Skip to main content

Advertisement

Log in

Risk assessment for humans using physiologically based pharmacokinetic model of diethyl phthalate and its major metabolite, monoethyl phthalate

  • Toxicokinetics and Metabolism
  • Published:
Archives of Toxicology Aims and scope Submit manuscript

Abstract

Diethyl phthalate (DEP) belongs to phthalates with short alkyl chains. It is a substance frequently used to make various products. Thus, humans are widely exposed to DEP from the surrounding environment such as food, soil, air, and water. As previously reported in many studies, DEP is an endocrine disruptor with reproductive toxicity. Monoethyl phthalate (MEP), a major metabolite of DEP in vivo, is a biomarker for DEP exposure assessment. It is also an endocrine disruptor with reproductive toxicity, similar to DEP. However, toxicokinetic studies on both MEP and DEP have not been reported in detail yet. Therefore, the objective of this study was to evaluate and develop physiologically based pharmacokinetic (PBPK) model for both DEP and MEP in rats and extend this to human risk assessment based on human exposure. This study was conducted in vivo after intravenous or oral administration of DEP into female (2 mg/kg dose) and male (0.1–10 mg/kg dose) rats. Biological samples consisted of urine, plasma, and 11 different tissues. These samples were analyzed using UPLC–ESI–MS/MS method. For DEP, the tissue to plasma partition coefficient was the highest in the kidney, followed by that in the liver. For MEP, the tissue to plasma partition coefficient was the highest in the liver. It was less than unity in all other tissues. Plasma, urine, and fecal samples were also obtained after IV administration of MEP (10 mg/kg dose) to male rats. All results were reflected in a model developed in this study, including in vivo conversion from DEP to MEP. Predicted concentrations of DEP and MEP in rat urine, plasma, and tissue samples using the developed PBPK model fitted well with observed values. We then extrapolated the PBPK model in rats to a human PBPK model of DEP and MEP based on human physiological parameters. Reference dose of 0.63 mg/kg/day (or 0.18 mg/kg/day) for DEP and external doses of 0.246 μg/kg/day (pregnant), 0.193 μg/kg/day (fetus), 1.005–1.253 μg/kg/day (adults), 0.356–0.376 μg/kg/day (adolescents), and 0.595–0.603 μg/kg/day (children) for DEP for human risk assessment were estimated using Korean biomonitoring values. Our study provides valuable insight into human health risk assessment regarding DEP exposure.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11
Fig. 12

Similar content being viewed by others

Abbreviations

DEP:

Diethyl phthalate

MEP:

Monoethyl phthalate

BBP:

Butyl benzyl phthalate

DBP:

Di-n-butyl phthalate

MBP:

Mono-n-butyl phthalate

DEHP:

Di-(2-ethylhexyl) phthalate

DiDP:

Diisodecyl phthalate

MEHP:

Mono-(2-ethylhexyl) phthalate

DiNP:

Diisononyl phthalate

DnOP:

Di-n-octyl phthalate

REACH:

Registration, evaluation, authorization and restriction of chemicals

CPSIA:

Consumer product safety improvement act

RoHS:

Restriction of hazardous substances directive

WHO:

World health organization

US EPA:

United states environmental protection agency

PBPK:

Physiologically based pharmacokinetic

UPLC–ESI–MS/MS:

Ultraperformance liquid chromatography–electrospray ionization-tandem mass spectrometer

MS:

Mass spectrometer

ESI:

Electrospray ionization

HPLC:

High performance liquid chromatography

IV:

Intravenous

MRM:

Multiple reaction monitoring

IS:

Internal standard

LLE:

Liquid–liquid extraction

QC:

Quality control

PP:

Protein precipitation

SD:

Standard deviation

CV:

Coefficient of variation

CL:

Clearance

Vd :

Volume of distribution

BTB:

Blood–testis barrier

NOAEL:

No observed adverse effect level

PK:

Pharmacokinetic

POD:

Points of departure

UF:

Uncertainty factor

RfD:

Reference dose

MOE:

Margin of exposure

References

  • Api A (2001) Toxicological profile of diethyl phthalate: a vehicle for fragrance and cosmetic ingredients. Food Chem Toxicol 39(2):97–108

    CAS  PubMed  Google Scholar 

  • Bekö G, Weschler CJ, Langer S, Callesen M, Toftum J, Clausen G (2013) Children’s phthalate intakes and resultant cumulative exposures estimated from urine compared with estimates from dust ingestion, inhalation and dermal absorption in their homes and daycare centers. PLoS ONE 8(4):e62442

    PubMed  PubMed Central  Google Scholar 

  • Bi Y, Deng J, Murry DJ, An G (2016) A whole-body physiologically based pharmacokinetic model of gefitinib in mice and scale-up to humans. AAPS J 18(1):228–238

    CAS  PubMed  Google Scholar 

  • Blount BC, Silva MJ, Caudill SP, Needham LL, Pirkle JL, Sampson EJ, Lucier GW, Jackson RJ, Brock JW (2000) Levels of seven urinary phthalate metabolites in a human reference population. Environ Health Perspect 108(10):979–982

    CAS  PubMed  PubMed Central  Google Scholar 

  • Calafat AM, McKee RH (2006) Integrating biomonitoring exposure data into the risk assessment process: phthalates [diethyl phthalate and di (2-ethylhexyl) phthalate] as a case study. Environ Health Perspect 114(11):1783–1789

    CAS  PubMed  PubMed Central  Google Scholar 

  • Campbell JL Jr, Yoon M, Ward PL, Fromme H, Kessler W, Phillips MB, Anderson WA, Clewell HJ III, Longnecker MP (2018) Excretion of Di-2-ethylhexyl phthalate (DEHP) metabolites in urine is related to body mass index because of higher energy intake in the overweight and obese. Environ Int 113:91–99

    CAS  PubMed  Google Scholar 

  • Cartwright CD, Owen SA, Thompson IP, Burns RG (2000) Biodegradation of diethyl phthalate in soil by a novel pathway. FEMS Microbiol Lett 186(1):27–34

    CAS  PubMed  Google Scholar 

  • Castillo M, Barcelo D, Pereira A, Neto FA (1999) Characterization of organic pollutants in industrial effluents by high-temperature gas chromatography–mass spectrometry. Trends Anal Chem 18(1):26–36

    CAS  Google Scholar 

  • Chambon P, Riotte M, Daudon M, Chambon-Mougenot R, Bringuier J (1971) Metabolism of dibutyl and diethyl phthalates in the rat. C R Séances Acad Sci 273(22):2165–2168

    CAS  Google Scholar 

  • Clewell RA, Kremer JJ, Williams CC, Campbell JJL, Andersen ME, Borghoff SJ (2008) Tissue exposures to free and glucuronidated monobutylyphthalate in the pregnant and fetal rat following exposure to di-n-butylphthalate: Evaluation with a PBPK model. Toxicol Sci 103(2):241–259

    CAS  PubMed  Google Scholar 

  • CPSC (2008) Consumer Product Safety Improvement Act (CPSIA) of 2008. US Consumer Production Safety Commission, Bethesda

    Google Scholar 

  • Dankovic D, Naumann B, Maier A, Dourson M, Levy L (2015) The scientific basis of uncertainty factors used in setting occupational exposure limits. J Occup Environ Hyg 12(sup1):S55–S68

    PubMed  PubMed Central  Google Scholar 

  • Davies B, Morris T (1993) Physiological parameters in laboratory animals and humans. Pharm Res 10(7):1093–1095

    CAS  PubMed  Google Scholar 

  • ECHA (2008) Guidance on information requirements and chemical safety assessment. Part D: exposure scenario building. Exposure Scenario Building, European Chemical Agency, Helsinki

    Google Scholar 

  • Elder RL (1984) The cosmetic ingredient review-a safety evaluation program. J Am Acad Dermatol 11(6):1168–1174

    CAS  PubMed  Google Scholar 

  • EPA (2006) Approaches for the application of physiologically based pharmacokinetic (pbpk) models and supporting data in risk assessment (Final Report). US Environmental Protection Agency, Washington

    Google Scholar 

  • EU (2002) Restriction of hazardous substances (RoHS) Directive. European Commission

  • FDA (2018) Bioanalytical method validation guidance for industry. US Department of Health and Human Services Food and Drug Administration, Rockville

    Google Scholar 

  • Feás CP, Alonso MCB, Peña-Vázquez E, Hermelo PH, Bermejo-Barrera P (2008) Phthalates determination in physiological saline solutions by HPLC–ES-MS. Talanta 75(5):1184–1189

    Google Scholar 

  • Field EA, Price CJ, Sleet RB, George JD, Marr MC, Myers CB, Schwetz BA, Morrissey RE (1993) Developmental toxicity evaluation of diethyl and dimethyl phthalate in rats. Teratology 48(1):33–44

    CAS  PubMed  Google Scholar 

  • Fisher JS (2004) Environmental anti-androgens and male reproductive health: focus on phthalates and testicular dysgenesis syndrome. Reproduction 127(3):305–315

    CAS  PubMed  Google Scholar 

  • Frederiksen H, Aksglaede L, Sorensen K, Skakkebaek NE, Juul A, Andersson A-M (2011) Urinary excretion of phthalate metabolites in 129 healthy Danish children and adolescents: estimation of daily phthalate intake. Environ Res 111(5):656–663

    CAS  PubMed  Google Scholar 

  • Frederiksen H, Nielsen JKS, Mørck TA, Hansen PW, Jensen JF, Nielsen O, Andersson A-M, Knudsen LE (2013) Urinary excretion of phthalate metabolites, phenols and parabens in rural and urban Danish mother–child pairs. Int J Hyg Environ Health 216(6):772–783

    CAS  Google Scholar 

  • Frederiksen H, Skakkebaek NE, Andersson AM (2007) Metabolism of phthalates in humans. Mol Nutr Food Res 51(7):899–911

    CAS  PubMed  Google Scholar 

  • Fromme H, Gruber L, Schuster R, Schlummer M, Kiranoglu M, Bolte G, Völkel W (2013) Phthalate and di-(2-ethylhexyl) adipate (DEHA) intake by German infants based on the results of a duplicate diet study and biomonitoring data (INES 2). Food Chem Toxicol 53:272–280

    CAS  PubMed  Google Scholar 

  • Gómez-Hens A, Aguilar-Caballos MP (2003) Social and economic interest in the control of phthalic acid esters. Trends Anal Chem 22(11):847–857

    Google Scholar 

  • Gerlowski LE, Jain RK (1983) Physiologically based pharmacokinetic modeling: principles and applications. J Pharm Sci 72(10):1103–1127

    CAS  PubMed  Google Scholar 

  • Goudah A, Hasabelnaby S (2010) Pharmacokinetics, plasma protein binding and bioavailability of moxifloxacin in Muscovy ducks after different routes of administration. Res Vet Sci 88(3):507–511

    CAS  PubMed  Google Scholar 

  • Gray LE Jr, Ostby J, Furr J, Price M, Veeramachaneni DR, Parks L (2000) Perinatal exposure to the phthalates DEHP, BBP, and DINP, but not DEP, DMP, or DOTP, alters sexual differentiation of the male rat. Toxicol Sci 58(2):350–365

    CAS  PubMed  Google Scholar 

  • Gray T, Gangolli SD (1986) Aspects of the testicular toxicity of phthalate esters. Environ Health Perspect 65:229–235

    CAS  PubMed  PubMed Central  Google Scholar 

  • Griffiths WC, Cámara PD, Saritelli A, Gentile J (1988) The in vitro serum protein-binding characteristics of bis-(2-ethylhexyl) phthalate and its principal metabolite, mono-(2-ethylhexyl) phthalate. Environ Health Perspect 77:151–156

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hansen SF, Carlsen L, Tickner JA (2007) Chemicals regulation and precaution: does REACH really incorporate the precautionary principle. Environ Sci Policy 10(5):395–404

    Google Scholar 

  • Hartmann C, Uhl M, Weiss S, Koch HM, Scharf S, König J (2015) Human biomonitoring of phthalate exposure in Austrian children and adults and cumulative risk assessment. Int J Hyg Environ Health 218(5):489–499

    CAS  PubMed  Google Scholar 

  • Hauser R, Calafat A (2005) Phthalates and human health. Occup Environ Med 62(11):806–818

    CAS  PubMed  PubMed Central  Google Scholar 

  • Hernández-Díaz S, Mitchell AA, Kelley KE, Calafat AM, Hauser R (2009) Medications as a potential source of exposure to phthalates in the US population. Environ Health Perspect 117(2):185–189

    PubMed  Google Scholar 

  • Howdeshell KL, Wilson VS, Furr J, Lambright CR, Rider CV, Blystone CR, Hotchkiss AK, Gray LE Jr (2008) A mixture of five phthalate esters inhibits fetal testicular testosterone production in the sprague-dawley rat in a cumulative, dose-additive manner. Toxicol Sci 105(1):153–165

    CAS  PubMed  Google Scholar 

  • Hu T-M, Hayton WL (2001) Allometric scaling of xenobiotic clearance: uncertainty versus universality. AAPS PharmSci 3(4):30–43

    PubMed Central  Google Scholar 

  • Huang P-C, Tsai C-H, Liang W-Y, Li S-S, Pan W-H, Chiang H-C (2015) Age and gender differences in urinary levels of eleven phthalate metabolites in general Taiwanese population after a DEHP episode. PLoS ONE 10(7):e0133782

    PubMed  PubMed Central  Google Scholar 

  • Igari Y, Sugiyama Y, Sawada Y, Iga T, Hanano M (1983) Prediction of diazepam disposition in the rat and man by a physiologically based pharmacokinetic model. J Pharmacokinet Biopharm 11(6):577–593

    CAS  PubMed  Google Scholar 

  • Ioku T, Mukaide A, Kitanaka H, Sakagami Y, Kamevama T (1976) In vitro distribution of drugs. Yakuri to Chiryo 4:510–514

    CAS  Google Scholar 

  • Janjua NR, Mortensen GK, Andersson A-M, Kongshoj B, Skakkebæk NE, Wulf HC (2007) Systemic uptake of diethyl phthalate, dibutyl phthalate, and butyl paraben following whole-body topical application and reproductive and thyroid hormone levels in humans. Environ Sci Technol 41(15):5564–5570

    CAS  PubMed  Google Scholar 

  • Jeong S-H, Jang J-H, Cho H-Y, Lee Y-B (2019) Simultaneous determination of diethyl phthalate and its major metabolite, monoethyl phthalate, in rat plasma, urine, and various tissues collected from a toxicokinetic study by ultrahigh performance liquid chromatography-tandem mass spectrometry. J Pharm Biomed Anal 173:108–119

    CAS  PubMed  Google Scholar 

  • Jones H, Rowland-Yeo K (2013) Basic concepts in physiologically based pharmacokinetic modeling in drug discovery and development. CPT Pharmacometrics Syst Pharmacol 2(8):1–12

    Google Scholar 

  • Kao ML, Ruoff B, Bower N, Aoki T, Smart C, Mannens G (2012) Pharmacokinetics, metabolism and excretion of 14C-monoethyl phthalate (MEP) and 14C-diethyl phthalate (DEP) after single oral and IV administration in the juvenile dog. Xenobiotica 42(4):389–397

    CAS  PubMed  Google Scholar 

  • Kasper-Sonnenberg M, Koch HM, Wittsiepe J, Brüning T, Wilhelm M (2014) Phthalate metabolites and bisphenol A in urines from German school-aged children: results of the Duisburg birth cohort and Bochum cohort studies. Int J Hyg Environ Health 217(8):830–838

    CAS  PubMed  Google Scholar 

  • Kawano M (1980) Toxicological studies on phthalate esters. Jpn J Hyg 35(4):684–692

    CAS  Google Scholar 

  • Kim S-J, Choi E-J, Choi G-W, Lee Y-B, Cho H-Y (2019) Exploring sex differences in human health risk assessment for PFNA and PFDA using a PBPK model. Arch Toxicol 93(2):311–330

    CAS  PubMed  Google Scholar 

  • Kim S-J, Shin H, Lee Y-B, Cho H-Y (2018) Sex-specific risk assessment of PFHxS using a physiologically based pharmacokinetic model. Arch Toxicol 92(3):1113–1131

    CAS  PubMed  Google Scholar 

  • Koch HM, Drexler H, Angerer J (2003) An estimation of the daily intake of di (2-ethylhexyl) phthalate (DEHP) and other phthalates in the general population. Int J Hyg Environ Health 206(2):77–83

    CAS  PubMed  Google Scholar 

  • Lake BG, Phillips JC, Linnell JC, Gangolli SD (1977) The in vitro hydrolysis of some phthalate diesters by hepatic and intestinal preparations from various species. Toxicol Appl Pharmacol 39(2):239–248

    CAS  PubMed  Google Scholar 

  • Laroche C, Aggarwal M, Bender H, Benndorf P, Birk B, Crozier J, Dal Negro G, De Gaetano F, Desaintes C, Gardner I (2018) Finding synergies for 3Rs–toxicokinetics and read-across: report from an EPAA partners' Forum. Regul Toxicol Pharmacol 99:5–21

    PubMed  Google Scholar 

  • Lovekamp-Swan T, Davis BJ (2003) Mechanisms of phthalate ester toxicity in the female reproductive system. Environ Health Perspect 111(2):139–145

    CAS  PubMed  PubMed Central  Google Scholar 

  • Martino-Andrade AJ, Chahoud I (2010) Reproductive toxicity of phthalate esters. Mol Nutr Food Res 54(1):148–157

    CAS  PubMed  Google Scholar 

  • Miura T, Uehara S, Mizuno S, Yoshizawa M, Murayama N, Kamiya Y, Shimizu M, Suemizu H, Yamazaki H (2019) Steady-state human pharmacokinetics of monobutyl phthalate predicted by physiologically based pharmacokinetic modeling using single-dose data from humanized-liver mice orally administered with dibutyl phthalate. Chem Res Toxicol 32(2):333–340

    CAS  PubMed  Google Scholar 

  • Moreau M, Leonard J, Phillips KA, Campbell J, Pendse SN, Nicolas C, Phillips M, Yoon M, Tan Y-M, Smith S, Pudukodu H, Isaacs K, Clewell H (2017) Using exposure prediction tools to link exposure and dosimetry for risk-based decisions: a case study with phthalates. Chemosphere 184:1194–1201

    CAS  PubMed  PubMed Central  Google Scholar 

  • Penalver A, Pocurull E, Borrull F, Marcé R (2001) Comparison of different fibers for the solid-phase microextraction of phthalate esters from water. J Chromatogr A 922(1–2):377–384

    CAS  PubMed  Google Scholar 

  • Philippat C, Mortamais M, Chevrier C, Petit C, Calafat AM, Ye X, Silva MJ, Brambilla C, Pin I, Charles M-A, Cordier S, Slama R (2012) Exposure to phthalates and phenols during pregnancy and offspring size at birth. Environ Health Perspect 120(3):464–470

    CAS  PubMed  Google Scholar 

  • Pilari S, Gaub T, Block M, Görlitz L (2017) Development of physiologically based organ models to evaluate the pharmacokinetics of drugs in the testes and the thyroid gland. CPT Pharmacometrics Syst Pharmacol 6(8):532–542

    CAS  PubMed  PubMed Central  Google Scholar 

  • Reddy BS, Rozati R, Reddy S, Kodampur S, Reddy P, Reddy R (2006) High plasma concentrations of polychlorinated biphenyls and phthalate esters in women with endometriosis: a prospective case control study. Fertil Steril 85(3):775–779

    CAS  PubMed  Google Scholar 

  • Sharma RP, Schuhmacher M, Kumar V (2018) Development of a human physiologically based pharmacokinetic (PBPK) model for phthalate (DEHP) and its metabolites: A bottom up modeling approach. Toxicol Lett 296:152–162

    CAS  PubMed  Google Scholar 

  • Sharma V, McNeill JH (2009) To scale or not to scale: the principles of dose extrapolation. Br J Pharmacol 157(6):907–921

    CAS  PubMed  PubMed Central  Google Scholar 

  • Wahl HG, Hoffmann A, Häring H-U, Liebich HM (1999) Identification of plasticizers in medical products by a combined direct thermodesorption–cooled injection system and gas chromatography–mass spectrometry. J Chromatogr A 847(1–2):1–7

    CAS  PubMed  Google Scholar 

  • Weng T-I, Chen M-H, Lien G-W, Chen P-S, Lin JC-C, Fang C-C, Chen P-C (2017) Effects of gender on the association of urinary phthalate metabolites with thyroid hormones in children: a prospective cohort study in Taiwan. Int J Hyg Environ Health 14(2):123

    Google Scholar 

  • WHO (2003) Diethyl phthalate. Concise international chemical assessment document 52. World Health Organization, Geneva, Switzerland

  • WHO (2010) Characterization and application of physiologically based pharmacokinetic models in risk assessment. IPCS harmonization project document No 9, World Health Organization, Geneva, Switzerland

  • Wittassek M, Koch HM, Angerer J, Brüning T (2011) Assessing exposure to phthalates—the human biomonitoring approach. Mol Nutr Food Res 55(1):7–31

    CAS  PubMed  Google Scholar 

  • Yasuhara A, Shiraishi H, Nishikawa M, Yamamoto T, Uehiro T, Nakasugi O, Okumura T, Kenmotsu K, Fukui H, Nagase M (1997) Determination of organic components in leachates from hazardous waste disposal sites in Japan by gas chromatography–mass spectrometry. J Chromatogr A 774(1–2):321–332

    Google Scholar 

  • Yue Y, Liu J, Liu R, Sun Y, Li X, Fan J (2014) The binding affinity of phthalate plasticizers-protein revealed by spectroscopic techniques and molecular modeling. Food Chem Toxicol 71:244–253

    CAS  PubMed  Google Scholar 

  • Zeman FA, Boudet C, Tack K, Barneaud AF, Brochot C, Pery AR, Oleko A, Vandentorren S (2013) Exposure assessment of phthalates in French pregnant women: results of the ELFE pilot study. Int J Hyg Environ Health 216(3):271–279

    CAS  PubMed  Google Scholar 

  • Zhao G, Peng C, Du W, Wang S (2014) Simultaneous determination of imperatorin and its metabolites in vitro and in vivo by a GC-MS method: application to a bioavailability and protein binding ability study in rat plasma. Biomed Chromatogr 28(7):947–956

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This research did not receive any specific grant from funding agencies in the public, commercial, or not-for-profit sectors.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Hea-Young Cho or Yong-Bok Lee.

Ethics declarations

Conflict of interest

The authors declare that they have no conflicts of interest relevant to this study.

Ethical approval

The animal experiment was approved by the Chonnam National University Animal Experimental Ethics Committee, Republic of Korea (approval number: CNU IACUC-YB-2017-45). In addition, this study was conducted according to revised Guidelines for Ethical Conduct in the Care and Use of Animals and the rules of Good Laboratory Practice.

Additional information

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary file1 (DOCX 334 kb)

Supplementary file2 (DOCX 54 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jeong, SH., Jang, JH., Cho, HY. et al. Risk assessment for humans using physiologically based pharmacokinetic model of diethyl phthalate and its major metabolite, monoethyl phthalate. Arch Toxicol 94, 2377–2400 (2020). https://doi.org/10.1007/s00204-020-02748-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00204-020-02748-9

Keywords

Navigation