Chromiumvi Erd Toxreview 9-30-10
Chromiumvi Erd Toxreview 9-30-10
Chromiumvi Erd Toxreview 9-30-10
www.epa.gov/iris
TOXICOLOGICAL REVIEW
OF
HEXAVALENT CHROMIUM
(CAS No. 18540-29-9)
September 2010
(Note: This document is a reassessment of the noncancer and cancer health effects
associated with the oral route of exposure only.)
NOTICE
This document is an External Review draft. This information is distributed solely for the
purpose of pre-dissemination peer review under applicable information quality guidelines. It has
not been formally disseminated by EPA. It does not represent and should not be construed to
represent any Agency determination or policy. It is being circulated for review of its technical
accuracy and science policy implications.
This document is a preliminary draft for review purposes only. This information is
distributed solely for the purpose of pre-dissemination peer review under applicable information
quality guidelines. It has not been formally disseminated by EPA. It does not represent and
should not be construed to represent any Agency determination or policy. Mention of trade
names or commercial products does not constitute endorsement or recommendation for use.
Figure 2-1. Definitions of bioaccessibility and biovailability used in this document .................... 4
Figure 2-2. Schematic of possible reaction processes (grey ovals) that determine
disposition and speciation of chromium contamination in the environment ............. 12
Figure 2-3. Stability diagram showing aqueous speciation of chromium at various
equilibrium potential (Eh, volts) and pH ................................................................... 13
Figure 3-1. Estimates of Cr(VI) sequestration or reduction by organs, cell populations and
fluids in the human body relevant to portal of entry uptake or effects on remote
distribution kinetics.................................................................................................... 22
Figure 3-2. Chromium excretion in urine and feces of Sprague-Dawley rats .............................. 32
Figure 3-3. Chromium concentrations in male and female F344 rats following chronic
drinking water consumption of Cr(VI) ...................................................................... 37
Figure 3-4. Chromium absorption and elimination in human volunteers after ingestion of a
single bolus dose in drinking water ........................................................................... 43
Figure 3-5. Biovailability and elimination half-life for chromium ingested by human
volunteers as a single bolus dose in drinking water ................................................... 43
Figure 3-6. Schematic of ingested Cr(VI) to internal dose in GI tissue and blood....................... 51
Figure 3-7. Observed and simulated plasma chromium concentrations predicted by the
PBPK model for a human subject ingesting Cr(VI) dissolved in orange juice
(CrVI-OJ) in the study of Kerger et al. (1996) .......................................................... 57
Figure 3-8. Schematic of structure for PBPK model of chromium in the rat and human ............ 59
Figure 3-9. Extended PBPK model structure to predict chromium distribution in the oral
cavity and GI tract tissue for rats and mice................................................................ 65
Figure 4-1. Ternary DNA adduct formation by chromium......................................................... 185
Figure B-1. Predicted and observed incidence of chronic inflammation of the liver in
female rats exposed to sodium dichromium dihydrate in drinking water for 2
years ......................................................................................................................... B-3
Figure B-2. Predicted and observed incidence of diffuse epithelial hyperplasia in the
duodenum of male mice exposed to sodium dichromium dihydrate in drinking
water for 2 years....................................................................................................... B-5
Figure B-3. Predicted and observed incidence of diffuse epithelial hyperplasia in the
duodenum of female mice exposed to sodium dichromium dihydrate in
drinking water for 2 years ........................................................................................ B-9
Figure B-4. Predicted and observed incidence of histiocytic cellular infiltration in the
livers of female mice exposed to sodium dichromium dihydrate in drinking
water for 2 years..................................................................................................... B-12
Figure B-5. Predicted and observed incidence of pancreas: acinus, cytoplasmic alteration
in female mice exposed to sodium dichromium dihydrate in drinking water for
2 years .................................................................................................................... B-14
The purpose of this Toxicological Review is to provide the scientific support and
rationale for the hazard identification and dose-response assessments in IRIS pertaining to
chronic exposure to hexavalent chromium via ingestion. It is not intended to be a comprehensive
treatise on the chemical or toxicological nature of hexavalent chromium. This document is a
reassessment of the noncancer and cancer health effects associated with the oral route of
exposure. A reassessment of the noncancer and cancer health effects of hexavalent chromium
associated with the inhalation route of exposure will be conducted at a later date.
Section 5, Dose-Response Assessments, is based largely on the work of four independent
groups that have recently evaluated the toxicity of hexavalent chromium via ingestion: (1) U.S.
EPA’s Office of Pesticide Programs (OPP), (2) the New Jersey Department of Environmental
Protection (NJDEP), (3) the California Environmental Protection Agency (CalEPA), and (4) the
Agency for Toxic Substances and Disease Registry (ATSDR). Section 5.1 relies, in part, on
work conducted by ATSDR and CalEPA, and the reference dose (RfD) generally relies on
ATSDR’s analysis for chronic oral exposure to hexavalent chromium. Section 5.3 was
developed, in part, based on work conducted by CalEPA and NJDEP, and the oral cancer slope
factor (CSF) generally relies on NJDEP’s analysis for cancer potency.
The intent of Section 6, Major Conclusions in the Characterization of Hazard and Dose
Response, is to present the major conclusions reached in the derivation of the reference dose,
reference concentration and cancer assessment, where applicable, and to characterize the overall
confidence in the quantitative and qualitative aspects of hazard and dose response by addressing
the quality of data and related uncertainties. The discussion is intended to convey the limitations
of the assessment and to aid and guide the risk assessor in the ensuing steps of the risk
assessment process.
For other general information about this assessment or other questions relating to IRIS,
the reader is referred to EPA’s IRIS Hotline at (202) 566-1676 (phone), (202) 566-1749 (fax), or
[email protected] (e-mail address).
CHEMICAL MANAGER/AUTHOR
AUTHORS
CONTRIBUTING AUTHORS
CONTRACTOR SUPPORT
REVIEWERS
This document has been provided for review to EPA scientists and interagency reviewers
from other federal agencies and White House offices.
This document presents background information and justification for the Integrated Risk
Information System (IRIS) Summary of the hazard identification and dose-response assessments
of ingested hexavalent chromium. IRIS Summaries may include oral reference dose (RfD) and
inhalation reference concentration (RfC) values for chronic and other exposure durations, and a
carcinogenicity assessment. This Toxicological Review of Hexavalent Chromium provides
documentation for oral toxicity values only (i.e., RfD and oral cancer slope factor). A
reassessment of the noncancer and cancer health effects of hexavalent chromium associated with
the inhalation route of exposure will be conducted at a later date.
The RfD and RfC, if derived, provide quantitative information for use in risk assessments
for health effects known or assumed to be produced through a nonlinear (presumed threshold)
mode of action. The RfD (expressed in units of mg/kg-day) is defined as an estimate (with
uncertainty spanning perhaps an order of magnitude) of a daily exposure to the human
population (including sensitive subgroups) that is likely to be without an appreciable risk of
deleterious effects during a lifetime. The inhalation RfC (expressed in units of mg/m3) is
analogous to the oral RfD, but provides a continuous inhalation exposure estimate. The
inhalation RfC considers toxic effects for both the respiratory system (portal-of-entry) and for
effects peripheral to the respiratory system (extrarespiratory or systemic effects). Reference
values are generally derived for chronic exposures (up to a lifetime), but may also be derived for
acute (≤24 hours), short-term (>24 hours up to 30 days), and subchronic (>30 days up to 10% of
lifetime) exposure durations, all of which are derived based on an assumption of continuous
exposure throughout the duration specified. Unless indicated otherwise, the RfD and RfC are
derived for chronic exposure durations.
The carcinogenicity assessment provides information on the carcinogenic hazard
potential of the substance in question and quantitative estimates of risk from oral and inhalation
exposure may be derived. The information includes a weight-of-evidence judgment of the
likelihood that the agent is a human carcinogen and the conditions under which the carcinogenic
effects may be expressed. Quantitative risk estimates may be derived from the application of a
low-dose extrapolation procedure. If derived, the oral slope factor is a plausible upper bound on
the estimate of risk per mg/kg-day of oral exposure. Similarly, an inhalation unit risk is a
plausible upper bound on the estimate of risk per μg/m3 air breathed.
Development of these hazard identification and dose-response assessments for hexavalent
chromium has followed the general guidelines for risk assessment as set forth by the National
Research Council (NRC, 1983). U.S. Environmental Protection Agency (U.S. EPA) Guidelines
and Risk Assessment Forum Technical Panel Reports that may have been used in the
development of this assessment include the following: Guidelines for the Health Risk
This toxicological review restricts its focus to oral exposure to hexavalent chromium
compounds. Hexavalent chromium compounds are a group of substances that contain chromium
in the hexavalent or +6 oxidation state. Hexavalent chromium compounds discussed in this
document include the following: chromium(VI) oxide, chromic acid, and selected salts of the
chromate (CrO42-) and dichromate (Cr2O72-) anions.
This section discusses sources of chromium in the environment and the physicochemical
properties of chromium compounds that determine their environmental bioaccessibility. It is
recognized that the definition of bioaccessibility and bioavailability has differed across
disciplines and can cause confusion especially in a regulatory context (Semple et al., 2004). For
the purposes of this toxicological review, bioaccessibility is defined as the ability of chromium to
be released (e.g., environmental solubilization and reduction or extracellular digestion and
reduction) from the environmental matrix to which it is bound, i.e., the fraction of the dose
ingested that becomes freely available for absorption via crossing a cellular membrane. As
shown in Figure 2-1, this would encompass processes A through C. Key determinants of
environmental bioaccessibility include the following: 1) type of contaminant, 2) contamination
level, 3) type of soil, 4) pH of soil, 5) aging of soil, and 6) metal speciation. Processes that
control the environmental chemistry of chromium include redox transformation,
precipitation/dissolution, and adsorption/desorption reactions.
Processes A through C can also occur internal to an organism, and the same determinants
of bioaccessibility function in that internal environment, e.g., pH in different sections of the
gastrointestinal (GI) lumen. Both environmental and internal bioaccessibility processes are
extracellular. For the purposes of this Toxicological Review, however, this latter internal,
extracellular bioaccessibility is discussed in Section 3 on toxicokinetics.
Bioavailability (processes D and E in Figure 2-1) is defined in this document as the
potential for chromium to cross cellular boundaries, i.e., the degree to which it becomes available
to the target tissue after administration. Key mechanisms of bioavailability that determine
internal tissue dose include the following: uptake through cell membranes, intracellular
Alloys with iron, nickel or cobalt are prepared from metallurgical grade ore (60% ≥
chromic oxide) for use in the production of a wide variety of steels including stainless steel,
austenite steel, and high-speed and high-temperature steels, and in other nonferrous alloys. The
chemical industry generally uses a lower grade chromite ore (≈ 45% chromic oxide) to
synthesize sodium chromate and dichromate from which most other chromium products are
prepared, including products used in pigment manufacture, plating/metal finishing, corrosion
inhibition, organic synthesis, leather tanning, and wood preservation. Other important
anthropogenic sources of chromium in the environment include fuel combustion, cement
production, and sewage sludge incineration/deposition (U.S. EPA, 1984).
Chromated copper arsenate (CCA) wood preservatives to prevent fungal decay and
infestations by wood-boring insects contain hexavalent chromium. The preservatives are used in
the industrial vacuum-impregnation of timber and supplied as pastes or water-based concentrates
that are diluted to between 1 and 10% w/w total salts (Cocker et al., 2006). Wood preservatives
such as CCA are regulated under the Control of Pesticide Regulations 1986, with only approved
compounds placed on the market. Cocker et al. (2006) showed that workers exposed to CCA
wood preservatives have urinary chromium levels significantly higher than nonoccupationally
exposed populations, but below occupational biological exposure indices for urinary chromium
based on inhalation exposures. Balasoiu et al. (2000) evaluated the influence of soil composition
and physicochemical characteristics on the retention and partitioning of chromium in nine CCA
artificially contaminated soils using a statistical mixture design to arrive at the different soil
compositions. Sequential extraction and modified solvent extraction were used to assess the
partitioning. Average metal retention in mineral soils was low (23%), but increased dramatically
Name Chromium(VI) oxidea Chromic acida,b Sodium chromate Sodium dichromate Sodium dichromate, dihydrate
CASRN 1333-82-0 7738-94-5 (H2CrO4); 7775-11-3 10588-01-9 7789-12-0
13530-68-2 (H2Cr2O7)
Synonyms Chromium oxide; Chromic(VI) acid; chromium Sodium chromate(VI); Sodium dichromate(VI); Dichromic acid, disodium salt,
(ChemID hexavalent chromium hydroxide oxide; dichromic acid chromium disodium sodium bichromate; dihydrate
Plus, oxide; chromic (H2Cr2O7) oxide; disodium dichromic acid, disodium
2008) trioxide; chromic chromate; rachromate; salt; bichromate of soda
anhydride; chromic chromic acid, disodium
acid anhydride (Anger salt; chromate of soda
et al., 2005)
Structure 2Na+ O OO 2-
2Na+ O OO 2-
(ChemID O O
Cr Cr Cr Cr 2H2O
Plus, O O
O O O O
2008)
Molecular 99.994 (Lide, 2008) 118.010 (H2CrO4) (Lide, 2008); 161.974 (Lide, 2008) 261.968 (Lide, 2008) 297.999 (Lide, 2008)
weight 218.001 (H2Cr2O7) (ChemID Plus)
Molecular CrO3 (ChemID Plus, H2CrO4; H2Cr2O7 (ChemID Plus, Na2CrO4 (ChemID Na2Cr2O7 (ChemID Plus, Na2Cr2O7•2H2O (ChemID Plus,
formula 2008) 2008) Plus, 2008) 2008) 2008)
Form Dark red, deliquescent Exists only as an aqueous solution Yellow, orthorhombic Light brown to orange- Orange-red, monoclinic,
bipyramidal prismatic (Lide, 2008); yellow to orange-red crystals (Anger et al., red plates (Anger et al., translucent needles (Anger et al.,
crystals, flakes, or (Anger et al., 2005) 2005) 2005) 2005)
granular powder
(O’Neil, 2006)
Stability/ Deliquescent; Strong oxidizing agent (Anger et al., Hygroscopic (Anger et Strongly hygroscopic; Very hygroscopic, deliquesces
reactivity decomposition begins 2005) al., 2005) decomposes above 400°C in air; decomposes above 85°C;
above 198°C (Anger et (Lide, 2008); strong strong oxidizing agent in acid
al., 2005); powerful oxidizing agent (Anger et solution (Lide, 2008; Anger et
oxidizer (O’Neil, 2006) al., 2005) al., 2005)
Melting 197°C (Lide, 2008) Not applicable 794°C (Lide, 2008) 357°C (Lide, 2008) Decomposes prior to melting
point (Lide, 2008)
Density 2.7 g/cm3 (Lide, 2008) Not applicable 2.72 g/cm (Lide, 2008) 2.52 g/cm (Anger et al., 2.35 g/cm3 (Lide, 2008)
3 3
2005)
Water 169 g/100 g H2O at Not applicable 87.6 g/100 g H2O at 187 g/100 g H2O at 25°C 272.9 g/100 g H2O (73.18 wt%)
solubility 25°C (Lide, 2008) 25°C (Lide, 2008) (Lide, 2008) at 20°C (Anger et al., 2005)
Other Soluble in alcohol and Not applicable Slightly soluble in Not available Soluble in acetic acid (Lide,
solubility mineral acids (Lewis, ethanol (Lide, 2008) 2008)
2007)
Name Potassium chromate Potassium dichromate Calcium chromate Ammonium dichromate Zinc chromate Lead chromate
CASRN 7789-00-6 7778-50-9 13765-19-0 7789-09-5 13530-65-9 7758-97-6
Synonyms Potassium Potassium dichromate(VI); Calcium Ammonium bichromate; Zinc chromate(VI) Lead chromate(VI);
(ChemID chromate(VI); bichromate of potash; chromate(VI); ammonium dichromate(VI); hydroxide; buttercup phoenicochroite;
Plus, bipotassium chromate; potassium bichromate; calcium chrome diammonium dichromate; yellow; chromic plumbous
2008) dipotassium chromate; dipotassium bichromate; yellow; calcium chromic acid, diammonium acid, zinc salt; zinc chromate; chromic
chromate of potash; dipotassium dichromate; monochromate; salt chrome yellow; zinc acid, lead salt;
tarapacaite; dipotassium dichromium gelbin; yellow teraoxychromate chrome yellow
chromic acid, heptaoxide; lopezite; ultramarine; chromic (O’Neil, 2006)
dipotassium salt dichromic acid, acid, calcium salt
dipotassium salt
Structure 2K+ O OO 2-
2NH4+ O OO 2-
(ChemID O O
Cr Cr Cr Cr
Plus, O O
O O O O
2008)
Molecular 194.191 (Lide, 2008) 294.185 (Lide, 2008) 156.07 (Lide, 2008) 252.065 (Lide, 2008) 181.403 (Lide, 323.2 (Lide, 2008)
weight 2008)
Molecular K2CrO4 (ChemID Plus, K2Cr2O7 (ChemID Plus, CaCrO4 (ChemID (NH4)2Cr2O7 (ChemID Plus, ZnCrO4 (ChemID PbCrO4 (ChemID
formula 2008) 2008) Plus, 2008) 2008) Plus, 2008) Plus, 2008)
Form Lemon yellow prisms Tabular or prismatic, bright Yellow monoclinic Large, bright, orange-red Yellow prisms Yellow-orange
(Anger et al., 2005) orange-red triclinic crystals or rhombic crystals crystals (Anger et al., 2005) (Lide, 2008) monoclinic crystals
(Anger et al., 2005) (O’Neil, 2006) (Lide, 2008)
Stability/ Nonhygroscopic Nonhygroscopic; Decomposes at Flammable; nonhygroscopic; Not available Not available
reactivity (Anger et al., 2005) decomposes at 500°C 1,000°C (Lide, decomposition begins upon
(Anger et al., 2005; Lide, 2008); oxidizing heating at 180°C (O’Neil,
2008) agent (Lewis, 2007) 2006). Strong oxidizing agent,
may explode in contact with
organic materials (Lewis,
2007)
Melting 974°C (Lide, 2008) 398°C (Lide, 2008) Decomposes prior to Decomposes prior to melting 316°C (Lide, 2008) 844°C (Lide, 2008)
point melting (Lide, 2008) (Lide, 2008)
Density 2.73 g/cm3 (Lide, 2.68 g/cm3 (Lide, 2008) 3.12 g/cm3 (Anger et 2.155 g/cm3 (Lide, 2008) 3.40 g/cm3 (Lide, 6.12 g/cm3 (Lide,
2008) al., 2005) 2008) 2008)
Name Potassium chromate Potassium dichromate Calcium chromate Ammonium dichromate Zinc chromate Lead chromate
Water 65.0 g/100 g H2O at 15.1 g/100 g H2O at 25°C 4.5 g/100 g H2O 35.6 g/100 g H2O at 20°C 3.08 g/100 g H2O 0.000017 g/100 g
solubility 25°C (Lide, 2008) (Lide, 2008) (4.3 wt%) at 0°C (Lide, 2008) (Lide, 2008) H2O at 20°C (Lide,
(Anger et al., 2005) 2008)
Other Insoluble in alcohol Insoluble in alcohol Soluble in dilute Soluble in alcohol (Lewis, Dissolves readily in Insoluble in acetic
solubility (O’Neil, 2006) (Lewis, 2007) acids; practically 2007) acids (Anger et al., acid; soluble in
insoluble in alcohol 2005); insoluble in solutions of fixed
(O’Neil, 2006) acetone (Lide, 2008) alkali hydroxides;
soluble in dilute
nitric acid (O’Neil,
2006)
a
Chromic acid is formed in aqueous solution when chromium(VI) oxide is dissolved in water; it cannot be isolated as a pure compound out of solution (Anger et
al., 2005; Page and Loar, 2004). The term chromic acid is sometimes used in reference to chromium(VI) oxide; however, it should be noted that there is a
structural difference between the anhydrous substance chromium(VI) oxide and the aqueous chromic acid that forms when the oxide is dissolved in water.
b
Chromic acid exists in solution as both H2CrO4 and H2Cr2O7 (Anger et al., 2005; Page and Loar, 2004; Cotton et al., 1999). H2CrO4 is the main species in basic
solutions (pH >6) while H2Cr2O7 is the main species in strongly acidic solutions (pH <1) (Anger et al., 2005; Page and Loar, 2004; Cotton et al., 1999). Both
species are present in equilibrium in solutions that have a pH value between 2 and 6 (Anger et al., 2005; Page and Loar, 2004; Cotton et al., 1999).
The environmental chemistry of chromium is complex. Figure 2-2 illustrates the possible
fates of chromium in soil/water systems. Chromium is known to undergo various chemical and
biological reactions in natural systems that govern its speciation, and in turn, environmental
behavior. Important reactions include oxidation/reduction, precipation/dissolution, and
adsorption/desorption. Both oxidation of Cr(III) and reduction of Cr(VI) can occur in geologic
and aquatic environments. Hexavalent chromium is a strong oxidizing agent and is readily
reduced in the presence of appropriate electron donors as shown here:
−
HCrO4 + 7 H + + 3e − ⇔ Cr 3+ + 4 H 2 O (2-1)
Chromium is a redox active soil contaminant with dramatic alterations in its mobility and
toxicity with changes in oxidation state (Fendorf et al., 2004; Rai et al., 1989). Chromium can
exist in oxidation states ranging from -2 to +6, but only +3 and +6 are typically found within the
range of pH and redox potential common in environmental systems (Shupack, 1991). Figure 2-3
depicts a generalized scheme of equilibrium potentials versus pH for chromium.
−
H 2 CrO4 ⇔ H + + HCrO4 K a1 = 10 0.6 (2-2)
− 2−
HCrO4 ⇔ H + + CrO4 K a 2 = 10 −5.9 (2-3)
Neuss and Rieman (YEAR, as cited in Katz and Salem, 1993) evaluated the acid
functions and arrived at Ka1 and Ka2 values of 1.8 x 10-1 and 3.2 x 10-7. From equations (2-2) and
(2-3) it can be observed that at very low pH values (pH < 1), H2CrO4 is the predominant species,
while between pH 0 and 5.9, the HCrO4- and Cr2O72- anions prevail (Shupack, 1991). At pH 6 or
above, CrO42- prevails. Thus, H2CrO4 and CrO42- should be most abundant in natural systems.
− 2−
2CrO4 + 2 H + ⇔ Cr2 O7 + H 2O K c = 1014.6 (2-4)
[Cr O ]
2 7
2−
= 1014.6
[CrO ] [H ]
(2-5)
2− 2 + 2
4
This reaction is pH sensitive as well, with dichromate favored at lower pH (Losi et al., 1994).
Solving equation (2-4) using values for chromate concentration and pH that would likely be
encountered in contaminated groundwater (5.2 mg/L and 7.0), the ratio of dichromate to
chromate would be 0.04. Thus, hexavalent chromium chemistry in environmental systems is
largely confined to that of the chromate anion.
Speciation of hexavalent (CrVI) and trivalent (CrIII) chromium will generally depend on
a variety of environmental parameters including: pH, concentration, and the ligands available in
the matrices (Katz, 1991). In most natural systems, Cr(VI) will be present as CrO42- and major
trivalent species may include hydroxides and various organic complexes (Losi et al., 1994). The
acid anhydride CrO3 and acid chloride CrO2Cl2, and a wide variety of metal chromates MCrO4
and metal dichromates MCr2O7 are typical hexavalant compounds (Katz and Salem, 1993).
Chromium(VI) may also form other species, including HCr2O7- and CrO42-, but their formation
requires Cr(VI) concentrations >10-2 M, which are not found commonly in natural waters (Baes
and Mesmer, 1986 as cited in Rai et al., 1989).
Natural occurrence of hexavalent chromium is rare as it is readily reduced by organic
matter in the environment (Ashley et al., 2003; Barceloux, 1999; U.S. EPA, 1984). Industrial
releases of hexavalent chromium compounds to surface water and soil can result in the transport
and leaching of these substances into groundwater, provided these substances remain under
oxidizing conditions (Loyuax-Lawniczak et al., 2001; Pellerin and Booker, 2000; James et al.,
1997). Hexavalent chromium compounds released to the environment by anthropogenic sources
may persist in natural waters and soils that contain low amounts of organic matter (Johnson et
al., 2006; Loyaux-Lawniczak et al., 2001; U.S. EPA, 1984). Hexavalent chromium compounds
are considered to be more soluble in water and have greater mobility in soil than other types of
chromium compounds (Loyuax-Lawniczak et al., 2001; James et al., 1997).
Whereas reduction of Cr(VI) is likely to occur in environmental systems, oxidation of
Cr(III) is not frequently observed with the exception of some oxidation in the presence of Mn4+
(Losi et al., 1994). Thus, Cr(III) is considered more stable in most natural systems. Trivalent
Table 2-4. Detection limits for methods commonly used in the analysis of
chromium in water and soil extracts
a
Bartlett (YEAR), see Gochfeld (1991)
b
Mehra and Frankenberger (1989)
c
MISSING
a
Mutti et al. (1979) and Tola et al. (1977) as cited in Paustenbach et al. (1997); bAnderson (1983), Gargas et al. (1994),
and Wiegand et al. (1988) as cited in Paustenbach et al. (1997); cGray and Sterling (1950), Wiegand et al. (1988), and
Kerger et al. (1996) as cited in Paustenbach et al. (1997); dCoogan et al. (1991) as cited in Paustenbach et al. (1997);
e
Wiegand et al. (1988) as cited in Paustenbach et al. (1997); fSaner et al. (1984) as cited in Paustenbach et al. (1997).
The internal environment of the GI tract, similar to the external environment discussed in
Section 2, can also affect the digestion, solubilization, and speciation of chromium compounds,
and thus impact their internal bioaccessibility (defined as the ability of chromium to be released
from the environmental matrix to which it is bound, i.e., the fraction of the dose ingested that
becomes freely available for absorption via crossing a cellular membrane), bioavailability
(defined as the potential for chromium to cross cellular boundaries, i.e., the degree to which it
becomes available to the target tissue after administration), and toxicokinetics within the body.
This section of the toxicological review discusses key determinants of these internal processes,
and why insights into how these processes vary across species and tissues are a critical
consideration for the mode of carcinogenic action of hexavalent chromium, Cr(VI).
As depicted in Figure 2-1 in the previous section, both environmental and internal
bioaccessibility processes are extracellular. Section 3.1 will discuss those physiological
processes, internal to the organism (but still extracellular), that impact bioaccessibility.
Bioavailability (processes D and E in Figure 2-1) will be discussed in Section 3.2. The available
data on the biochemistry of intracellular reactions involving chromium will be summarized in
Section 3.3. In Section 3.4, the impact of bioaccessibility and bioavailability on the toxicokinetic
component of the cancer mode of action for Cr(VI) will be discussed. These toxicokinetic
considerations are an important aspect of the mode of action and provide insights into the
evaluation of the tissue responses discussed in Section 4. Section 3.5 includes an evaluation and
discussion of available model structures to describe internal dosimetry of Cr(VI). Finally,
considerations of chromium essentiality versus toxicity are discussed in Section 3.6.
Binding by Uptake
gastric juice by intestinal rings
Tissue preparation (µg/ml)1 (µg/gm)1
Trivalent Cr51Cl3 without gastric juice – 0.9 ± 0.1
Plus gastric juice pH 7.0 1.8 ± 0.2 0.2 ± 0.1
Plus gastric juice pH 1.4 2.0 ± 0.2 0.2 ± 0.1
1
Mean result of 4 experiments ± SD
DeFlora and collaborators (DeFlora et al., 1997; DeFlora and Wetterhahn, 1989; De Flora
et al., 1987; Petrilli and DeFlora, 1982) performed a series of studies to estimate the ability of
various human physiological fluids and tissues to reduce or sequester Cr(VI). The summary data
shown in Table 3-2 and depicted in Figure 3-1 represent a synthesis of data from studies in that
laboratory with anatomical and physiological parameters as described in detail in the source
reference (DeFlora et al., 1997). These parameters were used to arrive at estimates of the overall
Cr(VI) reducing or sequestering capacity of human body compartments relative to oral and
inhalation exposures. The general term “sequestration” was used to connote when intact cells
were tested and the term “reduction” was used when cell homogenates or their subfractions were
tested in the presence of an exogenous NADPH-generating system, an S9 mix. Estimates of
overall Cr(VI) reducing or sequestering capacity were calculated by multiplying the specific
reducing activity of a given organ, cell population, or fluid expressed as µg Cr(VI) reduced per
unit of weight, volume, or number, by the average content of the same organ, cell population, or
fluid in the human body (De Flora et al., 1997). De Flora (2000) proposed that these reduction
capacities account for the limited toxicity for Cr(VI) after oral ingestion due to efficient
detoxication by saliva, gastric juice and intestinal bacteria; similarly, lung cancer is only induced
when Cr(VI) doses overwhelm the reductive capacity of the fluid of the epithelial lining,
pulmonary alveolar macrophages, and bronchial tree and peripheral lung parenchyma cells. De
Flora (2000) also suggested that efficient uptake and reduction of Cr(VI) in RBC explains the
lack of carcinogenicity at sites remote to the portal of entry.
Figure 3-1. Estimates of Cr(VI) sequestration or reduction by organs, cell populations and
fluids in the human body relevant to portal of entry uptake or effects on remote
distribution kinetics. See Table 3-2 and text for details on calculations.
Table 3-4. Distribution and retention of chromium in the rat after a single oral dose
In the second part of the same study, MacKenzie et al. (1959) administered 131 µg
chromium (as Na251CrO4) and blood was removed from the heart at 4 hour post exposure for
analysis of chromium in RBCs and plasma. In a third experiment, MacKenzie et al. (1959) also
evaluated the role of the stomach wall in absorption. Using the same dosing, the influence of the
stomach was bypassed by injecting chromium directly into the intestine, about 4 cm below the
stomach. The RBC and plasma were measured for radioactivity 4 hour post exposure as in the
second part of the study. Table 3-5 shows that the concentrations of chromium in whole blood,
plasma, and RBCs were greater after administration to the intestine than to the stomach (0
indicates no significant count above background).
Since RBC chromium is assumed to be in the hexavalent form and plasma chromium is assumed
to be in the trivalent form, it appeared that there was some reduction of the hexavalent form in
both fasted and nonfasted animals. Ratios for the counts in RBC to plasma (RBC:plasma) are
shown in Table 3-6.
Coogan et al. (1991a) exposed male F344 rats to hexavalent chromium as potassium
chromate (K2CrO4) dissolved in their drinking water at concentrations of 100 and 200 ppm for 3
or 6 weeks. Total chromium concentrations were measured in lung, liver, kidney, and blood by
AAS after acid digestion. Drinking water consumption was reduced at both concentrations for
the first 3 weeks and then only at the higher concentration the second 3 weeks. Chromium was
not detected in any lung samples. At both concentrations and durations, the order of tissue
chromium concentrations was as follows: kidney > liver > blood. Although a general trend of
increasing chromium content as a function of exposure duration existed for the liver, kidney, and
blood samples analyzed, only the kidney samples were significantly different between the 3- and
6-week sacrifices (p < 0.05). Blood chromium levels were not significantly different at either
sacrifice or at either concentration.
Witmer et al. (1991, 1989) performed several studies to evaluate the toxicity of
chromium contaminated soil samples from sites in Jersey City, NJ. In all studies, the organs
evaluated for chromium content included the liver, lung, spleen, kidney, muscle, brain, and
testes. An aliquot of blood from the abdominal aorta was also analyzed. Chromium was
identified in samples by the Baird ICP method (urine and feces) or by atomic absorption using a
graphite furnace (other tissues). In the initial pilot study, male Sprague Dawley rats were dosed
by gavage with 0, 20, 40 and 100 µmole/kg hexavalent chromium as Na2CrO4 · 4H2O in distilled
water for 7 days. Recovery of chromium was low, tissue burdens at the highest dose represented
only 1.7% of the amount administered. When tissue burden was expressed on a µg/g basis, the
kidney contained the highest amount with liver and blood also containing greater amounts than
the other samples. In the next experiment, doses of 120 µmole Cr/kg were administered to
Sprague-Dawley rats (n = 3/group) using four sources of chromium: 1) Na2CrO4, 2) CaCrO4, 3)
Figure 3-2. Chromium excretion in urine and feces of Sprague-Dawley rats. Rats were
gavaged with corn oil (control), calcium chromate, or chromium-contaminated soil at a level of
240 µmole Cr/kg daily for either 2 days (upper two panels) or 8 days (lower two panels). Urine
and feces were collected at 6, 12, and 24 hour after each dosing period. Chromium levels shown
are for each time period as well as for the total 24-hour period.
1
n = 6 per time point per exposure group
2
Non detect
Table 3-9. Chromium in tissues (μg/g wet tissue or μg/mL blood) of mice
and rats after ingesting K2CrO7 in drinking water (8 mg hexavalent
chromium/kg-day) for 4 or 8 weeks
Figure 3-3. Chromium concentrations in male and female F344 rats following chronic
drinking water consumption of Cr(VI). Bone (upper left), renal (upper right), and liver (lower
left) tissue burdens or total body burden (lower right) are mean ± SE. Means that do not share a
common superscript are significantly different (p ≤ 0.05).
Assuming a similar consumption and weight pattern in these satellite animals, it can be seen that
the mice generally consumed 1.5 to 1.8 more chromium as an average daily dose (mg/kg), which
may account to some degree for the larger tissue concentrations in this species observed in Table
3-10.
Forestomach
Day 6 0.098 ± 0.024 0.076 ± 0.004 0.122 ± 0.008 0.294 ± 0.029 0.285 ± 0.283
Day 13 0.091 ± 0.015 0.102 ± 0.034 0.171 ± 0.050 0.221 ± 0.055* 0.593 ± 0.159**
Day 182 0.089 ± 0.018 0.099 ± 0.003 0.338 ± 0.022* 0.574 ± 0.171* 1.654 ± 0.244**
Day 371 0.090 ± 0.015 0.118 ± 0.008 0.328 ± 0.081* 1.338 ± 0.444** 2.849 ± 0.975**
Mice
Glandular stomach
Day 6 0.306 ± 0.056 0.645 ± 0.253 1.258 ± 0.290* 2.450 ± 0.266** 5.785 ± 0.131**
Day 13 0.207 ± 0.053 0.324 ± 0.030 2.614 ± 0.190* 7.048 ± 1.751** 13.130 ± 2.604**
Day 182 0.305 ± 0.078 0.644 ± 0.035* 3.659 ± 0.547** 11.520 ± 3.017** 52.673 ± 12.310**
Day 371 0.731 ± 0.306 0.676 ± 0.104 2.807 ± 0.330* 9.994 ± 1.079* 49.867 ± 12.251**
Forestomach
Day 6 0.328 ± 0.132 0.683 ± 0.262 1.308 ± 0.553 1.102 ± 0.373 1.286 ± 0.116
Day 13 0.201 ± 0.094 0.288 ± 0.056 0.400 ± 0.044 2.030 ± 0.532* 3.849 ± 1.811*
Day 182 0.173 ± 0.064 0.444 ± 0.099 1.033 ± 0.102* 2.141 ± 0.643** 9.624 ± 3.638**
Day 371 0.320 ± 0.049 0.381 ± 0.077 1.271 ± 0.300* 1.812 ± 0.208* 7.442 ± 0.764**
1
n = 3; mean ± SE. Statistical tests performed on unrounded data.
*significantly different from the control group (p ≤ 0.05) and ** (p ≤ 0.01) by Shirley’s test
The dosing regimen was designed to achieve a steady-state urine concentration during dosing
and to ensure a return to baseline between dosing with Cr(VI) and Cr(III). Chromium picolinate
was delivered at a dose of 0.2 mg for the first seven days, considered a loading dose for this
dietary supplement. Cr(VI) was delivered as potassium dichromate (K2Cr2O4) on days 8 through
10 in a capsule at a dose of 0.005 mg Cr(VI), the U.S. EPA RfD for Cr(VI) at that time. Cr(III)
was then ingested as chromic oxide (Cr2O3) at 1.0 mg Cr(III)/kg/day, the U.S. EPA RfD level for
Cr(III) at that time, on days 14 through 16. Urine samples and measurements were performed as
previously described for Kerger et al. (1996, 1997). The ingestion of chromium picolinate
resulted in significantly elevated urine concentrations such that participants routinely exceeded
background. Ingestion of Cr(VI) yielded individual mean total urinary chromium levels of 1.2–
23 µg/L, and a pooled mean value of 2.4 µg/L. Ingestion of the Cr(III) compound yielded no
significant increases in urinary chromium concentrations, suggesting negligible absorption.
Paustenbach et al. (1996) evaluated uptake and elimination in a male, Caucasian
volunteer (age 44 years) who ingested deionized water containing potassium dichromate
(K2Cr2O4) in five daily portions (400 mL, 2 mg Cr(VI)/L each) for 17 days. Methods described
for the Kerger et al. (1996) study were used to sample and quantify chromium in blood and
urine. Bioavailability was estimated at 2% and the plasma elimination half-life at 36 hour, both
consistent with the previous studies. The time to achieve steady-state concentrations in urine and
blood was 7 days. Both plasma and RBC chromium concentrations returned rapidly to
background levels within a few days, again suggesting that concentrations of 10 mg Cr(VI)/L or
less in drinking water of humans appears to be completely reduced to Cr(III) prior to systemic
distribution.
Finley et al. (1997) extended this work by evaluating chromium kinetics in human
volunteers following repeated oral exposure to Cr(VI) concentrations ranging from 0.1 to 10.0
mg/L. Five healthy, adult (age 30 to 54 years), male Caucasian volunteers ingested a liter (in
three volumes of 333 ml at approximate 6-hour intervals) of deionized water containing Cr(VI)
concentrations of 0.1, 0.5, 1.0, 5.0 and 10.0 mg Cr(VI)/L. Potassium chromate (K2CrO4) was
used as the source of soluble Cr(VI). Other experimental methods and total chrome analyses in
the samples were as in Kerger et al. (1996); a study of the stability of the dosing solution
confirmed no measurable reduction of Cr(VI) occurred throughout the study. Each of the five
subjects demonstrated an increase in the amount of urinary chromium excreted, ranging from a
mean of 1.7% at the low dose (0.1 mg Cr(VI)/L) to 3.5% at the high dose (10.0 mg Cr(VI)/L). A
dose-related increase in plasma chromium began at the 5 mg Cr(VI)/day dose, with 2 subjects
not clearly increasing above baseline at either 5 or 10 mg Cr(VI)/day. The RBC chromium
Parameter Observation
Vmax 10.4–10.7
Km 1.04–1.68
Inhibition by O2 26–37%
Inhibition by TlCl3 96–100%
Inhibition by P450 inhibitors
Carbon monoxide None
Piperonyl butoxide None
Aminopyrine None
Figure 3-6. Schematic of ingested Cr(VI) to internal dose in GI tissue and blood.
Intracellular molecular mechanisms of biological disposition in GI tissue are expected in
other target tissues such as respiratory tract, liver, and kidney. See text for discussion.
1. General model structure taken from the existing PBPK model for lead (O’Flaherty,
1991a,b) with exclusion of any slow exchange in bone compartment. Relative
magnitudes of rapid surface exchange at bone surfaces and formation/resorption of bone
in juvenile and mature rats based on visual fit of model to data of Hopkins (1965), a study
in which Cr51Cl3 was administered by i.v. and radiolabel monitored for 72 days following
injection. The declining body burden data were fit with a three-term sum of
exponentials; the third term was presumed to be most closely related to loss of chromium
from bone, with bone data reported at 0.25, 4.0 and 24 hours.
2. Extension of time frame of model predictions. Starting value for plasma Cr(III) clearance
estimated from chromium body burden data of Hopkins (1965), the second term in that
data (described in Step 1) corresponding to a half-life of 5.9 days, results in a clearance
rate of 0.025 L Cr/day. Scaling by body weight0.75 resulted in whole-body clearance of
Cr(III) of 0.055 L Cr/day/kg. This value was later calibrated to be 0.065 to be more
consistent with other in vivo data sets after drinking water exposure (e.g., MacKenzie et
al., 1959).
4. Total clearance of Cr(III) was fractionated into clearances into bile, urine, and GI tract as
1%, 90% of the remainder, and 10% of the remainder, respectively; primarily based on
the data of Cikrt and Bencko (1979) who administered Cr(III) salt by i.v. Further work
on the model showed that the data of Bragt and Van Dura (1983) and Edel and Sabbioni
(1985) were not compatible with significant transintestinal and biliary excretion, so these
two fractions were subsequently set at 0 in final form.
5. Addition of RBC compartment in communication with arterial blood. The first-order rate
constants for loss of Cr(III) from RBC to plasma and transfer of Cr(III) from plasma to
the RBC were fixed on the basis of measured half-life of chromium association with RBC
(Bishop and Surgenor, 1964). These were not changed in further model development.
Estimates of the rate constant for transfer of Cr(VI) from plasma to RBC based on in
vitro data on human RBC suspended in either saline (Gray and Sterling, 1950) or blood
plasma (Weigand et al., 1985). The same half-life was initially used for transfer of
Cr(VI) between plasma and peripheral tissues, but these were changed for poorly
perfused tissues in order to fit the data of Weber (1983).
6. Total excretion clearance for Cr(VI) set and changed as above in Step 4 for Cr(III).
7. Percent of chromium dose excreted in the urine in 24 hour following i.v. injection of a
soluble salt of either Cr(III) or Cr(VI) in rats was reported by Cikrt and Bencko (1979) to
be independent of oxidation state of administered compound. Initial estimate of
excretion clearance of Cr(VI) was set equal to Cr(III) and the two values remained equal
in the course of model development.
8. Link of Cr(VI) model to Cr(III) model by reduction processes in all tissues. A single
value of the first-order reduction rate constant was provisionally assigned to the Cr(VI)
pools in the RBCs, peripheral tissues, and lung. This simplification proved satisfactory
and was retained in the final model. The value of the reduction rate constant was
determined by fitting the tissue concentration data of Weber (1983) in accordance with
the results of studies in which little or no reduction was observed in human plasma in
vitro (Gray and Sterling, 1950; Korallus et al., 1984).
9 Expansion of i.v. model to include GI uptake. First-order rate constant set at 0.01 per day
for GI absorption of Cr(III) estimated from the single bolus dose data of MacKenzie et al.
(1959) at 1.8%, shown to be in agreement with the estimate of Mertz et al. (1965) at 2–
3% for oral administration. The same study found 85% of an orally administered soluble
Cr(VI) salt had been reduced before it could reach the intestinal absorption site. Model
was run with inclusion of reduction pathway and the first-order reaction rate constant set
to give 85% reduction and 10% absorption.
10. Final structural development step was to expand the composite model to allow absorption
and elimination of chromium from the lung. Pulmonary clearance of Cr(VI) salts is not
dose-dependent within a reasonable dose range (Weber, 1983; Bragt and van Dura,
1983). Mucociliary transfer to the GI tract identified as second route of chromium
clearance. Both Cr(III) and Cr(VI) assigned to two lung pools in the model. Chromium
11. Adjustment of initial model parameter values by visual optimization to fit data from
O’Flaherty and Radike (1991). Published parameter estimates, especially for tissue
uptake and loss were refined by optimization of model predictions to the data set of
Weber (1983) in conjunction with the data sets of Bragt and Van Dura (1983) and Edel
and Sabbioni (1985). All were studies of radiolabeled chromium following i.t.
administration of soluble 51Cr(VI), or in one instance 51Cr(III), salts. The Weber (1983)
data set consisted of time course data in several tissues, whole blood, plasma, GI tract,
and carcass for 42 days after a single i.t. dose. The Bragt and Van Dura and Edel and
Sabbioni data sets had only limited tissue concentration measurements, but extensive
measurements of cumulative excretion in urine and feces.
To test the generalizeability of the final form of the model, the inhalation study of
Langard et al. (1978) was simulated. The study of Langard et al. (1978) consisted of a series of
inhalation exposures of rats to zinc chromate dust, 6 hour/day for 4 days followed by 4 days
during which urinary excretion was monitored and 37 days during which blood chromium levels
were monitored. Agreement of the O’Flaherty model against these data was reasonably good
despite the route extrapolation required. However, the model only fit the data of the MacKenzie
et al. (1958) drinking water study moderately well, as the nonlinear kidney and liver
oncentrations were over predicted. Modification of the uptake description to include a
Michaelis-Menton term may address this issue (see below).
The human model (O’Flaherty, 2001) was developed based on the rat using appropriate
scaling of physiological parameters and by estimating specific chromium-related parameters
using several studies in adult human volunteers administered chromium in drinking water
(described in Section 3.2 [Finley et al., 1997; Kerger et al., 1996; Paustenbach et al., 1996]).
Default values, determined by gender and age, were used for their body weights. The studies of
Kerger et al. (1996) and Finley et al. (1997) were used for calibration of the chromium-specific
parameters (e.g., clearance constants) in the model structure and the Paustenbach et al. (1996)
study was used for verification. In the absence of data on the magnitude of the rate of deposition
of Cr(III) or Cr(VI) in human bone, the fractional rates of deposition were assigned the values of
5 for Cr(III) and 15 for Cr(VI), the same values used in the rat model.
The human model generated reasonable time profiles to the data of Paustenbach et al.
(1996) despite the variability of the urinary excretion rates. Figure 3-7 illustrates the dependence
of the urinary excretion on the form of the chromium administered. As discussed for the data of
Kerger et al. (1996), when chromium has been administered as Cr(III) citrate rather than as the
inorganic salts of CrCl3 or K2Cr2O7, transfer of chromium from the blood to a compartment from
which excretion occurs is favored relative to other tissues. As a result, when the CrCl3 clearance
curve is applied and the excretion rates were optimized to fit urinary excretion rates, plasma
INHALATION
LUNG
EXPOSURE
POOL A
Cr(VI)→Cr(III)
WELL-PERFUSED
Cr(VI)→Cr(III)
POORLY-PERFUSED
Cr(VI)→Cr(III)
Cr(VI) →Cr(III)
RED CELLS
PLASMA
PLASMA
BONE
Cr(VI)→Cr(III)
ORAL
LIVER GI TRACT EXPOSURE
Cr(VI)→Cr(III) Cr(VI)→Cr(III)
KIDNEY
Cr(VI)→Cr(III)
FECAL EXCRETION
URINARY
EXCRETION
URINARY PATH
Figure 3-8. Schematic of structure for PBPK model of chromium in the rat
and human.
Rat Human
a
Parameter Cr(III) Cr(VI) Cr(III) Cr(VI) Definition
Absorption
KGI 0.01 0.04 0.25 2.5 First-order rate constant for absorption from the GI tract (Da-1)
KLU 0.2 2.0 NA NA First-order rate constant for absorption from the bioavailable
lung pool (pool A) (Da-1)
KMUCOA 0.8 0.8 NA NA First-order rate constant for mucociliary clearance from pool
A to the GI tract (Da-1)
KMUCOB 0.025 0.025 NA NA First-order rate constant for mucociliary clearance from the
nonbioavailable lung pool (pool B) to the GI tract (Da-1)
KLUAB 1.2 1.2 NA NA First-order rate constant for transfer from pool A to pool B
(Da-1)
FRLUNG NA NA 0.3 0.3 Fraction of inhaled chromium absorbed to blood
FRTRGI NA NA 0.7 0.7 Fraction of inhaled chromium transferred to GI tract
Distribution
b b
CR 5.0 15.0 NA NA Relative clearance of chromium into mineralizing bone
(L blood plasma cleared/L new bone formed)
KINRBC 0.0003 1.5 12.0 NA Clearance from plasma to red cell (L/Da)
KDIN 0.007 1.5 3.0 30.0 Clearance from plasma to kidney (L/Da)
LDIN 0.0001 1.5 3.0 30.0 Clearance from plasma to liver (L/Da)
WDIN 0.0001 1.5 3.0 30.0 Clearance from plasma to other well-perfused tissues (L/Da)
PDIN 0.0001 0.01 3.0 30.0 Clearance from plasma to poorly-perfused tissues (L/Da)
BDIN 0.0001 0.01 NAb NAb Clearance from plasma to bone (L/Da)
CR NA NA 5.0 15.0 Fraction deposition from blood to forming bone
KOUTRBC 0.0003 10.0 12.0 NA Clearance from red cell to plasma (L/Da)
KDOUT 0.001 10.0 3.0 30.0 Clearance from kidney to plasma (L/Da)
LDOUT 0.0003 10.0 3.0 30.0 Clearance from liver to plasma (L/Da)
WDOUT 0.001 10.0 3.0 30.0 Clearance from other well-perfused tissues to plasma (L/Da)
PDOUT 0.003 10.0 3.0 30.0 Clearance from poorly perfused tissues to plasma (L/Da)
BDOUT 0.003 10.0 NAb NAb Clearance from bone to plasma (L/Da)
Excretion
KFX 1.5 1.5 14.0 14.0 First-order rate constant for loss of chromium from intestinal
tract contents to the feces (Da-1)
QEC 0.065 0.065 NAc NAc Excretion clearance from the plasma (urinary clearance)
(L/kg/Da)
CLEARc NA NA 12.0 12.0 Parameter in expression for clearance from blood plasma to
urine (L/d)
MAXc NA NA 0.008 0.008 Parameter in expression for clearance from blood plasma to
urine (mg/d)
KMc NA NA 0.0008 0.0008 Parameter in expression for clearance from blood plasma to
urine (mg/L)
FB 0.0 0.0 NA NA Fraction of body burden secreted in the bile
FI 0.0 0.0 NA NA Fraction of body burden excreted via the GI tract
Rat Human
a
Parameter Cr(III) Cr(VI) Cr(III) Cr(VI) Definition
Reduction
KREDRC NA 0.7 NA 7.0 First-order rate constant for reduction of hexavalent chromium
to Cr(III) in the red cell (Da-1)
KREDBP NA NA NA 0.2 First-order rate constant for reduction of hexavalent chromium
to Cr(III) in blood plasma (Da-1)
KREDKL NA NA NA 500.0 First-order rate constant for reduction of hexavalent chromium
to Cr(III) in kidney (Da-1)
KREDGI NA 10.0 NA 100.0 First-order rate constant for reduction of hexavalent chromium
to Cr(III) in GI tract contents (Da-1)
KRED NA 0.5 NA 5.0 First-order rate constant for reduction of hexavalent chromium
to Cr(III) in all other tissues and in lung contents (Da-1)
Lag time for excretion of urine
FRHOLD 0.7 0.7 NA NA Fraction of urinary chromium not excreted immediately; that
is, temporarily held in pool
KHOLD 0.05 0.05 NA NA First-order rate constant for excretion from the retained urine
pool (Da-1)
FR 0.10 0.10 NA NA Fraction of chromium in retained urine that is associated with
the kidney
a
Parameter names are those for human model in cases where the reported rat and human parameter names were not
identical.
b
Exchanges between blood plasma and cortical and trabecular bone are simulated as functions of bone formation
and resorption rates.
c
MAX
QE = CLEAR −
KM + CBP , where QE is clearance from blood plasma to urine (L/d) and CBP is plasma
concentration of chromium (mg/L).
NA = not applicable
Sources: O’Flaherty (1996) (rat parameters); O’Flaherty et al. (2001) (human parameters).
Inhalation
Pool A
Lung
CrVI →CrIII
Well Perfused
Red Blood Cells
CrVI →CrIII
CrVI →CrIII
Plasma
Poorly Perfused
CrVI →CrIII
Drinking
Water
Bone
CrVI →CrIII
Buccal Cavity
Liver CrVI →CrIII
CrVI →CrIII
GI Tract
Kidney
CrVI →CrIII
CrVI →CrIII
Urinary
Excretion Fecal
Retained Excretion Oral
Urine Exposure
Feces
Lumen Cr Solution
+ Saliva
Cr(VI) Reduction Cr(III)
?
Epithelial
Tissue Reduction
Cr(VI) Cr(III)
?
Submucosal
Tissue Cr(VI) Cr(III)
Blood Flow
Liaoning Province, China (Kerger et al., 2009; Beaumont et al., 2008; Zhang and Li, 1997,
1987)
In 1987, Zhang and Li published a paper describing the soil and water contamination by
chromium in the vicinity of an alloy plant where chromium was smelted in the JinZhou area of
Liaoning Province, China (Zhang and Li, 1987). This paper was based on an earlier unpublished
report (Zhang and Li, 1980). A more detailed mortality analysis, which included variation in
cancer mortality rates among the five villages along the Nuer River, was published in 1997
(Zhang and Li, 1997) in the Journal of Occupational and Environmental Medicine. This study
has had a controversial history that culminated in the retraction, in 2006, of the latest report
(Zhang and Li, 1997) by the editors of the Journal of Occupational and Environmental Medicine
because “financial and intellectual input to the paper by outside parties was not disclosed”
(Brandt-Rauf, 2006). The financial and intellectual input in question were those from a
consulting firm that had (or may have had) financial ties with industry clients potentially liable
a
As reported by Beaumont et al. (2008).
b
As reported by Kerger et al. (2009).
c
As reported by Zhang and Li (1986), number of samples not stated.
d
Zhang and Li (1986) report this concentration as 70.5 mg/L, but Zhang and Li (1987), Beaumont et al. (2008), and
Kerger et al. (2009) report a concentration of 10.5 mg/L. The total number of samples and the range in
concentrations were not specified.
A mortality study was described first by Zhang and Li (1980) in an unpublished report
for the JinZhou health department, and later published in a Chinese journal (Zhang and Li,
1987). Mortality records for the period 1970–1978 were obtained from local police stations for
the five villages along the Nuer River, the district surrounding the ferrochromium alloy plant
(TangHeZi), and three other areas to the west (YaoTangHeZi) and north (North ThangHeZi,
North Nuer River) of the plant. TangHeZi and the other three areas were not affected by the
groundwater chromium contamination, and these areas serve as one of the comparison groups in
the analyses. Cause of death was abstracted by trained study staff and reviewed by Dr. Zhang
(Kerger et al., 2009). A study interview was also conducted (with unspecified surrogates), but
a
Area population figures are based on approximate 1975 data from Beaumont et al. (2008); group values are total person-yrs for the combined area.
b
As calculated by Beaumont et al. (2008). Nuer River Village stomach cancer rate was not included in the primary analysis by Beaumont et al. (2008) because it was
missing in the original (1980) report; an additional analysis used a rate of 28 as reported by Zhang and Li (1987).
c
Beaumont et al. (2008) estimate was 98,458 and Kerger et al. (2009) estimate was 98,850.
d
Beaumont et al. (2008) report this value as 26.14 in Table 2, but based on the calculation of the estimated age-adjusted rate, it appears that a value close to 26.3 was
used; Kerger et al. (2009) report this value as 26.4.
e
Beaumont et al. (2008) estimate was 252,277 and Kerger et al. (2009) estimate was 253,282.
f
As reported by Kerger et al. (2009).
Table 4-3. Risk ratios comparing cancer mortality rates in five villages
along a path of groundwater contamination from an alloy plant and other
comparison areas in western JinZhou, China from 1970 to 1978
a
TangHeZi, North TangHeZi, North Nuer River, and YaoTangHeZi.
b
Reported by Beaumont et al. (2008).
c
Reported by Kerger et al. (2009).
Kerger et al. (2009) also presented results of analyses of variation in cancer rates within
the five villages in the chromium contamination zone, using three measures of exposure
potential: distance from the plant, average hexavalent chromium concentrations in 1965, and
percent of wells with >0.05 mg/L hexavalent chromium in 1965 (these measures can be found in
Table 4-1). The analysis was based on Poisson regression of the log-transformed cancer rate in
relation to the exposure measures (separate models run for each measure). For the distance
a
Values are means ± SE; values in parenthesis are percent of control; n = 10 rats/group, with the following
exceptions: 1.7 mg hexavalent chromium/kg-d group females on d 23 and mo 3 (n = 9), 3.5 mg hexavalent
chromium/kg-d group females on d 23 (n = 8), 5.9 mg hexavalent chromium/kg-d group females on d 23 (n = 9),
and 20.9 mg hexavalent chromium/kg-d group females on d 23 and mo 3 (n = 9).
b
Significantly different (p ≤ 0.01) from the control group by Dunn’s or Shirley’s test.
c
Significantly different (p ≤ 0.05) from the control group by Dunn’s or Shirley’s test.
Results of clinical chemistry analyses in male and female rats exposed to sodium
dichromate dihydrate in drinking water showed treatment-related increases in serum liver
enzyme activities, bile acids, and serum creatine kinase activity and alterations in lipid
metabolism (Table 4-5) (NTP, 2007). Serum alanine aminotransferase (ALT) and sorbitol
dehydrogenase (SDH) activities were significantly increased compared to controls in all
treatment groups at 3 months, with less severe effects seen at 23 days. A consistent relationship
between severity and dose was not observed. In male rats, elevations of ALT and SDH activities
increased with increasing dose between 1.7 and 11.2 mg/kg-day, but less severe elevations were
observed at 20.9 mg/kg-day (Table 4-5). In females, increases in ALT and SDH activities were
generally indicative of a uniform effect across the dose range (Table 4-5). NTP (2007) suggested
that increases are consistent with hepatocellular injury or membrane leakage. At 3 months, bile
acids were significantly increased compared to controls at ≥11.2 mg hexavalent chromium/kg-
day in males and in all treatment groups (except 5.9 mg hexavalent chromium/kg-day) in
females; similar to serum liver enzymes, increases in bile acids were not consistently related to
dose. NTP (2007) suggested that increased bile acid was indicative of hepatic toxicity rather
than colestasis, as other markers of colestasis (e.g., alkaline phosphatase [AP] and 5N-
nucleotidase) were not affected by treatment. At 3 months, decreased serum cholesterol and
triglycerides, indicative of altered lipid metabolism, were observed; however, a consistent
relationship between severity and dose was not observed. At 3 months, dose-related increases in
serum creatine kinase activity, indicative of muscle damage, were observed in males and females
Table 4-5. Clinical chemistry effects in male and female F344/N rats
exposed to sodium dichromate dihydrate in drinking water for 3 months
a
Values are means ± SE; values in parenthesis are percent of control; n = 10 rats/group, with the following
exceptions: control group males (n = 9), 1.7 mg hexavalent chromium/kg-d group females (n = 9), and 20.9 mg
hexavalent chromium/kg-d group females (n = 9).
b
Significantly different (p ≤ 0.05) from the control group by Dunn’s or Shirley’s test.
c
Significantly different (p ≤ 0.01) from the control group by Dunn’s or Shirley’s test.
Table 4-6. Selected organ weights in male and female F344/N rats exposed
to sodium dichromate dihydrate in drinking water for 3 months
a
Values are means ± SE; n = 10 rats/group.
b
Significantly different (p ≤ 0.01) from the control group by Williams’s or Dunnett’s test.
c
Relative weight = mg organ weight/g body weight.
d
Significantly different (p ≤ 0.05) from the control group by Williams’s or Dunnett’s test.
Gross and microscopic examinations of male and female rats exposed to sodium
dichromate dihydrate in drinking water for 3 months showed nonneoplastic lesions of the
duodenum, glandular stomach, pancreatic lymph nodes, liver (females only), and bone marrow
(females only) (NTP, 2007); incidence data are summarized in Table 4-7. The incidence of
minimal-to-mild duodenal histiocytic cellular infiltration was increased in males and females at
≥3.5 and ≥1.7 mg hexavalent chromium/kg-day, respectively, compared to controls; incidence
increased with dose. Histiocytic cellular inflammation appeared as multifocal, randomly
scattered, small clusters of enlarged macrophages with pale foamy cytoplasm. Incidences of
nonneoplastic lesions of the glandular stomach (ulcer, focal regenerative hyperplasia, and focal
squamous hyperplasia, all mild-to-moderate in severity) were increased in rats in the highest
dose group. Microscopically, ulcers were characterized by complete loss of the lining of the
mucosal epithelium with necrosis, often extending through to the submucosa, and muscle layers;
mild to marked chronic inflammation (infiltrates of neutrophils, macrophages, lymphocytes, and
a
Number of animals with lesion/number of animals examined; parenthesis indicate average severity grade, with
1 = minimal; 2 = mild; 3 = moderate; 4 = severe.
b
Significantly different (p ≤ 0.01) from the control group by Fisher’s exact test.
c
Significantly different (p ≤ 0.05) from the control group by Fisher’s exact test.
Table 4-8. Selected organ weights in male and female B6C3F1 mice exposed
to sodium dichromate dihydrate in drinking water for 3 months
a
Values are means ± SE; n = 10 mice/group.
b
Significantly different (p ≤ 0.05) from the control group by Williams’s or Dunnett’s test.
c
Significantly different (p ≤ 0.01) from the control group by Williams’s or Dunnett’s test.
d
Relative weight = mg organ weight/g body weight.
a
Number of animals with lesion/number of animals examined; parenthesis indicate average severity grade, with
1 = minimal; 2 = mild; 3 = moderate; 4 = severe.
b
Significantly different (p ≤ 0.05) from the control group by Fisher’s exact test.
c
Significantly different (p ≤ 0.01) from the control group by Fisher’s exact test.
In conclusion, the NTP (2007) 3-month study in B6C3F1 mice exposed to sodium
dichromate dihydrate in drinking water identified adverse treatment-related hematological effects
(erythrocytic microcytosis) and histopathological changes to the small intestine (duodenal
epithelial hyperplasia and cellular histiocytic infiltration) and mesenteric lymph nodes (cellular
histiocytic infiltration). Based on histopathological changes (i.e., histiocytic cellular infiltration)
in the duodenum, EPA identified a LOAEL of 3.1 mg hexavalent chromium/kg-day for male and
female mice; in both sexes, a NOAEL was not identified because the effects were observed at the
lowest dose tested. Although a statistically significant decrease in MCV also was observed at
3.1 mg hexavalent chromium/kg-day in males and females, hematological effects (e.g.,
microcytosis) were not considered as the basis of the LOAEL, since decreases in MCV were
small (1–2%) at the lowest dose tested.
Finally, NTP (2007) conducted a comparative study in three strains of mice (B6C3F1,
BALB/c, and am3-C57BL/6) on the effects of exposure to sodium dichromate dihydrate in
drinking water for 3 months. This comparative study was conducted to investigate possible
strain differences in mice based on results of an earlier study reporting hepatotoxicity
(hepatocellular vacuolization) in BALB/c mice fed 32 mg hexavalent chromium/kg-day in the
a
Values are means ± SE; values in parenthesis are percent of control; n = 10 mice/group, with the following
exceptions: in B6C3F1 mice, controls (n = 7), 2.8 mg hexavalent chromium/kg-d group (n = 9), and 5.2 mg
hexavalent chromium/kg-d group (n = 9); in am3-C57BL/6 mice, 8.7 mg hexavalent chromium/kg-d group (n = 9).
b
Significantly different (p ≤ 0.01) from the control group by Dunn’s or Shirley’s test.
Microscopic examinations of gross lesions and masses and of selected tissues in male
B6C3F1, BALB/c, and am3-C57BL/6 mice exposed to sodium dichromate dihydrate in drinking
water for 3 months showed changes to the duodenum, liver, pancreas, and mesenteric lymph
nodes (NTP, 2007); incidence data are summarized in Table 4-11. Dose-related increases in the
incidences of hepatic glycogen depletion and pancreatic secretory depletion were also observed;
NTP (2007) stated that these lesions were likely due to depressed food consumption, which is
frequently observed when water consumption is decreased. The incidence of minimal-to-mild
histiocytic cellular infiltration of mesenteric lymph nodes was increased at 8.7 mg hexavalent
chromium/kg-day in am3-C57BL/6 mice, but not in B6C3F1 or BALB/c mice.
In the duodenum, dose-related increases in the incidences of minimal-to-mild histiocytic
cellular infiltration and epithelial hyperplasia were observed in all strains, with histopathological
changes of the duodenum observed in all exposure groups; severity increased with dose.
Microscopically, lesions were similar to those described above for male and female B6C3F1
mice. The incidences of histiocytic cellular infiltration and epithelial hyperplasia in the
duodenum, however, were smaller in the initial 3-month study in B6C3F1 mice than in this study.
For example, the incidences of histiocytic cellular infiltration in the initial study with male
a
Number of animals with lesion/number of animals examined; parenthesis indicate average severity grade, with
1 = minimal; 2 = mild; 3 = moderate; 4 = severe.
b
Significantly different (p ≤ 0.01) from the control group by Fisher’s exact test.
c
Significantly different (p ≤ 0.01) from the control group by Fisher’s exact test.
a
Values are means ± SE; values in parenthesis are percent of control; n = 10 rats/group, with the following
exceptions: control group on d 4 (n = 9), 0.77 mg hexavalent chromium/kg-d group on d 4 (n = 9), and 2.1 mg
hexavalent chromium/kg-d group in mo 12 (n = 8).
b
Significantly different (p ≤ 0.01) from the control group by Dunn’s or Shirley’s test.
c
Significantly different (p ≤ 0.05) from the control group by Dunn’s or Shirley’s test.
Table 4-13. Serum ALT activity in male F344/N rats exposed to sodium
dichromate dihydrate in drinking water for up to 12 months
a
Values are means ± SE; values in parenthesis are percent of control; n = 10 rats/group, with the following
exception of 0.77 mg hexavalent chromium/kg-d group on d 4 (n = 9). Note: clinical chemistry was not assessed in
female rats.
b
Significantly different (p ≤ 0.01) from the control group by Dunn’s or Shirley’s test.
c
Significantly different (p ≤ 0.05) from the control group by Dunn’s or Shirley’s test.
a
Number of animals with lesion/number of animals examined; parenthesis indicate average severity grade, with
1 = minimal; 2 = mild; 3 = moderate; 4 = severe.
b
Significantly different (p ≤ 0.01) from the control group by the Poly-3 test.
c
Significantly different (p ≤ 0.05) from the control group by the Poly-3 test.
a
Overall rate: number of animals with lesion/number of animals examined; parenthesis are the percent of animals
examined with lesion; brackets are days to first incidence; T: observed at terminal sacrifice. p-Value under
treatment group incidence data indicates statistically significant Poly-3 test for pairwise comparison between
control and exposed group. Statistical analysis using overall rates was only conducted if adjusted rates were not
determined.
b
Adjusted rate not reported.
c
Adjusted rate: Poly-3 estimated neoplasm incidence (expressed as percent of animals with neoplasm) adjusted for
intercurrent mortality. p-Value under control group indicates statistically significant positive Poly-3 trend test.
p-Value under treatment group incidence data indicates statistically significant Poly-3 test for pairwise comparison
between control and exposed groups, using adjusted rates.
a
Number of animals with lesion/number of animals examined.
b
Not statistically significant for positive trend (p > 0.05) by the Poly-3 test.
c
Significantly different from controls by the Poly-3 test (p < 0.05).
In conclusion, from the NTP (2008) 2-year drinking water toxicology and carcinogenicity
study on sodium dichromate dihydrate, EPA identified NOAEL and LOAEL values for
noncancer effects in male rats of 0.21 and 0.77 mg hexavalent chromium/kg-day, respectively,
a
Values are means ± SE; values in parenthesis are percent of control; n = 10 mice/group, with the exception of
1.4 mg hexavalent chromium/kg-d group on mo 12 (n = 9).
b
Significantly different (p ≤ 0.05) from the control group by Dunn’s or Shirley’s test.
c
Significantly different (p ≤ 0.01) from the control group by Dunn’s or Shirley’s test.
Gross and microscopic examinations of “core” study mice exposed to sodium dichromate
dihydrate in drinking water for 2 years showed nonneoplastic lesions of the small intestine, liver,
lymph nodes, and pancreas, and neoplastic lesions of the small intestine (NTP, 2008). Incidence
data for nonneoplastic lesions are summarized in Table 4-18. In the small intestine, statistically
significant increases in the incidences of minimal-to-mild diffuse epithelial hyperplasia of the
duodenum were observed in male and female mice in all treatment groups and of the jejunum in
females at 8.7 mg hexavalent chromium/kg-day, compared with controls. NTP (2008) noted that
diffuse epithelial hyperplasia is consistent with tissue regeneration following epithelial cell
damage. Incidences of minimal-to-mild histiocytic cellular infiltration of the duodenum were
a
Number of animals with lesion/number of animals examined; parenthesis indicate average severity grade, with
1 = minimal; 2 = mild; 3 = moderate; 4 = severe.
b
Significantly different (p ≤ 0.01) from the control group by the Poly-3 test.
c
Significantly different (p ≤ 0.05) from the control group by the Poly-3 test.
In the liver of female mice, dose-dependent increases were observed in the incidences of
histiocytic infiltration at all doses and of chronic inflammation in the 3.1 mg hexavalent
a
Overall rate: number of animals with lesion/number of animals examined; parentheses are the percent of animals
examined with lesion; brackets indicate the days to first incidence; T: observed at terminal sacrifice. p-Value
under treatment group incidence data indicates statistically significant Poly-3 test for pairwise comparison between
control and exposed group. Statistical analysis using overall rates were only conducted if adjusted rates were not
determined.
b
Adjusted rate not reported.
c
Adjusted rate: Poly-3 estimated neoplasm incidence (expressed as % of animals with neoplasm) adjusted for
intercurrent mortality. p-Value under control group indicates statistically significant positive Poly-3 trend test.
p-Value under treatment group incidence data indicates statistically significant Poly-3 test for pairwise comparison
between control and exposed groups, using adjusted rates.
d
Duodenum, jejunum, or ileum.
In conclusion, from the NTP (2008) 2-year toxicology and carcinogenicity study on
sodium dichromate dihydrate, EPA identified a LOAEL for noncancer effects of 0.38 mg
hexavalent chromium/kg-day in male and female B6C3F1 mice; a NOAEL value was not
identified because effects were seen at the lowest dose administered. In males, the LOAEL was
based on increased incidences of histopathological changes to the duodenum (diffuse epithelial
hyperplasia) and mesenteric lymph nodes (histiocytic cellular infiltration); in females, the
LOAEL was based on increased incidences of histopathological changes to the duodenum
(diffuse epithelial hyperplasia), mesenteric lymph nodes (histiocytic cellular infiltration), liver
(histiocytic cellular infiltration), and pancreas (depletion of cytoplasmic zymogen granules).
Although mild microcytic, hypochromic anemia was observed in female mice at ≥0.38 mg
hexavalent chromium/kg-day after 22 days of exposure, the severity of these effects decreased
over time, such that only small changes (<5%) were observed at ≥3.1 mg hexavalent
chromium/kg-day after 12 months of exposure; therefore, hematological effects were not
considered by EPA as the basis for the chronic LOAEL value in female mice. In addition to
noncancer effects, exposure of B6C3F1 mice to sodium dichromate dihydrate in drinking water
for 2 years resulted in significant increases in the incidences of neoplasms of the small intestine
in males and females at doses ≥2.4 and ≥3.1 mg hexavalent chromium/kg-day, respectively.
NTP (2008) concluded that results of this study provide clear evidence of carcinogenic activity
of sodium dichromate dihydrate in male and female B6C3F1 mice based on increased incidences
of neoplasms of the small intestine.
The lower body weights observed in male and female rats and mice exposed to
the two highest exposure concentrations were partly attributed to poor palatability
of the dosed water and consequent reductions in water consumption. However,
several lines of evidence suggest that the animals were not dehydrated. When
water consumption is adjusted for body weight (data not shown), dosed male and
female rats and female mice drank approximately the same quantities of water per
gram of body weight as the controls after the first 20 weeks on study. Male mice
exposed to 257.4 mg/L drank less water per gram of body weight than did the
controls throughout the study. Although mean body weights and water
consumption were reduced in the higher exposure concentration groups, the
average daily doses (mg sodium dichromate dihydrate per kilogram body weight)
were in the same proportions as the drinking water concentrations (mg/L) for
male and female rats and mice. Clinical observations related to dehydration
including loss of skin turgor, dry mucous membranes, retraction of eyes,
hypoactivity, and poor hair coats were not observed in rats or mice in the 2-year
studies of sodium dichromate dihydrate. Abnormalities in hematology and clinical
chemistry parameters that typically indicate dehydration include increases in
hematocrit, urine specific gravity, and serum concentrations of albumin, total
protein, and urea nitrogen. In the current 2-year studies, hematology and clinical
chemistry parameters were measured in male rats on days 4 and 22 and at months
3, 6, and 12. Significant decreases in hematocrit and serum concentrations of
albumin and total protein were observed in males exposed to 516 mg/L. Taken
together, these data suggest that the neoplastic and nonneoplastic effects of
sodium dichromate dihydrate were not associated with dehydration.
Li et al., 2001
Oral exposure of male rats to chromium(VI) oxide for 6 days resulted in adverse
reproductive effects, including reduced epididymal sperm counts and increased abnormal sperm
(Li et al., 2001). Groups of 8–11 male Wistar rats (60 days old) were administered
chromium(VI) oxide by gavage at doses of 0, 10, or 20 mg chromium(VI) oxide/kg-day
(equivalent to 0, 5.2, or 10.4 mg hexavalent chromium/kg-day, respectively) for 6 days. After
6 weeks, rats were sacrificed; testes and epididymis were removed and analyzed for epididymal
sperm count and abnormal sperm; and testes were prepared (fixed in formaldehyde, embedded in
paraffin, sliced, and stained with H&E) for histological evaluations of morphological
abnormalities and diameter of seminiferous tubules. Epididymal sperm counts were significantly
(p < 0.05) decreased by 76 and 80%, and the percentage of abnormal sperm was significantly
(p < 0.01) increased by 143 and 176% in the 5.2 and 10.4 mg hexavalent chromium/kg-day
groups, respectively. Treatment-related histopathological findings included decreased diameter
of seminiferous tubules and disruption of germ cell arrangement within seminiferous tubules in
both treatment groups. Based on decreased sperm counts and histopathological changes to the
testes, 5.2 mg hexavalent chromium/kg-day was identified by EPA as a LOAEL for male rats
NTP, 1996a,b
The NTP conducted studies to investigate the potential effects of dietary hexavalent
chromium as potassium dichromate on male reproductive organs in Sprague-Dawley rats (NTP,
1996b) and BALB/c mice (NTP, 1996a). The NTP studies were designed to expand or replicate
the Zahid et al. (1990) study (described above), and thereby provide data to either refute or
confirm findings of adverse male reproductive effects from hexavalent chromium exposure.
Groups of 24 male and 48 female Sprague-Dawley rats were exposed to diets containing
0, 15, 50, 100, or 400 mg potassium dichromate/kg diet (equivalent to 0, 5.3, 17.6, 35.3, or
141.2 mg hexavalent chromium/kg diet, respectively) daily for 9 weeks followed by an 8-week
recovery period (NTP, 1996b). Based on food consumption measured during the 9-week
treatment period, NTP (1996a, b) calculated average daily doses of 0, 1, 3, 6, or 24 mg potassium
dichromate/kg-day (equivalent to 0, 0.35, 1.1, 2.1, or 8.5 mg hexavalent chromium/kg-day) in
males and 0, 1, 3, 7, or 28 mg hexavalent chromium/kg-day (equivalent to 0, 0.35, 1.1, 2.5, or
9.9 mg hexavalent chromium/kg-day) in females for the 0, 15, 50, 100, or 400 mg potassium
dichromate/kg diet groups, respectively. Animals were examined twice daily for mortality and
NTP, 1997
The potential reproductive toxicity of dietary potassium dichromate was evaluated in
BALB/c mice in a continuous breeding study (NTP, 1997). Groups of 20 male and female pairs
(F0) were exposed to dietary potassium dichromate at 0, 100, 200, and 400 mg potassium
dichromate/kg diet (equivalent to 0, 17.6, 35.3, or 141.2 mg hexavalent chromium/kg diet,
respectively) for 13 weeks (1 week prior to and 12 weeks during cohabitation). During exposure
of the F0 generation, animals were examined daily for mortality and clinical signs of toxicity;
body weights and food consumption were measured periodically (4–5 times). Litters produced
during the cohabitation period were evaluated (i.e., total pups, live and dead pups, and sex),
weighed on postnatal day (PND) 1, and euthanized with no additional assessments; pregnancy
index (number of litters/breeding pair) was also determined. After the cohabitation period,
F0 breeding pairs were separated and continued on study diets; litters born during the post-
+ = positive results; (D) = study in D. melanogaster; (M) = study in laboratory mammal; ND = no data identified for this review
Results
Without
Endpoint Chemical form Test system activation With activation Reference
Reverse mutations Ammonium chromate S. typhimurium TA97, TA1538, NS De Flora et al., 1984
+
TA98, TA100
Reverse mutations Ammonium chromate S. typhimurium TA1537 ± NS De Flora et al., 1984
Reverse mutations Ammonium chromate S. typhimurium TA1535 – – De Flora et al., 1984
Reverse mutations Calcium chromate S. typhimurium TA97, TA1538, NS De Flora et al., 1984
+
TA98, TA100
Reverse mutations Calcium chromate S. typhimurium TA1537 ± NS De Flora et al., 1984
Reverse mutations Calcium chromate S. typhimurium TA1535 – – De Flora et al., 1984
Reverse mutations Calcium chromate S. typhimurium TA98 – ± Dunkel et al., 1984
Reverse mutations Calcium chromate S. typhimurium TA100, TA1535, – Dunkel et al., 1984
–
TA1537, TA1538
Reverse mutations Calcium chromate E. coli WP2 uvrA – ± Dunkel et al., 1984
Reverse mutations Chromic acid S. typhimurium TA102, TA2638 + ND Watanabe et al., 1998
Reverse mutations Chromic acid E. coli, WP2/pKM101, WP2 ND Watanabe et al., 1998
+
uvrA/pKM101
Reverse mutations Chromium trioxide S. typhimurium TA97, TA1538, NS De Flora et al., 1984
+
TA98, TA100
Reverse mutations Chromium trioxide S. typhimurium TA1537 ± NS De Flora et al., 1984
Reverse mutations Chromium trioxide S. typhimurium TA1535 – – De Flora et al., 1984
Reverse mutations Potassium chromate S. typhimurium TA102 + ND Marzin and Phi, 1985
Reverse mutations Potassium chromate S. typhimurium TA97, TA1538, NS De Flora et al., 1984
+
TA98, TA100
Reverse mutations Potassium chromate S. typhimurium TA1537 ± NS De Flora et al., 1984
Reverse mutations Potassium chromate S. typhimurium TA1535 – – De Flora et al., 1984
Reverse mutations Potassium chromate E. coli Hs30R + ND Nakamuro et al., 1978
Reverse mutations Potassium chromate E. coli WP2 hcr- try-, B/rWP2 + (WP2 hcr) ND Kanematsu et al., 1980
Reverse mutations Potassium chromate E. coli WP2(try-) + ND Venitt and Levy, 1974
Reverse mutations Potassium chromate E. coli WP2uvrA, CM571 + ND Seo and Lee, 1993
Results
Without
Endpoint Chemical form Test system activation With activation Reference
Reverse mutations Potassium dichromate S. typhimurium TA97, TA98, + Zeiger et al., 1992
+
TA100, TA1535, TA1537
Reverse mutations Potassium dichromate S. typhimurium TA102 + ND Marzin and Phi, 1985
Reverse mutations Potassium dichromate S. typhimurium TA100 + + Venier et al., 1982
Reverse mutations Potassium dichromate E. coli WP2 hcr- try-, B/rWP2 + (WP2 hcr) ND Kanematsu et al., 1980
Reverse mutations Potassium dichromate E. coli Hs30R + ND Nakamuro et al., 1978
Reverse mutations Potassium dichromate E. coli WP2, WP2uvrA, CM571 + ND Nishioka, 1975
Reverse mutations Potassium dichromate E. coli WP2uvrA, CM571 + ND Seo and Lee, 1993
Reverse mutations Potassium dichromate S. cerevisiae D7 + ND Singh, 1983
Reverse mutations Potassium dichromate S. typhimurium TA98 ± – Venier et al., 1982
Reverse mutations Potassium dichromate S. typhimurium TA1538 – – Venier et al., 1982
Reverse mutations Sodium chromate E. coli WP2(try-) + ND Venitt and Levy, 1974
Reverse mutations Sodium dichromate S. typhimurium TA102, TA2638 + ND Watanabe et al., 1998
Reverse mutations Sodium dichromate S. typhimurium TA102 + Bennicelli et al., 1983
+
Results
Without
Endpoint Chemical form Test system activation With activation Reference
Induction of SOS response Potassium chromate E. coli AB1157, GC2375, ND Llagostera et al., 1986
+
UA4202, PQ30
Induction of SOS response Potassium dichromate E. coli AB1157, GC2375, ND Llagostera et al., 1986
+
UA4202, PQ30
Induction of SOS response potassium dichromate E. coli PQ37, PQ35 + – Olivier and Marzin, 1987
Mutations Ammonium chromate S. typhimurium TA1978 (rec+), + ND Gentile et al., 1981
TA1538 (rec - )
Mutations Ammonium chromate B. subtilis + ND Gentile et al., 1981
Mutations Calcium chromate S. typhimurium TA97, TA98, – Brams et al., 1987
–
TA100
Mutations Chromic acid S. typhimurium TA1978 (rec+), + ND Gentile et al., 1981
TA1538 (rec - )
Mutations Chromic acid B. subtilis + ND Gentile et al., 1981
Mutations Potassium chromate S. typhimurium TA98, TA100, ND Arlauskas et al., 1985
+
TA1537
Mutations Potassium chromate S. typhimurium TA100 + ND Arlauskas et al., 1985
Mutations Potassium chromate E. coli WP2 uvrA pKm 101 + ND Arlauskas et al., 1985
Mutations Potassium chromate B. subtilis H17 + ND Nishioka, 1975
Mutations Potassium chromate S. typhimurium TA1535, TA1538 – ND Arlauskas et al., 1985
Mutations Potassium dichromate S. typhimurium TA 1535 pSK1002 + + Yamamoto et al., 2002
Mutations Potassium dichromate S. typhimurium TA100, TA1025, ND Le Curieux et al., 1993
+
TA98
Mutations Potassium dichromate S. typhimurium TA1978 (rec+), + ND Gentile et al., 1981
TA1538 (rec-)
Mutations Potassium dichromate E. coli WP2uvrA + ND Venier et al., 1987
Mutations Potassium dichromate B. subtilis + ND Gentile et al., 1981
Mutations Sodium dichromate B. subtilis + ND Gentile et al., 1981
Mutations Potassium dichromate B. subtilis NIG45, NIG17 + ND Matsui, 1980
Mutations Potassium dichromate B. subtilis H17 + ND Nishioka, 1975
Results
Without
Endpoint Chemical form Test system activation With activation Reference
Mutations Sodium dichromate S. typhimurium TA1978 (rec+), + ND Gentile et al., 1981
TA1538 (rec - )
Frame shift mutations Calcium chromate S. typhimurium TA98, TA1537 + ND Haworth et al., 1983
Frame shift mutation Potassium chromate S. typhimurium TA1537 + ND La Velle, 1986
Frame shift mutation Potassium chromate E. coli 343/358, /415, /435, /477 + ND La Velle, 1986
Frame shift mutations Potassium dichromate S. typhimurium TA97a, TA98 + + Tagliari et al., 2004
Frame shift mutations Potassium dichromate S. typhimurium TA100, TA1537, – ND Kanematsu et al., 1980
TA1538
Frame shift mutations Sodium dichromate S. typhimurium TA97, TA1978 + ND Bennicelli et al., 1983
Frame shift mutations Sodium dichromate S. typhimurium TA1537, TA1538 – ND Bennicelli et al., 1983
Frame shift mutations, base Calcium chromate S. typhimurium TA1537, TA98, + + Petrilli and De Flora, 1977
pair substitutions TA100
Frame shift mutations, base Calcium chromate S. typhimurium TA1535 – – Petrilli and De Flora, 1977
pair substitutions
Frame shift mutations, base Chromic acid S. typhimurium TA1537, TA98, + + Petrilli and De Flora, 1977
pair substitutions TA100
Frame shift mutations, base Chromic acid S. typhimurium TA1535 – – Petrilli and De Flora, 1977
pair substitutions
Frame shift mutations, base Potassium chromate S. typhimurium TA1537, TA98, + + Petrilli and De Flora, 1977
pair substitutions TA100
Frame shift mutations, base Potassium chromate S. typhimurium TA1535 – – Petrilli and De Flora, 1977
pair substitutions
Frame shift mutations, base Potassium dichromate S. typhimurium TA98 TA100, + + Bianchi et al., 1983
substitutions TA1535, TA1538
Frame shift mutations, base Sodium dichromate S. typhimurium TA1537, TA98, + + Petrilli and De Flora, 1977
pair substitutions TA100
Frame shift mutations, base Sodium dichromate S. typhimurium TA1535 – – Petrilli and De Flora, 1977
pair substitutions
Base pair substitutions Ammonium chromate S. typhimurium TA1537, TA1538, NS De Flora et al., 1984; De Flora,
±
TA98, TA100 1981
Base pair substitutions Ammonium chromate S. typhimurium TA1535 – – De Flora, 1981
Results
Without
Endpoint Chemical form Test system activation With activation Reference
Base pair substitutions Calcium chromate S. typhimurium TA100, TA1535 + ND Haworth et al., 1983
Base pair substitutions Calcium chromate S. typhimurium TA1537, TA1538, NS De Flora et al., 1984; De Flora,
±
TA98, TA100 1981
Base pair substitutions Calcium chromate S. typhimurium TA1535 – – De Flora, 1981
Base pair substitutions Chromic acid S. typhimurium TA1537, TA1538, ± NS De Flora et al., 1984; De Flora,
TA98, TA100 1981
Base pair substitutions Chromic acid S. typhimurium TA1537, TA1538, ± NS De Flora et al., 1984; De Flora,
TA98, TA100 1981
Base pair substitutions Potassium chromate S. typhimurium TA1537, TA1538, ± NS De Flora et al., 1984; De Flora,
TA98, TA100 1981
Base pair substitutions Potassium chromate S. typhimurium TA1535 – – De Flora, 1981
Base pair substitutions Potassium dichromate S. typhimurium TA100, TA102 + + Tagliari et al., 2004
Base pair substitutions Potassium dichromate S. typhimurium TA100, TA1535 – ND Kanematsu et al., 1980
Base pair substitutions Sodium dichromate S. typhimurium TA100, TA102, + ND Bennicelli et al., 1983
TA92
Base pair substitutions Sodium dichromate S. typhimurium TA1537, TA1538, ± NS De Flora et al., 1984; De Flora,
TA98, TA100 1981
Base pair substitutions Sodium dichromate S. typhimurium TA1535 – – De Flora, 1981
Base pair substitutions Sodium dichromate S. typhimurium TA1535 – ND Bennicelli et al., 1983
Reverse mutation, Potassium dichromate S. cerevisiae D7 + ND Kharab and Singh, 1985
induction of gene
conversion
Forward mutation Potassium dichromate Schizosaccharomyces pombe ± ND Bonatti et al., 1976
972, h-
Mitotic cross-over Chromic acid S. cerevisiae D7 + ND Fukunaga et al., 1982
Mitotic gene conversions Chromic acid S. cerevisiae D7 + ND Singh, 1983; Fukunaga et al.,
1982
Mitotic gene conversion, Sodium chromate S. cerevisiae D7 + ND Bronzetti and Galli, 1989
point reverse mutation
Results
Without
Endpoint Chemical form Test system activation With activation Reference
Mitotic gene conversion at Chromic acid S. cerevisiae D7 + ND Vashishat and Vasudeva, 1987
trp5 locus, reverse mutation
of ilvl-92 allele
Mitotic gene conversion at Potassium dichromate S. cerevisiae D7 + ND Vashishat and Vasudeva, 1987
trp5 locus, reverse mutation
of ilvl-92 allele
Induction of disomic and Potassium dichromate S. cerevisiae D1S13 + ND Sora et al., 1986
diploid spores
umu gene expression Potassium dichromate S. typhimurium TA1535 ± – Nakamura et al., 1987
DNA damage Potassium dichromate E. coli PQ37 + ND Le Curieux et al., 1993
DNA-protein crosslinks Potassium chromate E. coli DNA – ND Fornace et al., 1981
DNA polymerase arrest Sodium dichromate PSV2neo-based plasmid DNA – + Bridgewater et al., 1998, 1994
Results
Without With
Endpoint Chemical form Test system activation activation Reference
DNA damage Potassium Human lymphocytes + ND Blasiak and Kowalik, 2000
dichromate
DNA damage Potassium Human gastric mucosa + ND Trzeciak et al., 2000
dichromate
DNA damage Potassium Human peripheral blood lymphocytes + ND Trzeciak et al., 2000
dichromate
DNA damage Potassium Human lymphocytes, human lymphoblastoid TK-6 + ND Cemeli et al., 2003
dichromate cells
DNA damage Sodium Human gastric mucosa cells, rat gastric mucosa cells + ND Pool-Zobel et al., 1994
dichromate
DNA adducts, [32P] Potassium Calf thymus DNA – – Adams et al., 1996
postlabeling chromate (+1 mM
H2O2)
DNA fragmentation Potassium Human bronchial epithelial cells + ND Fornace et al., 1981
chromate
DNA fragmentation Potassium Human embryonic lung fibroblasts (IMR-90) + ND Fornace et al., 1981
chromate
DNA fragmentation Potassium Mouse L1210 leukemia cells + ND Fornace et al., 1981
chromate
DNA fragmentation sodium Chinese hamster ovary cells + ND Blankenship et al., 1997
chromate
DNA strand breaks Potassium Vero kidney fibroblasts, Pam 212 keratinocytes + ND Flores and Perez, 1999
dichromate
DNA strand breaks Sodium Rat primary lymphocytes + ND Gealy et al., 2007
dichromate
DNA strand breaks Sodium Rat hepatocytes + ND Gao et al., 1993
dichromate
DNA strand breaks Potassium Human lymphocytes + ND Depault et al., 2006
chromate
Results
Without With
Endpoint Chemical form Test system activation activation Reference
DNA strand breaks Potassium Human fibroblast + ND Fornace, 1982
chromate
DNA strand breaks Potassium Bacteriophage λ DNA + + Adams et al., 1996
chromate (+1mM
H2O2)
DNA strand breaks Sodium Rat primary lymphocytes + ND Elia et al., 1994
dichromate
DNA strand breaks Potassium Human lymphocytes, human gastric mucosa cells + ND Blasiak et al., 1999
dichromate
DNA-DNA crosslinks Sodium Human lung fibroblasts + ND Xu et al., 1996
chromate
DNA-protein crosslinks Potassium Human embryonic lung fibroblasts (IMR-90) + ND Fornace et al., 1981
chromate
DNA-protein crosslinks Potassium Human fibroblast + ND Fornace, 1982
chromate
DNA-protein crosslinks Potassium Chinese hamster cells (V79-UL) + ND Merk et al., 2000
chromate
DNA-protein crosslinks Potassium Mouse L1210 leukemia cells + ND Fornace et al., 1981
chromate
DNA-protein crosslinks Sodium Human HL-60 cells + ND Capellmann et al., 1995
chromate
Induced DNA methylation Potassium Chinese hamster V79 cells (hpr-1gpt+ transgenic cell + (T) ND Klein et al., 2002
chromate line G12)
Unscheduled DNA synthesis Sodium Rat hepatocytes + (T) ND Gao et al., 1993
dichromate
DNA synthesis inhibition Potassium HeLa S3 cells + ND Heil and Reifferscheid, 1992
chromate
DNA synthesis inhibition Potassium Mouse L cells + ND Nishio and Uyeki, 1985
dichromate
Results
Without With
Endpoint Chemical form Test system activation activation Reference
DNA polymerase arrest Sodium Human lung fibroblasts + ND Xu et al., 1996
chromate
Mutations at the HGPRT Potassium Chinese hamster ovary cells (AT3-2) + ND Paschin et al., 1983
locus dichromate
Mutations at the HGPRT Potassium Chinese hamster cells (V79) + ND Paschin et al., 1983
locus dichromate
Forward mutation Calcium Mouse lymphoma cells (L5178Y tk+/tk-) + + McGregor et al., 1987
chromate
Forward mutation Calcium Mouse lymphoma cells (L5178Y tk+) + + Mitchell et al., 1988
chromate
Forward mutation Calcium Mouse lymphoma cells (L5178Y tk+) + + Myhr and Caspary, 1988
chromate
Forward mutation Calcium Mouse lymphoma cells (L5178Y tk+) + + Oberly et al., 1982
chromate
Morphological Calcium Syrian hamster embryo cells + ND Elias et al., 1991
transformation chromate
Morphological Sodium Syrian hamster cells + ND DiPaolo and Casto, 1979
transformation chromate
dihydrate
Cell transformation Calcium Balb/3T3, Syrian hamster embryo, R-MuLV-RE cells + ND Dunkel et al., 1981
chromate
Transformations Potassium Rat liver epithelial cells + ND Briggs and Briggs, 1988
chromate
Chromosomal damage Calcium Chinese hamster ovary cells + ND Levis and Majone, 1979
chromate
Chromosomal damage Chromic acid Chinese hamster ovary cells + ND Levis and Majone, 1979
Chromosomal damage Potassium Chinese hamster ovary cells + ND Levis and Majone, 1979
chromate
Chromosomal damage Potassium Chinese hamster ovary cells + ND Seoane and Dulout, 1999
dichromate
Results
Without With
Endpoint Chemical form Test system activation activation Reference
Chromosomal damage Potassium Chinese hamster ovary cells + ND Levis and Majone, 1979
dichromate
Chromosomal damage Potassium Chinese hamster ovary cells + ND Majone and Levis, 1979
dichromate
Chromosomal damage Sodium Chinese hamster ovary cells + ND Levis and Majone, 1979
chromate
Chromosomal damage Sodium Chinese hamster ovary cells + ND Levis and Majone, 1979
dichromate
Chromosomal damage Sodium Chinese hamster ovary cells + ND Majone and Levis, 1979
dichromate
Chromosome aberrations Calcium Chinese hamster lung DON cells + ND Koshi and Iwasaki, 1983; Koshi,
chromate 1979
Chromosome aberrations Calcium Chinese hamster ovary cells (C3H10T1/2) + ND Sen et al., 1987
chromate
Chromosome aberrations Chromic acid BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
Chromosome aberrations Chromic acid Mouse mammary FM3A carcinoma cells + ND Umeda and Nishmura, 1979
Chromosome aberrations Chromic acid Chinese hamster lung DON cells + ND Koshi and Iwasaki, 1983; Koshi,
1979
Chromosome aberrations Potassium Human fibroblasts + ND MacRae et al., 1979
chromate
Chromosome aberrations Potassium Chinese hamster lung DON cells + ND Koshi and Iwasaki, 1983; Koshi,
chromate 1979
Chromosome aberrations Potassium Chinese hamster ovary cells + ND MacRae et al., 1979
chromate
Chromosome aberrations Potassium BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
chromate
Chromosome aberrations Potassium Mouse mammary FM3A carcinoma cells + ND Umeda and Nishmura, 1979
chromate
Chromosome aberrations Potassium Human fibroblasts + ND MacRae et al., 1979
dichromate
Results
Without With
Endpoint Chemical form Test system activation activation Reference
Chromosome aberrations Potassium Chinese hamster ovary cells + ND MacRae et al., 1979
dichromate
Chromosome aberrations Potassium BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
dichromate
Chromosome aberrations Potassium Mouse mammary FM3A carcinoma cells + ND Umeda and Nishmura, 1979
dichromate
Chromosome aberrations Sodium Human primary bronchial fibroblasts + ND Wise et al., 2004, 2002
chromate
Chromosome aberrations Sodium Human bronchial fibroblasts (WTHBF-6 cells) + ND Holmes et al., 2006
chromate
Chromosome aberrations Sodium Human bronchial epithelial cells (BEP2D cells) + ND Wise et al., 2006a
chromate
Chromosome aberrations Sodium Chinese hamster ovary cells + ND Blankenship et al., 1997
chromate
Chromosome aberrations Sodium Chinese hamster ovary cells (AA8 (parental), EM9 + ND Grlickova-Duzevik, 2006
chromate (XRCC1 mutant), and H9T3
Chromosome aberrations Sodium BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
dichromate
Chromosome and chromatid Potassium Human lymphocytes + ND Imreh and Radulescu, 1982
aberrations dichromate
Sister chromatid exchanges Calcium Human lymphocytes + ND Gomez-Arroyo et al., 1981
chromate
Sister chromatid exchanges Calcium Chinese hamster lung DON cells + ND Koshi and Iwasaki, 1983; Koshi,
chromate 1979
Sister chromatid exchanges Calcium Chinese hamster ovary cells + ND Levis and Majone, 1979
chromate
Sister chromatid exchanges Chromic acid Chinese hamster ovary cells + ND Levis and Majone, 1979
Sister chromatid exchanges Chromic acid Chinese hamster cells DON + ND Ohno et al., 1982
Sister chromatid exchanges Chromic acid Chinese hamster lung DON cells + ND Koshi and Iwasaki, 1983; Koshi,
1979
Results
Without With
Endpoint Chemical form Test system activation activation Reference
Sister chromatid exchanges Chromic acid BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
Sister chromatid exchanges Potassium Human fibroblasts + ND MacRae et al., 1979
chromate
Sister chromatid exchanges Potassium Chinese hamster lung DON cells + ND Koshi and Iwasaki, 1983; Koshi,
chromate 1979
Sister chromatid exchanges Potassium Chinese hamster ovary cells + ND Levis and Majone, 1979
chromate
Sister chromatid exchanges Potassium Chinese hamster ovary cells + ND MacRae et al., 1979
chromate
Sister chromatid exchanges Potassium Chinese hamster cells DON + ND Ohno et al., 1982
chromate
Sister chromatid exchanges Potassium BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
chromate
Sister chromatid exchanges Potassium Human lymphocytes + ND Gomez-Arroyo et al., 1981
dichromate
Sister chromatid exchanges Potassium Human lymphocytes + ND Imreh and Radulescu, 1982
dichromate
Sister chromatid exchanges Potassium Human fibroblasts + ND MacRae et al., 1979
dichromate
Sister chromatid exchanges Potassium Chinese hamster ovary cells + ND Levis and Majone, 1981
dichromate
Sister chromatid exchanges Potassium Chinese hamster ovary cells + ND Levis and Majone, 1979
dichromate
Sister chromatid exchanges Potassium Chinese hamster ovary cells + ND Majone and Levis, 1979
dichromate
Sister chromatid exchanges Potassium Chinese hamster ovary cells + ND MacRae et al., 1979
dichromate
Sister chromatid exchanges Potassium Chinese hamster cells DON + ND Ohno et al., 1982
dichromate
Results
Without With
Endpoint Chemical form Test system activation activation Reference
Sister chromatid exchanges Potassium BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
dichromate
Sister chromatid exchanges Potassium Mouse blastocysts + ND Iijima et al., 1983
dichromate
Sister chromatid exchanges Sodium Chinese hamster ovary cells + ND Levis and Majone, 1979
chromate
Sister chromatid exchanges Sodium BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
chromate
Sister chromatid exchanges Sodium Chinese hamster ovary cells + ND Levis and Majone, 1979
dichromate
Sister chromatid exchanges Sodium Chinese hamster ovary cells + ND Majone and Levis, 1979
dichromate
Sister chromatid exchanges Sodium BHK and Chinese hamster ovary cells + ND Bianchi et al., 1980
dichromate
Disruption of mitosis Sodium Human bronchial fibroblasts (WTHBF-6 cells) + ND Wise et al., 2006b
chromate
+ 43.6 mg
Cr(VI)/L
(am3-
C57BL/6)
B6C3F1 mouse Sodium dichromate dihydrate was administered in drinking – 349 mg
(10/sex/group) water for 3 mo at concentrations 0, 62.5, 125, 250, 500, or Cr(VI)/L
peripheral red 1,000 mg/L (0, 21.8, 43.6, 87.2, 174.5, or 349 mg hexavalent
blood cells chromium/L). NTP estimated daily doses at 0, 3.1, 5.2, 9.1,
15.7, or 27.9 mg hexavalent chromium/kg.
Oxidative DNA Female SKH-1 Sodium dichromate dihydrate was administered in drinking – 20 mg No measure of cytotoxicity; De Flora et al.,
damage, DNA hairless mouse water at concentrations of 0, 5, and 20 mg hexavalent Cr(VI)/L no weight changes in mice. 2008
protein forestomach, chromium/L for 9 mo. Using reference values for body
crosslinks glandular stomach, weight (0.0353 kg) and daily drinking water intake (0.0085
and duodenum L/d) for female B6C3F1 mice (U.S. EPA, 1988), doses of 1.20
cells and 4.82 mg hexavalent chromium/kg-d for the 5 and 20 mg
hexavalent chromium/L groups, respectively, were estimated.
DNA-protein crosslinks and oxidative DNA damage (8-oxo-
2’deoxyguanosine) were measured in forestomach, glandular
stomach, and duodenum cells.
Unscheduled Fischer 344 rat Potassium dichromate was administered at concentrations of – 20 mg No measure of cytotoxicity. Mirsalis et al.,
DNA synthesis hepatocytes 0, 1, 5, or 20 mg hexavalent chromium/L in drinking water ad Cr(VI)/L RDS not determined. 1996
libitum for 48 hours, while a second group was administered
single gavage doses (20 mL/kg) at the same concentrations.
Hepatocytes were collected from the rat livers and analyzed in
the in vivo-in vitro hepatocyte DNA repair assay.
Oral exposures (drinking water): rats
DNA-protein Male Fischer 344 Potassium chromate was administered in drinking water for 3 + 100 mg No cytotoxicity detected. Coogan et al.,
crosslinks rat liver and and 6 wks at 100 and 200 ppm hexavalent chromium. Liver Cr(VI)/L 1991
splenic and splenic lymphocytes were examined for DNA-protein
lymphocytes crosslinks; crosslinks were detected in liver, not lymphocytes.
Exposure type
Endpoint (chemical form) Cell type Test conditions Results Exposure levela Reference
DNA strand Occupational, Human Nineteen chromium plating workers in Italy (mean employment of + NS Gambelunghe
breaks chromium plating peripheral 6.3 yrs) and two groups of control subjects (18 hospital workers et al., 2003
(chromic acid) lymphocytes and 20 university personnel) gave pre- and postshift urine samples
and blood samples for analysis in the comet assay. Duration of
employment ranged from 4 mo to 14 yrs with a mean duration of
6.3 yrs. Mean chromium concentrations in urine were determined
to be 5.29 µg/g creatinine (preshift) and 7.31 µg/g creatinine
(postshift). Mean erythrocyte and lymphocyte concentrations in
the exposed workers were 4.94 µg/L and 50.3 µg/1012cells,
respectively. Air concentrations of chromium were not reported.
DNA strand Occupational, Human Urine and blood samples were taken from 10 exposed workers and – 0.001–0.055 mg Gao et al., 1994
breaks, production of peripheral 10 nonexposed workers at the end of a workweek at a bichromate hexavalent
hydroxylation of dichromate lymphocytes production plant in England. The mean duration of exposure was chromium/m3
deoxyguanosine (included exposure 15 yrs. Chromium concentrations in the factory ranged from (measured
to chromic acid, 0.001 to 0.055 mg hexavalent chromium/m3 (obtained from exposure range)
potassium personal and area samplers). Mean chromium concentrations in
dichromate and urine (5.97 µg/g creatinine), whole blood (5.5 µg/L), plasma
sodium (2.8 µg/L), and lymphocytes (1.01 µg/1010 cells) of exposed
dichromate) workers were significantly higher than in nonexposed workers.
DNA-protein Experimental oral Human Four adult volunteers ingested a single bolus dose of 5,000 µg – 71 µg hexavalent Kuykendall et
crosslinks exposure peripheral hexavalent chromium as potassium dichromate (approximately chromium/kg al., 1996
(potassium lymphocytes equivalent to 71 µg hexavalent chromium/kg, assuming a body
dichromate) weight of 70 kg). Blood samples were collected at 0, 60, 120,
180, and 240 mins after ingestion. Preingestion background
DNA-protein crosslink levels for each individual served as the
controls.
Chromosome Occupational, Human Blood from seven chromium electroplating workers at a Chinese + 8.1 µg Deng et al.,
aberrations, sister chromium peripheral electroplating facility (mean employment period of 12.8 yrs) and chromium/mm3b 1988
chromatid electroplating lymphocytes 10 control subjects were analyzed. Air samples from the
exchanges (chemical not electroplating room were collected, along with stool and hair
specified) samples to determine exposure. The mean chromium (total) air
concentration (by random air collection) was 8.1 µg/mm3, the
mean chromium concentration in stool samples was 8.5 µg/g stool,
and the mean chromium concentration in hair was 35.68 µg/g.
The valence of chromium that workers were exposed to was
unspecified.
Exposure type
Endpoint (chemical form) Cell type Test conditions Results Exposure levela Reference
Sister chromatid Occupational, Human whole Thirty-five chromium electroplating factory workers employed at + 5.99 mg Wu et al., 2001
exchanges chromium blood cells three electroplating plants in Tawain and 35 control subjects gave hexavalent
electroplating blood samples to analyze the frequency of sister chromatid chromium/m3
(chemical not exchange. Exposure duration ranged from 2 to 14 yrs with a mean
specified) of 6.5 yrs. Mean chromium exposure (determined by personal
monitoring samplers) was 5.99 mg hexavalent chromium/m3. The
mean urinary chromium concentration of the exposed workers was
3.67 µg/g creatinine.
Chromosomal Occupational, Human Thirty-eight male chromium plating factory workers in Italy were + NS Sarto et al.,
aberrations, sister chromium plating peripheral examined for urinary concentrations of chromium and 1982
chromatid (chromic acid) lymphocytes chromosomal aberrations and sister chromatid exchanges.
exchanges Chromium exposure levels were not reported. There were
35 unexposed control individuals.
Sister chromatid Occupational, Human The frequency of sister chromatid exchanges was determined in + NS Stella et al.,
exchanges chromium plating peripheral lymphocytes from 12 chromium plating workers in Italy and 1982
(chromic acid lymphocytes 10 control subjects. Exposure durations ranged from 0.5 to 18 yrs
fumes) (mean exposure duration was not reported). Hexavalent
chromium exposure levels and blood concentrations were not
reported.
Sister chromatid Occupational, Human Thirty-five chromium electroplating factory workers in Taiwan + NS Wu et al., 2000
exchanges chromium peripheral and 35 control subjects (matched for age and gender) gave blood
electroplating lymphocytes samples to determine sister chromatid exchange frequency. The
(chemical not mean duration of employment was 6.5 yrs. Exposure
specified) concentrations were not reported.
Chromosomal Occupational, Human Blood samples and buccal mucosal cells from 15 Bulgarian – Results reported Benova et al.,
aberrations, sister chromium plating peripheral chromium platers occupationally exposed were taken; exposure for combined 2002
chromatid (chemical not lymphocytes was estimated with personal air samplers and in urine samples. groups (0.0075 and
exchanges specified) and buccal Control subjects were matched with exposed individuals. 0.0249 mg
mucosal cells Duration of exposure ranged from 2 to >20 yrs; mean duration of chromium/m3)
exposure was not reported. Mean air concentration of total
chromium was 0.0075 mg chromium/m3 in the low-exposure
group and 0.0249 mg chromium/m3 in the high-exposure group
(number of workers in each exposure group was not reported).
Mean concentrations of chromium in urine were 18.63 µg/L (low)
and 104.22 µg/L (high)
Exposure type
Endpoint (chemical form) Cell type Test conditions Results Exposure levela Reference
Sister chromatid Occupational, Human Venous blood and urine sample were collected from 12 male – NS Nagaya et al.,
exchanges chromium plating peripheral chromium platers in Japan over a 5-yr period. No control subjects 1991
(chemical not lymphocytes were used in this study. Employment duration ranged from 6.6 to
specified) 25.1 yrs, with mean employment duration of 15.5 yrs. Exposure
concentrations were not reported. Urinary chromium
concentrations ranged from 1.2 to 57.0 µg/g with a mean urinary
chromium concentration of 17.9 µg/g creatinine. Sister chromatid
exchange frequency in lymphocytes was determined in blood-
urine paired samples.
Sister chromatid Occupational, Human Venous blood and urine sample were collected from 24 male – NS Nagaya, 1986
exchanges chromium plating peripheral chromium platers in Japan and 24 control subjects. Duration of
(chemical not lymphocytes employment ranged from 0.5 to 30.5 yrs with a mean employment
specified) of 11.6 yrs. Exposure concentrations were not reported. The
mean concentration of chromium in the urine was 13.1 µg/L.
Micronuclei Occupational, Human Forty electroplating workers in Bulgaria and 18 control subjects + 0.043 and Vaglenov et al.,
chromium peripheral gave blood samples to analyze for the frequency of micronuclei. 0.083 mg 1999
electroplating lymphocytes The workers were split into two groups based on levels of chromium/m3
(chemical not exposure. Mean air chromium (total) concentrations were 43 and
specified) 83 µg/m3 in the low- and high-exposure groups, respectively.
Duration of employment ranged from 4 to 25 yrs with mean
durations of 10.44 and 11.63 yrs in the low- and high-exposure
groups, respectively. Mean chromium concentrations in
erythrocytes and urine of the low-exposure group were 4.31 and
3.97 µg/L, respectively. The mean chromium concentrations in
erythrocytes and urine of the high-exposure group were 8.4 and
5.0 µg/L, respectively.
Micronuclei Occupational, Human Blood samples and buccal mucosal cells from 15 Bulgarian + Positive results Benova et al.,
chromium plating peripheral chromium platers occupationally exposed were taken. Exposure reported for 2002
(chemical not lymphocytes was estimated with personal air samplers and in urine samples. combined groups
specified) and buccal Control subjects were matched with exposed individuals. (0.0075 and 0.0249
mucosal cells Duration of exposure ranged from 2 to >20 yrs; mean duration of mg chromium/m3)
exposure was not reported. Mean air concentration of total
chromium was 0.0075 mg chromium/m3 in the low-exposure
group and 0.0249 mg chromium/m3 in the high-exposure group.
Mean concentrations of chromium in urine were 18.63 (low) and
104.22 µg/L (high).
Exposure type
Endpoint (chemical form) Cell type Test conditions Results Exposure levela Reference
Micronuclei Occupational, Human Sixteen exposed Italian electroplating factory workers and – NS Sarto et al.,
chromium plating buccal and 27 unexposed control subjects gave samples of exfoliated buccal 1990
(chromic acid) nasal cells and nasal swabs. Duration of exposure ranged from 0.5 to 23 yrs
with a mean duration of 8 yrs. Urine samples were collected at the
end of work days to determine chromium exposure. Urinary
chromium concentrations ranged from 2.5 to 88 µg/g creatinine;
the mean urinary chromium concentration was not reported.
Chromium levels in air were not determined.
a
All exposure levels associated with positive results, highest exposure level for negative results.
b
The exposure level of 8.1 µg chromium/mm3 is as reported by Deng et al. (1988); however, this appears to be a reporting error, as this concentration is equivalent to
8,100,000 mg chromium/m3.
DNA
O
OH
L L
O
H
L Cr(III) O
O H N NH
7
O P O L
O N
O N NH2
OH
O P O
O DNA
M: 0 or 1.4–
2.18 mg/kg-d
via drinking
water
Chronic studies
F344/N rat F, M F: 0.24, 0.94, 2 yrs F: ND 0.24 F: Increased incidence of chronic inflammation of the NTP, 2008
2.4, or 7.0 mg/ M: 0.21 0.77 liver.
kg-d via
drinking water M: Increased incidences of nonneoplastic
histopathological changes to the liver (basophilic foci),
M: 0.21, 0.77, duodenum (histiocytic cellular infiltrate), and mesenteric
2.1, or lymph nodes (histiocytic cellular infiltrate and
5.9 mg/kg-d via hemorrhage).
drinking water
B6C3F1 mouse F, M F: 0.38, 1.4, 2 yrs F: ND 0.38 F: Increased incidences of histopathological changes to NTP, 2008
3.1, or M: ND 0.38 the duodenum (diffuse epithelial hyperplasia), mesenteric
8.7 mg/kg-d via lymph nodes (histiocytic cellular infiltration), liver
drinking water (histiocytic cellular infiltration), and pancreas (depletion
of cytoplasmic zymogen granules).
M: 0.38, 0.91,
2.4, or M: Increased incidences of histopathological changes to
5.9 mg/kg-d via the duodenum (diffuse epithelial hyperplasia) and
drinking water mesenteric lymph nodes (histiocytic cellular infiltration).
Dog Not 0, 0.45, 2.25, 4 yrs ND ND No effects were observed. Doses in mg hexavalent Anwar et al.,
specified 4.5, 6.75, or chromium/kg-d could not be estimated. 1961
11.2 mg/L in
drinking water
M: 0, 0.35, 1.1,
2.1, or
8.5 mg/kg-d via
the diet
a
Unless otherwise noted, dose or concentration expressed as hexavalent chromium.
(5) Histiocytic cellular infiltration in the mesenteric lymph nodes of male mice
(6) Histiocytic cellular infiltration in the mesenteric lymph nodes of female mice, and
(7) Cytoplasmic cellular alteration of acinar epithelial cells in the pancreas of female
mice.
All of these effects occurred at the lowest doses tested (i.e., 0.24 mg/kg-day in female
rats and 0.38 mg/kg-day in male and female mice), and were considered as possible critical
effects for derivation of the RfD for hexavalent chromium. The incidences of these seven effects
across all treatment groups in NTP (2008) are shown in Table 5-1.
Dose
(mg hexavalent chromium/kg-d)
Endpoint 0 0.24 0.94 2.4 7.0
Female rats
Liver: chronic inflammation 12/50 21/50a 28/50b 35/50b 39/50b
Dose
(mg hexavalent chromium/kg-d)
0 0.38 0.91 2.4 5.9
Male mice
Duodenum: diffuse epithelial hyperplasia 0/50 11/50b 18/50b 42/50b 32/50a
Mesenteric lymph node: histiocytic cellular infiltration 14/47 38/47b 31/49b 32/49b 42/46a
Dose
(mg hexavalent chromium/kg-d)
0 0.38 1.4 3.1 8.7
Female mice
Duodenum: diffuse epithelial hyperplasia 0/50 16/50b 35/50b 31/50b 42/50b
Mesenteric lymph node: histiocytic cellular infiltration 3/46 29/48b 26/46b 40/50b 42/50b
Liver: histiocytic cellular infiltration 2/49 15/50b 23/50b 32/50b 45/50b
Pancreas: acinus, cytoplasmic alteration 0/48 6/50a 6/49a 14/50b 32/50b
a
Significantly different (p ≤ 0.05) from the control group by Dunn’s or Shirley’s test.
b
Significantly different (p ≤ 0.01) from the control group by Dunn’s or Shirley’s test.
Table 5-2. Summary of BMD10 and BMDL10 from the best fitting models for
lesions of the liver, duodenum, mesenteric lymph nodes, and pancreas in
female rats and male and female mice after exposure to sodium dichromate
dihydrate in drinking water for 2 years (NTP, 2008)
a
BMDs and BMDLs from dichotomous data are associated with a 10% extra risk; doses are in terms of mg
hexavalent chromium/kg-d.
b
None of the models provided an adequate fit to the data.
• A UF for LOAEL to NOAEL extrapolation was not used because the current
approach is to address this extrapolation as one of the considerations in selecting a
BMR for BMD modeling. In this case, a BMR represented by a 10% extra risk of
diffuse epithelial hyperplasia was selected under an assumption that it represents a
minimal biologically significant change.
For this assessment, the RfD of 0.0009 or 9 × 10-4 mg/kg-day for hexavalent chromium
was derived by dividing the BMDL10 (or POD) of 0.09 mg/kg-day by a composite uncertainty
factor of 100 (10 for extrapolation from animals to humans and 10 for human variability).
a
Intakes were reported by NTP (2008) based on drinking water intakes and mean body weights observed during the
study.
b
Number of animals with lesion/number of animals examined. Incidence estimates include all animals that were
examined for oral tumors unadjusted for survival.
c
Statistically significantly elevated above control at p < 0.05 using Fisher’s exact test.
a
Intakes were reported by NTP (2008) based on drinking water intakes and mean body weights observed during the
study.
b
Number of animals with lesion/number of animals examined. Incidence estimates include all animals that were
examined for oral tumors unadjusted for survival.
c
Statistically significantly elevated above control at p < 0.05 using Fisher’s exact test.
Also from the NTP (2008) study, incidence data for neoplastic lesions of the small
intestine in male and female B6C3F1 mice exposed to sodium dichromate dihydrate in drinking
water for 2 years are summarized in Table 4-19. In male mice, statistically significant increases
(p < 0.05) were observed in the incidences of adenomas or carcinomas combined in the small
intestine (duodenum, jejunum, and ileum) at hexavalent chromium doses ≥2.4 mg/kg-day (i.e., at
the two highest doses tested). Furthermore, significant positive trends were observed in the
incidences of duodenal adenomas, duodenal carcinomas, jejunal adenomas, small intestine
a
Intakes were reported by NTP (2008) based on drinking water intakes and mean body weights observed during
the study.
b
Calculated from reported percentages of mice with adenomas or carcinomas. Incidence estimates included all
animals that were examined for intestinal tumors and survived for at least 451 days. In each of the control and
first two dose groups, one animal died prior to day 451. In the high-dose group, two animals died prior to
day 451. None of these animals were found to have intestinal adenomas or carcinomas at the time of death.
c
Statistically significantly elevated above control at p < 0.05 using Fisher’s exact test.
a
Intakes were reported by NTP (2008) based on drinking water intakes and mean body weights observed during
the study.
b
Calculated from reported percentages of mice with adenomas or carcinomas. Incidence estimates included all
animals that were examined for intestinal tumors and survived for at least 451 days. In all of the dose groups
except the low-dose group, one animal died prior to day 451. None of these animals were observed to have
intestinal adenomas or carcinomas at the time of death.
c
Statistically significantly elevated above control at p < 0.05 using Fisher’s exact test.
where
WH = animal body weight (kg)
WA = human body weight (kg)
Using the above equation, the CSFs resulting from the fitting of the two-stage multistage
model in BMDS to the incidence of neoplasms in the small intestine of male or female mice
were 0.5 and 0.6 (mg/kg-day)-1, respectively, expressed in human equivalents.
Note that the partial risk for each age group is the product of the values in columns 2–5
(e.g., 10 × 0.5 × 0.0001 × 2/70 = 0.00001 for exposures from age 0 to <2 years), and the total
risk is the sum of the partial risks. Thus, a 70-year risk estimate for a constant average daily
dose of 0.0001 mg/kg-day starting at birth is 0.00008 or 8 × 10-5.
If calculating the cancer risk for a 30-year exposure to a constant average daily dose of
0.0001 mg hexavalent chromium/kg-day from ages 0 to 30 years, the duration adjustments would
Statistical ↓ CSF 25% if BMDL (default Size of bioassay results in sampling variability;
uncertainty at maximum likelihood approach for lower bound is 95% CI on administered dose.
POD estimation (i.e., calculating
BMD10) used rather reasonable upper
than lower bound bound CSF)
(BMDL10) for POD
Species/gender Human risk could ↓ or Male mouse tumors It was assumed that humans are as sensitive as the
combination ↑, depending on (adenomas or most sensitive rodent gender/species tested; true
relative sensitivity carcinomas of the correspondence is unknown. The carcinogenic
small intestine) response occurs across species. Generally, direct
site concordance is not assumed; consistent with
this view, some human tumor types are not found in
rodents and rat and mouse tumor types also differ.
Human relevance Lack of human Tumors with Hexavalent chromium is judged to be carcinogenic
of rodent tumor relevance of tumor significant dose- through a mutagenic mode of action and is a
data data would ↓ CSF response considered multisite carcinogen in rodents; therefore, the
for estimating carcinogenicity observed in rodent studies is
potential human assumed to be relevant to human exposure.
cancer response
Human Low-dose risk ↑ or ↓ to Considered No data are available to support the range of human
population an unknown extent qualitatively variability/sensitivity to hexavalent chromium.
variability in
metabolism and
response/
sensitive
subpopulations
6.2.2. Cancer—Oral
The mode of action is a key consideration in clarifying how risks should be estimated for
low-dose exposure. A linear low-dose extrapolation approach was used to estimate human
carcinogenic risk associated with hexavalent chromium exposures. This approach is supported
by the evidence for genotoxicity and a mutagenic mode of action.
The CSF for hexavalent chromium is based on tumor incidence data from the NTP (2008)
animal bioassay. The incidence of neoplasms in the small intestine of mice was used to derive
the CSF. Only animals that survived for at least 451 days, the time until appearance of the first
tumor, were considered at risk for tumor development.
BMD modeling was carried out using the multistage model in EPA’s BMDS (U.S. EPA,
2000b) to identify a POD. In applying the BMD approach to the derivation of a CSF, the lower
95% confidence bound on the dose corresponding to the BMR (defined as 10% extra risk of
small intestine tumors) was calculated. This lower confidence bound is referred to as the
BMDL. The CSF was calculated by dividing the BMR by the BMDL, and then converting this
CSF to human equivalents using body weight to the ¾ power scaling.
The CSF resulting from the fitting of the multistage model in BMDS to the incidence of
neoplasms in the small intestine of male and female mice was 0.5 (mg/kg-day)-1 and 0.6 (mg/kg-
day)-1, respectively, expressed in human equivalents. Because of the poorer fit of the multistage
model to the female mouse data, the cancer potency estimate of 0.5 (mg/kg-day)-1 based on the
male mouse data was selected as the CSF for hexavalent chromium.
Aaseth, J; Alexander, J; Norseth, T. (1982) Uptake of 51Cr-chromate by human erythrocytes-a role of glutathione.
Acta Pharmacol Toxicol (Copenh) 50(4):310–315.
ACGIH (American Conference of Governmental Industrial Hygienists). (2004) Threshold limit values for chemical
substances and physical agents and Biological exposure indices. Cincinnati, OH: American Conference of
Governmental Industrial Hygienists. ISBN 1-882417-54-2.
Acharya, S; Mehta, K; Krishnan, S; et al. (2001) A subtoxic interactive toxicity study of ethanol and chromium in
male Wistar rats. Alcohol 23:99–108.
Adams, SP; Laws, GM; Storer, RD; et al. (1996) Detection of DNA damage induced by human carcinogens in
acellular assays: potential application for determining genotoxic mechanisms. Mutat Res 368(3–4):235–248.
Aitio, A.; Järvisaleo, J.; Kiilunen, M.; Tossavainen, A.; Vaittinen, P. (1984) Urinary excretion of chromium as an
indicator of exposure to trivalent chromium sulphate in leather tanning. Int Arch Occup Environ Health 50:310 –
315.
Aitio, A.; Järvisaleo, J.; Kiilunen, M.; Kalliomaki, P.L.; Kalliomaki, K. (1988) Chromium: In Biological
monitoring of toxic metals. Eds. Clarkson, T.W.; Friberg, L.; Nordberg, G.F.; Sager, D.R. pp. 369 – 382. Plenum
Press, New York.
Aiyar, J; Berkovits, HJ; Floyd, RA. (1991) Reaction of hexavalent chromium with glutathione or with hydrogen
peroxide: identification of reactive intermediates and their role in hexavalent chromium-induced DNA damage.
Environ Health Perspect 92:53–62.
Alderson, MR; Rattan, NS; Bidstrup, L. (1981) Health of workmen in the chromate-producing industry in Britain.
Br J Ind Med 38:117-124.
Al-Hamood, MH; Elbetieha, A; Bataineh, H. (1998) Sexual maturation and fertility of male and female mice
exposed prenatally and postnatally to trivalent and hexavalent chromium compounds. Reprod Fertil Dev 10(2):179–
183.
Amlacher, E; Rudolph, C. (1981) The thymidine incorporation inhibiting screening system (TSS) to test
carcinogenic substances (a nuclear DNA synthesis suppressive short term test). Arch Geschwulstforsch 51(7):605–
610.
Amrani, S; Rizki, M; Creus, A; et al. (1999) Genotoxic activity of different chromium compounds in larval cells of
Drosophila melanogaster, as measured in the wing spot test. Environ Mol Mutagen 34(1):47–51.
Anger, G; Halstenberg, J; Hochgeschwender, K; et al. (2005) Chromium compounds. In: Ullmann's encyclopedia of
industrial chemistry. New York, NY: John Wiley & Sons, Inc., pp. 1–36.
Anwar, RA; Langham, CF; Hoppert, CA; et al. (1961) Chronic toxicity studies. III. Chronic toxicity of cadmium and
chromium in dogs. Arch Environ 3:456–460.
Arlauskas, A; Baker, RS; Bonin, AM; et al. (1985) Mutagenicity of metal ions in bacteria. Environ Res 36(2):379–
388.
Aruldhas, MM; Subramanian, S; Sekhar, P; et al. (2004) Microcanalization in the epididymis to overcome ductal
obstruction caused by chronic exposure to chromium - a study in the mature bonnet monkey (Macaca radiata
Geoffroy). Reproduction 128(1):127–137.
Aruldhas, MM; Subramanian, S; Sekar, P; et al. (2005) Chronic chromium exposure-induced changes in testicular
histoarchitecture are associated with oxidative stress: study in a non-human primate (Macaca radiata Geoffroy).
Hum Reprod 20(10):2801–2813.
Ashley, K; Howe, AM; Demange, M; et al. (2003) Sampling and analysis considerations for the determination of
hexavalent chromium in workplace air. J Environ Monit 5:707–716.
Asmatullah; Noreen, MA. (1999) Effect of oral administration of hexavalent chromium on total body weight,
chromium uptake and histological structure of mouse liver. Punjab Univ J Zool 14:53–63.
ATSDR (Agency for Toxic Substances and Disease Registry). (2008) Toxicological profile for chromium. Draft for
public comment. U.S. Department of Health and Human Services. U.S. Public Health Service, Atlanta, GA.
Baetjer, AM. (1950a) Pulmonary carcinoma in chromate workers. In: A review of the literature and report of cases.
Arch Ind Hyg Occup Med 2(5):487-504.
Baetjer, AM. (1950b) Pulmonary carcinoma in chromate workers. II. Incidence on basis of hospital records. Arch
Ind Hyg Occup Med 2(5):505-516.
Balasoiu, CF; Zagury, GJ; Deschenes, L. (2001) Partitioning and speciation of chromium, copper, and arsenic in
CCA-contaminated soils: Influence of soil composition. Sci Total Environ 280(1-3):239-55.
Banu, SK; Samuel, JB; Arosh, JA; et al. (2008) Lactational exposure to hexavalent chromium delays puberty by
impairing ovarian development, steroidogenesis and pituitary hormone synthesis in developing Wistar rats. Toxicol
Appl Pharmacol 232(2):180–189.
Baranowska-Dutkiewicz, B. (1981) Absorption of hexavalent chromium by skin in man. Arch Toxicol 47(1):47–50.
Barton, H.A.; Deisinger, P.J.; English, J.C.; Gearhart, J.M.; Faber, W.D.; Tyler, T.R.; Banton, M.I.; Teeguarden, J.;
Andersen, M.E. (2000) Family approach for estimating reference concentrations/doses for series of related organic
chemicals. Toxicol Sci 54:251 – 261.
Bataineh, H; Al-Hamood, MH; Elbetieha, A; et al. (1997) Effect of long-term ingestion of chromium compounds on
aggression, sex behavior and fertility in adult male rat. Drug Chem Toxicol 20(3):133–149.
Bataineh, HN; Bataineh, Z; Daradka, H. (2007) Short-term exposure of female rats to industrial metal salts: effect on
implantation and pregnancy. Reprod Med Biol 6(3):179–183.
Beaumont, JJ; Sedman, RM; Reynolds, SD; et al. (2008) Cancer mortality in a Chinese population exposed to
hexavalent chromium in drinking water. Epidemiology 19(1):12–23.
Bednar CM; Kies C. (1991) Inorganic contaminants in drinking water correlated with disease occurrence in
Nebraska. Water Resour Bull 27(4):631–635.
Bennicelli, C; Camoirano, A; Petruzzelli, S; et al. (1983) High sensitivity of Salmonella TA102 in detecting
hexavalent chromium mutagenicity and its reversal by liver and lung preparations. Mutat Res 122(1):1–5.
Benova, D; Hadjidekova, V; Hristova, R; et al. (2002) Cytogenetic effects of hexavalent chromium in Bulgarian
chromium platers. Mutat Res 514(1–2):29–38.
Beyersmann, D.; Hartwig, A. (2008) Carcinogenic metal compounds: Recent insights into molecular and cellular
mechanisms. Arch Toxicol 82:493 – 512.
Bianchi, V; Dal Toso, R; Debetto, P; et al. (1980) Mechanisms of chromium toxicity in mammalian cell cultures.
Toxicology 17(2):219–224.
Bick, RL; Girardi, TV; Lack, WJ; et al. (1996) Hodgkin’s disease in association with hexavalent chromium
exposure. Int J Hematol 64(3–4):257–262.
Bidstrup, PL. (1951) Carcinoma of the lung in chromate workers. Br J Med 8:302-305.
Bidstrup, PL; Case, RAM. (1956) Carcinoma of the lung in workmen in the bichromates-producing industry in
Great Britain. Br J Ind Med 13:260-264.
Bishop, C.; Surgenor, M. Eds. (1964) The red blood cell: A comprehensive treatise. Academic Press, New York.
Blade, LM; Yencken, MS; Wallace, ME; et al. (2007) Hexavalent chromium exposures and exposure-control
technologies in American enterprise: results of a NIOSH field research study. J Occup Environ Hyg 4:595–618.
Blankenship, LJ; Carlisle, DL; Wise, JP; et al. (1997) Induction of apoptotic cell death by particulate lead chromate:
differential effects of vitamins C and E on genotoxicity and survival. Toxicol Appl Pharmacol 146(2):270–280.
Blasiak, J; Kowalik, J. (2000) A comparison of the in vitro genotoxicity of tri- and hexavalent chromium. Mutat
Res 469(1):135–145.
Blasiak, J; Trzeciak, A; Malecka-Panas, E; et al. (1999) DNA damage and repair in human lymphocytes and gastric
mucosa cells exposed to chromium and curcumin. Teratog Carcinog Mutagen 19:19–31.
Borges, K.M.; Boswell, J.S.; Liebross, R.H.; Wetterhahn, K.E. (1991) Activation of chromium(VI) by thiols results
in chromium(V) formation, chromium binding to DNA and altered DNA conformation. Carcinogenesis 12:551 –
561.
Borneff, J; Engelhardt, K; Griem, W; et al. (1968) [Carcinogens in water and soil. XXII. Mouse drinking water
experiments with 3,4-benzopyrene and potassium chromate]. Arch Hyg Bakteriol 152(1):45–53.
Borthiry, GR; Antholine, WE; Myers, JM; et al. (2008) Reductive activation of hexavalent chromium by human
lung epithelial cells: generation of Cr(V) and Cr(V)-thiol species. J Inorg Biochem 102(7):1449–1462.
Bragt, PC; van Dura, EA. (1983) Toxicokinetics of hexavalent chromium in the rat after intratracheal administration
of chromates of different solubilities. Ann Occup Hyg 27(3):315–322.
Brams, A; Buchet, JP; Crutzen-Fayt, MC; et al. (1987) A comparative study, with 40 chemicals, of the efficiency of
the Salmonella assay and the SOS chromotest (kit procedure). Toxicol Lett 38(1–2):123–133.
Brandt-Rauf, P. (2006) Editorial retraction. Cancer mortality in a Chinese population exposed to hexavalent
chromium in water. J Occup Environ Med 48(7):749.
Bridges, CC; Zalups, RK. (2005) Molecular and ionic mimicry and the transport of toxic metals. Toxicol Appl
Pharmacol 204(3):274–308.
Bridgewater, LC; Manning, FC; Woo, ES; et al. (1994) DNA polymerase arrest by adducted trivalent chromium.
Mol Carcinog 9(3):122–133.
Bridgewater, LC; Manning, FC; Patierno, SR. (1998) Arrest of replication by mammalian DNA polymerases alpha
and beta caused by chromium-DNA lesions. Mol Carcinog 23(4):201–206.
Briggs, JA; Briggs, RC. (1988) Characterization of chromium effects on a rat liver epithelial cell line and their
relevance to in vitro transformation. Cancer Res 48(22):6484–6490.
Bronzetti, GL; Galli, A. (1989) Influence of NTA on the chromium genotoxicity. Toxicol Environ Chem 23:101–
104.
Brooks, B; O’Brien, TJ; Ceryak, S; et al. (2008) Excision repair is required for genotoxin-induced mutagenesis in
mammalian cells. Carcinogenesis 29(5):1064-1069.
Bukowksi, JA. (1991) Biological markers in chromium exposure assessment: Confounding variables. Arch Environ
Health 46(4):230–236.
Campbell J.L.; Tan,Y.; Clewell, H.J. (2009) Development of a PBPK model for hexavalent chromium in rats and
mice to estimate exposure to oral mucosa and small intestine. Toxicologist 108(1):98 (Abstract) Poster ID # 108.
Capellmann, M; Mikalsen, A; Hindrum, M; et al. (1995) Influence of reducing compounds on the formation of
DNA-protein cross-links in HL-60 cells induced by hexavalent chromium. Carcinogenesis 16(5):1135–1139.
Cavalleri, A; Minoia, C; Richelmi, P; et al. (1985) Determination of total and hexavalent chromium in bile after
intravenous administration of potassium dichromate in rats. Environ Res 37(2):490–496.
CDPH (California Department of Public Health). (2007) Chromium-6 in drinking water: sampling results. Health,
CDoP. Available online at http://ww2.cdph.ca.gov/certlic/drinkingwater/pages/chromium6sampling.aspx (accessed
May 10. 2010).
Cemeli, E; Carder, J; Anderson, D; et al. (2003) Antigenotoxic properties of selenium compounds on potassium
dichromate and hydrogen peroxide. Teratog Carcinog Mutagen 23(Suppl 2):53–67.
ChemIDplus. (2008) Chromium and compounds. Bethesda, MD: National Library of Medicine. Available online at
http://chem.sis.nlm.nih.gov/chemidplus/ (accessed May 10, 2010).
Cheng, L; Sonntag, DM; de Boer, J; et al. (2000) Chromium(VI) induces mutagenesis in the lungs of big blue
transgenic mice. J Environ Pathol Toxicol Oncol 19(3):239–249.
Chopra, A; Pereira, G; Gomes, T; et al. (1996) A study of chromium and ethanol toxicity in female Wistar rats.
Toxicol Environ Chem 53:91–106.
Chowdhury, AR; Mitra, C. (1995) Spermatogenic and steroidogenic impairment after chromium treatment in rats.
Indian J Exp Biol 33(7):480–484.
Cikrt, M; Bencko, V. (1979) Biliary excretion and distribution of 51Cr(III) and 51Cr(VI) in rats. J Hyg Epidemiol
Microbiol Immunol 23:241–246.
Clewell, HJ, III; Andersen, ME. (1985) Risk assessment extrapolations and physiological modeling. Toxicol Ind
Health 1(4):111–131.
Clochesy, JM. (1984) Chromium ingestion: a case report. J Emerg Nurs 10(6):281–282.
Cocker, J; Morton, J; Warren, N; Wheeler, JP; Garrod, ANI. (2006) Biomonitoring for chromium and arsenic in
timber treatment plant workers exposed to CCA wood preservatives. Ann Occup Hyg 50(5):517-625.
Coogan, T.P.; Squibb, K.S.; Motz, J.; Kinney, P.L.; Costa, M. (1991a) Distribution of chromium within cells of the
blood. Toxicol Appl Pharmacol 108:157 – 166.
Coogan, TP; Motz, J; Snyder, CA; et al. (1991b) Differential DNA-protein crosslinking in lymphocytes and liver
following chronic drinking water exposure of rats to potassium chromate. Toxicol Appl Pharmacol 109(1):60–72.
Corbett, GE; Finley, BL; Paustenbach, DJ; et al. (1997) Systemic uptake of chromium in human volunteers
following dermal contact with hexavalent chromium (22 mg/L). J Expo Anal Environ Epidemiol 7(2):179–189.
Costa, M.; Klein, C.B. (2008) Toxicity and carcinogenicity of chromium compounds in humans. Crit Rev Toxicol
36(2):155 – 163.
Cotton, FA; Wilkinson, G; Murillo, CA; et al. (1999) The elements of the first transition series. In: Advanced
inorganic chemistry. New York, NY: John Wiley & Sons, Inc., pp. 736–752.
Dai, H; Liu, J; Malkas, LH; et al. (2009) Chromium reduces the in vitro and fidelity of DNA replication mediated by
the human cell DNA synthesome. Toxicol Appl Pharmacol 236:154–165.
Danielsson, BRG; Hassoun, E; Dencker, L. (1982) Embryotoxicity of chromium: distribution in pregnant mice and
effects on embryonic cells in vitro. Arch Toxicol 51:233–245.
Davies, JM. (1978) Lung-cancer mortality in workers making chrome pigments. Lancet 1:384.
Davies, JM. (1979) Lung cancer mortality of workers in chromate pigment manufacture: An epidemiological survey.
J Oil Chem Assoc 62:157-163.
Davies, JM. (1984) Lung cancer mortality among workers making lead chromate and zinc chromate pigments at
three English factories. Br J Ind Med 41:158-169.
Decker, LE; Byerrum, RU; Decker, CF; et al. (1958) Chronic toxicity studies. I. Cadmium administered to drinking
water in rats. AMA Arch Ind Health 18(3):228–231.
De Flora, S. (1978) Metabolic deactivation of mutagens in the Salmonella-microsome test. Nature 271(5644):455–
456.
De Flora, S. (1981) Study of 106 organic and inorganic compounds in the Salmonella/microsome test.
Carcinogenesis 2(4):283–298.
De Flora, S. (2000) Threshold mechanisms and site specificity in chromium(VI) carcinogenesis. Carcinogenesis
21(4):533 – 541.
De Flora, S; Wetterhahn, KE. (1989) Mechanisms of chromium metabolism and genotoxicity. Life Sci 7:169–244.
De Flora, S; Bennicelli, C; Zanacchi, P; et al. (1984) Metabolic activation and deactivation of mutagens by
preparations of human lung parenchyma and bronchial tree. Mutat Res 139(1):9–14.
De Flora, S; Badolati, GS; Serra, D; et al. (1987) Circadian reduction of chromium in the gastric environment.
Mutat Res 192(3):169–174.
De Flora, S; Camoirano, A; Bagnasco, M; et al. (1997) Etimates of the chromium(VI) reducing capacity in human
body compartments as a mechanism for attenuating its potential toxicity and carcinogenicity. Carcinogenesis
18(3):531–537.
De Flora, S; Iltcheva, M; Balansky, RM. (2006) Oral chromium (VI) does not affect the frequency of micronuclei in
hematopoietic cells of adult mice and of transplacentally exposed fetuses. Mutat Res 610(1–2):38–47.
De Flora, S; D’Agostini, F, Balansky, R; et al. (2008) Lack of genotoxic effects in hematopoietic and
gastrointestinal cells of mice receiving chromium(VI) with the drinking water. Mutat Res 659:60–67.
Deng, C; Lee, HH; Xian, H; et al. (1988) Chromosomal aberrations and sister chromatid exchanges of peripheral
blood lymphocytes in Chinese electroplating workers: effect of nickel and chromium. J Trace Elem Exper Med
1:57–62.
Devi, KD; Rozati, R; Saleha Banu, B; et al. (2001) In vivo genotoxic effect of potassium dichromate in mice
leukocytes using comet assay. Food Chem Toxicol 39(8):859–865.
DiPaolo, JA; Casto, BC. (1979) Quantitative studies of in vitro morphological transformation of Syrian hamster
cells by inorganic metal salts. Cancer Res 39(3):1008–1013.
Donaldson, RM, Jr; Barreras, RF. (1966) Intestinal absorption of trace quantities of chromium. J Lab Clin Med
68(3):484–493.
Dunkel, VC; Pienta, RJ; Sivak, A; et al. (1981) Comparative neoplastic transformation responses of Balb/3T3 cells,
Syrian hamster embryo cells, and Rauscher murine leukemia virus-infected Fischer 344 rat embryo cells to chemical
compounds. J Natl Cancer Inst 67(6):1303–1312.
Dunkel, VC; Zeiger, E; Brusick, D; et al. (1984) Reproducibility of microbial mutagenicity assays: I. Tests with
Salmonella typhimurium and Escherichia coli using a standardized protocol. Environ Mutagen 6(Suppl 2):1–251.
Edel, J; Sabbioni, E. (1985) Pathways of Cr(III) and Cr(VI) in the rat after intratracheal administration. Hum
Toxicol 4(4):409–416.
Eizaguirre-Garcia, D; Rodriguez-Andres, C; Watt, GC; et al. (1999) A study of leukaemia in Glasgow in connection
with chromium-contaminated land. J Public Health Med 21(4):435–438.
Eizaguirre-Garcia, D; Rodriguez-Andres, C; Watt, GC. (2000) Congenital anomalies in Glasgow between 1982 and
1989 and chromium waste. J Public Health Med 22(1):54–58.
Elbetieha, A; Al-Hamood, MH. (1997) Long-term exposure of male and female mice to trivalent and hexavalent
chromium compounds: effect on fertility. Toxicology 116(1–3):39–47.
Elia, MC; Storer, RD; McKelvey, TW; et al. (1994) Rapid DNA degradation in primary rat hepatocytes treated with
diverse cytotoxic chemicals: analysis by pulsed field gel electrophoresis and implications for alkaline elution assays.
Environ Mol Mutagen 24(3):181–191.
Elsaieed, EM; Nada, SA. (2002) Teratogenicity of hexavalent chromium in rats and the beneficial role of ginseng.
Bull Environ Contam Toxicol 68(3):361–368.
Enterline, PE. (1974) Respiratory cancer among chromate workers. J Occup Med 16:523-526.
Febel, H; Szegedi, B; Huszar, S. (2001) Absorption of inorganic, trivalent and hexavalent chromium following oral
and intrajejunal doses in rats. Acta Vet Hung 49(2):203–209.
Fendorf, S; La Force, MJ; Li, G. (2004) Heavy metals in the environment: Temporal changes in soil partitioning of
and bioaccessibility of arsenic, chromium and lead. J Environ Qual 33:2049–2055.
Finley, BL; Johnson, EM; Holson, JF. (1993) Comment on “Comparative effects of trivalent and hexavalent
chromium on spermatogenesis of the mouse.” Toxicol Environ Chem 39:133–137.
Finley, BL; Scott, PK; Norton, RL; et al. (1996) Urinary chromium concentrations in humans following ingestion of
safe doses of hexavalent and trivalent chromium: implications for biomonitoring. J Toxicol Environ Health
48(5):479–499.
Finley, BL; Kerger, BD; Katona, MW; et al. (1997) Human ingestion of chromium (VI) in drinking water:
pharmacokinetics following repeated exposure. Toxicol Appl Pharmacol 142(1):151–159.
Flores, A; Perez, JM. (1999) Cytotoxicity, apoptosis, and in vitro DNA damage induced by potassium chromate.
Toxicol Appl Pharmacol 161(1):75–81.
Fornace, AJ, Jr.; Seres, DS; Lechner, JF; et al. (1981) DNA-protein cross-linking by chromium salts. Chem Biol
Interact 36(3):345–354.
Frentzel-Beyme, R. (1983) Lung cancer mortality of workers employed in chromate pigment factories. A
multicentric European epidemiological study. J Cancer Res Clin Oncol 105:183-188.
Fristedt, B; Lindqvist, B; Schuetz, A; et al. (1965) Survival in a case of acute oral chromic acid poisoning with acute
renal failure treated by haemodialysis. Acta Med Scand 177:153–159.
Fryzek, JP; Mumma, MT; McLaughlin, JK; et al. (2001) Cancer mortality in relation to environmental chromium
exposure. J Occup Environ Med 43(7):635–640.
Fukunaga, M; Kurachi, Y; Mizuguchi, Y. (1982) Action of some metal ions on yeast chromosomes. Chem Pharm
Bull 30:3017–3019.
Furst, A; Schlauder, M; Sasmore, DP. (1976) Tumorigenic activity of lead chromate. Cancer Res 36:1779-1783.
Fytianos, K. (2001) Speciation analysis of heavy metals in natural waters: A review. J AOAC Intl 84(6):1763–
1769.
Gambelunghe, A; Piccinini, R; Ambrogi, M; et al. (2003) Primary DNA damage in chrome-plating workers.
Toxicology 188(2–3):187–195.
Gammelgaard, B.; Jensen, K.; Steffansen, B. (1999) In vitro metabolism and permeation studies in rat jejunum:
Organic chromium compared to inorganic chromium. J Trace Elements Med Biol 13:82 – 88.
Gao, M; Levy, LS; Braithwaite, RA; et al. (1993) Monitoring of total chromium in rat fluids and lymphocytes
following intratracheal administration of soluble trivalent or hexavalent chromium compounds. Hum Exp Toxicol
12(5):377–382.
Gao, M; Levy, LS; Faux, SP; et al. (1994) Use of molecular epidemiological techniques in a pilot study on workers
exposed to chromium. Occup Environ Med 51(10):663–668.
Garcia, JD; Jennette, KW. (1981) Electron-transport cytochrome P-450 system is involved in the microsomal
metabolism of the carcinogen chromate. J Inorg Biochem 14(4):281–295.
Gargas, ML; Norton, RL; Paustenbach, DJ; et al. (1994) Urinary excretion of chromium by humans following
ingestion of chromium picolinate. Implications for biomonitoring. Drug Metab Dispos 22(4):522–529.
Gealy, R; Wright-Bourque, JL; Kraynak, AR; et al. (2007) Validation of a high-throughput in vitro alkaline
elution/rat hepatocyte assay for DNA damage. Mutat Res 629(1):49–63.
Gentile, JM; Hyde, K; Schubert, J. (1981) Chromium genotoxicity as influenced by complexation and rate effects.
Toxicol Lett 7(6):439–448.
Glaser, U.; Hochrainer, D.; Klöpper, H.; Kuhnen, H. (1985) Low level chromium (VI) inhalation effects on alveolar
macrophages and immune functions in Wistar rats. Arch Toxicol. 57(4):250 – 256.
Glaser, U; Hochrainer, D; Kloppel, H; et al. (1986) Carcinogenicity of sodium dichromate and chromium(VI/III)
oxide aerosols inhaled by male Wistar rats. Toxicology 42:219-232.
Goldman, M; Karotkin, RH. (1935) Acute potassium bichromate poisoning. Am J Med Sci 189:400–403.
Goode, EL; Ulrich, CM; Potter, JD. (2002) Polymorphisms in DNA repair genes and associations with cancer risk.
Cancer Epidemiol Biomarkers Prev 11(12):1513–1530.
Goodgame, D.M.L.; Joy, M.A. (1998) EPR stud of the Cr(V) and radical species produced in the reduction of
Cr(VI) by ascorbate. Inorg Chem Acta 135:115 – 118.
Graf, U; Wurgler, FE. (1996) The somatic white-ivory eye spot test does not detect the same spectrum of genotoxic
events as the wing somatic mutation and recombination test in Drosophila melanogaster. Environ Mol Mutagen
27(3):219–226.
Gray, S.J.; Sterling, K. (1950) The tagging of red cells and plasma proteins with radioactive chromium. J Clin
Invest 29:1604 – 1613.
Grlickova-Duzevik, E; Wise, SS; Munroe, RC; et al. (2006) XRCC1 protects cells from chromate-induced
chromosome damage, but does not affect cytotoxicity. Mutat Res 610(1-2):31–37.
Gruber, JE; Jennette, KW. (1978) Metabolism of the carcinogen chromate by rat liver microsomes. Biochem
Biophys Res Commun 82(2):700–706.
Guttmann, D; Poage, G; Johnston, T; et al. (2008) Reduction with glutathione is a weakly mutagenic pathway in
chromium(VI) metabolism. Chem Res Toxicol 21(11):2188–2194.
Gylseth, B; Gundersen, N; Langard, S. (1977) Evaluation of chromium exposure based on a simplified method for
urinary chromium. Scand J Work Environ Health 3:28–31.
Haguenor, JM; Dubois, G; Frimat, P; et al. (1981) Mortality due to bronch-pulmonary >cancer in a factory
producing pigments based on lead and zinc chromates. In: Prevention of occupational cancer - International
symposium, occupational safety and health series 46. Geneva, Switzerland: International Labour Office, pp. 168-
176. (French).
Hamilton, JW; Wetterhahn, KE. (1986) Chromium(VI)-induced DNA damage in chick embryo liver and blood cells
in vivo. Carcinogenesis 7(12):2085–2088.
Hantson, P; Van Caenegem, O; Decordier, I; et al. (2005) Hexavalent chromium ingestion: biological markers of
nephrotoxicity and genotoxicity. Clin Toxicol (Phila) 43(2):111–112.
Hasan, A. (2007) A case report: ammonium dichromate poisoning. Biomed Res 18(1):35–37.
Haworth, S; Lawlor, T; Mortelmans, K; et al. (1983) Salmonella mutagenicity test results for 250 chemicals.
Environ Mutagen 5(Suppl 1):1–142.
Hayashi, M; Sofuni, T; Ishidate, M, Jr. (1982) High-sensitivity in micronucleus induction of a mouse strain (MS).
Mutat Res 105(4):253–256.
Hayes, RB; Lilienfeld, AM; Snell, LM. (1979) Mortality in chromium chemical production workers: a prospective
study. Int J Epidemiol 8(4):365-374.
Hayes, RB; Sheffet, A; Spirtas, R. (1989) Cancer mortality among a cohort of chromium pigment workers. Am J Ind
Med 16:127-133.
Heil, J; Reifferscheid, G. (1992) Detection of mammalian carcinogens with an immunological DNA synthesis-
inhibition test. Carcinogenesis 13(12):2389–2394.
Hirose, T; Kondo, K; Takahashi, Y; et al. (2002) Frequent microsatellite instability in lung cancer from chromate-
exposed workers. Mol Carcinog 33(3):172–180.
Holmes AL, Wise SS, Sandwick SJ, et al. (2006) The clastogenic effects of chronic exposure to particulate and
soluble Cr(VI) in human lung cells. Mutat Res 610(1–2):8–13.
Holmes, A.; Wise, SS; Wise, Sr., JP (2008) Carcinogenicity of hexavalent chromium. Indian J Med Res 128:353 –
372.
Holmes, AL; Wise, SS; Pelsue, SC; et al. (2010) Chronic exposure to zinc chromate induces centrosome
amplification and spindle assembly checkpoint bypass in human lung fibroblasts. Chem Res Toxicol 23:386-395.
Hueper, WC. (1961) Environmental carcinogenesis and cancers. Cancer Res 21:842-857.
Hueper, WC; Payne, WW. (1962) Experimental studies in metal carcinogenesis: Chromium, nickel, iron, and
arsenic. Arch Environ Health 5:445-462.
Iijima, S; Spindle, A; Pedersen, RA. (1983) Developmental and cytogenetic effects of potassium dichromate on
mouse embryos in vitro. Teratology 27(1):109–115.
Imreh, S; Radulescu, D. (1982) Cytogenetic effects of chromium in vivo and in vitro. Mutat Res 97(3):192–193.
Iserson, KV; Banner, W; Froede, RC; et al. (1983) Failure of dialysis therapy in potassium dichromate poisoning. J
Emerg Med 1(2):143–149.
Itoh, S; Shimada, H. (1996) Micronucleus induction by chromium and selenium, and suppression by metallothionein
inducer. Mutat Res 367(4):233–236.
Itoh, S; Shimada, H. (1997) Clastogenicity and mutagenicity of hexavalent chromium in lacZ transgenic mice.
Toxicol Lett 91(3):229–233.
Itoh, S; Shimada, H. (1998) Bone marrow and liver mutagenesis in lacZ transgenic mice treated with hexavalent
chromium. Mutat Res 412(1):63–67.
Izzotti, A; Bagnasco, M; Camoirano, A; et al. (1998) DNA fragmentation, DNA-protein crosslinks, postlabeled
nucleotidic modifications, and 8-hydroxy-2’-deoxyguanosine in the lung but not in the liver of rats receiving
intratracheal instillations of hexavalent chromium. Chemoprevention by oral N-acetylcysteine. Mutat Res 400(1–
2):233–244.
James, BR; Petura, JC; Vitale, RJ; et al. (1997) Oxidation-reduction chemistry of chromium: relevance to the
regulation and remediation of chromate-contaminated soils. J Soil Contam 6(6):569–580.
Jannetto, PJ; Antholine, WE; Myers, CR. (2001) Cytochrome b5 plays a key role in human microsomal hexavalent
chromium reduction. Toxicology 159(3):119–133.
Jennette, KW. (1982) Microsomal reduction of the carcinogen chromate produces chromium(V). J Am Chem Soc
104:874–875.
Johnson, J; Schewel, L; Graedel, TE. (2006) The contemporary anthropogenic chromium cycle. Environ Sci
Technol 40:7060–7069.
Junaid, M; Murthy, RC; Saxena, DK. (1995) Chromium fetotoxicity in mice during late pregnancy. Vet Hum
Toxicol 37(4):320–323.
Junaid, M; Murthy, RC; Saxena, DK. (1996a) Embryotoxicity of orally administered chromium in mice: exposure
during the period of organogenesis. Toxicol Lett 84(3):143–148.
Kanematsu, N; Hara, M; Kada, T. (1980) Rec assay and mutagenicity studies on metal compounds. Mutat Res
77(2):109–116.
Kanojia, RK; Junaid, M; Murthy, RC. (1996) Chromium induced teratogenicity in female rat. Toxicol Lett
89(3):207–213.
Kanojia, RK; Junaid, M; Murthy, RC. (1998) Embryo and fetotoxicity of hexavalent chromium: a long-term study.
Toxicol Lett 95(3):165–172.
Kargacin, B; Squibb, KS; Cosentino, S; et al. (1993) Comparison of the uptake and distribution of chromate in rats
and mice. Biol Trace Elem Res 36:307–318.
Katz, AJ; Chiu, A; Beaubier, J; et al. (2001) Combining Drosophila melanogaster somatic-mutation-recombination
and electron-spin-resonance-spectroscopy data to interpret epidemiologic observations on chromium
carcinogenicity. Mol Cell Biochem 222(1–2):61–68.
Katz, SA. (1991) The analytical biochemistry of chromium. Environ Hlth Perspect 92:13–16.
Katz, SA; Salem, H. (1993) The toxicology of chromium with respet to its chemical speciation: A review. J Appl
Toxicol 13(3):217–224.
Kaufman, DB; DiNicola, W; McIntosh, R. (1970) Acute potassium dichromate poisoning. Treated by peritoneal
dialysis. Am J Dis Child 119(4):374–376.
Kaya, B; Creus, A; Velazquez, A; et al. (2002) Genotoxicity is modulated by ascorbic acid. Studies using the wing
spot test in Drosophila. Mutat Res 520(1–2):93–101.
Kerger, BD; Richter, RO; Chute, SM; et al. (1996a) Refined exposure assessment for ingestion of tap water
contaminated with hexavalent chromium: Consideration of exogenous and endogenous reducing agents. J Exposure
Anal Environ Epi 6(2):163–179.
Kerger, BD; Paustenbach, DJ; Corbett, GE; et al. (1996b) Absorption and elimination of trivalent and hexavalent
chromium in humans following ingestion of a bolus dose in drinking water. Toxicol Appl Pharmacol 141(1):145–
158.
Kerger, BD; Finley, BL; Corbett, GE; et al. (1997) Ingestion of hexavalent chromium in drinking water by human
volunteers: absorption, distribution, and excretion of single and repeated doses. J Toxicol Environ Health 50(1):67–
95.
Kerger, BD; Butler WJ; Paustenbach, DJ; et al. (2009) Cancer mortality in Chinese populations surrounding an alloy
plant with chromium smelting operations. J Toxicol Environ Health Part A 72(5):329–344.
Kharab, P; Singh, I. (1985) Genotoxic effects of potassium dichromate, sodium arsenite, cobalt chloride and lead
nitrate in diploid yeast. Mutat Res 155(3):117–120.
Kirpnick-Sobol, Z; Reliene, R; Schiestl, RH (2006) Carcinogenic Cr(VI) and the nutritional supplement Cr(III)
induce DNA deletions in yeast and mice. Cancer Res 66(7):3480-3484.
Klein, CB; Su, L; Bowser, D; et al. (2002) Chromate-induced epimutations in mammalian cells. Environ Health
Perspect 110(Suppl 5):739–743.
Knudsen, I. (1980) The mammalian spot test and its use for the testing of potential carcinogenicity of welding fume
particles and hexavalent chromium. Acta Pharmacol Toxicol (Copenh) 47(1):66–70.
Korallus, U. (1986) Chromium compounds: Occupational health, toxicological and biological monitoring aspects.
Toxicol Environ Chem 12:47–59.
Korallus, U; Lange H; Ness, A; et al. (1982) Relationships between precautionary measures and bronchial
carcinoma mortality in the chromate-producing industry. Arbeitsmedizin, Socialmedizin, Preventivmedizin.
17(7):159-167. (German - Eng. summary).
Kortenkamp, A; Oetken, G; Beyersmann, D. (1990) The DNA cleavage induced by a chromium(V) complex and by
chromate and glutathione is mediated by activated oxygen species. Mutat Res 232(2):155–161.
Koshi, K. (1979) Effects of fume particles from stainless steel welding on sister chromatid exchanges and
chromosome aberrations in cultured Chinese hamster cells. Ind Health 17:39–50.
Koshi, K; Iwasaki, K. (1983) Solubility of low-solubility chromates and their clastogenic activity in cultured cells.
Ind Health 21(2):57–65.
Koshi, K; Serita, F; Sawatari, K; et al. (1987) Cytogenetic analysis of bone marrow cells and peripheral blood
lymphocytes from rats exposed to chromium fumes by inhalation. Mutat Res 181:365.
Krishnan, K; Andersen, ME. (1994) Physiologically based pharmacokinetic modeling in toxicology. In: Hayes,
AW, ed. Principles and methods of toxicology. New York, NY: Raven Press Ltd., pp. 149–188.
Krishnan, K; Andersen, ME; Clewell, HJ; et al. (1994) Physiologically based pharmacokinetic modeling of chemical
structures. In: Yang, RSH, ed. Toxicology of chemical mixtures. New York, NY: Academic Press, pp. 399–437.
Kumpulainen, JT. (1992) Chromium content of foods and diets. Biol Trace Elem Res 32:9-18.
Kuykendall, JR; Kerger, BD; Jarvi, EJ; et al. (1996) Measurement of DNA-protein cross-links in human leukocytes
following acute ingestion of chromium in drinking water. Carcinogenesis 17(9):1971–1977.
Langård, S; Gundersen, N, Tsalev, DL; et al. (1978) Whole blood chromium and chromium excretion in the rat after
zinc chromate inhalation. Acta Pharmacol Toxicol (Copenh) 42(2):142–149.
Laskin, S; Kuschner, M; Drew, RT. (1970) Studies in pulmonary carcinogenesis. In: Hanna, Jr., MG;, Nettesheim,
P; and Gilbert, JR, eds.
LaVelle, JM. (1986) Potassium chromate potentiates frameshift mutagenesis in E. coli and S. typhimurium. Mutat
Res 171(1):1–10.
Lay, P.A.; Levina, A. (1998) Activation of molecular oxygen during the reactions of chromium(VI/V/IV) with
biological reductants: Implications for chromium-induced genotoxicities. J Am Chem Soc 120:6704 – 6714.
Le Curieux, F; Marzin, D; Erb, F. (1993) Comparison of three short-term assays: results on seven chemicals.
Potential contribution to the control of water genotoxicity. Mutat Res 319(3):223–236.
Levina, A.; Zhang, L.; Lay, P.A. (2003) Structure and reactivity of chromium(V) glutathione complex. Inorg Chem
42:767 – 784.
Levina, A; Lay, PA. (2005) Mechanistic studies of relevance to the biological activities of chromium. Coord Chem
Rev 249(3–4):281–298.
Levina, A.; Harris, H.H.; Lay, P.A. (2007) X-ray absorption and EPR spectroscopic studies of the
biotransformations of chromium(VI) in mammalian cells. Is chormodulin an artifact of isolation methods? J Am
Chem Soc 129:1065 – 1075.
Levis, AG; Majone, F. (1981) Cytotoxic and clastogenic effects of soluble and insoluble compounds containing
hexavalent and trivalent chromium. Br J Cancer 44(2):219–235.
Levy, LS; Martin, PA. (1983) The effects of a range of chromium-containing materials on rat lung. Sponsored by
Dry Color Manufacturers' Association and others. (Unpublished)
Lewalter, J; Korallus, U; Harzdorf, C; Wiedmann, H. (1985) Chromium bond detection in isolated erythrocytes: A
new principle of biological monitoring of exposure to hexavalent chromium. Int Arch Occup Environ Health
55:305–318.
Lewis, RJ. (2007) Chromium and compounds. In: Hawley's condensed chemical dictionary. New York, NY: John
Wiley & Sons, pp. 68–69, 216, 297, 1029–1030.
Li, H; Chen, Q; Li, S; et al. (2001) Effect of Cr(VI) exposure on sperm quality: human and animal studies. Ann
Occup Hyg 45(7):505–511.
Lide, DR. (2008) Chromium and compounds. In: Lide, DR; ed. CRC handbook of chemistry and physics. Boca
Raton, FL: CRC Press, pp. 4-46, 44-54, 44-58, 44-59, 44-70, 44-82, 44-89, 44-100.
Liebross, RH; Wetterhahn, E. (1992) In vivo formation of chromium(V) in chick embryo liver and red blood cells.
Carcinogenesis 13(11):2113–2120.
Lim, T.H.; Sargent, T. III.; Jusubov, N. (1983) Kinetics of trace element chromium (III) in the body. Am J Physiol
244(4):R445 – R454.
Liu, KJ; Shi, X; Jiang, JJ; et al. (1995) Chromate-induced chromium(V) formation in live mice and its control by
cellular antioxidants: an L-band electron paramagnetic resonance study. Arch Biochem Biophys 323(1):33–39.
Llagostera, M; Garrido, S; Guerrero, R; et al. (1986) Induction of SOS genes of Escherichia coli by chromium
compounds. Environ Mutagen 8(4):571–577.
Losi, ME; Amrhein, C; Frankenberger, Jr, WT. (1994) Environmental biochemistry of chromium. Rev Environ
Contam Toxicol 136:91–121.
Loubieres, Y; de Lassence, A; Bernier, M; et al. (1999) Acute, fatal, oral chromic acid poisoning. J Toxicol Clin
Toxicol 37(3):333–336.
Macfie, A; Hagan, E; Zhitkovich, A. (2010) Mechanism of DNA–protein cross-linking by chromium. Chem Res
Toxicol 23(2):341–347.
Machle, W; Gregorius, F. (1948) Cancer of the respiratory system in the United States chromate-producing industry.
Public Health Rep 63(35):1114-1127.
MacKenzie, RD; Byerrum, RU; Decker, CF; et al. (1958) Chronic toxicity studies. II. Hexavalent and trivalent
chromium administered in drinking water to rats. AMA Arch Ind Health 18(3):232–234.
MacKenzie, RD; Anwar, RA; Byerrum, RU; et al. (1959) Absorption and distribution of 51Cr in the albino rat. Arch
Biochem Biophys 79:200–205.
Macrae, WD; Whiting, RF; Stich, HF. (1979) Sister chromatid exchanges induced in cultured mammalian cells by
chromate. Chem-Biol Interact 26(3):281–286.
Mancuso, TF; Hueper, WC. (1951) Occupational cancer and other health Hazards in a chromate plant: A medical
appraisal. In: Lung cancers in chromate workers. Ind Med Surg 20(8):358-363.
Mancuso, TF. (1997) Chromium as an industrial carcinogen: Part 1. Am J Ind Med 31:129-139.
Marzin, DR; Phi, HV. (1985) Study of the mutagenicity of metal derivatives with Salmonella typhimurium TA102.
Mutat Res 155(1–2):49–51.
Matsui, S. (1980) Evaluation of a Bacillus subtilis rec-assay for the detection of mutagens which may occur in water
environments. Water Res 14(11):1613–1619.
McCarroll, N; Keshava, N; Chen, J; et al. (2010) An evaluation of the mode of action framework for mutagenic
carcinogens case study II: Chromium(VI). Environ Mol Mutagen 51:89–111.
McGregor, DB; Martin, R; Cattanach, P; et al. (1987) Responses of the L5178Y tk+/tk- mouse lymphoma cell
forward mutation assay to coded chemicals. I: Results for nine compounds. Environ Mutagen 9(2):143–160.
Merk, O; Reiser, K; Speit, G. (2000) Analysis of chromate-induced DNA-protein crosslinks with the comet assay.
Mutat Res 471(1–2):71–80.
Mertz, W; Roginski, EE; Feldman, FJ; et al. (1969) Dependence of chromium transfer into the rat embryo on the
chemical form. J Nutr 99(3):363–367.
Mertz, W. (1998) Chromium research from a distance: From 1959 to 1980. J Am College Nutrition 17(6):544 –
547.
Mikalsen, A; Alexander, J; Ryberg, D. (1989) Microsomal metabolism of hexavalent chromium. Inhibitory effect of
oxygen and involvement of cytochrome P450. Chem Biol Interact 69:175–192.
Miller, CA; Costa M. (1989) Analysis of proteins cross-linked to DNA after treatment of cells with formaldehyde,
chromate, and cis-diamminedichloroplatinum(II). Mol Toxicol 2:11–26.
Minoia, C; Cavalleri, A. (1988) Chromium in urine, serum and red blood cells in the biological monitoring of
workers exposed to different chromium valency states. Sci Total Environ 71(3):323–327.
Mirsalis, JC; Hamilton, CM; O’Loughlin, KG; et al. (1996) Hexavalent chromium at plausible drinking water
concentrations is not genotoxic in the in vivo bone marrow micronucleus or liver unscheduled DNA synthesis
assays. Environ Mol Mutagen 28(1):60–63.
Mitchell, AD; Rudd, CJ; Caspary, WJ. (1988) Evaluation of the L5178Y mouse lymphoma cell mutagenesis assay:
intralaboratory results for sixty-three coded chemicals tested at SRI International. Environ Mol Mutagen 12(Suppl
13):37–101.
Murthy, RC; Junaid, M; Saxena, DK. (1996) Ovarian dysfunction in mice following chromium (VI) exposure.
Toxicol Lett 89(2):147–154.
Myers, CR; Myers, JM. (1998) Iron stimulates the rate of reduction of hexavalent chromium by human microsomes.
Carcinogenesis 19(6):1029–1038.
Myhr, B; Caspary, W. (1988) Evaluation of the L5178Y mouse lymphoma cell mutagenesis assay; intralaboratory
results for sixty-three coded chemicals tested at Litton Bionetics, Inc. Environ Mol Mutagen 13(12):103–194.
Nagaya, T; Ishikawa, N; Hata, H; et al. (1991) Sister-chromatid exchanges in lymphocytes from 12 chromium
platers: a five-year follow-up study. Toxicol Lett 58(3):329–335.
Nakamura, SI; Oda, Y; Shimada, T; et al. (1987) SOS-inducing activity of chemical carcinogens and mutagens in
Salmonella typhimurium TA1535/pSK1002: examination with 151 chemicals. Mutat Res 192(4):239–246.
Nakamuro, K; Yoshikawa, K; Sayato, Y; et al. (1978) Comparative studies of chromosomal aberration and
mutagenicity of the trivalent and hexavalent chromium. Mutat Res 58(2–3):175–181.
National Institute for Occupational Safety and Health (NIOSH). (1975) Criteria for a recommended standard
occupational exposure to chromium (VI). U.S. Department of Health, Education, and Welfare, Washington, DC.
Nickens, K.P.; Patierno, S.R.; Ceryak, S. (2010) Chromium genotoxicity: A double-edged sword. Chemi Biol
Interact Nov 5;188(2):276-88. Epub 2010 Apr 27.
Nishio, A; Uyeki, EM. (1985) Inhibition of DNA synthesis by chromium compounds. J Toxicol Environ Health
15(2):237–244.
Nishioka, H. (1975) Mutagenic activities of metal compounds in bacteria. Mutat Res 31(3):185–189.
Norseth, T; Alexander, J; Aaseth, J; et al. (1982) Biliary excretion of chromium in the rat: a role of glutathione.
Acta Pharmacol Toxicol (Copenh) 51(5):450–455.
Nriagu, JO. (1988) Production and uses of chromium. In: Nriagu, J.O.; Nieboer, E. (eds) Chromium in natural and
human environments. John Wiley and Sons, New York, pp. 81–105.
NRC (National Research Council). (1983) Risk assessment in the federal government: managing the process.
Washington, DC: National Academy Press.
NRC. (2002) Bioavailability of contaminants in soils and sediments: Processes, tools and applications. National
Academies Press: Washington D.C.
NTP (National Toxicology Program). (1996a) Final report on the reproductive toxicity of potassium dichromate
(hexavalent) (CAS No. 7778-50-9) administered in diet to BALB/c mice. National Institute of Environmental
Health Sciences, National Toxicology Program. PB97125363.
NTP. (1996b) Final report on the reproductive toxicity of potassium dichromate (hexavalent) (CAS No. 7778-50-9)
administered in diet to SD rats. National Institute of Environmental Health Sciences, National Toxicology Program.
PB97125355.
NTP. (1997) Final report on the reproductive toxicity of potassium dichromate (CAS No. 7778-50-9) administered
in diet to BALB/c mice. National Institute of Environmental Health Sciences, National Toxicology Program.
PB97144919.
NTP. (2007) NTP technical report on the toxicity studies of sodium dichromate dihydrate (CAS No. 7789-12-0)
administered in drinking water to male and female F344/N rats and B6C3F1 mice and male BALB/c and am3-
C57BL/6 mice. Washington, DC: National Toxicology Program. Toxicity Report Series Number 72. Available
online at http://ntp.niehs.nih.gov/ntp/htdocs/ST_rpts/TOX72.pdf (accessed January 29, 2008).
NTP. (2008) NTP technical report on the toxicology and carcinogenesis studies of sodium dichromate dihydrate
(CAS No. 7789-12-0) in F344/N rats and B6C3F1 mice (drinking water studies). Washington, DC: National
Toxicology Program; NTP TR 546. Available online at http://ntp.niehs.nih.gov/files/546_web_FINAL.pdf
(accessed January 29, 2008).
Oberly, TJ; Piper, CE; McDonald, DS. (1982) Mutagenicity of metal salts in the L5178Y mouse lymphoma assay. J
Toxicol Environ Health 9(3):367–376.
O'Brien, TJ; Ceryak, S; Patierno, SR. (2003) Complexities of chromium carcinogenesis: role of cellular response,
repair and recovery mechanisms. Mutat Res 533(1–2):3–36.
O’Flaherty, E.J. (1991a) Physiologically based models for bone-seeking elements. I. Rat skeletal and bone growth.
Toxicol Appl Pharmacol 111:299 – 312.
O’Flaherty, E.J. (1991b) Physiologically based models for bone-seeking elements. II. Kinetics of lead disposition
in rats. Toxicol Appl Pharmacol 111:313 – 331.
O'Flaherty, EJ. (1993) Physiologically based models for bone-seeking elements. IV. Kinetics of lead disposition in
humans. Toxicol Appl Pharmacol 118(1):16–29.
O’Flaherty, EJ. (1995) Physiologically based models for bone-seeking elements. V. Lean absorption and disposition
in childhood. Toxicol Appl Pharmacol 131(2):297–308.
O’Flaherty, EJ. (1996) A physiologically based model of chromium kinetics in the rat. Toxicol Appl Pharmacol
138(1):54–64.
O’Flaherty, EJ; Kerger, BD; Hays, SM; et al. (2001) A physiologically based model for the ingestion of trivalent
chromium and hexavalent chromium by humans. Toxicol Sci 60(2):196–213.
O’Flaherty, E.J.; Radike, M.J. (1991) Pharmacokinetic modeling of trivalent and hexavalent chromium based on
ingestion and inhalation of soluble chromium compounds. Armstrong Laboratory, Wright-Patterson Air Force Base,
Dayton, OH. Final report. AL-TR-1991-0162.
Ohno, H; Hanaoka, F; Yamada, M. (1982) Inducibility of sister-chromatid exchanges by heavy-metal ions. Mutat
Res 104(1-3):141–145.
Ohsaki, Y; Abe, S; Kimura, K; et al. (1978) Lung cancer in Japanese chromate workers. Thorax 33:372-374.
Olivier, P; Marzin, D. (1987) Study of the genotoxic potential of 48 inorganic derivatives with the SOS chromotest.
Mutat Res 189(3):263–269.
O'Neil, MJ; Heckelman, PE; Koch, CB; et al. (2006) Chromium and compounds. In: O'Neil, MJ; Heckelman, PE;
Koch, CB; et al.; eds. The Merck index. Whitehouse Station, NJ: Merck & Co. Inc., pp. 86, 270, 371–372, 937,
1315–1316.
Quivryn, G.; Goulart, M.; Messer, J.; Zhitkovich, A. (2001) Reduction of Cr(VI) by cysteine : Significance in
human lymphocytes and formation of DNA damage in reactions with variable reduction rates. Mol Cell Biochem
222:107 – 118.
Page, BJ; Loar, GW. (2004) Chromium compounds. In: Kirk-Othmer encyclopedia of chemical technology. New
York, NY: John Wiley & Sons, pp. 526–571.
Partington, CN. (1950) Acute poisoning with potassium bichromate. Br Med J 2(4688):1097–1098.
Paschin, YV; Toropzev, SN. (1982) Chromosome damage induced in vivo by heavy metal ion detected by indirect
testing. Acta Biol Acad Sci Hung 33(4):419–422.
Paschin, YV; Zacepilova, TA; Kozachenko, VI. (1982) Induction of dominant lethal mutations in male mice by
potassium dichromate. Mutat Res 103(3-6):345–347.
Paschin, YV; Kozachenko, VI; Sal'nikova, LE. (1983) Differential mutagenic response at the HGPRT locus in V79
and CHO Chinese hamster cells after treatment with chromate. Mutat Res 122(3-4):361–365.
Patlolla, A; Armstrong, N; Tchounwou, PB. (2008) Cytogenetic evaluation of potassium dichromate toxicity in bone
marrow cells of Sprague-Dawley rats. In: Collery P; Maymard, I; Theophanides, T; et al., eds. Metal ions in biology
and medicine. Paris: John Libbey Eurotext, pp. 353–358.
Paustenbach, DJ; Hays, SM; Brien, BA; et al. (1996) Observation of steady state in blood and urine following
human ingestion of hexavalent chromium in drinking water. J Toxicol Environ Health 49(5):453–461.
Paustenbach, DJ; Panko, JM; Fredrick, MM; Finley, BL; Proctor, DM. (1997) Urinary chromium as a biological
maker of environmental exposure: What are the limitations? Reg Toxicol Pharm 26:S23–S34.
Paustenbach, DJ; Finley, BL; Mowat, FS; et al. (2003) Human health risk and exposure assessment of hexavalent
chromium in tap water. J Toxicol Environ Health A 66(14):1295–1339.
Payne, WW. (1960a) The role of roasted chromite ore in the production of cancer. Arch Environ Health 1:20-26.
Payne, WW. (1960b) Production of cancers in mice and rats by chromium compounds. Arch Ind Health 21:530-535.
Pechova, A.; Pavlata, L. (2007) Chromium is an essential nutrient: A review. Veterinari Medicina 52(1):1 – 18.
Pellerin, C; Booker, SM. (2000) Reflections of hexavalent chromium. Environ Health Perspect 108(9):A402–A407.
Peterson-Roth, E; Reynolds, M; Quievryn, G; et al. (2005) Mismatch repair proteins are activators of toxic
responses to chromium-DNA damage. Mol Cell Biol 25(9):3596–3607.
Petrilli, F.L.; de Flora, S. (1982) Interpretations on chromium mutagenecity and carcinogenicity. In: Sorsa, M.;
Vainio, H. (Eds.) Mutagens in our environment. New York, Alan R. Liss, Inc. pp. 453 – 464
Petrilli, FL; DeFlora, S. (1977) Toxicity and mutagenicity of hexavalent chromium on Salmonella typhimurium.
Appl Environ Microbiol 33:805–809.
Petrilli, FL; Rossi, GA; Camoirano, A; et al. (1986) Metabolic reduction of chromium by alveolar macrophages and
its relationships to cigarette smoke. J Clin Invest 77(6):1917–1924.
Plessi, M; Monzani, A. (1990) Survey of total and bioavailable chromium in grain and cereal by atomic absorption
spetrophotometry. J Assoc Off Anal Chem 73(5):7987-800.
Pool-Zobel, BL; Lotzmann, N; Knoll, M; et al. (1994) Detection of genotoxic effects in human gastric and nasal
mucosa cells isolated from biopsy samples. Environ Mol Mutagen 24(1):23–45.
Pratt, PF; Myers, CR. (1993) Enzymatic reduction of hexavalent chromium by human hepatic microsomes.
Carcinogenesis 14(10):2051–2057.
Quinteros, FA; Poliandri, AH; Machiavelli, LI; et al. (2007) In vivo and in vitro effects of chromium VI on anterior
pituitary hormone release and cell viability. Toxicol Appl Pharmacol 218(1):79–87.
Rafael, AI; Almeida, A; Santos, P; et al. (2007) A role for transforming growth factor-beta apoptotic signaling
pathway in liver injury induced by ingestion of water contaminated with high levels of Cr(VI). Toxicol Appl
Pharmacol 224(2):163–173.
Rai, D; Eary, LE; Zachara, JM. (1989) Environmental chemistry of chromium. The Science of the Total
Environment 86:15 –23.
Reichelderfer, TE. (1968) Accidental death of an infant caused by ingestion of ammonium dichromate. South Med J
61:96–97.
Rodriguez-Arnaiz, R; Martinez, RFM. (1986) Genetic effects of potassium dichromate and chromium trioxide in
Drosophila melanogaster. Cytologia (Tokyo) 51:421–425.
Rowbotham, AL; Levy, LS; Shuker, LK. (2000) Chromium in the environment: An evaluation of exposure of the
UK general population and possible adverse health effects. J Toxicol Environ Health B Crit Rev 3(3):145-178.
Royle, H. (1975) Toxicity of chromic acid in the chromium plating industry. Environ Res 10:141-163.
Ryden, E; Ekstrom, C; Hellmer, L; et al. (2000) Comparison of the sensitivities of Salmonella typhimurium strains
TA102 and TA2638A to 16 mutagens. Mutagenesis 15(6):495–502.
Salnikow, K; Zhitkovich, A. (2008) Genetic and epigenetic mechanisms in metal carcinogenesis and
cocarcinogenesis: nickel, arsenic and chromium. Chem Res Toxicol 21:28–44.
Salnikow, K; Zhitkovich, A; Costa, M. (1992) Analysis of the binding sites of chromium to DNA and protein in
vitro and in intact cells. Carcinogenesis 13(12):2341–2346.
Salnikov, K.; Zhitkovich, A. (2008) Genetic and epigenetic mechanisms in metal carcinogenesis and
cocarcinogenesis: Nickel, arsenic, and chromium. Chem Res Toxicol 21(1):28 – 44.
Sander, M; Cadet, J; Casciano, DA; et al. (2005) Proceedings of a workshop on DNA adducts: biological
significance and applications to risk assessment Washington, DC, April 13-14, 2004. Toxicol Appl Pharmacol
208:1–20.
Sano, T; Mitohara, I. (1978) Occupational cancer among chromium workers. Jpn J Chest Dis 37(2):90-101.
Sarto, F; Cominato, I; Bianchi, V; et al. (1982) Increased incidence of chromosomal aberrations and sister chromatid
exchanges in workers exposed to chromic acid (CrO3) in electroplating factories. Carcinogenesis 3(9):1011–1016.
Sarto, F; Tomanin, R; Giacomelli, L; et al. (1990) The micronucleus assay in human exfoliated cells of the nose and
mouth: application to occupational exposures to chromic acid and ethylene oxide. Mutat Res 244:345–351.
Saryan, LA; Reedy, M. (1988) Chromium determinations in a case of chromic acid ingestion. J Anal Toxicol
12(3):162–164.
Satoh, K; Fukuda, Y; Torii, K; et al. (1981) Epidemiologic study of workers engaged in the manufacture of
chromium compounds. J Occup Med 23(12):835-838.
Sayato, Y; Nakamuro, K; Matsui, S; et al. (1980) Metabolic fate of chromium compounds. I. Comparative behavior
of chromium in rat administered with NA251Cr4 and 51CrCl3. J Pharmacobiodyn 3(1):17–23.
Sekihashi, K; Sasaki, T; Yamamoto, A; et al. (2001) A comparison of intraperitoneal and oral gavage administration
in comet assay in mouse eight organs. Mutat Res 493(1-2):39–54.
Semple, KT; Doick, KJ; Jones, KC; et al. (2004) Defining bioavailability and bioaccessibility of contaminated soil
and sediment is complicated. Environ Sci Technol. Jun 15; 38(12):228A–231A.
Sen, P; Conway, K; Costa, M. (1987) Comparison of the localization of chromosome damage induced by calcium
chromate and nickel compounds. Cancer Res 47(8):2142–2147.
Seo, KS; Lee, CS. (1993) Mutagenicity of metal compounds using Escherichia. Misaengmul Hakhoechi 31(6):527–
531.
Shanker, AK; Cervantes, C; Loza-Tavera, H; et al. (2005) Chromium toxicity in plants. Environ Int 31:739–753.
Sharma, BK; Singhal, PC; Chugh, KS. (1978) Intravascular haemolysis and acute renal failure following potassium
dichromate poisoning. Postgrad Med J 54(632):414–415.
Shi, X; Dalal, NS. (1990) NADPH-dependent flavoenzymes catalyze one electron reduction of metal ions and
molecular oxygen and generate hydroxyl radicals. FEBS Lett 276(1–2):189–191.
Shi, X; Mao, Y; Knapton, AD; et al. (1994) Reaction of Cr(VI) with ascorbate and hydrogen peroxide generates
hydroxyl radicals and causes DNA damage: role of a Cr(IV)-mediated Fenton-like reaction. Carcinogenesis
15(11):2475–2478.
Shi, X; Ding, M; Ye, J; et al. (1999) Cr(IV) causes activation of nuclear transcription factor κB, DNA strand breaks
and dG hydroxylation via free radical reactions. J Inorg Biochem 75:37–44.
Shindo, Y; Toyoda, Y; Kawamura, K; et al. (1989) Micronucleus test with potassium chromate(VI) administered
intraperitoneally and orally to mice. Mutat Res 2223(4):403–406.
Shupack, SI. (1991) The chemistry of chromium and some resulting analytical problems. Environ Hlth Perspect
92:7–11.
Singh, I. (1983) Induction of reverse mutation and mitotic gene conversion by some metal compounds in
Saccharomyces cerevisiae. Mutat Res 117(1–2):149–152.
Smith, AH. (2008) Hexavalent chromium, yellow water, and cancer: a convoluted saga. Epidemiology 19(1):24–26.
Snow, ET. (1991) A possible role for chromium(III) in genotoxicity. Environ Health Perspect 92:75–81.
Sora, S; Agostoni Carbone, ML; Pacciarini, M; et al. (1986) Disomic and diploid meiotic products induced in
Saccharomyces cerevisiae by the salts of 27 elements. Mutagenesis 1(1):21–28.
Sorahan, T; Burgess, DC; Waterhouse, JA. (1987) A mortality study of nickel/chromium platers. Br J Ind Med
44:250-258.
Spano, MA; Frei, H; Wurgler, FE; et al. (2001) Recombinagenic activity of four compounds in the standard and
high bioactivation crosses of Drosophila melanogaster in the wing spot test. Mutagenesis 16(5):385–394.
Standeven, AM; Wetterhahn, KE. (1989) Chromium(VI) toxicity: uptake, reduction, and DNA damage. J Am Coll
Toxicol 8(7):1275–1283.
Standeven, A.M.; Wetterhahn, K.E. (1991) Ascorbate is the principal reductant of chromium(VI) in rat liver and
kidney ultrafiltrates. Carcinogenesis 12:1733 – 1737.
Standeven, A.M.; Wetterhahn, K.E. (1992) Ascorbate is the principal reductant of chromium(VI) in rat lung
ultrafiltrates and cytosols, and mediates chromium-DNA binding in vitro. Carcinogenesis 13:1319 – 1324.
Stearns, DM; Wetterhahn, KE. (1994) Reaction of chromium(VI) with ascorbate produces chromium(V),
chromium(IV), and carbon-based radicals. Chem Res Toxicol 7:219–230.
Stearns, DM; Courtney, KD; Giangrade, PH; et al. (1994) Hexavalent chromium reduction by ascorbate: role of
reactive intermediates in DNA damage in vitro. Environ Health Perspect 102(Suppl 3):21–25.
Stearns, D.M. (1999) Is chromium a trace essential metal? (2000) BioFactors 11:149 – 162.
Stella, M; Montaldi, A; Rossi, R; et al. (1982) Clastogenic effects of chromium on human lymphocytes in vitro and
in vivo. Mutat Res 101(2):151–164.
Stout, MD; Herbert, RA; Kissling, GE; et al. (2009) Hexavalent chromium is carcinogenic to F344/N rats and
B6C3F1 mice after chronic exposure. Environ Health Perspect 117(5):716–722.
Subramanian, S; Rajendiran, G; Sekhar, P; et al. (2006) Reproductive toxicity of chromium in adult bonnet monkeys
(Macaca radiata Geoffrey). Reversible oxidative stress in the semen. Toxicol Appl Pharmacol 215(3):237–249.
Sugden, KD; Martin BD. (2002) Guanine and 7,8-dihydro-8-oxo-guanine-specific oxidation in DNA by
chromium(V). Environ Health Perspect 110(Suppl 5):725–728.
Sugden, KD; Stearns, DM. (2000) The role of chromium(V) in the mechanism of chromate-induced oxidative DNA
damage and cancer. J Environ Pathol Toxicol Oncol 19(3):215–230.
Sugiyama, M; Ando, A; Nakao, K; et al. (1989) Influence of vitamin B2 on formation of chromium(V), alkali-labile
sites, and lethality of sodium chromate(VI) in Chinese hamster V-79 cells. Cancer Res 49:6180–6184.
Sutherland, JE; Zhitkovich, A; Kluz, T; et al. (2000) Rats retain chromium in tissues following chronic ingestion of
drinking water containing hexavalent chromium. Biol Trace Elem Res 74:41–53.
Suzuki, Y; Fukuda, K. (1990) Reduction of hexavalent chromium by ascorbic acid and glutathione with special
reference to the rat lung. Arch Toxicol 93:9 – 14.
Tagliari, KC; Cecchini, R; Vargas, VMF. (2004) Mutagenicity of chromium (VI) using the Salmonella
microsuspension bioassay. Rev Bras Toxicol 17(2):45–50.
Takahashi, Y; Kondo, K; Hirose, T; et al. (2005) Microsatellite instability and protein expression of the DNA
mismatch repair gene, hMLH1, of lung cancer in chromate-exposed workers. Mol Carcinog 42(3):150–158.
Taylor, FH. (1966) The relationship of mortality and duration of employment as reflected by a cohort of chromate
workers. Am J Public Health 56(2):218-229.
Thompson, RC; Hollis, OL. (1958) Irradiation of the gastrointestinal tract of the rat by ingested ruthenium-106. Am
J Physiol 194(2):308–312.
Tkaczyk, C.; Huk, O.L.; Mwale, F.; Antoniou, J.; Zukor, D.J.; Petit, A.; Tabrizian, M. (2010) Investigation of the
binding of Cr(III) complexes to bovine and human serum proteins: A proteomic approach. J Biomed Mater Res
94A:214 – 222.
Todd, GE. (1962) Tobacco manufacturer's standing committee research papers. No. 1. Statistics of Smoking in the
United Kingdom, 3rd ed. Tobacco Research Council, London.
Trivedi, B; Saxena, DK; Murthy, RC; et al. (1989) Embryotoxicity and fetotoxicity of orally administered
hexavalent chromium in mice. Reprod Toxicol 3(4):275–278.
Trzeciak, A; Kowalik, J; Malecka-Panas, E; et al. (2000) Genotoxicity of chromium in human gastric mucosa cells
and peripheral blood lymphocytes evaluated by the single cell gel electrophoresis (comet assay). Med Sci Monit
6(1):24–29.
Tsapakos, MJ; Hampton, TH; Wetterhahn, KE. (1983) Hexavalent chromium-induced DNA lesions and chromium
distribution in rat kidney, liver, and lung. Cancer Res 43(12 Pt 1):5662–5667.
Ueno, S; Kashimoto, T; Susa, N; et al. (2001) Detection of dichromate(VI)-induced DNA strand breaks and
formation of paramagnetic chromium in multiple mouse organs. Toxicol Appl Pharmacol 170(1):56–62.
U.S. EPA (Environmental Protection Agency). (1984) Health assessment document for chromium. Final report.
Cincinnati, OH: Environmental Criteria and Assessment Office. EPA600883014F.
U.S. EPA. (1986a) Guidelines for the health risk assessment of chemical mixtures. Federal Register 51(185):34014-
34025. Available online at http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (1986b) Guidelines for mutagenicity risk assessment. Federal Register 51(185):34006-34012. Available
online at http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (1988) Recommendations for and documentation of biological values for use in risk assessment.
Prepared by the Environmental Criteria and Assessment Office, Office of Health and Environmental Assessment,
Cincinnati, OH for the Office of Solid Waste and Emergency Response, Washington, DC; EPA 600/6-87/008.
Available online at http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (1991) Guidelines for developmental toxicity risk assessment. Federal Register 56(234):63798-63826.
Available online at http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (1994a) Interim policy for particle size and limit concentration issues in inhalation toxicity studies.
Federal Register 59(206):53799. Available online at http://www.epa.gov/iris/backgrd.html (accessed April 20,
2010).
U.S. EPA. (1994b) Methods for derivation of inhalation reference concentrations and application of inhalation
dosimetry. Office of Research and Development, Washington, DC; EPA/600/8-90/066F. Available online at
http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (1995) Use of the benchmark dose approach in health risk assessment. Risk Assessment Forum,
Washington, DC; EPA/630/R-94/007. Available online at
http://cfpub.epa.gov/ncea/raf/recordisplay.cfm?deid=42601 (accessed March 11, 2010).
U.S. EPA. (1996) Guidelines for reproductive toxicity risk assessment. Federal Register 61(212):56274–56322.
Available online at http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (1998) Guidelines for neurotoxicity risk assessment. Federal Register 63(93):26926–26954. Available
online at http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (2000a) Science policy council handbook: risk characterization. Office of Science Policy, Office of
Research and Development, Washington, DC; EPA 100-B-00-002. Available online at
http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (2000b) Benchmark dose technical guidance document. External review draft. Risk Assessment Forum,
Washington, DC; EPA/630/R-00/001. Available online at http://www.epa.gov/iris/backgrd.html (accessed April 20,
2010).
U.S. EPA. (2000c) Supplementary guidance for conducting for health risk assessment of chemical mixtures. Risk
Assessment Forum, Washington, DC; EPA/630/R-00/002. Available online at http://www.epa.gov/iris/backgrd.html
(accessed April 20, 2010).
U.S. EPA. (2002) A review of the reference dose and reference concentration processes. Risk Assessment Forum,
Washington, DC; EPA/630/P-02/0002F. Available online at http://www.epa.gov/iris/backgrd.html (accessed April
20, 2010).
U.S. EPA. (2005a) Guidelines for carcinogen risk assessment. Risk Assessment Forum, Washington, DC;
EPA/630/P-03/001B. Available online at http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (2006a) Science policy council handbook: peer review. Third edition. Office of Science Policy, Office of
Research and Development, Washington, DC; EPA/100/B-06/002. Available online at
http://www.epa.gov/iris/backgrd.html (accessed April 20, 2010).
U.S. EPA. (2006b) A Framework for Assessing Health Risk of Environmental Exposures to Children. National
Center for Environmental Assessment, Washington, DC, EPA/600/R-05/093F. Available online at
http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=158363 (accessed March 11, 2010).
Vaglenov, A; Nosko, M; Georgieva, R; et al. (1999) Genotoxicity and radioresistance in electroplating workers
exposed to chromium. Mutat Res 446(1):23–34.
Valko, M; Rhodes, CJ; Moncol, J; et al. (2006) Free radicals, metals and antioxidants in oxidative stress-induced
cancer. Chem Biol Interact 160:1–40.
Vashishat, RK; Vasudeva, M. (1987) Genotoxic potential of chromium salts in Saccharomyces cerevisiae. Ind J
Microbiol 27:35–36.
Venier, P; Montaldi, A; Majone, F; et al. (1982) Cytotoxic, mutagenic and clastogenic effects of industrial
chromium compounds. Carcinogenesis 3(11):1331–1338.
Venier, P; Gava, C; Zordan, M; et al. (1987) Interactions of chromium with nitrilotriacetic acid (NTA) in the
induction of genetic effects in bacteria. Toxicol Environ Chem 14(3):201–218.
Venitt, S; Levy, LS. (1974) Mutagenicity of chromates in bacteria and its relevance to chromate carcinogenesis.
Nature 250:493–495.
Vincent, J.B. (2004) Recent developments in the biochemistry of chromium(III). Biol Trace Elem Res 99(1 – 3):1
– 16.
Vitale, RJ; Mussoline, GR; Rinehimer, KA. (1997) Environmental monitoring of chromium in air, soil and water.
Regul Toxicol Pharmacol 26(1 Pt 2):S80-5.
Voitkun, V; Zhitkovich, A; Costa, M. (1998) Cr(III)-mediated crosslinks of glutathione or amino acids to the DNA
phosphate backbone are mutagenic in human cells. Nucleic Acids Res 26(8):2024–2030.
Vyskocil, A; Viau, C; Cizkova, M; et al. (1993) Kidney function in male and female rats chronically exposed to
potassium dichromate. J Appl Toxicol 13(5):375–376.
Wang, XF; Xing, ML; Shen, Y; et al. (2006) Oral administration of Cr(VI) induced oxidative stress, DNA damage
and apoptotic cell death in mice. Toxicology 228(1):16–23.
Wang, S; Chen, F; Zhang, Z; et al. (2004) NF-kB prevents cells from undergoing Cr(VI)-induced apoptosis. Mol
Cell Biochem 255:129-137.
Watanabe, K; Sakamoto, K; Sasaki, T. (1998) Comparisons on chemically-induced mutation among four bacterial
strains, Salmonella typhimurium TA102 and TA2638, and Escherichia coli WP2/pKM101 and WP2 uvrA/pKM101:
collaborative study II. Mutat Res 412(1):17–31.
Weber, H. (1983) Long-term study of the distribution of soluble chromate-51 in the rat after a single intratracheal
administration. J Toxicol Environ Health 11(4-6):749–764.
Wiegand, HJ; Ottenwalder, H; Bolt, HM. (1985) Fast uptake kinetics in vitro of 51Cr(VI) by red blood cells of man
and rat. Arch Toxicol 57(1):31–34.
Wiegand, HJ; Ottenwalder, H; Bolt, HM. (1987) Bioavailability and metabolism of hexavalent chromium
compounds. Toxicol Environ Chem 14:263–275.
Wild, D. (1978) Cytogenetic effects in the mouse of 17 chemical mutagens and carcinogens evaluated by the
micronucleus test. Mutat Res 56(3):319–327.
Wise, JP, Sr.; Wise, SS; Little, JE. (2002) The cytotoxicity and genotoxicity of particulate and soluble hexavalent
chromium in human lung cells. Mutat Res 517(1–2):221–229.
Wise, SS; Holmes, AL; Ketterer, ME; et al. (2004) Chromium is the proximate clastogenic species for lead
chromate-induced clastogenicity in human bronchial cells. Mutat Res 560(1):79–89.
Wise, SS; Holmes, AL; Wise, JP, Sr. (2006a) Particulate and soluble hexavalent chromium are cytotoxic and
genotoxic to human lung epithelial cells. Mutat Res 610(1–2):2–7.
Wise, SS; Holmes, AL; Xie, H; et al. (2006b) Chronic exposure to particulate chromate induces spindle assembly
checkpoint bypass in human lung cells. Chem Res Toxicol 19(11):1492–1498.
Wise, SS; Holmes, AL; Wise, JP, Sr. (2008) Hexavalent chromium-induced DNA damage and repair mechanisms.
Rev Environ Health 23(1):39–57.
Witmer, C.M.; Harris, R/; Shupack, S.I. (1991) Oral bioavailability of chromium from a specific site. Environ
Health Perspect 92:105 – 110.
Witmer, C.M.; Park, H-S. (1989) Mutagenicity and disposition of chromium. Sci. Total Environ 86: 131 – 148.
Wu, FY; Tsai, FJ; Kuo, HW; et al. (2000) Cytogenetic study of workers exposed to chromium compounds. Mutat
Res 464(2):289–296.
Wu, FY; Wu, WY; Kuo, HW; et al. (2001) Effect of genotoxic exposure to chromium among electroplating workers
in Taiwan. Sci Total Environ 279(1–3):21–28.
Xu, J; Bubley, GJ; Detrick, B; et al. (1996) Hexavalent chromium treatment of normal human lung cells results in
guanine-specific DNA polymerase arrest, DNA-DNA cross-links and S-phase blockade of cell cycle.
Carcinogenesis 17(7):1511–1517.
Yamamoto, A; Kohyama, Y; Hanawa, T. (2002) Mutagenicity evaluation of forty-one metal salts by the umu test. J
Biomed Mater Res 59(1):176–183.
Ye, J; Wang, S; Leonard, SS; et al. (1999) Role of reactive oxygen species and p53 in chromium(VI)-induced
apoptosis. J Biol Chem 274(49):34974–34980.
Yousef, MI; El-Demerdash, FM; Kamil, KI; et al. (2006) Ameliorating effect of folic acid on hexavalent chromium-
induced changes in reproductive performance and seminal plasma biochemistry in male rabbits. Reprod Toxicol
21(3):322–328.
Zahid, ZR; Al-Hakkak, ZS; Kadhim, AHH; et al. (1990) Comparative effects of trivalent and hexavalent chromium
on spermatogenesis of the mouse. Toxicol Environ Chem 25:131–136.
Zhang, JD; Li, XL. (1987) Chromium pollution of soil and water in Jinzhou. Zhonghua Yu Fang Yi Xue Za Zhi
21(5):262–264. (Chinese)
Zhang, JD; Li, S. (1997) Cancer mortality in a Chinese population exposed to hexavalent chromium in water. J
Occup Environ Med 39(4):315–319.
Zhang, Q; Kluz, T; Salnikow, K; et al. (2002) Comparison of the cytotoxicity, cellular uptake, and DNA-protein
crosslinks induced by potassium chromate in lymphoblast cell lines derived from three different individuals. Biol
Trace Elem Res 86:11–22.
Zhitkovich, A. (2005) Importance of chromium-DNA adducts in mutagenicity and toxicity of chromium(VI). Chem
Res Toxicol 18(3):3–11.
Zhitkovich, A; Voitkun, V; Costa, M. (1995) Glutathione and free amino acids form stable complexes with DNA
following exposure of intact mammalian cells to chromate. Carcinogenesis 16(4):907–913.
Zhitkovich, A; Song, Y; Quievryn, G; et al. (2001) Non-oxidative mechanisms are responsible for the induction of
mutagenesis by reduction of Cr(VI) with cysteine: role of ternary DNA adducts in Cr(III)-dependent mutagenesis.
Biochemistry 40:549–560.
Zimmering, S; Mason, JM; Valencia, R; et al. (1985) Chemical mutagenesis testing in Drosophila. II. Results of 20
coded compounds tested for the National Toxicology Program. Environ Mutagen 7(1):87–100.
Table B-1. Incidence data for nonneoplastic lesions from all treatment
groups of female F344/N rats and male and female B6C3F1 mice exposed to
sodium dichromate dihydrate in drinking water for 2 years (NTP, 2008)
Dose
(mg hexavalent chromium/kg-d)
0 0.24 0.94 2.4 7.0
Female rats
Liver: chronic inflammation 12/50 21/50a 28/50b 35/50b 39/50b
Dose
(mg hexavalent chromium/kg-d)
0 0.38 0.91 2.4 5.9
Male mice
Duodenum: diffuse epithelial 0/50 11/50b 18/50b 42/50b 32/50a
hyperplasia
Mesenteric lymph node: histiocytic 14/47 38/47b 31/49b 32/49b 42/46a
cellular infiltration
Dose
(mg hexavalent chromium/kg-d)
0 0.38 1.4 3.1 8.7
Female mice
Duodenum: diffuse epithelial 0/50 16/50b 35/50b 31/50b 42/50b
hyperplasia
Mesenteric lymph node: histiocytic 3/46 29/48b 26/46b 40/50b 42/50b
cellular infiltration
Liver: histiocytic cellular infiltration 2/49 15/50b 23/50b 32/50b 45/50b
Pancreas: acinus, cytoplasmic 0/48 6/50a 6/49a 14/50b 32/50b
alteration
a
Significantly different (p ≤ 0.05) from the control group by Dunn’s or Shirley’s test.
b
Significantly different (p ≤ 0.01) from the control group by Dunn’s or Shirley’s test.
BMD10 BMDL10
Model (mg/kg-d) (mg/kg-d) χ2 p-value AIC
a
Gamma 0.51 0.37 0.04 317.97
Logistic 0.84 0.65 0.01 321.45
Log-logisticb 0.22 0.14 0.37 312.57
Multistagec 0.51 0.37 0.04 317.97
Probit 0.88 0.70 0.01 321.80
Log-probitb 0.89 0.61 0.01 320.86
Quantal linear 0.51 0.37 0.04 317.97
Weibulla 0.51 0.37 0.04 317.97
a
Restrict power ≥1.
b
Slope restricted to >1.
c
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 3-degree polynomial is reported.
Log-Logistic
0.9
0.8
0.7
Fraction Affected
0.6
0.5
0.4
0.3
0.2
0 1 2 3 4 5 6 7
dose
13:34 04/08 2008
BMDs and BMDLs indicated are associated with a 10% extra risk, and are in
units of mg hexavalent chromium/kg-day.
full dataset (Table B-3). In order to achieve an adequately fitting model, the highest dose was
dropped. This is determined to be appropriate, as the area of concern is with the low-dose region
of the response curve. After dropping the highest dose, the gamma, log-logistic, multistage, log-
probit, quantal linear, and Weibull models provided adequate fits to the data (χ2 p-value > 0.1).
Comparing across models, a better fit is generally indicated by a lower Akaike’s Information
Criterion (AIC) (EPA, 2000b). As assessed by AIC, the 1-degree polynomial multistage model
provided the best fit to the data (Figure B-2). Based on the multistage model, the BMD
associated with a 10% extra risk was 0.16 mg hexavalent chromium/kg-day and its lower 95%
confidence limit (BMDL) was 0.13 mg hexavalent chromium/kg-day.
BMD10 BMDL10
Model (mg/kg-d) (mg/kg-d) χ2 p-value AIC
All doses
Gammaa 0.31 0.25 0.00 270.99
Logistic 0.90 0.74 0.00 296.25
Log-logisticb 0.15 0.12 0.00 247.93
Multistagec 0.31 0.25 0.00 270.99
Probit 0.90 0.76 0.00 296.18
Log-probitb 0.48 0.36 0.00 274.38
Quantal linear 0.31 0.25 0.00 270.99
Weibulla 0.31 0.25 0.00 270.99
Highest dose dropped (four doses modeled)
Gammaa 0.22 0.14 0.43 167.67
Logistic 0.47 0.39 0.03 177.09
Log-logisticb 0.26 0.15 0.20 169.23
Multistaged 0.16 0.13 0.52 166.34
Probit 0.45 0.37 0.04 176.19
Log-probitb 0.28 0.23 0.33 167.41
Quantal linear 0.16 0.13 0.52 166.34
Weibulla 0.22 0.14 0.47 167.50
a
Restrict power ≥1.
b
Slope restricted to >1.
c
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 3-degree polynomial is reported.
d
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; degree polynomial = 1.
1
Multistage
0.8
0.6
Fraction Affected
0.4
0.2
0
BMDL BMD
dose
15:45 04/08 2008
BMDs and BMDLs indicated are associated with a 10% extra risk, and are in
units of mg hexavalent chromium/kg-day.
Histiocytic cellular infiltration of the mesenteric lymph nodes in male mice. As assessed
by the χ2 goodness-of-fit statistic, none of the models provided an adequate fit (χ2 p-value ≥ 0.1)
to the full dataset (Table B-4). In order to achieve a statistically fit model, the highest dose was
dropped. This is determined to be appropriate, as the area of concern is with the low-dose region
of the response curve. Dropping the highest dose did not result in adequately fitting models, nor
did dropping the two highest doses. This dataset is considered not suitable for BMD modeling.
BMD10 BMDL10
Model (mg/kg-d) (mg/kg-d) χ2 p-value AIC
All doses
Gammaa 0.38 0.26 0.00 285.94
Logistic 0.53 0.39 0.00 286.38
Log-logisticb 0.16 0.08 0.00 284.48
Multistagec 0.43 0.26 0.00 287.88
Probit 0.56 0.43 0.00 286.35
Log-probitb 0.83 0.52 0.00 289.36
Quantal linear 0.38 0.26 0.00 285.94
Weibulla 0.38 0.26 0.00 285.94
Highest dose dropped (four doses modeled)
Gammaa 0.47 0.24 0.00 258.50
Logistic 0.61 0.35 0.00 259.04
Log-logisticb 0.21 0.08 0.00 256.81
Multistaged 0.47 0.24 0.00 258.50
Probit 0.63 0.37 0.00 259.08
Log-probitb 1.24 0.56 0.00 261.28
Quantal linear 0.47 0.24 0.00 258.50
Weibulla 0.47 0.24 0.00 258.50
Two highest doses dropped (three doses modeled)
Gammaa 0.11 0.07 0.00 187.77
Logistic 0.17 0.12 0.00 189.97
Log-logisticb 0.05 0.03 0.00 183.77
Multistagee 0.11 0.07 0.00 187.77
Probit 0.17 0.12 0.00 190.12
Log-probitb 0.17 0.11 0.00 190.37
Quantal linear 0.11 0.07 0.00 187.77
Weibulla 0.11 0.07 0.00 187.77
a
Restrict power ≥1.
b
Slope restricted to >1.
c
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 3-degree polynomial is reported.
d
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 2-degree polynomial is reported.
e
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 1-degree polynomial is reported.
BMD10 BMDL10
Model (mg/kg-d) (mg/kg-d) χ2 p-value AIC
All doses
Gammaa 0.34 0.27 0.00 275.34
Logistic 0.88 0.72 0.00 293.17
Log-logisticb 0.12 0.09 0.04 245.54
Multistagec 0.34 0.27 0.00 275.34
Probit 0.93 0.78 0.00 294.03
Log-probitb 0.52 0.38 0.00 279.54
Quantal linear 0.34 0.27 0.00 275.34
Weibulla 0.34 0.27 0.00 275.34
Highest dose dropped (four doses modeled)
Gammaa 0.20 0.16 0.00 213.41
Logistic 0.55 0.46 0.00 236.10
Log-logisticb 0.11 0.08 0.04 200.07
Multistaged 0.20 0.16 0.00 213.41
Probit 0.54 0.45 0.00 235.61
Log-probitb 0.29 0.24 0.00 220.04
Quantal linear 0.20 0.16 0.00 213.41
Weibulla 0.20 0.16 0.00 213.41
Two highest doses dropped (three doses modeled)
a
Gamma 0.12 0.09 0.87 126.06
Logistic 0.34 0.27 0.00 141.77
Log-logisticb 0.12 0.06 1.00 127.77
e
Multistage 0.12 0.09 0.87 126.06
Probit 0.32 0.26 0.00 140.65
Log-probitb 0.20 0.16 0.48 127.17
Quantal linear 0.12 0.09 0.87 126.06
Weibulla 0.12 0.09 0.87 126.06
a
Restrict power ≥1.
b
Slope restricted to >1.
c
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 3-degree polynomial is reported.
d
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 2-degree polynomial is reported.
e
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 1-degree polynomial is reported.
Multistage
0.8
0.7
0.6
Fraction Affected
0.5
0.4
0.3
0.2
0.1
0
BMDL BMD
dose
09:36 04/09 2008
BMDs and BMDLs indicated are associated with a 10% extra risk, and are in
units of mg hexavalent chromium/kg-day.
BMD10 BMDL10
Model (mg/kg-d) (mg/kg-d) χ2 p-value AIC
All doses
Gammaa 0.41 0.30 0.00 282.46
Logistic 0.77 0.61 0.00 290.18
Log-logisticb 0.09 0.06 0.00 263.55
Multistagec 0.41 0.30 0.00 282.46
Probit 0.85 0.69 0.00 291.41
Log-probitb 0.68 0.47 0.00 285.85
Quantal linear 0.41 0.30 0.00 282.46
Weibulla 0.41 0.30 0.00 282.46
Highest dose dropped (four doses modeled)
Gammaa 0.20 0.15 0.00 224.84
Logistic 0.40 0.33 0.00 230.81
Log-logisticb 0.07 0.05 0.00 215.19
Multistaged 0.20 0.15 0.00 224.84
Probit 0.40 0.34 0.00 230.85
Log-probitb 0.37 0.24 0.00 231.76
Quantal linear 0.20 0.15 0.00 224.84
Weibulla 0.20 0.15 0.00 224.84
Two highest doses dropped (three doses modeled)
a
Gamma 0.14 0.10 0.00 172.32
Logistic 0.31 0.24 0.00 178.99
Log-logisticb 0.07 0.04 0.00 164.47
Multistagee 0.14 0.10 0.00 172.32
Probit 0.30 0.23 0.00 178.74
Log-probitb 0.21 0.15 0.00 178.11
Quantal linear 0.14 0.10 0.00 172.32
Weibulla 0.14 0.10 0.00 172.32
a
Restrict power ≥1.
b
Slope restricted to >1.
c
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 3-degree polynomial is reported.
d
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 2-degree polynomial is reported.
e
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 1-degree polynomial is reported.
0.1) to the data (Table B-7). Based on the log-logistic model, the BMD associated with a 10%
extra risk was 0.17 mg hexavalent chromium/kg-day and its lower 95% confidence limit
(BMDL) was 0.12 mg hexavalent chromium/kg-day (Figure B-4).
Table B-7. BMD10 and BMDL10 values and goodness-of-fit statistics from
models fit to incidence data for histiocytic cellular infiltration in the liver of
female rats exposed to sodium dichromium dihydrate in drinking water for
2 years
BMD10 BMDL10
Model (mg/kg-d) (mg/kg-d) χ2 p-value AIC
a
Gamma 0.35 0.28 0.08 255.40
Logistic 0.85 0.70 0.00 267.56
Log-logisticb 0.17 0.12 0.44 251.36
Multistagec 0.35 0.28 0.08 255.40
Probit 0.88 0.75 0.00 268.64
Log-probitb 0.62 0.48 0.01 260.00
Quantal linear 0.35 0.28 0.08 255.40
Weibulla 0.35 0.28 0.08 255.40
a
Restrict power ≥1.
b
Slope restricted to >1.
c
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; no degree of polynomial
provided a fit, a 3-degree polynomial is reported.
1 Log-Logistic
0.8
Fraction Affected
0.6
0.4
0.2
0
BMDL BMD
0 1 2 3 4 5 6 7 8 9
dose
11:17 04/09 2008
BMDs and BMDLs indicated are associated with a 10% extra risk, and are in
units of mg hexavalent chromium/kg-day.
BMD10 BMDL10
Model (mg/kg-d) (mg/kg-d) χ2 p-value AIC
a
Gamma 0.92 0.72 0.13 206.82
Logistic 2.43 2.03 0.09 211.78
Log-logisticb 0.68 0.52 0.19 205.22
Multistagec 0.92 0.72 0.13 206.82
Probit 2.24 1.89 0.11 210.99
Log-probitb 1.77 1.40 0.11 209.99
Quantal linear 0.92 0.72 0.13 206.82
Weibulla 0.92 0.72 0.13 206.82
a
Restrict power ≥1.
b
Slope restricted to >1.
c
Restrict betas ≥0; lowest degree polynomial (up to n-2) with an adequate fit is reported; a 1-degree polynomial is
reported.
0.8 Log-Logistic
0.7
0.6
0.5
Fraction Affected
0.4
0.3
0.2
0.1
0
BMDL BMD
0 1 2 3 4 5 6 7 8 9
dose
11:41 04/09 2008
BMDs and BMDLs indicated are associated with a 10% extra risk, and are in
units of mg hexavalent chromium/kg-day.
The fit of the multistage model to the incidence of neoplasms in the small intestine of
male mice administered sodium dichromate dihydrate in drinking water for 2 years (NTP, 2008):
====================================================================
Multistage Cancer Model. (Version: 1.7; Date: 05/16/2008)
Input Data File:
M:\ChromiumVI\msc_MALE_MICE_INTESTINAL_TUMORS_NTP_2008_Setting.(d)
Gnuplot Plotting File:
M:\ChromiumVI\msc_MALE_MICE_INTESTINAL_TUMORS_NTP_2008_Setting.plt
Fri Feb 05 09:42:31 2010
====================================================================
Parameter Estimates
AIC: 161.73
BMD = 2.21769
BMDL = 1.16524
BMDU = 3.23024
====================================================================
Multistage Cancer Model. (Version: 1.7; Date: 05/16/2008)
Input Data File:
M:\ChromiumVI\msc_FEMALE_MICE_INTESTINAL_TUMORS_NTP_2008_Setting.(d)
Gnuplot Plotting File:
M:\ChromiumVI\msc_FEMALE_MICE_INTESTINAL_TUMORS_NTP_2008_Setting.plt
Fri Feb 05 09:54:51 2010
====================================================================
Background Beta(1)
Background 1 -0.62
Beta(1) -0.62 1
Parameter Estimates
AIC: 187.701
BMD = 1.33025
BMDL = 1.02757
BMDU = 1.93668