical opacity, after
lying mechanisms for arsenic-induced diabetes have adjustment for age, gender, diabetes mellitus, and
been reviewed by Tseng (2004). More recent studies sunlight exposure (See, 2000). There was a significant
(Paul et al., 2011) in mice exposed to inorganic arsenic association between arsenic in drinking water and pte-
in drinking water for 20 weeks and fed either a low-fat rygium, a fibrovascular growth of the bulbar conjunc-
or high-fat diet showed that a synergistic interaction tiva and underlying subconjunctival tissue that may
between arsenic exposure and a high-fat diet induced cause blindness, in southwestern Taiwan (Lin et al.,
obesity and glucose intolerance. In addition to these 2008).
suggestions, which are centered on altered molecu-
lar regulatory mechanisms, it should be noted that
7.3.10  Renal Effects
the metabolic impairments of carbohydrate metabo-
lism at the level of mitochondrial respiration (noted Proteinuria has been recognized as a marker for an
in Section 5.6) might represent an important basic increased risk of chronic renal disease. An association
aspect of altered glucose regulation by arsenic. This between exposure to arsenic in drinking water and
is reasonable, because mitochondria actively take up proteinuria has been reported in a Bangladeshi popu-
arsenic, thus concentrating this element at even low lation with low-to-moderate levels of arsenic exposure
dose levels. The mitochondrion is also known to be (Chen, 2011b)
the major intracellular source of ROS in cells. The spe-
cific inhibition of NAD-linked substrate respiration/
7.4  Chronic Cardiovascular Effects
loss of respiratory control by arsenic at low dose lev-
els has been shown to increase the formation of ROS, Table 4 shows chronic cardiovascular effects of
which will stimulate a number of the stress protein long-term exposure to arsenic through ingestion
and intracellular signaling mechanisms on a second- or inhalation. The cardiovascular effects included
ary basis. The overall concept with regard to arsenic electromyographic abnormalities, especially QT
602 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

TABLE 4  Chronic Cardiovascular Toxicity of Arsenic changes in the skin, as well as subjective symptoms
of ischemia (i.e. intermittent claudication, pain at
Organ System Symptoms and Signs
rest, and ischemic neuropathy). Not all patients are
Heart Arrythmias, pericarditis affected by black, mummified dry gangrene (Tseng
Peripheral artery Blackfoot disease (gangrene with et al., 1961). Extensive pathological study showed
spontaneous amputation), Raynaud that 30% of Blackfoot disease patients had histologi-
disease, acrocyanosis, intermittent
cal lesions compatible with thromboangiitis oblit-
Coronary artery Ischemic heart disease
Cerebral artery Cerebral infarction erans, and 70% showed changes of arteriosclerosis
Atherosclerosis Carotid atherosclerosis obliterans. Marked generalized atherosclerosis was
Blood pressure Hypertension observed in all autopsied cases of Blackfoot disease,
Microcirculation Microcirculation abnormalities and the fundamental vascular changes of the disease
represent an unduly developed severe arteriosclero-
Sources: WHO, 1981, 2001; ATSDR, 2007; Engel et al., 1994; Tseng sis (Yeh and How, 1963).
et al., 1995; Chen et al., 1997a; NRC, 1999; Chiou et al., 2005. The dose-response relationship between ingested
inorganic arsenic and Blackfoot disease has been
well documented in the endemic area of southwest-
ern Taiwan, where residents had used high-arsenic
prolongation and increased dispersion; peripheral, artesian well water for more than 50 years (Tseng,
coronary, and cerebral artery diseases; carotid athero- 1977). Patients affected with Blackfoot disease have
sclerosis; hypertension; and microcirculation abnor- a high prevalence of arsenic-induced skin lesions,
malities (Chen 2011b; Chen et al., 1997a; IARC, 2004; including hyperpigmentation, hyperkeratosis, and
NRC, 1999; Wang et al., 2007; WHO, 1981, 2001). skin cancers (Tseng, 1997). They also exhibit high
mortality from cancers of the lung, liver, bladder,
7.4.1  Cardiac Effects kidney, and prostate, as well as ischemic heart dis-
Characteristic arsenic-induced electromyographic ease (Chen et al., 1988b). In addition to the duration
abnormalities include decreased nerve conduc- of artesian well water consumption and the arsenic-
tion amplitude with little change in nerve conduc- induced skin lesions, the development of Blackfoot
tion velocity. Pericarditis and arrhythmias have also disease is also associated with undernourishment
been documented as arsenic-induced cardiac effects and a family history of Blackfoot disease (Chen
(Gorby, 1994; Morton and Dunnette, 1994). A dose- et al., 1988b).
response relationship has been reported between In Blackfoot disease, overt peripheral vascular dis-
cumulative arsenic exposure and heart rate-corrected ease was not reported in other arsenic-exposed pop-
increased QT dispersion and QT prolongation from ulations. However, comparable peripheral vascular
the endemic area of arseniasis in southwestern Tai- disorders with varying degrees of severity, including
wan (Wang, C.-H. et al., 2009). The arsenic-induced Raynaud disease, microangiopathies, vasospastic
QT dispersion is associated with atherosclerotic dis- tendency, and acrocyanosis, have also been reported
eases and predicts long-term cardiovascular mortal- among workers who were exposed to inorganic
ity in residents with previous exposure to arsenic arsenic through copper smelting (Lagerkvist et al.,
(Wang et al., 2010). 1986), pesticide handling, wall painting, and wood
burning; among vintners who had consumed arse-
nic-contaminated wine in Germany; among patients
7.4.2  Peripheral Vascular Diseases
treated with arsenic-containing drugs; and among
Blackfoot disease, a unique peripheral arterial dis- inhabitants exposed to high-arsenic drinking water
ease characterized by severe systemic arteriosclerosis, in Poland, Chile, Mexico, Argentina, Japan, Bangla-
as well as dry gangrene and spontaneous amputations desh, and Xinjiang, China (Garcia-Vargas et al., 1994;
of affected extremities and end stages, has been well Chen et al., 1997a; Engel et al., 1994; Hotta, 1989;
documented as a characteristic vascular disease asso- IARC, 2004; Jean et al., 2010; NRC, 1999; Rahman
ciated with long-term arsenic exposure (Chen et al., et al., 1999b; Wang and Huang, 1994; WHO, 1981,
1988b; Jean et al., 2010; Tseng, 1977). Diagnostic cri- 2001). Lynn et al. (2000) reported that NADH oxi-
teria for Blackfoot disease include objective signs of dase activation is involved in oxidative DNA dam-
ischemia (i.e. absence or diminution of arterial pulsa- age to human aorta vascular smooth muscle cells
tions, pallor on elevation, or rubor on dependency of produced by As(III). Such an underlying mechanism
ischemic extremities) and various degrees of ischemic is consistent with the broad spectrum of oxidative
28 Arsenic 603

damage produced by arsenical exposure in a num- the United States and Sweden, among chimney sweeps
ber of organ systems (NRC, 1999). in Sweden and Denmark, among glassblowers in
There was a decline in the incidence of Blackfoot Sweden, and among workers and neighboring residents
disease after the implementation of a surface water of an arsenic refinery in Japan (Chen et al., 1997a; Engel
supply system in the endemic area, and most newly et al., 1994; IARC, 2004; WHO, 1981, 2001).
developed cases were older residents who had con- A significant reverse dose-response relationship
sumed high-arsenic artesian well water (Wang et al., was observed between arsenic-induced ischemic heart
1985). A survey has shown a dose-response relation- disease and serum levels of α- and β-carotene after
ship between cumulative arsenic exposure and subclin- adjustment for age, gender, body mass index, and
ical peripheral vascular disorder detected by Doppler hypertension, as well as the ratio between total choles-
ultrasonography among seemingly normal subjects terol and high-density lipoprotein cholesterol (Hsueh
after the cessation of drinking artesian well water in et al., 1998).
the endemic area of Blackfoot disease in Taiwan. In a
recent ecological study, the prevalence of peripheral 7.4.4 Stroke
vascular disease among diabetic and nondiabetic resi-
A survey carried out in I-Lan County of northeast-
dents was significantly higher in arseniasis-endemic
ern Taiwan showed a significant dose-response rela-
than nonendemic areas in southwestern Taiwan (Wang
tionship between the arsenic concentration in drinking
et al., 2003). Arsenic methylation capacity is associated
water and the prevalence of strokes, especially cerebral
with the arsenic-related peripheral vascular disease
infarction (Chiou et al., 1997). The biological gradient
(Tseng et al., 2005).
of stroke by arsenic in drinking water remained sta-
In a recent study, an increased risk of erectile
tistically significant after adjustment for age, gender,
dysfunction associated with exposure to arsenic in
body mass index, disease status of hypertension and
drinking water was reported (Hsieh et al., 2008). Arse-
diabetes mellitus, cigarette smoking, and alcohol con-
nic-related oxidative stress has been suggested to be a
sumption. In an ecological study in southwestern Tai-
major cause of this male reproductive failure.
wan, increased mortality from stroke was observed
for residents who lived in arseniasis-endemic areas of
7.4.3  Ischemic Heart Diseases southwestern Taiwan (Tsai et al., 1999). In a recent eco-
logical study, the prevalence of coronary artery disease
Both ingested and inhaled inorganic arsenic have
among diabetic and nondiabetic residents was signifi-
been related to increased mortality from cardiovas-
cantly higher in arseniasis-endemic than nonendemic
cular disease, especially ischemic heart disease (Chen
areas in southwestern Taiwan (Wang et al., 2003). In an
et al., 1997a; Engel et al., 1994; IARC, 2004; Wang et al.,
ecological study, exposure to low levels of arsenic was
2007; WHO, 1981, 2001). Mortality from cardiovascu-
thought to be associated with a higher risk of incident
lar disease was significantly higher among residents
stroke (Lisabeth et al., 2010).
in endemic areas of Blackfoot disease than among
the general population in Taiwan (Wu et al., 1989). A
7.4.5  Carotid Atherosclerosis
significant dose-response relationship between the
ingested arsenic level and the risk of ischemic heart In a cross-sectional survey of residents in southwest-
disease was observed in cohort and case-control stud- ern Taiwan, a dose-response relationship between the
ies in southwestern Taiwan (Chen et al., 1996). In a prevalence of carotid atherosclerosis and long-term
recent ecological study, the prevalence of coronary exposure to arsenic from drinking well water was
artery disease among diabetic and nondiabetic resi- observed (Wang et al., 2002). The biological gradients
dents was significantly higher in arseniasis-endemic remained significant after adjustment for age, gender,
than nonendemic areas in southwestern Taiwan (Wang hypertension, diabetes mellitus, cigarette smoking,
et al., 2003). Myocardial infarction has been related to alcohol consumption, waist-to-hip ratio, and serum
high-arsenic drinking water in several autopsy studies levels of total cholesterol and low-density lipoprotein
in Antofagasta, Chile (Chen et al., 1997a; Engel et al., cholesterol. Arsenic methylation capability is associ-
1994). However, details of arsenic exposure and popu- ated with the risk of carotid atherosclerosis associ-
lation at risk were not available for further evaluation ated with arsenic exposure (Huang et al., 2009). More
of the dose-response relationship. recent studies in Taiwan (Hsieh et al., 2011) reported
The association between long-term exposure to arse- an increased risk of carotid atherosclerosis from arse-
nic and increased mortality from cardiovascular disease nic exposure and linked this effect to polymorphisms
has also been reported among copper smelter workers in in arsenic metabolism genes in a case-control study,
604 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

specifically the PNP A-T haplotype and at least vari- 7.4.7  Microcirculation Abnormality
ants of the AS3MT or GSTO1 haplotypes.
Based on laser Doppler flowmetry, seemingly nor-
mal men living in villages where Blackfoot disease was
7.4.6 Hypertension hyperendemic were found to have poorer peripheral
A cross-sectional study in southwestern Taiwan microcirculation than matched controls in nonendemic
reported an increased prevalence of hypertension areas (Tseng et al., 1995). However, the dose-response
among residents in the arseniasis-endemic area relationship between ingested inorganic arsenic and
than those in a nonendemic area, as well as a dose- abnormal peripheral microcirculation was not examined.
response relationship between ingested inorganic
7.4.7.1  Microvascular Diseases
arsenic and the prevalence of hypertension (Chen
et al., 1995). The biological gradient remained sig- In a recent ecological study, the prevalence of micro-
nificant after adjustment for age, gender, diabetes vascular diseases, including renal disease, retinopathy,
mellitus, proteinuria, body mass index, and serum and neurological disorders among diabetic and non-
triglyceride levels. Another study in Bangladesh diabetic residents was significantly higher in arsenia-
also found a dose-response relationship between sis-endemic than nonendemic areas in southwestern
ingested arsenic and hypertension prevalence Taiwan (Wang et al., 2003).
after adjustment for age, gender, and body mass A significant ecological correlation between the
index (Rahman et al., 1999b). Increased hyperten- arsenic content of drinking water and the mortality
sion prevalence was also observed among patients from cancer of the nasal cavity was reported recently
affected with arsenic-induced skin lesions in an (Chen and Wang, 1990). Although perforation of the
area in Chile where well water had a high arsenic nasal septum has been documented among smelter
concentration (Borgono et al., 1977; Zaldívar, 1980). workers exposed to high levels of inorganic arsenic
Arsenic-exposed workers were reported to have a (WHO, 1981), there were no other reports of an asso-
higher systolic blood pressure compared to unex- ciation between nasal cavity cancer risk and exposure
posed workers in Denmark (Jensen and Hansen, to arsenic through inhalation or ingestion.
1998). Arsenic methylation capability is associated
with a risk of arsenic-related hypertension in south-
7.5  Carcinogenic Effects
western Taiwan (Huang et al., 2007). More recent
studies from Taiwan (Wu et al., 2010, 2011) have Ingested and inhaled arsenic through occupational,
shown that the short GT-repeat polymorphic form of environmental, and medicinal exposure is involved in
HO-1 may play a protective role against the risk of the development of several cancers in humans with no
carotid artery atherosclerosis from arsenic exposure apparent organotropism, as shown in Table 5 (Chen
and more generally in blood pressure regulation et al., 1997b; IARC, 2004; NRC, 1999; WHO, 1981,
and cardiovascular mortality risk in hypertensive 2001). Ecological, case-control, and cohort studies
persons with exposure to agents such as arsenic. have shown a significant dose-response relationship

TABLE 5  Various Cancers with Increased Risks Caused by Arsenic Exposure Through Inhalation and Ingestion
Ingestion Inhalation

Cancer Site Drinking Water Contaminated Wine Fowler’s Solution Copper Smelter Pesticide Factory

Skin + + + +
Lung + + + + +
Urinary bladder + + + +
Kidney + + +
Nasal cavity +
Larynx +
Prostate +
Breast +
Hepatic angiosarcoma + + + + +
Hepatocellular carcinoma + + +
Gastrointestial tract + + +
Hematolymphatic system + + +
Brain and nervous system +
28 Arsenic 605

between arsenic in drinking water and increased risk could mediate this health outcome. Banerjee et al.
of cancers of the liver, nasal cavity, lung, skin, urinary (2011) reported that polymorphisms in the TNF and
bladder, kidney, and prostate in southwestern and IL10 [encoding interleukin-10 (IL-10)] gene promoters
northeastern Taiwan (Chen et al., 1988a, 1992, 2004; can modulate arsenic-induced skin lesions and other
Chiou et al., 1995, 2001). Based on sufficient evidence nonskin effects such as ocular and respiratory diseases.
that arsenic in drinking water causes cancers of the They found the GA/AA and TA/AA TNF alleles to be
urinary bladder, lung, and skin in humans, arsenic in associated with an increased risk of these disorders and
drinking water has been classified as a group 1 carcin- the −3575 IL10 allele to be associated with decreased
ogen to humans (IARC, 2004). It should be noted that IL-10 production. Sun et al. (2011) studied the possible
data from a number of laboratories are discussed in interaction between UV light and arsenic exposures in
Section 5.6.3. Recent studies (Luna et al., 2010; Jomova human skin cells. They reported evidence of interplay
et al., 2011; Flora, 2011) have implicated the formation between the oxidative stress effects of arsenic and UV
of arsenical-induced ROS as a common driver for the light on a number of important regulators of apoptosis,
production of cellular oxidative stress, including oxi- leading to enhanced cellular survival and carcinogenic
dative DNA damage, which could lead to cancer out- transformation. Subsequent studies by Sun et al. (2012)
comes in a number of tissues. Cellular responses to this examined the possible roles of stem cells in the devel-
oxidative stress include the induction of stress proteins opment of skin cancers in mice. These authors found
such as HO-1 (Liu et al., 2011). 2.5 times more stem cells in an arsenite-transformed
human skin cell line (HaCaT), suggesting a relation-
ship between a malignant phenotype and increased
7.5.1  Skin Cancer
numbers of stem cells. Other in vitro studies using the
There is clear evidence that long-term oral expo- HaCaT skin cell line (Komissarova and Rossman, 2010)
sure to inorganic arsenic through drinking water, showed that As3+-induced poly (ADP-ribosyl)lation of
grape wine, or medication increases the risk of skin the p53 tumor suppressor gene is a possible mecha-
cancer (Chen et al., 1997b; IARC, 2004; NRC, 1999; nism of arsenic-induced carcinogenesis.
WHO, 1981, 2001). Inhaled arsenic has also been docu-
mented to induce skin cancer among workers produc-
7.5.2  Lung Cancer
ing sheep-dip powder from sodium arsenite (Hill and
Faning, 1948). The largest study of arsenic-induced An increased risk of cancers of the lung and nasal
skin cancer was carried out in southwestern Taiwan, cavity has been associated with long-term exposure to
and a dose-response relationship between the arsenic inorganic arsenic through inhalation and ingestion in a
content of drinking water and the prevalence of skin dose-response relationship (Chen et al., 1997b; IARC,
cancer was observed (Tseng et al., 1968). In addition 2004; NRC, 1999; Pinto et al., 1978; WHO, 1981, 2001).
to long-term exposure to high-arsenic artesian well An excess of deaths caused by lung cancer has been
water, chronic liver disease and malnutritional status observed among workers exposed to inorganic arse-
have recently been reported to be associated with the nic through inhalation in the production and use of
development of arsenic-induced skin cancer (Hsueh pesticides, in gold mining, and in the smelting of non-
et al., 1995). The risk of skin cancer was significantly ferrous metals, especially copper. Individuals living
associated with elevated serum arsenic levels and within several kilometers of inorganic arsenic-emit-
decreased serum β-carotene level in a dose-response ting industries were also reported to have an increased
relationship, but no significant differences in serum risk of lung cancer (Chen et al., 1997b; WHO, 1981,
levels of selenium and zinc were observed between 2001). The dose-response relationship between inhaled
patients affected with arsenic-induced skin cancer and inorganic arsenic and lung cancer risk was mainly
matched healthy controls (Hsueh et al., 1997). Arsenic observed in two large cohorts of copper smelter work-
methylation capability has been found to be associated ers in Anaconda, Montana (Brown and Chu, 1983; Hig-
with the development of skin cancer. Increased skin gins et al., 1981; Lee-Feldstein, 1983), and in Tacoma,
cancer was observed among those who had an elevated Washington (Enterline and Marsh, 1982). The observed
proportion of MMA in the total urinary metabolites of effects might be confounded by cigarette smoking and
inorganic arsenic (Hsueh et al., 1997; Yu et al., 2000). exposures to other chemicals. An interaction between
An increased risk of arsenic-induced skin cancer was inhaled inorganic arsenic and cigarette smoking in
observed among those who had at least one null or the risk of developing lung cancer has been reported
variant genotype of GSTM1, GSTT1, or GSTP1 (Tseng, (Järup and Pershagen, 1991).
1999). More recent studies related to arsenic-induced An increased risk of lung cancer from exposure to
skin cancers have focused on molecular factors that > 100 μg/L arsenic in drinking water has been reported
606 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

among Bangladeshis who smoked (Mostafa et al., period following drinking-water arsenic exposures in
2008). A significant association between ingested arse- Chile and 25 years after reductions in exposures was
nic and lung cancer risk has been observed in patients reported by Yuan et al. (2010). Alterations in urinary
treated with arsenic-containing medicine, in vintners arsenic methylation profiles have also been reported
exposed to arsenic pesticide-contaminated grape wine, 15 years after cessation of drinking water exposures
and in persons exposed to inorganic arsenic from with a higher percentage of MMA(V) in the urine in
drinking water (Chen et al., 1997b; Guo, 2004; IARC, an elderly cohort subject from Taiwan (Huang et al.,
2004; NRC, 1999; WHO, 1981, 2001). A dose-response 2009), indicating that the arsenical methylation pro-
relationship between ingested inorganic arsenic and cess in humans is also dynamic and may change as a
lung cancer has been reported in cohort studies in function of age. Possible explanations for latency in
Taiwan (Chiou et al., 1995), Japan (Tsuda et al., 1995) arsenic-induced cancers may be related to epigenetic
and most recently in Argentina based on studies in mechanisms, which are discussed in the mechanisms
Cordoba, Argentina (Steinmaus et al., 2010), which of toxicity section of this chapter. Recent studies from
reported that persons exposed to arsenic in drinking Taiwan (Wang, C.-H. et al., 2009; Wang, Y. H. et al.,
water and whose percentage MMA was in the high- 2009; Chung et al., 2011) and the USA (Lesseur et al.,
est quartile had an elevated risk of lung cancer relative 2012) report a link between (1) susceptibility to arsenic-
to those in the lowest quartile. The data suggest the induced cancer of the bladder and urothelial system
importance of differences in arsenical metabolism in and (2) specific GST and AS3MT alleles. Wang, Y. H.
relation to the risk of lung cancer. Both cigarette smok- et al. (2009) reported an increased risk of urothelial
ing and signs of arseniasis (i.e. skin cancer or Blackfoot cancers in arsenic-exposed persons with diplotypes
disease) were reported to increase the risk of arsenic- GSTO1 and GSTO2. Persons with chemical exposures
induced lung cancer in these two studies. A synergis- from cigarette smoking, alcohol consumption, arsenic,
tic effect on the development of lung cancer between and occupational exposures had the highest risk factor
cigarette smoking and ingested arsenic was observed and were likely to have two or more genotypes/dip-
in the endemic areas of arseniasis in southwestern and lotypes of CYP2E1, GSTO1, and GSTO2. Subsequent
northeastern Taiwan (Chen et al., 2004). studies by Chung et al. (2011) focused on GSTO1 and
These results are consistent with the positive inter- GSTO2 alleles and found a positive significant asso-
active effects between benzo(a)pyrene and arsenic in ciation between total urinary arsenic, percentage of
producing lung cancers in hamsters after concomitant inorganic arsenic, percentage of MMA, and urothelial
exposures to both agents (Pershagen et al., 1984). cancers. The DMA percentage was inversely related to
urothelial cancers. The MMA percentage was found
to be associated with the wild-type form of GSTO1
7.5.3  Urothelial Cancer
(140ALA/ALA), compared to those with the GSTO1
Both inhaled and ingested inorganic arsenic have (Ala/Asp) or GSTO1 (140Asp/Asp) genotypes. The
been well documented to induce cancers of the bladder GSTO2 (Asp/Asp) allele was found to be inversely
and kidney, especially urothelial cancers (Chen et al., associated with urothelial cancer risk. In vitro studies
1997b, 1985, 1986; IARC, 2004; NRC, 1999; WHO, 2001). by Kojima et al. (2009) showed that arsenic biometh-
Recent studies by Yokohira et al. (2011) reported dose- ylation was a requirement for oxidative DNA damage
related As3+-induced effects on the bladder urothelium in UROtsa human urothelial cells exposed to As3+. In
of wild-type and AS3MT-knockout mice exposed to a case-control study of bladder cancer in New Hamp-
As3+ in drinking water for 4 weeks. More pronounced shire, Lesseur et al. (2012) studied a number of gene
cytotoxic effects were observed in AS3MT-knockout alleles associated with the outcomes of arsenic expo-
mice, but a no-effect level of 1.0 ppm was calculated sure and found elevated bladder cancer risk associated
for both strains. There have been several reports of with the GSTO2 (Asn142Asp) and GSTZ1 (Glu32Lys)
an increased risk of bladder cancer among patients homozygous variants. Karagas et al. (2012) screened
treated with Fowler’s solution (Chen et al., 1997b). In bladder cancer cases from an ongoing arsenic case-
a cohort study of patients treated with Fowler’s solu- control study and reported that polymorphisms in sol-
tion, a significant association between the cumulative uble carrier 39 member 2 (SLC39A2), a member of the
dose of arsenic intake and mortality from bladder can- ZIP family of metal transporters, and fibrous sheath
cer was observed (Cuzick et al., 1992). Moselle vint- interacting protein 1 (FSIP1) were potential modifiers
ners who consumed arsenic pesticide-contaminated of arsenic-related bladder cancer.
grape wine were found to have increased mortal- It should be noted that epigenetic mechanisms
ity from bladder cancer (Lüchtrath, 1983). Increased involving DNA methylation and attendant regulation
kidney cancer mortality following a 50-year latency of major gene families involved in arsenic-induced
28 Arsenic 607

cancers form an important area of current research to ingested and inhaled arsenic (Bates et al., 1992; Chen
(Reichard and Puga, 2010) because these processes et al., 1997b; Falk et al., 1981a,b; IARC, 2004; NRC, 1999;
may explain the observed long delays in the formation WHO, 1981, 2001). In these cases, exposure was from
of cancers following cessation of exposure, such as has ingestion of arsenic-contaminated wine, high-arsenic
been reported in Chile. Ecological studies in Taiwan drinking water, and Fowler’s solution, and from inha-
have well documented the dose-response relationship lation through copper smelting and the production and
between ingested arsenic from drinking water and application of arsenic-containing pesticides. Because
mortality from cancers of the urinary bladder and kid- hepatic angiosarcoma is a very rare disease, its associa-
ney (Chen and Wang, 1990; Chen et al., 1988a). A sig- tion with exposures to inorganic arsenic does not seem
nificant dose-response relationship between ingested likely to be a matter of chance.
inorganic arsenic from drinking water and the risk of Long-term exposure to inorganic arsenic through
bladder cancer has been documented in cohort studies ingestion and inhalation has been associated with the
in Taiwan (Chiou et al., 1995, 2001), Japan (Tsuda et al., development of hepatocellular carcinoma. Exposure to
1995), and, more recently, in Finland (Kurttio et al., ingested arsenic was from high-arsenic artesian well
1999), Argentina (Bates et al., 2004), and the United water and arsenic-contaminated grape wine, and that
States (Steinmaus et al., 2006). Those who show signs of inhaled arsenic was from copper smelting (Chen
of arseniasis (i.e. skin cancer or Blackfoot disease) et al., 1997b). Ecological studies have shown a dose-
were found to have an increased risk of bladder cancer response relationship between ingested inorganic
from drinking high-arsenic well water (Chiou et al., arsenic from drinking water and the risk of hepatocel-
1995; Tsuda et al., 1995). Cigarette smoking was sig- lular carcinoma among residents in the endemic area
nificantly associated with the development of arsenic- of arseniasis in southwestern Taiwan (Chen and Wang,
induced bladder cancer in northeastern Taiwan (Yang 1990; Chen et al., 1988a).
et al., 2014), but not in southwestern Taiwan (Chiou
et al., 1995). A significant dose-response relationship
7.5.5  Other Internal Cancers
between bladder cancer risk and arsenic exposure was
observed only among cigarette smokers in the United Inhaled and ingested inorganic arsenic have been
States (Bates et al., 1995). A decreased ratio between associated with an increased risk of gastrointestinal
urinary levels of DMA and MMA was associated with cancers, hematolymphatic malignancies, and malig-
an increased risk of bladder cancer (Chen et al., 2003). nant neoplasms of the nervous system (Chen et al.,
Studies by Steinmaus et al. (2006) support the findings 1997b; IARC, 2004; NRC, 1999; WHO, 1981, 2001).
from Taiwan and demonstrate that persons with higher Excess mortality from cancers of the digestive tract
urinary levels of MMA are at greater risk of developing has been observed among copper smelter workers,
arsenic-induced cancers. Moselle vintners, and residents in the endemic area
Inhaled inorganic arsenic has also been found to of arseniasis in Taiwan. Workers in copper smelters
be associated with the risk of mortality from bladder and pesticide manufacturing plants were reported to
cancer among copper smelter workers in the United have increased mortality from malignant neoplasms of
States and Japan (Bates et al., 1992; Chen et al., 1997b) lymphatic and hematopoietic tissues. Copper smelter
and among residents who lived near a plant roasting workers also had increased mortality from malignant
arsenopyrite in Japan and were exposed to inorganic neoplasms of the brain and nervous system.
arsenic through inhalation and ingestion (Hotta, 1989). There are no reports on the association between
There are no reports examining the dose-response rela- exposure to ingested inorganic arsenic and mortality
tionship between inhaled arsenic and bladder cancer. from malignant neoplasms of the hematolymphatic
Excess mortality from kidney cancer was observed and nervous systems. Ingested inorganic arsenic
among copper smelter workers and patients treated through drinking high-arsenic artesian well water has
with Fowler’s solution (Chen et al., 1997b). A dose- been found to be significantly associated with mortal-
response relationship between ingested inorganic ity from prostate cancer in a dose-response relationship
arsenic and kidney cancer mortality was also observed (Chen and Wang, 1990; Chen et al., 1988a,b). There are
among residents in the endemic area of arseniasis in no reports of an association between inhaled inorganic
Taiwan (Chen et al., 1988a, 1992; Chiou et al., 1995). arsenic and prostate cancer.
A significant ecological correlation between the arse-
nic content of drinking water and mortality from can-
7.5.4  Liver Cancer
cer of the nasal cavity was reported recently (Chen and
Both hepatic angiosarcoma and hepatocellular car- Wang, 1990). Although perforation of the nasal septum
cinoma have been associated with long-term exposure has been documented among smelter workers exposed
608 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

to high levels of inorganic arsenic (WHO, 1981), there (Yorifuji et al., 2011). Long-term exposure to rela-
are no other reports of an association between nasal tively low levels of arsenic from birth may increase
cavity cancer risk and exposure to arsenic through urinary cancer risk much later in life (Chen et al.,
inhalation or ingestion. 2010b). Other recent epidemiological studies (Smith
and Steinmaus, 2009) have highlighted strong asso-
ciations between (1) in utero and early life exposures
7.5.6  Lifetime Cancer Risk Induced by Arsenic
to arsenic in drinking water and (2) the development
As shown in Table 6, the lifetime risk of developing of a spectrum of adverse health effects later in life,
cancers of the skin, lung, bladder, liver, and kidney have including a number of cancers, chronic renal disease
been estimated on the basis of Armitage-Doll multistage and cardiovascular effects. Epidemiological studies
models. The lifetime risk of developing skin cancer from into arsenic-induced development and reproduc-
the ingestion of 1 μg/kg/day inorganic arsenic was esti- tive effects have been reviewed periodically (Chen
mated according to the prevalence of skin cancer among et al., 1997b; NRC, 1999; WHO, 1981, 2001). Babies
residents in an endemic area of arseniasis and an unex- born to female employees of a copper smelter who
posed control area (Tseng et al., 1968). The lifetime risk were exposed to inorganic arsenic through inhala-
of developing lung cancer from 1 μg/kg/day arsenic tion during pregnancy were reported to have an
through inhalation was based on the data from copper increased incidence of congenital malformation and
smelter workers in Anaconda, Montana (Brown and low birth weight; there was also an increase in spon-
Chu, 1983; Higgins et al., 1981; Welch et al. 1982; Lee- taneous abortion (Nordstrom et al., 1979a,b). Low
Feldstein, 1983), and in Tacoma, Washington (Enterline birth weight was also observed among newborns in
and Marsh, 1982; Enterline, et al., 1987). smelter areas relative to those in nonsmelter areas
An excessive risk of lung cancer has been reported (Tabacova et al., 1994).
from oral exposure to 100-300 μg/L arsenic (Chen An increased, but not statistically significant, risk of
et al., 2010a). The lifetime risks of developing cancer coarctation of the aorta was found to be associated with
of the lung, liver, bladder, and kidney from 1 μg/kg/ elevated arsenic levels in drinking water (Zierler et al.,
day inorganic arsenic through ingestion were based 1988). Arsenic in drinking water has been reported
on the mortality data of residents in the endemic to increase the mortality from congenital anomalies
area of arseniasis southwestern Taiwan (Chen et al., of the heart in females, and the mortality from con-
1992). The lifetime risks of developing cancers of the genital anomalies of the circulatory system for both
skin, lung, liver, bladder, and kidney from ingested sexes in the United States (Engel and Smith, 1994).
inorganic arsenic were within a sevenfold range of Arsenic in drinking water was reported to be associ-
magnitude. The risks were practically identical for ated with an increased risk of spontaneous abortion in
both men and women within a twofold range of two studies (Aschengrau et al., 1989; Borzsonyi et al.,
magnitude, indicating no gender difference in arse- 1992), and with a significantly increased risk of still-
nic-induced carcinogenic responses. Arsenic expo- birth in two studies (Borzsonyi et al., 1992; Ihrig et al.,
sure during infancy to contaminated milk powder 1998). Arsenic in drinking water was also reported
may lead to pancreatic and hematopoietic cancer to be associated with increased neonatal mortality in

TABLE 6  Lifetime Risk of Developing Cancers of Skin, Lung, Liver, Bladder, and Kidney Caused by 1 mg/kg/day
Inorganic Arsenic Through Ingestion and Inhalation
Lifetime Risk (per 1000)

Cancer Site Exposure Type Study Area Reference Male Female

Skin Ingestion Taiwan Tseng et al., 1968 3.0 2.1


Lung Inhalation Anacoda Higgins et al., 1981 17.0 –
Lee-Feldstein, 1983 9.8 –
Brown and Chu, 1983 4.6 –
Tacoma Enterline and Marsh, 1980 24.0 –
Ingestion Taiwan Chen et al., 1992 1.2 1.3
Bladder Ingestion Taiwan Chen et al., 1992 1.2 1.7
Kidney Ingestion Taiwan Chen et al., 1992 0.4 0.5
Liver Ingestion Taiwan Chen et al., 1992 0.4 0.4

Sources: EPA, 1984; Chen et al., 1992.


28 Arsenic 609

Chile (Hopenhayn-Rich et al., 2000). Similar findings 7.6.1  Developmental and Reproductive Effects
of increased rates of spontaneous abortion, stillbirth,
7.6.1.1  Teratogenic Effects
and infant mortality have been reported among popu-
lations consuming arsenic-contaminated well water in Until recently, there were very few reports on
Bangladesh (Milton et al., 2005) and West Bengal (von the teratogenicity of inorganic arsenic in humans.
Ehrenstein et al., 2005). Congenital malformations were observed in chil-
dren whose mothers worked, during pregnancy, at a
Swedish copper smelter and were exposed to arsenic,
7.6  Experimental System Cancer Studies other heavy metals, and sulfur dioxide. The observed
No studies of cancer incidence in humans after oral incidence was five times greater than that in children
exposure to organic arsenicals have been reported, but born to other mothers from the same region (Nord-
there are some animal studies into the carcinogenic- strom et al., 1979a,b). However, no conclusion can be
ity of organic arsenicals. In an early 2-year study of drawn as to whether arsenic is responsible for these
roxarsone (3-nitro-4-hydroxyphenylarsonic acid) tox- malformations. Teratogenic effects have been shown
icity in animals, no increase in tumor frequency was to occur after a single administration of a high dose
detected in dogs, rats, or mice given 1.5, 2.9, or 3.8 mg (6-10 mg As/kg body weight) of sodium arsenate to
arsenic/kg/day, respectively (Prier et al., 1963). Life- pregnant golden hamsters (Ferm, 1977). The com-
time studies of roxarsone at doses up to 1.4 mg arse- pound was given intravenously to the hamsters on
nic/kg/day yielded no evidence of carcinogenicity in the eighth day of gestation. Rates of both fetal reab-
male or female mice or female rats, although a slight sorption and malformation increased with increas-
increase in pancreatic tumors was noted in male rats ing doses of the arsenate. The teratogenic effects
(NTP, 1980). Carcinogenesis studies with inorganic were characterized by anencephaly, renal agenesis,
arsenic in hamsters (Inamasu et al., 1982; Pershagen and rib malformations. Similar teratogenic effects
et al., 1984) were also positive, particularly in com- have been induced in mice intraperitoneally injected
bination with other organic carcinogens (see Section with sodium arsenate at a dose of 11 mg As/kg body
7.7). Arnold et al. (2003) exposed male and female rats weight (Hood and Bishop, 1972) and in rats intraperi-
to 0, 50, 400, or 800 ppm MMA and male and female toneally injected with 5-12 mg As/kg body weight
B6C3F1 mice to 0, 10, 50, 200, or 400 ppm MMA in (Beaudoin, 1974). However, in all cases, the doses
the diet for 104 weeks; estimated average daily doses required to cause these effects resulted in significant
were up to 47.3 mg arsenic/kg/day for female rats maternal toxicity or even lethality. Recent studies in
and up to 48.5 mg arsenic/kg/day for female mice. mice, rats, and rabbits found no evidence of develop-
No treatment-related neoplastic changes were seen in mental effects at exposure levels that did not produce
either sex of either species. A similar lack of carcinoge- maternal toxicity (Holson et al., 1999, 2000; Nemec
nicity of MMA was reported by Shen et al. (2003), who et al., 1998; Stump et al., 1999).
exposed male F344 rats to 0, 50, or 200 ppm MMA(V)
7.6.2  Genotoxic Effects and Mutagenicity
in drinking water for 104 weeks. Wei et al. (1999, 2002)
exposed male F344 rats to 0, 12.5, 50, or 200 ppm DMA There have been a large number of studies into the
for 104 weeks in the diet; the average daily doses were genotoxic effects of arsenic. The results are mixed but,
0, 0.03, 0.14, or 0.53 mg arsenic/kg. Increases in the in general, it seems that the inorganic arsenicals are
number of animals with bladder tumors were seen in either inactive or weak mutagens (Jacobson-Kram and
the two highest dose groups. No increases in tumor Montalbano 1985), although they are able to produce
incidence were seen in other organs. Hayashi et al. chromosomal effects (aberrations, sister chromatid
(1998) reported that mice exposed to 400 ppm DMA exchange) in most systems. An increased incidence
for 50 weeks, but not those exposed to 50 or 200 ppm, of chromosome abnormalities has been observed in
showed an increased incidence of papillary adenomas lymphocytes of workers exposed to arsenic (Beckman
and an elevated average number of lung tumors per et al., 1977) and of patients therapeutically treated with
mouse. DMA, which is a major methylated metabo- arsenicals (Petres et al., 1977).
lite of inorganic arsenic, has been reported by Hayashi When cultured human lymphocytes were exposed
et al. (1998) to exhibit tumorigenicity and to stimu- to NaAsO2 (trivalent) and Na2HAsO4 (pentavalent),
late tumor progression in mice after drinking water significantly increased frequencies of chromosome
exposure at 400 ppm for up to 50 weeks. These data aberrations were found for trivalent, but not for pen-
are consistent with epidemiological studies of humans tavalent, arsenic (Nordenson et al., 1981). Sodium arse-
who developed lung cancer from the drinking water nite was found to be effective in increasing the rate of
exposures noted above. sister chromatid exchange, but cultured lymphocytes
610 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

obtained from 13 patients with Blackfoot disease did drinking water exposures to lead (25 mg Pb/L), cad-
not differ from those of healthy persons in terms of the mium (10 mg Cd/L), and arsenic (5.0 mgAs/L; the
frequency of sister chromatid exchange after exposure lowest observed effect level doses) for 30, 90, or 180
to sodium arsenite (Wen et al., 1981). Arsenical inter- days produced similar biochemical effects that showed
ference with normal DNA repair processes has also strong dependency on the duration of exposure (Fowler
been noted (Fong et al., 1980; Jung et al., 1969; Jung, et al., 2004). Please see Chapter 11, “Interactions and
1971; Rossman et al., 1977), suggesting that induction Mixtures in Metal Toxicology” and the review by Mad-
of cellular genome damage is a consequence. den and Fowler (2000) for a more complete discussion
In mutagenicity tests using Salmonella and Esch- on interactions among metals.
erichia coli systems and the Chinese hamster cell line Arsenic has been shown to cause an increase in
V79, both trivalent and pentavalent inorganic arsenic total plasma cholesterol in rats; coadministration of
failed to produce point mutations (Lofroth and Ames, chromium(III) counteracts this effect (Aguilar and
1978; Rossman et al., 1980). Arsenic trichloride, sodium Martinez-Para, 1997).
arsenite, and arsenic pentoxide, however, gave posi- A possible interaction among cigarette smoking,
tive results in the rec assay in Bacillus subtilis (Kada inhalation of arsenic, and the risk of lung cancer has
et al., 1980; Nishioka, 1975). not been extensively investigated. Smoking seemed to
Morphological transformation of Syrian hamster increase lung cancer risk synergistically in one study
embryo cells derived from 13- to 14-day-old embryos of smelter workers (Pershagen et al., 1981), although
occurred at concentrations of 2.5-5.0 μg/mL sodium the data are not adequate to exclude a simple additive
arsenate using a cell culture method (DiPaolo and interaction (Thomas and Whittemore, 1988). Cigarette
Casto, 1979). smoking has been shown to increase the occurrence
The genotoxic effects of organic arsenicals such as of lung cancer and urothelial carcinoma in people
DMA and roxarsone have been tested. They are shown exposed to high levels of arsenic in the drinking water
to be able to cause mitotic arrest, chromosome aber- (Chiou et al., 1995; Tsuda et al., 1995; Yang et al., 2014).
rations, mutations and DNA strand breaks (ATSDR, Coexposure to ethanol and arsenic may exacerbate
2007). the toxic effects of arsenic. Simultaneous exposure
of rats to ethanol (10% in drinking water) and arse-
nic for 6 weeks produced a significant increase in the
7.7  Interaction Between Arsenic and Other
concentration of arsenic in the kidney, a nonsignifi-
Compounds
cant increase or arsenic in the liver, and a significant
An interaction between arsenic and selenium has increase in the concentration of glutathione in the liver,
been described by Levander (1977). He concluded that compared with rats treated with either ethanol or arse-
arsenic has a protective effect against the toxicity of nic alone (Flora et al., 1997a,b). Histological damage to
selenium in several species. Likewise, selenium can the liver, but not to the kidneys, was increased in rats
decrease the effects of arsenic, including clastogenic- treated with both ethanol and arsenic compared with
ity, cytotoxicity, and teratogenicity (Babich et al., 1989; those receiving only arsenic.
Biswas et al., 1999; Holmberg and Ferm, 1969). The Please see Chapter 11 for a more detailed discussion.
mechanism of this mutual inhibition of effects is not
known, but may be related to the formation of a com-
plex that is excreted more rapidly than either arsenic 8  DOSE-EFFECT AND DOSE-RESPONSE
or selenium alone (Cikrt et al., 1988; Levander, 1977) or RELATIONSHIP IN ARSENIC POISONING
caused by selenium-induced changes in arsenic meth-
ylation (Styblo and Thomas 2001; Walton et al., 2003). The dose-effect relationship in acute exposure to
Combined exposure to arsenic and lead led to arsenic may be estimated from the 220 poisoning
additive effects on tissue respiration and functional cases associated with an episode of arsenic contamina-
changes in the central nervous system (Novakova, tion of soy sauce in Japan, reported by Mizuta et al.
1969). Mahaffey and Fowler (1977) first reported that (1956). The soy sauce was contaminated with approxi-
rats given cadmium and arsenic (50 mg/kg) in food for mately 0.1 mg As/mL, probably as calcium arsenate.
10 weeks showed lower serum alkaline phosphatase Arsenic intake in the affected individuals was esti-
levels than those treated with either metal alone. The mated by the researchers to be 3 mg/day (0.05 mg/
additive effects of dietary arsenic and lead (200 mg/ kg/day; assuming an average body weight of 55 kg
kg) in coproporphyrin excretion were also subse- for the Asian population). The duration of exposure
quently noted (Fowler and Mahaffey, 1978; Mahaffey was 2-3 weeks in most cases. The primary symptoms
et al., 1981). More recent studies in rats using combined were initially edema of the face and gastrointestinal
28 Arsenic 611

and upper respiratory symptoms, followed by skin water was converted to exposure doses by assuming
lesions and neuropathy in some patients. Other effects a water intake of 4.5 L/day, a body weight of 55 kg,
included mild anemia and leukopenia, mild degenera- and an estimation of arsenic intake of 0.003 mg/As/
tive liver lesions, and hepatic dysfunction, abnormal kg/day from food. The control, low, medium, and
electrocardiogram, and ocular lesions. An acute oral high exposure levels corresponded to doses of 0.0008,
guidance value of 0.005 mg/kg/day (ATSDR, 2007) 0.014, 0.038, 0.065 mg/kg/day, respectively. A clear
may be derived based on the lowest observed adverse dose-response relationship was observed for charac-
effect level (LOAEL) of 0.05 mg/kg/day, by applying teristic skin lesions: 0.0008 mg/kg/day was identified
an uncertainty factor of 10 for extrapolation from a as the NOAEL; at 0.014 mg/kg/day, hyperpigmenta-
LOAEL to a no observed adverse effect level (NOAEL) tion and keratosis of the skin were observed; and at
and 1 for intraindividual variability. This guidance 0.038-0.065  mg/kg/day, an increased incidence of
value is supported by the case of a husband and wife dermal lesions was observed. A chronic health guid-
in upstate New York who experienced gastrointestinal ance value of 0.0003 mg/kg/day may be derived from
symptoms (nausea, diarrhea, and abdominal cramps) the NOAEL of 0.0008 mg/kg/day; an uncertainty fac-
almost immediately after beginning the intermittent tor of 3 was applied in consideration of the fact that
consumption of arsenic-contaminated drinking water most of the control population was less than 20 years
at an estimated dosed of 0.05 mg/kg/day (Franzblau of age and the incidence of skin lesions increased as
and Lilis, 1989). The uncertainty factor of 1 for intra- a function of age, and because the estimates of water
individual variability reflects the fact that the data- intake and dietary arsenic intake are highly uncer-
base includes persons of different ethnicities and age tain (ATSDR, 2007). Schoof et al. (1998) estimated that
groups, including infants. dietary intakes of arsenic from rice and yams might
Hamamoto (1955) reported on Japanese infants who have been 15-211 μg/day (mean, 61 μg/day), based on
ingested milk contaminated with arsenic over a period arsenic analyses of foods collected in Taiwan in 1993-
of 33 days. The amount of arsenic ingested was about 1995. Use of the 50-μg/day estimate would result in
1.3-3.6 mg/day. When a total of about 80 mg arsenic an approximate doubling of the NOAEL (0.0016 mg/
had been ingested, these victims developed symptoms kg/day). This health guidance value is supported by
of arsenic poisoning, including pigmentation, swelling a number of well-conducted epidemiological studies
of the liver, and anemia. A total of 12,131 cases of poi- that identify reliable NOAELs and LOAELs for dermal
soning with 130 deaths have been reported: impaired effects. Southwick et al. (1981) identified a NOAEL of
learning and clinical functions such as hearing were 0.006-0.007 mg/kg/day for dermal lesions in several
described among those who survived (Yamashita small populations in Utah. Harrington et al. (1978)
et al., 1972). identified a NOAEL of 0.003 mg/kg/day for dermal
In areas in Cordoba, Argentina, with endemic effects in a small population in Alaska. Guha Mazum-
arsenicosis, the main symptoms described among der et al. (1988) identified a NOAEL of 0.009 mg/kg/
the inhabitants were symmetrical palmar and plantar day and a LOAEL of 0.006 mg/kg/day for pigmenta-
hyperkeratosis. Concentrations in drinking water have tion changes and hyperkeratosis in a small population
been reported to be 0.9-3.4 mg As/L (Wickstrom, 1972). in India. Haque et al. (2003) identified a LOAEL of
The dose-effect relationship in chronic exposure 0.002 mg/kg/day for hyperpigmentation and hyper-
may be estimated from reports by Tseng et al. (1968) keratosis in a case-control study in India. Cebrian et al.
and Tseng (1977), who investigated the incidence of (1983) identified a NOAEL of 0.0004 mg/kg/day and a
Blackfoot disease and dermal lesions in a large num- LOAEL of 0.022 mg/kg/day in two regions of Mexico.
ber of poor farmers (both males and females) exposed Borgono and Greiber (1972) and Zaldívar (1974) iden-
to high levels of arsenic in well water in Taiwan. A tified a LOAEL of 0.02 mg/kg/day for abnormal skin
control group consisting of over 7500 people was pigmentation in patients in Chile, and Borgono et al.
identified. The incidence of dermal lesions increased (1977) identified a LOAEL of 0.01 mg/kg/day for the
with dose, but individual doses were not provided. same effects in schoolchildren in Chile. Valentine et al.
Incidence data were provided based on stratifica- (1985) reported a NOAEL of 0.02 mg/kg/day for der-
tion of the exposure population into low (< 300 μg/L), mal effects in several small populations in California.
medium (300-600 μg/L), or high (> 600 μg/L) exposure Collectively, these studies indicate that the threshold
groups according to the arsenic concentration in the dose for hyperpigmentation and hyperkeratosis is
well water. Doses were calculated from group mean approximately 0.002 mg/kg/day.
arsenic concentrations in well water, assuming the Fierz (1965) discussed the dose-response relation-
intake parameters described by Abernathy et al. (1989). ship between skin cancer and the total dose of Fowl-
The arithmetic mean concentration of arsenic in well er’s solution ingested as medication. He reviewed 262
612 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

patients who had been treated for weeks or for several and reported that bladder cancer standardized mortal-
years with Fowler’s solution for various chronic der- ity ratio (SMRs) were consistently higher in counties
matoses. A total of 10-2600 mL of the solution had been with documented arsenic exposure; a later case-con-
administered to the patients. Among those patients trol study by the same authors (Bates et al., 2004) did
who developed late effects from this treatment, 40% not report statistically significant increases in blad-
had hyperkeratosis and 8% had skin cancer. A clear der cancers resulting from arsenic exposure, except in
positive relationship between the incidence of both individuals exposed for 50 years or longer. Guo (2000)
hyperkeratosis and cancer and the dose of arsenic was reported significantly increased rate differences for
indicated (Fierz, 1965). bladder cancer in men and women in Taiwan exposed
Dose-response relationships between skin cancer to 0.64 mg arsenic/L in the drinking water, but not at
and arsenic have been discussed by Tseng et al. (1968) lower exposure levels.
and Tseng (1977) for Taiwanese populations exposed to A study of arsenic-exposed individuals in north-
arsenic-contaminated well water. Out of a total popu- ern Chile reported significantly increased odd ratios
lation of 40,421, arsenical skin cancer was found in 428 for lung cancer among subjects with > 30 μg As/L
persons (10.6/1000). There were no patients younger drinking water (Ferreccio et al., 2000), although when
than 20 years of age. The minimal amount of arsenic adjusted for socioeconomic status, smoking, and other
in well water necessary to induce skin cancer was esti- factors, the increase was only significant at 60 μg/L or
mated to be less than 0.29 mg/L. Based on this study, greater. Guo (2004) reported significantly increased
the WHO Task Group on Environmental Health Crite- rate differences for lung cancer for Taiwanese men and
ria for arsenic (WHO, 1981) concluded that the lifetime women exposed to 0.64 mg arsenic/L or greater, with
risk for skin cancer is approximately 25% per mg As/L those older than 50 years of age being particularly at
drinking water. Based on the Taiwan skin cancer data risk. Nakadaira et al. (2002) suggested that even com-
using the multistage model, the U.S. Environmental paratively short exposure durations (less than 5 years)
Protection Agency (EPA) calculated a cancer unit risk might be sufficient for the development of arsenic-
for drinking water of 5 × 10-5 per μg/L (EPA, 2012). The induced lung cancer.
EPA is currently developing the Integrated Risk Infor- The NRC (1999, 2001) analyzed the Taiwan data and
mation System (IRIS) toxicological review of inorganic estimated the excess lifetime risk of bladder and lung
arsenic (cancer and noncancer effects of oral expo- cancer with various arsenic levels in drinking water: at
sures) (EPA, 2012). There is increasingly convincing a concentration of arsenic in drinking water of 3 μg/L,
evidence that long-term exposure to arsenic can result the lifetime risk estimates for bladder and lung can-
in the development of bladder cancer (Bates et al., cer combined are approximately 4 and 10 per 10,000
2004; Chen et al., 1992; Chiou et al., 1995, 2001; Cuzick using the Taiwan or U.S. background rates of these
et al., 1992; Guo, 2000), with transitional cell cancers cancers, respectively. The EPA has not yet calculated
being the most prevalent. Chiou et al. (1995) reported a unit risk value or slope factor for arsenic-induced
a dose-response relationship between long-term arse- internal tumors. As mentioned previously, the EPA is
nic exposure from drinking artesian well water and the currently developing the IRIS toxicological review of
incidence of lung cancer, bladder cancer, and cancers inorganic arsenic (cancer and noncancer effects of oral
of all sites combined (after adjustment for age, gen- exposures) (EPA, 2012).
der, and cigarette smoking) in four townships in Tai- Some data on dose-response relationships for respi-
wan exposed to inorganic arsenic in drinking water ratory cancer have been given by Pinto et al. (1977).
(0-1.14 mg/L). In a later follow-up study of the same They found an almost linear increase in respiratory
cohort, the increase in bladder cancer was found to be cancer with an increasing “exposure index.” From
statistically significant only in subjects exposed for 40 the data, it can be estimated that exposure to airborne
years or longer (Chiou et al., 2001). Cuzick et al. (1992) arsenic of about 50 μg/m3 for more than 25 years might
evaluated a cohort treated with Fowler’s solution in increase the risk for developing respiratory cancer
Lancashire, England, during the period 1945-1969 nearly threefold.
and followed through 1991; the cohort of 478 patients The data must be interpreted with caution because
showed a significant excess of bladder cancer, but no of the possible confounding factors. As for workers
excess of other causes of death. Of a subcohort of 142 engaged in production of insecticides such as lead
patients examined for signs of arsenicism around 1970, arsenate, calcium arsenate, copper acetoarsenite, and
all 11 subsequent cancer deaths occurred in those with magnesium arsenite, a positive dose-response rela-
signs of arsenicism (P = 0.0009). Hopenhayn-Rich et al. tionship between the degree of arsenic exposure and
(1996) investigated bladder cancer mortality for the lung cancer was indicated (Blejer and Wager, 1976;
years 1986-1991 in 26 counties of Cordoba, Argentina, Ott et al., 1974). The ratio of observed to expected
28 Arsenic 613

respiratory cancer deaths ranged from 0.6 in the lowest and reevaluated the effects of exposure. Relative risks
exposure category to 7.0 in the highest. However, this for respiratory cancer increased with increasing dura-
dose-response relationship for lung cancer should be tion. SMRs were significantly elevated after exposure
reevaluated after epidemiological adjustment for the to 0.58 mg/m3 (SMR 3.01, 95% CI 2.0-4.6) or 11.3 mg/
smoking histories of the workers. m3 (SMR 3.68, 95% CI 2.1-6.4) for 10 or more years, and
Järup et al. (1989) reported significantly increased after exposure to 0.29 mg/m3 (SMR 1.86, 95% CI 1.2-
lung cancer mortality [SMR 372, 95% confidence inter- 2.9) for 25 or more years.
val (CI) 304-450] based on 106 lung cancer deaths in a On the basis of the dose-response relationships
cohort of 3916 male workers employed for at least 3 between arsenic exposure and excess lung cancer
months between 1928 and 1967 at the Ronskar smelter mortality in workers at the Anaconda smelter and the
and followed for mortality through 1981. Workers ASARCO smelter, the EPA (2012) derived a unit risk
were separated into low, medium, and high arsenic estimate (the excess of lung cancer associated with life-
exposure groups, with mean TWA exposure estimates time exposure to 1 μg inhaled inorganic arsenic/m3) of
of 0.05, 0.2, and 0.4 mg/m3, respectively. Lung cancer 4.3 × 10−3 per μg/m3.
mortality was significantly increased in all three expo-
sure groups in a concentration-related fashion (SMR of 9  DIAGNOSIS, TREATMENT, AND
201, 353, and 480, respectively). A nested case-control PROGNOSIS
analysis of 102 lung cancer cases and 190 controls from
the cohort showed that lung cancer risk increased When specific exposures have occurred, poison
with increasing arsenic exposure in nonsmokers, light control centers and medical toxicologists should be
smokers, and heavy smokers (Järup and Pershagen, consulted for medical advice. There are several medi-
1991). cal toxicology texts that provide specific information
Enterline and Marsh (1982) reported a significant about clinical treatment after exposures to arsenic
increase in respiratory cancer mortality (SMR 189.4) (Ellenhorn, 1997; Goldfrank et al., 1998; Tintinalli et al.,
based on 104 observed respiratory cancer deaths, with 1996).
only 54.9 expected, over the years 1941-1976 in a cohort
of 2802 male workers employed for at least 1 year
between 1940 and 1964 at the ASARCO smelter. Enter- 9.1  Acute Poisoning
line et al. (1987) reanalyzed these data using improved 9.1.1  Inhalation Diagnosis
exposure estimates that incorporated historical mea-
surements of arsenic in the ambient and personal Acute intoxication caused by the inhalation of arse-
breathing zone of workers. Respiratory cancer mor- nic is unusual except in the case of arsine (see Section
tality was significantly increased in a concentration- 10), although respiratory symptoms may occur among
related fashion in the low (SMR 213.0), medium (SMR workers suddenly exposed to high concentrations of
312.1), and high (SMR 340.9) arsenic exposure groups, arsenic in smelters. Gerhardsson et al. (1988) reported
which had mean estimated TWA arsenic exposures of a case of a smelter worker who inhaled a large quan-
0.213, 0.564, and 1.487 mg/m3, respectively. tity of arsenic-containing dust (80% As2O3) and died
Respiratory cancer mortality was shown to be sig- several hours later. Autopsy findings disclosed wide-
nificantly increased (SMR 285) on the basis of 302 spread hemorrhages in the respiratory tree and con-
observed respiratory cancer deaths between 1938 and gestion of the major organs. Acute skin lesions such
1977 in a cohort of 8045 white male workers employed as contact and allergic dermatitis could be observed
for at least 1 year at the Anaconda smelter (Lee-Feld- around the eyes and mouth, as well as on the face
stein, 1986). Analysis of a subset of the Anaconda cohort and neck. The diagnosis of such cases is based on the
(n = 1800, including all 277 employees with heavy arse- knowledge of a history of arsenic exposure. The mea-
nic exposure) that included information on smoking surement of arsenic in the urine may be useful, pro-
and other occupational exposures showed that lung vided the specimen is taken within a few days after
cancer mortality increased with increasing TWA arse- exposure.
nic exposure, with a small nonsignificant increase in
the “low” group (SMR 138) exposed to 0.05 mg/m3 9.1.1.1  Treatment and Prognosis
and significant increases in the “medium” (SMR 303), Conservative treatment is usually applied to skin
“high” (SMR 375), and “very high” (SMR 704) groups lesions. Dimercaprol/BAL may be given in cases of
exposed to 0.3, 2.75, and 5.0  mg/m3, respectively. severe symptoms of the respiratory system or the
Lubin et al. (2000) recalculated the exposure concen- skin. Chelation therapy is most effective when insti-
tration on the basis of duration and time of exposure tuted within a few hours of exposure, and efficacy
614 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

decreases with increasing time after exposure (McFall normal limits despite the presence of this condition
et al., 1998; Peterson and Rumack, 1977). In gen- (Heyman et al., 1956).
eral, chelating agents should be used with caution Recognition of damage to the upper respiratory
because they may have serious side effects such as tract, specifically perforation of the nasal septum, may
pain, fever, hypotension, and nephrotoxicity. Some be quite helpful in the diagnosis of chronic arsenic poi-
water-soluble and less toxic analogs of BAL such as soning caused by somewhat higher exposures to inor-
dimercaptosuccinic acid (DMSA), dimercaptopro- ganic arsenic.
pyl phthalamadic acid, and dimercaptopropane sul- Liver damage, including cirrhosis and angiosar-
fonic acid are currently under investigation and may coma, which may be seen in an arsenic-exposed indi-
prove to be promising treatments for arsenic poison- vidual, may require differentiation in terms of the
ing (Aposhian et al., 1997; Guha Mazumder, 1996; cause-effect relationship. Among younger people
Kreppel et al., 1995). A recent article by Vantroyen exposed to inorganic arsenic-contaminated food and
et al. (2004) described a case of a massive arsenic tri- water, cardiac and pulmonary symptoms are useful
oxide overdose that was successfully treated by con- for diagnosis.
tinuous gastric irrigation with sodium bicarbonate,
forced diuresis, and the administration of BAL and 9.2.2  Treatment and Prognosis
DMSA.
BAL has been used for the treatment of chronic
arsenic poisoning, particularly in cases of dermatosis.
9.1.2  Ingestion Diagnosis
Pinto and McGill (1953) administered BAL to people
The determination of arsenic in the urine, hair, and, with arsenic dermatitis. They succeeded in augment-
in some cases, blood or stomach contents, is useful ing arsenic excretion into the urine, but not in improv-
for diagnosis if exposure to arsenic cannot be readily ing skin changes such as melanosis and keratosis. A
identified. randomized placebo trial of 2,3-DMSA as a therapy
for chronic arsenicosis caused by drinking contami-
9.1.2.1  Treatment and Prognosis nated water found no significant difference between
The treatment for acute arsenic poisoning by oral patients treated with 2,3-DMSA and those treated
ingestion is referred to in standard medical text- with a placebo (Guha Mazumder et al., 1998a). Apart
books (e.g. Haddad and Winchester, 1990). Prognosis from the often-questionable use of BAL, the treat-
will depend on both dose and elapsed time between ment of chronic arsenic poisoning should generally be
the ingestion of arsenic and the first treatment. In conservative.
some cases where the patient has recovered from Skin changes and neuropathy may persist for a
acute poisoning, dermatitis and peripheral neuritis number of years. The skin changes seen in simple
may persist for a relatively long time (Le Quesne and keratosis may progress into Bowen disease, which will
McLeod, 1977). spread over the entire body in multiple forms.

9.2  Chronic Poisoning 10 ARSINE


9.2.1 Diagnosis
Arsine (hydrogen arsenide, AsH3) is a colorless,
In the diagnosis of chronic arsenic poisoning, it is flammable gas with a slight garlic odor. It is generated
important to differentiate between senile pigmenta- whenever nascent hydrogen is liberated in material
tion, senile leukodermia, and arsenical dermatoses, containing arsenic. Because arsenic is present as an
as well as between peripheral polyneuritis caused by impurity in many metal ores, arsine may be generated
arsenic poisoning and that caused by other diseases. in metal industries, in nonferrous metal refineries, and
This is especially true for individuals exposed to arse- in the manufacture of silicon steel if the ores being pro-
nic through ingestion. cessed accidentally come into contact with acid. Arsine
Concentrations of arsenic in biological tissues may may also be released when hydrogen ions are formed
be within the normal range at the time of diagnosis. by hydrolysis and in the reaction of moisture with
In such cases, a well-designed epidemiological study arsenic-containing dross.
may be required if it is thought that a whole group The toxicity and toxicological mechanisms of arsine
of people might have been exposed to arsenic. White are quite different from those of other inorganic or
striae in the fingernails (Mees lines) are also a useful organic arsenic compounds. Arsine acts as a powerful
clue in the diagnosis of arsenical polyneuritis because hemolytic poison in cases of both acute and chronic
urine and hair arsenic concentrations may be within exposure.
28 Arsenic 615

Henderson and Haggard (1943) observed that the Arsine is used in doping the silicon-based chips and
lethal dose of arsine for humans was 250 mg/m3 for in producing group III semiconductors, such as GaAs
30 min, and that symptoms of poisoning occur after and InAs. Before the recent uses of arsine gas in the
a few hours of exposure to 3-10 mg/m3. Elkins (1959) semiconductor industry, the main concerns regard-
reported a serious, nonfatal, acute case of arsine poi- ing human health were related to its use as a war gas
soning in a person who had worked in an atmosphere and from industrial accidents (Fowler and Weissberg,
contaminated with an average of 0.5 mg arsine/m3. 1974). In the past, the chief clinical effects observed
Five workers, examined by Kipling and Fothergill in persons with acute occupational arsine exposure
(1964), developed typical arsine poisoning when were massive hemolysis followed by death from renal
working in a plant where an acetylene-like odor was failure. Patients typically presented with decreased
detected. This odor occurred during the slag-dissolving hematocrit values and red, “port wine”-colored urine
process that took place in a rotating drum. The concen- because of the presence of hemoglobin. Histopatho-
tration of arsine in this factory was 5 mg/m3. logical evaluations of postmortem kidney samples
Morse and Setterlind (1950) studied two fatal cases demonstrated the presence of renal tubular obstruc-
of arsine poisoning in which aluminum had been used tion from hemoglobin casts that resulted in renal fail-
to extract arsenic. The concentration of arsine in this ure. With the development of renal dialysis, mortality
instance was 70-300 mg/m3. from acute exposure has been greatly reduced in recent
Arsine poisoning is characterized by nausea, decades (Fowler and Weissberg, 1974).
abdominal colic, vomiting, backache, and shortness
of breath, followed by dark, blood-colored urine and
10.1  Experimental Model Studies
jaundice (Kipling and Fothergill, 1964). According to
Bulmer et al. (1940), the following symptoms appeared Prolonged inhalation exposures of rodents to arsine
after various lengths of exposure to arsine: short- at lower sublethal doses (Blair et al., 1990a,b; Fowler
ness of breath on exertion, general malaise, nausea, et al., 1989; Hong et al., 1989; Morrissey et al., 1990;
poor appetite, palpitation on exertion, and headache. Rosenthal et al., 1989) have been shown to produce a
In some groups of workers, a tingling sensation in regenerative anemia with immature erythrocytes and
the feet, chills, garlic breath, changes in complexion, the presence of Howell-Jolley bodies in the peripheral
weakness, vomiting, and drowsiness were observed. blood. Splenomegaly and the characteristic arsenical-
Jaundice was also noted in a number of people. Some induced porphyrinuria (Woods and Fowler, 1978)
of the workers with the longest exposure had anemia. dominated by increased excretion of uroporphyrins
Arsenic in the urine was almost always elevated and with reduced quantities of coproporphyrin (Fowler
ranged from 0.04 to 4.3 mg/L. Even after the workers et al., 1989) are also major manifestations of arsine tox-
had left the arsine-contaminated area, arsenic in urine icity to the hematopoietic system. Other effects include
ranged from 0.04 to 0.1 mg/L. Peripheral neuritis was alterations in immune system function that have been
seen in arsine-exposed workers. Liver disturbances noted in animals exposed to arsine for prolonged peri-
may accompany arsine poisoning. In most fatal cases, ods (Rosenthal et al., 1989), indicating concomitant
renal failure caused by blockage of the renal tubules perturbation of this important biological defense sys-
by hemoglobin casts seems to be the cause of death tem. No negative reproductive and developmental
(Fowler and Weissberg, 1974). Residual functional outcomes are seen in rodents exposed to arsine (Mor-
impairment of the kidneys has been reported in one rissey et al., 1990).
person after an acute episode of arsine poisoning Studies conducted in cell culture systems exposed
(Muehrcke and Pirani, 1968). to arsine in vitro (Ayala-Fierro and Carter, 2000;
The use of BAL for arsine poisoning has not had Carter et al., 2003; Hatlelid and Carter, 1997; Hatlelid
desirable results. Symptomatic therapy, which is gen- et al., 1996; Rael et al., 2000; Winski et al., 1997) have
erally used, is considered more reliable, particularly shown methemoglobin formation and ultrastruc-
in the treatment of anuria. Blood transfusions may be tural effects on erythrocyte membrane structure, with
necessary for severe anemia. Arsine toxicity and treat- concomitant loss of ion regulation capabilities with
ment have been reviewed extensively by Fowler and attendant red cell lysis. Based on these in vivo and
Weissberg (1974). in vitro experimental studies, arsine is clearly a highly
The mechanisms of arsine gas toxicity and more toxic chemical agent that exerts toxicity on a number
recent studies in experimental animal/in vitro model of organ systems. Target cells in the hematopoietic
systems have also been studied over the last 20 years, and immune systems and in the liver, and kidney are
primarily because of possible health effects related to clearly sensitive to arsine toxicity after either short-
its use in the semiconductor industry. term or more chronic exposures. The mechanisms by
616 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

which this agent produces hemolysis seem to require Bates, M.N., Rey, O.A., Biggs, M.L., et al., 2004. Am. J. Epidemiol.
oxygen and the internalization of arsine into the red 159 (4), 381–389.
Beauchemin, D., Siu, K.W.M., McLaren, J.W., et al., 1989. J. Anal. At.
cells with subsequent oxidation and inhibition of Spectrom. 4, 285–289.
membrane ion regulatory mechanisms. Pharmaco- Beaudoin, A.R., 1974. Teratology 10, 153–158.
logical factors such as dose and duration of expo- Beckman, G., Beckman, L., Nordenson, I., 1977. Environ. Health Per-
sure, as well as susceptibility factors such as age and spect. 19, 145–146.
nutritional status of those exposed, will also have an Bencko, V., Symon, K., 1969. J. Hyg. Epidemiol. Microbiol. Immunol.
13, 248–253.
impact on prognosis. Bencko, V., Symon, K., 1970. Atmos. Environ. 4, 157–161.
Bencko, V., Symon, K., 1977. Environ. Res. 13, 378–385.
Bencko, V., Dobisova, A., Macaj, M., 1971. Atmos. Environ 5, 275–279.
References Bencko, V., Benes, B., Cikrt, M., 1976. Arch. Toxicol. 36, 159–162.
Benramdane, L., Accominotti, M., Fanton, L., et al., 1999. Clin. Chem.
Abernathy, C.O., Marcus, W., Chen, C., et al. (1989). Report on ar- 45, 301–306.
senic (As) workgroup meetings. Memo to Peter Cook and Peter Berg, M., Tran, H.C., Nguyen, T.C., et al., 2001. Environ. Sci. Technol.
Preuss, U.S. EPA, 2nd February 1989. 35, 2621–2626.
Adair, B.M., Hudgens, E.E., Schmitt, M.T., et al., 2006. Environ. Res. Bertolero, F., Marafante, E., Rade, J.E., et al., 1981. Toxicology 20,
101 (2), 213–220. 35–44.
Adomako, E.E., et al., 2011. Environ. Pollut. 159, 2435–2442. Bettley, F.R., O’Shea, J.A., 1975. Br. J. Dermatol. 92, 563–568.
Adonis, M., Martínez, V., Marín, P., et al., 2005. Toxicol. Lett. 159, B’Hymer, C., Caruso, J.A., 2004. J. Chromatogr. A 1045, 1–13.
32–37. Biswas, S., Talukder, G., Sharma, A., 1999. Mutat. Res. 441, 155–160.
Aguilar, M.V., Martinez-Para, M.C., 1997. Ann. Nutr. Metab. 41, Blair, P.C., Thompson, M.B., Bechtold, M., et al., 1990a. Toxicology
189–195. 63 (1), 25–34.
Agusa, T., Kunito, T., Tue, N.M., 2012. Metallomics 4 (1), 91–100. Blair, P.C., Thompson, M.B., Morrissey, R.E., et al., 1990b. Fundam.
Ahsan, H., Chen, Y., Parvez, F., et al., 2006. Am. J. Epidemiol. 163 Appl. Toxicol. 14 (4), 776–787.
(12), 1138–1148. Blejer, H.P., Wager, W., 1976. Ann. N.Y. Acad. Sci. 271, 179–186.
Andreae, M.O., 1978. Deep Sea Res. 25, 391–402. Borgono, J.M., Greiber, R., 1972. In: Hemphill, D.D. (Ed.), Trace Sub-
Aposhian, H.V., Arroyo, A., Cebrian, M.E., et al., 1997. J. Pharmacol. stances in Environmental Health-V. University of Missouri Press,
Exp. Ther. 282, 192–200. Columbia, pp. 13–24.
Aposhian, H.V., Gurzau, E.S., Le, X.C., et al., 2000a. Chem. Res. Toxi- Borgono, J.M., Vicent, P., Venturino, H., et al., 1977. Environ. Health
col. 13, 693–697. Perspect. 19, 103–105.
Aposhian, H.V., Zheng, B., Aposhian, M.M., et al., 2000b. Toxicol. Borzsonyi, M., Bereczky, A., Rudnai, P., et al., 1992. Arch. Toxicol.
Appl. Pharmacol. 165, 74–83. 66, 77–78.
Apostoli, P., Bartoli, D., Alessio, L., et al., 1999. Occup. Environ. Med. Braman, R.S., 1977. Environ. Health Perspect. 19, 1–4.
56, 825–853. Braman, R.S., 1983. In: Fowler, B.A. (Ed.). Biological and Envi-
Argos, M., Kalra, T., Pierce, B.L., et al., 2011. Am. J. Epidemiol. 174, ronmental Effects of Arsenic, vol. 6. Elsevier, Amsterdam,
185–194. pp. 141–154.
Argüello, A., Cenget, D., Tello, E., 1938. Argent. Rev. Dermatosyphi- Braman, R.S., Foreback, C.C., 1973. Science 182, 1247–1249.
lol. 22, 461–487. Brooks, W.E., 2003. U.S. Geological Survey minerals yearbook.
Arnold, L.L., Eldan, M., van Gemert, M., et al., 2003. Toxicology 190, USGS, 7.1-7.4, tables 1–3.
197–219. Brooks, W.E., 2005. U.S. Geological Survey minerals yearbook.
Aschengrau, A., Zierler, S., Cohen, A., 1989. Arch. Environ. Health USGS, 24–25.
44, 283–290. Brown, C.C., Chu, K.C., 1983. J. Natl. Cancer Inst. 70, 455–463.
ATSDR (Agency for Toxic Substances and Disease Registry), 2007. Brune, D., Samsahl, K., Wester, P.O., 1966. Clin. Chim. Acta 13, 285–291.
Toxicological Profile for Arsenic. (Updated). Agency for Toxic Brune, D., Nordberg, G., Wester, P.O., 1980. Sci. Total Environ. 16 (1),
Substances and Disease Registry, Atlanta, GA. 41-228. 13–35.
Axelson, O., Dahlgren, E., Jansson, C.D., et al., 1978. Br. J. Ind. Med. Buchet, J.P., Lauwerys, R., 1985. Arch. Toxicol. 57, 125–129.
55, 8–15. Buchet, J.P., Lauwerys, R., 1988. Biochem. Pharmacol. 37, 3149–3153.
Ayala-Fierro, F., Carter, D.E., 2000. J. Toxicol. Environ. Health A 60 Buchet, J.P., Lauwerys, R., Roels, H., 1980. Int. Arch. Occup. Environ.
(1), 67–79. Health 46, 11–29.
Babich, H., Martin-Alguacil, N., Borenfreund, E., 1989. Toxicol. Lett. Buchet, J.P., Lauwerys, R., Roels, H., 1981a. Int. Arch. Occup. Envi-
45, 157–164. ron. Health 48, 71–79.
Bagla, P., Kaiser, J., 1996. Science 274, 174–175. Buchet, J.P., Lauwerys, R., Roels, H., 1981b. Int. Arch. Occup. Envi-
Baker, E.L., Hayes, C.G., Landrigan, P.J., et al., 1977. Am. J. Epide- ron. Health 48, 111–118.
miol. 106, 261–273. Buchet, J.P., Heilier, J.F., Bernard, A., et al., 2003. Int. Arch. Occup.
Banerjee, N., Nandy, S., Kearns, J.K., 2011. Toxicol. Sci. 121, 132–139. Env. Med. 76, 111–120.
Barnes, J.A., Collins, B.W., Dix, D.J., et al., 2002. Environ. Mol. Muta- Budavari, S., O’Neil, M.J., Smith, A., et al. (Eds.), 2001. The Merck
gen 40 (4), 236–242. Index: An Encyclopedia of Chemicals, Drugs, and Biologicals.
Basu, A., Mitra, S., Chung, J., et al., 2011. Environ. Health Perspect. thirteenth ed. Merck & Co., Inc., Whitehouse Station, NJ,
119, 1308–1313. pp. 440–462.
Bates, M.N., Smith, A.H., Hopenhayn-Rich, C., 1992. Am. J. Epide- Bulmer, F.M.R., Rothwell, H.E., Polack, S.S., et al., 1940. Ind. Hyg.
miol. 135, 462–476. Toxicol. 22, 111–124.
Bates, M.N., Smith, A.H., Cantor, K.P., 1995. Am. J. Epidemiol. 141, Burlo, F., Ramírez-Gandolfo, A., Signes-Pastor, A.J., et al., 2012.
525–530. J. Food Sci. 77, T15–T19.
28 Arsenic 617

Bustamente, J., Dock, L., Vahter, M., et al., 1997. Toxicology 118, Chen, C.L., Chiou, H.Y., Hsu, L.I., et al., 2010b. Cancer Epidemiol.
129–136. Biomarkers Prev. 19, 101–110.
Calatayud, M., Vázquez, M., Devesa, V., et al., 2012. Chem. Res. Toxi- Chen, Y.-C., Su, H.-J., Guo, Y.-L., et al., 2003. Cancer Cause Control
col. 25, 2654–2662. 14, 303–331.
Calderon, R.L., Hudgens, E., Le, C.X., et al., 1999. Environ. Health Chilvers, D.C., Peterson, P.J., 1987. In: Hutchinson, T.C., Meema,
Perspect. 107, 663–667. K.M. (Eds.), Lead, Mercury, Cadmium and Arsenic in the Envi-
Caldwell, K.L., Jones, R.L., Verdon, C.P., Jarrett, et al., 2009. J. Expo- ronment. John Wiley & Sons, New York, NY, pp. 279–301.
sure Sci. Enviro. Epi. 19, 59–68. Chiou, H.-Y., Hsueh, Y.-M., Liaw, K.-F., et al., 1995. Cancer Res. 55,
Calvert, C.C., 1975. Arsenical pesticides, 70–80. 1296–1300.
Cannon, R.J., Edmonds, J.S., Francesconi, K.A., et al., 1981. Aust. J. Chiou, H.-Y., Huang, W.-I., Su, C.-L., et al., 1997. Stroke 28,
Chem. 34, 787–798. 1717–1723.
Carapella, S.C., 1992. Kirk-Othmer Encyclopedia of Chemical Tech- Chiou, H.-Y., Chiou, S.-T., Hsu, Y.-H., et al., 2001. Am. J. Epidemiol.
nology. In: Kroschwitz, J.I., Howe-Grant, M. (Eds.). John Wiley 153, 411–418.
and Son, New York, NY. pp. 3, 624-33. Chiou, J.M., Wang, S.L., Chen, C.J., et al., 2005. Int. J. Epidemiol. 34
Carbonell-Barrachina, A.A., Wu, X., Ramírez-Gandolfo, A., et al., (4), 936–943.
2012. Environ. Pollut. 163, 77–83. Chiu, H.W., Lin, J.H., Chen, Y.A., et al., 2010. Autophagy 6, 353–365.
Carter, D.E., Aposhian, H.V., Gandolfi, A.J., 2003. Toxicol. Appl. Chung, C.J., Pu, Y.S., Su, C.T., et al., 2011. Sci. Total Environ. 409,
Pharmacol. 193 (3), 309–334. 465–470.
CDC (United States Centers for Disease Control and Prevention), 2013. Chung, J.S., Kalman, D.A., Moore, L.E., et al., 2002. Environ. Health
National Report on Human Exposure to Environmental Chemicals. Perspect. 110, 729–733.
http://www.cdc.gov/exposurereport/ (accessed 19.02.14). Cikrt, M., Mravcová, A., Malátová, I., et al., 1988. J. Hyg. Epidemiol.
Cebrian, M.E., Albores, A., Aguilar, M., et al., 1983. Hum. Toxicol. 2, Microbiol. Immunol. 32, 17–29.
121–133. Clement, W.H., Faust, S.D., 1973. Environ. Ltrs 5, 155–164.
Challenger, F., 1945. Chem. Rev. 36, 315–361. Concha, G., Nermell, B., Vahter, M., 1998a. Environ. Health Perspect.
Challenger, F., 1951. Adv. Enzymol. 12, 429–491. 106, 355.
Chang, W.-C., Chen, S.-H., Wu, H.-L., et al., 1991. Toxicology 69 (1), Concha, G., Vogler, G., Nermell, B., et al., 1998b. Int. Arch. Occup.
101–110. Environ. Health 71, 42–46.
Charbonneau, S.M., Spencer, K., Bryce, F., et al., 1978. Bull. Environ. Concha, G., Vogler, G., Nermell, B., et al., 2002. Int. Arch. Occup. En-
Contam. Toxicol. 20, 470–477. viron. Health 75, 576–580.
Charbonneau, S.M., Tarn, G.K.H., Bryce, F., et al., 1979. Toxicol. Lett. Conner, E.A., Yamauchi, H., Fowler, B.A., et al., 1993. J. Exposure
3, 107–113. Anal. Environ. Epidemiol. 3, 431–440.
Chen, B., Burt, C.T., Goering, P.L., et al., 1986. Biochem. Biophys. Res. Conner, E.A., Yamauchi, H., Fowler, B.A., 1995. Chem. Biol. Interact.
Comm. 139, 228–234. 96, 273–285.
Chen, B., Hu, B., He, M., et al., 2012. J. Chromatogr.A 1227, 19–28. Cooney, R.V., Mumma, R.O., Benson, A.A., 1978. Proc. Natl. Acad.
Chen, C.-J., 2011a. In: Chen, C.-J., Chiou, H.-Y. (Eds.), Health Haz- Sci. USA 75, 4262–4264.
ards of Environmental Arsenic Poisoning: From Epidemic to Crecelius, E.A., 1977a. Bull. Environ. Contam. Toxicol. 18, 227–230.
Pandemic. World Scientific Publishing, Singapore, pp. 1–20. Crecelius, E.A., 1977b. Environ. Health Perspect. 19, 147–150.
Chen, C.-J., 2011b. Health Hazards of Environmental Arsenic Csanaky, I., Nemeit, B., Gregus, Z., 2003. Toxicology 183 (1-3), 77–91.
Poisoning: From Epidemic to Pandemic. In: Chen, C.-J., Chiou, Cui, X., Kobayashi, Y., Hayakawa, T., et al., 2004. Toxicol. Sci. 82 (2),
H.-Y. (Eds.), World Scientific Publishing, Singapore, pp. 33–49. 478–487.
Chen, C.-J., Wang, C.-J., 1990. Cancer Res. 50, 5470–5474. Cui, X., Wakai, T., Shirai, Y., et al., 2006. Hum. Pathol. 37 (3), 298–311.
Chen, C.-J., Chen, C.-W., Wu, M.-M., et al., 1992. Br. J. Canada 66, Cullen, W.R., 1998. In: Brunnett, J.F., Milolajczyk, M. (Eds.), Arsenic
888–892. in the Environment. Kluwer Academic Publishers, The Nether-
Chen, C.-J., Chiou, H.-Y., Chiang, M.-H., et al., 1996. Arterioscler lands, pp. 123–124.
Thromb. Vasc. Biol. 16, 504–510. Cuzick, J., Sasieni, P., Evans, S., 1992. Am. J. Epidemiol. 136, 417–421.
Chen, C.-J., Chiou, H.-Y., Huang, W.-I., et al., 1997a. In: Abernathy, Dastgiri, S., Mosaferi, M., Fizi, M.A., et al., 2010. J. Health Popul.
C.O., Calderon, R.L., Chappell, W.R. (Eds.), Arsenic: Exposure Nutr. 28 (1), 14–22.
and Health Effects. Chapman & Hall, London, pp. 124–134. Davis, W.C., Zeisler, R., Sieber, J.R., et al., 2010. Anal. Bioanal. Chem.
Chen, C.-J., Hsueh, Y.-M., Chiou, H.-Y., et al., 1997b. Arsenic: 396, 3041–3050.
Exposure and Health Effects. In: Abernathy, C.O., Calderon, R.L., Davison, R.L., Natusch, D.F.S., Wallace, J.R., et al., 1974. Environ. Sci.
Chappell, W.R. (Eds.), Chapman & Hall, London, pp. 232–242. Technol. 8, 1107–1113.
Chen, C.-J., Chuang, Y.-C., Lin, T.-M., et al., 1985. Cancer Res. 45, Del Razo, L.M., García-Vargas, G.G., Valenzuela, O.L., et al., 2011.
5895–5899. Environ. Health 10, 73.
Chen, C.-J., Chuang, Y.-C., You, S.L., et al., 1986. B. J. Canada 53, Demesmay, C., Olle, M., Porthault, M., et al., 1994. Anal. Chem. 348,
399–405. 205–210.
Chen, C.-J., Kuo, T.-L., Wu, M.-M., 1988a. Lancet February 20, Diaz-Villaseñor, A., Sanchez-Soto, M.C., Cebrian, M.E., et al., 2006.
414–415. Toxicol. Appl. Pharmacol. 214 (1), 30–34.
Chen, C.-J., Wu, M.-M., Lee, S.-S., et al., 1988b. Arteriosclerosis 8, DiPaolo, J.A., Casto, B.C., 1979. Cancer Res. 39, 1008–1013.
452–460. Dong, Z., 2002. Environ. Health Perspect. 110 (Suppl. 5), 757–759.
Chen, C.-J., Hsueh, Y.-M., Lai, M.-S., et al., 1995. Hypertension 25, Dousova, B., Erbanova, L., Novak, M., 2007. Sci. Total Environ. 387,
53–60. 185–193.
Chen, C.-L., Hsu, L.-I., Chiou, H.-Y., et al., 2004. JAMA 292, Drobna, Z., Naranmandura, H., Kubachka, K.M., et al., 2009. Chem.
2984–2990. Res. Toxicol. 22, 1713–1720.
Chen, C.L., Chiou, H.Y., Hsu, L.I., et al., 2010a. Environ. Res. 110, Drobna, Z., Waters, S.B., Devesa, V., et al., 2005. Toxicol. Appl. Phar-
455–462. macol. 207 (2), 147–159.
618 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

Drobna, Z., Walton, F.S., Harmon, A.W., et al., 2010. Toxicol. Appl. Focazio, M.J., Welch, A.H., Watkins, S.A., et al., 2000. Water–Resourc-
Pharmacol. 245, 47–56. es Investigations Report 99-4279. USGS, Reston, VA.
Durum, W.H., Hem, J.D., Heidel, S.G., 1971. Geological survey circu- Fong, K., Lee, F., Bockrath, R., 1980. Mutat. Res. 70, 151–156.
lar, 643. U.S. Department of Interior, Washington, DC. Fowler, B.A. (Ed.). 1983. Biological and Environmental Effects of Ar-
Edmonds, J.S., Francesconi, K.A., 1981a. Mar. Pollut. Bull. 12, 92–94. senic, vol. 6. Elsevier, Amsterdam, pp. 1–281.
Edmonds, J.S., Francesconi, K.A., 1981b. Chemosphere 10, Fowler, B.A., Mahaffey, K.R., 1978. Environ. Health Perspect. 25,
1041–1044. 87–90.
Edmonds, J.S., Francesconi, K.A., Cannon, J.T., et al., 1977. Tetrahe- Fowler, B.A., Weissberg, J.B., 1974. N. Engl. J. Med. 291, 1171–1174.
dron Lett. 18, 1543–1546. Fowler, B.A., Woods, J.S., 1979. Toxicol. Appl. Pharmacol. 50,
Edmonds, J.S., Francesconi, K.A., Healy, P.C., et al., 1982. Chem. Soc. 177–187.
Perkin Trans. 1, 2989–2993. Fowler, B.A., Fay, R.C., Walter, R.L., et al., 1975. Environ. Health
Eichler, T., Ma, W., Kelly, C., et al., 2006. Toxicol. Sci. 90 (2), 392–399. Perspect. 12, 71–76.
Eichler, T.E., Randsom, R.F., Smoyer, W.E., 2005. Toxicol. Sci. 84 (1), Fowler, B.A., Woods, J.S., Schiller, C.M., 1979. Lab Invest. 41, 313–320.
120–128. Fowler, B.A., Moorman, M.P., Adkins Jr., B., et al., 1989. Hazard As-
Elkins, H.B., 1959. The Chemistry of Industrial Toxicology, second sessment and Control Technology in Semiconductor Manufac-
ed. John Wiley and Sons, New York. turing. Lewis Publishers, Chelsea, MI, pp. 85–89.
Ellenhorn, M.J., 1997. Ellenhorn’s Medical Toxicology. Diagnosis and Fowler, B.A., Whittaker, M.H., Lipsky, M., et al., 2004. Biometals 17
Treatment of Human Poisoning. Williams & Wilkins, Baltimore, (5), 567–568.
MD. 1538–1542. Fowler, B.A., Conner, E.A., Yamauchi, H., 2005. Toxicol. Appl. Phar-
Engel, R.R., Smith, A.H., 1994. Arch. Environ. Health 49, 418–427. macol. 206, 121–130.
Engel, R.R., Hopenhayn-Rich, C., Receveur, O., et al., 1994. Epide- Franzblau, A., Lilis, R., 1989. Arch. Environ. Health 44, 385–390.
miol. Rev. 16, 184–209. Fujihara, J., Soejima, M., Yasuda, T., et al., 2010. Toxicol Appl Phar-
Enterline, P.E., Marsh, G.M., 1980. Amer.J Indus. Med. 1, 251–259. macol. 243 (3), 292–299.
Enterline, P., Marsh, G., 1982. Am. J. Epidemiol. 116, 895–911. Fujihara, J., Soejima, M., Yasuda, T., et al., 2011. Toxicol Appl Phar-
Enterline, P.E., Henderson, V.L., Marsh, G.M., 1987. Am. J. Epide- macol. 256 (2), 174–178.
miol. 125, 929–938. Fujino, Y., Guo, X., Liu, J., et al., 2005. J. Expo. Anal. Environ. Epide-
EPA (U.S. Environmental Protection Agency), 1982. Office of Water miol. 15 (2), 147–152.
Regulations and Standards. PB85221711. EPA440485005. 1.1– Gabor, S., Coldea, V., 1977. Environ. Health Perspect. 19, 107–108.
4.68. Gallagher, R.E., 1998. N. Engl. J. Med. 339, 1389–1391.
EPA., 1984. US Environmental Protection Agency, EPA-60018-83- Garb, L.G., Hine, C.H., 1977. J. Occup. Med. 19 (8), 567–568.
021F, Assessment for Inorganic Arsenic, Final Report. Garbarino, J.R., Bednar, A.J., Rutherford, D.W., et al., 2003. Environ.
EPA, 2001. Fed. Regist 66, 6976–7066. Sci. Technol. 37, 1509–1514.
EPA., 2005a. STORET data warehouse. U.S. Environmental Protec- García-Montalvo, E.A., Valenzuela, O.L., Sánchez-Peña, L.C., 2011.
tion Agency. http://www.epa.gov/storet/dbtop.html(accessed Toxicol. Mech. Methods. 21 (9), 649–655.
19.02.14). Garcia-Vargas, G.G., Hernandez-Zavala, A., 1996. Biomed. Chro-
EPA. (2005b). U.S. Environmental Protection Agency. Technology matogr. 10, 278–284.
Transfer Network. National Air Toxics Assessment. Pollutant- Garcia-Vargas, G.G., Del Razo, L.M., Cebrian, M.E., et al., 1994. Hu-
Specific Data Tables. http://www.epa.gov/ttnatw01/nata/tabl man Exp. Toxicol. 13, 839–847.
emis.html (accessed 19.02.14). Gerhardsson, L., Dahlgren, E., Eriksson, A., et al., 1988. Scand. J.
EPA., 2012. U.S. Environmental Protection Agency. Arsenic, Inor- Work Environ. Health 14 (2), 130–133.
ganic. Integrated Risk Information System (IRIS). National Cen- Ghafghazi, T., Ridlington, J.W., Fowler, B.A., 1980. Toxicol. Appl.
ter for Environmental Assessment, Washington, D.C. http://ww Pharmacol. 55, 126–130.
w.epa.gov/ncea/iris/subst/0278.htm. Goldbaum, O., Richter-Landsberg, C., 2001. J. Neurochem. 78 (6),
Erry, B.V., Macnair, M.R., Meharg, A.A., et al., 1999. Bull. Environ. 1233–1242.
Contam. Toxicol. 63, 567–574. Goldfrank, R.L., Flomenbaum, N.E., Lewin, N.A., et al., 1998. In:
Falk, H., Caldwell, G.G., Ishak, K.G., et al., 1981a. Am. J. Ind. Med. Goldfrank’s Toxicologic Emergencies, sixth ed. Appleton and
2, 43–50. Lange, Stamford, CT, pp. 1261–1273.
Falk, H., Herbert, J.T., Edmonds, L., et al., 1981b. Cancer 47, 382–391. Gomez-Rubio., P., Meza-Montenegro, M.M., Cantu-Soto, E., et al.,
Farzaneh, P., Allameh, A., Pratt, S., et al., 2005. Altern. Lab. Anim. 33 2010. J. Appl. Toxicol. 30 (3), 260–270.
(2), 105–110. Gong, Z., Lu, X., Ma, M., et al., 2002. Talanta 58, 77–96.
Felix, K., Manna, S.K., Wise, K., et al., 2005. J. Biochem. Mol. Toxicol. Gong, G., O’Bryant, S.E., 2012. Environ. Res. 113, 52–57.
19 (2), 67–77. Gonsebatt, M.E., Vega, L., Montero, R., et al., 1994. Mutat. Res. 313,
Ferguson, J.F., Gavis, J., 1972. Water Res. 6, 1259–1274. 293–299.
Ferm, V.H., 1977. Environ. Health Perspect. 19, 215–217. Gorby, M.S., 1994. In: Nriagu, J.O. (Ed.), Advance in Environmental
Ferreccio, C., Gonzalez, C., Milosavjlevic, V., et al., 2000. Epidemiol- Science and Technology. John Wiley, New York, pp. 1–16.
ogy 11, 673–679. Gottschalg, E., Moore, N.E., Ryan, A.K., et al., 2006. Chem. Biol. In-
Fierz, U., 1965. Dermatologica 131, 41–58. teract. 161 (3), 251–261.
Flora, S.J., 2011. Free Radic. Biol. Med. 51, 257–281. Goussetis, D.J., Altman, J.K., Glaser, H., et al., 2010. J. Biol. Chem.
Flora, S.J., Dube, S.N., Vijayaraghavan, R., et al., 1997a. Biol. Trace 285, 29989–29997.
Elem. Res. 58, 197–208. Grantham, D.A., Jones, J.F., 1977. J. Am. Water Works Assoc. 69,
Flora, S.J.S., Pant, S.C., Malhotra, P.R., et al., 1997b. Alcohol 14, 653–657.
563–568. Guha Mazumder, D.N., 1996. J. Indian Med. Assoc. 94, 41–42.
Flora, S.J., Bhadauria, S., Pant, S.C., et al., 2005. Life Sci. 77 (18), Guha Mazumder, D., Dasgupta, U.B., 2011. Kaohsiung J. Med. Sci.
2324–2337. 27, 360–370.
28 Arsenic 619

Guha Mazumder, D.N., Chakraborty, A.K., Ghose, A., et al., 1988. Hood, R.D., Bishop, S.L., 1972. Arch. Environ. Health 24, 62–65.
Bull. WHO 66, 499–506. Hopenhayn, C., Huang, B., Christian, J., et al., 2003. Environ. Health
GuhaMazumder, D.N., Ghoshal, U.C., Saha, J., et al., 1998a. Clin. Perspect. 111, 1888–1891.
Toxicol. 36, 643–690. Hopenhayn-Rich, C., Biggs, M.L., Fuchs, A., et al., 1996. Epidemiol-
Guha Mazumder, D.N., Haque, R., Ghosh, N., et al., 1998b. Int. J. ogy 7, 117–124.
Epidemiol. 27, 871–877. Hopenhayn-Rich, C., Browning, S.R., Hertz-Picciotto, I., et al., 2000.
Guha Mazumder, D.N., Steinmaus, C., Bhattacharya, P., et al., 2005. Environ. Health Perspect. 108, 667–673.
Epidemiology 16 (6), 760–765. Hotta, N., 1989. Jpn. J. Constit. Med. 53, 49–70.
Guo, H.R., 2000. Environ. Geochem. Health 22, 83–91. Hour, T.-C., Pu, Y.-S., Lin, C.-C., et al., 2006. Anticancer Res. 26 (1A),
Guo, H.R., 2004. Cancer Causes Control 15, 171–177. 375–378.
Guo, X., Fujino, Y., Kaneko, S., et al., 2001. Mol. Cell Biochem. 222 Hsieh, F.-I., Hwang, T.-S., Hsieh, Y.-C., et al., 2008. Environ. Health
(1-2), 137–140. Perspect. 116, 532–536.
Guo, X., Fujino, Y., Ye, X., et al., 2006. Int. J. Environ. Res. Public Hsieh, Y.-C., Lien, L.-M., Chung, W.-T., et al., 2011. Environ. Res. 111,
Health 3 (3), 262–267. 804–810.
Haddad, L.M., Winchester, J.F., 1990. Clinical Management of Poi- Hsu, L.I., Chen, W.P., Yang, T.Y., et al., 2011. J. Biomed. Sci. 18, 51.
soning And Drug Overdose, second ed. W.B. Saunders Company, Hsu, Y.-H., Li, S.-Y., Chiou, H.-Y., et al., 1997. Mutat. Res. 386,
Philadelphia, PA, pp. 1024–1028. 241–251.
Hamadani, J.D., Tofail, F., Nermell, B., et al., 2011. Int. J. Epidemiol. Hsueh, Y.-M., Cheng, G.-S., Wu, M.-M., et al., 1995. Br. J. Cancer 71,
40, 1593–1604. 109–114.
Hamamoto, E., 1955. Nihon Iji Shinpo 1649, 3–12 (In Japanese). Hsueh, Y.-M., Chiou, H.-Y., Huang, Y.-L., et al., 1997. Cancer Epide-
Hammer, D.I., Finklea, J.F., Hendrickson, R.H., et al., 1971. Am. J. miol. Biomarkers Prev. 6, 589–596.
Epidemiol. 93, 84–92. Hsueh, Y.-M., Wu, W.-L., Huang, Y.-K., et al., 1998. Atherosclerosis
Han, S.G., Castranova, V., Vallyathan, V., 2005. Mol. Cell Biochem. 141, 249–257.
277 (1-2), 153–164. Huang, Y.-K., Tseng, C.-H., Huang, Y.-L., et al., 2007. Toxicol. Appl.
Haque, R., Mazumder, D.N., Samanta, S., et al., 2003. Epidemiology Pharmacol. 218, 135–142.
14, 174–182. Huang, Y.-L., Hsueh, Y.-M., Huang, Y.-K., et al., 2009. Sci. Total Envi-
Harrington, J.M., Middaugh, J.P., Morse, D.L., et al., 1978. Am. J. Epi- ron. 407, 2608–2614.
demiol. 108, 377–385. Hueber, D.M., Winefordner, J.D., 1995. Anal. Chem. Acta 316,
Hatlelid, K.M., Carter, D.E., 1997. J. Toxicol. Environ. Health 50 (5), 129–144.
463–474. Hughes, M.F., Kenyon, E.M., Edwards, B.C., et al., 2003. Toxicol.
Hatlelid, K.M., Brailsford, C., Carter, D.E., 1996. J. Toxicol. Environ. Appl. Pharmacol. 191, 202–210.
Health 9 47 (2), 145–157. Hughes, M.F., Devesa, V., Adair, B.M., et al., 2005. Toxicol. Appl.
Hayakawa, T., Kobayashi, Y., Cui, X., 2005. Arch. Toxicol. 79 (4), Pharmacol. 2, 186–197.
183–191. Hughes, M.F., Edwards, B.C., Herbin-Davis, K.M., et al., 2010. Toxi-
Hayashi, H., Kanisawa, M., Yamanaka, K., et al., 1998. Cancer Lett. col. Appl. Pharmacol. 249, 217–223.
125, 82–88. Hung, D.Q., Nekrassova, O., Compton, R.G., 2004. Talanta 64,
Healy, S.M., Casarez, E.A., Ayala-Fierro, F., et al., 1998. Toxicol. Appl. 269–277.
Pharmacol. 148, 65–70. Hunter, F.T., Kip, A.F., Irvine, J.W., 1942. J. Pharmacol. Exp. Ther. 76,
Henderson, Y., Haggard, H.W., 1943. Noxious Gases. Reinholdt, 207–220.
New York. IARC (International Agency for Research on Cancer), 1982. IARC
Heydorn, K., 1970. Clin. Chim. Acta 28, 349–357. Monographs, Supplement 4. Evaluation of the Carcinogenic
Heyman, A., Pfeiffer Jr., J.B., Willett, R.W., et al., 1956. N. Engl. J. Risk of Chemicals to Humans, Chemicals, Industrial Processes
Med. 254, 401–409. and Industries Associated with Cancer in Humans. International
Higgins, I., Welch, K., Oh, M., et al., 1981. Am. J. Ind. Med. 2, 33–41. Agency for Research on Cancer, Lyon, 292 pp.
Hill, A.B., Faning, E.L., 1948. Br. J. Ind. Med. 5, 1–6. IARC., 2004. IARC Monographs Evaluation of Carcinogenic Risks to
Hindmarsh, J.T., 2002. Clin. Biochem. 35, 1–11. Humans: Some Drinking-water Disinfectants and Contaminants,
Hinwood, A.L., Sim, M.R., Jolley, D., et al., 2003. Environ. Health Per- including Arsenic, vol. 84. International Agency for Research on
spect. 111, 187. Cancer, Lyon, 512 pp.
Hirano, S., Kobayashi, Y., Cui, X., et al., 2004. Toxicol. Appl. Pharma- IARC., 2006. IARC Monographs. Evaluation of the Carinogenic Risks
col. 198 (3), 458–467. to Humans: Cobalt in Hard Metals and Cobalt Sulfate, Gallium
Hisanaga, A., 1982. Fukuoka Acta Med. 73, 46–63. Arsenide, Indium Phosphide and Vanadium Pentoxide, vol. 86.
Hogan, R.B., Eagle, H., 1944. J. Pharmacol. Exp. Ther. 80, 93–113. International Agency for Research on Cancer, Lyon, 330 pp.
Holak, W., 1969. Anal. Chem. 41, 1712–1713. IARC, 2012. 512 pp. A Review of Human Carcinogens. C. Metals,
Holland, R.H., Wilson, R.H., Acevedo, A.R., et al., 1959. Acta Union Arsenic, Fibres and Dusts, Monograph on the Evaluation of Car-
Int. Contra Cancrum 15, 608–611. cinogenic Risks to Humans, vol. 100C. International Agency for
Hollins, J.G., Charbonneau, S.M., Bryce, F., et al., 1979. Toxicol. Lett. Research on Cancer, Lyon, France.
4, 7–13. Ihrig, M.M., Shalat, S.L., Baynes, C., 1998. Epidemiology 9, 290–294.
Holmberg, R.E., Ferm, V.H., 1969. Arch. Environ. Health 18, 873–877. Inamasu, T., 1983. Toxicol. Appl. Pharmacol. 71, 142–152.
Holson, J.F., Stump, D.G., Ulrich, C.E., et al., 1999. Toxicol. Sci. 51, Inamasu, T., Hisanaga, A., Ishinishi, N., 1982. Toxicol. Lett. 12, 1–5.
87–97. Irgolic, K.J., Woolson, E.A., Stockton, R.A., et al., 1977. Environ.
Holson, J.F., Stump, D.G., Clevidence, K.J., et al., 2000. Food Chem. Health Perspect. 19, 61–66.
Toxicol. 38, 459–466. Islam, K., Haque, A., Karim, R., et al., 2011. Environ. Health 10, 64.
Hong, H.L., Fowler, B.A., Boorman, G.A., 1989. Toxicol. Appl. Phar- Ito, H., Okamoto, K., Kato, K., 1998. Biochim. Biophys. Acta 1397 (2),
macol. 97 (1), 173–182. 223–230.
620 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

Jacobson-Kram, D., Montalbano, D., 1985. Environ. Mutagen 7, Lai, M.-S., Hsueh, Y.-M., Chen, C.-J., et al., 1994. Am. J. Epidemiol.
787–804. 139, 484–492.
Jarrett, J.M., Jones, R.L., Caldwell, K.L., Verdon, C.P., 2007. Atomic Lam, H.K., Li, K., Chik, K.W., et al., 2005. Int. J. Oncol. 27 (2), 537–545.
Spectro. 28, 113–122. Landsberger, S., Wu, D., 1995. Sci. Total Environ. 173, 323–337.
Järup, L., Pershagen, G., 1991. Am. J. Epidemiol. 134, 545–551. Laskey, T., Sun, W., Kadry, A., et al., 2004. Environ. Health Perspect.
Järup, L., Pershagen, G., Wall, S., 1989. Am. J. Ind. Med. 15, 31–41. 112, 18–21.
Jean, J.S., Bundschuh, J., Chen, C.J., et al., 2010. The Taiwan Crisis: Lau, A.T., He, Q.-Y., Chiu, J.-F., 2004. Biochem. J. 382, 641–650.
a showcase of the global arsenic problem. CRC Press, London. Le, X.C., Lu, X., Li, S.-F., 2004. Anal. Chem. 76, 27A–33A.
Jelinek, C.F., Corneliussen, P.E., 1977. Environ. Health Perspect. 19, Le Quesne, P.M., McLeod, J.G., 1977. J. Neurol. Sci. 32, 437–451.
83–87. Lee, C., Lee, Y.M., Rice, R.H., 2005. Chem. Biol. Interact. 155 (1-2),
Jensen, G.E., Hansen, M.L., 1998. Analyst 123, 77–80. 43–54.
Jomova, K., Jenisova, Z., Feszterova, M., et al., 2011. J. Appl. Toxicol. Lee-Feldstein, A., 1983. J. Natl Cancer Inst. 70, 601–610.
31, 95–107. Lee-Feldstein, A., 1986. J. Occup. Med. 28, 296–302.
Jou, M.J., Peng, T.I., Reiter, R.J., et al., 2004. J. Pineal Res. 37 (1), 55–70. Lerman, S., Clarkson, T.W., 1983. Fundam. Appl. Toxicol. 3, 309–314.
Jung, E., 1971. Z. Haul. Geschlechtskr 46, 35–36. Lerman, S., Clarkson, T.W., Gerson, R.J., 1983. Chem. Biol. Interact.
Jung, E., Trachsel, B., Immich, H., 1969. Ger. Med. Man 14, 614–616. 45, 401–406.
Jutooru, I., Chadalapaka, G., Sreevalsan, S., et al., 2010. Exp. Cell Res. Lesseur, C., Gilbert-Diamond, D., Andrew, A.S., et al., 2012. Toxicol.
316, 2174–2188. Lett. 210, 100–106.
Kada, T., Hirano, K. and Shirasu, Y. (1980). In F.J. de Serres and A. Levander, O.A., 1977. Environ. Health Perspect. 19, 159–164.
Hollaender (eds.) Chemical mutagens (pp. 149–173). Springer U.S. Lewis, A.S., Reid, K.R., Pollock, M.C., et al., 2012. J. Air Waste Man-
Kadowaki, K., 1960. Osaka City Med. J. 9, 2083. ag. Assoc. 62, 2–17.
Kala, S.V., Kala, G., Prater, C.I., 2004. Chem. Res. Toxicol. 17 (2), Li, C.Y., Lee, J.S., Ko, Y.G., et al., 2000. J. Biol. Chem. 275 (33),
243–249. 25666–25671.
Karagas, M.R., Andrew, A.S., Nelson, H.H., et al., 2012. Hum. Genet. Li, J., Waters, S.B., Drobna, Z., et al., 2005. Toxicol. Appl. Pharmacol.
131, 453–461. 204 (2), 164–169.
Karthiheyan, S., Hirata, S., 2003. Anal. Lett. 36, 2355–2366. Li, M., Cai, J.F., Chiu, J.F., 2002. J. Cell Biochem. 87 (1), 29–38.
Kato, K., Ito, H., Okamoto, K., 1997. Cell Stress Chaperones 2 (3), Liebscher, K., Smith, H., 1968. Arch. Environ. Health 17, 881–890.
199–209. Lin, C.J., Wu, M.H., Hsueh, Y.M., et al., 2005. Cancer Chemother.
Katsura, K., 1958. Shikoku Igaku Zasshi 12, 706–720. Pharmacol. 55 (2), 170–178.
Kawakami, Y., 1967. Onsen Kagaku 17, 58–82 (In Japanese). Lin, W., Wang, S.-L., Wu, H.-J., et al., 2008. Environ. Health Perspect.
Kedderis, G.L., Elmore, A.R., Crecelius, E.A., et al., 2006. Chem. Biol. 116, 952–955.
Interact. 161 (2), 139–145. Lindberg, A.L., Sohel, N., Rahman, M., et al., 2010. Environ. Health
Kenyon, E.M., Del Razo, L.M., Hughes, M.F., et al., 2005. Toxicol. Sci. Perspect. 118, 533–538.
85 (1), 468–475. Lindgren, A., Vahter, M., Dencker, L., 1982. Acta Pharmacol. Toxicol.
Khalil, S., Luciano, J., Chen, W., et al., 2006. J. Cell Physiol. 207 (2), 51, 253–265.
562–569. Lindgren, A., Danielsson, B.R.G., Demcker, L., et al., 1984. Acta Phar-
Kilburn, K.H., 1997. In: Abernathy, C.O., Calderon, R.L., Chappell, macol. Toxicol. 54, 311–320.
W.R. (Eds.), Arsenic: Exposure and Health Effects. Chapman & Lisabeth, L.D., Ahn, H.J., Chen, J.J., et al., 2010. Stroke 41, 2499–2504.
Hall, London, pp. 159–175. Liu, J., Kadiiska, M.B., Liu, Y., et al., 2001. Toxicol. Sci. 61, 314–320.
Kim, Y., Lee, B.K., 2011. Sci. Total Environ. 409, 4054–4062. Liu, L., Trimarchi, J.R., Navarro, P., et al., 2003. J. Biol. Chem. 278 (34),
Kipling, M.D., Fothergill, R., 1964. Br. J. Ind. Med. 21, 74–77. 31998–32004.
Kobayashi, Y., Cui, X., Hirano, S., 2005. Toxicology 211 (1-2), 115–123. Liu, S.X., Davidson, M.M., Tang, X., et al., 2005. Cancer Res. 65 (8),
Kodama, Y., Ishinishi, N., Kunitake, E., et al., 1976. In: Nordberg, G.F. 3236–3242.
(Ed.), Effects and Dose-Response Relationships of Toxic Metals. Liu, Y., Liang, Y., Zheng, T., et al., 2011. J. Neurooncol. 104 (2), 449–458.
Elsevier, Amsterdam, pp. 464–470. Loffredo, C.A., Aposhian, H.V., Cebrian, M.E., et al., 2003. Environ.
Kojima, C., Ramirez, D.C., Tokar, E.J., et al., 2009. J. Natl. Cancer Inst. Res. 92, 85–91.
101 (24), 1670–1681. Lofroth, G., Ames, B.N., 1978. Mutat. Res. 53, 65–66.
Komissarova, E.V., Rossman, T.G., 2010. Toxicol. Appl. Pharmacol. Lubin, J.H., Pottern, L.M., Stone, B.J., et al., 2000. Am. J. Epidemiol.
243, 399–404. 151, 554–565.
Kraus, T., Quidenus, G., Schaller, K.H., 2000. Arch. Environ. Contam. Lüchtrath, H., 1983. J. Cancer Res. Clin. Oncol. 105, 173–182.
Toxicol. 38, 116–121. Lugo, G., Cassady, G., Palmisano, P., 1969. Am. J. Dis. Child. 117,
Kreiss, K., Zack, M.M., Landrigan, P.J., et al., 1983. Arch. Environ. 328–330.
Health 38 (2), 116–121. Luna, A.L., Acosta-Saavedra, L.C., Lopez-Carrillo, L., et al., 2010.
Kreppel, H., Reichl, F.X., Kleine, A., et al., 1995. Fundam. Appl. Toxi- Toxicol. Appl. Pharmacol. 245, 244–251.
col. 26. 293–245. Lunde, G., 1973. Acta Chem. Scand 27, 1586–1594.
Kubota, R., Kunito, T., Agusa, T., et al., 2006. Environ. Monit. 8 (2), Lunde, G., 1975. J. Sci. Food Agric 26, 1247–1255.
293–299. Lundgren, K.D., 1954. Nord. Hyg. Tidskr 3, 66–82.
Kuo, T., 1968. Rep. Inst. Pathol. Natl Taiwan Univ. 20, 7–13. Luten, J.B., Riekwal-Booy, G., Rauchbaar, A., 1982. Environ. Health
Kuo, T.T., Hu, S., Lo, S.K., et al., 1997. Hum. Pathol. 28 (7), 786–790. Perspect. 45, 165–167.
Kurosawa, S., Yasuda, K., Taguchi, M., et al., 1980. Agric. Biol. Chem. Lynn, S., Gurr, J.R., Lai, H.T., et al., 2000. Circ. Res. 86, 514–519.
44, 1993–1994. Madden, E.F., Akkerman, M., Fowler, B.A., 2002. J. Biochem. Mol.
Kurttio, P., Pukkala, E., Kahelin, H., et al., 1999. Environ. Health Per- Toxicol. 16 (1), 24–32.
spect. 107, 705–710. Madden, E.F., Fowler, B.A., 2000. Drug Chem. Toxicol. 23, 1–12.
Lagerkvist, B., Linderhold, H., Nordberg, G.F., 1986. Environ. Res. Maden, N., Singh, A., Smith, L.S., et al., 2011. J. Community Health
39, 465–474. 36, 76–82.
28 Arsenic 621

Mahaffey, K.R., Fowler, B.A., 1977. Environ. Health Perspect. 19, Morrissey, R.E., Fowler, B.A., Harris, M.W., et al., 1990. Fundam.
165–171. Appl. Toxicol. 15 (2), 350–356.
Mahaffey, K.R., Capar, S.G., Gladen, B.C., et al., 1981. J. Lab. Clin. Morse, K.M., Setterlind, A.N., 1950. Arch. Ind. Hyg. Occup. Med. 2,
Med. 98, 463–481. 148–169.
Mahieu, P., Buchet, J.P., Roels, H.A., et al., 1981. Clin. Toxicol. 18 (9), Morton, W.E., Dunnette, D.A., 1994. Advances in Environmental
1067–1975. Science and Technology. In: Nriagu, J.O. (Ed.). John Wiley, New
Maiti, S., Chatterjee, A.K., 2001. Arch. Toxicol. 75 (9), 531–537. York. pp. 27, 17-34.
Mandal, B.K., Suzuki, K.T., 2002. Talanta 58, 201–235. Mosser, D.D., Caron, A.W., Bourget, L., et al., 2000. Mol. Cell Biol. 20
Mandal, B.K., Ogra, Y., Suzuki, K.T., 2001. Chem. Res. Toxicol. 14, (19), 7146–7159.
371–378. Mostafa, M.G., McDonald, J.C., Cherry, N.M., 2008. Occup. Environ.
Mandal, B.K., Ogra, Y., Suzki, K.T., 2003. Toxicol. Appl. Pharmacol. Med. 65, 765–768.
189, 73–83. Muehrcke, R.C., Pirani, C.L., 1968. Ann. Intern. Med. 68, 853–866.
Mandal, B.K., Ogra, Y., Anzai, K., et al., 2004. Toxicol. Appl. Pharma- Mukherjee, S.C., Saha, K.C., Pati, S., et al., 2005. Clin. Toxicol. 43 (7),
col. 198, 307–318. 835–848.
Mao, G., Guo, X., Kang, R., et al., 2010. Chemosphere 80, 978–981. Muñoz, E., Palmero, S., 2005. Talanta 65, 613–620.
Mappes, R., 1977. Int. Arch. Occup. Environ. Health 40, 267–272. Munro, I.C., 1976. Clin. Toxicol. 9, 647–663.
Marafante, E., Vahter, M., 1987. Environ. Res. 42, 72–82. Muscarella, D.E., Bloom, S.E., 2002. Toxicol. Sci. 68 (1), 82–92.
Marafante, E., Rade, J., Sabbioni, E., 1981. Clin. Toxicol. 18 (11), Myers, D.J., Osteryoung, J., 1973. Anal. Chem. 45, 267–271.
1335–1341. Nafees, A.A., Kazi, A., Fatmi, Z., et al., 2011. Environ. Geochem.
Marafante, E., Bertolero, F., Edel, J., et al., 1982. Sci. Total Environ. Health 33, 203–216.
24, 27–39. Nakadaira, H., Endoh, K., Katagiri, M., et al., 2002. J. Occup. Envi-
Marafante, E., Vahter, M., Dencker, L., 1984. Sci. Total Environ. 34, ron. Med. 44. 291–199.
223–240. Nakazato, T., Tao, H., Taniguchi, T., et al., 2002. Talanta 58, 121–132.
Marafante, E., Vahter, M., Norin, H., et al., 1987. J. Appl. Toxicol. 7, Namgung, U., Xia, Z., 2000. J. Neurosci. 20 (17), 6442–6451.
111–117. Nemec, M.D., Holson, J.F., Farr, C.H., et al., 1998. Reprod. Toxicol.
Marawi, I., Wang, J., Caruso, J.A., 1994. Analytica. Chimica. Acta 291, 12, 647–658.
127–136. Nemeti, B., Gregus, Z., 2005. Toxicol. Sci. 85 (2), 847–858.
Martindale, W., 1977. In: Wade, A., Reynolds, J.E.F. (Eds.), The Extra Ng, J.C., Qi, C.M., Moore, M.R., 2002. Cell. Mol. Biol. 48, 111–123.
Pharmacopoeia, twentyseventh ed. The Pharmaceutical Press, Ng, J.C., Wang, J.P., Zheng, B., et al., 2005. Toxicol. Appl. Pharmacol.
London, pp. 1721–1723. 206, 176–184.
Maruyama, Y., Komiya, K., Manri, T., 1970. Radioisotopes 19, 44–46. NIOSH (National Institute for Occupational Safety and Health). (2006a).
Mass, M.J., Tennant, A., Roop, B.C., et al., 2001. Chem. Res. Toxicol. www.cdc.gov/niosh/idlh/7440382.html (accessed 19.02.14).
14 (4), 355–361. NIOSH., 2006b. International Chemical Safety Cards. (accessed
Matsui, M., Nishigori, C., Toyokuni, S., et al., 1999. J. Invest. Derma- 19.02.14) www.cdc.gov/niosh/ipcsneng/neng0222.html.
tol. 113 (1), 26–31. Nishioka, H., 1975. Mutat. Res. 31, 185–189.
McBride, B.C., Merilees, H., Cullen, W.R., et al., 1978. Organometals Nixon, D.E., Neubauer, K.R., Eckdahl, S.J., et al., 2004. Spectrochi-
and Organometalloids. In: Brinckman, F.E., Bellama, J.M. (Eds.). mica Acta Part B 59, 1377–1387.
ACS Symposium Series 82. American Chemical Society, Wash- Nordenson, I., Sweins, A., Beckman, L., 1981. Scand. J. Work Envi-
ington, DC, pp. 94–115. ron. Health 7, 277–281.
McCabe, L.J., Symons, J.M., Lee, R.D., et al., 1970. J. Am. Water Works Nordstrom, S., Beckman, L., Nordenson, I., 1979a. Hereditas 90,
Assoc. 62, 670–687. 291–296.
McFall, T.L., Richards, S., Mathews, G., 1998. J. Spinal Cord Med. Nordstrom, S., Beckman, L., Nordenson, I., 1979b. Hereditas 90,
21, 142–147. 297–302.
Mealey, J., Brownell, G.L., Sweet, W.H., 1959. Am. Med. Assoc. Arch. Norin, H., Christakopoulos, A., 1982. Chemosphere 11, 287–298.
Neurol. Psychiat. 81, 310–320. Norin, H., Vahter, M., 1981. Scand. J. Work Environ. Health 7, 38–44.
Menzel, D.B., Ross, M., Oddo, S.V., et al., 1994. Environ. Geochem. Novakova, S., 1969. Gig. Sanit 34, 81–85.
Health 16, 209–218. NRC (National Academy of Sciences National Research Council),
Merwin, I., Pruyne, P.T., Ebel, J.G., et al., 1994. Chemosphere 29, 1999. Arsenic in Drinking Water. National Research Council. 310
1361–1367. pp. National Academy Press, Washington, DC.
Milton, A.H., Smith, W., Rahman, B., et al., 2005. Epidemiology 16 NRC., 2001. Arsenic in Drinking Water 2001 Update. National Research
(1), 82–86. Council. National Academy Press, Washington, DC, 244 pp.
Mitchell, R.A., Chang, B.F., Huang, C.H., et al., 1971. Biochemistry Nriagu, U.O., Lin, T.-S., 1995. Sci. Total Environ. 172, 223–228.
10 (11), 2049–2054. NTP (National Toxicology Program), 1980. National Toxicology Pro-
Mizoi, M., Takabayashi, F., Nakano, M., et al., 2005. Toxicol. Lett. 158 gram—technical report series no. 345. U.S. Department of Health
(2), 87–94. and Human Services, Public Health Service, National Institutes
Mizuta, N., Mizuta, M., Ito, F., et al., 1956. Bull. Yamaguchi Med. Sch. of Health, Research Triangle Park, NC. NTP-TR-345; NIH Pub
4, 131–150. no. 89-2800, pp. 1–198.
Mohri, T., Hisanaga, A., Ishinishi, N., 1990. Fd. Chem. Toxic. 28, Nunes, J.A., Batista, B.L., Rodrigues, J.L., et al., 2010. J. Toxicol. Envi-
521–529. ron. Health 73, 878–887.
Molin, L., Wester, P.O., 1976. Scand. J. Clin. Lab. Invest. 36, 679–682. Nygren, O., Nilsson, C.-A., Lindahl, R., 1992. Ann. Occup. Hyg. 36,
Moreda-Piñeiro, J., Alonso-Rodríguez, E., Moreda-Piñeiro, A., et al., 509–517.
2010. Anal. Chim. Acta 679, 63–73. O’Bryant, S.E., Edwards, M., Menon, C.V., et al., 2011. Int. J. Environ.
Moreda-Piñeiro, A., Moreda-Piñeiro, J., Herbello-Hermelo, P., et al., Res. Public Health 8, 861–874.
2011. J. Chromatogr. A. 1218, 6970–6980. Offergelt, J.A., Roels, H., Buchet, J.P., et al., 1992. Br. J. Ind. Med. 49,
Moreira, J.C., 1996. Sci. Total Environ. 188, S61–S71. 387–393.
622 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

Ott, M.G., Holder, B.B., Gordon, H.L., 1974. Arch. Environ. Health Reay, P.P., Asher, C.J., 1977. Anal. Biochem. 78, 557–560.
29, 250–255. Reichard, J.F., Puga, A., 2010. Epigenomics 2, 87–104. Reed, D. J.
Pacyna, J.M., 1987. Lead, Mercury, Cadmium and Arsenic in the En- (2001). Curr. Protocols. Toxicol. 6, 601-608.
vironment. In: Hutchinson, T.C., Meema, K.M. (Eds.). John Wiley Robertson, F.N., 1989. Environ. Geochem. Health 11, 171–185.
and Sons, New York, NY, pp. 297–298. Rose, M., Knaggs, M., Owen, L., et al., 2001. J. Anal. At. Spectrom.
Paiva, L., Hernández, A., Martínez, V., etal, 2010. Environ. Res. 110 16, 1101–1106.
(5), 463–468. Rosenthal, G.J., Fort, M.M., Germolec, D.R., et al., 1989. Toxicology
Papaconstantinou, A.D., Brown, K.M., Noren, B.T., et al., 2003. Birth 1, 113–127.
Defects Res. B. Dev. Reprod. Toxicol. 68 (6), 456–464. Rossi, M.R., Somji, S., Garrett, S.H., et al., 2002. Environ. Health Per-
Parvez, F., Chen, Y., Brandt-Rauf, P.W., et al., 2010. Thorax 65, 528–533. spect. 110, 1225–1232.
Parvez, F., Wasserman, G.A., Factor-Litvak, P., et al., 2011. Environ. Rossman, T.G., Meyn, M.S., Troll, W., 1977. Environ. Health Perspect.
Health Perspect. 119, 1665–1670. 19, 229–233.
Patty, F.A., 1962. Industrial Hygiene and Toxicology, second ed. In- Rossman, T.G., Stone, D., Molina, M., et al., 1980. Environ. Mutagen
terscience Publishers, John Wiley and Sons, New York. 2, 371–379.
Paul, D.S., Walton, F.S., Saunders, R.J., et al., 2011. Environ. Health Rossner Jr., P., Binkova, B., Sram, R.J., 2003. Mutat. Res. 542 (1-2),
Perspect. 119, 1104–1109. 105–116.
Pellizzari, E.D., Clayton, C.A., 2006. Environ. Health Perspect. 114, Rowland, I.R., Davies, M.J., 1982. J. Appl. Toxicol. 2, 294–299.
220–227. Sagner, G., 1973. Schriftenr. Ver. Wasser Boden Lufthyg 40, 189–208.
Penrose, W.R., Conacher, H.B.S., Black, R., et al., 1977. Environ. Sampayo-Reyes, A., Hernández, A., El-Yamani, N., et al., 2010. Toxi-
Health Perspect. 19, 53–59. col Sci. Sep. 117 (1), 63–71.
Pershagen, G., Wall, S., Taube, A., et al., 1981. Scand. J. Work Environ. Sandell, E.B., 1959. Colorimetric Determination of Trace Metals,
Health 7, 302–309. third ed. Interscience Publishers, New York, 1032 pp.
Pershagen, G., Nordberg, G., Bjorklund, N.E., 1984. Environ. Res. 34, Sanders, J.G., Riedel, G.F., Osman, R.W., 1994. In: Nriagu, J.O. (Ed.),
227–241. Arsenic in the Environment. Part I. John Wiley and Sons, New
Peters, H.A., Crost, W.A., Woolson, E.A., et al., 1984. JAMA 251, York, pp. 289–308.
2393–2396. Satterlee, H.S., 1956. N. Engl. J. Med. 254, 1149–1154.
Peters, R.A., 1955. Bull. Johns Hopkins Hosp. 97, 1–20. Schiller, C.M., Walden, R., Fowler, B.A., 1981. Biochem. Pharmacol.
Peterson, R.G., Rumack, B.H., 1977. J. Pediatr. 9, 661–666. 30, 168–170.
Petres, J., Baron, D., Hagedorn, M., 1977. Environ. Health Perspect. Schoof, R.A., Yost, L.J., Crecelius, K., et al., 1998. Hum. Ecol. Risk
19, 223–227. Assess 4, 117–135.
Pi, J., He, Y., Bortner, C., et al., 2005. Int. J. Cancer 116 (1), 20–26. Schoof, R.A., Eickhoff, J., Yost, L.J., et al., 1999a. Arsenic Exposure
Pinto, S.S., McGill, C.M., 1953. Ind. Med. Surg. 22, 281–287. and Health Effects. In: Chappell, W.R., Abernathy, C.O., Calde-
Pinto, S.S., Varner, M.O., Nelson, K.W., et al., 1976. J. Occup. Med. raon, R.L. (Eds.). Elsevier Science, Amsterdam, pp. 81–88.
18, 677–680. Schoof, R.A., Yost, L.J., Eickhoff, J., et al., 1999b. Food Chem. Toxicol.
Pinto, S.S., Enterline, P.E., Henderson, V., et al., 1977. Environ. Health 37, 839–846.
Perspect. 19, 127–130. Schrenk, H.H., Schreibeis Jr., L., 1958. Am. Ind. Hyg. Assoc. J. 19,
Pinto, S.S., Henderson, V., Enterline, P.E., 1978. Arch. Environ. Health 225–228.
33, 325–331. See, L. C. (2000). Doctoral Thesis. National Taiwan University, Tai-
Polissar, L., Lowry-Coble, K., Kalman, D.A., et al., 1990. Environ. pei, Taiwan.
Res. 53, 29–47. Sen, B., Wang, A., Hester, S.D., et al., 2005. Toxicology 215 (3), 214–226.
Pomroy, C., Charbonneau, S.M., McCullough, R.S., et al., 1980. Toxi- Sheehy, J.W., Jones, J.H., 1993. Am. Ind. Hyg. Assoc. J. 54, 61–69.
col. Appl. Pharmacol. 53 (3), 550–556. Shen, J., Wanibuchi, H., Salim, E.I., et al., 2003. Toxicol. Appl. Phar-
Prier, R.F., Nees, P.O., Derse, P.H., 1963. Toxicol. Appl. Pharmacol. macol. 193, 335–345.
5, 526–542. Shen, J., Wanibuchi, H., Waalkes, M.P., et al., 2006. Toxicol. Appl.
Pucer, A., Castino, R., Mirkovic, B., et al., 2010. J. Biol. Chem. 391, Pharmacol. 210 (3), 171–180.
519–531. Shen, Z.X., Chen, G.Q., Ni, J.H., et al., 1997. Blood 89, 3354–3360.
Raab, A., Feldmann, J., 2005. Anal. Bioanal. Chem. 381, 332–338. Sheppard, B.S., Caruso, J.A., Heitkemper, D.T., et al., 1992. Analyst
Rael, L.T., Ayala-Fierro, F., Carter, D.E., 2000. Toxicol. Sci. 55 (2), 117, 971–975.
468–477. Shimizu, M., Hochadel, J.F., Fulmer, B.A., et al., 1998. Toxicol. Sci. 45
Rahman, A., Vahter, M., Ekström, E.C., et al., 2011. Environ. Health (2), 204–211.
Perspect. 119, 719–724. Shinjo, K., Takeshita, A., Sahara, N., et al., 2005. Intern. Med. 44 (8),
Rahman, M., Axelson, O., 1995. Occup. Environ. Med. 52, 773–774. 818–824.
Rahman, M., Wingren, G., Axelson, O., 1996. Scand. J. Work Environ. Siripitayakunkit, U., Visudhiphan, P., Pradipasen, M., et al., 1999. In:
Health 22, 146–149. Chappell, W.R., Abernathy, C.O., Calderon, R.L. (Eds.), Arsenic
Rahman, M., Tondel, M., Ahmad, S.A., et al., 1998. Am. J. Epidemiol. Exposure and Health Effects. Elsevier Science, Amsterdam, pp.
148, 198–203. 141–149.
Rahman, M., Tondel, M., Ahmad, S.A., et al., 1999b. Hypertension Smith, A.H., Marshall, G., Yuan, Y., et al., 2006. Environ. Health Per-
33, 74–78. spect. 114, 1293–1296.
Rahman, M., Tondel, M., Chowdhury, I.A., et al., 1999a. Occup. En- Smith, A.H., Steinmaus, C.M., 2009. Annu. Rev. Public Health 30,
viron. Med. 56, 277–281. 107–122.
Ramanathan, K., Anusuyadevi, M., Shila, S., et al., 2005. Toxicol. Lett. Smith, C.J., Livingston, S.D., Doolittle, D.J., 1997. Food Chem. Toxi-
156 (2), 297–306. col. 35, 1107–1130.
Ramos, O., Carrizales, L., Yanez, L., et al., 1995. Environ. Health Per- Smith, D.M., Patel, S., Raffoul, F., et al., 2010. Cell Death Differ 17,
spect. 103 (Suppl.), 85–88. 1867–1881.
Ratnaike, R.N., 2003. Postgrad. Med. J. 79, 391–396. Smith, H., 1964. J. Forensic Sci. Soc. 4, 192–199.
28 Arsenic 623

Smith, R.S., Alexander, R.B., Wolman, M.G., 1987. Science 235, Tsai, S.-Y., Chou, H.-Y., The, H.-W., et al., 2003. Neurotoxicology 24,
1607–1615. 747–753.
Smith, T.J., Crecelius, E.A., Reading, J.C., 1977. Environ. Health Per- Tseng, C.-H., 2004. Toxicol. Appl. Pharmacol. 197 (2), 67–83.
spect. 19, 89–93. Tseng, C.-H., Chong, C.-K., Chen, C.-J., 1995. Int. J. Microcirc. 15,
Somogyi, A., Beck, H., 1993. Environ. Health Perspect. 101, 45–52. 21–27.
Song, X., Geng, Z., Li, X., Zhao, Q., et al., 2011. Biochimie 93 (2), Tseng, C.-H., Chong, C.-K., Chen, C.-J., 1997. Angiology 48, 321–335.
369–375. Tseng, C.-H., Tai, T.-Y., Chong, C.-K., et al., 2000. Environ. Health
Southwick, J.W., Western, A.E., Beck, M.M., et al., 1981. U.S. Environ- Perspect. 108, 847–885.
mental Protection Agency, Health Effects Research Laboratory. Tseng, C.-H., Tseng, C.-P., Chiou, H.-Y., 2002. Toxicol. Lett. 133 (1),
EPA600181064. PB82108374. 69–76.
Southwick, J.W., Western, A.E., Beck, M.M., 1983. In: Lederer, W., Tseng, C.H., Huang, Y.K., Huang, Y.L., et al., 2005. Toxicol. Appl.
Fensternheim, R. (Eds.), Arsenic: Industrial, Biomedical, Envi- Pharmacol. 206, 299–308.
ronmental Perspectives. Van Nostrand Reinhold, New York, Tseng, H.P., Wang, Y.H., Wu, M.M., et al., 2006. J. Health Popul. Nutr.
pp. 210–225. 24, 182–189.
Stanhill, A., Levin, V., Hendel, A., et al., 2006. Oncogene 25 (10), Tseng, M.-P. (1999). Master’s Thesis. National Taiwan University,
1485–1495. Taipei, Taiwan.
Steinmaus, C., Carrigan, K., Kalman, D., et al., 2005a. Environ. Tseng, W.-P., 1977. Environ. Health Perspect. 19, 109–119.
Health Perspect. 113 (9), 1153–1159. Tseng, W.-P., Chen, W.-Y., Sung, J.-L., et al., 1961. Memoirs College
Steinmaus, C., Yuan, Y., Kalman, D., et al., 2005b. Cancer Epidemiol. Med. Natl. Taiwan Univ. 7, 1–17.
Biomarkers Prev. 14 (4), 919–924. Tseng, W.P., Chu, H.M., How, S.W., et al., 1968. J. Natl. Cancer Inst.
Steinmaus, C., Bates, M.N., Yuan, Y., et al., 2006. J. Occup. Environ. 40, 453–463.
Med. 48 (5), 478–488. Tsou, T.-C., Tsai, F.-Y., Hsieh, Y.-W., et al., 2005. Toxicol. Appl. Phar-
Steinmaus, C., Yuan, Y., Kalman, D., et al., 2010. Toxicol. Appl. Phar- macol. 208 (3), 277–284.
macol. 247, 138–145. Tsuda, T., Babazono, A., Yamamoto, E., et al., 1995. Am. J. Epidemiol.
Stevens, J.R., Hall, L.L., Farmer, J.D., et al., 1977. Environ. Health Per- 141, 198–209.
spect. 19, 151–157. Uchino, T., Roychowdhury, T., Ando, M., et al., 2006. Food Chem.
Stump, D.G., Holson, J.F., Fleeman, T.L., et al., 1999. Teratology 60, Toxicol. 44, 455–461.
283–291. Vahter, M., 1981. Environ. Res. 25, 286–293.
Styblo, M., Thomas, D.J., 1995. Biochem. Pharma. 49 (7), 971–977. Vahter, M., 1994. In: Chappell, W.R., Abernathy, C.O., Calderon, C.R.
Styblo, M., Thomas, D.J., 2001. Toxicol. Appl. Pharmacol. 172, 52–61. (Eds.), Arsenic Exposure and Health. Science and Technology
Sullivan, R.J. (1969) Air pollution aspect of arsenic and its com- Letters. Northwood, pp. 171–180.
pounds. Nat. Air Polllution Control Admin. Consumer Protec- Vahter, M., Envall, J., 1983. Environ. Res. 32, 14–24.
tion and Env. Health Services. U.S. DHEW. Contract PH-22-68-25 Vahter, M., Marafante, E., 1983. Chem. Biol. Interact. 47, 29–44.
to Litton Systems, Bethesda, MD. Vahter, M., Marafante, E., 1985. Arch. Toxicol. 57, 119–124.
Sumino, K., Hayakawa, K., Shibata, T., et al., 1975. Arch. Environ. Vahter, M., Norin, H., 1980. Environ. Res. 21, 446–457.
Health 30, 487–494. Vahter, M., Marafante, E., Lindgren, A., et al., 1982. Arch. Toxicol.
Sun, Y., Tokar, E.J., Waalkes, M.P., 2012. Toxicol. Sci. 125, 20–29. 51, 65–77.
Suzuki, K.T., Tomita, T., Ogra, Y., et al., 2001. Chem. Res. Toxicol. 14 Vahter, M., Marafante, E., Dencker, L., 1983. Sci. Total Environ. 30,
(12), 1604–1611. 197–211.
Suzuki, K.T., Mandal, B.K., Ogra, Y., 2002. Talanta 58, 111–119. Vahter, M., Marafante, E., Dencker, L., 1984. Arch. Environ. Contam.
Tabacova, S., Baird, D. D., Balabaeva, L., et al. (1994). 15(8), 873-881. Toxicol. 13, 259–264.
Takahashi, K., Yamauchi, H., Yamato, N., et al., 1988. Appl. Or- Vahter, M., Friberg, L., Rahnster, B., et al., 1986. Int. Arch. Occup.
ganomet. Chem. 2, 309–314. Environ. Health 57, 79–91.
Tam, G.K., Charbonneau, S.M., Bryce, F., et al., 1978. Anal. Biochem. Vahter, M., Concha, G., Nemell, B., et al., 1995. Eur. J. Pharmacol.
86, 505–511. 293, 455–462.
Tam, G.K., Charbonneau, S.M., Bryce, F., et al., 1979. Toxicol. Appl. Valentine, J.L., Reisbord, L.S., Kang, H.K., et al., 1985. Proceedings of
Pharmacol. 50, 319–322. the Fifth International Symposium on Trace Elements in Man and
Tam, G.K., Charbonneau, S.M., Bryce, F., et al., 1982. Bull. Environ. Animals. Commonwealth Agricultural Bureaux, Slough, UK,
Contam. Toxicol. 28, 669–673. pp. 289–294.
Tanner, S.D., Baranov, V.I., Volkopf, U., 2000. J. Anal. At. Spectrom Valenzuela, O.L., Borja-Aburto, V.H., Garcia-Vargas, G.G., et al.,
15, 1261–1269. 2005. Environ. Health Perspect. 113 (3), 250–254.
Taylor, A., Branch, S., Halls, T.J., et al., 2004. J. Anal. At. Spectrom Vallee, B.L., Uliner, D.D., Wacker, W.E.C., 1960. AMA Arch. Ind.
19, 505–556. Health 21, 132–151.
Thomas, D.C., Whittemore, A.S., 1988. Am. J. Ind. Med. 13, 131–147. Vantroyen, B., Heilier, J.F., Meulemans, A., et al., 2004. J. Toxicol.
Tian, X., Ma, X., Qiao, D., et al., 2005. Mol. Cell Biochem. 277 (1-2), Clin. Toxicol. 42, 889–895.
33–42. Vasak, V., Sedivec, V., 1952. Chem. Listy 46, 341–344.
Tintinalli, J.E., Ruiz, E., Krone, R.L. (Eds.), 1996. Emergency Medi- Verdon, C.P., Caldwell, K.L., Fresquez, M.R., Jones, R.L., 2009. Anal.
cine. A Comprehensive Study, fourth ed. American College of Bioanal. Chem. 393, 939–947. Vondracek, V. (1963). Cesk. Hyg. 8,
Emergency Physicians. The McGraw-Hill Companies, Inc., New 333-339.
York, NY. von Ehrenstein, O.S., Guha Mazumder, D.N., Yuan, Y., et al., 2005.
Tondel, M., Rahman, M., Magnuson, A., et al., 1999. Environ. Health Am. J. Epidemiol. 162, 533–541.
Perspect. 107, 727–729. Waalkes, M.P., Liu, J., Ward, J.M., et al., 2004. Toxicology 198 (1-2),
Townsend, A.T., 1999. Fresenius J. Analytical Chem. 364, 521–526. 31–38.
Tsai, S.-M., Wang, T.-N., Ko, Y.-C., 1999. Arch. Environ. Health 54, Walsh, L.M., Sumner, M.E., Keeney, D.R., 1977. Environ. Health Per-
186–193. spect. 19, 67–71.
624 Bruce A. Fowler, C.-H. Selene J. Chou, Robert L. Jones, Dexter W. Sullivan Jr, and C.-J. Chen

Walter, R.L., Willis, R.D., Gutknecht, W.F., et al., 1974. Anal. Chem. Wyatt, C.J., Fimbres, C., Romo, L., et al., 1998a. Environ. Res. 76, 114–119.
46, 843–855. Wyatt, C.J., Lopez Quiroga, V., Acosta, R.T., et al., 1998b. Environ
Walton, F.S., Waters, S.B., Jolley, S.L., et al., 2003. Chem. Res. Toxicol. Res. 78, 19–24.
16, 261–265. Yaglom, J.A., Ekhterae, D., Gabai, V.L., et al., 2003. J. Biol. Chem. 278
Wang, C.-H., Jeng, J.-S., Yip, P.-K., et al., 2002. Circulation 105, (50), 50483–950486.
1804–1809. Yamashita, N., Doi, M., Nishio, M., et al., 1972. Nihon Eiseigaku
Wang, C.-H., Hsiao, C.-K., Chen, C.-L., et al., 2007. Toxicol. Appl. Zasshi 27, 364–399 (EPA translation No. TR108-74).
Pharmacol. 222, 315–326. Yamauchi, H., Yamamura, Y., 1979a. Ind. Health 17, 79–83.
Wang, C.-H., Chen, C.-L., Hsiao, C.-K., et al., 2009. Toxicol. Appl. Yamauchi, H., Yamamura, Y., 1979b. Jpn. J. Ind. Health 21, 47–54.
Pharmacol. 239, 320–324. Yamauchi, H., Yamamura, Y., 1984. Toxicol. Appl. Pharmacol. 74 (1),
Wang, C.-H., Chen, C.-L., Hsiao, C.-K., et al., 2010. Cardiovasc. Toxi- 134–140.
col. 10, 17–26. Yamauchi, H., Yamamura, Y., 1985. Toxicology 34, 113–121.
Wang, J.-D., Yang, H.-L., Wu, H.-Y., 1985. Blackfoot Dis. Res. Report Yamauchi, H., Takahashi, K., Yamamura, Y., 1986. Toxicoogy 240,
23, 30–46. 237–246.
Wang, L.F., Huang, J.Z., 1994. Advance in Environmental Science Yamauchi, H., Yamato, N., Yamamura, Y., 1988. Bull. Environ. Con-
and Technology. In: Nriagu, J.O. (Ed.). John Wiley, New York, tam. Toxicol. 40, 280–286.
pp. 159–172. Yamauchi, H., Takahashi, K., Yamamura, Y., 1989. Toxicol. Environ.
Wang, S.-L., Chiou, J.-M., Chen, C.-J., et al., 2003. Environ. Health Chem. 22, 69–76.
Perspect. 111, 155–159. Yamauchi, H., Kaise, T., Takahashi, K., et al., 1990. Fundam. Appl.
Wang, Y.H., Yeh, S.D., Shen, K.H., et al., 2009. Toxicol. Appl. Pharma- Toxicol. 14 (2), 399–407.
col. 241, 111–118. Yamauchi, H., Takahashi, K., Yamamura, Y., et al., 1992. Toxicol.
Wang, Z.Y., 2001. Cancer Chemother. Pharmacol. 48, S72–S76. Appl. Pharmacol. 116, 240–247.
Wasserman, G.A., Parvez, F., Ahsan, H., et al., 2004. Environ. Health Yamauchi, H., Yoshida, T., Aikawa, H., et al., 1998. The Toxicologist
Perspect. 112, 1329–1333. 42, 321.
Waters, S.B., Devesa, V., Fricke, M.W., et al., 2004. Chem. Res. Toxicol. Yang, L., Wang, W., Hou, S., et al., 2002. Environ. Geochem. Health
17 (12), 1621–1629. 24, 337–348.
Watrous, R.M., McCaughey, M.B., 1945. Ind. Med. 14, 639–646. Yang, T.Y., Hsu, L.I., Chiu, A.W., et al., 2014. Environ Res. 128, 57–63.
Webb, J.L., 1966. Enzyme and Metabolic Inhibitors. Academic Press, Yeh, S., How, S.-W., 1963. Rep. Inst. Pathol. Natl. Taiwan Univ. 14,
New York, pp. 595–793. 25–73.
Wedepohl, K.H., 1991. Metals and Their Compounds in the Environ- Yokohira, M., Arnold, L.L., Pennington, K.L., et al., 2011. Toxicol. Sci.
ment. In: Merian, E. (Ed.). VCH, New York, NY, pp. 3–17. 121, 257–266.
Wei, M., Wanibuchi, H., Yamamoto, S., et al., 1999. Carcinogenesis Yoo, D.R., Chong, S.A., Nam, M.J., 2009. Cancer Genomics Pro-
20, 1873–1876. teomics 6, 269–274.
Wei, M., Wanibuchi, H., Morimura, K., et al., 2002. Carcinogenesis Yorifuji, T., Tsuda, T., Doi, H., et al., 2011. Environ. Health Prev. Med.
23, 1387–1397. 16, 164–170.
Welch, A.H., Lico, M.S., Hughes, J.L., 1988. Groundwater 26, 333–347. Yoshida, K., Kuroda, K., Inoue, Y., et al., 2001. Appl. Organomet.
Welch, K., Higgins, I., Oh, M., et al., 1982. Arch. Environ. Health 37, Chem. 15, 539–547.
325–335. Yu, H.S., Liao, W.T., Chai, C.Y., 2006. J. Biomed. Sci. 13 (5), 657–666.
Wen, W., Lieu, T., Chang, H., et al., 1981. Hum. Genet. 59, 201–203. Yu, R.-C., Hsu, K.-H., Chen, C.-J., et al., 2000. Cancer Epidemiol. Bio-
Wendt, P.H., Van Dolah, R.F., Bobo, M.Y., et al., 1996. Arch. Environ. markers Prev. 9, 1259–1262.
Contam. Toxicol. 31, 24–37. Yuan, Y., Marshall, G., Ferreccio, C., et al., 2010. Epidemiology 21,
Wester, R.C., Maibach, H.I., Sedik, L., et al., 1993. Fundam. Appl. 103–108.
Toxicol. 20, 336–340. Yukawa, M., Suzaki-Yasumoto, M., Amaho, K., et al., 1980. Arch. En-
WHO (World Health Organization), 1981. Environmental Health viron. Health 35, 36–44.
Criteria, Arsenic. World Health Organization, Geneva, pp. 1–174. Zakharyan, R.A., Wildfang, E., Aposhian, H.V., 1996. Toxicol. Appl.
WHO, 2001. Environmental Health Criteria, 224, Arsenic and Arse- Pharmacol. 140, 77–84.
nic Compounds, second ed. World Health Organization, Geneva, Zakharyan, R.A., Tsaprailis, G., Chowdhury, U.K., et al., 2005. Chem.
pp. 385–392. Res. Toxicol. 18 (8), 1287–1295.
Wickstrom, G., 1972. Work Environ. Health 9, 2–8. Zaldívar, R., 1974. Beitr. Pathol. 151, 384–400.
Wijeweera, J.B., Gandolfi, A.J., Parrish, A., et al., 2001. Toxicol. Sci. Zaldívar, R., 1980. Zentralbl. Bakteriol. Parasitenk. Infektionskr.
61 (2), 283–294. Hyg. Abt. I, Orig. Reihe B 170, 44–56.
Winski, S.L., Barber, D.S., Rael, L.T., et al., 1997. Fundam. Appl. Toxi- Zhang, T.C., Schmitt, M.T., Mumford, J.L., 2003. Carcinogenesis 24
col. 38 (2), 123–128. (11), 1811–1817.
Wood, T.C., Salavaggione, O.E., Mukherjee, B., et al., 2006. J. Biol. Zhang, W., Wang, L., Fan, Q., 2011. Oncol. Rep. 26, 621–628.
Chem. 281 (11), 7364–7373. Zhang, Y., Cao, E.H., Liang, X.Q., et al., 2003a. Eur. J. Pharmacol. 474
Woods, J.S., Fowler, B.A., 1978. Toxicol. Appl. Pharmacol. 43, 361–371. (2-3), 141–147.
Wu, M.-M., Kuo, T.-L., Hwang, Y.-H., et al., 1989. Am. J. Epidemiol. Zheng, P.Z., Wang, K.K., Zhang, Q.Y., et al., 2005. Proc. Natl. Acad.
130, 1123–1132. Sci. USA 102 (21), 7653–7658.
Wu, M.-M., Chiou, H.-Y., Lee, T.-C., et al., 2010. J. Biomed. Sci. 17, 70. Zheng, Y., Wu, J., Ng, J.C., et al., 2002. Toxicol. Lett. 133, 77–82.
Wu, M.-M., Chiou, H.-Y., Chen, C.-L., et al., 2011. Atherosclerosis 19, Zheng, Y., Yamaguchi, H., Tian, C., et al., 2005. Oncogene 24 (20),
704–708. 3339–3347.
Wu, W., Graves, L.M., Jaspers, I., et al., 1999. Am. J. Physiol. 277, Zierler, S., Theodore, M., Cohen, A., 1988. Int. J. Epidemiol. 17, 589–594.
L924–L931. Zoeteman, B.C.J., Brinkmann, F.J.J., 1976. In: Amavis, W., Hunter,
Wu, W., Jaspers, I., Zhang, W., et al., 2002. Am. J. Physiol. Lung Cell W.J., Smeets, J.G.P.M. (Eds.), Hardness of Drinking Water and
Mol. Physiol. 282 (5), L1040–L1048. Public Health. Pergamon Press, Oxford, pp. 173–202.

You might also like