Abstract
Background
Dichloroacetate (DCA) is known to environmental scientists as a by-product of water chlorination and as a metabolite of industrial solvents, including Superfund chemicals. In contrast, DCA is studied by clinical investigators for its therapeutic potential in several life-threatening conditions, including genetic mitochondrial diseases, pulmonary arterial hypertension, and cancer. Thus, DCA holds an almost unique position at the interface between environmental science and allopathic medicine.
Objective
I critically reviewed laboratory and clinical pharmacological research on DCA to address questions about the current and future status of DCA in relation to human health.
Discussion
Recent information on the clinical toxicogenetics of DCA is interpreted particularly in light of its use as an investigational drug. Adverse effects from chronic DCA exposure have been identified in several target organs in animals. However, in humans, toxicity has so far been limited to reversible effects on the nervous system and liver. DCA is primarily biotransformed to glyoxylate by the bifunctional enzyme glutathione transferase zeta1 and maleylacetoacetate isomerase (GSTz1/MAAI), which also catalyzes the penultimate step in the phenylalanine and tyrosine catabolic pathway. DCA is a suicide inhibitor of GSTz1/MAAI, which can result in delayed plasma clearance of DCA and the accumulation of potentially toxic tyrosine intermediates. Age and GSTz1/MAAI haplotype can markedly affect the toxicokinetics of DCA in humans and rodents.
Conclusions
I have defined new potential avenues of research that focus on discrete human populations that may be at increased health risk or that may receive increased health benefit from chronic exposure to DCA at both environmentally and clinically relevant concentrations.
Keywords: cancer, dichloroacetate, glutathione transferase zeta, hereditary tyrosinemia, maleylacetoacetate isomerase, mitochondrial disease, peripheral neuropathy, pyruvate dehydrogenase, toxicogenetics
To many readers of this journal, dichloroacetate (DCA) has a bad reputation that is mainly the result of its association with Superfund chemicals and other halogenated hydrocarbons of ill repute (Stacpoole et al. 1998b), the findings of numerous animal studies implicating DCA in a variety of moderate to life-threatening toxicities [International Agency for Research on Cancer (IARC) 2004], and the ubiquity of this molecule in our biosphere that makes at least some degree of chronic exposure inevitable (Ammini and Stacpoole 2003; IARC 2004). Despite these concerns, the possibility that DCA might have a positive impact on certain human populations has been evident for half a century (Stacpoole 1989). Over time, DCA’s pharmacological profile has created an interesting dialectic between environmental toxicologists, who consider it to be a hazard to humans, and clinical investigators, who are pursuing its therapeutic potential. Most ironic is the growing controversy over the relevance of DCA to human cancer, in which it is viewed as both a likely cause (IARC 2004) and a possible treatment (Michelakis et al. 2010).
The DCA dilemma has been framed by three broad fields of science: epidemiological studies of populations exposed to environmental levels of measured by-products of water disinfection or Superfund chemicals that are precursors of DCA; mechanism-based toxicological experiments in which inbred rodent strains receive doses of DCA usually vastly higher than those to which most humans are exposed in the environment; and research in which subjects are administered DCA as a drug at doses similar to those used in animal toxicological investigations. Together, the data generated by such research address three questions that are at the heart of the DCA dilemma: Is DCA a significant environmental health hazard? Are certain populations at increased health risk or benefit? What are the important areas of future research that will help confirm, modify, or reject current opinions about DCA’s impact on human health?
Is DCA a significant environmental health hazard? Human populations have been exposed to DCA for generations, primarily by ingesting chlorinated drinking water that may contain up to 160 μg/L (Miller and Uden 1983). Thus, consuming 2 L of municipal water may result in a DCA dose of approximately 2–4 μg/kg body weight in a 70-kg human. However, studies of worldwide exposure to chlorinated drinking water in relation to human disease did not include an assessment of DCA exposure levels (Obolensky et al. 2003). Dermal absorption of DCA may occur when bathing or swimming in chlorinated pools or upon atmospheric exposure, although there are no good estimates of the contribution of dermal absorption to tissue or plasma levels of the compound. DCA is also considered a metabolite of the degreasing agent trichloroethylene (TCE) and related solvents, most of which are released into the atmosphere (Agency for Toxic Substances and Disease Registry 1997). TCE emissions and contamination of surface water vary widely across the United States but correlate broadly with population density and provide an estimated combined median daily intake by the general population of slightly > 13 μg/day (U.S. Geological Survey 2006). Both enzymatic (Lash et al. 2000) and nonenzymatic (Guengerich 2004) catalysis of TCE to DCA have been reported. However, the quantitative significance of either pathway is controversial (Ketcha et al. 1996; Merdink et al. 1998; Templin et al. 1995), which contributes to uncertainty about the relevance of environmental exposure levels of DCA to human health.
Nevertheless, considerable weight has been given to assigning causality to the oxidative metabolites of TCE [see Supplemental Material, Figure 1 (doi:10.1289/ehp.1002554)] in evaluating the toxicity of DCA. Data from population-based pharmacokinetic modeling of the toxicokinetics and risk assessment of TCE and its metabolites (Chiu et al. 2009; Evans et al. 2009) indicate that TCE exposure has been associated with toxicity to the kidney and liver and to the nervous, immune, and reproductive systems. How much of this information is relevant to the assessment of human risk from DCA exposure? Strong evidence from case–control studies indicates an association between TCE exposure and renal cancer in humans (Brüning et al. 2003; Charbotel et al. 2006), but no experimental or clinical evidence is currently available to implicate DCA as a cause of this malignancy. Indeed, no epidemiological investigations have studied the carcinogenicity of DCA in humans. Lee et al. (2003) found a much weaker epidemiological association between environmental exposure to TCE and liver cancer. In this case, however, the relationship between TCE and DCA becomes highly significant, because DCA is unquestionably a liver carcinogen in certain inbred strains of mice and rats (reviewed by IARC 2004; Stacpoole et al. 1998b). Most of the studies that have investigated the carcinogenic potential of DCA have used drinking water or, occasionally, gavage to administer the compound for weeks or months at effective doses usually of 75 mg/kg/day or more [see Supplemental Material, Text A (doi:10.1289/ehp.1002554)]. Regardless of the precise mechanism(s) that underlies DCA’s carcinogenic potential, it is clear that hepatotoxicity in rodents from DCA is obtainable only at doses thousands of times greater than those to which humans are normally exposed from the combined effects of the atmosphere and chlorinated drinking water. This fact, together with current data from population studies, does not support the postulate that exposure to DCA from environmental sources represents a general hazard to humans. Whether chronic exposure to DCA at concentrations encountered in the environment or in the clinic is problematic to select human populations is addressed below.
Are certain populations at increased health risk or benefit? As a medicinal agent, DCA has been administered as the sodium salt in doses ranging from 10 to > 100 mg/kg/day by intravenous and oral routes for a few days to > 10 years [see Supplemental Material, Text B (doi:10.1289/ehp.1002554)]. These clinical studies have reported occasional asymptomatic increases in serum concentrations of alanine aminotransferase and aspartate aminotransferase that seldom exceeded two to three times the pretreatment values of these enzymes and that returned to pretreatment levels within a few weeks after the DCA dose was reduced or stopped (Barshop et al. 2004; Berendzen et al. 2006; Stacpoole et al. 1998b). A more worrisome toxicity of DCA is reversible peripheral neuropathy, an effect replicated in rodents, dogs, and humans (Stacpoole 1989). Clinical features include tingling of the fingers and toes and weakness of the facial and distal muscles of the extremities, accompanied by reduction in nerve conduction velocity in the sural and tibial nerves [see Supplemental Material, Text C (doi:10.1289/ehp.1002554)].
One possible explanation for the hepatic and neurological toxicity of DCA relates to the unusual consequences of its biotransformation. DCA is dehalogenated to glyoxylate by the cytosolic zeta-1 family isoform of glutathione transferase (GSTz1), which is identical to maleylacetoacetate isomerase (MAAI), the penultimate enzyme in the phenylalanine/tyrosine catabolic pathway [see Supplemental Material, Figure 2 (doi:10.1289/ehp.1002554)]. DCA causes a mechanism-based inhibition of GSTz1/MAAI (Anderson et al. 1999), an effect so profound as to significantly delay the plasma clearance of DCA at both environmental (micrograms per kilogram per day) and clinical (micrograms per kilogram per day) exposure levels in humans (Henderson et al. 1997; Jia et al. 2006; Schultz and Shangraw 2006; Stacpoole et al. 1998a) [see Supplemental Material, Text D (doi:10.1289/ehp.1002554)].
Genetic or DCA-induced ablation of GSTz1/MAAI causes accumulation of the MAAI substrates maleylacetone and, presumably, maleylacetoacetate (MAA) (Ammini et al. 2003; Cornett et al. 1999; Lim et al. 2004; Tzeng et al. 2000), although the latter compound is too unstable to be detected by gas chromatography–mass spectrometry. Depending on exposure level, recovery of basal GSTz1/MAAI enzyme activity occurs over days to months after the withdrawal of DCA (Guo et al. 2006; Saghir and Schultz 2002). The toxicological significance of inhibition of MAAI is highlighted by the consequences of loss of function mutations in fumarylacetoacetate (FAA) hydrolase, the terminal enzyme of phenylalanine and tyrosine catabolism. Deficiency of this enzyme causes heredity tyrosinemia type 1 (hepatorenal tyrosinemia), an autosomal recessive disease that has an estimated prevalence of 100,000 persons worldwide (Mitchell et al. 2001). In this disease, accumulation of the substrates for both the hydrolase and MAAI-catalyzed reactions are thought to be responsible for the dramatically increased risk of hepatocellular carcinoma in affected young children, because of the ability of MAA and FAA to form adducts (Lantum et al. 2002). This property is also shared by glyoxylate (Anderson et al. 2004), the product of the GSTz1/MAAI-mediated dehalogenation of DCA.
Another important consequence of tyrosinemia type 1 is the diversion of carbon precursors to succinylacetone, high urinary levels of which are diagnostic for the disease. Succinylacetone is a competitive inhibitor of δ-aminolevulinate dehydratase (enzyme–inhibitor dissociation constant Ki = 0.3 μM; Sassa and Kappas 1983), which catalyzes an early step in heme biosynthesis. Inhibition of the dehydratase causes accumulation of δ-aminolevulinate, a neurotoxin that has been implicated in several peripheral neuropathies, including lead poisoning (see Felitsyn et al. 2008). Increased concentrations of δ-aminolevulinate or subsequent perturbations in heme metabolism or both are thought to be the cause of the neurological manifestations of tyrosinemia type 1, including peripheral neuropathy (Lindberg et al. 1999; Mitchell et al. 2001). Given the effects of DCA on GSTz1/MAAI function, it is noteworthy that increased urinary concentrations of maleylacetone and δ-aminolevulinate have been measured in children who were treated for months or years with DCA. For some of these children, maleylacetone levels approached or exceeded those found in children with hereditary tyrosinemia type 1 (Shroads AL, Langaee T, Coats BS, Kurtz TL, Bullock JR, Weithorn D, Gong Y, Wagner DA, Ostrov DA, Johnson JA, Stacpoole PW, unpublished observations) [see Supplemental Material, Text E (doi:10.1289/ehp.1002554)].
Recently, age and GSTz1/MAAI genotype have emerged as important determinants of clinical toxicity from DCA. For example, in a randomized clinical trial of 43 children (mean age at entry, 5.6 years) with congenital forms of lactic acidosis, Stacpoole et al. (2006) found no difference in clinical or biochemical adverse effects, including hematological, hepatic, and peripheral nerve function, between those children who received 25 mg/kg/day of sodium DCA and those who received placebo for 6 months. In contrast, a controlled clinical trial of 30 patients with mitochondrial encephalomyopathy, lactic acedosis, and stroke-like episodes (MELAS) whose mean age at entry was 30 years, ended prematurely because of drug-associated neurotoxicity (Kaufmann et al. 2006). In examining the plasma kinetics of DCA in a representative sample of children and adults from both trials, a striking age-dependent decrease was discovered in plasma DCA clearance that was replicated in rats (Shroads et al. 2008); this finding confirmed and extended earlier observations in rodents (James et al. 1998; Schultz et al. 2002). Furthermore, delayed clearance of DCA in rats was associated with increased urinary excretion of DCA, maleylacetone, and monochloroacetate (a putative neurotoxin and a very minor product of DCA metabolism) and with a decrease in urinary oxalate, an end product of DCA biotransformation [see Supplemental Material, Figure 1 and Text F (doi:10.1289/ehp.1002554)].
Age alone does not account entirely for variability in the kinetics and biotransformation of DCA. The human GSTz1/MAAI gene contains four nonsynonymous single-nucleotide polymorphisms, with products that show different activities toward DCA and variable frequencies among ethnic and racial groups (Lim et al. 2004; Stacpoole et al. 2008; Shroads AL, Langaee T, Coats BS, Kurtz TL, Bullock JR, Weithorn D, Gong Y, Wagner DA, Ostrov DA, Johnson JA, Stacpoole PW, unpublished observations). Using 1,2-13C-DCA, several researchers were able to clearly segregate “slow” and “fast” metabolizers of DCA, based on GSTz1/MAAI (Shroads AL, Langaee T, Coats BS, Kurtz TL, Bullock JR, Weithorn D, Gong Y, Wagner DA, Ostrov DA, Johnson JA, Stacpoole PW, unpublished observations). Those who metabolized DCA slowly showed markedly delayed plasma clearance and biotransformation, increased excretion of unmetabolized drug [see Supplemental Material, Figure 3 (doi:10.1289/ehp.1002554)], and increased urinary accumulation of maleylacetone and δ-aminolevulinate. Atomic modeling of the effect of GSTz1/MAAI polymorphism showed that variants associated with slow DCA metabolism induced structural changes in the enzyme homodimer leading to protein instability or abnormal protein–protein interactions. These data suggest that GSTz1/MAAI haplotype may predict, at least in part, the toxicogenetics of DCA. Moreover, such information may be used prospectively to adjust dosing and mitigate the risk of adverse events from DCA.
Despite the potential toxicity of DCA, its therapeutic potential continues to be investigated. Its principal site of pharmacodynamic action is the mitochondrial pyruvate dehydrogenase (PDH) complex (Stacpoole 1989), a gatekeeper enzyme for regulating carbohydrate fuel metabolism in cells [see Supplemental Material, Figure 4 (doi:10.1289/ehp.1002554)]. Under aerobic conditions, PDH catalyzes the rate-determining step in glucose oxidation by irreversibly decarboxylating pyruvate to acetyl coenzyme A, thereby “priming” the Krebs cycle with glucose carbon. In addition, both the PDH-catalyzed reaction and several enzymes of the Krebs cycle generate reducing equivalents in the forms of nicotine adenine dinucleotide and flavin adenine dinucleotide that are used by the respiratory chain for the reduction of molecular oxygen to water and for ATP synthesis from ADP. PDH is reversibly phosphorylated and inactivated by a family of PDH kinase (PDK) isoforms that exhibit variable tissue expression (Sugden and Holness 2003). DCA exerts differential inhibition on PDKs to maintain PDH in its unphosphorylated, catalytically active form and exerts greatest inhibition (Ki = 0.2 mM) (Bowker-Kinley et al. 1998) toward the ubiquitously expressed PDK2. Because pyruvate is in equilibrium with lactate, it follows that acquired or congenital deficiencies of PDH may lead to mitochondrial energy failure and lactate accumulation. Accordingly, DCA has been used to improve mitochondrial energetics and to mitigate tissue or systemic lactic acidosis (see Stacpoole et al. 1992, 2006 and references therein).
Invoking the same site and mechanism of action described above, several laboratories have reported DCA’s selective proapoptotic and antiproliferative effects when administered in vitro to a variety of cultured human tumor cells or to human tumors implanted into immunocompromised rodents (reviewed by Michelakis et al. 2008). The rationale for considering DCA as an anticancer drug is predicted on the well-established finding that solid tumors derive most of their energy from glycolysis, rather than through mitochondrial oxidative phosphorylation, even in the presence of adequate oxygen availability. This phenomenon is called the Warburg effect and is now known to be a consequence of the combined transactivation of glucose membrane transporters, glycolytic enzymes, and PDK (Vander Heiden et al. 2009). Consequently, PDH activity and oxidative phosphorylation are inhibited in most cancers. By inhibiting PDK in tumor cells, DCA stimulates PDH and oxidative phosphorylation, decreases tumor lactate concentration, and initiates a caspase-mediated chain of reactions leading to apoptosis of tumor, but not of host, cells (Bonnet et al. 2007) [see Supplemental Material, Text G (doi:10.1289/ehp.1002554)].
Primary arterial hypertension represents another neoplastic (but nonmalignant) condition in which mitochondrial dysfunction and hyperproliferation of cells lining the pulmonary vasculature lead to increased pulmonary vascular resistance, failure of the right ventricle of the heart, and premature death. In animal models of primary arterial hypertension, DCA has been shown to prevent or reverse mitochondrial damage and cellular proliferation, improve lung function, and increase survival by its ability to stimulate oxidative phosphorylation and induce apoptosis in proliferating cells (Bonnet et al. 2006; McMurtry et al. 2004; Michelakis et al. 2002). Results from clinical trials of DCA in this disease have not yet been reported [see Supplemental Material, Text H (doi:10.1289/ehp.1002554)].
What are important areas of future research? Few chemicals have integrated the disciplines of environmental science and clinical research as effectively as DCA. Recent studies investigating the toxicogenetics of this compound when administered as a drug have important implications for human populations chronically exposed to environmentally relevant concentrations. The findings are particularly germane to persons in contact with water sources contaminated with TCE or other industrial solvents or who are administered drugs that may be biotransformed to DCA (reviewed by Stacpoole et al. 1998b). The recent discovery of a mitochondrial GSTz1/MAAI (Li et al. 2011) indicates that both the biotransformation of DCA and the site of its major pharmacodynamic action, the PDH complex, occur within the matrix of this organelle. This finding raises intriguing questions about whether the stimulation of PDH activity and, subsequently, the respiratory chain by DCA could generate sufficient amounts of reactive oxygen species to cause oxidative damage to mitochondrial GSTz1/MAAI and further impair DCA metabolism.
New epidemiological research on DCA should focus more selectively on populations that may be predisposed to DCA-associated health complications because of age or genotype. For example, individuals with an inborn error of the phenylalanine and tyrosine catabolic pathway could suffer additional adverse consequences when exposed to clinically, and perhaps even environmentally, relevant doses of DCA. Individually, these diseases are rare. However, perhaps the most common of these conditions, hyperphenylalaninemia (which includes phenylketonuria), has a worldwide annual incidence that varies widely from approximately 3 to > 300 per million births (Scriver et al. 2001). Another population at potential risk is persons prone to hyperoxaluria. For example, primary hyperoxaluria has a prevalence of > 1 per 10,000 persons and may cause widespread calcium oxalate crystal deposition (Danpure 2001). Such individuals could be at heightened risk for developing calcium oxalate deposits were they to receive clinical doses of DCA over long periods of time.
Likewise, humans who metabolize DCA slowly may also be genetically predisposed to exhibit reduced flux through the phenylalanine and tyrosine catabolic pathway. Chronic consumption of a high-protein diet by such individuals could lead to an accumulation of toxic tyrosine intermediates, with adverse consequences for hepatic, neurological, and other organ system failure. Testing this postulate could involve studies of stable isotope amino acid kinetics in relation to GSTz1/MAAI haplotype. Finally, careful consideration must be given to age and genotype when conducting future treatment-oriented research with DCA, particularly cancer trials involving adults.
DCA has so far been proven to be neither a major environmental health hazard nor a panacea for human ills. Future clinical and environmental research on this unusual molecule should be focused increasingly on understanding its impact on biological processes in discrete subpopulations of humans with known GSTz1/MAAI genotype. We continue to move incrementally, but steadily, from puzzled observations about the pharmacology of DCA to established conclusions about its effect on human health.
Footnotes
Supplemental Material is available online (doi:10.1289/ehp.1002554 via http://dx.doi.org/).
The author thanks K. St. Croix for her editorial assistance.
This work was supported in part by grants from the National Institutes of Health [R01-ES07355, R01-ES014617, and U54RR025208-01 (Clinical and Translational Science Award)].
References
- Agency for Toxic Substances and Disease Registry. Toxic Substances Portal: Trichloroethylene (TCE). Toxicological Profile for Trichloroethylene. Atlanta, GA: Centers for Disease Control and Prevention; 1997. [[accessed 9 September 2010]]. Available: http://www.atsdr.cdc.gov/toxprofiles/tp.asp?id=173&tid=30. [Google Scholar]
- Ammini CV, Fernandez-Canon J, Shroads AL, Cornett R, Cheung J, James MO, et al. Pharmacologic or genetic ablation of maleylacetoacetate isomerase increases levels of toxic tyrosine catabolites in rodents. Biochem Pharmacol. 2003;66:2029–2038. doi: 10.1016/j.bcp.2003.07.002. [DOI] [PubMed] [Google Scholar]
- Ammini CV, Stacpoole PW. Biotransformation, toxicology and pharmacogenomics of dichloroacetate. In: Gribble GW, editor. Natural Production of Organohalogen Compounds. Handbook of Environmental Chemistry 3/P. Berlin: Springer; 2003. pp. 215–234. [Google Scholar]
- Anderson WB, Board PG, Anders MW. Glutathione transferase zeta-catalyzed bioactivation of dichloroacetic acid: reaction of glyoxylate with amino acid nucleophiles. Chem Res Toxicol. 2004;17:650–662. doi: 10.1021/tx034099+. [DOI] [PubMed] [Google Scholar]
- Anderson WB, Board PG, Gargano B, Anders MW. Inactivation of glutathione transferase zeta by dichloroacetic acid and other fluorine-lacking alpha-haloalkanoic acids. Chem Res Toxicol. 1999;12:1144–1149. doi: 10.1021/tx990085l. [DOI] [PubMed] [Google Scholar]
- Barshop BA, Naviaux RK, McGowan KA, Levine F, Nyhan WL, Loupis-Geller A, et al. Chronic treatment of mitochondrial disease patients with dichloroacetate. Mol Genet Metab. 2004;83:138–149. doi: 10.1016/j.ymgme.2004.06.009. [DOI] [PubMed] [Google Scholar]
- Berendzen K, Theriaque D, Shuster J, Stacpoole PW. Therapeutic potential of dichloroacetate for pyruvate dehydrogenase complex deficiency. Mitochondrion. 2006;6:126–135. doi: 10.1016/j.mito.2006.04.001. [DOI] [PubMed] [Google Scholar]
- Bonnet S, Archer SL, Allalunis-Turner J, Haromy A, Beaulieu C, Thompson R, et al. A mitochondria-K+ channel axis is suppressed in cancer and its normalization promotes apoptosis and inhibits cancer growth. Cancer Cell. 2007;11:37–51. doi: 10.1016/j.ccr.2006.10.020. [DOI] [PubMed] [Google Scholar]
- Bonnet S, Michelakis ED, Porter CJ, Andrade-Navarro MA, Thébaud B, Bonnet S, et al. An abnormal mitochondrial-hypoxia inducible factor-1alpha-Kv channel pathway disrupts oxygen sensing and triggers pulmonary arterial hypertension in fawn hooded rats: similarities to human pulmonary arterial hypertension. Circulation. 2006;113:2630–2641. doi: 10.1161/CIRCULATIONAHA.105.609008. [DOI] [PubMed] [Google Scholar]
- Bowker-Kinley MM, Davis WI, Wu P, Harris RA, Popov KM. Evidence for existence of tissue-specific regulation of the mammalian pyruvate dehydrogenase complex. Biochem J. 1998;329:191–196. doi: 10.1042/bj3290191. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brüning T, Pesch B, Wiesenhütter B, Rabstein S, Lammert M, Baumuller A, et al. Renal cell cancer risk and occupational exposure to trichloroethylene: results of a consecutive case–control study in Arnsberg, Germany. Am J Ind Med. 2003;43:274–285. doi: 10.1002/ajim.10185. [DOI] [PubMed] [Google Scholar]
- Charbotel B, Fevotte J, Hours M, Martin JL, Bergeret A. Case–control study on renal cell cancer and occupational exposure to trichloroethylene. Part II: epidemiological aspects. Ann Occup Hyg. 2006;50:777–787. doi: 10.1093/annhyg/mel039. [DOI] [PubMed] [Google Scholar]
- Chiu WA, Okino MS, Evans MV. Characterizing uncertainty and population variability in the toxicokinetics of trichloroethylene and metabolites in mice, rats, and humans using an updated database, physiologically based pharmacokinetic (PBPK) model, and Bayesian approach. Toxicol Appl Pharmacol. 2009;241:36–60. doi: 10.1016/j.taap.2009.07.032. [DOI] [PubMed] [Google Scholar]
- Cornett R, James MO, Henderson GN, Cheung J, Shroads AL, Stacpoole PW. Inhibition of glutathione S-transferase zeta and tyrosine metabolism by dichloroacetate: a potential unifying mechanism for its altered biotransformation and toxicity. Biochem Biophys Res Commun. 1999;262:752–756. doi: 10.1006/bbrc.1999.1287. [DOI] [PubMed] [Google Scholar]
- Danpure CJ. Primary hyperoxaluria. In: Scriver C, Beaudet A, Sly W, Valle D, Childs B, Kinzler K, et al., editors. Metabolic and Molecular Bases of Inherited Disease. 8th ed. New York: McGraw-Hill; 2001. pp. 3323–3367. [Google Scholar]
- Evans MV, Chiu WA, Okino MS, Cladwell JC. Development of an updated PBPK model for trichloroethylene and metabolites in mice, and its application to discern the role of oxidative metabolism in TCE-induced hepatomegaly. Toxicol Appl Pharmacol. 2009;236:329–340. doi: 10.1016/j.taap.2009.02.013. [DOI] [PubMed] [Google Scholar]
- Felitsyn N, McLeod C, Shroads AL, Stacpoole PW, Notterpek L. The heme precursor delta-aminolevulinate blocks peripheral myelin formation. J Neurochem. 2008;106:2068–2079. doi: 10.1111/j.1471-4159.2008.05552.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Guengerich FP. Metabolism of trichloroethylene and covalent binding of reaction products. Proceedings of Symposium on New Scientific Research Related to the Health Effects of Trichloroethylene; 26–27 February, 2004; Washington, DC. 2004. [[accessed 19 December 2010].]. Available: http://cfpub.epa.gov/ncea/cfm/recordisplay.cfm?deid=75934. [Google Scholar]
- Guo X, Dixit V, Lui H, Shroads AL, Henderson AL, James MO, et al. Inhibition and recovery of rat hepatic glutathione S-transferase zeta and alteration of tyrosine metabolism following dichloroacetate treatment and withdrawal. Drug Metab Dispos. 2006;34:36–42. doi: 10.1124/dmd.105.003996. [DOI] [PubMed] [Google Scholar]
- Henderson GN, Curry SH, Derendorf H, Wright EC, Stacpoole PW. Pharmacokinetics of dichloroacetate in adult patients with lactic acidosis. J Clin Pharmacol. 1997;37:416–425. doi: 10.1002/j.1552-4604.1997.tb04319.x. [DOI] [PubMed] [Google Scholar]
- IARC (International Agency for Research on Cancer) Dichloroacetic acid. IARC Monogr Eval Carcinog Risks Hum. 2004;84:359–402. [Google Scholar]
- James MO, Yan Z, Cornett R, Jayanti VM, Henderson GN, Davydova N, et al. Pharmacokinetics and metabolism of [14C]-dichloroacetate (DCA) in the male Sprague-Dawley rats: identification of glycine conjugates, including hippurate, as urinary metabolites of dichloroacetate. Drug Metab Dispos. 1998;26:1134–1143. [PubMed] [Google Scholar]
- Jia M, Coats BS, Chadha M, Frentzen B, Perez-Rodriguez J, Chadik PA, et al. Human kinetics of orally and intravenously administered low dose 1, 2-13C-dichloroacetate. J Clin Pharmacol. 2006;46:1449–1459. doi: 10.1177/0091270006292627. [DOI] [PubMed] [Google Scholar]
- Kaufmann P, Engelstad K, Wei Y, Jhung S, Sano MC, Shungu DC, et al. Dichloroacetate causes toxic neuropathy in MELAS: a randomized, controlled clinical trial. Neurology. 2006;66:324–330. doi: 10.1212/01.wnl.0000196641.05913.27. [DOI] [PubMed] [Google Scholar]
- Ketcha MM, Stevens DK, Warren DA, Bishop CT, Brashear WT. Conversion of trichloroacetic acid to dichloroacetic acid in biological samples. J Anal Toxicol. 1996;20:236–241. doi: 10.1093/jat/20.4.236. [DOI] [PubMed] [Google Scholar]
- Lantum HB, Liebler DC, Board PG, Anders MW. Alkylation and inactivation of human glutathione transferase zeta (hGSTZ1-1) by maleylacetone and fumarylacetone. Chem Res Toxicol. 2002;15:707–716. doi: 10.1021/tx025503s. [DOI] [PubMed] [Google Scholar]
- Lash LH, Fisher JW, Lipscomb JC, Parker JC. Metabolism of trichloroethylene. Environ Health Perspect. 2000;108:177–200. doi: 10.1289/ehp.00108s2177. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee JH, Chung CW, Ma YC, Wang GS, Chen PC, Hwang YH, et al. Increased mortality odds ratio of male liver cancer in a community contaminated by chlorinated hydrocarbons in groundwater. Occup Environ Med. 2003;60:364–369. doi: 10.1136/oem.60.5.364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li W, James MO, McKenzie S, Stacpoole PW. Mitochondrion is a novel site of biotransformation of dichloroacetate by glutathione transferase zeta1. J Pharmacol Exp Ther. 2011;330:87–94. doi: 10.1124/jpet.110.173195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lim CE, Matthaei KI, Blackburn AC, Davis RP, Dahlstrom JE, Koina ME, et al. Mice deficient in glutathione transferase zeta/maleylacetoacetate isomerase exhibit a range of pathological changes and elevated expression of alpha, mu, and pi class glutathione transferases. Am J Pathol. 2004;165:679–693. doi: 10.1016/S0002-9440(10)63332-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lindberg RL, Martini R, Baumgartner M, Erne B, Borg J, Zielasek J, et al. Motor neuropathy in porphobilinogen deaminase-deficient mice imitates the peripheral neuropathy of human acute porphyria. J Clin Invest. 1999;103:1127–1134. doi: 10.1172/JCI5986. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McMurtry MS, Bonnet S, Wu X, Dyck JR, Haromy A, Hashimoto K, et al. Dichloroacetate prevents and reverses pulmonary hypertension by inducing pulmonary artery smooth muscle cell apoptosis. Circ Res. 2004;95:830–840. doi: 10.1161/01.RES.0000145360.16770.9f. [DOI] [PubMed] [Google Scholar]
- Merdink JL, Gonzalez-Leon A, Bull RJ, Schultz IR. The extent of dichloroacetate formation from trichloroethylene, chloral hydrate, trichloroacetate, and trichloroethanol in B6C3F1 mice. Toxicol Sci. 1998;45:33–41. doi: 10.1006/toxs.1998.2500. [DOI] [PubMed] [Google Scholar]
- Michelakis ED, McMurtry MS, Wu XC, Dyck JR, Moudgil R, Hopkins TA, et al. Dichloroacetate, a metabolic modulator, prevents and reverses chronic hypoxic pulmonary hypertension in rats: role of increased expression and activity of voltage-gated potassium channels. Circulation. 2002;105:244–250. doi: 10.1161/hc0202.101974. [DOI] [PubMed] [Google Scholar]
- Michelakis ED, Sutendra G, Dromparis P, Webster L, Harmony A, Niven E, et al. Metabolic modulation of glioblastoma with dichloroacetate. Sci Trans Med. 2010;2:31–34. doi: 10.1126/scitranslmed.3000677. [DOI] [PubMed] [Google Scholar]
- Michelakis ED, Webster L, Mackey JR. Dichloroacetate (DCA) as a potential metabolic-targeting therapy for cancer. Br J Cancer. 2008;99:989–994. doi: 10.1038/sj.bjc.6604554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller JW, Uden PC. Characterization of nonvolatile aqueous chlorination products of humic substances. Environ Sci Technol. 1983;17:150–157. doi: 10.1021/es00109a006. [DOI] [PubMed] [Google Scholar]
- Mitchell GA, Grompe M, Lambert M, Tanguay RM. Hypertyrosinemia. In: Scriver C, Beaudet A, Sly W, Valle D, Childs B, Kinzler K, Vogelstein B, editors. Metabolic and Molecular Bases of Inherited Disease. 8th edition. II. New York: McGraw-Hill; 2001. pp. 1777–1806. [Google Scholar]
- Obolensky A, Shukairy H, Blank V. Occurrence of haloacetic acids in ICR finished water and distribution systems. In: McGuire MJ, McLain JL, Obolensky A, editors. Information Collection Rule Analysis Data. Denver, CO: AWWA Research Foundation; 2003. pp. 111–140. [Google Scholar]
- Saghir SA, Schultz IR. Low-dose pharmacokinetics and oral bioavailability of dichloroacetate in naive and GST-zeta-depleted rats. Environ Health Perspect. 2002;110:757–763. doi: 10.1289/ehp.02110757. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sassa S, Kappas A. Hereditary tyrosinemia and the heme biosynthetic pathway. Profound inhibition of delta-aminolevulinic acid dehydratase activity by succinylacetone. J Clin Invest. 1983;71:625–634. doi: 10.1172/JCI110809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schultz IR, Merdink JL, Gonzalez-Leon A, Bull RJ. Dichloroacetate toxicokinetics and disruption of tyrosine catabolism in B6C3F1 mice: dose-response relationships and age as a modifying factor. Toxicology. 2002;173:229–247. doi: 10.1016/s0300-483x(02)00034-3. [DOI] [PubMed] [Google Scholar]
- Schultz IR, Shangraw RE. Effect of short-term drinking water exposure to dichloroacetate on its pharmacokinetics and oral bioavailability in human volunteers: a stable isotope study. Toxicol Sci. 2006;92:42–50. doi: 10.1093/toxsci/kfj193. [DOI] [PubMed] [Google Scholar]
- Scriver R, Levy H, Donlon J. Hyperphenylalaninemia: phenylalanine hydroxylase deficiency. In: Scriver C, Beaudet A, Sly W, Valle D, Childs B, Kinzler K, et al., editors. Metabolic and Molecular Bases of Inherited Disease. 8th ed. New York: McGraw-Hill; 2001. pp. 1667–1724. [Google Scholar]
- Shroads AL, Guo X, Dixit V, Liu H-P, James MO, Stacpoole PW. Age-dependent metabolism of dichloroacetate in rats: possible relevance to human toxicity. J Pharmacol Exp Ther. 2008;324:1163–1171. doi: 10.1124/jpet.107.134593.. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stacpoole PW. The pharmacology of dichloroacetate. Metabolism. 1989;38:1124–1144. doi: 10.1016/0026-0495(89)90051-6. [DOI] [PubMed] [Google Scholar]
- Stacpoole PW, Henderson GN, Yan Z, Cornett R, James MO. Pharmacokinetics, metabolism and toxicology of dichloroacetate. Drug Metab Rev. 1998b;30:499–539. doi: 10.3109/03602539808996323. [DOI] [PubMed] [Google Scholar]
- Stacpoole PW, Henderson GN, Yan Z, James MO. Clinical pharmacology and toxicology of dichloroacetate. Environ Health Perspect. 1998a;106:989–994. doi: 10.1289/ehp.98106s4989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stacpoole PW, Kerr DS, Barnes C, Bunch ST, Carney PR, Fennell EM, et al. A controlled clinical trial of dichloroacetate for treatment of congenital lactic acidosis in children. Pediatrics. 2006;117:1519–1531. doi: 10.1542/peds.2005-1226. [DOI] [PubMed] [Google Scholar]
- Stacpoole PW, Kurtz TL, Han Z, Langaee T. Role of dichloroacetate in the treatment of genetic mitochondrial diseases. Adv Drug Deliv Rev. 2008;60:1478–1487. doi: 10.1016/j.addr.2008.02.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Stacpoole PW, Wright EC, Baumgartner TG, Bersin RM, Buchalter S, Curry SH, et al. A controlled clinical trial of dichloroacetate treatment of lactic acidosis in adults. The Dichloroacetate-Lactic Acidosis Study Group. N Engl J Med. 1992;327:1564–1569. doi: 10.1056/NEJM199211263272204. [DOI] [PubMed] [Google Scholar]
- Sugden MC, Holness MJ. Recent advances in mechanisms regulating glucose oxidation at the level of the pyruvate dehydrogenase complex by PDKs. Am J Physiol Endocrinol Metab. 2003;284:E855–E862. doi: 10.1152/ajpendo.00526.2002. [DOI] [PubMed] [Google Scholar]
- Templin MV, Stevens DK, Stenner RD, Bonate PL, Tuman D, Bull RJ. Factors affecting species differences in the kinetics of metabolites of trichloroethylene. J Toxicol Environ Health. 1995;44:435–447. doi: 10.1080/15287399509531972. [DOI] [PubMed] [Google Scholar]
- Tzeng HF, Blackburn AC, Anders MW. Polymorphism- and species-dependent inactivation of glutathione transferase zeta by dichloroacetate. Chem Res Toxicol. 2000;13:231–236. doi: 10.1021/tx990175q. [DOI] [PubMed] [Google Scholar]
- U.S. Geological Survey. Volatile Organic Compounds in the Nation’s Ground Water and Drinking-Water Supply Wells. Circular 1292. Reston, VA: U.S. Geological Survey; 2006. [Google Scholar]
- Vander Heiden MG, Cantley LC, Thompson CB. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science. 2009;324:1029–1033. doi: 10.1126/science.1160809. [DOI] [PMC free article] [PubMed] [Google Scholar]