throbber
Articles
`
`Low-Dose Pharmacokinetics and Oral Bioavailability of Dichloroacetate in
`Naive and GSTζ-Depleted Rats
`Shakil A. Saghir* and Irvin R. Schultz
`Battelle Pacific Northwest National Laboratory, Richland, Washington, USA
`
`We studied the pharmacokinetics of dichloroacetate (DCA) in naive rats and rats depleted of glu-
`tathione S-transferase-zeta (GSTζ), at doses approaching human daily exposure levels. We also
`compared in vitro metabolism of DCA by rat and human liver cytosol. Jugular vein-cannulated
`male Fischer-344 rats received graded doses of DCA ranging from 0.05 to 20 mg/kg (intra-
`venously or by gavage), and we collected time-course blood samples from the cannulas. GSTζ
`activity was depleted by exposing rats to 0.2 g/L DCA in drinking water for 7 days before initia-
`tion of pharmacokinetic studies. Elimination of DCA by naive rats was so rapid that only 1–20
`mg/kg intravenous and 5 and 20 mg/kg gavage doses provided plasma concentrations above the
`method detection limit of 6 ng/mL. GSTζ depletion slowed DCA elimination from plasma,
`allowing kinetic analysis of doses as low as 0.05 mg/kg. DCA elimination was strongly dose
`dependent in the naive rats, with total body clearance declining with increasing dose. In the
`GSTζ-depleted rats, the pharmacokinetics became linear at doses ≤ 1 mg/kg. Virtually all of the
`dose was eliminated through metabolic clearance; the rate of urinary elimination was < 1
`mL/hr/kg. At higher oral doses (≥ 5 mg/kg in GSTζ-depleted and 20 mg/kg in naive rats), sec-
`ondary peaks in the plasma concentration appeared long after the completion of the initial
`absorption phase. Oral bioavailability of DCA was 0–13% in naive and 14–75% in GSTζ-
`depleted rats. Oral bioavailability of DCA in humans through consumption of drinking water was
`predicted to be very low and < 1%. The use of the GSTζ-depleted rat as a model for assessing the
`kinetics of DCA in humans is supported by the similarity in pharmacokinetic parameter estimates
`and rate of in vitro metabolism of DCA by human and GSTζ-depleted rat liver cytosol. Key
`words: animal study, dichloroacetic acid, drinking water disinfection by-products, halogenated
`acetic acids, human risk assessment, human in vitro metabolism, low-dose pharmacokinetics, oral
`bioavailability, rat in vitro metabolism, toxicology. Environ Health Perspect 110:757–763 (2002).
`[Online 13 June 2002]
`http://ehpnet1.niehs.nih.gov/docs/2002/110p757-763saghir/abstract.html
`
`Dichloroacetate (DCA) is a drinking water
`disinfectant by-product commonly identified
`in municipal water supplies. Concentrations
`of DCA in finished drinking water have been
`reported as high as 133 µg/L (1), although
`concentrations < 25 µg/L are more common
`(2,3). DCA is a metabolite of certain chlori-
`nated industrial solvents and of several phar-
`maceuticals (4). DCA has also been used for
`decades as an investigational drug to treat
`numerous cardiovascular and metabolic dis-
`orders in humans, for example, diabetes,
`hypercholesterolemia, and amelioration of
`lactic acid during liver transplantation (4–6).
`Recently, DCA has been used in clinical tri-
`als to treat congenital or acquired lactic
`acidosis in children (5,7). Human exposure
`to DCA ranges from ~1 to 4 µg/kg/day
`through consumption of drinking water and
`up to 50 mg/kg/day from the use of DCA as
`a therapeutic drug (4).
`DCA is rapidly and completely absorbed
`from the gastrointestinal (GI) tract and is
`extensively metabolized both in rodents and
`in humans, with glyoxylate, oxalate, glyco-
`late, and CO2 being the major metabolites
`(8,9). Only a small percentage (< 3%) of the
`dose is excreted as the parent compound
`(8–10). DCA metabolism occurs primarily in
`
`the liver (11), mediated through a recently
`characterized class of glutathione S-trans-
`ferase, GSTζ (GSTZ1–1) (12). DCA is also
`a mechanism-based inhibitor of GSTζ, and
`prolonged exposure to DCA in rodents
`causes both reduction in metabolism and
`depletion of immunoreactive GSTζ protein
`levels from the liver (9,13–15). Thus, in
`repeated dosings of DCA, the first dose is
`always cleared more rapidly than are subse-
`quent doses because of the inactivation of
`GSTζ. This has prevented measurement of
`oral bioavailability of DCA using a crossover
`experimental design because the second dose
`is always eliminated more slowly regardless of
`the route of administration (16).
`DCA has been associated with a number
`of toxic effects in animals exposed to high
`doses, including testicular abnormalities,
`birth defects, and liver cancer (17–20).
`Consumption of chlorinated water has been
`linked to increased risk for certain cancers in
`humans (21) without any specific correlation
`with DCA or other haloacetates. DCA pre-
`sents an interesting dilemma for risk asses-
`sors because it has a history of safe use as a
`therapeutic, but it has created regulatory
`concern because of its carcinogenicity in ani-
`mals. The U.S. Environmental Protection
`
`Agency (EPA) has classified DCA as a likely
`human carcinogen based on the hepatocar-
`cinogenic effects observed in rodents (22).
`Because of the prevalence of halogenated
`acetic acids in finished drinking water and
`their possible link to human cancer, the U.S.
`EPA has set standards permitting a com-
`bined total of 60 µg/L of five common halo-
`genated acetic acids (HAA5) in drinking
`water. The goal of the U.S. EPA is the vir-
`tual elimination of DCA from drinking
`water under stage I regulations (22).
`Previous pharmacokinetic studies of
`DCA have focused on therapeutic (i.e., mil-
`ligram per kilogram) doses. Also, the effects
`of GSTζ depletion on DCA disposition have
`not been studied in detail. Therefore, we
`designed this study to determine the phar-
`macokinetics and oral bioavailability of
`DCA in cohorts of naive and GSTζ -
`depleted rats using a range of doses down to
`50 µg/kg. We also compared the in vitro
`metabolism of DCA in human liver cytosol
`with cytosol obtained from naive and GSTζ-
`depleted rats. We made this comparison to
`aid in determining the appropriateness of
`using the GSTζ-depleted rat model for
`understanding low-dose pharmacokinetics of
`haloacetates in humans.
`Materials and Methods
`Chemicals. We purchased DCA (> 99%
`pure as free acid) from Fluka Chemical
`Corp. (Milwaukee, WI). Reagent-grade
`methyl-tert-butyl ether (MTBE) was pur-
`chased from Fisher Scientific (Pittsburgh,
`PA). We prepared diazomethane from
`
`Address correspondence to I.R. Schultz, Battelle
`MSL, 1529 West Sequim Bay Road, Sequim, WA
`98382 USA. Telephone: (360) 681-4566. Fax:
`(360) 681-3681. E-mail: ir_schultz@pnl.gov
`*Current address: Dow Chemical Company,
`Toxicology and Environmental Research and
`Consulting, Midland, MI, USA.
`We thank G. Muñiz, Y. Rivera, E. Robershotte,
`and N. Flintoff for their help in various parts of the
`study.
`Research described in this article has been funded
`wholly or in part by the U.S. Environmental
`Protection Agency through STAR grant R82594.
`The article has not been subjected to the U.S. EPA’s
`required peer and policy review and therefore does
`not necessarily reflect the views of the U.S. EPA,
`and no official endorsement should be inferred.
`This work was presented in part at the 40th
`annual meeting of the Society of Toxicology, San
`Francisco, CA, 25–29 March, 2001.
`Received 17 October 2001; accepted 22 January
`2002.
`
`Environmental Health Perspectives • VOLUME 110 | NUMBER 8 | August 2002
`
`757
`
`ALKERMES EXHIBIT 2032
`Amneal Pharmaceuticals LLC v. Alkermes Pharma Ireland Limited
`IPR2018-00943
`
`Page 1 of 7
`
`

`

`Articles • Saghir and Schultz
`
`N-methyl-N-nitrosoguanidine following
`Aldrich Technical Information Bulletin AL
`121 (23). All other chemicals were of the
`purest grade available and were obtained
`from standard sources. All dosing solutions
`were prepared in 0.9% (w/v) NaCl and pH
`adjusted to 7.0 with NaOH.
`Animals and treatment. The Institutional
`Animal Care and Use Committee of Battelle,
`Pacific Northwest National Laboratory
`approved the animal care and experimental
`protocols, and animal care and treatment was
`conducted in accordance with their estab-
`lished guidelines.
`For pharmacokinetic experiments, we
`purchased 8- to 10-week-old male Fischer
`344 rats (185 ± 29 g body weight, mean ±
`SD; n = 41 naive rats) fitted with a jugular
`vein cannula from Taconic Laboratories
`(Germantown, NY). We also purchased 6
`noncannulated rats from Charles River
`Laboratories (Raleigh, NC) for the prepara-
`tion of liver cytosol. We housed jugular
`vein–cannulated rats individually, whereas we
`housed three noncannulated rats per cage.
`Each cage contained wood-chip bedding and
`stainless steel wire tops, and rats were housed
`under standard conditions (22°C, 40–60%
`relative humidity, 12-hr light/dark cycle). We
`allowed rats a minimum of 48 hr for recovery
`from transport before use in experiments.
`Initially, rats were provided with deion-
`ized water and Purina rat chow (St Louis,
`MO) ad libitum. We used deionized water
`throughout the experiments to avoid
`unwanted exposure to haloacetates, which
`can be present in drinking water sources and
`may cause some inactivation of GSTζ .
`Animals were fasted overnight before the
`administration of DCA. We dosed naive ani-
`mals intravenously (iv) or by gavage
`(4–6/dose group) with 1, 5, or 20 mg/kg
`DCA and housed them in polycarbonate
`metabolism cages. After the initial dosing
`experiments, we then provided the same
`individual rats with 0.2 g/L DCA in their
`drinking water for 7 days to deplete/inacti-
`vate GSTζ activity (henceforth GSTζ
`depleted). We also pretreated one group of
`three noncannulated rats for 7 days with 0.2
`g/L DCA in their drinking water to deplete
`GSTζ. We then switched the GSTζ-depleted
`animals to non-DCA–fortified water
`overnight (16 hr) to allow residual DCA to
`be cleared from the body. This treatment
`protocol was previously shown to reduce
`GSTζ activity by > 90% in rat liver cytosol
`(24), and the experimental results presented
`in this study further confirm this finding. We
`then dosed GSTζ-depleted rats (4–6 per dose
`group) iv with 0.05, 0.25, 1, 5, or 20 mg/kg
`or gavaged them with 0.25, 1, 5, or 20
`mg/kg DCA. We gavaged two additional
`GSTζ-depleted rats with 100 mg/kg DCA to
`
`estimate oral bioavailability at this highest
`dose using earlier reported iv data of
`Gonzalez-Leon et al. (9). We administered
`dosing solutions at a volume of 1 mL/kg.
`Sample collection and analysis. We col-
`lected serial blood samples (0.075–0.125 mL)
`from individual rats through the jugular vein
`cannula using a 1-mL syringe coated with
`sodium heparin. After each blood sample, we
`flushed the cannula with ~0.2 mL of
`heparinized saline (40 U/mL heparin). We
`obtained plasma by centrifugation, mixed it
`with 0.2 mL of ice-cold 0.1 M sodium acetate
`buffer (pH 5.2), and stored the plasma at
`–20°C until analysis. We determined actual
`plasma volumes gravimetrically using tared
`vials and assuming plasma density of 1.0. A
`typical blood sampling schedule after iv dos-
`ing was 0, 3, 6, 10, 15, 20, 25, and 30 min
`and variously thereafter up to 24 hr, depend-
`ing on the dose and pretreatment. For orally
`dosed animals, we added an additional 1-min
`sample. Sampling continued until plasma
`concentrations were expected to have declined
`below the method detection limit (MDL) for
`DCA. We calculated the MDL as described
`by Glaser et al. (25) using nine replicate
`plasma samples. The MDL for DCA in naive
`and GSTζ-depleted rat plasma was 6 and 10
`ng/mL, respectively. The MDL for plasma
`removed from GSTζ rats was slightly higher
`because of elevated background levels. We
`collected urine from each rat for 24 hr, mixed
`an aliquot with sodium acetate buffer, and
`stored the samples at –20°C until analysis.
`For experiments measuring diurnal
`changes in plasma DCA levels, we provided
`four rats with 0.2 g/L DCA water and col-
`lected time-course blood samples up to 24
`hr, after which we gave animals non-DCA
`water and collected the last blood samples 11
`hr later. We allowed animals used in the
`kinetic experiments an additional 5 hr on
`non-DCA water (16 hr total depuration
`before administering DCA). This additional
`time was to ensure that residual DCA con-
`centration in plasma approached back-
`ground values without significant resynthesis
`of the GSTζ enzyme (26). We verified resid-
`ual concentration after depuration by mea-
`suring DCA levels in plasma collected before
`dosing (time 0) and comparing them with
`levels in non-DCA treated rats.
`We analyzed all plasma samples antici-
`pated to contain > 100 ng DCA using a pre-
`viously described method (27). Briefly, we
`added 0.025 mL (0.2 µg) internal standard
`(dibromoacetic acid) to samples (plasma and
`urine), acidified them by adding 0.025 mL
`50% sulfuric acid (v/v), and extracted in vari-
`ous volumes (0.2–1.0 mL) of MTBE
`depending on the dose and sampling time.
`We extracted samples anticipated to contain
`< 100 ng DCA in 0.2 mL MTBE. We then
`
`concentrated the extracted DCA by reducing
`the volume of MTBE to 0.01–0.02 mL
`under a gentle stream of N2. We converted
`the free acid to the methyl ester by adding
`0.01–0.02 mL ethereal diazomethane (previ-
`ously diluted 1:10 with MTBE). We then
`analyzed samples by gas chromatography
`with electron-capture detection (Hewlett-
`Packard 5890-Series II, Avondale, PA). The
`additional preconcentration step increased
`the MDL for DCA by 50- to 75-fold when
`compared to our previous method. We deter-
`mined stability of DCA in urine by fortifying
`freshly collected urine from a naive rat with
`DCA (10 µg); the fortified samples were
`either stored at –20°C or left at room tem-
`perature for 24 hr. We then analyzed DCA
`as described above and compared the results.
`Degradation of DCA in urine was negligible;
`> 90% could be recovered from urinary sam-
`ples left at room temperature for 24 hr.
`Kinetic analysis. The methods we used
`to analyze the concentration–time profiles of
`DCA were similar to those used by Schultz
`et al. (27). Briefly, we analyzed the individ-
`ual plasma profiles after both iv and oral
`administration by noncompartmental meth-
`ods to obtain estimates of total body clear-
`ance, apparent volume of distribution at
`steady state (Clb, Vss, for iv doses only), and
`the mean residence time (MRT) using the
`standard equations for these parameters that
`are incorporated into the WinNonlin
`program (Pharsight Corp., Cary, NC).
`WinNonlin calculates the area under the
`curve (AUC0→∞) by the linear trapezoidal
`method with the terminal portion of the
`curve extrapolated from time 0 to infinity by
`Cp,t/β, where Cp,t is the concentration of
`DCA in plasma at the last observation and β
`is the slope of the terminal phase determined
`by linear regression. WinNonlin calculated
`the elimination half-life (t1/2,β) as 0.693/β.
`We calculated renal clearance as Clr =
`Χu0→24/AUC0→24, where Χu0→24 is the total
`amount of DCA recovered in the urine after
`24 hr. We also report the observed peak
`plasma concentration of DCA (Cmax) and the
`time of its occurrence (Tmax) after oral dos-
`ing. We calculated the oral bioavailability
`from the ratios of the average values for
`AUC0→∞ for the oral and iv doses, and calcu-
`lated the mean absorption time (MAT) as the
`difference between the MRToral and MRTiv.
`Preparation of liver cytosol. We prepared
`rat liver cytosol from male F-344 rats (8–10
`weeks old; n = 3 naive and 3 GSTζ depleted)
`by differential centrifugation as described by
`Okita and Okita (28). We purchased two
`human liver sections and a pooled S-9 frac-
`tion obtained from 10 donors from the
`International Institute for the Advancement
`of Medicine (Exton, PA). The human liver
`section designated Human 1 was from a
`
`758
`
`VOLUME 110 | NUMBER 8 | August 2002 • Environmental Health Perspectives
`
`Page 2 of 7
`
`

`

`Articles • Kinetics of DCA in naive and GSTζ-depleted rats
`
`blood flow (18%) to the liver (33). For
`humans, we assumed total cardiac output to
`be 312 L/hr and liver blood flow to be 22%
`of this value [from Astrand (34) and
`Williams and Leggett (35), respectively].
`The calculated Qh for a 70 kg human was
`1.01 L/hr/kg. The unbound fraction for
`DCA in rat plasma was 0.94 ± 0.07 (27) and
`we assumed it to be the same for humans.
`Statistics. We assessed significant differ-
`ences between pharmacokinetic parameter
`estimates from the different treatment groups
`by Student’s t-test. We also performed analy-
`sis of variance on the individual Clb values to
`determine if they were significantly different.
`We considered a p-value of ≤ 0.05 to be sta-
`tistically significant.
`Results
`Plasma DCA levels while receiving DCA
`treatment. To study the pharmacokinetics of
`DCA in rats with reduced metabolism, we
`exposed animals to DCA (0.2 g/L) in drink-
`ing water for 7 days to effectively deplete the
`GSTζ enzyme. Periodically, we monitored
`consumption of water and plasma DCA lev-
`els to verify concentrations during the expo-
`sure and residual levels after 16 hr of
`washout. The average consumption of water
`by rats was 80 mL/kg/day, corresponding to
`a daily DCA dose of around 16 mg/kg.
`Figure 1 shows the diurnal plasma levels of
`DCA in rats provided with 0.2 g/L DCA-
`fortified water. DCA plasma levels were
`much higher during the dark cycle. We
`
`observed peak plasma levels at 500 hr (1 hr
`before lights on), which declined thereafter.
`The levels of DCA in plasma started to
`climb again 2 hr before the lights were
`turned off, corresponding to the increased
`activity (drinking/eating). We found the
`minimum plasma concentration of DCA at
`1600 hr. During an 11-hr depuration period
`after removal of DCA-fortified drinking
`water, plasma levels of DCA rapidly dropped
`to 10 ± 2 ng/mL (Figure 1). We provided
`animals used in kinetic experiments an addi-
`tional 5 hr of washout (16 hr total depura-
`tion) that allowed DCA plasma levels to fall
`below 10 ng/mL.
`Intravenous administration. Figure 2
`shows the mean (± SE) plasma concentra-
`tion–time profiles. The decline of DCA from
`plasma of naive rats was so rapid that the low-
`est dose that could be used for kinetic analysis
`was 1 mg/kg, which had a plasma elimination
`half-life of approximately 4 min (Figure 2A,
`Table 1). In contrast, elimination of DCA
`from the plasma of GSTζ-depleted rats was
`much slower, allowing kinetic analysis of doses
`as low as 0.05 mg/kg (Figure 2B inset). Visual
`inspection of the plasma concentration–time
`profiles and the pharmacokinetic parameters
`presented in Table 1 for the naive rats indicate
`that DCA declined from plasma in a monoex-
`ponential manner. Decline of DCA from
`plasma of the GSTζ-depleted rats became
`biexponential at the higher doses (Figure 2B).
`Table 1 summarizes the kinetic analysis
`of DCA for the naive and GSTζ-depleted
`
`Light /dark cycle
`
`DCA removed
`
`20
`
`10
`
`1
`
`0.1
`
`Concentration in plasma (µg/mL)
`
`0.01
`
`DCA in water
`
`2400
`
`400
`
`800
`
`1200
`
`1600
`Clock time
`
`2000
`
`2400
`
`400
`
`800
`
`Figure 1. Diurnal plasma concentration profile of DCA in rats administered 0.2 g/L DCA in drinking water
`and its elimination upon removal of the DCA-fortified water. Each point indicates mean ± SE (n = 4).
`
`69-year-old white male (body weight 90 kg;
`height, 1.78 m) who died of cardiopul-
`monary arrest; the Human 2 liver section was
`from a 68-year-old white female (body
`weight 73 kg; height 1.63 m) who died of a
`brain stem infarction. The pooled S9 desig-
`nated Human Pooled was prepared from
`liver tissue obtained from 10 white male
`donors of 8, 40, 40, 48, 51, 52, 53, 58, 63,
`and 64 years of age, who died of cardiovascu-
`lar disease, brain hemorrhage, anoxia, anoxia,
`head injury, anoxia, stroke, head injury,
`anoxia, and head injury, respectively. We
`prepared cytosol from the liver sections as
`described for rats by Okita and Okita (28)
`and from pooled human S-9 by centrifuga-
`tion at 100,000 × g for 1 hr. Liver sections
`had been perfused with University of
`Wisconsin medium and contained viable
`hepatocytes. We stored aliquots of cytosol at
`–70°C until use, and determined protein
`concentrations as described by Bradford (29).
`DCA depletion in cytosol and determi-
`nation of intrinsic metabolic clearance. We
`measured the depletion of added DCA using
`rat and human hepatic cytosol. Incubation
`mixtures consisted of 1–4 mg/mL protein,
`0.1 M phosphate buffer (pH 7.4), and 1.4
`mM glutathione in a final incubation vol-
`ume of 3 mL. We preincubated solutions for
`2 min at 37°C in a shaking water bath and
`started the reaction by adding 0.025 mg
`DCA prepared in 0.1 M phosphate buffer
`(pH 7.4). We removed a 0.15-mL aliquot
`from each incubate at various times (0.2–60
`min) and added it to a mixture of 0.25 mL
`0.1 M sodium acetate and 0.05 mL of 50%
`H2SO4 to stop the reactions. We added
`internal standard to each aliquot and
`extracted and analyzed DCA as described
`above. We plotted the loss of DCA against
`time and calculated AUC as described above.
`We calculated the intrinsic metabolic clear-
`ance (Clint) by dividing the initial amounts
`of DCA (at time 0.2 min) in the incubation
`medium with that of the AUC values (30).
`We scaled up the Clint to a whole animal/
`human by calculating the amount of cytoso-
`lic protein per gram of liver for rat and
`humans. For rats, we used measured liver and
`body weights. For humans, we assumed liver
`to be 2.5% of the body weight as reported by
`Davies and Morris (31). We calculated the
`hepatic clearance as
`


`
`Cl
`Cl
`
`,
`
`)int
`
`[1]
`
`=
`
`h
`

`(
`+
`
`Q
`Q
`
`u
`
`f
`f
`
`Cl
`h
`
`h
`u
`int
`where Qh is the liver blood flow and fu is the
`unbound fraction of DCA in plasma. We
`assumed the total cardiac output for F-344
`rats was 17.38 L/hr/kg [from Hachamovitch
`et al. (32)]. We calculated the Qh to be 3.13
`L/hr/kg by adjusting for the percentage
`
`Environmental Health Perspectives • VOLUME 110 | NUMBER 8 | August 2002
`
`759
`
`Page 3 of 7
`
`

`

`disproportionate increase in the AUC0→∞
`between the doses (Table 2). Maximum
`plasma concentrations were reached within
`5–10 min in naive and 8–45 min in GSTζ-
`depleted rats (Table 2, Figure 3). In the
`GSTζ-depleted rats, peak plasma concentra-
`tions (Cmax) were 4- to 6-fold higher than in
`the naive rats. The higher Cmax and longer
`MRT in GSTζ -depleted rats was also
`reflected by a 22- to 56-fold increase in the
`AUC0→∞ (Table 2). The MAT in both
`naive and GSTζ-depleted rats was increased
`in a dose dependent manner.
`The oral bioavailability of DCA was sig-
`nificantly reduced in naive rats. At doses of 5
`and 20 mg/kg, bioavailability was only 10%
`and 13%, respectively. At a higher dose of
`100 mg/kg, the oral bioavailability reached
`81% (Table 2). Bioavailability at 1 mg/kg
`could not be calculated because of the lack
`
`of detectable concentrations of DCA in
`plasma. In GSTζ-depleted rats, the oral
`bioavailability was 14%, 29%, 31%, and
`75% at the 0.25, 1, 5, and 20 mg/kg doses,
`respectively, and became 100% at 100
`mg/kg (Table 2).
`Correlation between dose and kinetic
`parameters. Figure 4 presents the correlation
`between dose and percent oral bioavailabil-
`ity. The relationship between dose and oral
`bioavailability of DCA for the GSTζ-
`depleted rats was best defined by saturable
`mechanism using a hyperbolic distribution
`with a correlation coefficient (r 2) of 0.90
`(Figure 4). The relationship between dose
`and oral bioavailability for the naive rats was
`less clear, and Figure 4 shows only the
`observed data.
`Cytosolic metabolism of DCA. The results
`of in vitro experiments using liver cytosol
`
`A
`
`100
`
`B
`
`1.0
`
`0.1
`
`0.01
`
`0.0
`
`0.5
`Time (hr)
`
`1.0
`
`10
`
`1
`
`0.1
`
`Plasma concentration (µg/mL)
`
`0.01
`
`0.005
`
`20 mg/kg
`5 mg/kg
`1 mg/kg
`0.25 mg/kg
`0.05 mg/kg
`
`100
`
`10
`
`1
`
`0.1
`
`Plasma concentration (µg/mL)
`
`0.01
`
`0.005
`
`0.0
`
`0.5
`
`1.5
`
`2.0
`
`0
`
`2
`
`4
`
`8
`
`10
`
`12
`
`6
`1.0
`Time (hr)
`Time (hr)
`Figure 2. The plasma concentration–time profiles of DCA after iv administration of a series of doses of
`1–20 mg/kg to naive (A) and 0.05–20 mg/kg to GSTζ-depleted rats (B). Data shown are mean ± SE (n =
`4–6). Error bars that fit within the data point are not shown. To avoid crowding of data points on the y-
`axis, the line was slightly shifted to the left.
`
`Table 1. Pharmacokinetic parameters of DCA after iv administration of a range of doses in naive and
`GSTζ-depleted adult male F-344 rats.
`Dose
`AUC0→∞
`(mg/kg)
`(µg/mL/hr)
`Naive rats
`1
`5
`20
`100b,c
`GSTζ-depleted rats
`0.05
`0.25
`1
`5
`20
`100c
`NR, not reported.
`aClrenal was < 0.7 mL/hr with the exception of 100 mg/kg (2.9 ± 0.5 mL/hr for naive and 8.9 ± 3.3 mL/hr for GSTζ-depleted)
`doses. bData from Schultz et al. (27). cData from Gonzalez-Leon et al. (9). dNot significantly different from each other: p ≥ 0.4.
`
`Vss
`(mL/kg)
`
`Clb
`(mL/hr/kg)a
`
`MRT
`(hr)
`
`t 1/2
`(hr)
`
`0.07 ± 0.01
`0.08 ± 0.01
`0.14 ± 0.01
`2.10 ± 0.86
`
`0.23 ± 0.05
`0.18 ± 0.03
`0.19 ± 0.04
`0.64 ± 0.04
`3.45 ± 0.09
`NR
`
`0.07 ± 0.001
`0.08 ± 0.003
`0.15 ± 0.01
`2.4 ± 0.15
`
`0.19 ± 0.05
`0.17 ± 0.02
`0.20 ± 0.05
`0.50 ± 0.03
`1.81 ± 0.09
`10.8 ± 2.0
`
`No.
`
`5
`6
`5
`5
`
`4
`6
`5
`4
`4
`6
`
`0.15 ± 0.01
`1.24 ± 0.05
`13.8 ± 0.85
`433 ± 233
`
`0.04 ± 0.01
`0.15 ± 0.03
`0.61 ± 0.02
`8.21 ± 0.50
`136.6 ± 3.4
`2,410 ± 406
`
`508 ± 68.6
`415 ± 47.2
`223 ± 111.0
`618 ± 319.0
`
`277 ± 33.4
`454 ± 57.3
`261 ± 13.6
`392 ± 31.4
`513 ± 18.5
`582 ± 146
`
`6,554 ± 356
`5,265 ± 636
`1,571 ± 97
`267 ± 105
`
`1,326 ± 342d
`1,816 ± 288d
`1,640 ± 57d
`614 ± 39
`168 ± 22
`43 ± 8
`
`Articles • Saghir and Schultz
`
`rats. In general, the kinetics of DCA were sim-
`ilar to previous descriptions (9,27): rapid elim-
`ination by the naive rats with GSTζ depletion
`causing an increase in t1/2,β and MRT and a
`decrease in the total body clearance (Clb).
`DCA was essentially eliminated through
`metabolism by both naive and GSTζ-depleted
`rats. The renal clearance of DCA accounted
`for < 1% of the total body clearance at most
`doses (Table 1). The steady-state volume of
`distribution (Vss) did not appear to be affected
`by GSTζ depletion or with dose, ranging non-
`systematically between 223 and 618 mL/kg
`(Table 1). The main pharmacokinetic parame-
`ter that was affected by treatment and dose
`was Clb. In naive rats, we observed nonlinear
`kinetics throughout the dosing regimen (Table
`1). In GSTζ-depleted rats, however, the phar-
`macokinetics became linear at doses < 1
`mg/kg; Clb was not different (p ≥ 0.4) at these
`lower doses (Table 1).
`Oral administration. The average
`plasma concentration–time profiles of DCA
`after gavage dosing are presented in Figure 3
`and a summary of the pharmacokinetic para-
`meters is presented in Table 2. DCA was
`rapidly absorbed after oral dosing and
`detected in plasma within 1 min after dosing
`(Figure 3). In naive rats, the greater capacity
`for metabolism limited the doses that could
`be used. Pilot experiments using a dose of 1
`mg/kg failed to detect plasma concentrations
`above the MDL (6 ng/mL) because DCA
`was apparently completely metabolized
`before reaching the general circulation
`(Table 2). The decline in the plasma concen-
`tration of DCA after the initial peak
`appeared to be monoexponential at lower
`doses (5 mg/kg in the naive and ≤ 1 mg/kg
`in the GSTζ-depleted rats; Figure 3). At
`higher doses (20 mg/kg in naive rats and ≥ 5
`mg/kg in GSTζ-depleted rats), DCA dis-
`played complex plasma concentration–time
`profiles, with secondary plasma peaks
`appearing between 4–8 hr after dosing, long
`after completion of the initial absorption
`phase (Figure 3). This observation is consis-
`tent with a previous report of the absorption
`of DCA in naive rats gavaged with a 100
`mg/kg dose (27). In GSTζ-depleted rats, the
`secondary plasma peak was less apparent at
`the highest gavaged dose of 20 mg/kg
`(Figure 3B). This observation implies that a
`complex dose–response relationship exists in
`GSTζ-depleted rats between the oral dose
`and appearance of the secondary plasma
`peaks, with both very low and high doses
`displaying a less pronounced secondary peak.
`This relationship may also apply to naive
`rats, although the high dose needed to
`obscure the secondary peak is apparently
`> 100 mg/kg.
`The complex plasma concentration–time
`profile at the higher doses contributed to the
`
`760
`
`VOLUME 110 | NUMBER 8 | August 2002 • Environmental Health Perspectives
`
`Page 4 of 7
`
`

`

`Articles • Kinetics of DCA in naive and GSTζ-depleted rats
`
`were consistent with in vivo kinetic analysis;
`that is, GSTζ depletion significantly
`decreases DCA metabolism/elimination. The
`rate of DCA metabolism by naive rat cytosol
`was significantly faster (p < 0.01) than that in
`the GSTζ-depleted rats (Figure 5). The
`intrinsic metabolic clearance (Clint) of DCA
`by the human liver cytosol closely resembled
`that in GSTζ-depleted rats (Figure 5) and
`was not statistically different (p > 0.3). Table
`3 presents the predicted hepatic clearance
`(Clh) and extraction efficiency (Ess) of DCA
`by rats and humans. We derived these pre-
`dicted values using in vitro Clint of DCA by
`the rat and human liver cytosol. The Clh and
`Ess of DCA by naive rats was 3-fold higher
`than that by the GSTζ-depleted rats. We
`predict humans to have somewhat lower Clh
`and similar E ss of DCA compared with
`GSTζ-depleted rats (Table 3).
`
`Discussion
`The results of this study demonstrate that
`elimination of DCA in naive rats exhibits
`nonlinear behavior at all doses that allowed
`pharmacokinetic analysis. The Clb continued
`to increase at lower iv doses and exceeded 6.5
`L/hr/kg at the 1 mg/kg dose. The cardiac out-
`put in F-344 rats of body size comparable to
`those used in this study has been reported to
`be 17.38 L/hr/kg (32). Therefore, the clear-
`ance of DCA in naive rats at low doses is at
`least 38% of cardiac output, which would
`exceed liver blood flow and implies extensive
`extrahepatic elimination of DCA occurs. In
`contrast, the pharmacokinetics of DCA in
`GSTζ-depleted rats becomes linear at doses
`≤ 1 mg/kg (Table 1); the Clb ranged between
`1.33 and 1.82 L/hr/kg after iv doses of
`0.05–1 mg/kg. Assuming liver blood flow is
`3.13L/hr/kg (i.e., fraction of cardiac output to
`
`B
`
`0.1
`
`0.01
`0.0
`
`0.5
`Time (hr)
`
`1.0
`
`30
`
`10
`
`1
`
`0.1
`
`Plasma concentration (µg/mL)
`
`0.01
`
`0.005
`
`A
`
`20 mg/kg
`5 mg/kg
`1 mg/kg
`0.25 mg/kg
`
`30
`
`10
`
`1
`
`0.1
`
`Plasma concentration (µg/mL)
`
`0.01
`
`0.005
`
`0
`
`2
`
`6
`
`8
`
`0
`
`4
`
`8
`
`16
`
`20
`
`24
`
`12
`4
`Time (hr)
`Time (hr)
`Figure 3. The plasma concentration–time profiles of DCA after oral administration of 5 and 20 mg/kg to
`naive rats (A) and a series of doses ranging from 0.25 to 20 mg/kg to GSTζ-depleted rats (B). Data shown
`are mean ± SE (n = 4–6). Error bars that fit within the data point are not shown. To avoid crowding of data
`points on the y-axis, the line was slightly shifted to the left.
`
`the liver is 0.18) (33) and liver metabolism
`accounts for the bulk of DCA elimination (in
`GSTζ-depleted rats), DCA clearance appears
`to correspond to 42–58% of liver blood flow.
`This finding indicates that DCA is moder-
`ately extracted by liver under linear kinetics
`by GSTζ-depleted rats. At higher doses,
`metabolism becomes saturated, and liver
`extraction decreases.
`The complex plasma concentration–time
`profiles of DCA observed after some oral
`doses (Figure 3) agreed with those in an ear-
`lier report (27). The extent of the secondary
`peaks appears to be reduced at lower doses
`and was absent after doses that only pro-
`duced detectable levels of DCA until 4 hr
`after dosing (5 and 20 mg/kg in this study).
`
`Naive
`GST ζ-depleted
`
`100
`
`90
`
`80
`
`70
`
`60
`
`50
`
`40
`
`30
`
`20
`
`10
`
`0
`
`Oral bioavailability (%)
`
`75
`
`100
`
`0
`
`25
`
`50
`Dose (mg/kg)
`Figure 4. Correlation of dose and oral bioavailability
`of DCA for the naive and GSTζ-depleted rats.
`Y = 106.8x ÷ (8.32 + x). r2 = 0.90. To avoid crowding
`of data points on the y-axis, the line was slightly
`shifted to the left.
`
`Human 1 Human 2 Human
`pooled
`
`*
`
`3.5
`
`3.0
`
`2.5
`
`2.0
`
`1.5
`
`1.0
`
`0.5
`
`Intrinsic metabolic clearance
`
`(mL/hr/mg protein)
`
`0.0
`
`Naive rat
`
`GST ζ-
`depleted
`rat
`Figure 5. Intrinsic metabolic clearance of DCA
`calculated from the progress metabolism experi-
`ment using liver cytosol from naive and GSTζ-
`depleted rats (n = 3). Human hepatic cytosol was
`from two donors or pooled from 10 individuals. For
`rats, each bar represents the mean ± SE (n = 3);
`for humans, each bar represents the mean ± SE of
`three experiments. Error bars that fit within the
`data point are not shown.
`*p < 0.01.
`
`Table 2. Pharmacokinetic parameters of DCA after oral administration of a range of doses in naive and
`GSTζ-depleted adult male F-344 rats.a
`Dose
`AUC0→∞
`(mg/kg)
`(µg/mL/hr)
`Naive rats
`1
`5
`20
`50b
`100c
`GSTζ-depleted rats
`0.25
`1
`5
`20
`100
`
`Tmax
`(hr)
`
`Cmax
`(µg)
`
`MRT
`(hr)
`
`MAT Bioavailability
`(hr)
`(%)
`
`BD
`0.12 ± 0.01
`1.82 ± 0.10
`11.7 ± 1.68
`218 ± 74.5
`
`0.02 ± 0.01
`0.18 ± 0.03
`2.58 ± 1.05
`103 ± 14.0
`2,730
`
`BD
`0.09 ± 0.02
`0.17 ± 0.01
`0.27 ± 0.04
`8.0
`
`0.13 ± 0.01
`0.12 ± 0.02
`0.25 ± 0.03
`0.75 ± 0.25
`3.32
`
`BD
`0.36 ± 0.07
`2.91 ± 0.24
`9.29 ± 1.87
`27.4
`
`0.08 ± 0.01
`0.26 ± 0.01
`1.66 ± 0.31
`17.0 ± 1.90
`151
`
`0
`0.28 ± 0.04
`1.70 ± 0.49
`NR
`6.70 ± 1.44
`
`0.39 ± 0.03
`1.05 ± 0.07
`1.80 ± 0.19
`4.63 ± 0.69
`12.5
`
`0
`0.20
`1.56
`NR
`4.5
`
`0.19
`0.86
`1.16
`1.18
`ND
`
`0
`9.68
`13.20
`ND
`80.93
`
`14.0
`29.4
`31.4
`75.0
`100
`
`No.
`
`3
`6
`6
`4
`5
`
`6
`4
`4
`4
`2
`
`Abbreviations: BD, below detection (< MDL); MAT, mean, absorption, time; ND, not determined; NR

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket