U.S. flag

An official website of the United States government

NCBI Bookshelf. A service of the National Library of Medicine, National Institutes of Health.

IARC Working Group on the Evaluation of Carcinogenic Risks to Humans. Re-evaluation of Some Organic Chemicals, Hydrazine and Hydrogen Peroxide. Lyon (FR): International Agency for Research on Cancer; 1999. (IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, No. 71.)

Cover of Re-evaluation of Some Organic Chemicals, Hydrazine and Hydrogen Peroxide

Re-evaluation of Some Organic Chemicals, Hydrazine and Hydrogen Peroxide.

Show details

Tris(2,3-dibromopropyl) phosphate

Data were last reviewed in IARC (1979) and the compound was classified in IARC Monographs Supplement 7 (1987).

1. Exposure Data

1.1. Chemical and physical data

1.1.1. Nomenclature

  • Chem. Abstr. Serv. Reg. No.: 126-72-7
  • Chem. Abstr. Name: 2,3-Dibromo-1-propanol phosphate (3:1)
  • IUPAC Systematic Name: 2,3-Dibromo-1-propanol phosphate
  • Synonyms: Phosphoric acid, tris(2,3-dibromopropyl) ester; Tris

1.1.2. Structural and molecular formulae and relative molecular mass

Image 905a

1.1.3. Chemical and physical properties of the pure substance

1.2. Production and use

Production of tris(2,3-dibromopropyl) phosphate in the United States in 1975 was estimated to be between 4100 and 5400 tonnes. There are no reports of current production anywhere other than for research purposes (WHO, 1995).

Tris(2,3-dibromopropyl) phosphate has been used as a flame retardant for plastics and in synthetic textiles and fibres, which have been fabricated into children's clothing (Lewis, 1993).

1.3. Occurrence

1.3.1. Occupational exposure

Occupational exposures to tris(2,3-dibromopropyl) phosphate may have occurred during its production in the textile and polyurethane foam industries (IARC, 1979).

1.3.2. Environmental occurrence

Environmental release in the past has been shown to result from textile finishing plants and laundering of the finished product (United States National Library of Medicine, 1997).

Tris(2,3-dibromopropyl) phosphate was found in the air and soil in the United States in the 1970s. None was found in samples taken from various water and soil sources in Japan at this time. General population exposures may have occurred from the use of clothing treated with the compound (WHO, 1995).

1.4. Regulations and guidelines

The American Conference of Governmental Industrial Hygienists (ACGIH) (1997) has not proposed any occupational exposure limit for tris(2,3-dibromopropyl) phosphate. Finland, Sweden and France have a carcinogen notation (United States National Library of Medicine, 1997).

No international guideline for tris(2,3-dibromopropyl) phosphate in drinking-water has been established (WHO, 1993).

2. Studies of Cancer in Humans

In a cohort mortality study in the United States, a group of 628 male workers was classified as exposed to tris(2,3-dibromopropyl) phosphate either on a ‘routine’ or ‘nonroutine’ basis; 36 deaths occurred in this group (35 expected), seven of which were due to cancer compared with 6.6 that would have been expected (Wong et al., 1984).

3. Studies of Cancer in Experimental Animals

Tris(2,3-dibromopropyl) phosphate was tested for carcinogenicity in one experiment in mice and in one in rats by oral administration and in one experiment in female mice by skin application. In mice, following oral administration, it produced benign and malignant tumours of the forestomach and lung in animals of both sexes, benign and malignant liver tumours in females and benign and malignant tumours of the kidney (of the tubule cells) in males. In rats, it produced benign and malignant tumours of the kidney (of the tubule cells) in males and benign kidney tumours (of the tubule cells) in females. After skin application to female mice, it produced tumours of the skin, lung, forestomach and oral cavity (IARC, 1979).

3.1. Oral administration

Rat: A group of 50 male Fischer 344 rats, four weeks of age, was administered 100 mg/kg bw tris(2,3-dibromopropyl) phosphate [purity unspecified] dissolved in vegetable oil by gavage on five days per week for four weeks. After four weeks, the group was divided into three subgroups of 20, 15 and 15 rats. The first group received no further exposure; in the second subgroup, tris(2,3-dibromopropyl) phosphate administration was continued for 48 weeks; the third subgroup received vegetable oil (vehicle) alone for the remainder of the experiment. Two control groups consisted of 27 rats treated with vegetable oil (vehicle) alone and seven rats which received no treatment. The study was terminated at 52 weeks. Rats were killed at various time intervals to study the reversibility of tris(2,3-dibromopropyl) phosphate-induced lesions. In the rats treated for 52 weeks with tris(2,3-dibromopropyl) phosphate, one developed a kidney adenocarcinoma and 3/5 rats surviving at 52 weeks had adenomas of the descending colon (Reznik et al., 1981). [The Working Group noted the short duration of the experiment.]

3.2. Carcinogenicity of metabolites

3.2.1. 2,3-Dibromo-1-propanol

Mouse: Groups of 50 male and 50 female B6C3F1 mice, eight weeks of age, were administered skin applications of 0, 88 or 177 mg/kg bw 2,3-dibromo-1-propanol (98% pure) in 95% ethanol on five days per week for 36–39 weeks (males) or 39–42 weeks (females). The study was terminated at 36–39 weeks (males) and 39–42 weeks (females) because sera from sentinel mice housed in the same room as the study animals were found to be positive for antibodies to lymphocytic choriomeningitis virus. As shown in Table 1, increased incidences of skin papillomas, forestomach papillomas and fore-stomach carcinomas were observed in both sexes. Hepatocellular adenomas were seen in 1/50 control, 2/50 low-dose and 9/50 high-dose (p < 0.05) male mice; no data for liver were reported in females (Eustis et al., 1995).

Table 1. Increased tumour incidences in mice administered 2,3-dibromo-1-propanol by skin application.

Table 1

Increased tumour incidences in mice administered 2,3-dibromo-1-propanol by skin application.

Rat: Groups of 50 male and 50 female Fischer 344/N rats, eight weeks of age, were administered skin applications of 0, 188 or 375 mg/kg bw 2,3-dibromo-1-propanol (98% pure) in 95% ethanol on five days per week for 48–51 weeks (males) or 52–55 weeks (females). The study was terminated at 48–51 weeks for males and 52–55 weeks for females because of reduced survival of the high-dose groups and because sentinel mice housed in the same room as the rats tested positive for lymphocytic choriomeningitis virus. As shown in Table 2, there were increased incidences of skin neoplasms (all types), squamous-cell carcinomas of the skin, basal-cell tumours [not further specified] of the skin, squamous-cell carcinomas of the oral mucosa, squamous-cell papillomas of the oesophagus, squamous-cell papillomas of the forestomach, adenocarcinomas of the small intestine, adenomatous polyps of the large intestine, adenomas of the nasal mucosa, Zymbal gland adenocarcinomas and liver carcinomas (Eustis et al., 1995).

Table 2. Increased tumour incidences in rats administered 2,3-dibromo-1- propanol by skin application.

Table 2

Increased tumour incidences in rats administered 2,3-dibromo-1- propanol by skin application.

3.2.2. Bis(2,3-dibromopropyl) phosphate

Rat: Groups of 40 male and 40 female Wistar rats, five weeks of age, were administered the magnesium salt of bis(2,3-dibromopropyl) phosphate [purity not specified] mixed in the diet at concentrations of 0 (control), 80 (low-dose), 400 (mid-dose) or 2000 (high-dose) mg/kg diet (ppm) for 24 months. Oesophageal papillomas were observed in 0/40 control, 0/40 low-dose, 6/40 mid-dose (p < 0.05) and 2/40 high-dose males and in 0/40 control, 0/40 low-dose, 0/40 mid-dose and 6/40 high-dose (p < 0.05) females. Papillomas of the forestomach were seen in 0/40 control, 0/40 low-dose, 8/40 mid-dose (p < 0.05) and 17/40 high-dose (p < 0.01) males and in 0/40 control, 0/40 low-dose, 4/40 mid-dose and 20/40 high-dose (p < 0.01) females. Adenocarcinomas of the small intestine were observed in 0/40 control, 0/40 low-dose, 2/40 mid-dose and 14/40 high-dose (p < 0.01) males and in 0/40 control, 0/40 low-dose, 0/40 mid-dose and 9/40 high-dose (p < 0.01) females. Hepatocellular carcinomas were observed in 0/40 control, 1/40 low-dose, 7/40 mid-dose (p < 0.05) and 24/40 high-dose (p < 0.01) female rats (Takada et al., 1991).

4. Other Data Relevant to an Evaluation of Carcinogenicity and its Mechanisms

4.1. Absorption, distribution, metabolism and excretion

4.1.1. Humans

No data were available to the Working Group.

4.1.2. Experimental systems

The excretion balance and tissue distribution of radiolabelled tris(2,3-dibromopropyl) phosphate in rats were examined by Lynn et al. (1980, 1982) and Nomeir and Matthews (1983). After intravenous administration of 1.76 mg/rat, Lynn et al. (1980) recovered 57% of the dose in the urine in five days and identified the diester bis(2,3-dibromopropyl) phosphate as a minor urinary metabolite (7.8% of urinary 14C). In further work, Lynn et al. (1982) recovered a total of 86% of the dose in the excreta (58% urine, 9% faeces, 19% as expired 14CO2) with a further 9% in the carcass. Bile-duct-cannulated rats excreted 34% of the dose in the bile in 24 h, 20% being eliminated in the first hour after dosing. No unchanged tris(2,3-dibromopropyl) phosphate was detected in the urine, but dibromopropanol was present in addition to the diester previously reported. On high-performance liquid chromatography, numerous 14C peaks remained unidentified.

Nomeir and Matthews (1983) compared the disposition of tris(2,3-dibromo[1-14C]propyl) phosphate given intravenously and orally to rats at a dose of 1.4 mg/kg bw (2 µmol/kg bw). Absorption from the gastrointestinal tract was extensive and rapid, the tissue levels of 14C being essentially identical after both routes of administration. After 24 h, 24% of an oral dose and 17% of an intravenous dose were present in urine and 11% (oral dose), 7% (intravenous dose) in faeces, with 21% (oral dose) and 26% (intravenous dose) present in seven tissues examined. A further 20% was recovered as exhaled 14CO2 after intravenous dosing; no data were reported on exhalation after oral dosing. The tissue distribution of 14C was widespread, with an elimination half-life of 60 h from most tissues and 91 h from liver and kidney.

Lynn et al. (1982) detected three major 14C-containing compounds in plasma and the pattern was dominated by bis(2,3-dibromopropyl) phosphate as early as 5 min after intravenous dosing. 2,3-Dibromopropanol was also detected up to 8 h after dosing. The elimination of bis(2,3-dibromopropyl) phosphate was biphasic, with half-lives of 6 and 36 h and it was detected up to five days after dosing.

Tissue distribution was examined at five time points, with separate determinations of total 14C, tris(2,3-dibromopropyl) phosphate and bis(2,3,-dibromopropyl) phosphate. The results confirmed the rapid disappearance of tris(2,3-dibromopropyl) phosphate, this being detected only at 5 and 30 min. Bis(2,3-dibromopropyl) phosphate was the major component in blood, lung, muscle and fat and had a long elimination period. At five days after dosing, there was significant retention of 14C in the kidney, this comprising various polar components with some bis(2,3-dibromopropyl) phosphate also detected. The extensive biliary excretion of tris(2,3-dibromopropyl) phosphate-related radioactivity and low faecal elimination of the radiolabel indicate that enterohepatic circulation contributes to the retention of 14C in the body (Lynn et al., 1982).

In addition to the previously reported bis(2,3-dibromopropyl) phosphate and 2,3-dibromopropanol, Nomeir and Matthews (1983) characterized four additional metabolites by mass spectrometry, that arose from further hydrolysis and dehydrobromination of the 2,3-dibromopropane moiety, namely 2-bromo-2-propenyl-2,3-dibromopropyl phosphate, bis(2-bromo-2-propenyl) phosphate, 2,3-dibromopropyl phosphate and 2-bromo-2-propenyl phosphate. All six metabolites were found in 24-h urine and 3-h bile, with bis(2-bromo-2-propenyl) phosphate and 2-bromo-2-propenyl phosphate predominating in urine, while bis(2,3-dibromopropyl) phosphate and 2-bromo-2-propenyl-2,3-dibromopropyl phosphate were the major metabolites identified in bile; 67% of urinary and 47% of biliary 14C were accounted for by a variety of unidentified metabolites.

In vitro, liver microsomes from rat, mouse, hamster and guinea-pig all activate tris(2,3-dibromopropyl) phosphate resulting in covalent binding to protein. This binding was cytochrome P450-dependent and was inhibited by glutathione. In vivo in rats, the kidney was the principal target organ for covalent binding to protein and, at high doses, to DNA. This binding was enhanced by pretreatment with polychlorinated biphenyls but not sodium phenobarbital and was partly prevented by cobalt chloride (CoCl2) pretreatment. There was much less binding to liver protein and DNA and minimal binding to muscle (Søderlund et al., 1981, 1982a).

Nelson et al. (1984) and Søderlund et al. (1984) identified the proximate mutagenic metabolite of tris(2,3-dibromopropyl) phosphate as 2-bromoacrolein and used stable isotope techniques in microsomal incubations to show that it is formed by cytochrome P450-dependent oxidative debromination at C-3 of one of the 2,3-dibromopropyl groups followed by β-elimination to break the phosphoester bond. Søderlund et al. (1984) also showed the evolution of bromide ion release in microsomes in a glutathione-dependent reaction.

These studies have been extended by Pearson et al. (1993a,b), who showed that a number of metabolites contribute to protein binding in addition to 2-bromoacrolein. The major metabolic pathway leading to protein binding is C-2 oxidation of the 2,3-dibromopropyl groups, giving a reactive α-bromoketone which might either alkylate proteins directly or be hydrolysed to bis(2,3,-dibromopropyl) phosphate and an α-bromo-α′-hydroxyketone which could mediate the alkylation of protein.

4.2. Toxic effects

4.2.1. Humans

In a cohort of 3579 white male chemical workers with potential exposures to brominated compounds including tris(2,3-dibromopropyl) phosphate, no significant overall or cause-specific mortality excess was detected (Wong et al., 1984).

4.2.2. Experimental systems

Tris(2,3-dibromopropyl) phosphate caused extensive acute renal tubule necrosis at doses of 175 mg/kg bw and higher in male rats. Treatment also resulted in hepatotoxicity, but this effect was less pronounced and occurred at higher doses (Søderlund et al., 1980). Administration of radioactively labelled tris(2,3-dibromopropyl) phosphate to rats as a single intraperitoneal dose of 250 mg/kg bw led to pronounced binding of radioactivity to kidney but not to liver protein 9 h later (Dybing et al., 1980). Morales and Matthews (1980) and Lynn et al. (1982) also showed that the kidney accumulated the highest rate of radioactivity after injection of [14C]tris(2,3-dibromopropyl) phosphate, compared with other organs. Dybing and Søderlund (1980) treated rats intraperitoneally with 250 mg/kg bw unlabelled tris(2,3-dibromopropyl) phosphate and determined parameters of kidney and liver toxicity 24 h later. The treatment resulted in increased plasma urea and creatinine levels. Kluwe et al. (1981) reported that tris(2,3-dibromopropyl) phosphate treatment of rodents resulted in decreased non-protein sulfhydryl content in the liver, but not in the kidney as the major target organ.

In young male Fischer 344 rats treated with 100 mg/kg bw tris(2,3-dibromopropyl) phosphate by gavage, severe tubular nephrosis was observed, starting from the corticomedullar junction and spreading to the peripheral cortex (Reznik et al., 1981). A single intraperitoneal dose of 154 mg/kg bw tris(2,3-dibromopropyl) phosphate given to male Sprague-Dawley rats caused cortical damage, significant increases in serum creatinine level and a depression of para-aminohippurate uptake in cortical slices (Elliott et al., 1982).

Cunningham et al. (1993, 1994) fed male Fischer 344 rats a diet containing 0, 50 or 100 ppm (mg/kg) tris(2,3-dibromopropyl) phosphate for 14 days. In the kidney, the treatment induced significant cell proliferation that was localized in the renal outer medulla region. The proliferation rates of the inner medulla, the cortex and the liver were not increased.

In mice, hamsters and guinea-pigs, no clear evidence of renal damage was found at doses of 500–1000 mg/kg bw tris(2,3-dibromopropyl) phosphate (Søderlund et al., 1982a), which were clearly nephrotoxic to rats. Analysis of protein binding of radiolabelled tris(2,3-dibromopropyl) phosphate showed that binding to kidney protein also was much higher in rats than in the other species investigated.

4.3. Reproductive and developmental effects

4.3.1. Humans

No data were available to the Working Group.

4.3.2. Experimental systems

In a study by Seabaugh et al. (1981), pregnant Sprague-Dawley rats received 0, 5, 25 or 125 mg/kg bw tris(2,3-dibromopropyl) phosphate by gavage on days 6–15 of gestation. Weight gain during gestation was significantly decreased in the animals treated with 125 mg/kg bw per day, but no other compound-related toxic or teratogenic effect was observed.

4.4. Genetic and related effects

The genotoxicity of tris(2,3-dibromopropyl)phosphate has been reviewed (van Beerendonk et al., 1994a).

4.4.1. Humans

No data were available to the Working Group.

4.4.2. Experimental systems (see Table 3 for references)

Table 3. Genetic and related effects of tris(2,3-dibromopropyl) phosphate.

Table 3

Genetic and related effects of tris(2,3-dibromopropyl) phosphate.

Tris(2,3-dibromopropyl) phosphate is mutagenic in Salmonella typhimurium in the presence of a metabolic activation system and in V79 Chinese hamster lung cells. With or without metabolic activation, it produces sister chromatid exchanges in the latter system and morphological transformation in C3H 10T½ and Syrian hamster embryo cells. It binds covalently to proteins and DNA, and causes DNA single strand breaks in mammalian cells in vitro and in vivo. It is mutagenic (in somatic and germ cells), clastogenic and recombinogenic in Drosophila melanogaster and induces bone-marrow micronuclei in mice and hamsters, liver micronuclei in rats and gene mutations in mouse kidney in vivo.

4.4.3. Mechanistic aspects

Mechanistic and metabolic studies have suggested that the genotoxicity of tris(2,3-dibromopropyl) phosphate may be mediated by its conversion to reactive metabolites, the most important of which may be 2-bromoacrolein (Nelson et al., 1984; Søderlund et al., 1984). 2-Bromoacrolein and 2,3-dibromopropanal are mutagenic in Salmonella typhimurium TA100, with or without metabolic activation, cause single-strand breaks in DNA of a rat hepatoma cell line and morphological transformation of Syrian hamster embryo cells (Gordon et al., 1985). Furthermore, 2-bromoacrolein forms adducts with DNA which block DNA replication in vitro. It also induces DNA–protein cross-links in Drosophila melanogaster (van Beerendonk, 1992, 1994c).

An equimolar dose of the metabolite bis(2,3-dibromopropyl) phosphate was markedly more nephrotoxic and led also to damage of the descending loop of Henle. Intraperitoneal injection of bis(2,3-dibromopropyl) phosphate to Sprague-Dawley rats resulted in necrosis of the renal cortex, which was less severe in female than in male rats (Elliott et al., 1983). Renal dysfunction, as indexed by serum creatinine level and in-vitro renal cortical uptake of para-aminohippurate and N-methylnicotinamide, was similar in males and females. Evidence for the role of bis(2,3-dibromopropyl) phosphate and mono(2,3-dibromopropyl) phosphate as nephrotoxic metabolites of tris(2,3-dibromopropyl) phosphate was provided by Lynn et al. (1982), Søderlund et al. (1982b) and Fukuoka et al. (1988). In isolated proximal tubule cells from rat kidney, 100 µM bis(2,3-dibromopropyl) phosphate inhibited the uptake of α-methylglucose, a parameter that the authors used to assess cytotoxicity (Boogard et al., 1989).

5. Summary of Data Reported and Evaluation

5.1. Exposure data

During the 1970s, tris(2,3-dibromopropyl) phosphate was produced in low volumes, with occupational exposure likely to have occurred in its production and use in the textile industry. It does not appear to have been produced since then. The primary exposure to the general population appears to have been through wearing clothing treated with the chemical.

5.2. Human carcinogenicity data

A small cohort study of workers exposed to tris(2,3-dibromopropyl) phosphate was uninformative.

5.3. Animal carcinogenicity data

Tris(2,3-dibromopropyl) phosphate was tested for carcinogenicity in mice and rats by oral administration. In mice, it produced benign and malignant tumours of the fore-stomach and lung in animals of each sex, benign and malignant liver tumours in females and benign and malignant tumours of the kidney in males. In rats, it produced benign and malignant tumours of the kidney in males and benign kidney tumours in females. In a study of limited duration in male rats, benign tumours of the colon were reported. After skin application to female mice, it produced tumours of the skin, lung, forestomach and oral cavity.

A metabolite of tris(2,3-dibromopropyl) phosphate, bis(2,3-dibromopropyl) phosphate, was tested for carcinogenicity in rats by oral administration and another metabolite, 2,3-dibromo-1-propanol, was tested in mice and rats by skin application. They produced a variety of tumours, including skin, forestomach and hepatocellular tumours, in mice and rats and tumours of the oesophagus, intestine, nasal mucosa and Zymbal glands in rats.

5.4. Other relevant data

Tris(2,3-dibromopropyl) phosphate and its metabolites bis(2,3-dibromopropyl)phosphate and mono(2,3-dibromopropyl) phosphate are nephrotoxic in rodents.

Tris(2,3-dibromopropyl) phosphate is mutagenic in bacteria and causes genetic damage in cultured mammalian cells, Drosophila melanogaster and mice, probably via metabolism to a number of intermediates of which 2-bromoacrolein may be particularly important.

5.5. Evaluation

There is inadequate evidence in humans for the carcinogenicity of tris(2,3-dibromopropyl) phosphate.

There is sufficient evidence in experimental animals for the carcinogenicity of tris(2,3-dibromopropyl) phosphate.

Overall evaluation

Tris(2,3-dibromopropyl)phosphate is probably carcinogenic to humans (Group 2A).

In making the overall evaluation, the Working Group took into consideration that tris(2,3-dibromopropyl) phosphate is consistently active in a wide range of mammalian in-vivo and in-vitro test systems.

6. References

  • American Conference of Governmental Industrial Hygienists (1997) 1997 Threshold Limit Values for Chemical Substances and Physical Agents and Biological Exposure Indices, Cincinnati, OH.
  • de Boer J.G., Mirsalis J.C., Provost G.S., Tindall K.R., Glickman B.W. Spectrum of mutations in kidney, stomach and liver from lacI/transgenic mice recovered after treatment with tris(2,3-dibromopropyl)phosphate. Environ. mol. Mutag. 1996;28:418–423. [PubMed: 8991072]
  • Boogaard P.J., Mulder G.J., Nagelkerke J.F. Isolated proximal tubular cells from rat kidney as an in vitro model for studies on nephrotoxicity. II. α-Methylglucose uptake as a sensitive parameter for mechanistic studies of acute toxicity by xenobiotics. Toxicol. appl. Pharmacol. 1989;101:144–157. [PubMed: 2799813]
  • Budavari, S., ed. (1996) The Merck Index, 12th Ed., Whitehouse Station, NJ, Merck & Co., p. 1661.
  • Cunningham M.L., Elwell M.R., Matthews H.B. Site-specific cell proliferation in renal tubular cells by the renal tubular carcinogen tris(2,3-dibromopropyl)phosphate. Environ. Health Perspect. 1993;101 Suppl. 5:253–258. [PMC free article: PMC1519429] [PubMed: 8013416]
  • Cunningham M.L., Elwell M.R., Matthews H.B. Relationship of carcinogenicity and cellular proliferation induced by mutagenic noncarcinogens vs carcinogens. III. Organophosphate pesticides vs tris(2,3-dibromopropyl)phosphate. Fundam. appl. Toxicol. 1994;23:363–369. [PubMed: 7835536]
  • Dybing E., Soderlund E. Nephrotoxicity of the flame retardant tris(2,3-dibromopropyl)phosphate. Arch. Toxicol. 1980;Suppl. 4:219–222. [PubMed: 6933906]
  • Dybing E., Søderlund E.J., Nelson S.D. Irreversible macromolecular binding of the flame retardant tris(2,3-dibromopropyl)phosphate in vitro and in vivo. Develop. Toxicol. environ. Sci. 1980;8:265–268. [PubMed: 7308025]
  • Elliott W.C., Lynn R.K., Houghton D.C., Kennish J.M., Bennett W.M. Nephrotoxicity of the flame retardant, tris(2,3-dibromopropyl)phosphate, and its metabolites. Toxicol. appl. Pharmacol. 1982;62:179–182. [PubMed: 7064152]
  • Elliott W.C., Koski J., Houghton D.C., Hunter-Baines J., Bennett W.M., Lynn R.K. Bis(2,3-dibromopropyl)phosphate nephrotoxicity: effect of sex and CoCl2 pretreatment. Life Sci. 1983;32:1107–1117. [PubMed: 6827891]
  • Eustis S.L., Haseman J.K., Mackenzie W.F., Abdo K.M. Toxicity and carcinogenicity of 2,3-dibromo-1-propanol in F344/N rats and B6C3F1 mice. Fundam. appl. Toxicol. 1995;26:41–50. [PubMed: 7657061]
  • Fukuoka M., Takahashi T., Naito K., Takada K. Comparative studies on nephrotoxic effects of tris(2,3-dibromopropyl) phosphate and bis(2,3-dibromopropyl) phosphate on rat urinary metabolites. J. appl. Toxicol. 1988;8:43–52. [PubMed: 3356864]
  • Gordon W.P., Søderlund E.J., Holme J.A., Nelson S.D., Iyer L., Rivedal E., Dybing E. The genotoxicity of 2-bromoacrolein and 2,3-dibromopropanal. Carcinogenesis. 1985;6:705–709. [PubMed: 4006056]
  • Holme J.A., Søderlund E.J., Hongslo J.K., Nelson S.D., Dybing E. Comparative genotoxicity studies of the flame retardant tris(2,3-dibromopropyl)phosphate and possible metabolites. Mutat. Res. 1983;124:213–224. [PubMed: 6686285]
  • IARC (1979) IARC Monographs on the Evaluation of the Carcinogenic Risk of Chemicals to Humans, Volume 20, Some Halogenated Hydrocarbons, Lyon, 575–588. [PubMed: 397179]
  • IARC (1987) IARC Monographs on the Evaluation of Carcinogenic Risks to Humans, Suppl. 7, Overall Evaluations of Carcinogenicity: An Updating of IARC Monographs Volumes 1 to 42, Lyon, pp. 369–370. [PubMed: 3482203]
  • Kluwe W.M., McNish R., Smithson K., Hook J.B. Depletion by 1,2-dibromoethane, 1,2-dibromo-3-chloropropane, tris(2,3-dibromopropyl)phosphate, and hexachloro-1,3-butadiene of reduced non-protein sulfhydryl groups in target and non-target organs. Biochem. Pharmacol. 1981;30:2265–2271. [PubMed: 6794573]
  • Lewis, R.J., Jr. (1993) Hawley's Condensed Chemical Dictionary, 12th Ed., New York, Van Nostrand Reinhold, p. 1189.
  • Lynn R.K., Wong K., Dickinson R.G., Gerber N., Kennish J.M. Diester metabolites of the flame retardant chemicals, tris(1,3-dichloro-2-propyl) phosphate and tris(2,3-dibromopropyl)phosphate in the rat: identification and quantification. Res. Commun. chem. Path. Pharmacol. 1980;28:351–360. [PubMed: 7394328]
  • Lynn R.K., Garvie-Gould C., Wong K., Kennish J.M. Metabolism, distribution and excretion of the flame retardant tris(2,3-dibromopropyl) phosphate (Tris-BP) in the rat: identification of mutagenic and nephrotoxic metabolites. Toxicol. appl. Pharmacol. 1982;63:105–119. [PubMed: 7071866]
  • McCoy E., Hyman J., Rosenkranz H.S. A new mutagenic and genotoxic response of the flame retardant tris(2,3-dibromopropyl)phosphate. Mutat. Res. 1980;77:209–214. [PubMed: 6991918]
  • Morales N.M., Matthews H.B. In vivo binding of the flame retardants tris(2,3-dibromopropyl) phosphate and tris(1,3-dichloro-2-propyl) phosphate to macromolecules of mouse liver, kidney and muscle. Bull. environ. Contam. Toxicol. 1980;25:34–38. [PubMed: 6161654]
  • Nelson S.D., Omichinski J.G., Lyer L., Gordon W.P., Soderlund E.J., Dybing E. Activation mechanism of tris(2,3-dibromopropyl) phosphate to the potent mutagen, 2-bromoacrolein. Biochem. Biophys. Res. Commun. 1984;121:213–219. [PubMed: 6375666]
  • Nomeir A.A., Matthews H.B. Metabolism and disposition of the flame retardant tris(2,3-dibromopropyl) phosphate in the rat. Toxicol. appl. Pharmacol. 1983;67:357–369. [PubMed: 6845365]
  • Pearson P.G., Omichinski J.G., McClanahan R.H., Søderlund E.J., Dybing E., Nelson S.D. Metabolic activation of tris(2,3-dibromopropyl) phosphate to reactive intermediates. I. Covalent binding and reactive metabolite formation in vitro. Toxicol. appl. Pharmacol. 1993a;118:186–195. [PubMed: 8441997]
  • Pearson P.G., Omichinski J.G., Holme J.A., McClanahan R.H., Brunborg G., Søderlund E.J., Dybing E., Nelson S.D. Metabolic activation of tris(2,3-dibromopropyl) phosphate to reactive intermediates. II. Covalent binding, reactive metabolite formation and differential metabolite-specific DNA damage in vivo. Toxicol. appl. Pharmacol. 1993b;118:196–204. [PubMed: 8441998]
  • Provost G.S., Mirsalis J.C., Rogers B.J., Short J.M. Mutagenic response to benzene and tris(2,3-dibromopropyl)phosphate in the lambda lacI transgenic mouse mutation assay: a standardized approach to in vivo mutation analysis. Environ. mol. Mutag. 1996;28:342–347. [PubMed: 8991062]
  • Reznik G., Reznik-Schüller H.M., Rice J.M., Hague B.F. Pathogenesis of toxic and neoplastic renal lesions induced by the flame retardant tris(2,3-dibromopropyl)phosphate in F344 rats, and development of colonic adenomas after prolonged oral administration. Lab. Invest. 1981;44:74–83. [PubMed: 7453132]
  • Sala M., Gu Z.G., Moens G., Chouroulinkov I. In vivo and in vitro biological effects of the flame retardants tris(2,3-dibromopropyl)phosphate and tris(2-chlorethyl)orthophosphate. Eur. J. Cancer clin. Oncol. 1982;18:1337–1344. [PubMed: 6891932]
  • Salamone M.F., Katz M. Mutagenicity of tris(2,3-dibromopropyl)phosphate in mammalian gonad and bone marrow tissue. J. natl Cancer Inst. 1981;66:691–695. [PubMed: 6939915]
  • Schechtman L.M., Kiss E., McCarvill J., Nims R., Kouri R.E., Lubet R.A. A method for the amplification of chemically induced transformation in C3H/10T1/2 clone 8 cells: its use as a potential screening assay. J. natl Cancer Inst. 1987;79:487–497. [PubMed: 3114533]
  • Seabaugh V.M., Collins T.F.X., Hoheisel C.A., Bierbower G.W., McLaughlin J. Rat teratology study of orally administered tris-(2,3-dibromopropyl) phosphate. Food Cosmet. Toxicol. 1981;19:67–72. [PubMed: 7262733]
  • Shimada T., Iwasaki M., Martin M.V., Guengerich F.P. Human liver microsomal cytochrome P-450 enzymes involved in the bioactivation of procarcinogens detected by umu gene response in Salmonella typhimurium TA 1535/pSK1002. Cancer Res. 1989;49:3218–3228. [PubMed: 2655891]
  • Søderlund E.J., Nelson S.D., Dybing E. Mutagenic activation of tris(2,3-dibromopropyl)phosphate: the role of microsomal oxidative metabolism. Acta pharmacol. toxicol. 1979;45:112–121. [PubMed: 40390]
  • Søderlund E., Dybing E., Nelson S.D. Nephrotoxicity and hepatotoxicity of tris(2,3-dibromopropyl)phosphate in the rat. Toxicol. appl. Pharmacol. 1980;56:171–181. [PubMed: 7466817]
  • Søderlund E.J., Nelson S.D., Dybing E. In vitro and in vivo covalent binding of the kidney toxicant and carcinogen tris(2,3-dibromopropyl)phosphate. Toxicology. 1981;21:291–304. [PubMed: 7302950]
  • Søderlund E.J., Nelson S.D., von Bahr C., Dybing E. Species differences in kidney toxicity and metabolic activation of tris(2,3-dibromopropyl)phosphate. Fundam. appl. Toxicol. 1982a;2:187–194. [PubMed: 7185616]
  • Søderlund E., Nelson S.D., Dybing E. Mutagenicity and nephrotoxicity of two tris(2,3-dibromopropyl)phosphate metabolites: bis(2,3-dibromopropyl)phosphate and 2,3-dibromopropylphosphate. Acta pharmacol. toxicol. 1982b;51:76–80. [PubMed: 7124413]
  • Søderlund E.J., Gordon W.P., Nelson S.D., Omichinski J.G., Dybing E. Metabolism in vitro of tris(2,3-dibromopropyl) phosphate: Oxidative debromination and bis(2,3-dibromopropyl) phosphate formation as correlates of mutagenicity and covalent protein binding. Biochem. Pharmacol. 1984;33:4017–4023. [PubMed: 6508849]
  • Søderlund E.J., Dybing E., Holme J.A., Hongslo J.K., Rivedal E., Sanner T., Nelson S.D. Comparative genotoxicity and nephrotoxicity studies of the two halogenated flame retardants tris(1,3-dichloro-2-propyl)phosphate and tris(2,3-dibromopropyl)phosphate. Acta pharmacol. toxicol. 1985;56:20–29. [PubMed: 3883695]
  • Søderlund E., Brunborg G., Dybing E., Trygg B., Nelson S.D., Holme J.A. Organ-specific DNA damage of tris(2,3-dibromopropyl)phosphate and its diester metabolite in the rat. Chem.-biol. Interact. 1992;82:195–207. [PubMed: 1568270]
  • Takada K., Naito K., Kobayashi K., Tobe M., Kurokawa Y., Fukuoka M. Carcinogenic effects of bis(2,3-dibromopropyl)phosphate in Wistar rats. J. appl. Toxicol. 1991;11:323–331. [PubMed: 1783736]
  • United States National Library of Medicine (1997) Hazardous Substances Data Bank (HSDB), Bethesda, MD [Record No. 2581]
  • van Beerendonk G.J.M., Nivard M.J.M., Vogel E.W., Nelson S.D., Meerman J.H.N. Formation of thymidine adducts and cross-linking potential of 2-bromoacrolein, a reactive metabolite of tris(2,3-dibromopropyl)phosphate. Mutagenesis. 1992;7:19–24. [PubMed: 1635451]
  • van Beerendonk G.J.M., Nelson S.D., Meerman J.H.N. Metabolism and genotoxicity of the halogenated alkyl compound tris(2,3-dibromopropyl)phosphate. Hum. exp. Toxicol. 1994a;13:861–865. [PubMed: 7718306]
  • van Beerendonk G.J.M., Nivard M.J.M., Vogel E.W., Nelson S.D., Meerman J.H.N. Genotoxicity of the flame retardant tris(2,3-dibromopropyl)phosphate in the rat and Drosophila: effects of deuterium substitution. Carcinogenesis. 1994b;15:1197–1202. [PubMed: 8020156]
  • van Beerendonk G.J.M., van Gog F.B., Vrieling H., Pearson P.G., Nelson S.D., Meerman J.H.N. Blocking of in vitro DNA replication by deoxycytidine adducts of the mutagen and clastogen 2-bromoacrolein. Cancer Res. 1994c;54:679–685. [PubMed: 8306329]
  • Verschueren, K. (1996) Handbook of Environmental Data on Organic Chemicals, 3rd Ed., New York, Van Nostrand Reinhold, pp. 1867.
  • Vogel E.W., Nivard M.J.M. Performance of 181 chemicals in a Drosophila assay predominantly monitoring interchromosomal mitotic recombination. Mutagenesis. 1993;8:57–81. [PubMed: 8450769]
  • WHO (1993) Guidelines for Drinking Water Quality, 2nd Ed., Vol. 1, Recommendations, Geneva.
  • WHO (1995) Tris(2,3-dibromopropyl)phosphate and Bis(2,3-dibromopropyl)phosphate (Environmental Health Criteria 173), Geneva, International Programme on Chemical Safety.
  • Wong O., Brocker W., Davis H.V., Nagle G.S. Mortality of workers potentially exposed to organic and inorganic brominated chemicals, DBCP, Tris, PBB, and DDT. Br. J. ind. Med. 1984;41:15–24. [PMC free article: PMC1009230] [PubMed: 6318800]
  • Zeiger E., Pagano D.A., Nomeir A.A. Structure-activity studies on the mutagenicity of tris(2,3-dibromopropyl)phosphate (Tris-BP) and its metabolites in Salmonella. Environ. Mutag. 1982;4:271–277. [PubMed: 7094911]
©International Agency for Research on Cancer, 1999.
Bookshelf ID: NBK499429

Views

  • PubReader
  • Print View
  • Cite this Page
  • PDF version of this title (4.1M)

Related information

  • PMC
    PubMed Central citations
  • PubMed
    Links to PubMed

Recent Activity

Your browsing activity is empty.

Activity recording is turned off.

Turn recording back on

See more...