Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2020 Feb 1.
Published in final edited form as: Toxicol Appl Pharmacol. 2018 Dec 21;364:97–105. doi: 10.1016/j.taap.2018.12.010

Toxicokinetics of Benzo[a]pyrene in Humans: Extensive Metabolism as Determined by UPLC-Accelerator Mass Spectrometry Following Oral Micro-Dosing

Erin Madeen a,b,1, Lisbeth K Siddens a,b,c, Sandra Uesugi c, Tammie McQuistan d, Richard A Corley b,e, Jordan Smith b,e, Katrina M Waters b,e, Susan C Tilton a,b, Kim A Anderson a,b, Ted Ognibene f, Kenneth Turteltaub f,g, David E Williams a,c,*
PMCID: PMC6369707  NIHMSID: NIHMS1517550  PMID: 30582946

Abstract

Benzo[a]pyrene (BaP), is a known human carcinogen (International Agency for Research on Cancer (IARC) class 1). The remarkable sensitivity (zepto-attomole 14C in biological samples) of accelerator mass spectrometry (AMS) makes possible, with de minimus risk, pharmacokinetic (PK) analysis following [14C]-BaP micro-dosing of humans. A 46 ng (5 nCi) dose was given thrice to 5 volunteers with minimum 2 weeks between dosing and plasma collected over 72 hours. [14C]-BaPeq PK analysis gave plasma Tmax and Cmax values of 1.25 hours and 29–82 fg/mL, respectively. PK parameters were assessed by non- compartment and compartment models. Intervals between dosing ranged from 20–420 days and had little impact on intra-individual variation. DNA, extracted from peripheral blood mononuclear cells (PBMCs) of 4 volunteers, showed measurable levels (LOD ~ 0.5 adducts/1011 nucleotides) in two individuals 2–3 hours post-dose, approximately three orders of magnitude lower than smokers or occupationally-exposed individuals. Little or no DNA binding was detectable at 48–72 hours. In volunteers the allelic variants CYP1B1*1/*1, *1/*3 or *3/*3 and GSTM1*0/0 or *1 had no impact on [14C]-BaPeq PK or DNA adduction with this very limited sample. Plasma metabolites over 72 hours from two individuals (one CYP1B1*1/*1 and one CYP1B1*3/*3) were analyzed by UPLC-AMS. In both individuals, parent [14C]-BaP was a minor constituent even at the earliest time points and metabolite profiles markedly distinct. AMS, coupled with UPLC, could be used in humans to enhance the accuracy of pharmacokinetics, toxicokinetics and risk assessment of environmental carcinogens.

Keywords: Benzo[a]pyrene, Pharmacokinetics, Polycyclic Aromatic Hydrocarbons, Accelerator Mass Spectrometry, Micro-Dosing, Metabolites

Introduction

Benzo[a]pyrene (BaP), the most studied polycyclic aromatic hydrocarbon (PAH), is a high priority (8th/785) environmental chemical of concern for human exposure according to the Agency for Toxic Substances and Disease Registry (ATSDR, 2013). In preclinical models, BaP exposure results in multiple toxicities including cardiovascular, developmental, immunological and reproductive as well as multiple cancers (International Agency for Research on Cancer (IARC), 2010; World Health Organization (WHO), 1998; U.S. Environmental Protection Agency (EPA), 2017). The EPA (2017) recently modified the oral BaP cancer risk slope factor (an upper bound estimate of risk) from 7.3 to 1 (mg/kg-day)−1 which, based on an estimated daily exposure (non-smoking adult in the U.S.) of 270–750 ng, would correspond to a 3.9×10−6-1.1×10−5 (1 in 93,000 to 1 in 260,000) lifetime excess risk of developing cancer. BaP is used by the EPA as the reference compound for determination of cancer risk for environmental PAH mixtures (Relative Potency Factor (RPF) with BaP=1) (EPA, 2010).

Formation of covalent DNA adducts, following exposure to BaP, has long been used as a biomarker in preclinical models (Godschalk et al., 1997; Ross et al., 1995; Zuo et al., 2014), or in vitro and ex vivo studies with human cells, postmortem or biopsy tissue (Alexandrov et al., 2002; Lodovici et al., 1998; Monien et al., 2015; Schults et al., 2013; Shiilzaki et al., 2017) and human epidemiology studies, primarily with occupationally exposed populations or smokers (Borska et al., 2014; Boysen and Hecht, 2003; Divi et al., 2002; Kriek et al., 1993; Tuominen et al., 2002). The major adduct associated with mutation and carcinogenesis (both rodents and humans) is formed from reaction of (+)-anti-BaP-7,8-dihydrodiol-9,10-epoxide ((+)-anti-BaPDHDE), the most mutagenic of four possible enantiomers) and the N2 position of guanine is the preferred site for adduction (Jernström and Gräslund 1994). In A/J mice, often used in lung cancer studies, BaP-induction of adenomas correlates with time-integrated DNA adducts (Ross et al., 1995). In the Muta Mouse model oral administration of BaP leads to an order of DNA adduction (spleen ≥ liver > lung > glandular stomach ≥ colon > forestomach) distinct from that of target organ mutation sensitivity (colon > forestomach > spleen > glandular stomach > liver = lung) or tumor incidence (forestomach > spleen > lung) (Hakura et al., 1998a; b; 1999; 2000). Zuo et al., (2014) enhanced the prediction of target organs by also incorporating BaP-induced alteration in gene expression. Assessment of DNA adducts in peripheral blood mononuclear cells (PBMCs), a relatively non-invasive approach, of smokers or as a result of occupational exposures yields BaP-DNA adducts in the 1–50 per 108 nucleotide range (Alexandrov et al., 2002; Kriek et al., 1993; Pavanello et al., 2004).

Accelerator mass spectrometry (AMS), due to its remarkable sensitivity to 14C, is finding increasing use in study of environmental carcinogens (Enright et al., 2016; Felton and Turteltabaub, 1994; Ognibene et al., 2004). Human subjects have been dosed with [14C]-labeled 2-amino-3-methylimidazo[4,5-f]quinolone (IQ), 2-amino-3,8-dimethylimidazo[4,5-b]quinoxaline (MeIQx), 2-amino-3-methyl-6-phenylimidazo[4,5-b]pyridine (PhIP), tamoxifen, aflatoxin B1, dibenzo[def,p]chrysene (DBC) and BaP (Brown et al., 2006; Enright et al., 2016; Hummel et al., 2018; Jubert et al., 2009; Lightfoot et al., 2000; Madeen et al., 2015; 2016). The majority of these studies with carcinogens have assayed for target organ DNA binding (Lightfoot et al., 2000; Felton and Turteltaub, 1994) (dosing prior to surgical resection) but some have examined pharmacokinetics (Hummel et al., 2018; Madeen et al., 2015; 2016). The graphite combustion technique allows for measurement of total [14C] but not chemical identification.

In the present study a dose of 46 ng of [14C]-BaP was given orally to five volunteers on three separate cycles with a minimum two week “washout” interval. Blood was collected over 72 hours to determine pharmacokinetics. Covalent binding of [14C]-BaP to DNA from PBMCs was assayed in 4 of the 5 individuals over time and preliminary analysis of the [14C]-BaP metabolite profile in two individuals conducted by a recently developed UPLC-AMS interface developed for biological samples (Ognibene et al., 2015; Thomas et al., 2011).

Materials and Methods

This study was conducted under FDA-IND #117175 and IRB approval #5644 from Oregon State University and LLNL-approved IRB Protocol 2017–008.

Volunteers:

Subjects of both sexes, 21–65 years of age, were recruited. All subjects were white not of Hispanic origin. Exclusion criteria included women that were pregnant or could become pregnant, smoking (tobacco or other substances) in the past 3 months or living with one or more smokers, regular use of medications that affect gut motility or nutrient absorption, a history of gastrointestinal surgery or gastrointestinal disorder, current or history of kidney or liver disease, prior high-dose 14C exposure from medical tests or occupational PAH exposure (e.g. roofers, asphalt pavers, fire-fighters, etc.). We did not recruit individuals considered to be members of vulnerable populations, including those 20 years of age or younger, pregnant women, prisoners, non-English speakers, non-literate subjects or adults lacking capacity to consent. Following receipt of a signed informed consent, volunteers were given a physical exam by the study physician and blood chemistry analysis performed (including a pregnancy test for women). The demographics of the five individuals participating in the present study are given in Table 1. Three male and two females subjects 30–63 years of age with BMIs of 18.5–36.5 were enrolled and dosed with [14C]-BaP on three separate dosings to follow a 0–72 hour time course. In order to minimize the contribution of daily BaP exposure due to this study, for two weeks prior to micro-dosing of [14C]-BaP and during the 72 hour study, subjects were requested to refrain from consuming smoked meats or cheeses and charcoal-broiled meat. A dietary questionnaire was completed by each subject for the 3 days prior to dosing. Levels of dietary consumption of BaP were estimated using the guidelines of Dieziel et al., (2011).

Table 1.

Volunteera Demographics and CYP1B1 and GSTM1 Genotypes

Age Gender Height (cm) Weight (kg) BMI CYP1B1 *1 and *3 GSTM1*1 and *0
BaP002b 63 M 175 99.8 32.5 *3/*3 *0/*0
BaP003 38 M 190 131.8 36.5 *1/*1 *0/*0
BaP004 46 F 173.5 55.6 18.5 *1/*3 *0/*0
BaP009 55 F 157 77.8 31.6 *1/*1 *0/*0
BaP014 30 M 183 68.5 20.5 *1/*3 *1/*0 or *1/*1c
a

Race/ethnicity- All volunteers were Caucasian, not Hispanic

b

The numbers are non-continuous as 10 other enrolled subjects did not meet eligibility criteria (screen fail) or voluntarily withdrew prior to beginning the study or completing 3 cycles.

c

Does not distinguish between *1/*1 or *1/*0

Dosing and Blood Collections:

Subjects fasted overnight before Day 1 of each cycle. Each cycle was performed thrice with a minimum two week “washout” between cycles. The actual interval between cycles varied considerably (20–420 days) based on subject availability. A catheter was placed in an appropriate vein and a time zero blood sample taken prior to swallowing a capsule containing 46 ng (5 nCi) of [7-14C]-BaP (27 nCi/nmol), with water. The [7-14C]-BaP was from American Radiolabeled Chemicals, Inc. (St. Louis, MO, custom synthesis) and purified by reverse phase HPLC, capsules prepared and blood (8 mL at each time point) drawn at 0.25 0.5, 0.75, 1.0, 1.5, 2, 2.5, 3, 4, 8, 24, 48 and 72 hours as described in Hummel et al., (2018).

Plasma Extraction and Determination of [14C]-BaPeq by AMS:

Aliquots of plasma (0.75 mL or 1.5 ml for metabolic profiles) from each time point were extracted within two hours after collection according to the method of Crowell et al., (2011). The protocol is described in detail in Hummel et al., (2018). Samples were evaporated and flame-sealed in a quartz tube containing Cu(II) and combusted to 900°C, producing CO2 which was then transferred to a septa sealed glass tube containing Zn and Co and heated to 525°C, producing graphite on the Co catalyst. The graphite was loaded into an aluminum sample holder for AMS analysis, performed on the 250 kV single stage AMS, constructed and maintained by the Center for Accelerator Mass Spectrometry at LLNL (ratio of 14C:C with a precision of 3% and sensitivity of 0.7 attomol 14C per mg total carbon) (Ognibene et al., 2015). As in previous studies (Madeen et al., 2015; 2016; Hummel et al., 2018) biochemical samples are measured 4–10 times with at least 10,000 14C counts or for 30 seconds (each replicate) (Ognibene et al., 2015).

Pharmacokinetic Analysis:

Pharmacokinetics of total BaP equivalents (BaP and BaP metabolites, [14C]-BaPeq) were evaluated using non-compartmental and compartmental analyses. Area under the curve (AUC) values to the last measured time point and extrapolated to infinity (using last 3 time points) were calculated using the trapezoidal rule (Gibaldi and Perrier 1982). Mean resident times were calculated as a ratio of AUC under the 1st moment curve extrapolated to infinity to AUC extrapolated to infinity. Non-compartmental half-lives were calculated as the product of mean resident times with the natural log of two. A two-compartment pharmacokinetic model was used to evaluate the time course of [14C]-BaPeq (Equations 13), where “A0”, “A1”, and “A2” represent the amount (fg) of [14C]-BaPeq in the absorption, central, and peripheral compartments, respectively; “ka”, “ke”, “k12”, and “k21” are first-order rate constants. The concentration of the central compartment (C1) was calculated by normalizing the amount with the volume of distribution (V1, L, Equation 4).

dA0dt=(ka×A0) (1)
dA1dt=(ka×A0)(ke×A1)(k12×A1)+(k21×A2) (2)
dA2dt=(k12×A1)(k21×A2) (3)
C1=A1V1 (4)

Model parameters were optimized using a maximum log likelihood objective and the Nelder-Mead algorithm. Initial values were set by adjusting parameters visually. Software used to statistically analyze data was “R: A language and environment for statistical computing” Version 3.2.3 (R Foundation for Statistical Computing, Vienna, Austria).

DNA Extraction from PBMCs and Determination of BaPeq Covalent Binding to DNA:

Peripheral blood mononuclear cells (PBMCs) were isolated from blood of each volunteer at each time point and DNA isolated for determination of total [14C]-BaPeq bound using the graphite AMS assay as previously described (Felton and Turteltaub, 1994). DNA (average yield 24 ± 8 μg/mL whole blood) was extracted with a DNA Isolation Kit for Mammalian Blood (Roche Diagnostics, Indianapolis, IN). The DNA pellet was washed twice with 100% and once with 70% ethanol prior to solubilizing in nuclease-free water for determination of the concentration and purity via a Nanodrop ND-1000 (Thermo Scientific, Wilmington, DE). Samples were prepared as 200 ng/μL solutions. Twenty μg of DNA were analyzed by graphite AMS. The limit of adduct detection (LOD) for the 20 μg DNA sample was 0.2 fg (10 fg [14C]-BaP adducts/mg DNA) or approximately 0.5 adducts/1011 nucleotides.

Genotyping for CYP1B1*1 vs.*3 and GSTM1*1 vs. GSTM1*0 Alleles:

CYP1B1*1 and *3 alleles were distinguished by Sanger sequencing. PCR products were amplified from PMBC DNA (extraction described above) in a 20 μL reaction containing 200 ng DNA, 250 nM forward and reverse primers, and 10 μL SsoFast™ Supermix (Bio-Rad). The product was cleaned with Exo-SAP-IT™ (Applied Biosystems) per manufacturer’s instructions and sequenced at the Oregon State University Center for Genome Research and Biocomputing on an ABI 3730 capillary sequencer using ABI’s Big Dye Terminator chemistry. Sequences were visualized using FinchTV v. 1.5 (Supplemental Fig. 1). GSTM1 nulls were determined using a conventional PCR amplification. PCR components were the same as described above using GSTM1 specific primers to detect homozygotes or heterozygotes for a GSTM1 allele. Primers for human beta globin (HBB) were used as a control in the event an individual was a GSTM1 null. See Supplemental Table 1 for a list of primer sequences. Products were visualized on 10% TBE gels (Supplemental Fig. 2).

UPLC-AMS Analysis of [14C]-BaP and Metabolites Extracted from Human Plasma:

The following BaP metabolite standards were obtained from the PAH repository of the Oregon State University Superfund Research Program (http://limsweb.science.oregonstate.edu/, accessed 8–22-2018); BaP-9,10-diol, BaP-7,8-diol, BaP-1,6-quinone, BaP-3,6-quinone, BaP-6,12-quinone, BaP-7,8-dione, 3-hydroxy-BaP, 7-hydroxy-BaP, 9-hydroxy-BaP, BaP and DBC (internal standard). Resolution of BaP and metabolites was performed with a Waters Acquity H series UPLC. The column utilized was a Waters Acquity UPLC BEH C18 1.7 μm, 2.1 × 50 mm (Part # 186002350) with a guard column (part #186003981). Solvent A was HPLC-grade H2O with 0.3% formic acid and solvent B was HPLC grade (PEG free) acetonitrile. Initial conditions were 40% B for 2 minutes followed by a linear gradient to 60% B over 5 minutes with a hold at 60% for an addition 5 minutes following which solvent B was increased by a linear gradient to 80% over 10 minutes, and then to 100% over 5 minutes followed by a 3 minute gradient return to 40% B. The samples were maintained at 20°C in an autosampler. The column temperature was maintained at 28°C. Five μL was the injection volume and the column was eluted at 0.25 mL/minute. BaP and metabolite standards were prepared in acetonitrile and stored at −80°C until analysis. The plasma extracts of [14C]-BaP and metabolites (100 μL) in ethyl acetate were evaporated (Speed Vap, vacuum only with no centrifugation) to dryness and the residue redissolved in acetonitrile and transferred to the autosample vials. BaP and metabolites (Supplemental Table 2) were analyzed by a Waters Acquity UPLC PDA detector. Analysis of plasma extract containing [14C]-BaP and metabolites was by collection of the UPLC eluate onto a nickel moving wire (periodically indented to hold solvent) that passed through a cleaner prior to deposition of the eluate. The wire with eluate is passed through a drying oven followed by a combustion oven to convert the sample to 14CO2 before entering the 250 kV AMS. The 250 kV [14C]-AMS detector employed has previously been validated as an analytical instrument for pharmaceutical development (Keck et al. 2010). With respect to the moving wire interface with the UPLC and AMS we have previously established the LLOQ at 200 zmol [14C] (Thomas et al. 2011). Thus, with a specific activity of [14C]-BaP (27 nCi/nmol) and a molecular weight of 252 ng/nmol, the LLOQ is 1.2 fg. The accuracy was 1–3% with a precision (coefficient of variation) between 1–6% (Keck et al. 2010). A more complete description of the system is provided by Thomas et al., (2011) and https://www.youtube.com/watch?v=oFi17Vho44k (accessed 8–22-2018).

Results

Pharmacokinetics of [14C]-BaPeq

Pharmacokinetic parameters in plasma of volunteers were compared as a function of the interval between dosing. The relatively small difference between cycles (with the exception of 1 dose with subject BaP002) indicates little intra-individual variation in BaP pharmacokinetics, even with up to 60 weeks between runs, and variation did not correlate to days between doses (Fig. 1). The small number of individuals analyzed does not allow for any conclusions with respect to impact of gender, age, race/ethnicity, or BMI (Table 1). Although not statistically significant (due to the small cohort), compared to the 6-membered ring PAH, [14C]-DBC, (29 ng), 46 ng of [14C]-BaP was taken up faster (Tmax of 1.25 and 2.25 hours for [14C]-BaPeq and [14C]-DBCeq, respectively, p=0.09), but reached similar Cmax levels (70 ± 20 and 69 ± 40 fg/mL, respectively) (Table 2). The T1/2 (non-compartmental) was 70 ± 27 and 37 ± 30 hours, respectively for [14C]-BaPeq and [14C]-DBCeq (not statistically significant, p=0.11) (Table 2). The average AUC0–72hr and AUC0−∞ for [14C]-BaPeq were 1400 and 2615 (fg/mL × hr), respectively (Table 2 and Fig. 1), both similar to [14C]-DBCeq. A two-compartmental model yielded a T1/2α and T1/2β for plasma [14C]-BaPeq of 1.1 ± 0.3 and 89 ± 42 hours, respectively and the volume of distribution ranged from 235–1146 L. These kinetic constants were not significantly different from [14C]-DBCeq. Overall, these comparisons demonstrate similar kinetic profiles in plasma of total (parent [14C]-PAH and metabolites) following exposures to [14C]-BaP or [14C]-DBC but the extent of metabolism of [14C]-BaP is markedly greater.

Fig. 1. Time Intervals Between Dose and Plasma Levels of [14C]-BaPeq Determined by the Graphite Method over 72 Hours.

Fig. 1.

Volunteers were fasted overnight and dosed (9:00–10:00 AM) with 46 ng [7-14C]-BaP (5 nCi) in a food-grade cellulose capsule with 100 mL of water. Blood was drawn at 0. 0.25, 0.5, 0.75, 1, 1.5, 2, 2.5, 3, 4, 8, 12, 24, 48 and 72 hours. Plasma was extracted with ethyl acetate and 14C/C determined by the graphite method at the Center for Accelerator Mass Spectrometry (AMS), Lawrence Livermore National Laboratory. The 1 kv AMS has a precision of 3% and sensitivity of 0.7 attomol 14C per mg carbon. Additional experimental details are provided in Materials and Methods. The levels of [14C]-BaPeq in fg/mL plasma are depicted over 72 hours for each of the three dosing or cycles for the five volunteers. Results are shown for the first (C1-●), second (C2-■) and third cycles (C3-▲). The legend inset gives days elapsed, at the beginning of C2 and C3, since the first dose (C1). The raw data can be found as an Excel file in the Supplementary Materials.

Table 2.

Plasma Pharmacokinetics of [14C]-BaPeq and [14C]-DBCeq Following Micro-Dosinga

[14C]-PAH Tmax (hr) Cmax (fg/mL) AUC0–72hr (fg × hr × mL−1) AUC0- (fg × hr × mL−1) T1/2 (hr)b T1/2α (hr)c T1/2β (hr)c V (L)c
[14C]-BaP 1.25±0.4 4 (0.75–2.0) 70± 20 (29–82) 1400±483 (649–2060) 2615±962 (1769–4306) 70±27 (31114) 1.1±0.3 (0.7–1.6) 89±42 (38–156) 452 ±386 (235–1146)
[14C]-DBCd 2.25±0.95 (1.5–4.0) 69±40 (23–124) 1345±920 (315–2851) 2193±1947 (315–4892) 37±30 (9–93) 1.4±0.3 (1.0–1.9) 42±16e (20-70) 327±183 (116–646)
a

Mean ± SE with (range), of 3 separate dosing of 5 volunteers for [14C]-BaPeq (Hummel et al., 2018) and 6 volunteers for [14C]-DBCeq (Madeen et al., 2015)

b

Non-compartmental model

c

Two-compartmental model

d

Data taken from Madeen et al., (2015)

e

One outlier of 2683 hours was not used in the calculation

Time Course of Covalent Binding of [14C]-BaPeq to PBMC DNA

In four of the five volunteers, PBMCs were isolated from plasma at various time points up to 72 hours post-dose to assess covalent binding of [14C]-BaPeq to DNA. In two volunteers, (BaP002 and BaP003), no DNA adduction was detected at any time point (Fig. 2, top panel). With volunteers BaP004 and BaP009 a small amount of adduction is seen at 2 and 2.5 hours, respectively (Fig. 2). With the possible exception of the 24 hour time point for volunteer BaP004, no DNA adduction is observed 24–72 hours post-dosing consistent with previous studies with both [14C]-DBCeq (Madeen et al., 2015; 2016) and [14C]-BaPeq (Hummel et al., 2018). A recently developed ultrasensitive high-resolution mass spectrometric assay (Villata et al., 2017) achieved a similar LOD (1 adduct/1011 nucleotides) with BaP in human lung.

Fig. 2. Time Intervals Between Dose and PBMC [14C]-BaPeq DNA Adducts Determined by the Graphite Method over 72 Hours.

Fig. 2.

PBMCs were isolated and nuclear DNA extracted, at each of the time points for plasma collection, as described in Materials and Methods. The time course data for volunteers BaP002 (●), BaP003 (■) and BaP004 (▲) was following a single dosing of 46 ng, 5 nCi [14C]-BaP (top panel) and from two separate dosing to volunteer BaP009 (36 day interval between dosing BaP009a-● and BaP009b-■) (bottom panel). Twenty μg DNA was analyzed with an LOD of 10 fg [14C]-BaP/mg DNA, equivalent to 0.5 adducts/1011 nucleotides). The data are expressed as the number of [14C]-BaPeq covalent adducts per 1011 nucleotides.

Time Course of [14C]-BaP Metabolites in Plasma of Two Volunteers

The [14C]-BaP metabolite profile for volunteers BaP002 and BaP009 was assessed over the 72 hour time course utilizing the moving wire UPLC-AMS system developed by the Biomedical Research group at LLNL (Thomas et al., 2011; Ognibene et al., 2015; Madeen et al., 2016). Unlike the case with [14C]-DBC (Madeen et al., 2016), [14C]-BaP (21.86 min) was a minor component even at the earliest time points post-dosing (Fig. 3). Two major metabolites dominated the profile of each volunteer. With volunteer BaP002, the earliest (60–90 minutes) metabolite (M2, 1.41 min) to appear post-dosing in plasma eluted in the region of the gradient where BaP-dihydrodiols (could not identify based on retention time) and a later (Cmax of 3 hours) eluting metabolite (M4, 7.66 min) in the region where quinones (could not distinguish between the 1,6-, 3–6- or 6,12-quinone) are seen. In the plasma extract from volunteer BaP009, M2 is also a major metabolite and is seen in plasma with a time course similar to volunteer BaP002. A second major metabolite (M5, 12.86 min) which has a Cmax 45 minutes post-dosing, with a smaller peak at 90 minutes, elutes with a retention time matching 3-hydroxy-BaP (Fig. 3).

Fig. 3. [14C]-BaP and Metabolite Profile by UPLC-AMS as a Function of Time Post-Dosing for Volunteers BaP002 and BaP009.

Fig. 3.

Blood was collected from volunteers BaP002 and BaP009 at intervals from 0–72 hours and plasma isolated and extracted with ethyl acetate. The extract was taken up in acetonitrile and [14C]-BaP and metabolites resolved utilizing UPLC interfaced via a “moving wire” into a 250 kV AMS. The UPLC conditions and operation of the “moving wire” interface were as described in Materials and Methods. Metabolites 1–3 (M1●, M2■ and M3▲) eluted early in the gradient (Supplemental Table 2) and are possibly dihydrodiols or tetrols. The metabolite designated “quinone” (M4▼) eluted at 7.66 minutes. At this time we cannot, with confidence, designate this peak as either the 1,6-, 3,6- or 6,12-quinone of [14C]-BaP. This peak is unlikely [14C]-BaP-7,8-dione as that standard elutes somewhat later in the gradient as a single peak (Supplemental Fig. 3). The single phenol (M5♦) observed co-elutes with 3-hydroxy-[14C]-BaP. The metabolite identities are tentative and based on similar retention times to the BaP standards available (see Materials and Methods for list). Parent [14C]-BaP (♦) was well resolved from the metabolites and was a minor component in the profiles from both volunteers even at early time points.

CYP1B1 (*1/*1, *1/*3 and *3/*3) and GSTM1 (*1 and *0/*0) Allelic Variants

This study is not sufficiently powered to correlate genotype to pharmacokinetic constants, metabolite profiles or formation of covalent [14C]-BaPeq-DNA adducts in PBMCs. We did observe that, in the two-compartment model, k12 (r2 =0.86, p=0.023) and kelα (r2 =0.75, p=0.056) rates increased with the number of CYP1B1*3 alleles whereas the t1/2α decreased (r2 = 0.671, p = 0.090) (data not shown). Both volunteers BaP002 and BaP009 were genotyped as GSTM1 nulls and CYP1B1*3/*3 and CYP1B1*1/*1, respectively (Table 1 and Supplemental Figs. 1 and 2).

Discussion

In the present report five volunteers, fasted overnight, were given capsules on three separate occasions with the same dose of [14C]-BaP. It should be noted that (with the exception of BaP002) there was markedly low intra-individual variation in the PK profile of [14C]-BaPeq independent of the interval between doses. As in rodent models (Crowell et al., 2011), BaP is fairly rapidly eliminated from plasma. We found that at doses 5–15-times lower than the estimated daily exposure to BaP in the U.S., very little parent compound was present in plasma, even at the earliest time points, perhaps indicative of extensive hepatic and/or intestinal first-pass metabolism.

Comparison of PK parameters of [14C]-BaPeq to [14C]-DBCeq by graphite AMS displayed few marked differences. It should be recognized that our PK parameters, determining by graphite AMS, incorporate all [14C]-BaP (parent and metabolites), not just the parent PAH; thus PK parameters are impacted by protein binding, partitioning, transport, etc. Incorporation of the recently developed UPLC “moving wire” AMS interface, that allows for speciation of [14C]-PAHs and metabolites, demonstrated a marked difference in extent of metabolism. An interesting observation, yet unexplained, is that, when comparing [14C] recovered (volunteers BaP002 and BaP009) as [14C]-BaPeq using graphite (Figure 1) to UPLC-AMS (Figure 3), the latter is 26–33% lower than the former (based on AUC0–72hr). Compared to [14C]-DBC, [14C]-BaP was extensively metabolized such that parent [14C]-BaP was a minor component even at the earliest time points. The more rapid metabolism of BaP, compared to DBC, is consistent with in vitro studies at much higher concentrations (Crowell et al., 2014; Smith et al., 2017) whereas the rates of phase 1 and phase 2 metabolism of BaP-7,8-DHD and DBC-11,12-DHD are similar (Smith et al., 2017).

The metabolic profile of [14C]-BaP exhibited some differences and similarities in the two individuals examined. In one individual (BaP002), a BaP quinone, not seen in the profile from BaP009, was observed although the second of the two major metabolites was the same and markedly more polar, probably a dihydrodiol (possibly a tetrol for which no standard was examined). Another striking difference was the lack of formation of any phenols with BaP002 whereas 3-hydroxy-BaP was a major metabolite found in plasma from BaP009. The later eluting smaller 3-hydroxy-BaP peak in plasma may be indicative of enterohepatic recirculation.

PAHs, such as BaP and DBC, have multiple mechanisms of carcinogenesis. Metabolic activation by CYPs in the 1 family (especially CYP1B1) to bay or fjord region epoxides are rapidly hydrolyzed by epoxide hydrolase to two stereo isomers ((+/−)-BaP-7,8-DHD or (+/−)-DBC-11,12-DHD) followed by a second CYP1-epoxygenation to the potent electrophilic anti- or cis-dihydrodiol-epoxides (4 enantiomers each of BaP-7,8-DHD-9,10-E or DBC-11,12-DHD-13,14-E). BaP readily forms covalent DNA adducts, predominantly the N2-(7,8,9-trihydroxy-7,8,9,10-tetrahydrobenzo[a]pyrene-C10-yl)-2’-deoxyguanosine (N2-BaP-DHDE-dG) whereas DBC-11,12-DHD-13,14-E has a preference for adduction at N6-adenosine. Additional mechanisms of carcinogenesis involve reduction of the DHD by aldo-keto reductase (AKR) to electrophilic and redox active catechols (Penning, 2014; Quinn and Penning, 2008). In our UPLC system the BaP-7,8-dione had a retention time of 9.34 minutes and was well resolved from the three quinones and phenols (we did not have a standard of the BaP-7,8-catechol). With PAHs, one-electron oxygenations by peroxidases also produce electrophiles (Zamzow et al., 1989). Carcinogenicity can also result from PAH-dependent alteration of expression of genes important in tumor production through binding to the aryl hydrocarbon receptor (AhR) (Shimizu et al., 2000).

DBC, a six-membered PAH with both a bay- and fjord-region, is a potent carcinogen in preclinical rodent models (probable compared to BaP. In the study with [14C]-DBC, the major plasma metabolite observed was tentatively identified as DBC-11,12-DHD yet we found no evidence of covalent DNA adduction in PBMCs (Madeen et al., 2016). In this study with [14C]-BaP, although extensive metabolism was evident, little or no covalent DNA adduction was observed 48–72 hours after dosing. Two out of four volunteers examined over the 72 hour time-course showed [14C]-BaP DNA binding at an early time point (2 and 2.5 hr for volunteers BaP004 and BaP009, respectively), which we originally believed was random noise, however, DNA from PBMCs from BaP009 gave similar results over 2 separate dosing time-courses. We are continued to examine this issue with a planned dose-response study.

Can studies such as this be useful in prediction of cancer risk from truly environmentally relevant levels of exposure to carcinogenic PAHs or other carcinogens? There are limitations to this study including sample size and a single dosage not reflective of steady-state concentrations with daily exposures. The EPA has set an oral cancer slope factor (upper bound for risk) for BaP of 1 (mg/kg-day)−1. The lifetime risk-specific dose equating to 1 cancer in 106 is 1.0 × 10−6 mg/kg-day which is approximately twice the risk if the dose used here (5.2 × 10−7 mg/kg) were administered every day of the volunteer’s lives. If we utilized covalent DNA adduction to predict cancer susceptibility (Bartsch et al., 1998; Kriek et al., 1993; Lee et al., 2002), the limited DNA adduct data we have to date would indicate a risk markedly less than 1 in 106. Due to extensive metabolism of [14C]-BaP, observed by UPLC-AMS, the plasma Cmax following our single dose of 46 ng ranged from 9–15 fM (two volunteers). Utilizing our PBPK model (Crowell et al., 2011), the estimated Cmax in liver would be 0.12–0.2 pM, over 106-fold lower than the calculated Km (0.725 μM) in human liver microsomes (Crowell et al., 2014) and the IC50 in a BaP competitive binding assay with human AhR (Flaveny et al., 2009). Thus it would seem unlikely that BaP would impact the expression of AhR-dependent gene expression following this exposure although no alterations in the transcriptome were examined in this study.

It is also useful to relate our data to studies with human cells in vitro and BaP plasma concentrations and degree of PBMC DNA adduction found in vivo with human populations. Peak plasma levels of [14C]-BaP in this study are more than 3 orders of magnitude lower than the BaP median level of 4–19 pg/mL from a study by Pleil et al., (2010), examining a healthy student population and about 30,000 fold lower than concentrations found to have no adverse impact on Hepa1c1c7 hepatocytes (Madureira et al., 2014). The levels of BaP-DNA adducts in PBMCs from occupationally exposed workers have been reported to be 1–10 adducts per 108 nucleotides (Boysen and Hecht, 2003; Carstensen et al., 1999; Divi et al., 2002; Pavanello et al., 2005). The levels of covalent DNA adduction in PBMCs from volunteers in our study were at least 3 orders of magnitude lower, again indicating little to no measurable risk from this exposure.

Somewhat surprisingly, history of exposure (smoking, occupational) has been reported as not significantly elevating BaP-DHDE-DNA adducts in PBMCs or other tissues (Boysen and Hecht, 2003; Carstensen et al., 1999). Other studies report modestly higher levels in smoker’s compared to non- or ex-smokers (Lodovici et al., 1998; Villalta et al., 2017) Examination of 10 human lung biopsy samples by Monien et al., (2015) found no detectable (LOD 1–3 adducts per 108 nucleotides) BaP-DHDE-dG adducts. Recent advances in methodology, leading to increased specificity and sensitivity for assaying BaP-DNA adducts, have improved (LOD of ~ 1 adduct in 1011 nucleotides). Villalta et al., (2017) reported levels of (predominantly 7R,8S,9S,10R) BaP-DHDE-N2-dG adducts in lung biopsies from smokers and non-smokers as 3.1 and 1.3 adducts per 1011 nucleotides, respectively.

Another potential use of UPLC-AMS technology, if expanded, is identification of resistant and susceptible individuals. Measures of BaP-DNA adducts in humans is likely driven by gene-environment interactions. A number of studies have documented enhanced cancer susceptibility for individuals expressing the GSTM1*0 (null) allele (approximately 50% in Caucasians) (Alexandrov et al., 2002; Boysen and Hecht, 2003; Pavanello et al., 2002, 2005; Rojas et al., 1998; Wenzlaff et al., 2005a). As all volunteers examined for [14C]-BaP DNA adduction in PBMCs were GSTM1 nulls, this study could not examine the impact of GSTM1 expression.

Studies suggest individuals expressing the CYP1B1*3 allelic variant (about 39% in Caucasians) have an enhanced risk of some cancers including lung (Shah et al., 2008; Wenzlaff et al., 2005b; Xu et al., 2012), ovarian (Goodman et al., 2001), and head and neck cancer in smokers (Thier et al., 2002), but not for colorectal cancer (Xie et al., 2012). An inverse relationship is seen for prostate cancer (Beuten et al., 2008). Individuals with both CYP1B1*3 (or CYP1A1*2, not examined in this study) and GSTM1*0 alleles have an enhanced lung cancer risk compared to either alone (Alexandrov et al., 2002; Shah et al., 2008).

The CYP1B1.3 (L432V) enzyme is expressed at about 50% the level of CYP1B1.1, is less responsive to induction by BaP (human lymphocytes) and exhibits a 25% lower Kcat for BaP epoxygenation to the 7,8-DHD (Aklillu et al., 2005; Helmig et al., 2014). These observations would seem to be inconsistent with an enhanced lung cancer risk if CYP1B1-dependent formation of BaP-7,8-DHD-9,10-E were critical. The two individuals examined for [14C]-BaP metabolic profiles in plasma were homozygous for either CYP1B1*1 or CYP1B1*3. The extent of metabolism and profile of metabolites was distinct suggesting further genotype-dependent studies are warranted. The same could be said for the GSTM1 allelic variant.

A limitation of this study was failure to positively identify BaP metabolites. Work is in progress at LLNL to send the UPLC sample (spiked with unlabeled metabolites standards) elute through a splitter, a UV/VIS scanning detector and conventional MS/MS for positive identification of peaks with the same retention time as the AMS 14C peaks.

These studies complement and expand PK determinants of PAHs such as BaP and DBC (Crowell et al., 2014) and their metabolites (Smith et al., 2017) in vitro with human liver microsomes of BaP at concentrations at least 6 orders of magnitude greater than the Cmax in liver (based on the PBPK model from Crowell et al., 2011)). Such an approach is consistent with the philosophy of “the best model for humans is humans”.

Supplementary Material

1
2
3
4
5
6

HIGHLIGHTS.

  • There is little intra-individual variation in T1/2, Cmax or Tmax of [14C]-benzo[a]pyrene

  • [14C]-Benzo[a]pyrene is rapidly absorbed from GI and eliminated from plasma

  • UPLC-accelerator mass spectrometry shows benzo[a]pyrene is rapidly metabolized

  • Little or no covalent adduction to DNA in PBMCs occurs (< 1 in 1011 base pairs)

  • Two individuals showed marked differences in the profile of metabolites in plasma

Acknowledgments

The authors would like to thank Alexandria Van Scoyk, Hannah You, Jessica Hummel, Chelsea Meedom, and Youngjoo Lee for their contribution of extracting samples for accelerator mass spectrometry. This study was funded by Public Health Service grants P42ES016465, K.C. Donnelly Supplement P42ES016465, R01ES028600 and T32ES07060. Accelerator Mass Spectrometry was performed at the Research Resource for Biomedical Accelerator Mass Spectrometry operated at Lawrence Livermore National Laboratory under the U.S. Department of Energy contract DE-AC52-07NA27344 and National Institutes of Health grant P41 GM103483.

Footnotes

The authors have no conflicts of interest to disclose.

Compliance with Ethical Standards

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. Informed consent was obtained from all individual participants included in this study.

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final citable form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  1. Aklillu E, Øvrebø S, Botnen IV, Otter C, Ingelman-Sundberg M (2005) Characterization of common CYP1B1 variants with different capacity for benzo[a]pyrene-7,8-dihydrodiol epoxide formation from benzo[a]pyrene. Cancer Res 65:5105–11, doi: 10.1158/0008-5472.CAN-05-0113. [DOI] [PubMed] [Google Scholar]
  2. Alexandrov K, Cascorbi I, Rojas M, Bouvier G, Kriek E, Bartsch H (2002) CYP1A1 and GSTM1 genotypes affect benzo[a]pyrene DNA adducts in smokers’ lung: comparison with aromatic/hydrophobic adduct formation. Carcinogenesis 23:1969–77, PMID:. [DOI] [PubMed] [Google Scholar]
  3. ATSDR (2013), http://www.atsdr.cdc.gov/spl/resources/ATSDR_2013_SPL_Detailed_Data_Table.pdf., accessed 08–22–2018.
  4. Bartsch H, Rojas M, Alexandrov K, Risch A (1998) Impact of adduct determination on the assessment of cancer susceptibility. Recent Res Cancer Res 154:86–96, PMID:. [DOI] [PubMed] [Google Scholar]
  5. Beuten J, Gelfond JAL, Byrne JJ, Balic I, Crandall A, Johnson-Pais TL, Thompson IM, Price DK, Leach RJ (2008) CYP1B1 variants are associated with prostate cancer in non-Hispanic and Hispanic Caucasians. Carcinogenesis 29:1751–7, doi: 10.1093/carcin/bgm300. [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Borska L, Andrys C, Krejsek J, Palicka V, Chmelarova M, Hamakova K, Kremlacek J, Fiala Z (2014) Oxidative damage to nucleic acids and benzo(a)pyrene-7,8-diol-9,10-epoxide-DNA adducts and chromosomal aberration in children with psoriasis repeatedly exposed to crude coal tar ointment and UV radiation. Oxid Med Cell Longev 2014:302528, doi: 10.1155/2014/302528. [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Boysen G, Hecht SS (2003) Analysis of DNA and protein adducts of benzo[a]pyrene in human tissues using structure-specific methods. Mutat Res 543:17–30, 10.1016/S1383-5742(02)00068-6. [DOI] [PubMed] [Google Scholar]
  8. Brown K, Tompkins EM, White INH (2006) Applications of accelerator mass spectrometry for pharmacology and toxicological research. Mass Spectrom Rev 25:127–4, doi: 10.1002/mas.20059. [DOI] [PubMed] [Google Scholar]
  9. Carstensen U, Hou S-M, Alexandrie A-K, Högstedt B, Tagesson C, Warholm M, Rannug A, Lambert B, Axmon A, Hagmar L (1999) Influence of genetic polymorphisms of biotransformation enzymes on gene mutations, strand breaks of deoxyribonucleic acid, and micronuclei in mononuclear blood cells and urinary 8-hydroxydeoxyguanosine in potroom workers exposed to polyaromatic hydrocarbons. Scand J Work Environ Health 25:351–60, doi: 10.5271/sjweh.445. [DOI] [PubMed] [Google Scholar]
  10. Crowell SR, Amin SG, Anderson KA, Krishnegowda G, Sharma AK, Soelberg JJ, Williams DE, Corley RA (2011) Preliminary physiologically based pharmacokinetic models for benzo[a]pyrene and dibenzo[def,p]chrysene in rodents. Toxicol Appl Pharmacol 257:365–76, doi: 10.1016/j.taap.2011.09.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  11. Crowell SR, Hanson-Drury S, Williams DE, Corley RA (2014) In vitro metabolism of benzo[a]pyrene and dibenzo[def,p]chrysene in rodent and human hepatic microsomes. Toxicol Lett 228:48–55, doi: 10.1016/j.toxlet.2014.04.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Dieziel NC, Strickland PT, Platz EA, Abubaker S, Buckley TJ (2011) Comparison of standard methods for assessing dietary intake of benzo[a]pyrene. Cancer Epidemiol Biomarkers Prev 20:962–70, doi: 10.1158/1055-9965.EPI-10-1344. [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Divi RL, Beland FA, Fu PP, Von Tungeln LS, Schoket B, Camara JE, Ghei M, Rothman N, Sinha R, Poirier MC (2002) Highly sensitive chemiluminescence immunoassay for benzo[a]pyrene-DNA adducts: validation by comparison with other methods, and use in human biomonitoring. Carcinogenesis 23:2043–9, PMID:. [DOI] [PubMed] [Google Scholar]
  14. Enright HA, Malfatti MA, Zimmermann M, Ognibene T, Henderson P, Turteltaub KW (2016) Use of accelerator mass spectrometry in human health and molecular toxicology. Chem Res Toxicol 29:1976–86, doi: 10.1021/acs.chemrestox.6b00234. [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. EPA (2010) Development of a relative potency factor (RPF) approach for polycyclic aromatic hydrocarbon (PAH) mixtures (External Review Draft), U.S. EPA, Washington, D.C., EPA/635/R-08/012A, 622 p. [Google Scholar]
  16. EPA (2017) Toxicological Review of Benzo[a]pyrene, Integrated Risk Information System, National Center for Environmental Assessment, Office of Research and Development, EPA, Washington, D.C., [CASRN 50–32-8]. EPA/635/R-17–003, https://cfpub.epa.gov/ncea/iris2/chemicalLanding.cfm?substance_nmbr=136 (accessed 08–22–2018). [Google Scholar]
  17. Felton JS, Turteltaub KW (1994) Accelerator mass spectrometry for measuring low-dose carcinogen binding to DNA. Environm Hlth Perspect 102:450–2, doi: 10.1289/ehp.94102450. [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Flaveny CA, Murray IA, Chiaro CR, Perdew GH (2009) Ligand selectivity and gene regulation by the human aryl hydrocarbon receptor in transgenic mice. Molec Pharmacol 75:1412–20, doi: 10.1124/mol.109.054825. [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Gibaldi M, Perrier D (1982) Pharmacokinetics, M. Dekker, New York. [Google Scholar]
  20. Godschalk RW, Vermeer IT, Kriek E, Floot B, Schilderman PA, Moonen EJ, Kleinjans JC, van Schooten FJ (1997) Comparison of 32P-postlabeling and HPLC-FD analysis of DNA adducts in rats acutely exposed to benzo(a)pyrene. Chem-Biol Interact 104:41–54, 10.1016/S0009-2797(97)03765-4. [DOI] [PubMed] [Google Scholar]
  21. Goodman MT, McDuffie K, Kolonel LN, Terada K, Donlon TA, Wilkens LR, Guo C, Le Marchand L (2001) Case-control study of ovarian cancer and polymorphisms in genes involved in catechol estrogen formation and metabolism. Cancer Epidemiol Biomarkers Prev 10:209–16, PMID:. [PubMed] [Google Scholar]
  22. Hakura A, Sonoda J, Tsutsui Y, Mikami T, Imade T, Shimada M, Yaguchi S, Yamanaka M, Tomimatsu M, Tsukidate K (1998a) Toxicity profiles of benzo[a]pyrene in the male LacZ transgenic mouse (MutaMouse) following oral administration for 5 consecutive days. Regul Toxicol Pharmacol 27:273–9, doi: 10.1006/rtph.1998.1218. [DOI] [PubMed] [Google Scholar]
  23. Hakura A, Tsutsui Y, Sonoda J, Kai J, Imade T, Shimada M, Sugihara Y, Mikami T (1998b) Comparison between in vivo mutagenicity and carcinogenicity in multiple organs by benzo[a]pyrene in the LacZ transgenic mouse (Muta Mouse). Mutat Res 398:123–30, 10.1016/S0027-5107(97)00248-0. [DOI] [PubMed] [Google Scholar]
  24. Hakura A, Tsutsui Y, Sonoda J, Mikami T, Tsukidate K, Sagami F, Kerns WD (1999) Multiple organ mutation in the lacZ transgenic mouse (Muta mouse) 6 months after oral treatment (5 days) with benzo[a]pyrene. Mutat Res 426:71–7, 10.1016/S0027-5107(99)00046-9. [DOI] [PubMed] [Google Scholar]
  25. Hakura A, Tsutsui Y, Sonoda J, Tsukidate K, Mikami T, Samami F (2000) Comparison of the mutational spectra of the LacZ transgene in four organs of the MutaMouse treated with benzo[a]pyrene: target organ specificity. Mutat Res 447:239–47, 10.1016/S0027-5107(99)00213-4. [DOI] [PubMed] [Google Scholar]
  26. Helmig S, Wenzel S, Maxeiner H, Schneider J (2014) CYP1B1 mRNA inducibility due to benzo(a)pyrene is modified by the CYP1B1 L432V gene polymorphism. Mutagenesis 29:237–40, doi: 10.1093/mutage/geu010. [DOI] [PubMed] [Google Scholar]
  27. Hummel JM, Madeen EP, Siddens LK, Uesugi SL, McQuistan T, Anderson KA, Turteltaub KW, Ognibene TJ, Bench G, Krueger SK, Harris S, Smith J, Tilton SC, Baird WM, Williams DE (2018) Pharmacokinetics of [14C]-benzo[a]pyrene (BaP) in humans: impact of co-administration of smoked salmon and BaP dietary restriction. Fd Chem Toxicol 115:136–47, doi: 10.1016/j.fct.2018.03.003. [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. IARC (2010), Some non-heterocyclic polycyclic aromatic hydrocarbons and some related exposures. Monographs on the evaluation of carcinogenic risks to humans; Lyon, France. [PMC free article] [PubMed] [Google Scholar]
  29. Jernström B, Gräslund A (1994) Covalent binding of benzo[a]pyrene 7,8-dihydrodiol 9,10-epoxides to DNA: molecular structures, induced mutations and biological consequences. Biophys Chem 49:185–99, 10.1016/0301-4622(93)E0087-L. [DOI] [PubMed] [Google Scholar]
  30. Jubert C, Mata J, Bench G, Dashwood R, Pereira C, Tracewell W, Turteltaub K, Williams D, Bailey G (2009) Effects of chlorophyll and chlorophyllin on low-dose aflatoxin B1 pharmacokinetics in human volunteers. Cancer Prev Res 2:1015–22, doi: 10.1158/1940-6207.CAPR-09-0099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Keck BD, Ognibene T, Vogel JS (2010) Analytical validation of accelerator mass spectrometry for pharmaceutical development. Bioanalysis 2:469–85, doi: 10.4155/bio.10.14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Kriek E, Van Schooten FJ, Hillebrand MJX, Van Leeuwen FE, Den Engelse L, De Looff AJA, Dijkmans APG (1993) DNA adducts as a measure of lung cancer risk in humans exposed to polycyclic aromatic hydrocarbons. Environm Hlth Perspect 99:71–5, doi: 10.1289/ehp.939971. [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Lee BM, Yoo SD, Kim S (2002) A proposed methodology of cancer risk assessment modeling using biomarkers. J Toxicol Environ Health A 65:341–54, doi: 10.1080/15287390252808019. [DOI] [PubMed] [Google Scholar]
  34. Lightfoot TJ, Coxhead JB, Cupid BC, Nicholson S, Garner RC (2000) Analysis of DNA adducts by accelerator mass spectrometry in human breast tissue after administration of 2-amino-1-methyl-6-phenylimidzao[4,5-b]pyridine and benzo[a]pyrene. Mutat Res 472:119–27, 10.1016/S1383-5718(00)00134-0. [DOI] [PubMed] [Google Scholar]
  35. Lodovici M, Akpan V, Giovannini L, Migliani F, Dolara P (1998) Benzo[a]pyrene diol-epoxide DNA adducts and levels of polycyclic aromatic hydrocarbons in autoptic samples from human lungs. Chem-Biol Interact 116:199–212, 10.1016/S0009-2797(98)00091-X. [DOI] [PubMed] [Google Scholar]
  36. Madeen E, Corley RA, Crowell S, Turteltaub K, Ognibene T, Malafatti M, McQuistan TJ, Garrad M, Sudakin D, Williams DE (2015) Human in vivo pharmacokinetics of [14C]dibenzo[def,p]chrysene by accelerator mass spectrometry following oral microdosing. Chem Res Toxicol 28:126–34, doi: 10.1021/tx5003996. [DOI] [PMC free article] [PubMed] [Google Scholar]
  37. Madeen EP, Ognibene TJ, Corley RA, McQuistan TJ, Henderson MC, Baird WM, Bench G, Turteltaub KW, Williams DE (2016) Human microdosing with carcinogenetic polycyclic aromatic hydrocarbons: in vivo pharmacokinetics of dibenzo[def,p]chrysene and metabolites by UPLC accelerator mass spectrometry. Chem Res Toxicol 29:1641–50, doi: 10.1021/acs.chemrestox.6b00169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  38. Madureira DJ, Weiss FT, Midwoud PV, Helbling DE, Sturla SJ, Schirmer K (2014) Systems toxicology approach to understand the kinetics of benzo[a]pyrene uptake, biotransformation, and DNA adduct formation in a liver cell model. Chem Res Toxicol 27:443–53, doi: 10.1021/tx400446q. [DOI] [PubMed] [Google Scholar]
  39. Monein BH, Schumacher F, Herrmann K, Glatt H, Turesky RJ, Chesné C (2015) Simultaneous detection of multiple DNA adducts in human lung samples by isotope-dilution UPLC-MS/MS. Analy Chem 87:641–8, doi: 10.1021/ac503803m. [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Ognibene TJ, Bench G, Brown TA, Vogel JS (2004) The LLNL accelerator mass spectrometry system for biochemical 14C-measurements. Nucl Instr Meth Phys Res 23:12–5, doi: 10.1021/ac026334j. [DOI] [Google Scholar]
  41. Ognibene TJ, Haack KW, Bench G, Brown TA, Turteltaub KW (2015) Operation of the “small” BioAMS spectrometers at CAMS: past and future prospect. Nucl Instr Meth Phys Res Section B 361:54–7, doi: 10.1016/j.nimb.2015.05.019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Pavanello S, Siwinska E, Mielzynska D, Clonfero E (2004) GSTM1 null genotype as a risk factor for anti-BPDE-DNA adduct formation in mononuclear white blood cells of coke-oven workers. Mutat Res 558:53–62, doi: 10.1016/j.mrgentox.2003.10.019. [DOI] [PubMed] [Google Scholar]
  43. Pavanello S, Pulliero A, Siwinska E, Mielzynska D, Clonfero E (2005) Reduced nucleotide excision repair and GSTM1-null genotypes influence anti-B[a]PDE-DNA adduct levels in mononuclear white blood cells of highly PAH-exposed coke oven workers. Carcinogenesis 26:169–75, doi: 10.1093/carcin/bgh303. [DOI] [PubMed] [Google Scholar]
  44. Penning TM (2014) Human aldo-keto reductases and the metabolic activation of polycyclic aromatic hydrocarbons. Chem Res Toxicol 27:1901–17, doi: 10.1021/tx500298n. [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Pleil JD, Stiegel MA, Sobus JR, Tabucchi S, Ghio AJ, Madden MC (2010) Cumulative exposure assessment for trace-level polycyclic aromatic hydrocarbons (PAHs) using blood and plasma analysis. J Chromatogr B Analyt Technol Biomed Life Sci 878:1753–60, doi: 10.1016/j.jchromb.2010.04.035. [DOI] [PubMed] [Google Scholar]
  46. Quinn AM, Penning TM (2008) Comparisons of (±)-benzo[a]pyrene-trans-7,8-dihydrodiol activation by human cytochrome P450 and aldo-keto reductase enzymes: effect of redox state and expression levels. Chem Res Toxicol 21:1086–94, doi: 10.1021/tx700345v. [DOI] [PubMed] [Google Scholar]
  47. Rojas M, Alexandrov K, Cascorbi I, Brockmöller J, Likhachev A, Pozharisski K, Bouvier G, Auburtin G, Mayer L, Koop-Schneider A, Roots I, Bartsch H. (1998) High benzo[a]pyrene diol-epoxide DNA adduct levels in lung and blood cells from individuals with combined CYP1A1 MspI/Msp-GSTM1*0/*0 genotypes. Pharmacogenetics 8:109–18, PMID:. [PubMed] [Google Scholar]
  48. Ross JA, Nelson GB, Wilson KH, Rabinowitz JR, Galati A, Stoner GD, Nesnow S, Mass MJ (1995) Adenomas induced by polycyclic aromatic hydrocarbons in strain A/J mouse lung correlate with time-integrated DNA adduct levels. Cancer Res 55:1039–44, PMID:. [PubMed] [Google Scholar]
  49. Schults MA, Chiu RK, Nagle PW, Wilms LC, Kleinjans JC, van Schooten FJ, Godschalk RW (2013) Genetic polymorphisms in catalase and CYP1B1 determine DNA adduct formation by benzo[a]pyrene ex vivo. Mutagenesis 28:181–5, doi: 10.1093/mutage/ges070. [DOI] [PubMed] [Google Scholar]
  50. Shah PP, Singh AP, Singh M, Mathur N, Mishra BN, Pant MC, Parmar D (2008) Association of functionally important polymorphisms in cytochrome P4501B1 with lung cancer. Mutat Res 643:4–10, doi: 10.1016/j.mrfmmm.2008.05.001. [DOI] [PubMed] [Google Scholar]
  51. Shiizaki K, Kawanishi M, Yagi T (2017) Modulation of benzo[a]pyrene-DNA adduct formation by CYP1 inducer and inhibitor. Genes Environ 39:14, doi: 10.1186/s41021-017-0076-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Shimizu Y, Nakatsuru Y, Ichinose M, Takahashi Y, Kume H, Mimura J, Fujii-Kuriyama Y, Ishikawa T (2000) Benzo[a]pyrene carcinogenicity is lost in mice lacking the aryl hydrocarbon receptor. Proc Natl Acad Sci (USA) 97:779–82, 10.1073/pnas.97.2.779. [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Smith JN, Mehinagic D, Nag S, Crowell SR, Corley RA (2017) In vitro metabolism of benzo[a]pyrene-7,8-dihydrodiol and dibenzo[def,p]chrysene-11,12 diol in rodent and human hepatic microsomes. Toxicol Lett 269:23–32, doi: 10.1016/j.toxlet.2017.01.008. [DOI] [PubMed] [Google Scholar]
  54. Thier R, Brüning T, Roos PH, Bolt HM (2002) Cytochrome P450 1B1, a new keystone in gene-environment interactions related to human head and neck cancer? Arch Toxicol 76:249–56, doi: 10.1007/s00204-002-0349-3. [DOI] [PubMed] [Google Scholar]
  55. Thomas AT, Ognibene T, Daley P, Turteltaub K, Radousky H, Bench G (2011) Ultrahigh efficiency moving wire combustion interface for online coupling of high-performance liquid chromatography (HPLC). Analy Chem 83:9413–7, doi: 10.1021/ac202013s. [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Tuominen R, Baranczewski P, Warholm M, Hagmar L, Möller L, Rannug A (2002) Susceptibility factors and DNA adducts in peripheral blood mononuclear cells of aluminum smelter workers exposed to polycyclic aromatic hydrocarbons. Arch Toxicol 76:178–86, doi: 10.1007/s00204-002-0331-0. [DOI] [PubMed] [Google Scholar]
  57. Villalta PW, Hochalter JB, Hecht SS (2017) Ultrasensitive high-resolution mass spectrometric analysis of a DNA adduct of the carcinogen benzo[a]pyrene in human lung. Analy Chem 89:12,735–42, doi: 10.1021/acs.analchem.7b02856. [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Wenzlaff AS, Cote ML, Bock CH, Land SJ, Schwartz AG (2005a) GSTM1, GSTT1, and GSTP1 polymorphisms, environmental tobacco smoke exposure and risk of cancer among never smokers: a population based study. Carcinogenesis 26:395–401, doi: 10.1093/carcin/bgh326. [DOI] [PubMed] [Google Scholar]
  59. Wenzlaff AS, Cote ML, Bock CH, Land SJ, Santer SK, Schwartz DR, Schwartz AG (2005b) CYP1A1 and CYP1B1 polymorphisms and risk of lung cancer among never smokers: a population-based study. Carcinogenesis 26:2207–12, doi: 10.1093/carcin/bgi191. [DOI] [PubMed] [Google Scholar]
  60. WHO, Benzo[a]pyrene (1998)WHO FoodAdditives Series 28 in http://www.inchem.org/documents/ehc/ehc/ehc202.htm#SubSectionNumber:7.1.6 (accessed 08–22–2018), United Nations environment programme, international labour organization, World Health Organization, international programme on chemical safety, environmental health criteria 202, selected non-heterocyclic, polycyclic aromatic hydrocarbons, ISBN 92 4 157202 7, (NLM Classification: QD 341.H9), ISSN 0250–863X.
  61. Xie Y, Liu GQ, Miao XY, Liu Y, Zhou W, Zhong DW (2012) CYP1B1 leu431val polymorphism and colorectal cancer risk among Caucasians: a meta-analysis. Tumour Biol 33:809–16, doi: 10.1007/s13277-011-0298-7. [DOI] [PubMed] [Google Scholar]
  62. Xu W, Zhou Y, Hang X, Shen D (2012) Current evidence on the relationship between CYP1B1 polymorphisms and lung cancer risk: a meta-analysis. Mol Biol Rep 39:2821–9, doi: 10.1007/s11033-011-1041-6. [DOI] [PubMed] [Google Scholar]
  63. Zamzow D, Jankowiak R, Cooper RS, Small GJ, Tibbels SR, Cremonesi P, Devanesan P, Rogan EG, Cavalieri EL (1989) Fluorescence line narrowing spectrometric analysis of benzo[a]pyrene-DNA adducts formed by one-electron oxidation. Chem Res Toxicol 2:29–34, PMID:. [DOI] [PubMed] [Google Scholar]
  64. Zuo J, Brewer DS, Arlt VM, Cooper CS, Phillips DH (2014) Benzo[a]pyrene-induced DNA adducts and gene expression profiles in target and non-target organs for carcinogenesis in mice. BMC Genomics 15:880, doi: 10.1186/1471-2164-15-880. [DOI] [PMC free article] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

1
2
3
4
5
6

RESOURCES