Draft Screening Assessment Phthalate Substance Grouping: Sections 9 and 10 (2024)

Environment Canada
Health Canada
October 2017

Back to Sections 1-8

Appendices

Table of contents

  • 9. Potential to cause harm to human health
    • 9.1 Exposure assessment
    • 9.2 Health effects assessment
    • 9.3 Characterization of risk to human health
    • 9.4 Uncertainties in evaluation of cumulative risk to human health
  • 10. Conclusion
  • References

9. Potential to cause harm to human health

9.1 Exposure assessment

9.1.1 Short-chain phthalates

DMP

Exposure estimates were calculated using human biomonitoring data as well as data on DMP occurrence in indoor air, soil, dust, food and cosmetics. They are presented in the SCP SOS report (Environment Canada, Health Canada 2015a) and summarized below. Since the publication of the SOS report, DMP has been analyzed in foods included in the 2013 Canadian Total Diet Study (TDS). While it was not quantified in any food composites above the method detection limit in Canada (average MDL=1.13 ng/g; Cao et al. 2015), it was reported in indoor air in homes in the United States (Tran and Kannan 2015), in agricultural soils in Canada (Khosravi and Price 2015) and in various studies internationally. However, these reported values did not change the previously presented exposure estimates derived using environmental media and food data (Environment Canada, Health Canada 2015a).

The highest exposed group according to biomonitoring data (all sources, Maternal Infant Research on Environmental Chemicals - Child Development Plus study [MIREC-CD Plus]) are male children aged 2 to 3 years, with median and 95th percentile intakes of 0.19 and 0.66 μg/kg bw/day, respectively. For older populations (3 years and over) the highest exposed group (all sources, National Health and Nutrition Examination Survey [NHANES]) is males aged 12 to 19 years, with median and 95thpercentile intakes of 0.042 and 0.29 μg/kg bw/day, respectively.

The subpopulation with the highest exposure to DMP from environmental media and food consisted of breastfed infants, with total daily intake of 0.019 and 0.26 μg/kg bw/dayestimated on the basis of central tendency and upper-bounding concentrations, respectively.

Estimated daily intake of DMP from use of diaper cream (infants aged 0 to 0.5 years, dermal) was 2.7 μg/kg bw/day (lower-end exposure scenario) and 8.2 μg/kg bw/day (upper-bounding exposure scenario). For adults aged 20 years and over, estimated intakes from the use of hairsprays and hair dyes were 6.6 μg/kg bw/day (lower end) and 20 μg/kg bw/day (upper bound) and 140 μg/kg bw/event (lower end) and 420 (upper bound) μg/kg bw/event, respectivelyFootnote 5.

For more details, please refer to the SCP SOS report (Environment Canada, Health Canada 2015a).

9.1.2 Medium-chain phthalates and additional phthalates

Medium-chain phthalates

For detailed information on all 10 medium-chain phthalates in this section, please refer to the MCP SOS report (Environment Canada, Health Canada 2015b).

DIBP

Exposure estimates were calculated using biomonitoring data as well as data on DIBP occurrence in air, drinking water, dust, food, plastic items and cosmetics. They are presented in the MCP SOS report (Environment Canada, Health Canada 2015b) and summarized below. Since the publication of the SOS report, DIBP has been analyzed in foods included in the 2013 Canadian TDS. DIBP was detected in 27 of 159 composite food samples at an average MDL of 7.25 ng/g (mean of positives = 8.26 ng/g, range of 2.41 to 39.8 ng/g; Cao et al. 2015). DIBP has also been detected in indoor air in homes in the United States (Tran and Kannan 2015) and in various studies internationally. However, these reported values did not significantly change the previously presented exposure estimates derived using biomonitoring and environmental media data (Environment Canada, Health Canada 2015b).

The highest exposed group according to biomonitoring data (all sources, Canadian Health Measures Survey [CHMS]) is male children aged 6 to 11 years, with estimated mean intakes of 1.5 μg/kg bw/day and median and 95th percentile intakes of 0.76 and 5.3 μg/kg bw/day, respectively. For older populations (12 years and over), the highest exposed group (all sources, CHMS) is 20-to-49-year-old females, with mean intakes of 0.56 μg/kg bw/day and median and 95th percentile intakes of 0.46 and 1.4 μg/kg bw/day, respectively.

The subpopulation with the highest exposure from environmental media and food consisted of breastfed infants, with total daily intakes of 1.6 and 5.9 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively. Daily intakes for DIBP for infants (0 to 1.5 years) from exposure from mouthing plastic toys and childcare articles were estimated at 62.8 and 251 μg/kg bw/day on the basis of lower-end and upper-bounding exposure scenarios, respectively. Estimated daily intakes for infants from dermal exposure to plastic items, based on a lower-end and upper-bounding exposure scenarios, were 30.7 and 245.3 μg/kg bw/day, respectively. For adults exposed to plastic items (females aged 20 years and over), estimated daily intakes, based on lower-end and upper-bounding exposure scenarios, were 30.8 and 96.3 μg/kg bw/day, respectively. Estimated daily intake of DIBP from use of body lotions was 0.03 μg/kg bw/day for adults aged 20 to 59 years.

DCHP

Exposure estimates were calculated using data on DCHP occurrence in dust and food. Since the publication of the MCP SOS report, DCHP has been analyzed in foods included in the 2013 Canadian TDS but was detected in only 1 of 159 composite food samples, at a concentration of 64.9 ng/g (average MDL=1.58 ng/g; Cao et al. 2015). However, this new information did not significantly change the exposure estimates presented previously (Environment Canada, Health Canada 2015b).

The subpopulation with the highest exposure from environmental media and food consisted of children (aged 6 months to 4 years), with total daily intakes of 0.0018 and 0.15 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively. For populations aged 12 years and over, the highest exposed group is adolescents (aged 12 to 19 years), with total daily intakes of less than 0.001 and 0.065 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively.

Intakes were not calculated on the basis of biomonitoring data, because the majority of samples were measured below the limit of detection (CHMS, MIREC-CD Plus, Maternal Infant Research on Environmental Chemicals study [MIREC], Plastics and Personal Care Product Use in Pregnancy survey [P4]).

DMCHP

Exposure estimates were calculated using data on DMCHP occurrence in dust. They are presented in the MCP SOS report (Environment Canada, Health Canada 2015b) and summarized below.

The subpopulation with the highest exposure from dust consisted of infants (0 to 6 months old) with total daily intake of 0.0027 and 0.054 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively. For populations aged 12 years and over, total daily intakes are less than 0.001 μg/kg bw/day.

DBzP

Exposure estimates were calculated using data on DBzP occurrence in dust. Since the publication of the MCP SOS report, DBzP has been analyzed in foods included in the 2013 Canadian TDS and was not quantified above the MDL (average MDL=12.7; Cao et al. 2015). Exposure estimates for DBzP are presented in the MCP SOS report (Environment Canada, Health Canada 2015b) and summarized below.

The subpopulation with the highest exposure from dust consisted of infants (0 to 6 months old), with total daily intakes of 0.016 and 0.097 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively. For populations aged 12 years and over, total daily intakes of less than 0.001 and 0.0011 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively, are observed for all age groups.

B84P

Exposure estimates were calculated using data on B84P occurrence in dust and plastic items. They are presented in the MCP SOS report (Environment Canada, Health Canada 2015b) and summarized below.

The subpopulation with the highest exposure from dustFootnote 6 consisted of infants (0 to 6 months old), with total daily intakes of 0.0063 and 0.047 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively.

Estimated daily intakes for infants from dermal exposure to B84P from plastic items, calculated on the basis of lower-end and upper-bounding exposure scenarios, were 2.7 and 21.6 μg/kg bw/day, respectively. For adults exposed to plastic items (females aged 20 years and older), lower-end and upper-bounding estimates of daily intake were 2.7 and 8.5 μg/kg bw/day, respectively.

DIHepP

Exposure estimates were calculated using data on DIHeP occurrence in dust. They are presented in the MCP SOS report (Environment Canada, Health Canada 2015b) and summarized below.

The subpopulation with the highest exposure from dust consisted of infants (0 to 6 months old), with total daily intakes of 0.096 and 1.1 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively (for more details refer to Environment Canada, Health Canada 2015b). For populations aged 12 years and over, the highest exposed group is adolescents (aged 12 to 19 years), with total daily intakes of 0.0011 and 0.013 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively.

B79P

Exposure estimates were calculated using data on B79P occurrence in dust and plastic items. They are presented in the MCP SOS report (Environment Canada, Health Canada 2015b) and summarized below.

The subpopulation with the highest exposure from dust consisted of infants (0 to 6 months old), with total daily intakes of 0.0063 and 0.047 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively. For populations aged 12 years and over, total daily intakes are less than 0.001 μg/kg bw/day.

Estimated daily intakes for infants from dermal exposure to B79P from plastic items, as calculated on the basis of lower-end and upper-bounding exposure scenarios, were 2.7 and 21.6 μg/kg bw/day, respectively. For adults exposed to plastic items (females aged 20 years and over), estimated daily intakes, based on lower-end and upper-bounding exposure scenarios, were 2.7 and 8.5 μg/kg bw/day respectively.

DINP

Exposure estimates were calculated using biomonitoring data as well as data on DINP occurrence in dust, food, and plastic items. They are presented in the DINP SOS report (Environment Canada, Health Canada 2015c) and summarized below. Recently, DINP has been reported internationally in soils (Tran et al. 2015), indoor air (Blanchard et al. 2014, Takeuchi et al. 2014), settled dust (Blanchard et al. 2014; Luongo and Östman 2015) and tap water (Yang et al. 2014). However, these reported values did not change the exposure estimates presented previously.

The highest exposed group according to biomonitoring data (all sources, NHANES) is male children aged 6 to 11 years, with mean intakes of 4.6 μg/kg bw/day, and median and 95thpercentile intakes of 4.2 and 25 μg/kg bw/day, respectively. For older populations (aged 12 years and over), the highest exposed group (all sources, NHANES) is 12-to-19-year-old males, with mean intakes of 3.0 μg/kg bw/day, and median and 95thpercentile intakes of 2.6 and 33 μg/kg bw/day, respectively. For adults aged 20 years and over, daily intakes are 2.8 μg/kg bw/day (mean), 2.4 μg/kg bw/day (median), and 24 μg/kg bw/day (95th percentile) for males and 2.3 μg/kg bw/day (mean), 1.9 μg/kg bw/day (median), and 23 μg/kg bw/day (95th percentile) for females.

The subpopulation with the highest exposure from dust and food consisted of children (aged 6 months to 4 years), with total daily intakes of 1.8 and 19.7 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively. For populations aged 12 years and over, the highest exposed group is adolescents (aged 12 to 19 years), with total daily intakes of 1.0 and 11.4 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively.

Daily intakes for DINP for infants (aged 0 to 1.5 years) from mouthing plastic toys and childcare articles, were estimated on the basis of lower-end and upper-bounding exposure scenarios, to be 30 and 120 μg/kg bw/day, respectively. Estimated daily intakes for infants from dermal exposure to plastic items, based on lower-end and upper-bounding exposure scenarios, were 1.1 and 8.6 μg/kg bw/day, respectively. For females aged 20 years and over, estimated daily intakes from dermal exposure to plastic items, based on lower-end and upper-bounding exposure scenarios, were 1.1 and 3.4 μg/kg bw/day, respectively.

CHIBP, BCHP and BIOP

Given the absence of reporting to the s. 71 industry survey (CHIBP, BCHP and BIOP), non-detection in dust (CHIBP and BCHP), non-detection in products used by consumers (BCHP; emission chamber study, NRC 2012), negligible modelled indoor air concentrations (CHIBP), and the absence of information as to their presence in product databases (CHIBP, BCHP and BIOP), general population exposure to CHIBP, BCHP and BIOP from environmental media and products used by consumers is expected to be negligible (Environment Canada, Health Canada 2015b).

Additional medium-chain phthalates

BBP
Biomonitoring

Monobenzylphthalate (MBzP), the main monoester metabolite of BBP, was monitored in the CHMS Cycle 1 (2007-2009) and Cycle 2 (2009-2011), with 100% detection in all samples (Health Canada 2011b, 2013). Additionally, MBzP was measured as part of the First Nations Biomonitoring Initiative (FNBI) of the Assembly of First Nations (n=492 for on-reserve and Crown-land populations aged 20 years and over). Urine concentrations for MBzP (geometric mean) were reported to be statistically higher in FNBI samples than those observed in the CHMS population (AFN 2013).

MBzP was also monitored by Health Canada in three cohort surveys: P4 (n = 31 women and their infants, 542 individual spot samples, women provided multiple urine samples over two visits), MIREC (n = 1742 pregnant women, spot urine samples), and MIREC-CD Plus (197 children aged 2 to 3 years, 1 spot sample per individual). All three surveys reported high detection frequencies of MBzP (100%, 99% and 97%, respectively) (personal communication from Environmental Health Sciences and Radiation Directorate [EHSRD], Health Canada, to Existing Substances Risk Assessment Bureau [ESRAB], Health Canada, October 2013, 2014, unreferenced; Arbuckle et al. 2014).

Finally, in the United States, the NHANES also monitored MBzP in urine during survey years 1999 to 2012 and reported high detection frequencies (CDC 2014).

Using the CHMS, P4, MIREC and MIREC-CD Plus datasets, reverse dosimetry intake estimates were generated. Metabolite concentrations were adjusted for urine dilution using the creatinine correction method, a commonly used method for phthalate biomonitoring assessment (Fromme 2007; Frederiksen et al. 2013; Christensen et al. 2014; US CPSC CHAP 2014). Daily creatinine excretion rates for participants were estimated using the Mage equation. Biomonitoring intakes are presented in Table 9-1 below (see Appendix C for further information on the methodology).

Table 9-1. Biomonitoring daily intakes (μg/kg bw/day) for BBP a
Age group Study Male/Female n Arithmetic mean 50th 75th 95th
1-4 months P4 males and females 48 0.607 0.253 0.513 1.802
2-3 years MIREC-CD Plus males and females 198 0.873 0.379 0.853 2.97
3-5 years CHMS males and females 519 1.4 0.76 1.6 4.5
6-11 years CHMS males 261 1.2 0.79 1.6 3.4
6-11 years CHMS females 253 0.93 0.61 1.1 2.7
12-19 years CHMS males 255 0.53 0.36 0.59 1.4
12-19 years CHMS females 255 0.46 0.28 0.53 1.6
18+ years MIREC females (pregnant) 1727 0.53 0.27 0.53 1.60
19+ years P4 females (pregnant) 31c 1.0 0.31 0.86 3.01
20-49 years CHMS males 290 0.33 0.2 0.35 0.97b
20-49 years CHMS females 286 0.37 0.19b 0.36b 1.2
50-79 years CHMS males 211 0.22 0.13 0.23 0.61b
50-79 years CHMS females 216 0.24 0.15 0.3 -

- = No data.
a. Data for males and females from: P4 and MIREC pregnant women, P4 infants, MIREC-CD Plus children (preliminary results), and CHMS (Cycle 2).
b. Use data with caution.
c. n = 31 women, 542 individual spot samples, women provided multiple urine samples over two visits.

The highest exposed group according to biomonitoring data (all sources, CHMS) is children aged 3 to 5 years with median and 95th percentile intakes of 0.76 and 4.5 μg/kg bw/day, respectively. For older populations, the highest exposed group (all sources, P4) is pregnant women (aged 19 years and over) with median and 95th percentile intakes of 0.31 and 3.01 μg/kg bw/day, respectively.

Environmental media and food

The subpopulation with the highest exposure to BBP from environmental media and food consisted of children (aged 0.5 to 4 years ), with total daily intakes of 0.58 and 2.99 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively (Appendix D, Table D-1a).

Indoor air and dust

One study measured BBP in indoor air in Canadian homes; all samples were less than the limit of detection (LOD) (LOD was not reported; Zhu et al. 2007). Recently, BBP has also been measured in indoor air in homes in the United States (detected in 100% of 20 samples from homes in Albany, NY, method quantification limit = 0.20 ng/m3, median: 2.99 ng/m3, maximum: 24.7 ng/m3; Tran and Kannan 2015) and internationally (Fromme et al. 2004; Rudel et al. 2010; Bergh et al. 2011a; Pei et al. 2013; Blanchard et al. 2014; Lin et al. 2014; Takeuchi et al. 2014). As no Canadian indoor air survey reported BBP above the LOD, median (2.99 ng/m3) and maximum (24.7 ng/m3) concentrations from the US study (Tran and Kannan 2015) were used to estimate the general population daily intake of BBP from indoor air (Appendix D, Table D-1a).

BBP was surveyed in the Canadian House Dust Study (CHDS) and was detected in 100% of homes (range: 0.6 to 944 μg/g, median: 42.3 μg/g, 95th percentile: 512 μg/g) (Kubwabo et al. 2013). Internationally, BBP has also been reported in house dust (Fromme et al. 2004; Bornehag et al. 2005; Kolarik et al. 2008; Langer et al. 2010; Bergh et al. 2011a; Guo and Kannan 2011; Hsu et al. 2012; Kang et al. 2012; Gevao et al. 2013; Orecchio et al. 2013; Papadopoulos et al. 2013; Blanchard et al. 2014; Lin et al. 2014; Dodson et al. 2015; Luongo and Östman 2015).

BBP has applications as a plasticizer in the manufacturing of automobiles and automobile parts (ECHA 2012; Environment Canada 2014). For the general population, indirect exposure (e.g., off-gassing) is considered a relevant source, but no data on this exposure source has been identified, which is currently an uncertainty in the draft screening assessment.

The Canadian survey (Kubwabo et al. 2013) was identified as the key study for exposure characterization, and median (42.3 μg/g) and 95th percentile (512 μg/g) concentrations were used to estimate the Canadian general population daily intake of DBP from dust (Appendix D, Table D-1a).

Food, infant formula and breast milk

In Canada, BBP presence in food was measured in samples from the 2013 Canadian TDS (Cao et al. 2015). BBP was detected in 32 of 159 food composite samples (average MDL = 3.10 ng/g), with a mean concentration of 12.4 ng/g and a range of 1.86 to 82.7 ng/g (Cao et al. 2015). Milk, soft drinks, and fruit juice are the major sources of intake for all population groups. In addition, BBP presence in food was monitored as part of the Canadian Food Inspection Agency's (CFIA) 2013-2014 and 2014-2015 Food Safety Action Plan (FSAP) surveys (personal communication from the Food Directorate, Health Canada, to ESRAB, Health Canada, April 2014; unreferenced). BBP was detected in less than 1% of 1518 (LOD: 0.1 μg/g) packaged and processed food samples at concentrations ranging from 0.3 to 1.9 μg/g, with a mean of the positive samples of 0.75 μg/g.

Additionally, breast milk was monitored as part of the MIREC study. BBP was not detected in any samples (n=305; MDL=0.00741 μg/g; personal communication from the Food Directorate, Health Canada, to ESRAB, Health Canada, October 2014; unreferenced). Internationally, BBP was detected in breast milk from one study in Sweden (Högberg et al. 2008) and not detected in another study in Germany (Fromme et al. 2011). However, BBP readily metabolizes to MBzP in the human body (Koch and Calafat 2009; Frederiksen et al. 2011), and as a result, BBP, as the parent compound, may not be found in breast milk in high quantities.

MBzP was measured in breast milk as part of the P4 study (n = 31 women, 56 breast milk samples collected from study participants). It was detected in 34% of breast milk samples, with the median value reported as below the LOD (0.018 μg/L) and the maximum value reported as 0.16 μg/L (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013; unreferenced). Internationally, MBzP has been detected in breast milk (Mortensen et al. 2005; Högberg et al. 2008).

In the P4 study, MBzP was detected in less than 5% of samples of infant formula (n=23; LOD=0.018 μg/L) (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013; unreferenced). The CFIA surveillance data from the 2013-2014 and 2014-2015 FSAP surveys did not detect BBP (LOD = 0.1 μg/g) in any samples of infant foods (n=44), infant formula (n=59) or infant cereals (n=19). Internationally, BBP was not detected in infant formulas in two European studies (Sørensen 2006; Bradley et al. 2013a).

Using Canadian TDS data, probabilistic estimates of dietary intakes were derived for BBP and results are outlined in Appendix D, Table D-1b (for methodology for estimating probabilistic intakes, see Appendix E). Results for BBP in breast milk and infant formula that were below the LOD (i.e., 0.018 μg/L) were set to half the LOD (Appendix D, Table D-1a).

Ambient air, drinking water and soil

No Canadian data were identified for BBP in ambient air. Rudel et al. (2010) measured outdoor air outside homes in the United States (detected in 5% of 43 samples [method reporting limit = 6 ng/m3], median: not reported, maximum: 8.5 ng/m3). Internationally, BBP has been reported in ambient air (Li and Wang 2015). Rudel et al. (2010) was identified as the relevant study for exposure characterization (sampling from North America), and half the method reporting limit (method reporting limit [MRL] = 6 ng/m3) and maximum (8.5 ng/m3) concentrations were used to estimate potential exposures to BBP via ambient air (Appendix D, Table D-1a).

No Canadian data were identified for BBP in drinking water. BBP was analyzed for, but not detected in, Canadian bottled water (MDL: 0.085 μg/L; Cao 2008). Similarly, BBP was not detected in any samples of well water intended to be bottled in Spain (Bono-Blay et al. 2012). Internationally, BBP has been detected in bottled water (Jeddi et al. 2015, Lv et al. 2015), drinking water (Liu et al. 2015), and surface waters (Net et al. 2015, Selvaraj et al. 2015). It was not detected in any samples of tap water or river water in Spain (Dominguez-Morueco et al. 2014). In the absence of Canadian or North American data on levels of BBP in tap water, half the MDL (i.e., 0.085 μg/L) for BBP in bottled water was used to estimate the general population daily intake from drinking water (Cao et al. 2008) in Canada (Appendix D, Table D-1a).

Khosravi and Price (2015) reported various phthalates in agricultural soils in both control and biosolid amended soils collected in Nova Scotia, Canada. BBP was reported at 0.13 ng/g (control soil) and 2.4 ng/g (biosolid amended soil) (Khosravi and Price 2015). Internationally, BBP has been detected in soils (Cheng et al. 2015; Tran et al. 2015; Wang et al. 2015). Conversely, Hongjun et al. (2014) did not detect BBP in any samples of urban, suburban or rural soils in China. The reported concentration of BBP in control agricultural soils (0.13 ng/g; Khosravi and Price 2015) was used to estimate potential exposures to BBP via soil in Canada (Appendix D, Table D-1a).

Products used by consumers

Globally, BBP may also be present in a wide variety of manufactured items, including childcare articles, children's toys, do-it-yourself (DIY) products, paints and exercise balls (HPD 1993- ; ECHA 2012; Korfali et al. 2013;AGDH 2015). In Canada, BBP has also been reported as a plasticizer used in various types of manufactured items (NRC 2012; See Table 2-2; Environment Canada 2014).

Toys and childcare articles

Health Canada has surveyed soft vinyl toys and childcare articles over a number of years for multiple phthalates, including BBP, and it was detected in only one sample from the 2008 survey (below the restricted limit) and has not been detected since (Health Canada 2007, 2009, 2012, 2014). BBP was also monitored in toys purchased in Canada, but made elsewhere; it was detected at 0.001% to 0.02% (Stringer et al. 2000). Internationally, BBP was monitored in toys in India. It was detected in 3 of 24 toy samples, all at less than 0.1% (Johnson et al. 2011).

Currently, Canada (like the United States and the European Union) has regulations in place limiting the amount of certain phthalates (including BBP) in toys and childcare articles (Phthalates Regulations under the Canada Consumer Product Safety Act [Canada 2016]). The European Union (EU)'s Rapid Alert System for Non-Food (RAPEX) database also shows low reporting of toys tested in violation of the regulation for BBP (RAPEX 2015), and the United States Consumer Product Safety Commission (US CPSC) recently stated that less than 10% of exposures to BBP for infants and children would result from mouthing of toys and childcare articles (US CPSC CHAP 2014).

Given the absence of BBP in toys and childcare articles reported in Canadian and international monitoring studies, exposure is expected to be negligible and oral intakes from mouthing toys and/or childcare articles were not estimated.

Cosmetics

On the basis of notifications submitted under the Cosmetic Regulations, BBP is not expected to be present in cosmetics in Canada (personal communication from the Consumer Product Safety Directorate [CPSD], Health Canada, to ESRAB, Health Canada, July 2015; unreferenced). Internationally, BBP has been detected in various types of cosmetics and personal care productsFootnote 7 (Guo and Kannan 2013; Guo et al. 2013; Bao et al. 2015). In contrast, Liang et al. (2013) did not detect BBP in cosmetics in China. This presence may be due to potential migration from packaging. A summary of recent studies measuring concentrations of BBP in cosmetics and personal care products reported in North America is outlined in Table 9-2.

Table 9-2. Concentrations of BBP in cosmetics and personal care products
Detection frequency and product typesa Concentration
(μg/g)
Reference
(country)
12% of 41 rinse-off products ND - 0.18 Guo and Kannan 2013
(United States)
13% of 109 leave-on products ND - 78.3 Guo and Kannan 2013
(United States)
5% of 20 baby products ND - 0.14 Guo and Kannan 2013
(United States)

a. Limits of detection: Koniecki et al. 2011 (0.1 μg/g), Guo and Kannan 2013 (0.01 μg/g)

BBP was not reported as used in Canada. The United States reported low detection frequencies (5% to 13%), and the majority of the concentrations in all studies were in the sub-ppm range. Exposure to BBP from personal care products and cosmetics is therefore not considered significant. In addition, NICNAS (AGDH 2015) and US CPSC CHAP (2014) reported that the cosmetic use of BBP is likely to be rare and they did not assess exposure from this source. For that reason, exposure estimates from this source were not generated.

Other products used by consumers

Globally, BBP may be found in paint products (US CPSC CHAP 2014; AGDH 2015). This use has also been reported in Canada (Environment Canada 2014). The US CPSC CHAP (2014) reported that aerosol paints may contribute to greater than 10% of exposure to BBP for adults, infants, toddlers and children.

In addition, BBP may have applications in the production of articles that may come in contact with skin (ECHA 2012; AGDH 2015). However, BBP was reported in only 1 of 35 samples of children's clothing (Brigden et al. 2013). The US CPSC CHAP (2014) did not evaluate dermal exposure to items containing BBP.

However, since exposure to BBP from these uses is considered to be captured by the available Canadian biomonitoring data estimates of exposure were not calculated.

DBP
Biomonitoring

Mono-n-butyl phthalate (MnBP), the major monoester metabolite of DBP, was monitored in the CHMS Cycle 1 (2007-2009) and Cycle 2 (2009-2011) with 100% detection in all samples (Health Canada 2011b, 2013). Additionally, MnBP was measured as part of the First Nations Biomonitoring Initiative (n=492 for on-reserve and Crown-land populations aged 20 years and over). Urine concentrations for MnBP (geometric mean) were reported to be similar in FNBI samples to those reported in the CHMS (AFN 2013).

MnBP was also monitored by Health Canada in three cohort surveys: P4, MIREC and the MIREC-CD Plus. All three surveys reported high detection frequencies of MnBP (100%, 99% and 100%, respectively; personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013, 2014, unreferenced; Arbuckle et al. 2014). An additional metabolite of DBP (MHBP) was also monitored and detected at high frequency (92% and 100%, respectively) in the P4 and MIREC-CD Plus studies (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013, 2014; unreferenced).

Using the CHMS, P4, MIREC and MIREC-CD Plus datasets, reverse dosimetry intake estimates were generated as previously described. Biomonitoring intakes are presented in Table 9-3 below (see Appendix C for further information on the methodology).

Table 9-3. Biomonitoring daily intakes (μg/kg bw/day) for DBP a
Age group Study Male/Female n Arithmetic mean 50th 75th 95th
1-4 months P4 males and females 48 0.830 0.572 1.126 1.900
2-3 years MIREC-CD Plus males and females 192 1.19 0.939 1.39 2.71
3-5 years CHMS males and females 519 2.4 1.7 2.5 5.3b
6-11 years CHMS males 260 - 1.3 2.3b -
6-11 years CHMS females 253 - 1.3 2.1 5.3b
12-19 years CHMS males 255 1.4 0.85 1.4 3.2b
12-19 years CHMS females 255 0.84 0.71 1.1 1.8
18+ years MIREC females (pregnant) 1728 1.24 0.66 1.04 2.66
19+ years P4 females (pregnant) 31c 1.39 0.55 0.96 4.11
20-49 years CHMS males 290 0.86 0.58 0.9 1.8b
20-49 years CHMS females 284 0.91b 0.55 0.79 0.6b
50-79 years CHMS males 210 0.6 0.43 0.67 1.5
50-79 years CHMS females 216 0.69 0.51 0.72 1.7b

- = No data.
a. Data for males and females from: P4 and MIREC pregnant women, P4 infants, MIREC-CD Plus children (preliminary results), and CHMS (Cycle 2).
b. Use data with caution.
c. n = 31 women, 542 individual spot samples, women provided multiple urine samples over two visits.

The highest exposed group according to biomonitoring data (all sources, CHMS) is children aged 3 to 5 years, with median and 95th percentile intakes of 1.7 and 5.3 μg/kg bw/day, respectively. For older populations, the highest exposed group (all sources, P4) is pregnant women (aged 19 years and over), with median and 95th percentile intakes of 0.55 and 4.11 μg/kg bw/day, respectively.

Environmental media and food

The subpopulation with the highest exposure to DBP from environmental media and food consisted of children (aged 0.5 to 4 years), with total daily intakes of 0.88 and 2.96 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively (Appendix D, Table D-2a).

Indoor air and dust

One study measured DBP in indoor air in Canadian homes (LOD was not reported; Zhu et al. 2007). They reported air concentrations of 200 ng/m3 (median) and a range of 130 to 1100 ng/m3 (Zhu et al. 2007). Recently, DBP has also been measured in indoor air in homes in the United States (detected in 100% of 20 samples from homes in Albany, NY, method quantification limit = 0.10 ng/m3, median: 22.6 ng/m3, maximum: 111 ng/m3; Tran and Kannan 2015) and internationally (Blanchard et al. 2014; Lin et al. 2014; Takeuchi et al. 2014). The Canadian indoor air survey concentrations (median: 200 ng/m3 and maximum: 1100 ng/m3; Zhu et al. 2007) were used to estimate the general population daily intake of DBP from indoor air in Canada (Appendix D, Table D-2a).

DBP was surveyed in the CHDS and was detected in 99% of homes (range: ND-1392 μg/g, median: 16.8 μg/g, 95thpercentile: 95.4 μg/g; Kubwabo et al. 2013). Internationally, DBP has also been reported in house dust (Dodson et al. 2015; Luongo and Östman 2015).

DBP has applications as a plasticizer in the manufacturing of automobiles and automobile parts (ECHA 2012; AGDH2013a). For the general population, indirect exposure (e.g., off-gassing) is considered a relevant source, but no data on this exposure source has been identified, which is currently an uncertainty in the draft screening assessment.

The Canadian survey (Kubwabo et al. 2013) was identified as the key study for exposure characterization, and median (16.8 μg/g) and 95th percentile (95.4 μg/g) concentrations were used to estimate the Canadian general population daily intake of DBP from dust (Appendix D, Table D-2a).

Food, infant formula and breast milk

In Canada, DBP presence in food was monitored in samples from the 2013 Canadian TDS (Cao et al. 2015). DBP was detected in 44 of 159 food composite samples (average MDL=16.6 ng/g) with a mean concentration of the positive samples of 23.2 ng/g and a range of 6.21 - 208 ng/g (Cao et al. 2015). Milk, soft drinks, bread, and ice cream are the major sources of intake for the overall population. In addition, DBP presence in food was monitored as part of the CFIA 2013-2014 and 2014-2015 FSAP surveys (personal communication from the Food Directorate, Health Canada, to ESRAB, Health Canada, April 2014; unreferenced). DBP was detected in 14% of 1518 (LOD: 0.1 μg/g) packaged and processed food samples. DBP concentrations ranged from 0.26 to 4.3 μg/g with a mean concentration of the positive samples of 0.76 μg/g.

MnBP was measured in breast milk as part of the P4 study (n = 31 women, 56 breast milk samples collected from study participants, LOD=0.057 μg/L). It was detected in 100% of breast milk samples, with median and maximum values reported as 0.656 and 5.18 μg/L, respectively (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013; unreferenced). Internationally, MnBP has been measured in breast milk in Europe (Fromme et al. 2011).

Breast milk was also monitored as part of the MIREC study. DBP was detected in 21 samples (n=305, median=0.0129 μg/g, range= less than MDL-0.030 μg/g, MDL=0.0149 μg/g; personal communication from Food Directorate, Health Canada, to ESRAB, Health Canada, October 2014; unreferenced). Internationally, DBP was detected in breast milk from one study in Sweden (Högberg et al. 2008) and Germany (Fromme et al. 2011). However, DBP readily metabolizes to MnBP in the human body (Koch and Calafat 2009) and would therefore not be expected in breast milk in high frequency.

Recently, an analysis of 23 infant formula samples in the P4 study showed 80% detection of MnBP (LOD: 0.057 μg/L, median: 0.299 μg/L, maximum: 1.16 μg/L) (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013; unreferenced). However, measurement of this metabolite in infant formula was affected by high field blank levels (possible contamination); and therefore, was not used to quantify intakes. The CFIA surveillance data did detect DBP in 3 samples of infant foods (n=20, median: ND, 95th percentile: 0.675 μg/g), 12 samples of infant formula (n=32, median: ND, 95thpercentile: 1.12 μg/g) and 4 samples of infant cereals (n=7, median: 0.42 μg/g, 95th percentile: 1.79 μg/g; personal communication from the Food Directorate, Health Canada, to ESRAB, Health Canada, April 2014; unreferenced). Internationally, DBP has been detected in infant formulas in Italy and Europe (Cirillo et al. 2015), but not in the United Kingdom (Bradley et al. 2013a).

Using TDS data, probabilistic estimates of dietary intakes for DBP were derived for the Canadian general population and results are outlined in Appendix B, Table B-2b (the methodology for estimating probabilistic intakes is provided in Appendix E). Median (0.656 μg/L) and maximum (5.18 μg/L) values of MnBP (metabolite of DBP) in breast milk measured in the P4 study was used for risk characterization (Appendix D, Table D-2a).

Ambient air, drinking water and soil

No Canadian data were identified for DBP in ambient air. Rudel et al. (2010) measured outdoor air outside homes in the United States (detected in 35% of 43 samples [MRL = 7 ng/m3], maximum: 32 ng/m3). Internationally, DBP has been reported in ambient air (Li and Wang 2015). Rudel et al. (2010) was identified as the relevant study for exposure characterization (sampling from North America), and half the MRL (7 ng/m3) and maximum (32 ng/m3) concentrations were used to estimate potential exposures to DBP via ambient air (Appendix D, Table D-2a).

No Canadian data were identified for DBP in drinking water. In Canada, Cao et al. (2008) surveyed phthalates in bottled carbonated and non-carbonated water and detected and quantified DBP in all 11 samples (range: 0.075 - 1.72 μg/L). Internationally, DBP was also detected in bottled water samples (Jeddi et al. 2015; Lv et al. 2015), surface waters (Li et al. 2015; Net et al. 2015; Selvaraj et al. 2015), and tap water (Liu et al. 2015). In the absence of Canadian and North American data on levels of DBP in tap water, mean (0.357 μg/L) and maximum (1.72 μg/L) concentrations of DBP in bottled non-carbonated water were used to estimate the general population daily intake from drinking water (Cao et al. 2008) in Canada (Appendix D, Table D-2a).

Khosravi and Price (2015) reported various phthalates in agricultural soils in both control and biosolid amended soils collected in Nova Scotia, Canada. DBP was reported at 0.14 ng/g (control soil) and 1.1 ng/g (biosolid amended) (Khosravi and Price 2015). Internationally, DBP has been detected in soils (Cheng et al. 2015; Tran et al. 2015; Wang et al. 2015). The reported concentration of DBP in control agricultural soils (0.14 ng/g; Khosravi and Price 2015) was used to estimate potential exposures to DBP via soil in Canada (Appendix D, Table D-2a).

Products used by consumers

Globally, DBP may also be present in a wide variety of manufactured items, including childcare articles, children's toys, DIY products, gloves and exercise balls (HPD 1993- ; Stringer et al. 2000; ECHA 2012; Chao et al. 2013; Korfali et al. 2013; AGDH 2013a). In Canada, DBP has also been reported as a plasticizer used in various types of manufactured items (NRC 2012; See Table 2-2; Environment Canada 2014).

Toys and childcare articles

Numerous studies have examined the concentrations of DBP in childcare articles and toys (Stringer et al. 2000; Biedermann-Brem et al. 2008; Johnson et al. 2011; Korfali et al. 2013). A summary of DBP reported in toys and childcare articles available in Canada are presented in Table 9-4.

Table 9-4. Percent content of DBP in various soft vinyl toys and childcare articles available in Canada
Detection frequency Percent content Reference
0 of 117 samples less than 0.1% Health Canada 2014
(Canada)
1 of 62 samples greater than 0.1% Health Canada 2012
(Canada)
1 of 38 samples greater than 0.1% Health Canada 2009
(Canada)
4 of 72 samples greater than 0.1% Health Canada 2007
(Canada)

Currently, Canada (like the United States and the European Union) has regulations in place limiting the amount of certain phthalates (including DBP) in toys and childcare articles (Phthalates Regulations under the Canada Consumer Product Safety Act [Canada 2016]). The EU's RAPEX database also shows low reporting of toys tested in violation of the regulation for DBP (RAPEX 2015), and the US CPSC recently stated that less than 10% of exposures to DBP for infants and children would result from mouthing of toys and childcare articles (US CPSC CHAP 2014).

Given the absence of DBP in toys and childcare articles reported in Canadian monitoring studies and the low frequency and low percent content of DBP reported in international monitoring studies, exposure is expected to be negligible and oral intakes from mouthing toys and/or childcare articles were not estimated.

Cosmetics

On the basis of notifications submitted under the Cosmetic Regulations, DBP is likely to be present in cosmetics in Canada, specifically nail polishes (personal communication from the CPSD, Health Canada, to ESRAB, Health Canada, July 2015; unreferenced).

Koniecki et al. (2011) reported DBP in cosmetics purchased in Canada, including hair sprays, mousses, nail polishes, skin cleansers and baby shampoos. Internationally, DBP has been detected in various types of cosmetics and personal care products (Guo and Kannan 2013; Guo et al. 2013; Liang et al. 2013). A summary of recent studies measuring concentrations of DBP in cosmetics and personal care products reported in North America is outlined in Table 9-5.

Table 9-5. Concentrations of DBP in cosmetics and personal care products
Detection frequency and product typesa Concentration
(μg/g)
Reference
(country)
8% of 85 fragrance, haircare and deodorant products ND-36 Koniecki et al. 2011
(Canada)
7% of 69 nail polish, lotion and skin cleanser products ND-24304 Koniecki et al. 2011
(Canada)
2% of 98 baby products ND-1.8 Koniecki et al. 2011
(Canada)
17% of 41 rinse-off products ND-0.69 Guo and Kannan 2013
(United States)
39% of 109 leave-on products (including nail polishes) ND-27400 Guo and Kannan 2013
(United States)
20% of 20 baby products ND-0.22 Guo and Kannan 2013
(United States)

a. Limits of detection: Koniecki et al. 2011 (0.1 μg/g), Guo and Kannan 2013 (0.01 μg/g)

Given that the North American studies report that detection frequencies are low (2% to 8% in Canada, 17 to 39% in US) and the majority of the concentrations in all three studies are in the sub-ppm range, exposure from personal care products and cosmetics are not considered significant. The exception is nail polishes, as both North American studies showed high concentrations of DBP in nail polishes (Koniecki et al. 2011 and Guo and Kannan 2013). In addition, the US CPSC CHAP (2014) indicated that nail polish as a product type would likely contribute to greater than 10% of exposure to DBP, and NICNAS (AGDH 2013a) reported that this type of product was most likely to contain the highest amount of DBP. Therefore, estimates of dermal exposure from nail polishes as a worst-case representative scenario are presented in Table 9-6.

Table 9-6. Estimates of dermal exposure from nail polish use a
Product type Concentration
(μg/g)
Intake
(μg/kg bw/day)
Nail polish Mean: 5280;
Max: 27400b
Mean: 0.16;
Max: 0.83

a. Applied a 10% dermal absorption factor. See Appendix H in Environment Canada, Health Canada 2015b for approach to characterizing dermal absorption to medium-chain phthalates.
b. Guo and Kannan 2013.

Estimates of exposure to DBP from nail polishes were 0.16 and 0.83 μg/kg bw/day for mean and maximum concentrations, respectively.

Other products used by consumers

Globally, DBP may be found in paint products (AGDH 2013a). This use has also been reported in Canada (Environment Canada 2014). In addition, DBP may have applications in the production of articles that may come in contact with skin (ECHA 2012; AGDH 2013a; Environment Canada 2014). DBP was reported in 23 of 35 samples (less than 3.0-120 mg/kg) of children's clothing (Brigden et al. 2013). DBP is also found as a non-medicinal ingredient in 1 topical natural health product in Canada (LNHPD modified 2014). The US CPSC CHAP (2014) did not evaluate dermal exposure to items containing DBP.

Exposure to DBP from these uses is considered to be captured by the available Canadian biomonitoring data, therefore estimates of exposure were not calculated.

DEHP
Biomonitoring

Several metabolites of DEHPFootnote 8 were monitored in the CHMS Cycle 1 (2007-2009) and Cycle 2 (2009-2011) with greater than 99% detection in all samples (Health Canada 2011b, 2013). Additionally, these metabolites were measured as part of the First Nations Biomonitoring Initiative (n=492 for on-reserve and Crown-land populations aged 20 years and over). Urine concentrations for MEHP, MEHHP and MEOHP (geometric means) were all reported to be statistically lower in FNBI samples than those reported in CHMS (AFN 2013).

Metabolites of DEHP were also monitored by Health Canada in three cohort surveys: P4 study (n = 31 women and their infants, 542 individual spot samples, women provided multiple urine samples over two visits, monitored 5 metabolites of DEHPFootnote 9), MIREC study (n = 1742 pregnant women, spot urine samples, monitored 3 metabolites of DEHPFootnote 10) and the MIREC-CD Plus study (n = 197 children aged 2 to 3 years, 1 spot sample per individual, monitored 5 metabolites of DEHPFootnote 11). All three surveys reported high detection frequencies (greater than 90%) of all metabolites monitored (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013, 2014, unreferenced; Arbuckle et al. 2014).

Using the CHMS, P4, MIREC and MIREC-CD Plus datasets, reverse dosimetry intake estimates were generated as previously described. Biomonitoring intakes are presented in Table 9-7 below (see Appendix C for further information on the methodology).

Table 9-7. Biomonitoring daily intakes (μg/kg bw/day) for DEHP a
Age group Study Male/female n Arithmetic mean 50th 75th 95th
1-4 months P4 males and females 48 0.81 0.42 0.69 1.4
2-3 years MIREC-CD Plus males and females 198 3.4 2.6 4.0 8.9
3-5 years CHMS males and females 509 5.3 4 6 12b
6-11 years CHMS males 256 4.3 3 4.8 12
6-11 years CHMS females 250 3.2 2.3 3.2 8.1b
12-19 years CHMS males 249 2.1 1.4 2.4 5.6b
12-19 years CHMS females 250 2 1.2 1.8 4
18+ years MIREC females (pregnant) 1713 3.4 1.6 2.7 8.4
19+ years P4 females (pregnant) 31c 2.2 1.6 2.3 5.2
20-49 years CHMS males 284 1.6 1 1.8 4.9b
20-49 years CHMS females 274 1.4 1.0 1.5 2.7
50-79 years CHMS males 205 1.3 0.88 1.3 -
50-79 years CHMS females 209 1.2 0.94 1.3 2.6

- = No data
a. Data for males and females from: P4 and MIREC pregnant women, P4 infants, MIREC-CD plus children (preliminary results), and CHMS (Cycle 2)
b. Use data with caution.
c. n = 31 pregnant women, 542 individual spot samples, women provided multiple urine samples over two visits.

The highest exposed group based on biomonitoring data (all sources, CHMS) is children aged 3 to 5 years, with median and 95th percentile intakes of 4.0 and 12 μg/kg bw/day, respectively. For older populations (19 years and over), the highest exposed group (all sources, MIREC) is pregnant females aged 18 years and over, with median and 95th percentile intakes of 1.6 and 8.4 μg/kg bw/day, respectively. However, the higher exposure estimates for pregnant women relative to non-pregnant women could be a result of multiple factors (different populations sampled, different sample sizes, etc.) as the results were obtained from different studies. Therefore a correlation between pregnancy and higher DEHP levels cannot be made.

Environmental media and food

The subpopulation with the highest exposure to DEHP from environmental media and food consisted of children (aged 0.5 to 4 years), with total daily intakes of 10.45 and 27.57 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively (Appendix D, Table D-3a).

Indoor air and dust

One study measured DEHP in indoor air in Canadian homes and reported air concentrations of 88 ng/m3 (median) and a range of 8.8 to 2100 ng/m3 (LOD was not reported; Zhu et al. 2007). Recently, DEHP has also been measured in indoor air in homes in the United States (detected in 100% of 20 samples from homes in Albany, NY, method quantification limit = 0.10 ng/m3, median: 17.4 ng/m3, maximum: 132 ng/m3; Tran and Kannan 2015) and internationally (Blanchard et al. 2014; Lin et al. 2014; Takeuchi et al. 2014). The Canadian indoor air survey concentrations (median: 88 ng/m3 and maximum: 2100 ng/m3; Zhu et al. 2007) were used to estimate the general population daily intake of DEHP from indoor air in Canada (Appendix D, Table D-3a).

DEHP was surveyed in the CHDS and was detected in 100% of homes (range: 35.9-3836 μg/g, median: 462 μg/g, 95thpercentile: 1880 μg/g) (Kubwabo et al. 2013). Internationally, DEHP has also been reported in house dust (Dodson et al. 2015; Luongo and Östman 2015).

DEHP has applications as a plasticizer in the manufacturing of automobiles and automobile parts (AGDH 2010; ECHA 2012; Environment Canada 2014a). For the general population, indirect exposure (e.g., off-gassing) is considered a relevant source, but no data on this exposure source has been identified, which is currently an uncertainty in the draft screening assessment.

The Canadian survey (Kubwabo et al. 2013) was identified as the key study for exposure characterization, and median (462 μg/g) and 95th percentile (1880 μg/g) concentrations were used to estimate the Canadian general population daily intake of DBP from dust in Canada (Appendix D, Table D-3a).

Food, infant formula and breast milk

In Canada, DEHP presence in food was measured in samples from the 2013 Canadian TDS (Cao et al. 2015). DEHP was detected in 111 of 159 food composite samples (average MDL=39.0 ng/g) with a range of concentrations from 14.4 to 714 ng/g (Cao et al. 2015). Milk, fruits and vegetables are the major sources of intake for all population groups over 1 year of age. Infant formula is the major source of intake for infants under the age of 1 year.

Regarding fruits and vegetables, detectable levels of DEHP (i.e., concentrations above the LOD) were found in most of the composite samples (Cao et al. 2015). The source of DEHP in fruit and vegetable samples is unclear, but could be linked to their packaging. For example, some fruits or vegetables may have been packaged in plastic. Furthermore, certain composite samples included canned or jarred products where coatings and gaskets on jar lids may be potential sources of DEHP in foods. Environmental contamination and agricultural practices are other possible contributors to DEHP residues in fruits and vegetables. Nonetheless, the DEHP results for composite fruit and vegetable samples from the 2013 TDS are considered unusual and unlikely to represent the frequency and magnitude of DEHP concentrations that are typically found in these types of foods (personal communication from Food Directorate, Health Canada, to ESRAB, Health Canada, April 2016; unreferenced). Health Canada's Food Directorate also analyzed composite fruit and vegetable samples from the 2014 TDS in order to further understand the distribution of DEHP concentrations in fruits and vegetables sold in CanadaFootnote12. Relative to the 2013 TDS data, the 2014 TDS results detected DEHP approximately twofold less frequently in fruit and vegetable composite samples (68% in 2013 versus 30% in 2014). In addition, the mean concentrations in positive samples from 2014 are more than 5 times lower than those from the 2013 TDS samples (61 μg/kg in the 2014 versus 331 μg/kg in 2013). Therefore, the dietary exposures estimates presented herein, which are based only on the 2013 TDS results, are expected to overestimate actual dietary exposure to DEHP (personal communication from Food Directorate, Health Canada, to ESRAB, Health Canada, April 2016; unreferenced).

DEHP presence in food was also monitored as part of the CFIA 2013-2014 and 2014-2015 FSAP surveys (personal communication from Food Directorate, Health Canada, to ESRAB, Health Canada, April 2014; unreferenced). DEHP was detected in 8% of 1518 (LOD: 0.05 μg/g) packaged and processed food samples. DEHP concentrations ranged from 0.27 to 76.2 μg/g, with a mean of the samples of 2.09 μg/g.

Breast milk was monitored as part of the MIREC study. DEHP was detected in 23 samples (n=305, mean of positives=0.0977 μg/g, range= less than MDL-0.236 μg/g, average MDL=0.0668 μg/g; personal communication from Food Directorate, Health Canada, to ESRAB, Health Canada, October 2014; unreferenced). Internationally, DEHP has been detected in breast milk in Sweden (Högberg et al. 2008) and Germany (Fromme et al. 2011), but was not detected in Italy (Guerranti et al. 2013). However, DEHP readily metabolizes to multiple metabolites in the human body (Koch and Calafat 2009) and would therefore not be expected to be found in breast milk in high quantities.

Three metabolites of DEHPFootnote 13 were measured in breast milk as part of the P4 study (n = 31 women, 56 breast milk samples collected from study participants). MEHP, MEHHP and MEOHP were detected in 100, 16 and 8% of breast milk samples, respectively (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013; unreferenced). LODs were reported to be 0.1, 0.019 and 0.017 μg/L, respectively. The median and maximum reported levels of MEHP were 1.26 and 17.05 μg/L, respectively (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013; unreferenced). Internationally, MEHP has also been detected in breast milk in Europe (Mortensen et al. 2005; Högberg et al. 2008; Fromme et al. 2011; Guerranti et al. 2013).

Recently, an analysis of 23 infant formula samples in the P4 study showed 90% detection of MEHP (LOD: 0.1 μg/L) and 8% detection of MEHHP (LOD: 0.019 μg/L). Median and maximum values of MEHP were reported as 0.469 and 2.154 μg/L, respectively. MEOHP was not detected (LOD: 0.017 μg/L) (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013; unreferenced). However, measurement of these metabolites in infant formula was affected by high field blank levels (possible contamination) and therefore was not used to quantify intakes. The CFIA surveillance data did detect DEHP in 1 sample of infant cereal (n=7). It was not detected in any samples of infant formula (n=32) or infant foods (n=20) (personal communication from Food Directorate, Health Canada, to ESRAB, Health Canada, April 2014; unreferenced). Internationally, DEHP has been detected in infant formulas in Italy, the UK and Europe (Sørensen 2006; Bradley et al. 2013a; Cirillo et al. 2015).

Using TDS data, probabilistic estimates of dietary intakes for DEHP were derived for the Canadian general population. The results are outlined in Appendix D, Table D-3b and the methodology for estimating probabilistic intakes is provided in Appendix E. Median (1.26 μg/L) and maximum (17.05 μg/L) values of MEHP (metabolite of DEHP) in breast milk measured in the P4 study were used for exposure characterization (Appendix D, Table D-3a). Note that exposure estimates based on food intake are significantly higher than those based on biomonitoring results (see Table 9-7 and Table D-3b). This is expected to be due to detection of frequent and elevated concentrations of DEHP in highly consumed foods from the 2013 TDS, such as certain fruits and vegetables, as previously discussed.

Ambient air, drinking water and soil

No Canadian data were identified for DEHP in ambient air. Internationally, DEHP has been reported in outdoor air (Li and Wang 2015). Rudel et al. (2010) measured outdoor air outside homes in the United States (detected in 14% of 43 samples [MRL= 40 ng/m3], median: not reported, maximum: 230 ng/m3). Rudel et al. (2010) was identified as the relevant study for exposure characterization (sampling from North America), and half the MRL (40 ng/m3) and maximum (230 ng/m3) concentrations were used to estimate potential exposures to DEHP via ambient air (Appendix D, Table D-3a).

No Canadian data were identified for DEHP in drinking water. In Canada, Cao et al. (2008) surveyed phthalates in bottled carbonated and non-carbonated water and detected and quantified DEHP in all 11 samples (range: 0.052 to 0.338 μg/L). Internationally, DEHP was also detected and quantified in bottled water (Jeddi et al. 2015), drinking water (Liu et al. 2015), and surface waters (Li et al. 2015; Net et al. 2015; Selvaraj et al. 2015). In the absence of Canadian and North American data on levels of DEHP in tap water, mean (0.102μg/L) and maximum (0.338 μg/L) DEHP concentrations in bottled non-carbonated water were used to estimate the general population daily intake from drinking water (Cao et al. 2008) in Canada (Appendix D, Table D-3a).

Khosravi and Price (2015) reported various phthalates in agricultural soils in both control and biosolid amended soils collected in Nova Scotia, Canada. DEHP was reported at 0.06 ng/g (control soil) and 4.3 ng/g (biosolid amended) (Khosravi and Price 2015). DEHP has also been detected in soils internationally (Cheng et al. 2015; Tran et al. 2015; Wang et al. 2015a). The reported concentration of DEHP in control agricultural soils (0.06 ng/g; Khosravi and Price 2015) was used to estimate potential exposures to DEHP via soil in Canada (Appendix D, Table D-3a).

Products used by consumers

Globally, DEHP may be present in a wide variety of manufactured items, including childcare articles, children's toys, DIY products, electronics, textiles, and gloves (HPD 1993- ; Stringer et al. 2000; AGDH 2010; US CPSC 2010; ECHA 2012; Chao et al. 2013; Korfali et al. 2013). In Canada, DEHP has also been reported as a plasticizer used in various types of manufactured items (NRC 2012; See table 2-2; Environment Canada 2014).

DEHP may be found in paint products (AGDH 2010). The use has also been reported in Canada (Environment Canada 2014).

Globally, DEHP may have applications in the production of articles that may come in contact with skin (HPD 1993- ; AGDH 2010; ECHA 2012; NRC 2012; Chao et al. 2013; US CPSC CHAP 2014). The US CPSC CHAP (2014) reported that dermal exposure to items containing DEHP (i.e., play pen, change pad) would contribute to greater than 10% of exposure to DEHP for infants and children.

Exposure to DEHP from these uses is considered to be captured by the available Canadian biomonitoring data, therefore estimates of exposure were not calculated.

Toys and childcare articles

Numerous studies have examined the concentrations of DEHP in childcare articles and toys (Stringer et al. 2000; Biedermann-Brem et al. 2008; Johnson et al. 2011; Korfali et al. 2013). A summary of DEHP reported in toys and childcare articles available in Canada is presented in Table 9-8.

Table 9-8. Percent content of DEHP in various toys and childcare articles available in Canada
Detection frequency Percent content Reference
1 of 117 samples 6.9% Health Canada 2014
(Canada)
6 of 62 samples greater than 0.1-37% Health Canada 2012
(Canada)
15 of 38 samples greater than 0.1-54% Health Canada 2009
(Canada)
33 of 72 samples greater than 0.1-22.8% Health Canada 2007
(Canada)

Currently, Canada (like the United States and the European Union) has regulations in place limiting the amount of certain phthalates (including DEHP) in toys and childcare articles (Phthalates Regulations under the Canada Consumer Product Safety Act [Canada 2016]). The EU's RAPEX database also shows low reporting of toys tested in violation of the regulation for DEHP (RAPEX 2015), and the US CPSC recently stated that less than 10% of exposures to DEHP for infants and children would result from mouthing of toys and childcare articles (US CPSC CHAP 2014).

Given the absence of DEHP in toys and childcare articles reported in Canadian monitoring studies and the low frequency and low percent content of DEHP reported in international monitoring studies, exposure was not quantified and oral intakes from mouthing toys and/or childcare articles were not estimated.

Cosmetics

On the basis of notifications submitted under the Cosmetic Regulations, DEHP is not expected to be present in cosmetics in Canada (personal communication from the CPSD, Health Canada, to ESRAB, Health Canada, July 2015; unreferenced). However, DEHP has been detected in various types of cosmetics and personal care products in Canada (Koniecki et al. 2011) and internationally (Guo and Kannan 2013; Guo et al. 2013). Liang et al. (2013) did not detect DEHP in cosmetics in China. DEHP has also been reported in some fragrance products (0-46ppm; Not too Pretty 2015). This presence may be due to potential migration from packaging. A summary of recent studies measuring concentrations of DEHP in cosmetics and personal care products reported in North America is outlined in Table 9-9.

Table 9-9. Concentrations of DEHP in cosmetics and personal care products
Detection frequency and product typesa Concentration
(μg/g)
Reference
(country)
5% of 85 fragrance, haircare and deodorant products ND - 521 Koniecki et al. 2011
(Canada)
4% of 69 nail polish, lotion and skin cleanser products ND - 1045 Koniecki et al. 2011
(Canada)
1% of 98 baby products ND - 15 Koniecki et al. 2011
(Canada)
76% of 41 rinse-off products ND - 6.15 Guo and Kannan 2013
(United States)
66% of 109 leave-on products ND - 135 Guo and Kannan 2013
(United States)
40% of 20 baby products ND - 8.22 Guo and Kannan 2013
(United States)

a. Limits of detection: Koniecki et al. 2011 (0.1 μg/g), Guo and Kannan 2013 (0.01 μg/g)

DEHP is on the List of Prohibited and Restricted Cosmetic Ingredients in Canada (Health Canada 2011a) and was not reported as used in Canada (July 2015 email from the CPSD to ESRAB, Health Canada). Additionally, the Canadian study reported that detection frequencies are low (1% to 5%) and a majority of the concentrations in all studies are in the sub-ppm range. Consequently, exposure to DEHP from personal care products and cosmetics is not considered to be significant. Exposure estimates from this source were not generated.

Adult toys

No Canadian use of DEHP in adult toys was reported under the CEPA section 71 industry survey (Environment Canada 2014). Global use patterns suggest that there is potential for use of DEHP in adult toys. DEHP was reported in 8 of 15 adult toys sampled in the EU, with concentrations ranging from 0.73 to 702 mg/g (Bavarian State Ministry of the Environment and Public Health 2012). The Danish Environmental Protection Agency published a report assessing exposure to DEHP from these products and derived intakes of 1.7 and 47 μg/kg bw/day, for normal-case and worst-case exposures, respectively (Nilsson et al. 2006).

Medical devices

Globally, DEHP is reported to be used in medical applications (ECHA 2012). Medical devices may therefore be a potential source of DEHP exposure. Currently, Canada has guidance in place for DEHP in medical devices. However, given the lack of data and uncertainty in quantifying estimates of exposure from medical devices, this exposure source was not quantified and is currently an uncertainty in this draft screening assessment.

DnHP

No data was identified for DnHP in air or water; therefore, intakes from these sources were not estimated.

DnHP was surveyed in the CHDS and was detected in 98% of homes (range: ND-264 μg/g, median: 3.8 μg/g, 95thpercentile: 62 μg/g) (Kubwabo et al. 2013). DnHP has also been reported in house dust in the United States (Dodson et al. 2015). The Canadian survey (Kubwabo et al. 2013) was identified as the key study for exposure characterization, and median (3.8 μg/g) and 95th percentile (62 μg/g) concentrations were used to estimate the Canadian general population daily intake of DnHP from dust (Appendix D, Table D-4a).

No Canadian or North American data were identified for DnHP in soil. DnHP has been detected in 100% of urban (n=17; median=0.018 μg/g, maximum=0.019 μg/g), suburban (n=28; median=0.016 μg/g, maximum=1.227 μg/g) and rural (n=37; median=0.016 μg/g, maximum=0.1 μg/g) soils in China (Hongjun et al. 2014). Given the limited data available on DnHP in soil, exposure intakes from this source were not estimated.

No Canadian data for DnHP in foods were found. One US study looked at many phthalates, including DnHP, in a variety of food items (Schecter et al. 2013). Data from this US study were employed to generate dietary exposure estimates using a probabilistic approach (Appendix D, Table D-4b).

The subpopulation with the highest exposure from dust and food consisted of infants (0 to 6 months old), with total daily intakes of 0.019 and 0.31 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively (Appendix D, Table D-4a).

Given the absence of reporting to the CEPA section 71 industry survey (Environment Canada 2014), and the lack of information as to DnHP presence in product databases, general population exposure to DnHP from products used by consumers is expected to be negligible.

DIOP

No data was identified for DIOP in air, water, soil or food; therefore, intakes from these sources were not estimated.

DIOP was surveyed in the CHDS and was detected in 87% of homes (range: ND-1165 μg/g, median: 6.6 μg/g, 95thpercentile: 28.6 μg/g) (personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, August 2014; unreferenced). These data were identified as the key study for exposure characterization, and median (6.6 μg/g) and 95thpercentile (28.6 μg/g) concentrations were used to estimate the Canadian general population daily intake of DIOP from dust (Appendix D, Table D-5).

In Canada, DIOP has also been reported as a plasticizer used in various types of manufactured items (See Table 5-1; Environment Canada 2014). However, given the low volume of DIOP reported in Canada (See table 4-2), exposure estimates from products used by consumers were not estimated.

The subpopulation with the highest exposure from dust consisted of infants (0 to 6 months old), with total daily intakes of 0.033 and 0.14 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively (Appendix D, Table D-5).

9.1.3 Long-chain phthalates

For detailed information on long-chain phthalates included in this section, please refer to the SOS report on long-chain phthalates esters (Environment Canada, Health Canada 2015d).

DIDP

Exposure estimates were calculated using biomonitoring results as well as data on DIDP occurrence in dust, food and plastic items. They are presented in the LCP SOS report (Environment Canada, Health Canada 2015d) and summarized below. Recently, DIDP has been reported internationally in soils (Tran et al. 2015), indoor air (Takeuchi et al. 2014), settled dust (Luongo and Östman 2015), and tap water (Yang et al. 2014). However, these reported values did not change the exposure estimates presented previously (Environment Canada, Health Canada 2015d).

The highest exposed group according to biomonitoring data (all sources, NHANES) is male children aged 6 to 11 years, with median and 95th percentile intakes of 1.4 and 4.4 μg/kg bw/day respectively. For older populations, the highest exposed group (all sources, NHANES) is adults over 20 years of age, with median and 95th percentile intakes of 0.76 and 4.4 μg/kg bw/day, respectively, for males, and 0.65 and 4.9 μg/kg bw/day, respectively, for females.

The subpopulation with the highest exposure from dust and food consisted of infants (6 months to 4 years old), with total daily intakes of 0.514 and 2.87 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively. For populations aged 12 years and over, the highest exposed group is adolescents (aged 12 to 19 years), with total daily intakes of 0.075 and 0.726 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively.

Estimated daily intakes for infants from dermal exposure to DIDP from plastic items, based on lower-end and upper-bounding exposure scenarios, were 0.27 and 2.16 μg/kg bw/day, respectively. For adults exposed to plastic items (females aged 20 years and over), estimated daily intakes, based on lower-end and upper-bounding exposure scenarios, were 0.27 and 0.85 μg/kg bw/day, respectively.

DUP

Exposure estimates were calculated using data on DUP occurrence in dust and plastic items. Since the publication of the LCP SOS report, DUP has been analyzed in foods included in the 2013 Canadian Total Diet Study and was not quantified above the method detection limit (average MDL= 6.97 ng/g; Cao et al. 2015). Exposure estimates for DUP are presented in the LCP SOS report (Environment Canada, Health Canada 2015d) and summarized below.

The subpopulation with the highest exposure from dust consisted of infants (0 to 6 months old), with total daily intakes of 0.0198 and 0.349 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively. For populations aged 12 years and over, the highest exposed group is adolescents (aged 12 to 19 years), with total daily intakes of less than 0.001 and 0.004 μg/kg bw/day estimated on the basis of central tendency and upper-bounding concentrations, respectively.

Estimated daily intakes for infants from dermal exposure to DUP from plastic items, derived on the basis of lower-end and upper-bounding exposure scenarios, were 2.7 and 21.6 μg/kg bw/day, respectively. For adults exposed to plastic items (females aged 20 years and over), estimated daily intakes, based on lower-end and upper-bounding exposure scenarios, were 2.7 and 8.5 μg/kg/day, respectively.

9.2 Health effects assessment

As described in detail in the SOS reports (Environment Canada, Health Canada 2015a,b,c,d), critical effects of phthalates (i.e., medium-chain phthalates) consist of adverse effects on the development of the male reproductive system following exposure. The spectrum of effects on male reproductive development has been described as the "rat phthalate syndrome" (RPS) and although primarily studied in rats, it has also been demonstrated in other species. The effects include alterations of feminization parameters (decreased anogenital distance [AGD] in pups and nipple retention [NR] in juveniles), reproductive tract malformations (cryptorchidism [CRY], hypospadias [HYP], testicular pathological changes [TP]) and effects on fertility (sperm counts, motility and quality at adulthood). Similar to the SOS reports, the hazard assessment is structured to present information at three different life stages (gestational exposure [GD0-21], (pre)pubertal-pubertal [PND1-55] and adult [PND55+], with particular focus on the male gender because of the varying degrees of sensitivity at different life stages. When there was limited information or absence of data for a particular phthalate at a specific life stage or exposure period (as it is the case for DMP, DIBP, DMCHP, DBzP, B84P, B79P, CHIBP, BCHP, BIOP, and DUP), read-across was applied based on health effects of the closest analogue(s) (Health Canada 2015). In addition, potential effects of phthalates on humans were evaluated following the same approach as previously described in the SOS reports (Environment Canada, Health Canada 2015a,b,c,d).

9.2.1 Short-chain phthalates

DMP

The SCP SOS (Environment Canada, Health Canada 2015a) summarizes the health effects literature related to DMP and its analogue DEP. No new animal hazard data were identified after the literature cut-off date of the SCP SOS.

Tables 9-10, 9-11 and 9-12 provide critical endpoints and corresponding no-observed-effects level (NOEL) and/or lowest-observed effects level (LOEL) values for DMP, as previously described in SCP SOS (Environment Canada, Health Canada 2015a) that will be used for risk characterization.

Table 9-10. Summary results of reproductive and/or developmental effects studies based on oral exposure to DMP
Life stage Species Effect
(mg/kg-bw/day)
LOEL
(mg/kg-bw/day)
NOEL
(mg/kg-bw/day)
Reference
In utero Rat No developmental effects observed. No effects on RPS parameters
(GD14-PND3)
NA 750 Gray et al. (2000);
Furr et al. (2014)
(Pre)pubertal Rat
(7 d)
A significant decrease in serum and testicular testosterone*, dihydrotestosterone concentrations and ↑ absolute, relative liver weight 1862
(LOAEL)
N/A Oishi and Hiraga (1980a)
Adult Rat
DEP
(F0, 8 wk)
↓ serum testosterone, transient increases in abnormal and tailless sperm at the mid dose, (not high), ↓ in absolute epididymis, adrenal weights 1016 197 Fujii et al. (2005)

*These results are of uncertain adversity since no other effects in testes were noted (no changes in testes weights, no inhibition of spermatogenesis, and no testicular atrophy).
N/A = Not applicable

Table 9-11. Summary results of reproductive and/or developmental effects studies based on dermal exposure to DMP
Life stage Species Effect
(mg/kg-bw/day)
LOEL
(mg/kg-bw/day)
NOEL
(mg/kg-bw/day)
Reference
In utero Rat
(GD1-20)
LOEL (Maternal) = 2380
(slight ↓ body weight), no effects on pups.
N/A 2380 Hansen and Meyer (1989)
(Pre)pubertal Rat
DEP
(4 wk)
Systemic LOEL = 1332
↑ relative kidney and liver (2278) weights, no testicular pathology observed
N/A 2278 NTP (1995)
Adult Rat
DEP
(2 yr)
Systemic LOAEL =743
↓ absolute brain weight, no testicular pathology observed
N/A 743 NTP (1995)

N/A = Not applicable

Table 9-12. Summary table of critical systemic effects after dermal exposure to DMP
Endpoint Species Effect LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Subchronic Rat
(90 days)
Changes in nervous system and renal function in males 1250 200 Timofieyskaya (1976)
Chronic Rats
DEP
(2 years)
Small but significant decrease in absolute brain weight in males 743 230 NTP (1995)
Chronic Mice
DEP
(2 years)
Decrease in mean body weight in females 834 415 NTP (1995)
Evidence in humans

A literature update was also conducted to identify any recent human data for short-chain phthalates. The updated search was focused on reproductive and developmental endpoints in males because these endpoints were identified as critical health endpoints in the SOS reports. Studies were further evaluated and scored for quality using a consistent evaluation metric (Downs and Black 1998). For the health outcomes evaluated (i.e., sex hormone levels, anogenital distance, birth measures, male infant genitalia, preterm birth and gestational age, altered male puberty, gynecomastia, changes in sem*n parameters, pregnancy loss, and altered time to pregnancy), there was inadequate evidence or no evidence of associations between DMP and the reported outcomes (Table 9-13). More detail is provided in Health Canada (2016a) available upon request.

Table 9-13. Summary of levels of evidence of associations between short-chain phthalates and health outcomes
Outcome DMP
(MMP)
Sex hormone levels IA (7)
Anogenital distance NA (1)
Birth measures NA (4)
Male infant genitalia NA (1)
Preterm birth and gestational age NA (2)
Altered male puberty IA (3)
Gynecomastia NM
Changes in sem*n parameters IA (5)
Pregnancy loss NA (2)
Altered time to pregnancy IA (1)

() = Number of studies
NM = Not measured in studies of quartile 2 and above (See Health Canada [2016a] for more details).
NA = No evidence of association.
IA = Inadequate evidence of an association.
LA = Limited evidence of an association.
MMP = Monomethyl phthalate.

9.2.2 Medium-chain phthalates and additional phthalates

9.2.2.1 Medium-chain phthalates
DIBP

The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects literature related to DIBP. No new animal hazard studies were identified for this phthalate after the literature cut-off date of the MCP SOS.

Table 9-14 provides critical endpoints and corresponding no-observed-adverse-effects level (NOAEL) and/or lowest-observed-adverse-effects level (LOAEL) values for DIBP, as previously described in MCP SOS (Environment Canada, Health Canada 2015b), that will be used for risk characterization. DIBP has low systemic toxicity as described in MCP SOS (Environment Canada, Health Canada 2015b); hence, no systemic critical effects were established.

Table 9-14. Summary of critical effect levels for reproductive and/or developmental effects after oral exposure to DIBP
Life stage during which exposure occurred Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
In utero (GD12-21) Rat ↓ AGD, ↓ NR, effects on fertility and other RPS effects;
decreased testicular testosterone production
250 125 Saillenfait et al. 2008;
Furr et al. 2014
(Pre)pubertal Rat ↑ apoptotic spermatogenic cells;
↓ testes weight and vimentin filament disorganization in Sertoli cells
500 300 Zhu et al. 2010
Adult Rat
(DBP)
Testicular pathology, effects on sperm count and motility, and decreased ROW 500 250 Srivastava et al. 1990a;
Zhou et al. 2011c
DCHP

The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects literature related to DCHP. A literature update identified new developmental and genotoxicity studies, which are summarized in Tables 9-15 and 9-16. In Li et al. (2016), rats were exposed to 0, 10, 100 or 500 mg/kg-bw/day DCHP from GD12-21. The body weight of male pups was significantly reduced in treated animals. DCHP dose-dependently increased abnormal fetal cell aggregation and decreased fetal Leydig cell size, cytoplasmic size and nuclear size at all doses tested. Decreases in AGD and increased incidences of MNGs were noted at 100 mg/kg-bw/day and above. In another study, the authors also examined the effects of in utero DCHP exposure on offspring development (Ahbab and Barlas 2015). Rats were exposed to 0, 20, 100 or 500 mg/kg-bw/day DCHP from GD6-19. Maternal effects were not observed. An increase in resorption was observed in all treated groups. A decrease in AGD and testicular pathological changes were observed starting from 20 mg/kg-bw/day. The LOAELs of theses studies are lower than the lowest NOAEL previously identified in the MCP SOS for DCHP (Environment Canada, Health Canada 2015b). As described in the MCP SOS for DCHP, a decrease in AGD was observed at higher doses in both F1 (at 511 mg/kg-bw/day) and F2 (from 107 mg/kg-bw/day) generations in a two-generation study with a NOAEL of 21 mg/kg-bw/day (Hoshino et al. 2005), suggesting transgenerational effects induced by DCHP. The dose level of 16 to 21 mg/kg-bw/day from this two-generation study was also identified as the NOAEL for developmental effects by the Australian National Industrial Chemicals Notification and Assessment Scheme (NICNAS) (AGDH 2008) and the US CPSC CHAP (2014). Different stains of rats were used by Ahbab and Barlas (2015) than by Hoshino et al. (2005) and Li et al. (2016) (Wistar vs Sprague Dawley, respectively). The lowest LOAELs identified for developmental toxicity were 10 to 20 mg/kg-bw/day estimated on the basis of testicular pathological changes from 10 mg/kg-bw/day, reduced AGD, and increased resorption from 20 mg/kg-bw/day (Ahbab and Barlas 2015; Li et al. 2016).

Table 9-15. Summary of new DCHP developmental studies identified since the publication of the MCP SOS report. Effects from gestational exposure to DCHP in male offspring (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
CAS RN
Testosterone levelsa
(T, S)
Feminization parametersb Reproductive tract malformations and/or fertilityc Other developmental parametersd Maternal effects
DCHP
Sprague Dawley rats;
0, 10, 100, 500;
gavage;
GD12-21 (Li et al. 2016)
100 (T)
NM (S)
100 (AGD)
NM (NR)
NM (PPS)
NM (CRY)
NM (HYP)
10e (TP ↑ abnormal fetal leydig cell aggregation, ↓ fetal Leydig cell size, cytoplasmic size and nuclear size, ↑ MNGs at 100)
NM (FER)
10e (↓ BW)
NM (ROW)
NE (FV)
NM (EMB)
NM (ESV)
NE
DCHP
Wistar albino rats;
0, 20, 100, 500;
gavage;
GD6-19 (Ahbab and Barlas 2015)
NM (T)
100 (S)
20e, f (AGD)
NM (NR)
NM (PPS)
NM (CRY)
NM (HYP)
20e (TP- ↑ atrophic and small seminiferous tubules, ↓ no. of germ cells in tubules, cells detach from tubule wall, ↑ Leydig cell clusters)
NM (FER)
20e (↑ BW @ 20, 100)
NM (ROW)
NE (FV)
20e, f (EMB-resorption)
NM (ESV)
NE

NM = not measured.
NE = no effect observed at the dose range tested. When NE is presented alone in the first four columns of effects, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Testosterone levels measured (can include quantity/production) at varying days post-birth. T=Testicular testosterone; S=Serum testosterone.
b. Feminization parameters can include anogenital distance (AGD), nipple retention (NR), preputial separation (PPS).
c. Malformations include: cryptorchidism (CRY), hypospadias (HYP), testicular pathology (TP), including multinucleated gonocytes (MNG), and/or reproductive effects such as fertility (FER) in offspring (sperm number, motility) or reproductive success at adult stage after in utero exposure.
d. Other developmental effects include: decreases in overall fetal body weight (BW), decreases in reproductive organ weight (ROW), fetal viability (FV), embryotoxicity (EMB), or on the incidence of external, skeletal or visceral malformations (ESV).
e. Lowest dose measured in the study.
f. Absolute AGD, AGD relative to BW and AGD relative to cube food of BW at birth were reported in the study and reached statistically significant different from control animals at 20 mg/kg-bw/day (Ahbab and Barlas 2015).

Table 9-16. Summary of new literature of DCHP identified since the publication of the MCP SOS report
Endpoint Study details Results Reference
Genotoxicity (in vivo) TUNEL assay
Species/strain: female Wistar albino rat
Route: oral (gavage)
Dose and duration: 0, 20, 100 or 500 mg/kg-bw/day during GD6-19 (10/dose)
Testicular samples were collected from different life stages (prepubertal PD20, pubertal PD21, adult PD90) of the pups (n=8-10/group).
Dose-response increase in apoptotic cells was observed in prepubertal and pubertal life stages, but not in the adult life stage Ahbab et al. 2014
Genotoxicity (in vivo) Comet assay
Species/strain: female Wistar albino rat
Route: oral (gavage)
Dose and duration: 0, 20, 100 or 500 mg/kg-bw/day during GD6-19 (10/dose)
Testicular and blood samples were collected at the adult life stage (PD90) of the pups (n=7-10/group)
Increase in DNA breakage was only observed in the low doses with no clear dose-response pattern Ahbab et al. 2014

As described in the MCP SOS for DCHP (Environment Canada, Health Canada 2015b), only one limited repeat-dose oral exposure study with DCHP was identified in sexually immature animals. That study was determined to be limited and was not used to characterize risk of DCHP for this life stage. Therefore, results from the two-generation study (Hoshino et al. 2005) described in the MCP SOS were used, particularly observations reported in the F1 males exposed in uteroto DCHP through lacation and via diet from PND21 for at least 10 weeks until necropsy. The NOAEL for the reproductive toxicity of DCHP at the prepubertal-pubertal life stage was 18 mg/kg-bw/day in F1 males, based on observed decreases in spermatid head counts and testicular atrophy starting from 90 mg/kg-bw/day. Reduced body weight gain and reduced food consumption were observed in F1 males at 90 mg/kg-bw/day and above. It is recognized that this effect level is conservative as animals had been exposed during early development before the prepubertal-pubertal life stage.

Tables 9-17 and 9-18 provide the critical endpoints and corresponding NOAEL and/or LOAEL values for DCHP that will be used for risk characterization.

Table 9-17. Summary of critical effect levels for reproductive and/or developmental effects after oral exposure to DCHP
Life stage Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
In utero Rat Testicular pathological changes, decreased AGD and increased resorption 10-20 N/A Ahbab and Barlas 2015;
Li et al. 2016
(Pre)pubertal Rat Decrease in spermatid head counts, testicular atrophy, reduced body weight gain and reduced food consumption in F1 males 90 18 Hoshino et al. 2005
Adult Rat Slight focal seminiferous tubule atrophy in 1 male at highest dose, with decreased body weight gain 402
(6000 ppm LOEL)
80
(1200 ppm)
Hoshino et al. 2005

N/A = Not applicable.

Table 9-18. Summary table of critical systemic effects after oral exposure to DCHP
Endpoint Species Effect LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Subchronic Rat
(90 days)
Increase in liver weight
(females)
75 25 de Ryke and Willems 1977
DMCHP

As described in the MCP SOS (Environment Canada, Health Canada 2015b), DCHP was used as an analogue as no studies examining the potential health effects of DMCHP were identified for any species or gender. No new literature on DMCHP was identified after the literature cut-off date of the MCP SOS. See the above section for a summary of the critical health effects used for this phthalate.

DBzP

The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects literature related to DBzP and its analogue MBzP. No new literature was identified after the literature cut-off date of the MCP SOS. Table 9-19 provides the critical endpoints and corresponding NOAEL and/or LOAEL values for DBzP, as previously described in MCP SOS (Environment Canada, Health Canada 2015b), that will be used for risk characterization.

Table 9-19. Summary of critical effect levels after oral exposure to DBzP using MBzP as closest analogue
Life stage Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
In utero Rat
(MBzP)
↓ AGD and ↑ cryptorchidism 250 167a
(systemic toxicity LOAEL based on ↓ food consumption, ↓ BW)
Ema et al. 2003
(Pre)pubertal/adult Rat
(MBzP)
↓ sperm count (20%) 250
(LOEL)
N/A Kwack et al. 2009

N/A = Not applicable.
a. Maternal toxicity at this dose, but considered not a factor in selection of adverse effects in male offspring.

B84P

The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects literature related to B84P and its analogues. No new literature was identified for B84P after the literature cut-off date of the MCP SOS. Tables 9-20 and 9-21 provide the critical endpoints and corresponding NOAEL and/or LOAEL values for B84P, as previously described in MCP SOS (Environment Canada, Health Canada 2015b) that will be used for risk characterization.

Table 9-20. Summary of critical effect levels for reproductive and/or developmental effects after oral exposure to B84P
Life stage Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
In utero Rat
(BBP)
Decreased body weight (F1/F2 males and females) and ↓ AGD at birth in F2 malesa;
↓ testicular testosterone
100 50 Aso et al. 2005;
Nagao et al. 2000;
Tyl et al. 2004;
Furr et al. 2014
(Pre)pubertal Rat
(MBzP)
↓ sperm counts and sperm motility 250
(LOEL)
N/A Kwack et al. 2009
(Pre)pubertal Rat
(BBP)
↓ sperm counts and sperm motility 500
(LOEL)
N/A Kwack et al. 2009
Adult Rat
(BBP)
reduced absolute epididymal weight, hyperplasia of the Leydig cells in the testes and decreased spermatozoa in the lumina of the epididymis 400 200 Aso et al. 2005

N/A = Not applicable.
a. A statistically significant increase of AGD in F1 and decreased pup weight on PND0 in F2 female offspring at 100 mg/kg-bw/day and above was also reported.

Table 9-21. Summary table of critical systemic effects after oral exposure to B84P
Endpoint Species Effect LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Subchronic Rat
(3 months)
(BBP)
Histopathological changes in the pancreas, gross pathological alterations in the liver and a significant increase in relative kidney weight in male rats 381 151 Hammond et al. 1987
DIHepP

The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects literature related to DIHepP. No new literature was identified after the literature cut-off date of the MCP SOS. Tables 9-22 and 9-23 provide the critical endpoints and corresponding NO(A)EL and/or LO(A)EL values for DIHepP, as previously described in MCP SOS (Environment Canada, Health Canada 2015b), that will be used for risk characterization.

Table 9-22. Summary of critical effect levels for reproductive and/or developmental effects after oral exposure to DIHepP
Life stage Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
In utero Rat Significant reduction in AGD in male F2 pups 309-750 64-168 McKee et al. 2006
(Pre)pubertal Rat Significant reduction in AGD; delayed preputial separation, nipple retention, hypospadias and cryptorchidism in F1 pups 419-764 227-416 McKee et al. 2006
Adult Rat No adverse effects up to the highest dose level tested N/A 404-623 McKee et al. 2006

N/A = Not applicable.

Table 9-23. Summary table of critical systemic effects after oral exposure to DIHepP
Endpoint Species Effect LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Subchronic Rat Increased liver and kidney weights with histopathological findings in F0 222-716 50-162 McKee et al. 2006
Chronic Rat Increased liver and kidney weights associated with centriobular hypertrophy in males and females in F1 227-750 50-168 McKee et al. 2006
B79P

The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects literature related to B79P and its analogues. New key studies were identified after the literature cut-off date of the MCP SOS, which are described in the following sections and will be used for risk characterization.

Early development: in utero exposure to B79P

Three oral rat studies were found focusing on effects of B79P during development, including one study that was previously described in the MCP SOS. These studies all examined the effects of B79P when administered during gestation in pregnant rats during the fetal masculinization programming window GD15-17. Summaries of the studies are described in Table 9-24 below.

In an industry submitted developmental study, Tyl et al. (2012) exposed Sprague-Dawley rats to B79P via the diet from GD6-20, at target concentrations of 0, 250, 750 or 3750 ppm (reported intakes: 0, 19, 58 and 288 mg/kg-bw/day). There were no effects on maternal body weight, although food consumption was significantly increased in a dose-related manner in the two highest dose groups for the exposure. At necropsy, both absolute and relative liver weights were significantly increased at the highest dose; no adverse effects were observed at histological examination. There was no effect on the mean number of live fetuses per litter or fetal body weight. There were no differences among groups in the incidence (fetal or litter) of either external malformations/variations or of skeletal malformations. Similarly, there were no differences in the total number of malformations (external, visceral, skeletal). The authors concluded that there had been maternal toxicity at the highest dose only, with increased absolute and relative liver weights and "no adverse findings in the prenatal offspring at any dose." Parameters specific to RPS (AGD, NR, PPS, etc) were not reported.

In a second industry submitted study, Tyl et al. (2013) also conducted a two-generation reproductive toxicity study with Sprague-Dawley rats exposed to B79P at 0, 250, 750, 2500 and 5000 ppm (estimated doses: 0, 19, 56, 188 and 375 for males and 0, 17, 50, 167 and 333 mg/kg-bw/day for females). F0 and F1 male and female rats were exposed for 10 weeks premating and 2 weeks mating. Female rats were also exposed throughout gestation (approximately 3 weeks) and lactation (3 weeks). There were no differences in maternal or offspring lactational parameters or in F1 litter parameters on PND 0-4. No effects were observed in AGD across groups. No differences were observed in acquisition of PPS in F1 males. There was no evidence of effects on mating, production of litters, or litter size. Other parameters specific to RPS, specifically NR, HYP and CRY, were not reported.

In the F2 generation, there were no differences in total litter size, live litter size, live birth ratio, survival ratio, AGD, pup body weight or gross pathology. Relative liver weight was increased in both F2 males. The authors concluded that B79P was "clearly not a reproductive toxicant in either sex" at exposures up to 333-375 mg/kg-bw/day. However, a NOAEL of 50-56 mg/kg-bw/day was assigned for systemic toxicity on the basis of reduced body weights and altered organ weights and organ/body weight ratios, affecting the liver and kidney, which were free from gross and histopathologic changes (Tyl et al. 2013).

Table 9-24. Effects from gestational exposure to B79P in male offspring (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
CAS RN
Testosterone levelsa
(T, S)
Feminization parametersb Reproductive tract malformations and/or fertilityc Other developmental parametersd Maternal effects
B79P
SD Rats: 0, 750, 3750, 7500 ppm;
est. 0, 50, 250, 500 (diet) GD6 - PND 21
Cited in REACH Dossier;
ECHA 2013a
NM 50e (at PND1 both sexes) (AGD)
250 (at PND21 for males)
500f (NR)
NR (PPS)
50e, f (CRY)
250f (HYP, epispadias)
500 (TP)
NM (FER)
50e (BW @ lactation)
NM (ROW)
NE (FV)
NE (EMB)
NR (ESV)
250 (↑ liver and kidney weights, ↓ body wt)
SD Rats: 0, 250, 750, 2500, 5000 ppm;
F0 Female intake during gestation est. 0, 17, 50, 167, 333 (diet) 2-gen
Tyl et al. (2013)
NM NE (AGD
@PND0)
NEg (NR)
NE (PPS)
NM (CRY)
NM (HYP)
NE (TP-adult)
NE (FER)
167 (BW 11.6%@PND7-14)
NE (ROW)
NE (FV)
NE (EMB)
NE (ESV)
177 (OW - ↑ rel liver wt)
336 (↓ abs brain, spleen, thymus weight)
17 (↓ body wt gain at this dose only during gestation)
167 (↑ relative liver and kidney weights, no histopathological findings)
333 (small ↑ bilateral cortical vacuolation of adrenal glands)
SD Rats: 0, 250, 750, 2500, 5000 ppm;
F1 Female intake during gestation est. 0, 17, 50, 167, 333 (diet) 2-gen
Tyl et al. (2013)
NM NE (AGD)
NM (NR)
NM (PPS)
NM (CRY)
NM (HYP)
NE (TP)
NE (FER)
NE (BW)
NE (ROW)
NE (FV)
NE (EMB)
NE (ESV)
333 (OW - ↑ relative liver weight)
NE (BW)
167 (↑ relative and absolute liver weights, no histopathological findings)
333 (small ↑ bilateral cortical vacuolation of adrenal glands in 2 females)
B79P
SD Rats: 0, 250, 750, 3750 ppm;
est. 0, 16.7, 50, 250 (diet) GD6-20
Tyl et al. (2012)
NM NM (AGD)
NM (NR)
NM (PPS)
NM (CRY)
NM (HYP)
NM (TP)
NM (FER)
NE (BW)
NM (ROW)
NE (FV)
NE (EMB)
NE (ESV)
50 (↑ food cons)
250 (↑ absolute and relative liver weights (7.6%, no histopathological findings)

NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone in the first 4 columns of effects, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Testosterone levels measured (can include quantity/production) at varying days post-birth. T=Testicular testosterone; S=Serum testosterone.
b. Feminization parameters can include anogenital distance (AGD), nipple retention (NR), preputial separation (PPS).
c. Malformations include: cryptorchidism (CRY), hypospadias (HYP), testicular pathology (TP), and/or reproductive effects such as fertility (FER) in offspring (sperm number, motility) or reproductive success at adult stage after in utero exposure.
d. Other developmental effects include: decreases in overall fetal body weight at PND 1 (BW), decreases in reproductive organ weight (ROW), fetal viability (FV), embryotoxicity (EMB), or on the incidence of external, skeletal or visceral malformations (ESV). Other organ weights (OW).
e. Lowest dose tested in the study.
f. CYP, HYP and NR effects were observed on PND21, but not on PND75 where these animals were exposed until PND21 (ECHA 2013b).
g. NR result was based the executive summary of the study saying there were no effects in both F1 and F2. No additional info on NR was found in the rest of the report (Tyl et al. 2013).

Overall, the highest NOAEL identified for developmental toxicity of B79P after gestational exposure was 50 mg/kg-bw/day based on decreased AGD in male pups and increased incidence of epispadias on PND21 observed at the next dose level of 250 mg/kg-bw/day (REACH dossier; ECHA 2013b). A decrease in AGD on PND1 and a slight increase in the incidence of CRY were also observed at 50 mg/kg-bw/day and evidence of retained nipples was observed at 500 mg/kg-bw/day on PND21. It should be noted that there is uncertainty regarding this critical NOAEL as no AGD effect was observed on PND 1 in both F1 and F2 in a recent two-generation reproductive toxicity study up to 333 mg/kg-bw/day (Tyl et al. 2013). Both studies (REACH dossier; ECHA 2013b; Tyl et al. 2013) were conducted using the same strain of rat. No information was provided by Tyl et al. (2013) to explain this discrepancy. The NOAEL for maternal systemic toxicity was considered to be 50 mg/kg-bw/day based on decrease body weight gain during gestation and liver and kidney effects at 250 mg/kg-bw/day (Tyl et al. 2013).

Exposure to B79P at prepubertal-pubertal life stage

In the MCP SOS, the critical effect level for the reproductive toxicity of B79P was based on read across from closest analogues as no studies were identified for this life stage. Since the publication of the MCP SOS report, an unpublished two-generation reproductive toxicity study was submitted by industry as described above; therefore, results from the two-generation study were considered, particularly observations reported in the F1 males that were exposed in utero, through lactation to B79P and via diet until necropsy at PND126 (Tyl et al. 2013).

There were transient reductions on male body weight gain at 2500 ppm (188 mg/kg-bw/day) on PND56-63, but no effects on terminal body weights or body weight gains at the end of treatment (PND126). No effects on reproductive organ weights were observed at any dose, nor were there any histopathological findings reported. No effect on sperm counts or motility; however, a small, statistically significant increase in abnormal sperm was noted at all dose groups (19 mg/kg-bw/day and above), but there were no effects on reproductive performance of F1 males with the exception of a significantly longer precoital interval (p less than 0.05) at the highest dose (375 mg/kg-bw/day). Changes in abnormal sperm were 1.45, 1.72, 1.96, 1.91, and 2.31% in 0, 19, 56, 188, and 375 mg/kg-bw/day, respectively, which reflect a 16% to 37% increase compared to control. The authors noted these percent abnormal sperm values were within the laboratory's historical control values for this rat strain and supplier. In addition, these values, although showing a dose-responsive nature, are also within typical ranges for untreated males of this strain in other laboratories (Kato et al. 2006; Matsumoto et al. 2008).

Systemic effects were limited to statistically significant increases in relative liver weights at 56 mg/kg-bw/day and 375 mg/kg-bw/day, but not at 188 mg/kg-bw/day with no histopathological findings. There were no changes in absolute or relative adrenal glands, but observations of small, increased incidences of bilateral cortical vacuolation at the highest dose tested (375 mg/kg-bw/day; 9 males).

There were no differences in offspring lactational parameters. There were no differences in F1 litter parameters on PND 0-4. No differences were observed in AGD across groups. No differences were observed in acquisition of PPS in F1 males.

Overall, the NOEAL for the reproductive toxicity of B79P at the prepubertal-pubertal life stage was 375 mg/kg-bw/day since no adverse health effects were observed at the highest dose level tested in F1 males in the two-generation study by Tyl et al. (2013). The lowest LOEL for systemic effects was 375 mg/kg-bw/day based on small, increased incidences of bilateral cortical vacuolation in adrenal glands in the same above cohort.

Oral exposure to B79P at the mature male adult stage

In the MCP SOS, the critical effect level for the reproductive toxicity of B79P was based on read-across from closest analogues as no studies were identified for this life stage. One industry-submitted two-generation study was available after the literature cut-off date of the MCP SOS.

Reproductive effects from a 14-week exposure of adult F0 males to B79P in diet from the two-generation study described in the section above showed no adverse effects in fertility or reproductive organs (Tyl et al. 2013). A small, statistically significant increase in abnormal sperm was noted at 56 mg/kg-bw/day and above, but there were no effects on sperm count, sperm motility or reproductive performance of F0 males (Table 9-25). Changes in abnormal sperm were 1.35, 1.59, 1.73, 1.95, and 2.16% in 0, 19, 56, 188, and 375 mg/kg, respectively, which reflect a 22% to 38% increase compared to control. The authors noted these percent abnormal sperm values were within the laboratory’s historical control values for this rat strain and supplier. In addition, these values, although showing a dose-responsive nature, are within typical ranges for untreated males of this strain in other laboratories (Kato et al. 2006; Matsumoto et al. 2008). One male in the highest dose group (375 mg/kg-bw/day) developed a misshapen bilateral testes. There was no evidence of effects on mating, production of litters, or litter size.

Table 9-25. Reproductive effects from exposure to B79P in adult males (mg/kg-bw/day)
Strain and species; dose (mg/kg-bw/day);
route;
duration (reference) CAS RN
Life stage at the start of dosing
(age)
Hormone levelsa
(T, S, LH)
Fertilityb Reproductive tract pathologyc Other effectsd
SD male Rats: 0, 250, 750, 2500, 5000 ppm;
F0 est. 0, 19, 56, 188, 375 (diet) 14 wks
Tyl et al. (2013)
6-7 weeks NM 56 (↓ % abnormal sperm, but no effect on reproduction) 375 (1 male with bilateral misshapen testes)
no histopathological findings
  1. NE (BW)
  2. NE (ROW)
  3. 188 (ST- ↑ absolute and relative liver weight, relative kidney weight, no histopathological findings)

NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Hormone level can include quantity/production of testicular testosterone (T),serum testosterone (S), or leutinizing hormone (LH).
b. Fertility parameters include: sperm number, motility, morphology, viability, stages of spermatogenesis, or reproductive success at adult stage after in utero exposure.
c. Reproductive tract pathology includes: any observations based on histopathological examination of the testes such as, but not limited to, multinucleated gonocytes (MNGs), necrosis, hyperplasia, clustering of small Leydig cells, vacuolisation of Sertoli cells, decrease in Leydig cell number, an increase in Leydig cell size, focal dysgenesis, and/or seminiferous tubule atrophy.
d. Other effects include: Decreased overall body weight (BW), decreased reproductive organ weight (ROW) and systemic toxicity (ST).

Overall, the NOAEL for reproductive toxicity identified for B79P at the adult life stage was 375 mg/kg-bw/day since no adverse health effects were observed at the highest dose level tested in F0 adult male rats in a two generation diet study after 14-week exposure to B79P (Tyl et al. 2013).

Reproductive Developmental Effects: Oral exposure to B79P in females

In a previously described two-generation study by Tyl et al. (2013), F0 females exhibited a significant increase in absolute and relative liver weights at both 167 and 333 mg/kg-bw/day. There were no differences in maternal or offspring lactational parameters and no differences in F1 litter parameters on PND 0-4. For F1 females, there were no differences in body weight at acquisition of vagin*l patency; reproductive performance, fertility and fecundity were equivalent across all groups. There was no evidence of effects upon evidence of mating, production of litters of litter size for the production of F2 offspring. At weaning and termination of the F2 pups on PND 21, F1 females had equivalent vagin*l cytology parameters and body weights. Absolute and relative liver weights were increased at 167 and 333 mg/kg-bw/day.

Table 9-26 provides a summary of critical effects for the reproductive and/or developmental effects of B79P which will be used for risk characterization.

Table 9-26. Summary of critical effects levels for reproductive and/or developmental effects after oral exposure to B79P
Life stage during which exposure occurred Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
In utero
(GD6-21)
Rat ↓ AGD, ↑ epispadis 250 50 ECHA 2013a
(Pre)pubertal Rat No adverse effects at the highest dose level tested N/A 375 Tyl et al. 2013
Adult Rat No adverse effects at the highest dose level tested N/A 375 Tyl et al. 2013

N/A = Not applicable.

For systemic effects, critical effect levels were based on its analogue DINP, as previously described in MCP SOS (Environment Canada, Health Canada 2015b) (Table 9-27).

Table 9-27. Summary table of critical systemic effects after oral exposure to B79P
Endpoint Species Effect LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Chronic Rat
(DINP)
Increase in absolute and relative liver and kidney weight with increase in histopathological changes in both organs in both male and females 152-184 15-18 Lington et al. 1997
DINP

The DINP SOS (Environment Canada, Health Canada 2015c) summarizes the health effects literature related to DINP. Since the publication of the DINP SOS report, a recent developmental study was identified (Table 9-28). Li et al. (2015b) examined the effects of in utero DINP exposure on offspring development. Rats were exposed to 0, 10, 100, 500 or 1000 mg/kg-bw/day DINP during GD12-21. No maternal toxicity was observed except in one dam at the highest dose level, which died on GD21.5.

Testicular pathological changes in Leydig cells (clusters) were first observed at 10 mg/kg-bw/day and MNGs were observed starting from 100 mg/kg-bw/day. The LOEL of 10 mg/kg-bw/day of this study is lower than the lowest NOAEL (50 mg/kg-bw/day) and LOAEL (159-395 mg/kg-bw/day) (Waterman et al. 2000; Clewell 2011 cited in ECHA 2013; Clewell et al. 2013) identified in the DINP SOS (Environment Canada, Health Canada 2015c). The NOAEL of 50 mg/kg-bw/day was also established by multiple agencies on the basis of the same studies (AGDH 2012; ECHA 2013b; US CPSC CHAP 2014). It should be noted that similar MNGs and Leydig cell clusters effects were observed in Clewell (2011a, 2013) at 250 mg/kg-bw/day and above. At higher dose levels, other RPS-related parameters include decreased serum testosterone levels, decreased AGD, NR, effects in sperm and other histopathological effects in the testes were observed.

Overall, the lowest LOEL identified for developmental toxicity was 10 mg/kg-bw/day based on testicular pathological changes (i.e., MNGs and Leydig cell clusters) observed at the next dose level of 100 mg/kg-bw/day (Li et al. 2015b).

Table 9-28. Effects from gestational exposure to DINP in male offspring (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
CAS RN
Testosterone levelsa
(T, S)
Feminization parametersb Reproductive tract malformations and/or fertilityc Other developmental parametersd Maternal effects
DINP
SD rats; 0, 10, 100, 500, 1000;
gavage;
GD12-21 (Li et al. 2015b)
1000 (T)
NM (S)
NEf (AGD)
(NR)
(PPS)
NM (CRY)
NM (HYP)
100f (TP-focal testis dysgenesis, MNGs, ↓ clusters of Leydig cells @ 10)
NM (FER)
10e, NDR (BW)
(ROW)
NE (FV)
NM (EMB)
NM (ESV)
NE (except 1/6 dam @ 1000 died at GD21.5 with all fetuses alive obtained through surgery)

NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Testosterone levels measured (can include quantity/production) at varying days post-birth. T = Testicular testosterone; S = Serum testosterone.
b. Feminization parameters can include anogenital distance (AGD), nipple retention (NR), preputial separation (PPS).
c. Malformations include: cryptorchidism (CRY), hypospadias (HYP), testicular pathology (TP), including multinucleated gonocytes (MNG), and/or reproductive effects such as fertility (FER) in offspring (sperm number, motility) or reproductive success at adult stage after in utero exposure.
d.Other developmental effects include: decreases in overall fetal body weight (BW), decreases in reproductive organ weight (ROW), fetal viability (FV), embryotoxicity (EMB), or on the incidence of external, skeletal or visceral malformations (ESV).
e. Lowest dose measured in the study.
f. Absolute AGD and AGD adjusted with cube root of pup weight were reported. Frequency of large cell clusters (i.e., greater than 16 cells per cluster) was significantly increased from control (0.16%) to 6%, 11%, 14% and 14% in the 10, 100, 500 and 1000 mg/ kg bw/day groups, respectively (Li et al. 2015b).

Table 9-29 provides critical endpoints and corresponding NOAEL and/or LOAEL values for DINP which will be used for risk characterization for reproductive and developmental endpoint.

Table 9-29. Summary of critical effect levels for reproductive and/or developmental effects after oral exposure to DINP
Life stage Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
In utero Rat ↑ MNGs, ↑ Leydig cell clusters/aggreation 100 10
(LOEL)
Li et al. 2015b
(Pre)pubertal Rat
(castrated)
Decreased absolute seminal vesicle and LABC weights 500
(LOEL)
100
(NOEL)
Lee and Koo 2007
Adult Rat Reduced relative and absolute reproductive organ weights 742
(LOEL)
276 Moore 1998

For other health effects, two neurotoxicity studies were identified (Table 9-30). Table 9-31 provide critical endpoints and corresponding NOAEL and/or LOAEL value for DINP which will be used for risk characterization for systemic effects.

Table 9-30. Summary of new studies identified since the publication of DINP SOS report
Endpoint Study details Results Reference
Neurotoxicity Species/strain: male kummi*ng mouse
Route: oral
Dose and duration: 0, 1.5, 15 or 150 mg/kg-bw/day
(n = 10/dose)
LOAEL = 150 mg/kg bw/ day based on reduced body weight gain, impaired cognitive ability in Morris water maze (MWM) test, histological alterations in pyramidal cells in the hippocampus Peng 2015
Neurotoxicity Species/strain: male kummi*ng mouse
Route: oral
Dose and duration: 0, 0.2, 2, 200 or 200 mg/kg-bw/day
(n = 7/dose)
LOAEL = 20 mg/kg bw/ day based on histopathological alterations in pyramidal cells in the hippocampus, oxidative stress and inflammation in the brain. Impaired cognitive ability was observed in MWM test and anxiety in Open field test at the 200 mg/kg-bw/day. Ma et al. 2015
Table 9-31. Summary table of critical non-cancer effects after oral exposure to DINP
Endpoint Species Effect LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Chronic Rat
(2 years)
Increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs 152-184 15-18 Lington et al. 1997
CHIBP, BCHP and BIOP

As described in the MCP SOS (Environment Canada, Health Canada 2015b), no studies examining the potential reproductive/developmental health effects of CHIBP, BCHP and BIOP were identified. An updated literature search conducted after the literature cut-off date of the MCP SOS did not identify any new studies. As described in the exposure assessment, general population exposure to CHIBP, BCHP and BIOP from environmental media and products used by consumers is expected to be negligible. Therefore, risk to human health for this substance is not expected.

Evidence in humans

A literature update was conducted to identify any recent human data for medium-chain phthalates since the publication of the MCP SOS report. The search was focused on reproductive and developmental endpoints in males as these endpoints were identified as critical health endpoints in the SOS reports. Studies were further evaluated and scored for quality using a consistent evaluation metric (Downs and Black 1998). For the health outcomes evaluated (i.e., sex hormone levels, AGD, birth measures, male infant genitalia, preterm birth and gestational age, altered male puberty, gynecomastia, changes in sem*n parameters, pregnancy loss, and altered time to pregnancy), there was limited evidence of association between DINP and sex hormone levels (Main et al. 2006; Joensen et al. 2012; Mouritsen et al. 2013; Meeker and Ferguson 2014; Specht et al. 2014; Axelsson et al. 2015a; Jensen et al. 2015; Lenters et al. 2015a; Pan et al. 2015) or sem*n parameters (Joensen et al. 2012; Jurewicz et al. 2013; Specht et al. 2014; Axelsson et al. 2015a; Lenters et al. 2015a; Pan et al. 2015). There was inadequate evidence or no evidence of association for the other medium-chain phthalates in the Phthalate Substance Grouping and the outcomes (Table 9-32). More detail is provided in Health Canada (2016a).

Table 9-32. Summary of levels of evidence of associations between medium-chain phthalates in the Phthalate Substance Grouping and health outcomes
Outcome DIBP
(MIBP)
DCHP
(MCHP)
DINP
(MINP/MCOP, etc.)
Sex hormone levels IA (4) NM LA (9)
Anogenital distance NA (2) NM IA (2)
Birth measures NA (3) NM NA (2)
Male infant genitalia NM NM NA (2)
Preterm birth and gestational age NA (2) NM NA (1)
Altered male puberty IA (1) NM NM
Gynecomastia NA (1) NM NA (1)
Changes in sem*n parameters IA (3) NM LA (6)
Pregnancy loss IA (3) NM NA (1)
Altered time to pregnancy NA (1) NA (1) NA (1)

() = Number of studies.
NM = Not measured in studies of quartile 2 and above (See Health Canada [2016a] for more details).
NA = No evidence of association.
IA = Inadequate evidence of an association.
LA = Limited evidence of an association.
MIBP = mono-iso-butyl phthalate.
MINP = monoisononyl phthalate.

9.2.2.2 Additional phthalates

As previously discussed in the cumulative risk assessment (CRA) approach document (Environment Canada, Health Canada 2015e), additional phthalates identified to have common adverse effects within the RPS as a result of alterations will be considered to be included into the CRA. After both the exposure (see section 6.1) and hazard filters were considered, BBP, DBP, DEHP, DnHP and DIOP were included in the CRA. Reproductive and development effects in males (i.e., RPS-related parameters) of these five additional phthalates were evaluated and summarized in the following sections. The RPS-related parameters were previously described in detail in the MCP SOS (Environment Canada, Health Canada 2015b).

BBP

The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the reproductive and development effects of BBP in section 9.2.8.1, as BBP was identified as an analogue to B84P. Since the literature cut-off date of the MCP SOS, a recent developmental study was identified. Ahmad et al. (2014) examined the effects of in utero BBP exposure on offspring development (Table 9-33). Significant decrease in fetal body weight was observed at 4 to 20 mg/kg-bw/day during development from PND1 to 75. However, the percentage decrease in body weight was 2.5% to 5% compared to untreated control animals. Maternal effects were observed at 4 to 20 mg/kg-bw/day with decrease in maternal body weight and increase in gestation length. RPS effects were observed at 100 mg/kg-bw/day with decrease in serum testosterone level, sperm effects and decrease in reproductive organ weights.

Table 9-33. Effects from gestational exposure to BBP in male offspring (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Testosterone levelsa
(T, S)
Feminization parametersb Reproductive tract malformations and/or fertilityc Other developmental parametersd Maternal effects
Albino rats; 0 (untreated), 4, 20, 100; oral;
GD14-parturition (up to GD23.5) (Ahmad et al. 2014f)
NM (T)
100 (S)
NE (AGD)
NM (NR)
NM (PPS)
NE (CRY)
NM (HYP)
NM (TP)
100 (FER - ↓ sperm count, ↓ sperm motility, ↑ abnormal sperm)
4e, f (BW on PND1 and PND21, 20 on PND75)
100(ROW-others- epididymis, prostate)
NE (FV)
NM (EMB)
NM (ESV)
20f (BW)
4e (↑ gestation length)

NM = Not measured.
NE = No effect observed at the dose range tested.
a. Testosterone levels measured (can include quantity/production) at varying days post-birth. T = Testicular testosterone; S = Serum testosterone.
b. Feminization parameters can include anogenital distance (AGD), nipple retention (NR), preputial separation (PPS).
c. Malformations include: cryptorchidism (CRY), hypospadias (HYP), testicular pathology (TP), including multinucleated gonocytes (MNG), and/or reproductive effects such as fertility (FER) in offspring (sperm number, motility) or reproductive success at adult stage after in utero exposure.
d. Other developmental effects include: decreases in overall fetal body weight (BW), decreases in reproductive organ weight (ROW), fetal viability (FV), embryotoxicity (EMB), or on the incidence of external, skeletal or visceral malformations (ESV).
e. Lowest dose measured in the study.
f. Results were based on statistical analysis compared with untreated control. The study also presented data of vehicle control and positive control. Maternal effects were only presented in graphical format in the study (Ahmad et al. 2014).

Table 9-34 summarizes the critical endpoints and corresponding NOAEL and/or LOAEL values for BBP which will be used for risk characterization.

Table 9-34. Summary of critical effect levels for reproductive and or developmental effects after oral exposure to BBP
Life stage Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
In utero Rat Decreased body weight (F1/F2 males and females) and ↓ AGD at birth in F2 malesa;
↓ testicular testosterone
100 50 Aso et al. 2005;
Nagao et al. 2000;
Furr et al. 2014
(Pre)pubertal Rat ↓ sperm counts (20%) and sperm motility 500
(LOEL)
N/A Kwack et al. 2009
Adult Rat reduced absolute epididymal weight, hyperplasia of the Leydig cells in the testes and decreased spermatozoa in the lumina of the epididymis 400 200 Aso et al. 2005;
NTP 1997

N/A = Not applicable.
a. A statistically significant increase of AGD in F1 and decreased pup weight on PND0 in F2 female offspring at 100 mg/kg-bw/day and above was also reported.

DBP
Early development: in utero exposure

The European Commission classified DBP as a Category 1B reproductive toxicant (presumed human reproductive toxicant) as defined in the EU regulation on classification, labelling and packaging of substances and mixtures (ECHA 2015a).

A literature search identified many studies examining the toxicity of DBP during gestation in rodents. For the purposes of characterizing effects during early male development, only studies in which effects of DBP were observed at doses below 250 mg/kg-bw/day in rats and 500 mg/kg-bw/day in mice following in utero exposure during the masculinization programming window are reported here. Summaries of the studies are described below and in Table 9-35.

Overall, adverse effects in the parameters used to describe RPS in male rat offspring after in utero exposure to DBP include decreased testicular testosterone levels, delayed PPS, decreased AGD, NR, CRY, gross and testicular malformations, and effects on fertility.

A search of the available literature revealed nine studies examining the effects of gestational exposure of DBP during the masculinization programming window in mice. The majority did not examine the parameters used to describe RPS, with only two reporting any testicular pathology and one examining testicular testosterone levels (Marsman 1995; Gaido et al. 2007, Saffarini et al. 2012).

The reproductive-developmental effects of DBP do not appear to be restricted to rodents, as one study was identified in rabbits exhibiting testicular pathological changes and sperm effects (Higuchi et al. 2003). A full summary of the health effects associated with gestational exposure to DBP is summarized in Health Canada 2016b. Table 9-35 presents a list of key studies with effects from gestational exposure to DBP in male offspring.

Table 9-35. Key studies with effects from gestational exposure to DBP in male offspring (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route; duration (reference)
Testosterone levelsa
(T, S)
Feminization parametersb Reproductive tract malformations and/or fertilityc Other developmental parametersd Maternal effects
Albino rats;
0 (untreated), 2, 10, 50 DBP;
oral;
GD14-parturition (up to GD23.5) (Ahmad et al. 2014e)
NM (T)
NE (S)
NE (AGD)
NM (NR)
NM (PPS)
NE (CRY)
NM (HYP)
NM (TP)
50 (FER- ↓ sperm count, ↓ sperm motility, ↑ abnormal sperm)
10 (BW-sig. but less than 5%)
50 (ROW)
NE (FV)
NM (EMB)
NM (ESV)
2e (↓ BW, ↑ gestational length)
SD rats;
0, 0.1, 1, 10, 50, 100 DBP;
gavage;
GD12-19 (Lehmann et al. 2004)
50 (T)
NM (S)
NM NM NR (BW)
NM (ROW)
NM (FV)
NM (EMB)
NM (ESV)
NR
CD(SD)IGS rats;
0, 20, 200, 2000, 10000 ppm, est. 0, 1.5-3, 14-29, 148-291, 712-1372 based on AGDH (2008) DBP;
diet;
GD15-21 (Lee et al. 2004)
NM 712-1372 (AGD)
712-1372f (NR)
NE (PPS)
NM (CRY)
NM (HYP)
148-291 (TP TP- loss of germ cell development, aggregated foci of Leydig cells)
1.5-29 (FER-sig. ↑ incidence; 148-291 sig.↑ severity of reduced spermatocyte development)
712-1372NS (BW)
712-1372 (ROW)
NE (FV)
NM (EMB)
NM (ESV)
LOEL= 712-1372 (↓ BW)
SD rats;
0.1, 1, 10, 30, 50, 100, 500 DBP;
gavage;
GD12-21 (Boekelheide et al. 2009)
NM NM NM (CRY)
NM (HYP)
50, 30 (TP)
A. Disorganized seminif. tubules,
B. Cell number within testes
NM (FER)
NM NM
SD rats;
0, 0.1, 0.5, 1%, est. F1 [Task 4] 0, 52, 256, 509 DBP;
diet;
"second generation fertility" (F2 pup results) (Wine et al. 1997)
NM NM NM 256 (BW)
509 (ROW)
509 (FV)
509 (EMB)
NM (ESV)
LOEL= 509 (↓ body weight at wk 17)
Dutch-belted rabbits;
0, 400 DBP;
gavage;
GD15-29 (Higuchi et al. 2003)
NM (T)
400g (↓ S at 6 weeks only)
NM 400g, NS (CRY- 1/17 rabbits)
400g , NS (HYP- 1/17 rabbits)
400g (TP- germinal epithelium loss, semini. epithelium with desquamation or focal vacuolation)
400f (FER- sperm concentration and morphology)
NE (BW)
400 (ROW- at 12 weeks)
NM (FV)
NM (EMB)
NM (ESV)
NM

NE = No effect observed at the dose range tested. When NE is presented alone in the first 4 columns of effects, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
NM = Not measured.
NR = Results not recorded (but measurement was stated in the methods and materials).
NS = Not statistically significant.
a. Testosterone levels measured (can include quantity/production) at varying days post-birth. T=Testicular testosterone; S=Serum testosterone.
b. Feminization parameters can include anogenital distance (AGD), nipple retention (NR), preputial separation (PPS).
c. Malformations include: cryptorchidism (CRY), hypospadias (HYP), testicular pathology (TP), and/or reproductive effects such as fertility (FER) in offspring (sperm number, motility) or reproductive success at adult stage after in utero exposure.
d. Other developmental effects include: decreases in overall fetal body weight at PND 1 (BW), decreases in reproductive organ weight (ROW), embryotoxicity (EMB), fetal viability (FV), or on the incidence of external, skeletal or visceral malformations (ESV).
e. Results were based on statistical analysis compared with untreated control. The study also presented data of vehicle control and positive control. Maternal effects were only presented in graphical format in the study (Ahmad et al. 2014).
f. Nipple retention was observed as follows: Number of animals identified (%) were 0, 4, 13, 15, and 100 in controls, 20, 200, 2000, and 10,000 ppm groups (Lee et al. 2004).
g. Lowest dose measured in the study.

Overall, the highest NOAEL identified for developmental toxicity of DBP after gestational exposure was 10 mg/kg-bw/day based on effects on fertility (decreased sperm count and motility and increase levels of abnormal sperm) (Ahmad et al. 2014) and a decrease in male offspring testicular testosterone levels at birth at the next dose of 50 mg/kg-bw/day (Lehmann et al. 2004). This NOAEL is supported by evidence from other studies observing decreases in both the tubular and interstitial cell populations and altered seminiferous tubule morphometry along with other mild effects on spermatocyte development at similar dose levels (Lee et al. 2004; Boekelheide et al. 2009). At lower doses, there was a slight reduction in spermatocyte development (1.5-3 mg/kg-bw/day dose range); however, the severity of this effect at this dose was minimal to slight and only minimal at the second higher dose of 14-19 mg/kg-bw/day (Lee et al. 2004). The lowest LOEL for maternal toxicity of DBP was 509 mg/kg-bw/day based on reductions in body weight gain (less than 10%) in exposed dams (Wine et al. 1997). One study in rabbits indicates that DBP has also effects in other species, but it is unknown if these effects would occur at lower doses (Higuchi et al. 2003).

Exposure at prepubertal/pubertal life stages

A literature search identified many studies examining the reproductive toxicity of DBP in young rodents. For the purposes of brevity, only studies where effects of DBP were observed at doses below 500 mg/kg-bw/day in rodents were evaluated in this screening assessment.

Overall, adverse effects in reproductive parameters observed in (pre)pubertal males after short-termexposure to DBP include changes in serum and testicular testosterone levels, histopathological effects in the testes, and potential effects in fertility (spermatogenesis, sperm motility, and number). The majority of studies available used the rat as a model for evaluation, but only within a relatively high dose range (250 to 1000 mg/kg-bw/day) which limits the interpretation of the potential reproductive toxicity of DBP in this species. One study conducted in mice examined effects at lower dose levels and reported RPS effects at dose levels lower than those in rats (Moody et al. 2013). One study was conducted in rabbits reporting evidence of testicular pathology (Higuchi et al. 2003). A full summary of the health effects associated with propubertal/pubertal exposure to DBP is summarized in Health Canada (2016b). Table 9-36 presents a list of key studies with effects from prepubertal/pubertal exposure to DBP in males.

Table 9-36. Key studies with effects from exposure to DBP in prepubertal/pubertal males (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Life stage at the start of study
(age)
Hormone levelsa
(T, S, LH)
Fertilityb Reproductive tract pathologyc Other effectsd
C57BL/6 mice;
0, 1, 10, 50, 100, 250, 500 DBP;
gavage;
PND 4-14, 10 days (Moody et al. 2013)
Prepubertal
(PND 4)
NM (T)
500 (S)
NM (LH)
500 (FSH)
10 (delayed spermatogenesis) 100 (↑ immature Sertoli cell and disorganization)
1e,f (AGD relative to trunk length; 50NDR abs AGD; 500 AGD relative to BW @ PND14)
NE (BW)
50 (ROW)
NP (ST)
Wistar rats;
0, 250, 500, 1000 DBP;
gavage;
15 days (Srivastava et al. 1990a)
Prepubertal
(PND 35)
NM 250e,g (defective spermatogenesis) 250e,g (shrunken tubules) 500 (BW)
500 (ROW)
NM (ST)
SD rats;
0, 250, 500, 1000, 2000 DBP;
gavage;
30 days (Xiao-feng et al. 2009)
Prepubertal
(PND 35)
NM (T)
500 (S)
1000 (↑ GC)
250 (↓ spermatogenic cells, b/c of ↓ LC no) 250 (↓ LC number);
500 (histopathological changes in the testes)
NP (BW)
500 (ROW)
NM (ST)
C57BL/6 mice;
0, 1, 10, 50, 100, 250, 500 DBP;
gavage;
PND4-8, 3 days (Moody et al. 2013)
Prepubertal
(PND 4)
NM NM 100 (↑ immature Sertoli cell and disorganization) NE (BW)
500 (ROW)
500 (ST- ↑ heart weight)
Dutch-belted rabbits;
0, 400 DBP;
gavage;
15 days (Higuchi et al. 2003)
Prepubertal
(PND 28)
NM (T)
NE (S)
NM (LH)
400e (sperm morphology defects, NE on mating behaviour) 400e (TP- germinal epithelium loss, semini. epithelium with desquamation or focal vacuolation, 1/11 (CRY)
NP (HYP)
NE (BW)
400e (ROW- ↓ sex accessory organ at 12 weeks only)
400e (ST - ↑ thyroid weight)

NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
NDR = No dose relationship.
TPr = Testosterone propionate.
NP = Not reported.
a. Hormone levels can include quantity/production of testicular testosterone (T), serum testosterone (S), leutinizing hormone (LH), glucocorticoid hormone (GC), or follicle stimulating hormone (FSH).
b. Fertility parameters include sperm number, motility, morphology, viability, stages of spermatogenesis, or reproductive success at adult stage after in utero exposure.
c. Reproductive tract pathology includes: testicular pathology (TP): any observations based on histopathological examination of the testes such as, but not limited to, multinucleated gonocytes/germ cells (MNGs), necrosis, hyperplasia, clustering of small Leydig cells (LC), vacuolisation of Sertoli cells, decrease in Leydig cell number, an increase in Leydig cell size, focal dysgenesis, and/or seminiferous tubule atrophy. Anogenital distance (AGD), cryptorchidism (CRY), hypospadias (HYP).
d. Other effects include: Decreased overall body weight (BW), decreased reproductive organ weight (ROW) and systemic toxicity (ST).
e. Lowest dose tested.
f. AGD results were presented in graphical format only (Moody et coll., 2013).
g. Testis of rats after 250 mg/kg DBP treatment showed approximately 5% shrunken tubules with spongy appearance and defective spermatogenesis (Srivastava et coll., 1990a).

Overall, the lowest LOEL identified for reproductive toxicity of DBP at the prepubertal-pubertal life stage was 10-50 mg/kg-bw/day based on delayed spermatogenesis in male mice exposed to DBP for 10 days at this dose and above (Moody et al. 2013). At the next dose level of 50 mg/kg-bw/day, significant reduction in absolute AGD was observed on PND14 mice, but the effect was not significant when AGD was measured relative to body weight. An increase in immature Sertoli cell and disorganization was observed at 100 mg/kg-bw/day. No comparison can be made for rats as there were no studies available at similar dose ranges. The lowest dose tested in rats was 250 mg/kg-bw/day with observations of defective spermatogenesis, shrunken tubules, decrease in spermatogenic cells and Leydig cell numbers (Srivastava et al. 1990a; Xiao-feng et al. 2009). The lowest LOEL for systemic toxicity for mice was 500 mg/kg-bw/day based on increased relative heart weight after 3 days of DBP treatment. This effect was lost by 14 days (Moody et al. 2013). One study in rabbits indicates that DBP also effects other species, but it is unknown if these effects would occur at lower doses (Higuchi et al. 2003).

Exposure at the mature male adult stage

The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects of DBP in sexually mature adult rats (PND55+) as DBP was identified as an analogue to DIBP. Since the literature cut-off date of the MCP SOS (Environment Canada, Health Canada 2015b), new literature was identified (Table 9-37). DBP (0, 200, 400 or 600 mg/kg-bw/day) was administered to rats by oral gavage for 15 days (Aly et al. 2015). At the lowest dose level of 200 mg/kg-bw/day, decrease in serum testosterone level, decrease in sperm count, and decrease in sperm motility were observed. In addition, histopathological examination of the testes indicated degeneration with absence of spermatogenic series in the lumen of some seminiferous tubules starting from 200 mg/kg-bw/day. However, the results from this study are limited as systemic effect and clinical signs of the animals were not measured. In another study, adverse effects (testicular pathological changes and effects on sperm) were observed starting from 500 mg/kg bw/day (Nair 2015).

Table 9-37. Effects from exposure to DBP in adults males (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Life stage at the start of study
(age)
Hormone levelsa
(T, S, LH)
Fertilityb Reproductive tract pathologyc Other effectsd
Wistar rat;
0, 200, 400, 600;
gavage;
15 days (Aly et al. 2015)
~13 weeks NM (T)
200e (↓ S)
NM (LH)
200e (↓ sperm count, ↓ sperm motility) 200e (degeneration with absence of spermatogenic series in the lumen of some seminiferous tubules) NM (BW)
200e (ROW)
NM (ST)
Wistar rats;
0, 500, 1000, 1500 DBP;
oral;
7 days (Nair 2015)
Adult (age not reported, 120-122 g) NM 500e (FER- ↓ sperm density, karyorrhexis in spermatocytes,) 500e (TP- atrophy of Leydig cells) NM (BW)
NM (ST)
NE (ROW)

NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Hormone level can include quantity/production of testicular testosterone (T), serum testosterone (S), or leutinizing hormone (LH).
b. Fertility parameters include: sperm number, motility, morphology, viability, stages of spermatogenesis, or reproductive success after mating.
c. Reproductive tract pathology includes: any observations based on histopathological examination of the testes such as, but not limited to, multinucleated gonocytes (MNGs), necrosis, hyperplasia, clustering of small Leydig cells, vacuolisation of Sertoli cells, decrease in Leydig cell number, an increase in Leydig cell size, focal dysgenesis, and/or seminiferous tubule atrophy.
d. Other effects include: Decreased overall body weight (BW), decreased reproductive organ weight (ROW) and systemic toxicity (ST).
e. Lowest dose measured in the study.

Table 9-38 provides critical endpoint and corresponding NOAEL and/or LOAEL value for DBP reproductive and/or developmental effects in mature adult male rats.

Table 9-38. Summary of critical effect level for reproductive and/or developmental effects in mature adult male rats after oral exposure to DBP
Life stage Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Adult Rat Testicular pathology, effects on sperm count, density, and motility, and decreased ROW 500 250 Srivastava et al. 1990b;
Zhou et al. 2011c;
Nair 2015
DEHP
Early development: in utero exposure

The European Commission classified DEHP as a Category 1B reproductive toxicant (presumed human reproductive toxicant) as defined in the EU regulation on classification, labelling and packaging of substances and mixtures (ECHA 2015b).

A literature search identified many studies examining the toxicity of DEHP during gestation in rodents. For the purposes of the CRA, only studies covering the masculinization programming window where effects of DEHP were observed at doses at and below 50 mg/kg-bw/day in rats and 100 mg/kg bw/day in mice were evaluated in this draft screening assessment.

Overall, adverse effects in the parameters used to describe RPS in male rat offspring after in utero exposure to DEHP include decreased serum and testicular testosterone levels, delayed PPS, AGD, increase incidences of NR and CRY, gross testicular malformations, and effects in fertility. A full summary of the health effects associated with gestational exposure to DEHP is summarized in Health Canada (2016b). Table 9-39 presents a list of key studies with effects from gestational exposure to DEHP in male offspring.

Table 9-39. Key studies with effects from gestational exposure to DEHP in male offspring (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Testosterone levelsa (T, S) Feminization parametersb Reproductive tract malformations and/or fertilityc Other developmental parametersd Maternal effects
Crl:CD BR SD rats;
1.5 (Con), 10, 30, 100, 300, 1000, 7500, 10000 ppm (est. F0: 0.12, 0.78, 2.4, 7.9, 23, 77, 592, 775);
diet;
6 weeks prior to mating - PND35 of last of three litters (Wolfe and Layton 2003)
NM 592 (AGD)
NE (NR)
592 (PPS)
592 (CRYe)
NP (HYP)
23 (TP- low incidence of small and/or aplastic epididymis and testes, small seminal vesicle, minimal seminiferous tubule atrophy; small prostate @ 77)
592 (FER- ↓ sperm measured);
775 (FER- ↓ epididymal sperm density)
775 (BW)
592 (ROW- in adults)
592 (FV)
NM (EMB)
NM (ESV)
LOEL= 592
(↑ absolute and relative liver weights, relative kidney weight, ↑ food consumption during gestation, ↓ food consumption during lactation)
Crl:CD BR SD rats;
1.5 (Con), 10, 30, 100, 300, 1000, 7500, 10000 ppm (est. F1: 0.09, 0.48, 1.4, 4.9, 14, 48, 391, 543);
diet;
6 weeks premating - PND35 of last of three litters (Wolfe and Layton 2003)
NM 391 (AGD)
NE (NR)
0.48 (PPS)
1.4 (CRYe)
NP (HYP)
14 (TP- low incidence of small and/or aplastic epididymis and testes; seminiferous tubule atrophy at 391)
4.9NDR (FER- abnormal sperm morphology);
391 (FER- ↓ sperm, ↓ motile percentage, ↓ epididymal sperm density)
391 (BW)
391 (ROW- in adults)
543 (FV- no offspring)
543 (EMBe)
NM (ESV)
LOEL= 391
(↑ absolute and relative liver weights and ↑ food consumption)
Crl:CD BR SD rats;
1.5 (Con), 10, 30, 100, 300, 1000, 7500 ppm (est. F2: 0.1, 0.47, 1.4, 4.8, 14, 46, 359);
diet;
6 weeks premating - PND35 of last of three litters (Wolfe and Layton 2003)
NM 359 (AGD)
359 (NR)
359 (PPS)
359 (CRYe)
NP (HYP)
NP (TP)
359 (FER- ↓ sperm, ↓ epididymal sperm density)
NE (BW)
359 (ROW- in adults)
NP (FV)
359 (EMBe)
NM (ESV)
LOEL= 359
(↑ absolute and relative liver weights, ↑ food consumption)
Wistar rats;
0, 3, 10, 30, 100, 300, 600, 900;
gavage;
GD7-PND16 (Christiansen et al. 2010)
NM 10 (AGDf)
10 (NRf)
NM (PPS)
NM (CRY)
NM (HYP)
300 (TP- immature testes, delayed seminiferous epithelium development, focal Leydig cell hyperplasia, ↓ seminiferous tubule diameter, ↓ germ cells)
NM (FER)
300 (BW)
10 (ROWf)
10NDR (FV)
NM (EMB)
NM (ESV)
NE
Wistar rats;
0, 0.015-1.215; 5, 15, 45, 135, 405;
gavage;
GD6-PND21 (Andrade et al. 2006a,b)
NE (T- PND1)
0.045NDR (↑ S)
0.015e (↑ ), 405 (↓ ) (AGD- PND22)
405 (NR)
15 (PPS)
5g (CRY)
NE (HYP)
135 (TP- MNG, ↓ germ cell layers);
405 (TP- ↓ germ cell differentiation in seminiferous tubules, ↓ tubule diameter, ↓ lumen)
15g (FER- ↓ sperm pro, 25%);
0.045 (FER- ↑ abnormalitiesNDR)
0.045NDR (↑ BW, PND1)
5 (↑ ROWg transient)
NE (FV)
NE (EMB)
NM (ESV)
NE

NDR = No dose response relationship.
NP = Not reported.
NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone in the first 4 columns of effects, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Testosterone levels measured (can include quantity/production) at varying days post-birth. T=Testicular testosterone; S=Serum testosterone.
b. Feminization parameters can include anogenital distance (AGD), nipple retention (NR), preputial separation (PPS).
c. Malformations include: cryptorchidism (CRY), hypospadias (HYP), testicular pathology (TP), including multinucleated gonocytes (MNG), and/or reproductive effects such as fertility (FER) in offspring (sperm number, motility) or reproductive success at adult stage after in utero exposure. TTM = transabdominal testicular migration
d. Other developmental effects include: decreases in overall fetal body weight (BW), decreases in reproductive organ weight (ROW), fetal viability (FV), embryotoxicity (EMB), or on the incidence of external, skeletal or visceral malformations (ESV).
e. CRY was based on significant delay in testes descent. EMB was based on an additional crossover mating trial conducted using the 7500 and 10000 ppm males and females (Wolfe and Layton 2003).
f. the effect levels for AGD and NR were based on 1 set out of 2 sets of experiments. Similar effects were observed at higher dose level in the other set of experiment. The effect level for ROW was based on combining the results of the 2 sets of experiments (Christiansen et al. 2010).
g. CRY was based on undescended (ectopic) testes observed in three animals, exposed to either 5, 135 and 405 mg/kg-bw/day (one case in each dose). In all three cases, undescended testes were unilateral (right side) and located in the superficial inguinal pouch. The ROW effect level reported by author was adjusted for body weight by covariance analysis (Andrade et al. 2006a,b).

Overall, the highest NOAEL identified for developmental toxicity of DEHP after gestational exposure was 4.8 mg/kg-bw/day based on small and/or aplastic epididymis, testicular pathology and other RPS effects observed in F1 and F2 at the next dose level of 14 mg/kg-bw/day in a multigenerational reproductive toxicity study (Wolfe and Layton 2003; Blystone et al. 2010). This effect level was also established by other jurisdictions (ECJRC 2008; Danish EPA 2012; US CPSC CHAP 2014; EFSA 2005). At similar dose levels of 10-15 mg/kg-bw/day, decreases in AGD, increased NR, decreased reproductive organ weights, and delayed PPS were observed in rat offspring in other studies (Andrade et al. 2006a,b; Christiansen et al. 2010). The lowest NOEL for maternal toxicity of DEHP was 359 mg/kg-bw/day based on increase in liver weight and food consumption.

Exposure at prepubertal/pubertal life stages

A literature search identified many studies examining the reproductive toxicity of DEHP in young rodents. Results from repeated-dose oral exposure studies in sexually immature rats (PND1-55) have shown that administration of DEHP can cause reproductive effects in male rats.

Overall, adverse effects observed in (pre)pubertal males after short-termexposure to DEHP include changes in serum and testicular testosterone levels, histopathological effects in the testes, and potential effects in fertility (spermatogenesis, sperm motility, and number). Reproductive effects observed in prepubertal or pubertal mice after exposure to DEHP occurred at higher doses than rats. Other species were also found to be less sensitive than rats to DEHP. No significant effects were observed in Cynomolgus monkeys, marmosets and Syrian hamsters treated with DEHP. A summary of the health effects associated with prepubertal/pubertal exposure to DEHP is summarized in Health Canada (2016b). Table 9-40 presents key studies with effects from prepubertal/pubertal exposure to DEHP in males.

Table 9-40. Key studies of effects from exposure to DEHP in (pre)pubertal males (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Life stage at the start of dosing (age) Hormone levelsa
(T, S, LH)
Fertilityb Reproductive tract pathologyc Other effectsd
SD rats;
0, 10, 100, 1000, 2000 (fatal);
gavage;
5 days (Dostal et al. 1988)
Prepubertal
(PND21)
NM (T)
NM (S)
NM (LH)
NM 1000 (loss of spermatocytes in Sertoli cell cytoplasm, degenerating spermatocytes) 1000 (BW-sig. @ 10, NS @ 100)
100 (ROW)
NM (ST)
SD rats;
0, 200, 500, 1000;
gavage;
5 days (fertility group mated at 8-15 wks (mated to F344)) (Dostal et al. 1988)
Postnatal
(PND6)
NM 200e (↓ spermatid heads at 13 weeks of age; @ 1000 at 19 weeks of age) 500 (↓ Sertoli cell number 24h after last dose) 1000
(BW-24h after last dose)
500 (ROW-24h after last dose)
NM (ST)
SD rats;
0, 300, 600;
gavage;
21 days (Cammack et al. 2003)
Postnatal
(PND3-5)
NM NE (PND90) 300 (PND25; testicular changes, such as partial depletion of the germinal epithelium and/or decreased diameter of the seminiferous tubules) Less severe @PND 90 600 (BW)
300 (ROW- PND25, PND 90)
300 (ST-↑ rel. liver weight)
Wistar rats;
0, 1, 3, 10, 30, 100, 300;
gavage;
40 days (Tonk et al. 2012)f
Prepubertal
(PND10)
NM (T)
3.9(↓ S-BMDL5)
14(↑ LH-BMDL5)
9.5 (sperm count, BMDL5) 300f (TP- lesions; sertoli cell vacuolization) NE (BW)
84 (ROW - BMDL5)
4.4 (ST-↑ rel. liver weight - BMDL5)
Wistar rats;
0, 1, 3, 10, 30, 100, 300;
gavage;
40 days (Tonk et al. 2012)f
Pubertal-adult
(PND50)
NM (T)
3.9 (S-BMDL5)
62 (LH-BMDL5)
55 (BMDL5, sperm count) 300f (TP- lesions; sertoli cell vacuolization) NE (BW)
517(ROW BMDL5)
4.4 (ST-↑ rel. liver weight - BMDL5) (ST)

NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Hormone levels can include quantity/production of testicular testosterone (T), serum testosterone (S), leutinizing hormone (LH), glucocorticoid hormone (GC), or follicle stimulating hormone (FSH).
b. Fertility parameters include sperm number, motility, morphology, viability, stages of spermatogenesis, or reproductive success at adult stage after in utero exposure.
c. Reproductive tract pathology includes: testicular pathology (TP): any observations based on histopathological examination of the testes such as, but not limited to, multinucleated gonocytes/germ cells (MNGs), necrosis, hyperplasia, clustering of small Leydig cells (LC), vacuolisation of Sertoli cells, decrease in Leydig cell number, an increase in Leydig cell size, focal dysgenesis, and/or seminiferous tubule atrophy.
d. Other effects include: Decreased overall body weight (BW), decreased reproductive organ weight (ROW) and systemic toxicity (ST).
e. Lowest dose measured in the study.
f. Data were represented as 5% lower confidence bound of benchmark dose (BMDL5). In juvenile males, preputial separation was delayed at the 300 mg/kg bw dose group. Testicular pathological changes were only examined at the 300 mg/kg-bw/day dose group (Tonk et al. 2012).

Overall, the highest NOAEL identified for reproductive toxicity of DEHP at the prepubertal-pubertal life stage was 10 mg/kg-bw/day based on a significant decrease in absolute testis weight at the next dose level of 100 mg/kg-bw/day in rats exposed to DEHP for 5 days from PND21 (Dostal et al. 1988). At 200 mg/kg-bw/day, a decrease in spermatid heads was observed at 13 weeks of age in rats exposed for 5 days from PND6 (Dostal et al. 1988). Testicular pathological changes were observed at 300 mg/kg-bw/day and higher in rats exposed to DEHP for 21 days from PND3-5 (Cammack et al. 2003) and rats exposed for 40 days from PND10 or PND50 (Tonk et al. 2012).

Exposure at the mature male adult stage

Studies examining the potential reproductive toxicity of DEHP at the adult male life stage (PND55+) were identified in rats, mice, marmosets and ferrets.

In rats, chronic exposure to DEHP (104 weeks to 2 years) resulted in inhibition of spermatogenesis starting from 10-29 mg/kg-bw/day (Ganning 1991; David et al. 2000a). In animals treated for less than a chronic duration (2-13 weeks), inhibition of spermatogenesis, decrease sperm count and decrease in sperm motility was observed at 300-900 mg/kg-bw/day (Poon et al. 1997; Wolfe and Layton 2003; Kwack et al. 2009; Tonk et al. 2012; Abd-Ellah et al. 2016). Decrease in testis weight and testicular pathological changes were generally observed at 300 mg/kg-bw/day and above.

Adult mice, ferrets, and marmosets were found to be less sensitive than rats to DEHP. Systemic effects in mice such as a decrease in relative kidney weight and an increase in absolute liver weight were observed at 99 mg/kg-bw/day whereas effects on sperm and testicular pathological changes were observed at 292 mg/kg-bw/day in a chronic study (David et al. 2000b). In Ferrets, testicular pathological changes, decreases in body weight and increases in liver weight were observed at a much higher dose level of 1200 mg/kg-bw/day (Lake et al. 1976). In marmoset, only peroxisome proliferator-activated receptor (PPAR) effects in liver were observed at 500 mg/kg-bw/day (Kurata et al. 1998). A summary of the health effects associated with adult life stage exposure to DEHP is summarized in Health Canada (2016b). Table 9-41 presents key study with effects from adult life stage exposure to DEHP in males.

Table 9-41. Effects from exposure to DEHP in adults males (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Life stage at the start of dosing (age) Hormone levelsa
(T, S, LH)
Fertilityb Reproductive tract pathologyc Other effectsd
Fischer 344 rats;
0, 100, 500, 2500, 12500 ppm (est. 0, 5.8, 28.9, 146.6, 789);
diet;
104 weeks (David et al. 2000a)
6 weeks NM (T)
NM (S)
NM (LH)
28.9 (↓ spermatogenesis) ↓ interstitial cell tumors of testes at 789 789 (BW- ↓ from wk 1)
789 (ROW)
146.6 (ST- ↑ rel/abs. kidney, liver weight, ↑ rel. lung weight; 789 - liver and kidneye histopath effects, pancreas, ↑ pituitary castration cells) ↑ spongiosis hepatis at 146.6 and 789)

NM = Not measured.
a. Hormone level can include quantity/production of testicular testosterone (T), serum testosterone (S), or leutinizing hormone (LH).
b. Fertility parameters include: sperm number, motility, morphology, viability, stages of spermatogenesis, or reproductive success after mating.
c. Reproductive tract pathology includes: any observations based on histopathological examination of the testes such as, but not limited to, multinucleated gonocytes (MNGs), necrosis, hyperplasia, clustering of small Leydig cells, vacuolisation of Sertoli cells, decrease in Leydig cell number, an increase in Leydig cell size, focal dysgenesis, and/or seminiferous tubule atrophy.
d. Other effects include: Decreased overall body weight (BW), decreased reproductive organ weight (ROW) and systemic toxicity (ST).
e. Other systemic effects include: chronic progressive nephropathy observed in all male groups with increased severity at 789 mg/kg-bw/day. Significant ↑ of hyperplasia and adenomas of pancreas in males only at 789 mg/kg-bw/day (David et al. 2000a).

Overall, the highest NOAEL identified for reproductive toxicity of DEHP at the adult life stage was 5.8 mg/kg-bw/day based on a decrease in spermatogenesis at the next dose level of 29 mg/kg-bw/day in male adult rats exposed chronically to DEHP for 104 weeks (David et al. 2000a).

DnHP
Early development: in utero exposure

The European Commission classified DnHP as a Category 1B reproductive toxicant (presumed human reproductive toxicant) as defined in the EU regulation on classification, labelling and packaging of substances and mixtures (ECHA 2015c).

A literature search identified a number of recent studies examining the toxicity of DnHP during gestation in rodents. For the purposes of the CRA, only studies covering the masculinization programming window in males were evaluated in this draft screening assessment.

Overall, adverse effects in the parameters used to describe RPS in male rat offspring after in utero exposure to DnHP include decreased serum and testicular testosterone levels, delayed PPS, AGD, retention of nipple/areolae (NR), increase incidences of CRY and hypospadias, gross testicular malformations, and effects in fertility. Two studies were conducted in mice where embryotoxicity and effects on fetal viability were observed. A summary of the health effects associated with gestational exposure to DnHP is summarized in Health Canada (2016b). Table 9-42 presents a list of key studies with effects from gestational exposure to DnHP in male offspring.

Table 9-42. Key studies with effects from gestational exposure to DnHP in male offspring (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Testosterone levelsa
(T, S)
Feminization parametersb Reproductive tract malformations and/or fertilityc Other developmental parametersd Maternal effects
SD rats;
0, 5, 20, 50, 100, 125, 250, 500, 625;
gavage;
GD12-19 (Saillenfait et coll., 2013a)
20 (T- ↓ by 17% on GD19; ED50= 67.4 mg/kg)
NM (S)
NM (AGD)
NM (NR)
NM (PPS)
NM (CRY)
NM (HYP)
500e (TP - abnormal distribution of Leydig cells on GD19, ↓ number of Leydig cell clusters, ↑ size of Leydig cell clusters, other effects)
NM (FER)
NM (BW)
NM (ROW)
NM (FV)
NM (EMB)
NM (ESV)
NP
Wistar rats;
0, 20, 100, 500;
oral gavage;
GD6-19 (Ahbab and Barlas 2015)
20f (↓ T:450 pg/ml testosterone compared to control, p less than 0.05) 20f (↓ AGD/cube root of body weight ratio, control at approx. 2.4 mm/g1/3 and 20 mg/kg/day at approx. 2.0 mm/g1/3, p less than 0.05) NM (CRY)
NM (HYP)
20f (TP - atrophic and small seminiferous tubules, decreased germ cells in tubules, detached cells from tubular wall)
NM (FER)
20f (↓ 15.9% BW; at 100, ↓ 20.5% BW; at 500, ↑ 13.6% BW)
NM (ROW)
NE (FV)
20f (EMB-resorption based on number of offspring, not by percentage)
NM (ESV)
NE

NP = Results not recorded (but measurement was stated in the methods and materials).
NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone in the first 4 columns of effects, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Testosterone levels measured (can include quantity/production) at varying days post-birth. T=Testicular testosterone; S=Serum testosterone.
b. Feminization parameters can include anogenital distance (AGD), nipple retention (NR), preputial separation (PPS).
c. Malformations include: cryptorchidism (CRY), hypospadias (HYP), testicular pathology (TP), including multinucleated gonocytes (MNG), and/or reproductive effects such as fertility (FER) in offspring (sperm number, motility) or reproductive success at adult stage after in utero exposure. TTM = transabdominal testicular migration
d. Other developmental effects include: decreases in overall fetal body weight (BW), decreases in reproductive organ weight (ROW), fetal viability (FV), embryotoxicity (EMB), or on the incidence of external, skeletal or visceral malformations (ESV).
e. TP was only examined in the control and 500 mg/kg-bw/day dose groups (Saillenfait et al. 2013).
f. Lowest dose measured in the study.

Overall, the highest NOAEL identified for developmental toxicity of DnHP after gestational exposure was 5 mg/kg-bw/day based on decreased serum or testicular testosterone levels, decreased AGD at birth in males and testicular pathological changes observed at 20 mg/kg-bw/day or higher in rats (Ahbab and Barlas 2015; Saillenfait et al. 2013a).

Exposure at prepubertal/pubertal life stages

A literature search identified four studies examining the reproductive toxicity of DnHP in young sexually immature rats (PND1-55). These studies were generally tested at high dose levels and did not describe RPS-related parameters other than one study that was tested at lower dose levels using castrated male rats. A summary of the health effects associated with prepubertal/pubertal exposure to DnHP is summarized in Health Canada (2016b). Since available studies were limited, DEHP was identified, using the same chemical categories and read-across approach as that used for other phthalates, as the "closest analogue" phthalate, taking into consideration the similarities in monoester metabolism as well as the length and nature of the ester chains (Health Canada 2015).

As described earlier in the DEHP section, the highest NOAEL identified for reproductive toxicity of DEHP at the prepubertal-pubertal life stage was 10 mg/kg-bw/day based on significant decreases in absolute testis weight at the next dose level of 100 mg/kg-bw/day in rats exposed to DEHP for 5 days from PND21 (Dostal et al. 1988). Therefore, the critical effect level of 10 mg/kg-bw/day will be used to characterize the risk of developmental toxicity of DnHP for this life stage.

Exposure at the mature male adult stage

Two studies examining the potential reproductive toxicity of DnHP at the adult male life stage (PND55+) were identified in rodents. One study was conducted in rats where RPS-related parameters were not measured. Another study was conducted in mice where fertility effects and testicular pathology were examined. A summary of the health effects associated with adult stage exposure to DnHP is summarized in Health Canada (2016b). Table 9-43 presents the key study with effects from exposure to DnHP in adult males.

Table 9-43. Effects from exposure to DnHP in mature adult males (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Age at the start of dosing Hormone levelsa
(T, S, LH)
Fertilityb Reproductive tract pathologyc Other effectsd
COBS Crl: CD-1, (IRC)BR outbred albino mice; 0, 0.3, 0.6, 1.2%;
est. 0, 390, 780, 1560 (based on dose conversion by HC 1994);
(crossover mating trial and necropsy of highest dose only);
oral diet;
7 days premating and 98 days cohabitation (total 105 days exposure)
(Lamb et al. 1987)
PND 42 NM 390e (↓ fertility at mating, litters/pair, live pups/litter, proportion of pups born alive, live pup weight; 780, produced 1 litter; 1560, infertile)
1560f (sperm parameters, ↓ % motile sperm, sperm concentration, % abnormal sperm)
1560f (extensive atrophy of seminiferous tubules, tubules were lined mostly by Sertoli cells, no normal spermatogenesis observed, in 3 of 18 mice, observed microscopic changes in seminal vesicles 1560f (↓ 10.27% BW in F0 male, calculated from 36.19g at 1560 dose compared to 40.33 for control)
1560f (↓ ROW, i.e. left testis and epididymis, right testis, right epididymis, prostate, seminal vesicles)
1560 (ST -↓ liver, kidney and adrenal weight)

NM = Not measured.
a. Hormone levels can include quantity/production of testicular testosterone (T), serum testosterone (S), or leutinizing hormone (LH).
b. Fertility parameters include sperm number, motility, morphology, viability, stages of spermatogenesis, or reproductive success after mating.
c. Reproductive tract pathology includes: any observations based on histopathological examination of the testes such as, but not limited to, multinucleated gonocytes (MNGs), necrosis, hyperplasia, clustering of small Leydig cells, vacuolisation of Sertoli cells, decrease in Leydig cell number, an increase in Leydig cell size, focal dysgenesis, and/or seminiferous tubule atrophy.
d. Other effects include: Decreased overall body weight (BW), decreased reproductive organ weight (ROW) and systemic toxicity (ST).
e. Lowest dose tested.
f. Reproductive tract pathology, BW and ROW parameters were only examined in the control and the 1560 mg/kg-bw/day dose groups (Lamb et al. 1987).

Overall, no NOAEL was identified and the lowest LOAEL identified for reproductive toxicity of DnHP was 390 mg/kg-bw/day in mice based on adverse effects in fertility (decreases in fertility, the number of litters per pair, the number of live pups per litter and the proportion of pups born alive) treated with DnHP for 105 days during the adult male life stage (Lamb et al. 1987).

DIOP
Early development: in utero exposure

The European Commission classified DIOP as a Category 1B reproductive toxicant (presumed human reproductive toxicant) as defined in the EU regulation on classification, labelling and packaging of substances and mixtures (ECHA 2015d).

Three studies were identified that were all conducted by Saillenfait et al. (2013b) during the masculinization programming window. Adverse effects in the parameters used to describe RPS in male rat offspring after in utero exposure to DIOP included decreased testicular testosterone levels, decreased AGD, increased incidence of NR and CRY, gross testicular malformations, and effects in fertility. A summary of the health effects associated with gestational exposure to DIOP is summarized in Health Canada (2016b). Table 9-44 presents the key study with effects from gestational exposure to DnHP in male offspring.

Table 9-44. Effects from gestational exposure to DIOP in male offspring (mg/kg-bw/day)
Strain and species;
dose (mg/kg-bw/day);
route;
duration (reference)
Testosterone levelsa
(T, S)
Feminization parametersb Reproductive tract malformations and/or fertilityc Other developmental parametersd Maternal effects
DIOP
SD Rats;
0, 100, 500, 1000;
gavage;
GD12-21 (Saillenfait et al. 2013b)
NM NM (AGD)
1000 (NR @ PND 68-84)
NM (PPS)
1000 (CRY @ PND 68-84)
1000 (HYP @ PND 68-84)
500 (TP-one incidence each of unilaterally enlarged testis, abnormal epididymis, underdeveloped seminal vesicles and prostate)
500 (FER-hypospermatogenesis)
NE (BW)
500 (ROW)
1000 (FV @PND21)
NE (EMB)
NM (ESV)
1000 (BW)

NM = Not measured.
NE = No effect observed at the dose range tested. When NE is presented alone in the first 4 columns, all parameters in the footnote description were measured and no statistically significant effects were observed in the endpoints at the dose range administered.
a. Testosterone levels measured (can include quantity/production) at varying days post-birth. T=Testicular testosterone; S=Serum testosterone.
b. Feminization parameters can include anogenital distance (AGD), nipple retention (NR), preputial separation (PPS).
c. Malformations can include cryptorchidism (CRY), hypospadias (HYP), testicular pathology (TP), and/or reproductive effects such as fertility (FER) in offspring (sperm number, motility) or reproductive success at adult stage after in utero exposure. TTM = transabdominal testicular migration.
d. Other developmental effects include decreases in overall fetal body weight at PND 1 (BW), decreases in reproductive organ weight (ROW), embryo/fetal viability (FV), average litter size (ALS), or on the incidence of external, skeletal or visceral malformations (ESV).

Overall, the highest NOAEL identified for developmental toxicity of DIOP after gestational exposure was 100 mg/kg-bw/day based on testicular pathological changes, effects on fertility and decreased testis weight at the next dose level of 500 mg/kg-bw/day. Maternal decrease in body weight was observed at 1000 mg/kg-bw/day (Saillenfait et al. 2013b).

Exposure at prepubertal/pubertal life stages

There were no repeated-dose oral exposure studies in sexually immature animals (PND1-55) with DIOP. DIHepP was identified as the appropriate analogue to use for read across. The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects literature related to DIHepP. Table 9-45 provides the critical endpoints and corresponding NOAEL and/or LOAEL values for DIOP.

Table 9-45. Summary of critical effect levels for reproductive and/or developmental effects after oral exposure to DIOP based on its analogue
Life stage Species Effects LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
(pre)pubertal Rat
(DIHepP)
Significant reduction in AGD;
delayed preputial separation, nipple retention, hypospadias and cryptorchidism in F1 pups
419-764 227-416 McKee et al. 2006
adult Rat
(DIHepP)
No adverse effects observed up to the highest dose level tested NA 404-623 McKee et al. 2006

NA = Not applicable.

Oral exposure at the mature male adult stage

There were no repeated-dose oral exposure studies in adult animals with DIOP. DIHepP was identified as the appropriate analogue to use for read across. The MCP SOS (Environment Canada, Health Canada 2015b) summarizes the health effects literature related to DIHepP. Table 9-45, described in the above section, provides the critical endpoints and corresponding NOAEL and/or LOAEL values for DIOP.

Evidence in humans

A literature search was conducted to identify any human data for additional phthalates. The search was focused on reproductive and developmental endpoints in males as these endpoints were identified as critical health endpoints in the SOS reports. Studies were further evaluated and scored for quality using a consistent evaluation metric (Downs and Black 1998).

For the health outcomes evaluated (i.e., sex hormone levels, anogenital distance, birth measures, male infant genitalia, preterm birth and gestational age, altered male puberty, gynecomastia, changes in sem*n parameters, pregnancy loss, and altered time to pregnancy), there was limited evidence of association between DEHP and sex hormone levels (Pan et al. 2006; Meeker et al. 2009; Li et al. 2011; Mendiola et al. 2011, 2012; Joensen et al. 2012; Araki et al. 2014; Ferguson et al. 2014a; Pant et al. 2014; Su et al. 2014; Chang et al. 2015; Fong et al. 2015; Jensen et al. 2015; Pan et al. 2015; Wang et al. 2016), birth measures (Zhang et al. 2009; Philippat et al. 2012; de co*ck et al. 2014; Zhao et al. 2014; Lenters et al. 2015b; Xie et al. 2015; Zhao et al. 2015; Casas et al. 2016), or sem*n parameters (Zhang et al. 2006; Pant et al. 2008; Jurewicz et al. 2013; Huang et al. 2014; Pant et al. 2014; Specht et al. 2014; Axelsson et al. 2015a; Axelsson et al. 2015b; Lenters et al. 2015a; Pan et al. 2015; Wang et al. 2015b; Thurston et al. 2016). There was inadequate evidence or no evidence of association between the remaining phthalates and the outcomes (Table 9-46). More detail is provided in Health Canada (2016a).

Table 9-46. Summary of levels of evidence of associations between additional phthalates and health outcomes
Outcome BBP
(MBP, MBzP)
DBP
(MBP, etc.)
DEHP
(MEHP, MEOHP, MEHHP, MECPP, MCMHP)
DnHP
(MnHP)
DIOP
Sex hormone levels IA (9) NA (1) LA (17) NM NM
Anogenital distance NA (3) NM IA (4) NM NM
Birth measures IA (4) IA (2) LA (11) NM NM
Male infant genitalia NA (1) NM NA (2) NM NM
Preterm birth and gestational age IA (4) NM IA (5) NM NM
Altered male puberty NA (2) NM IA (4) NM NM
Gynecomastia NA (1) NM NA (2) NM NM
Changes in sem*n parameteres IA (8) NA (1) LA (11) NM NM
Pregnancy loss NA (3) NM IA (4) NM NM
Altered time to pregnancy IA (2) NA (1) NA (2) NM NM

() = Number of studies.
NM = Not measured in studies of quartile 2 and above (See Health Canada [2016a] for more details).
NA = No evidence of association.
IA = Inadequate evidence of an association.
LA = Limited evidence of an association.
MBP = Monobutyl phthalate.
MBzP = Monobenzyl phthalate.
MCMHP = Mono[2-(carboxymethyl)hexyl] phthalate.
MEHP = Mono(2-ethyl hexyl)phthalate.
MEOHP = Mono(2-ethyl-5-oxohexyl) phthalate.
MEHHP = Mono(2-ethyl-5-hydroxyhexyl) phthalate.
MECPP = Mono(2-ethyl-5-carboxypentyl) phthalate.
MnHP = Mono-h-hexyl phthalate.

9.2.3 Long-chain phthalates

DIDP

The LCP SOS (Environment Canada, Health Canada 2015d) summarizes the health effects literature related to DIDP. No new animal hazard studies were identified after the literature cut-off date of the LCP SOS.

Table 9-47 provides critical endpoints and corresponding NOAEL and/or LOAEL values for DIDP, as previously described in LCP SOS (Environment Canada, Health Canada 2015d).

Table 9-47. Summary of critical systemic effects after oral exposure to DIDP
Endpoint Species Effect LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Short term Rat Increase in liver weight in males accompanied with histological changes at the highest dose 300 300
(females)
BIBRA 1986
Subchronic Dog Increase in liver weight accompanied with histological changes. 75 15 Hazleton Laboratories 1968b
Chronic Rat Histopathological changes in the liver in males. 22 N/A Cho et al. 2008

N/A = Not applicable.

DUP

The LCP SOS (Environment Canada, Health Canada 2015d) summarizes the health effects literature related to DUP. No new literature was identified after the literature cut-off date of the LCP SOS. Table 9-48 provides critical endpoints and corresponding NOAEL and/or LOAEL values for DUP, as previously described in LCP SOS (Environment Canada, Health Canada 2015d).

Table 9-48. Summary of critical systemic effects associated with oral exposure to DUP
Endpoint Species Effect LOAEL
(mg/kg-bw/day)
NOAEL
(mg/kg-bw/day)
Reference
Short term Rat Decreased body weight gain and increased liver and kidney weights accompanied with liver lesions 1145 282 Barber et al. 1987
Subchronic Rat
(DnOP)
Increases in liver enzyme activities and histological effects in the liver and thyroid less than or equal to 350-403 37 Poon et al. 1997
Evidence in humans

A literature update was also conducted to identify any recent human data for long-chain phthalates. The search was focused on reproductive and developmental endpoints in males as these endpoints were identified as critical health endpoints in the SOS reports. No information is currently available on the potential reproductive-developmental effects of DUP in humans. Studies identified for DIDP were further evaluated and scored for quality using a consistent evaluation metric (Downs and Black 1998).

For the health outcomes evaluated (i.e., sex hormone levels, anogenital distance, birth measures, male infant genitalia, preterm birth and gestational age, altered male puberty, gynecomastia, changes in sem*n parameters, pregnancy loss, and altered time to pregnancy), there was no evidence of association between any of the long-chain phthalates evaluated and the outcomes (Table 9-49). More detail is provided in Health Canada (2016a).

Table 9-49. Summary of levels of evidence of associations between long-chain phthalates and health outcomes
Outcome DIDP
(MIDP/MCINP)
Sex hormone levels NA (1)
Anogenital distance NA (1)
Birth measures NA (1)
Male infant genitalia NM
Preterm birth and gestational age NA (1)
Altered male puberty NM
Gynecomastia NM
Changes in sem*n parameteres NM
Pregnancy loss NA (1)
Altered time to pregnancy NM

() = Number of studies.
NM = Not measured in studies of quartile 2 and above (See Health Canada [2016a] for more details).
NA = No evidence of association.
IA = Inadequate evidence of an association.
LA = Limited evidence of an association.
MIDP = Monoisodecyl phthalate.
MCINP = Mono(carboxyisononyl) phthalate.

9.3 Characterization of risk to human health

9.3.1 Short-chain phthalates

DMP

Table 9-50 provides all relevant exposure and hazard values for DMP, as well as resultant margins of exposure (MOEs), for determination of risk, which were previously described in the SCP SOS (Environment Canada, Health Canada 2015a). Overall, the MOEs for DMP are considered to be adequate to account for uncertainties in the exposure and health effect databases.

Table 9-50. Summary of MOEs to DMP for subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
Level and basis for NOAEL
(mg/kg-bw/day)
MOEd
Children (males) 2-3 years: biomonitoring, MIREC CD Plus 0.19
(0.66)
NOAEL = 230
(chronic dermal, DEP)
Decrease in absolute brain weight in males
(NTP 1995)
greater than 1 million
(340 000)
Infants 0-6 months, breast milk fed: environmental media and food, oral and inhalation 0.019
(0.26)
LOAEL = 1862
(pubertal, 7 days oral, DMP)
↓ serum and testicular testosterone, dihydrotestosterone concentrations and ↑ absolute, relative liver weight
(Oishi and Hiraga 1980) (no NOAEL)
greater than 1 million
Infants 0-6 months: diaper cream, dermal 2.7a
(8.2)a
NOAEL = 200
(subchronic dermal, DMP)
Changes in nervous system and renal function in males
(Timofieyskaya 1976)
74 000
(24 000)
Adults (females) 20+ years: biomonitoring, NHANES 0.027
(0.26)
NOAEL = 415
(chronic dermal, DEP)
Decrease in BW of 8% in females
(NTP 1995)
greater than 1 million
Adolescents (males) 12-19 years: biomonitoring, NHANES 0.042
(0.29)
NOAEL = 230
(chronic dermal, DEP)
Decrease in absolute brain weight in males
(NTP 1995)
greater than 1 million
(790 000)
Adolescents 12-19 years: environmental media and food, oral and inhalation 0.0085
(0.091)
NOAEL = 750
(in utero oral DMP)
Highest dose tested for potential RPS effects
(Gray et al. 2000; Furr et al. 2014)
greater than 1 millionb
Adults 20+ years: hairspray, dermal 66ac
(200)a
NOEL = 230
(chronic dermal, DEP)
Decrease in absolute brain weight in males
(NTP 1995)
3500
(1150)
Adults 20+ years: hair dye, dermal 1400ac
(4200)a
NOAEL = 2380
(short term dermal, DMP)
slight ↓ body weight in dams
(Hansen and Meyer 1989)
1700
(570)

a. External dermal exposure estimates.
b. This margin is also protective for potential effects of DMP (based on effects observed with DEP) on males of this age group which occur at higher doses.
c. Lower-bound estimate: based on minimum concentration.
d. Margin of exposure: central tendancy and (upper bounding).

9.3.2 Medium-chain phthalates, additional phthalates and cumulative risk assessment

The critical effects of concern of medium-chain phthalates consisted of adverse effects on the development of the male reproductive system following gestational exposure, with a particular focus on RPS-related parameters that were identified in the rat, the most sensitive species. These parameters are considered adverse and relevant for characterizing risk of exposure of the general Canadian population to this subgrouping of phthalates. Please see MCP SOS (Environment Canada, Health Canada 2015b) for a general summary and rationale.

In cases where limited rat studies were available, effect levels in other species (i.e., mice) that were lower than in rats were used for risk characterization. Evidence in humans, based on the Downs and Black scoring system (Downs and Black 1998), indicated limited evidence of association between DINP and sex hormone levels or sem*n parameters and between DEHP and sex hormone levels, birth measures or sem*n parameters. This supports the selection of the mode of action for risk characterization.

In the following sections, human health risk from exposure to medium-chain phthalates within the Phthalate Grouping is characterized on an individual basis, followed by a cumulative risk assessment to address the potential risk of concurrent exposure to medium-chain phthalates exhibiting a similar mode of action.

9.3.2.1 Individual risk characterization of the original medium-chain subgroup
DIBP

Table 9-51 provides all relevant exposure and hazard values for DIBP, as well as resultant MOEs, for determination of risk, which were previously described in the MCP SOS (Environment Canada, Health Canada 2015b). Overall, the MOEs for DIBP are considered to be adequate to account for uncertainties in the exposure and health effect databases. From the information available, there is evidence that DIBP elicits effects on the developing male reproductive system, indicative of RPS, which suggests that DIBP has a common mode of action with other phthalates in this grouping.

Table 9-51. Summary of MOEs to DIBP for relevant subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
Level and basis for oral NOAEL
(mg/kg-bw/day)
MOEc
Children (males and females) 6-11 years: biomonitoring, CHMS 1.5
(5.3)
NOAEL = 300
Testicular pathology at 500 mg/kg-bw/day (7d)
(Zhu et al. 2010)
200 000
(60 000)
Infants 0-6 months (breastfed): environmental media and food 1.6
(5.9)
NOAEL = 300
Testicular pathology at 500 mg/kg-bw/day (7d)
(Zhu et al. 2010)
200 000
(50 000)
Infants/children (0-18 months)a: contact plastic articles, dermal 30.7b
(245.3)
NOAEL = 300
Testicular pathology at 500 mg/kg-bw/day (7d)
(Zhu et al. 2010)
10 000
(1200)
Infants (0-18 months): mouthing toys, oral 62.8b
(251.0)
NOAEL = 300
Testicular pathology at 500 mg/kg-bw/day (7d)
(Zhu et al. 2010)
5000
(1200)
Adults (females) 20-49 years: biomonitoring, CHMS 0.56
(1.4)
NOAEL = 125
Reduced AGD, NR, effects on fertility and other TDS effects at the next highest dose (250 mg/kg-bw/day)
(Saillenfait et al. 2008; Furr et al. 2014)
220 000
(89 000)
Adults 20-59 yearsa: chronic body lotion, dermal 0.030 NOAEL = 125
Reduced AGD, NR, effects on fertility and other TDS effects at the next highest dose (250 mg/kg-bw/day)
(Saillenfait et al. 2008; Furr et al. 2014)
greater than 1 million
Adults (20+): contact plastic articles, dermal 30.8b
(96.3)
NOAEL = 125
Reduced AGD, NR, effects on fertility and other TDS effects at the next highest dose (250 mg/kg-bw/day)
(Saillenfait et al. 2008; Furr et al. 2014)
4060
(1300)

TDS = human testicular dysgenesis syndrome.
a. Estimate adjusted on the basis of 10% dermal absorption of DBP.
b. Estimated lower-end exposure.
c. Margin of exposure: central tendancy and (upper bounding).

DCHP

Table 9-52 provides all relevant exposure and hazard values for DCHP, as well as resultant MOEs, for determination of risk, which were previously described in the MCP SOS (Environment Canada, Health Canada 2015b). Overall, the MOEs for DCHP are considered to be adequate to account for uncertainties in the exposure and health effect databases. From the information available, there is evidence that DCHP has effects on the developing male reproductive system, indicative of RPS, which suggests that DCHP has a common mode of action with other phthalates in the grouping.

Table 9-52. Summary of MOEs to DCHP for relevant subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
Level and basis for oral NOAEL
(mg/kg-bw/day)
MOEb
Children 6 months to 4 years: indoor air and dust, dermal and inhalation 0.0018
(0.15)
NOAEL = 25
Increased relative liver weight (females), accompanied by histological changes in the liver and kidneys in both sexes at the two highest doses tested (subchronic)
(de Ryke and Willems 1977)
greater than 1 million
(170 000)
Adolescents 12-19a years: indoor air and dust, dermal and inhalation less than 0.001
(0.065)
LOAEL = 10-20
Testicular pathological changes after in utero exposure during GD12-21 (Li et al. 2016).
Reduced AGD, testicular pathology and increased resorption after in utero exposure during GD6-19 (Ahbab and Barlas 2015).
Antiandrogenic effects (decreased AGD and retained nipples, decreased testosterone production) in F1 and F2 males at higher dose levels in a two-generation study in rats
(Hoshino et al. 2005)
greater than 1 million
(155 000-310 000)

a. MOEs were calculated for non-pregnant individuals (male and female) and pregnant females for this age group.
b. Margin of exposure: central tendancy and (upper bounding).

DMCHP

Table 9-53 provides all relevant exposure and hazard values for DMCHP, as well as resultant MOEs, for determination of risk, which were previously described in the MCP SOS (Environment Canada, Health Canada 2015b). Overall, the MOEs for DMCHP are considered to be adequate to account for uncertainties in the exposure and health effect databases. From the information available, there is evidence that DMCHP has effects on the developing male reproductive system, indicative of RPS, which suggests that DMCHP has a common mode of action with other phthalates in the grouping.

Table 9-53. Summary of MOEs to DMCHP for relevant subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
Level and basis for oral NOAEL
(mg/kg-bw/day)
MOEb
Infants 0-6 months: dust ingestion, oral 0.0027
(0.054)
NOAELDCHP = 25
Increased relative liver weight (females), accompanied by histological changes in the liver and kidneys in both sexes at the two highest doses tested (subchronic)
(de Ryke and Willems 1977)
greater than 1 million
(460 000)
Adolescents 12-19a years: dust ingestion, oral less than 0.001 LOAELDCHP = 10-20
Testicular pathological changes after in utero exposure during GD12-21 (Li et al. 2016).
Reduced AGD, testicular pathology and increased resorption after in utero exposure during GD6-19 (Ahbab and Barlas 2015).
Antiandrogenic effects (decreased AGD and retained nipples, decreased testosterone production) in F1 and F2 males at higher dose levels in a two-generation study in rats
(Hoshino et al. 2005)
greater than 1 million
Adults 20+a years: dust ingestion, oral less than 0.001 LOAELDCHP = 10-20
Testicular pathological changes after in utero exposure during GD12-21 (Li et al. 2016).
Reduced AGD, testicular pathology and increased resorption after in utero exposure during GD6-19 (Ahbab and Barlas 2015).
Antiandrogenic effects (decreased AGD and retained nipples, decreased testosterone production) in F1 and F2 males at higher dose levels in a two-generation study in rats
(Hoshino et al. 2005)
greater than 1 million

a. MOEs were calculated for non-pregnant individuals (male and female) and pregnant females for this age group.
b. Margin of exposure: central tendancy and (upper bounding).

DBzP

Table 9-54 provides all relevant exposure and hazard values for DBzP, as well as resultant MOEs, for determination of risk, which were previously described in the MCP SOS (Environment Canada, Health Canada 2015b). Overall, the MOEs for DBzP are considered to be adequate to account for uncertainties in the exposure and health effect databases. From the information available, there is evidence that DBzP has effects on the developing male reproductive system, indicative of RPS, which suggests that DBzP has a common mode of action with other phthalates in the grouping.

Table 9-54. Summary of MOEs to DBzP for relevant subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
Level and basis for oral NOAEL
(mg/kg-bw/day)
MOEb
Infants 0-6 months: dust ingestion, oral 0.016
(0.097)
LOAELMBzP = 167
decrease in body weight gain and food consumption
(Ema et al. 2003)
greater than 1 million
Adolescents 12-19a years: dust ingestion, oral less than 0.001
(0.0011)
NOAELMBzP = 167
anti-androgenic effects in utero
LOAELMBzP = 167
decrease in body weight gain and food consumption
(Ema et al. 2003)
greater than 1 million

a. MOEs were calculated for non-pregnant individuals (male and female) and pregnant females for this age group.
b. Margin of exposure: central tendancy and (upper bounding).

B84P

Table 9-55 provides all relevant exposure and hazard values for B84P, as well as resultant MOEs, for determination of risk, which were previously described in the MCP SOS (Environment Canada, Health Canada 2015b). Overall, the MOEs for B84P are considered to be adequate to account for uncertainties in the exposure and health effect databases. From the information available, there is evidence that B84P has effects on the developing male reproductive system, indicative of RPS, which suggests that B84P has a common mode of action with other phthalates in the grouping.

Table 9-55. Summary of MOEs to B84P for relevant subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
Levels and basis for oral NOAEL
(mg/kg-bw/day)
MOEd
Infants (0-18 months): exposure to plastic articles, dermal 2.7c
(21.6)
NOAEL (BBP)= 151b
Histopathological changes in the pancreas, gross pathological alterations in the liver and significant increase in relative kidney weight in male rats at next highest dose of 381 mg/kg-bw/day (subchronic) (NTP 1997)
56 000
(6990)
Infants 0-6 months: dust ingestion, oral 0.0063
(0.047)
NOAEL (BBP)= 151b
Histopathological changes in the pancreas, gross pathological alterations in the liver and significant increase in relative kidney weight in male rats at next highest dose of 381 mg/kg-bw/day (subchronic) (NTP 1997)
greater than 1 million
Adults (20+): exposure to plastic articles, dermal 2.7c
(8.5)
NOAEL (BBP) = 50
Decreased pup body weight (male and female) and ↓ AGD at birth in F2 males at next highest dose of 100 mg/kg-bw/day;
decreased fetal testosterone (Aso et al. 2005; Nagao et al. 2000; Tyl et al. 2004; Furr et al. 2014)
19 000
(5900)

a. MOEs were calculated for non-pregnant individuals (male and female) and pregnant females for these age groups.
b. NOAEL (BBP prepubertal) = 300 (testicular pathology at 500 mg/kg-bw/day [7d]) is at higher doses than the systemic effects.
c. Estimated lower-end exposure, adjusted for dermal absorption (10%).
d. Margin of exposure: central tendancy and (upper bounding).

DIHepP

Table 9-56 provides all relevant exposure and hazard values for DIHepP, as well as resultant MOEs, for determination of risk, which were previously described in the MCP SOS (Environment Canada, Health Canada 2015b). Overall, the MOEs for DIHepP are considered to be adequate to account for uncertainties in the exposure and health effect databases. From the information available, there is evidence that DIHepP has effects on the developing male reproductive system, indicative of RPS, which suggests that DIHepP has a common mode of action with other phthalates in the grouping.

Table 9-56. Summary of MOEs to DIHepP for relevant subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
Level and basis for oral NOAEL
(mg/kg-bw/day)
MOEb
Infants 0-6 months: dust ingestion, oral 0.096
(1.1)
NOAEL = 50-162
Increased liver and kidney weights with histopathological findings at 222-716 mg/kg-bw/day
(McKee et al. 2006)
520 000 - greater than 1 million
(45 000-150 000)
Adolescents 12-19a years: dust ingestion, oral 0.0011
(0.013)
NOAEL = 50-168
Significant reduction in AGD and body weight in male F2 pups after in utero exposure to DIHepP at the next highest dose tested in rats (309-750 mg/kg-bw/day) and liver and kidney effects at the next highest dose
(227-750 mg/kg-bw/day) in F1 rats (McKee et al. 2006)
greater than 1 million

a. MOEs were calculated for non-pregnant individuals (male and female) and pregnant females for this age group.
b. Margin of exposure: central tendancy and (upper bounding)

B79P

Table 9-57 provides all relevant exposure and hazard values for B79P, as well as resultant MOEs, for determination of risk. Overall, the MOEs for B79P are considered to be adequate to account for uncertainties in the exposure and health effect databases. From the information available, there is evidence that B79P has effects on the developing male reproductive system, indicative of RPS, which suggests that B79P has a common mode of action with other phthalates in the grouping.

Table 9-57. Summary of MOEs to B79P for relevant subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
MOEb based on an oral NOAELDINP of 15 mg/kg-bw/day from Lington et al. 1997
Infants (0-18 months): exposure to plastic articles, dermal 2.7a
(21.6)
5600
(690)
Adults (20+): contact with plastic articles, dermal 2.7a
(8.5)
5600
(1800)
Infants 0-6 months: dust ingestion, oral 0.0063
(0.047)
greater than 1 million
(319 149)
Adolescents 12-19a years: dust ingestion, oral less than 0.001 greater than 1 million

a. Estimated lower-end exposure.
b. Margin of exposure: central tendancy and (upper bounding)

DINP

Table 9-58 provides all relevant exposure and hazard values for DINP, as well as resultant MOEs, for determination of risk. Overall, the MOEs for DINP are considered to be adequate to account for uncertainties in the exposure and health effect databases. From the information available, there is evidence that DINP has effects on the developing male reproductive system, indicative of RPS, which suggests that DINP has a common mode of action with other phthalates in the grouping.

Table 9-58. Summary of MOEs to DINP for subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
Level and basis for oral NOAEL
(mg/kg-bw/day)
MOEd
Children (females) 6-11 years: biomonitoring, 95thpercentile, NHANESb 3.8
(26)
NOAEL = 15
Increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs at 152-184 (Lington et al. 1997)
4000
(580)
Infants/children 6 months to 4 years: food and dust, oral 1.8
(19.7)
NOAEL = 15
Increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs at 152-184 (Lington et al. 1997)
8300
(760)
Infants (0 to 18 months): mouthing plastic toys and articles, oral 30c
(120)
NOAEL = 15
Increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs at 152-184 (Lington et al. 1997)
500
(125)e
Infants (0 to 18 months): exposure to plastic articles, dermal 1.1c
(8.6)
NOAEL = 15
Increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs at 152-184 (Lington et al. 1997)
14 000
(1700)
Adults (females) 20+ years: biomonitoring: 95thpercentile, NHANESb 2.3
(23)
LOEL/NOAEL = 10-15
↑ MNGs, ↑ Leydig cell clusters/aggregation starting from 100 mg/kg-bw/day after in utero exposure in GD12-21 (Li et al. 2015b),
increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs at 152-184 (Lington et al. 1997)
4300 to 6500
(430 to 650)
Adults (males) 20+a years: biomonitoring, 95th percentile, NHANESb 2.8
(24)
NOAEL = 15
Increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs at 152-184 (Lington et al. 1997)
5400
(630)
Adolescents 12 to 19 years: food and dust, oral 1.0
(11.4)
LOEL/NOAEL = 10-15
↑ MNGs, ↑ Leydig cell clusters/aggregation starting from 100 mg/kg-bw/day after in utero exposure in GD12-21 (Li et al. 2015b),
increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs at 152-184 (Lington et al. 1997)
10 000-15 000
(880-1300)
Adults (females) 20+ayears: exposure to plastic articles, dermal 1.1c
(3.4)
LOEL/NOAEL = 10-15
↑ MNGs, ↑ Leydig cell clusters/aggregation starting from 100 mg/kg-bw/day after in utero exposure in GD12-21 (Li et al. 2015b),
increase in liver and kidney weights, increased peroxisomal enzyme levels and histological changes in both organs at 152-184 (Lington et al. 1997)
9100-14 000
(2900-4400)

a. MOEs were calculated for non-pregnant individuals (male and female) and pregnant females for this age group.
b. The highest intakes at the 95th percentile (33 μg/kg bw/day:12-to-19-year-old males and 27 μg/kg/day: 12-to-19-year old females) were not brought forward to risk characterization because the relative standard error of the data was greater than 30%. For children aged 6 to 11 years, 26 μg/kg bw/d (RSE greater than 30%) was carried forward to risk characterization in order to be protective of this age group and because of the absence of low variability data, at the upper percentiles, for another comparable age group. For more details, see Environment Canada, Health Canada 2015c.
c. Estimated lower end exposure.
d. Margin of exposure: central tendancy and (upper bounding)
e. Migration rates used to estimate exposure were based on concentrations in toys (12.9 - 77%) that are higher than concentrations observed in recent Health Canada surveys (see Table 9-3, Environment Canada, Health Canada 2015c). Currently, Canada (like the United States and the European Union) have regulations (0.1%) in place limiting the amount of certain phthalates (including DINP) in toys and childcare articles.

CHIBP, BCHP and BIOP

An examination of the potential developmental and reproductive toxicity of CHIBP, BCHP and BIOP using appropriate analogues for read-across revealed that these medium-chain phthalates have the potential to have significant effects on the developing male, in addition to systemic effects (liver, kidney).

On the basis of the information available, it can be concluded that CHIBP, BCHP and BIOP meet the criteria for inclusion in the evaluation of the potential cumulative risk of phthalates on the developing male reproductive system given the evidence of the effects of their analogues; however, there is no exposure at this time. Consequently, the risk to human health for these substances is not expected.

Although the above MOEs for the medium-chain phthalates in the Phthalate Substance Grouping described in this section are considered adequate on an individual basis, this does not address the potential risk of concurrent exposure to these substances and other phthalates exhibiting a similar mode of action. Hence, all 10 medium-chain phthalates in the Phthalate Substance Grouping will be included for risk characterization in a cumulative context.

9.3.2.2 Cumulative risk assessment

The human health approach for a CRA for this Grouping has been described in detail in the "Proposed approach for cumulative risk assessment of certain phthalates under the Chemicals Management Plan" document (Environment Canada, Health Canada 2015e).

Human health-focused approaches for quantifying the cumulative risk of phthalates have been conducted by several national organizations including the Australian Department of Health (AGDH 2012, 2013, 2014a,b), the Danish Environmental Protection Agency (Danish EPA 2011), and the recently completed assessment by the United States Chronic Health Advisory Panel (US CPSC CHAP 2014). This draft screening assessment uses a tiered approach, following the schematic of the World Health Organization (WHO) and the International Program on Chemical Safety (IPCS) Framework for Risk Assessment of Combined Exposures to Multiple Chemicals (WHO 2009; Meek et al. 2011). This framework exercises the default assumption that the substances being assessed act by dose addition and the evaluation of cumulative risk involves multiple tiers where each higher level is increasingly dependent on additional data. The lower tier begins with simple assumptions and/or surrogate data for both hazard and exposure and refinement to a higher tier occurs if required and as the data allows.

For exposure characterization, if there is sufficient evidence of co-occurrence, then a CRA can be considered. The exposure approach is described in the CRA approach document (Environment Canada, Health Canada 2015e).

In summary, the pivotal information that determined substances for inclusion into the CRA were industry data collected under section 71 of CEPA (Environment Canada 2014), detection in North American biomonitoring surveys (CHMS, MIREC, MIREC-CD Plus, P4, NHANEs; Health Canada 2011b; Health Canada 2013; personal communication from EHSRD, Health Canada, to ESRAB, Health Canada, October 2013, 2014, unreferenced; Arbuckle et al. 2014; CDC 2014), and detection in household dust (CHDS; Kubwabo et al. 2013). For phthalate parent compounds (i.e., DEHP, DIBP, DBP, BBP, and DINP), with close to a 100% detection in various biomonitoring surveys, there was sufficient evidence for co-exposure; these substances were therefore assessed for cumulative risk.

A significant number of phthalates within the medium-chain phthalates subgroup have not been monitored in biomonitoring samples, but are found in commerce in Canada. These substances (i.e., DIHepP, B79P, B84P, DCHP, and DIOP) were included in the assessment of cumulative risk on the basis of their in-commerce status coupled with close to 100% detection in dust samples from Canadian homes.

Finally, given reporting limits and difficulties in determining import activity of substances, the CEPA section 71 survey may not have captured all in-commerce activity. As a result, substances that fit the profile of non-reporting to section 71 and close to 100% detection in house dust samples were also assessed for cumulative risk (i.e., DMCHP, DBzP, and DnHP).

There were three populations of interest for which exposure estimates were considered for cumulative risk: pregnant women and women of childbearing age (characterized as women aged twelve and up), infants (covered by age groups 0 to 6 months, 6 months to 4 years and 3 to 5 yearsFootnote 14), and children (covered by age groups 6 months to 4 years and 5 to 11 years). Adolescents and adult males were covered by this approach as they had generally lower estimates of phthalate exposure and are considered less sensitive to the reproductive effects of medium-chain phthalates compared to younger males (i.e. children) (NAS 2008).

In terms of exposure estimates considered for cumulative risk, biomonitoring data, which generally captures all potential exposure sources and routes (environmental media, food and products available to consumers) was considered a primary source and estimated monitoring data (chronic exposures resulting from environmental media and food) was considered as a supporting source of information. The US CPSC CHAP (2014) also used biomonitoring data as the primary source of exposure in their CRA. Regardless of route of exposure, phthalates, in general, are not considered acute toxicants, with LD50 levels from dermal exposure being at minimum two- to five-fold higher than oral values, which in turn are also high (Draize et al. 1948; David et al. 2001; Monsanto Company 1970 cited in US EPA 2006, 2010). Since phthalates metabolize relatively quickly showing no accumulation, and excretion is rapid, within hours to days (Phokha et al. 2002; Clewell et al. 2009), acute exposures were not considered relevant for a CRA. It should also be noted that no other jurisdictions have addressed phthalate exposures and risk from acute, one-time, exposures (AGDH 2011; ECHA 2013b; US CPSC CHAP 2014).

Upper-bound exposure estimates were used to estimate individual phthalate levels in the CRA to account for the uncertainties associated with the exposure data. The upper-bound exposure estimates used to estimate cumulative risk for relevant populations for the 16 medium-chain phthalates are summarized in Appendix F (Tables F-1, F-2, F-3 and F-4).

On the basis of the available information on the common adverse effects (RPS) and the observed differences in potencies within the medium-chain phthalates, a lower-tiered hazard characterization using the hazard index (HI) was considered to be the most appropriate approach. The HI method was selected because it offers the benefit of being simple and flexible and allows for an indication of which substance or substances in the CRA, or which source and route, may be the predominant contributor(s) to the overall risk. Identification of the substances or sources and routes that are drivers of the CRA is beneficial for informing risk management.

An HI is the summation of the individual hazard quotients (HQs) for each individual substance to determine the overall cumulative risk of the substance group of interest. The HQ of each substance is the ratio of exposure to a reference value (RfV) which is calculated by dividing the critical effect level identified in the hazard database by a defined uncertainty factor [UF]). The HI equation is as follows: HI = ∑HQ = ∑ (exposure/RfV). HI values of the medium-chain phthalates are calculated for the three subpopulations that represented the highest exposure groups.

Critical effect levels have been established at three life stages (i.e., in utero, prepubertal/pubertal, and adult) because of different sensitivity to the adverse effects of phthalates on development and reproduction at different life stages. The critical effect levels identified for the in utero life stage were used to calculate the HI value for pregnant woman and for infants. The critical effect levels identified for the prepubertal/pubertal life stage were used to calculate the HI value for children. The critical effect levels of the in utero and prepubertal/pubertal life stages for the medium-chain phthalates as well as the calculated RfVs are summarized in Appendix F (Table F-5 and F-6).

Given the fact that medium-chain phthalates have similar physical-chemical properties, have similar toxicological effects, and show an overall similarity in strength of their health effect databases (especially in regards to in utero exposure), the same default UF for the two corresponding subpopulations was used to calculate HQ values for each phthalate. A similar approach was adopted by another jurisdiction (US CPSC CHAP 2014). For the calculation of the total HI for pregnant women/women of childbearing age and infants, an UF of 100 (10 for intraspecies and 10 for interspecies differences) was used to calculate the RfV for the critical effect levels identified at the in utero life stage. For children, a default UF of 300 was used to calculate the RfV for the critical effects levels identified for the prepubertal-pubertal life stage. An additional UF of 3 was applied in this case on the basis of the limitations of the health effects database for the prepubertal life stage (quality and quantity of the studies currently available), and taking into consideration the variability in exposure duration in the different studies as well as the possibility that animals might also have been exposed to medium-chain phthalates in utero. The HI values for pregnant women/women of childbearing age, infants, and children are presented in Table 9-59 (also see Appendix F; tables F-7, F-8 and F-9).

Table 9-59. Hazard index values for subpopulations with the highest exposure
Population of interest Calculated HI with exposure estimates based on biomonitoring (upper bound) Calculated HI with exposure estimates based on environmental media and food (upper bound)
HI for pregnant women and women of childbearing age 0.24 0.23
HI for infants 0.37 0.82
HI for children 0.54 0.61

Individual phthalates that had the highest contribution to the cumulative risk were identified. Tables F-7, F-8 and F-9 (in Appendix F) show that the same three phthalate compounds (DINP, DBP, DEHP) resulted in the majority of cumulative risk, regardless of the age group or source of exposure data (biomonitoring or environmental media and food). Biomonitoring exposure estimates were generally considered more representative of potential exposures, including products available to consumers (regardless of source, route or duration). As a result, the HI values calculated using biomonitoring were considered more realistic but still conservative, because they were calculated using upper-bound exposure estimates.

Children and infants had higher HI values compared to pregnant women and women of childbearing age (aged 12 years and over). HI values calculated using biomonitoring data were lower than those calculated using environmental and dietary monitoring data for infants and children. In the case of DEHP, the high exposures from food were expected to be as a result of the unexpected presence of DEHP in fruits and vegetables which would have overestimated actual dietary exposure to DEHP. Nonetheless, this conservative, lower-tiered HI approach indicated no concern for potential cumulative risk of medium-chain phthalates for the general Canadian population, specifically the more sensitive subpopulations (pregnant women/women of childbearing age, infants, and children) at current exposure levels.

An HI value greater than 1 would indicate the need for further investigation or refinement. The HI values for the three subpopulations with the highest exposures were all below 1. Therefore, further refinement to a higher-tiered assessment is not necessary at this time. While risk is low at current levels for the cumulative risk of medium-chain phthalates (Phthalate Substance Grouping: DIBP, CHIBP, BCHP, DCHP, DBzP, B79P, DMCHP, DIHepP, BIOP, B84P, DINP; Additional phthalates: DPrP, DBP, BBP, DnHP, 79P, DIOP, DEHP), an increase in exposure levels could represent a potential risk to human health.

9.3.3 Long-chain phthalates

DIDP

Table 9-60 provides all relevant exposure and hazard values for DIDP, as well as resultant MOEs, for determination of risk, which were previously described in the LCP SOS (Environment Canada, Health Canada 2015d). Overall, the MOEs for DIDP are considered to be adequate to account for uncertainties in the exposure and health effect databases.

Table 9-60. Summary of MOEs to DIDP for subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
MOEc based on an oral LOAEL of 22 mg/kg-bw/day from Cho et al. 2008
Children (males) 6-11 years: biomonitoring, mean
(95th percentile), NHANES
1.4
(4.4)
16 000
(5000)
Infants (0-18 months)a: Exposure to plastic articles, dermal 0.27b
(2.16)
81 000
(10 000)
Children 6 months to 4 years: food and dust, oral 0.514
(2.87)
43 000
(7700)
Adolescents 12-19 years: food and dust, oral 0.075
(0.726)
290 000
(30 000)
Adults (males) 20+ years: biomonitoring, mean
(95th percentile), NHANES
0.76
(4.4)
29 000
(5000)
Adults (females) 20+ years: biomonitoring, mean
(95th percentile), NHANES
0.65
(4.9)
34 000
(4500)
Adults 20-59 years: food and dust, oral 0.068
(0.715)
320 000
(31 000)
Adults 20+a years: exposure to plastic articles, dermal 0.27b
(0.85)
81 000
(26 000)

a. Estimate adjusted on the basis of 1% dermal absorption of DIDP.
b. Estimated lower end exposure.
c. Margin of exposure: central tendancy and (upper bounding)

DUP

Table 9-61 provides all relevant exposure and hazard values for DUP, as well as resultant MOEs, for determination of risk, which were previously described in the LCP SOS (Environment Canada, Health Canada 2015d). Overall, the MOEs for DUP are considered to be adequate to account for uncertainties in the exposure and health effect databases.

Table 9-61. Summary of MOEs to DUP for subpopulations with highest exposure
Age group and exposure scenario Central tendency (upper bounding) estimate of exposure
(μg/kg bw/day)
MOEc based on an oral NOAEL of 37 mg/kg-bw/day from Poon et al. 1997
(DnOP)
Infants 0-6 months: dust, oral 0.0198
(0.349)
greater than 1 million
(110 000)
Infants (0-18 months)a:
exposure to plastic articles, dermal
2.7b
(21.6)
14 000
(1700)
Adolescents/Adults 12-19 years: dust, oral less than 0.001
(0.004)
greater than 1 million
Adults 20+a years: exposure to plastic articles, dermal 2.7b
(8.5)
14 000
(4400)

a. Estimate adjusted on the basis of 10% dermal absorption as default.
b. Estimated lower end exposure.
c. Margin of exposure: central tendancy and (upper bounding)

9.4 Uncertainties in evaluation of cumulative risk to human health

Uncertainties specific to short-chain, medium-chain, long-chain phthalates and DINP are summarized in the SOS reports (Environment Canada, Health Canada 2015b-e).

The key sources of uncertainty related to the CRA are presented in the table below.

Table 9-62. Sources of uncertainty in the cumulative risk characterization
Key source of uncertainty Impact
Data availability (multiple species, both sexes, sensitive exposure periods) and data quality for certain phthalates +/-
The unknown relevance of the available human epidemiological data studies implicating the potential hazard that certain phthalates pose to humans +
The inherent limitations in the use of biomonitoring data for risk characterization related to methods, the chemical-specific variability in levels and metabolites as well as lack of availability of data for certain phthalates +/-
The exclusion of estimates of exposure for products available to consumers in the CRA of phthalates, even if biomonitoring estimates would likely capture all exposure sources/routes, including exposure from products available to consumers -
The potential toxico*kinetic or toxicodynamic differences between species and between the individual chemicals +/-
The application of default uncertainty factors for a specific life stage even if some databases are more robust than others as well as the use of life-stage specific studies to determine HIs for children exposed after birth (prepubertal database less robust) +
The overall cumulative risk of phthalates based on other adverse effects observed after exposure to this grouping as a whole regardless of chain length -

+ = uncertainty with potential to cause over-estimation of exposure/risk, - = uncertainty with potential to cause under-estimation of exposure risk, +/- = unknown potential to cause over or under estimation of risk.
Considering the above sources of uncertainty, it is anticipated that the cumulative risk characterization of this grouping would not be sensitive to refinement at this time if additional data were provided, as the lower-tiered HI approach with several conservative assumptions indicated no concern for human health.

10. Conclusion

Considering all available lines of evidence presented in this draft screening assessment, there is low risk of harm to organisms and the broader integrity of the environment from 13 of the phthalates in the Phthalate Substance Grouping (DMP, DIBP, CHIBP, BCHP, DCHP, DBzP, DMCHP, DIHepP, BIOP, B84P, DINP, DIDP and DUP). However, there is risk of harm to organisms, but not to the broader integrity of the environment, from 1 phthalate included in the Phthalate Substance Grouping, B79P, and 1 additional phthalate, DEHP. DEHP was previously assessed by Environment Canada and Health Canada in 1994 under the Priority Substances Assessment Program. The assessment concluded that DEHP posed a risk to human health in Canada. However, a conclusion for the environment could not be determined because of insufficient information.

It is proposed to conclude that 13 substances in the Phthalate Substance Grouping do not meet the criteria under paragraphs 64(a) or (b) of CEPA as they are not entering the environment in a quantity or concentration or under conditions that have or may have an immediate or long-term harmful effect on the environment or its biological diversity or that constitute or may constitute a danger to the environment on which life depends. It is proposed to conclude that B79P and DEHP meet the criteria under paragraph 64(a) of CEPA as they are entering or may enter the environment in a quantity or concentration or under conditions that have or may have an immediate or long-term harmful effect on the environment or its biological diversity. However, it is proposed to conclude that B79P and DEHP do not meet the criteria under paragraph 64(b) of CEPA as they are not entering the environment in a quantity or concentration or under conditions that constitute or may constitute a danger to the environment on which life depends.

On the basis of the information presented in this draft screening assessment, it is proposed that the 14 phthalates in the Phthalate Substance Grouping do not meet the criteria under paragraph 64(c) of CEPA as they are not entering the environment in a quantity or concentration or under conditions that constitute or may constitute a danger in Canada to human life or health.

Therefore, it is proposed to conclude that B79P and DEHP meet one or more of the criteria set out in section 64 of CEPA. B79P and DEHP have been determined to not meet the persistence and bioaccumulation criteria as set out in the Persistence and Bioaccumulation Regulations of CEPA.

References

Abd-Ellah MF, Aly HA, Mokhlis HA, Abdel-Aziz AH. 2016. Quercetin attenuates di-(2-ethylhexyl) phthalate-induced testicular toxicity in adult rats. Hum Exp Toxicol. 35(3):232-243.

[ACC] American Chemistry Council. 2006. High Production Volume (HPV) Chemical Challenge Program: Test Plan for the Phthalate Ester Category [Internet]. Phthalate Esters Panel. (HPV Testing Group). Report No.: 201-16554A.

ACD/Percepta[prediction module]. 2012. Toronto (ON): Advanced Chemistry Development.

Adams WJ, Biddinger GR, Robillard KA, Gorsuch JW. 1995. A summary of the acute toxicity of 14 phthalate esters to representative aquatic organisms. Environ Toxicol Chem. 14(9):1569-1574.

Adeleke AA, Okedeyi OO, Yusuf KA. 2011. Flame ionization gas chromatographic determination of phthalate esters in water, surface sediments and fish species in the Ogun river catchments, Ketu, Lagos, Nigeria. Environ Monit Assess. 172:561-569.

[AFN] The Assembly of First Nations. 2013. First Nations Biomonitoring Initiative. National Results (2011). June 2013. [accessed 2014 Feb 26].

[AGDH] Australian Government Department of Health. 2008. Phthalates Hazard Compendium. A summary of physiochemical and human health hazard data for 24 ortho-phthalate chemicals. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS).

[AGDH] Australian Government Department of Health. 2008a. Dimethyl phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Existing Chemical Hazard Assessment Report.

[AGDH] Australian Government Department of Health. 2008b. Butylbenzyl Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Existing Chemical Hazard Assessment Report.

[AGDH] Australian Government Department of Health. 2008c. Diisononyl phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Existing Chemical Hazard Assessment Report.

[AGDH] Australian Government Department of Health. 2008d. Diisoheptyl Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Existing Chemical Hazard Assessment Report.

[AGDH] Australian Government Department of Health. 2008e. Diisobutyl Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Existing Chemical Hazard Assessment Report.

[AGDH] Australian Government Department of Health. 2008f. Diisodecyl phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Existing chemical hazard assessment report.

[AGDH] Australian Government Department of Health. 2010. Diethylhexyl Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Priority Existing Chemical (PEC) Assessment Report No.32.

[AGDH] Australian Government Department of Health. 2012. Diisononyl Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Priority Existing Chemical (PEC) Assessment Report No. 35.

[AGDH] Australian Government Department of Health. 2013a. Dibutyl Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Priority Existing Chemical (PEC) Assessment Report.

[AGDH] Australian Government Department of Health. 2013b. Butyl Benzyl Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Priority Existing Chemical (PEC) Assessment Report No. 40.

[AGDH] Australian Government Department of Health. 2014a. Dimethyl Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Priority Existing Chemical (PEC) Assessment Report No. 37.

[AGDH] Australian Government Department of Health. 2014b. Di(methoxyethyl) Phthalate. Sydney (AU): Department of Health, National Industrial Chemicals Notification and Assessment Scheme (NICNAS). Priority Existing Chemical (PEC) Assessment Report No. 38.

Ahbab MA, Barlas N. 2013. Developmental effects of prenatal di-n-hexyl phthalate and dicyclohexyl phthalate exposure on reproductive tract of male rats: Postnatal outcomes. Toxicol Lett. 51:123-136.

Ahbab MA, Undeger U, Barlas N, Basaran N. 2014. In utero exposure to dicyclohexyl and di-n-hexyl phthalate possess genotoxic effects on testicular cells of male rats after birth in the comet and TUNEL assays. Hum Exp Toxicol. 33(3):230-239.

Ahbab MA, Barlas N. 2015. Influence of in utero di-n-hexyl phthalate and dicyclohexyl phthalate on fetal testicular development in rats. Toxicol Lett. 233:125-137.

Ahmad R, Gautam AK, Verma Y, Sedha S, Kumar S. 2014. Effects of in utero di-butyl phthalate and butyl benzyl phthalate exposure on offspring development and male reproduction of rat. Environ Sci Pollut Res Int. 21(4):3156-3165.

Aly HA, Hassan MH, El-Beshbishy HA, Alahdal AM, Osman AM. 2015. Dibutyl phthalate induces oxidative stress and impairs spermatogenesis in adult rat. Toxicol Ind Health. 32(8).

Andrade AJM, Grande SW, Talsness CE, Gericke C, Grote K, Golombiewsky A, Sterner-Kock A, Chahoud I. 2006a. A dose response study following in utero and lactational exposure to di-(2-ethylhexyl) phthalate (DEHP): Reproductive effects on adult male offspring rats. Toxicology. 228:85-97.

Andrade AJM, Grande SW, Talsness CE, Grote K, Golombiewsky A, Sterner-Kock A, Chahoud I. 2006b. A dose-response study following in utero and lactational exposure to di-(2-ethylhexyl) phthalate (DEHP): effects on androgenic status, developmental landmarks and testicular histology in male offspring rats. Toxicology. 225(1):64-74.

Aoki KA. 2010. A study of the anti-androgenic effects of the phthalate ester, din-butyl phthalate, on two freshwater fish species, the fathead minnow and the three-spined stickleback. PhD thesis. Brunel University, Institute for the Environment. London (UK): Brunel University.

Aoki KAA, Harris CA, Katsiadaki I, Sumpter JP. 2011. Evidence suggesting that di-N-butyl phthalate has antiandrogenic effects in fish. Environ Toxicol Chem. 30(6):1338-1345.

Araki A, Mitsui T, Miyash*ta C, Nakajima T, Naito H, Ito S, Sasaki S, Cho K, Ikeno T, Nonomura K, Kishi R. 2014. Association between maternal exposure to di(2-ethylhexyl) phthalate and reproductive hormone levels in fetal blood: The Hokkaido Study on Environment and Children's Health. PLOS ONE. 9(10):1-10.

Arbuckle TE, Davis K, Marro L, Fisher M, Legrand M, LeBlanc A, Gaudreau E, Foster WG, Choeurng V, Fraser WD, the MIREC Study Group. 2014. Phthalate and bisphenol A exposure among pregnant women in Canada - results from the MIREC study. Environ Int. 68:55-65.

Aso S, Ehara H, Miyata K, Hosyuyama S, Shiraishi K, Umano T, Minobe Y. 2005. A two-generation reproductive toxicity study of butyl benzyl phthalate in rats. J Toxicol Sci. 30:39-58.

[ASTER] Assessment Tools for the Evaluation of Risk. 1999. Duluth (MN): US Environmental Protection Agency, Mid-Continent Ecology Division. [restricted access].

Ateşşahin A, urk GT, Karahan I, Yilmaz S, Ceribasi AO, Bulmus O. 2006. Lycopene prevents adriamycin-induced testicular toxicity in rats. Fertil Steril. 85(1):1216-1222.

Axelsson J, Rylander L, Rignell-Hydbom A, Lindh CH, Jönsson BAG, Giwercman A. 2015a. Prenatal phthalate exposure and reproductive function in young men. Environ Res. 138:264-270.

Axelsson J, Rylander L, Rignell-Hydbom A, Jönsson BAG, Lindh CH, Giwercman A. 2015b. Phthalate exposure and reproductive parameters in young men from the general Swedish population. Environ Int. 185:54-60.

Bao J, Wang M, Ning X, Zhou Y, He Y, Yang J, Gao X, Li S, Ding Z, Chen B. 2015. Phthalate concentrations in personal care products and the cumulative exposure to female adults and infants in Shanghai. J Toxicol Environ Health A. 78:325-341.

Barber ED, Astill BD, Moran EJ, Schneider BF, Gray TJ, Lake BG, Evans JG. 1987. Peroxisome induction studies on seven phthalate esters. Toxicol Ind. Health. 3(2):7-24.

Bavarian State Ministry of the Environment and Public Health. 2012. Submission ofinformation via the public consultation on ECHA's draft review report by the Bavarian State Ministry of the Environment and Public Health (Bayerisches Staatsministerium für Umwelt und Gesundheit). Comment reference number 19.

Bergh C, Torgrip R, Emenius G, Östman C. 2011a. Organophosphate and phthalate esters in air and settled dust - a multi-location indoor study. Indoor Air. 21:67-76.

Bergh C, Aberg K, Svartengren M, Emenius G, Östman C. 2011b. Organophosphate and phthalate esters in indoor air: a comparison between multi-storey buildings with high and low prevalence of sick building symptoms. J Environ Monit. 13:2001-2009.

Bergh C, Luongo G, Wise S, Östman C. 2012. Organophosphate and phthalate esters in standard reference material 2585 organic contaminants in house dust. Anal Bioanal Chem. 402:51-59.

[BIBRA] British Industrial Biological Research Association. 1986. A 21 Day Feeding Study of Di-isodecyl Phthalate to Rats: Effects on the Liver and Liver Lipids. Report No. 0495/5/85 [cited in ECJRC 2003].

Biedermann-Brem S, Biedermann M, Pfenninger S, Bauer M, Altkofer W, Rieger K, Hauri U, Droz C, Grob K. 2008. Plasticizers in PVC toys and childcare products: What succeeds the phthalates? Market survey 2007. Chromatographia. 68:227-234.

Blanchard O, Glorennec P, Mercier F, Bonvallot N, Chevrier C, Ramalho O, Mandin C, Le Bot B. 2014. Semivolatile organic compounds in indoor air and settled dust in 30 French dwellings. Environ Sci Technol. 48:3959-3969.

Blystone CR, Kissling GE, Bishop JB, Chapin RE, Wolfe GW, Foster PMD. 2010. Determination of the di-(2-ethylhexyl) phthalate NOAEL for reproductive development in the rat: importance of the retention of extra animals to adulthood. Toxicol Sci. 116(2):640-646.

Boekelheide K, Kleymenova E, Liu K, Swanson C, Gaido KW. 2009. Dose-dependent effects on cell proliferation, seminiferous tubules, and male germ cells in the fetal rat testis following exposure to di(n-butyl) phthalate. Microsc Res Tech. 72(8):629-638.

Bono-Blay F, Guart A, de la Fuente B, Pedemonte M, Pastor MC, Borrell A, Lacorte S. 2012. Survey of phthalates, alkylphenols, bisphenol A and herbicides in Spanish source waters intended for bottling. Environ Sci Pollut Res Int. 19(8):3339-3349. DOI 10.1007/s11356-012-0851-y.

Bornehag CG, Lundgren B, Weschler CH, Sigsgaard T, Hagerhed-Engman L, Sundell J. 2005. Phthalates in indoor dust and their association with building characteristics. Environ Health Perspect. 113:1399-1404.

Bradlee CA, Thomas P. 2003. Aquatic toxicity of phthalate esters. In: Staples CA, editor. The Handbook of Environmental Chemistry: Phthalate Esters. Springer-Verlag Berlin Heidelberg. Volume 3. Part Q. pp. 263-298.

Bradley EL, Burden RA, Leon I, Mortimer DN, Speck DR, Castle L. 2013a. Determination of phthalate diesters in foods. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 30(4):722-734.

Bradley EL, Burden RA, Bentayeb K, Driffield M, Harmer N, Mortimer DN, Speck DR, Ticha J, Castle L. 2013b. Exposure to phthalic acid, phthalate diesters and phthalate monoesters from foodstuffs: UK total diet study results. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 30(4):735-742.

Brigden K, Hetherington S, Wang M, Santillo D, Johnston P. 2013. Hazardous chemicals in branded textile products on sale in 25 countries/regions during 2013. Greenpeace Research Laboratories Technical Report 06/2013.

Brown D, Thompson RS, Stewart KM, Croudace CP, Gillings E. 1996. The effect of phthalate ester plasticisers on the emergence of the midge (Chironomus riparius) from treated sediments. Chemosphere. 32(11):2177-2187.

Buccafusco RJ, Ells SJ, LeBlanc GA. 1981. Acute toxicity of priority pollutants to bluegill (Lepomis macrochirus). Bull Environ Contam Toxicol. 26:446-452.

Call DJ, Markee TP, Geiger DL, Brooke LT, VandeVenter FA, Cox DA, Genisot KI, Robillard KA, Gorsuch JW, Parkerton TF, Reiley MC, Ankley GT, Mount DR. 2001a. An assessment of the toxicity of phthalate esters to freshwater benthos. 1. Aqueous exposures. Environ Toxicol Chem. 20(8):1798-1804.

Call DJ, Cox DA, Geiger DL, Genisot KI, Markee TP, Brooke LT, Polkinghorne CN, VandeVenter FA, Gorusuch JW, Robillard KA, Parkerton TF, Reiley MC, Ankley GT, Mount DDR. 2001b. An assessment of the toxicity of phthalate esters to freshwater benthos. 2. Sediment exposures. Environ Toxicol Chem. 20(8):1805-1815.

Cammack JN, White RD, Gordon D, Gass J, Hecker L, Conine D, Bruen US, Friedman M, Echols C, Yeh TY, Wilson DM. 2003. Evaluation of reproductive development following intravenous and oral exposure to DEHP in male neonatal rats. Int J Toxicol. 22:159-174.

Canada. 1999. Canadian Environmental Protection Act, 1999. S.C. 1999, c. 33. Canada Gazette, Part III, vol. 22, no. 3.

Canada. 2016. Phthalates Regulations. SOR/2016-188.

Canada. Department of the Environment. 2013. Canadian Environmental Protection Act, 1999: Notice with respect to certain phthalate substances. Canada Gazette, Part I, vol. 147, no. 28, p. 1801-1821.

Cao X. 2008. Determination of phthalates and adipate in bottled water by headspace solid-phase microextraction and gas chromatography/mass spectrometry. J Chromatogr A. 1178(1):231-238.

Cao X, Zhao W, Churchill R, Hilts C. 2014. Occurrence of di-(2-ethylhexyl) adipate and phthalate plasticizers in samples of meat, fish, and cheese and their packaging films. J Food Prot. 77(4):610-620.

Cao X, Zhao W, Dabeka R. 2015. Di-(2-ethylhexyl) adipate and 20 phthalates in composite food samples from the 2013 Canadian Total Diet Study. Food Addit Contam Part A Chem Anal Control Expo Risk Assess. 32(11):1893-1901. DOI:10.1080/19440049.2015.1079742.

Carnevali O, Tosti L, Speciale C, Peng C, Zhu Y, Maradonna F. 2010. DEHP impairs zebrafish reproduction by affecting critical factors in oogenesis. PLOS ONE. 5(4):1-5.

Casas M, Valvi D, Ballesteros-Gomez A, Gascon M, Fernández MF, Garcia-Esteban R, Iñiguez C, Martínez D, Murcia M, Monfort N, Luque N, Rubio S, Ventura R, Sunyer J, Vrijheid M. 2016. Exposure to bisphenol A and phthalates during pregnancy and ultrasound measures of fetal growth in the INMA-Sabadell cohort. Environ Health Perspect. 124(4):521-528.

[CDC] Centers for Disease Control and Prevention (US). National Center for Health Statistics (NCHS). 2013. National Health and Nutrition Examination Survey Data 1999-2010. Hyattsville, MD: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention.

[CDC] Centers for Disease Control and Prevention (US). National Center for Health Statistics (NCHS). 2014. National Health and Nutrition Examination Survey Data 1999-2012. Hyattsville, MD: U.S. Department of Health and Human Services, Centers for Disease Control and Prevention.

Chang BV, Yang CM, Cheng CH, Yuan SY. 2004. Biodegradation of phthalate esters by two bacteria strains. Chemosphere. 55(4):533-538.

Chang BV, Liao GS, Yuan SY. 2005. Anaerobic degradation of diethyl phthalate, di-n-butyl phthalate, and di-(2-ethylhexyl) phthalate from river sediment in Taiwan. Chemosphere. 58(11):1601-1607.

Chang JW, Yan BR, Chang MH, Tseng SH, Kao YM, Chen JC, Lee CC. 2014. Cumulative risk assessment for plasticizer-contaminated food using the hazard index approach. Environ Pollut. 189:77-84.

Chang W-H, Li S-S, Wu M-H, Pan H-A, Lee C-C. 2015. Phthalates might interfere with testicular function by reducing testosterone and insulin-like factor 3 levels. Human Reprod. 30(11):2658-2670.

Chao KP, Huang CS, Huang ML. 2013. Direct Extraction of phthalate esters from polymeric gloves materials. Adv Mater Res. 804:114-117.

Chen X, Xu S, Tan T, Lee ST, Cheng SH, Lee FWF, Xu SJL, Ho KC. 2014. Toxicity and estrogenic endocrine disrupting activity of phthalates and their mixtures. Int J Environ Res Public Health. 11(3):3156-3168.

Cheng X, Ma L, Xu D, Cheng H, Yang G, Luo M. 2015. Mapping of phthalate esters in suburban surface and deep soils around a metropolis-Beijing, China. J Geochem Explor. 155:56-61.

[CHMS] 2010. Canadian Health Measures Survey (CHMS) Data User Guide: Cycle 1. [accessed 2014].

[CHMS] 2012. Canadian Health Measures Survey (CHMS) Data User Guide: Cycle 2. [accessed 2014].

Cho WS, Han BS, Ahn B, Nam KT, Choi M, Oh SY, Kim SH, Jeong J, Jang DD. 2008. Peroxisome proliferator di-isodecyl phthalate has no carcinogenic potential in Fischer 344 rats. Toxicol Lett. 178:110-116.

Christensen KLY, Makris SL, Lorber M. 2014. Generation of hazard indices for cumulative exposure to phthalates for use in cumulative risk assessment. Regul Toxicol Pharmacol. 69(3):380-389.

Christiansen S, Boberg J, Axelstad M, Dalgaard M, Vinggaard AM, Metzdorff SB, Hass U. 2010. Low-dose perinatal exposure to di(2-ethylhexyl) phthalate induces anti-androgenic effects in male rats. Reprod Toxicol. 30:313-321.

Christiansen LB, Pedersen KL, Pedersen SN, Korsgaard B, Bierregaard P. 2000. In vivo comparison of xenoestrogens using rainbow trout vitellogenin induction as a screening system. Environ Toxicol Chem. 19(7):1867-1874.

Cirillo T, Latini G, Castaldi MA, Dipaola L, Fasano E, Esposito F, Scognamiglio G, Di Francesco F, Cobellis L. 2015. Exposure to di-2-ethylhexyl phthalate, di-N-butyl phthalate and bisphenol A through infant formulas. J Agric Food Chem. 63:3303-3310.

Clara M, Windhofer G, Hartl W, Braun K, Simon M, Gans O, Scheffknecht C, Chovanec A. 2010. Occurrence of phthalates in surface runoff, untreated and treated wastewater and fate during wastewater treatment. Chemosphere. 78:1078-1084.

Clewell RA, Kremer JJ, Williams CC, Campbell JL, Sochaski MA, Andersen ME, Borghoff SJ. 2009. Kinetics of selected di-n-butyl phthalate metabolites and fetal testosterone following repeated and single administration in pregnant rats. Toxicology. 255(1-2):80-90.

Clewell RA. 2011. Pharmaco*kinetics and foetal testes effects after diisononyl phthalate administration in rat gestation. The Hamner Protocol #09016 Final Report, DINP Phase I study. The Hamner Institutes for Life Sciences, Research Triangle Park, NC 27709-2137. Sponsored by ExxonMobil Biomedical Sciences Inc. [as cited in ECHA 2013].

Clewell RA, Sochaski M, Edwards K, Creasy DM, Willson G, Anderson ME. 2013. Disposition of diiosononyl phthalate and its effects on sexual development of the male fetus following repeated dosing in pregnant rats. Reprod Toxicol. 35:56-69.

[CMA] Chemical Manufacturers Association. 1984. Acute toxicity of thirteen phthalate esters to fathead minnow (Pimephales promelas) under flowthrough conditions. Springborn Bionomics Inc. report. Washington (DC): Chemical Manufacturers Association [cited in ECJRC 2004].

Corradetti B, Stronati A, Tosti L, Manicardi G, Carnevali O, Bizzaro D. 2013. Bis-(2-ethylhexyl) phthalate impairs spermatogenesis in zebrafish (Danio rerio). Reprod Biol. 13(3):195-202.

Cousins IT, Mackay D, Parkerton TF. 2003. Physical-chemical properties and evaluative fate modelling of phthalate esters. In: Hutzinger O, ed. The handbook of environmental chemistry. Volume 3. Anthropogenic compounds. Part Q. Berlin (DE): Springer-Verlag. pp. 263-298.

[Danish EPA] Danish Environmental Protection Agency. 2011. Annex XV: Restriction Report Proposal for a restriction. Danish EPA Copenhagen Version 2.

David RM. 2000. Exposure to phthalate esters. Environ Health Perspect. 108:A440.

David RM, Moore MR, Finney DC, Guest D. 2000a. Chronic toxicity of di(2-ethylhexyl) phthalate in tats. Toxicol Sci. 55:433-443.

David RM, Moore MR, Finney DC, Guest D. 2000b. Chronic toxicity of di(2-ethylhexyl) phthalate in mice. Toxicol Sci. 58:377-385.

David RM, McKee RH, Butala JH, Barter RA, Kayser M. 2001. Esters of aromatic mono-, di-, and tricarboxylic acids, aromatic diacids and di-, tri-, or polyalcohols. In: Patty's Toxicology. p. 80.

De co*ck M, de Boer MR, Lamoree M, Legler J, van de Bor M. 2014. First year growth in relation to prenatal exposure to endocrine disruptors - a Dutch prospective cohort study. Int J Environ Res Public Health. 11(7):7001-7021.

de Ryke D, Willems MI. 1977. Sub-chronic (90 day) toxicity study with dicyclohexyl phthalate in Albino rats. TNO Report R5228 (confidential) [as cited in MAFF 1987].

De Solla SR, Langlois VS. 2014. Toxicity and endocrine disruption of phthalates in fish and amphibians. SRAD Progress Report 2013-2014. 19 p.

Dodson RE, Camann DE, Morello-Frosch R, Brody JG, Rudel RA. 2015. Semivolatile organic compounds in homes: strategies for efficient and systematic exposure measurement based on empirical and theoretical factors. Environ Sci Technol. 49(1):113-122.

Dominguez-Morueco N, Gonzalex-Alonso S, Valcarcel Y. 2014. Phthalate occurrence in rivers and tap water from central Spain. Sci Total Environ. 500-501:139-146.

Dostal LA, Chapin RE, Stefansky SA, Harris MW, Schewetz BA. 1988. Testicular toxicity and reduced Sertoli cell numbers in neonatal rats by di(2-ethylhexyl) phthalate and the recovery of fertility as adults. Toxicol Appl Pharmacol. 95:104-121.

Downs SH, Black N. 1998. The feasibility of creating a checklist for the assessment of the methodological quality both of randomised and non-randomised studies of health care interventions. J Epidemiol Community Health. 52:377-84.

[DPD] Drug Product Database [database]. [modified 2014]. Ottawa (ON): Health Canada. [accessed 2014 Sep 9].

Draize J, Alvarez E, Whitesell M, Woodard G, Hagan E, Nelson A. 1948. Toxicological investigations of compounds proposed for use as insect repellents: A. local and systemic effects following topical skin application; B. acute oral toxicity C. pathological examination. J Pharmacol Exp Ther. 93(1):26-39.

[EC] European Commission. 2009. Addendum to Risk Assessment from 2007: Benzyl butyl phthalate: EC No: 201-662-7: CAS: 85-68-7. Rapporteur Member State Norway. 28 p.

[ECCC] Environment and Climate Change Canada. 2016. Supporting documentation: Information in support of the draft screening assessment for phthalates. Ottawa (ON): ECCC.

[ECCC, HC] Environment and Climate Change Canada, Health Canada. [modified 2007 Apr 20]. Categorization. Ottawa (ON): Government of Canada. [accessed 2014 Jun 10].

[ECHA] European Chemicals Agency. c2007-2014a. Registered substances database; search for CAS RN 84-69-5 [DIBP]. Helsinki (FI): ECHA. [accessed 2014 Sep].

[ECHA] European Chemicals Agency. c2007-2014b. Registered substances database; search for CAS RN 84-61-7 [DCHP]. Helsinki (FI): ECHA. [accessed 2014 Sep].

[ECHA] European Chemicals Agency. c2007-2014c. Registered substances database; search for CAS RN 68515-40-2 [B79P]. Helsinki (FI): ECHA. [accessed 2014 Sep].

[ECHA] European Chemicals Agency. c2007-2015. Registered substances database; search for CAS RN 85-68-7 [BBP]. Helsinki (FI): ECHA. [updated 2016 Mar 24; accessed 2016 Apr 4].

[ECHA] European Chemicals Agency. 2009. Addendum to risk assessment from 2007: benzyl butyl phthalate. Helsinki (FI): ECHA. Rapporteur Member State Norway. 28 p.

[ECHA] European Chemicals Agency. 2012a. Registration information on identified uses related to article categories. Search result of CASRN [84-74-2].

[ECHA] European Chemicals Agency. 2012b. Registration information on identified uses related to article categories. Search result of CASRN [117-81-7].

[ECHA] European Chemicals Agency. 2012c. Registration information on identified uses related to article categories. Search result of CASRN [84-75-3].

[ECHA] European Chemicals Agency. 2012d. Registration information on identified uses related to article categories. Search result of CASRN [27554-26-3].

[ECHA] European Chemicals Agency. 2013a. REACH Dossier on B79P.

[ECHA] European Chemicals Agency. 2013b. Evaluation of new scientific evidence concerning DINP and DIDP. Final Review Report.

[ECHA] ECHA European Chemicals Agency. 2015a. Substance name: Di-butyl phthalate. EC number: 201-557-4. CAS RN 84-74-2. Summary of Classification and Labelling. European Chemicals Agency.

[ECHA] ECHA European Chemicals Agency. 2015b. Substance name: Di-ethylhexyl phthalate. EC number: 204-211-0. CAS RN 117-81-7. Summary of Classification and Labelling. European Chemicals Agency.

[ECHA] ECHA European Chemicals Agency. 2015c. Substance name: Dihexyl phthalate. EC number: 201-559-5. CAS RN 84-75-3. Summary of Classification and Labelling. European Chemicals Agency.

[ECHA] ECHA European Chemicals Agency. 2015d. Substance name: Diisooctyl phthalate. EC number: 248-523-5. CAS RN 27554-26-3. Summary of Classification and Labelling. European Chemicals Agency.

[ECJRC] European Commission Joint Research Centre. 2003. European Union risk assessment report: CAS: 68515-49-1, 26761-40-0: 1,2-benzenedicarboxylic acid, di-C9-11-branched alkyl esters, C10-rich and di-"isodecyl" phthalate (DIDP). Report No.: EUR 20785 EN. Luxembourg: Office for Official Publications of the European Communities.

[ECJRC] European Commission Joint Research Centre. 2004. European Union risk assessment report: CAS No: 84-74-2: Dibutyl phthalate. Report No.: EUR 19840 EN. Luxembourg: Office for Official Publications of the European Communities.

[ECJRC] European Commission Joint Research Centre. 2008. European Union risk assessment report: CAS: 117-81-7: bis(2-ethylhexyl)phthalate (DEHP). Report No.: EUR 20785 EN. Luxembourg: Office for Official Publications of the European Communities.

[ECOSAR] ECOlogical Structure Activity Relationships Class Program [estimation model]. 2009. Ver. 1.00. Washington (DC): US Environmental Protection Agency, Office of Pollution Prevention and Toxics; Syracuse (NY): Syracuse Research Corporation.

[EFSA] European Food Safety Authority. 2005. Opinion of the scientific panel on food additives, flavourings, processing aids, and materials in contact with food (AFC) related to bis(2-ethylhexyl)phthalate (DEHP) for use in food contact materials. EFSA Journal 3(9):243, 20 pp. doi:10.2903/j.efsa.2005.243.

Ema M, Miyawaki E, Hirose A, Kamata E. 2003. Decreased anogenital distance and increased incidence of undescended testes in fetuses of rats given monobenzyl phthalate, a major metabolite of butyl benzyl phthalate. Reprod Toxicol. 17:407-412.

Engelhardt G, Tillmanns G, Wallnofer PR, Hutzinger O. 1977. Biodegradation of di-iso-butyl phthalate and related dialkyl phthalates by Penicillium lilacinum. Chemosphere. 6:347-354.

Environment Canada. 2014. Data for phthalate substances collected under the Canadian Environmental Protection Act, 1999, section 71: Notice with respect to certain phthalate substances. Data prepared by: Environment Canada, Existing Substances Program.

Environment Canada and Health Canada. 1993. Priority Substances List assessment report: di-n-octyl phthalate. Ottawa (ON): Environment Canada, Health Canada.

Environment Canada and Health Canada. 1994a. Priority Substances List assessment report: bis(2-ethylhexyl) phthalate. Ottawa (ON): Environment Canada, Health Canada.

Environment Canada and Health Canada. 1994b. Priority Substances List assessment report: dibutyl phthalate. Ottawa (ON): Environment Canada, Health Canada.

Environment Canada and Health Canada. 2000. Priority Substances List assessment report: butylbenzylphthalate. Ottawa (ON): Environment Canada, Health Canada.

Environment Canada, Health Canada. 2015a. State of the Science Report: the Phthalate Substance Grouping: Short-Chain Phthalate Ester: 1,2-Benzenedicarboxylic acid, dimethyl ester (DMP). Chemical Abstracts Service Registry Number: 131-11-3. Gatineau (QC): Environment Canada, Health Canada: Existing Substances Program.

Environment Canada, Health Canada. 2015b. State of the Science Report: the Phthalate Substance Grouping: Medium-Chain Phthalate Esters. Chemical Abstracts Service Registry Numbers: 84-61-7; 84-64-0; 84-69-5; 523-31-9; 5334-09-8;16883-83-3; 27215-22-1; 27987-25-3; 68515-40-2; 71888-89-6. Gatineau (QC): Environment Canada, Health Canada: Existing Substances Program.

Environment Canada, Health Canada. 2015c. State of the Science Report: the Phthalate Substance Grouping: 1,2-Benzenedicarboxylic acid, diisononyl ester; 1,2-Benzenedicarboxylic acid, di-C8-10-branched alkyl esters, C9-rich (DINP). Chemical Abstracts Service Registry Numbers: 28553-12-0, 68515-48-0. Gatineau (QC): Environment Canada, Health Canada: Existing Substances Program.

Environment Canada, Health Canada. 2015d. State of the Science Report: the Phthalate Substance Grouping: Long-Chain Phthalate Esters. 1,2-Benzenedicarboxylic acid, diisodecyl ester (diisodecyl phthalate; DIDP) and 1,2-Benzenedicarboxylic acid, diundecyl ester (diundecyl phthalate; DUP). Chemical Abstracts Service Registry Numbers: 26761-40-0, 68515-49-1; 3648-20-2. Gatineau (QC): Environment Canada, Health Canada: Existing Substances Program.

Environment Canada, Health Canada. 2015e. Proposed approach for cumulative risk assessment of certain phthalates under the Chemicals Management Plan. August 2015. Gatineau (QC): Health Canada, Environment Canada.

[EPI Suite] Estimation Programs Interface Suite for Microsoft Windows [estimation model]. 2010. Ver. 4.10. Washington (DC): US Environmental Protection Agency, Office of Pollution Prevention and Toxics; Syracuse (NY): Syracuse Research Corporation.

European Commission. 2000. IUCLID Dataset. Ispra (IT): European Commission, Joint Research Centre, Institute for Health and Consumer Protection, European Chemicals Bureau.

Fatoki OS, Bomman M, Ravandhalala L, Chimuka L, Genthe B, Adeniyi A. 2010. Phthalate ester plasticizers in freshwater systems of Venda, South Africa and potential health effects. Water SA. 36(1):117.

Fankhauser-Noti A, Grob K. 2006. Migration of plasticizers from PVC gaskets of lids for glass jars into oily foods: amount of gasket material in food contact, proportion of plasticizer migrating into food and compliance testing by simulation. Trends Food Sci Technol. 17:105-12.

Ferguson KK, McElrath TF, Meeker JD. 2014. Environmental phthalate exposure and preterm birth. JAMA Pediatr 168(1):61-7.

Ferreira ID, Morita DM. 2012. Ex-situ bioremediation of Brazilian soil contaminated with plasticizers process wastes. Braz J Chem Eng. 29:77-86.

Fierens T, Servaes K, Van Holderbeke M, Geerts L, De Henauw S, Sioen I, Vanerman G. 2012a. Analysis of phthalates in food products and packaging materials sold on the Belgian market. Food Chem Toxicol. 50:2575-2583.

Fong J-P, Lee F-J, Lu I-S, Uang S-N, Lee C-C. 2015. Relationship between urinary concentrations of di(2-ethylhexyl) phthalate (DEHP) metabolites and reproductive hormones in polyvinyl chloride production workers. Occup Environ Med. 72:346-353.

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

Frederiksen H, Nielsen JKS, Morck TA, Hansen PW, Jensen JF, Nielsen O, Andersson AM, 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. doi: 10.1016/j.ijheh.2013.02.006.

Fromme H, Lahrz T, Piloty M, Gebhart H, Oddoy A, Rüden H. 2004. Occurrence of phthalates and musk fragrances in indoor air and dust from apartments and kindergartens in Berlin (Germany). Indoor Air. 14(3):188-195.

Fromme H, Gruber L, Schlummer M, Wolz G, Böhmer S, Angerer J, Mayer R, Liebl B, Bolte G. 2007. Intake of phthalates and di (2-ethylhexyl) adipate: results of the integrated exposure assessment survey based on duplicate diet samples and biomonitoring data. Environ Int. 33(8):1012-1020.

Fromme H, Gruber L, Seckin E, Raab U, Zimmermann S, Kiranoglu M, Völkel W. 2011. Phthalates and their metabolites in breast milk - results from the Bavarian Monitoring of Breast Milk (BAMBI). Environ Int. 37(4):715-722.

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.

Fujii S, Yabe K, Furukawa M, Hirata M, Kiguchi M, Ikka T. 2005. A two-generation reproductive toxicity study of diethyl phthalate (DEP) in rats. J Toxicol Sci 30:97-116.

Furr J, Lambright C, Wilson V, Foster P, Gray L.2014. A short-term in vivo screen using fetal testosterone production, a key event in the phthalate adverse outcome pathway, to predict disruption of sexual differentiation. Toxicol Sci. 140(2):403-432.

Furtmann K. 1994. Phthalates in surface water - a method for routine trace level analysis. Fresenius J Anal Chem. 348:291-296.

Gaido KW, Janran BH, Liu D, Wallace DG, Borghoff S, Johnson KJ, Hall SJ, Boekelheide K. 2007. Fetal mouse phthalate exposure shows that gonocyte multinucleation is not associated with decreased testicular testosterone. Toxicol Sci. 97(2):491-503.

Ganji S, Karigar C, Pujar B.1995. Metabolism of dimethyl terephthalate by Aspergillus niger. Biodegradation. 6:61-66.

Ganning AE, Olsson MJ, Brunk U, Dallner G. 1991. Effects of prolonged treatment with phthalate ester on rat liver. Pharmacol Toxicol. 68:392-401.

Geiger DL, Brooke LT, Call DJ. 1985. Acute toxicities of organic chemicals to fathead minnows (Pimephales promelas) Vol 2. Center for Lake Superior Environmental Studies, Univ. of Wisconsin-Superior, Superior (WI). 326 pp.

Gevao B, Al-Ghadban AN, Bahloul M, Uddin S, Zafar J. 2013. Phthalates in indoor dust in Kuwait: implication for non-dietary human exposure. Indoor Air. 23(2):126-133.

Gray LE, Ostby J, Furr J, Price M, Veermachaneni DNR, 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. 58:350-365.

Guart A, Bono-Blay F, Borrell A, Lacorte S. 2014. Effect of bottling and storage on the migration of plastic constituents in Spanish bottled waters. Food Chem. 156:73-80.

Guerranti C, Sbordoni I, Fanello EL, Borghini F, Corsi I, Focardi SE. 2013. Levels of phthalates in human milk samples from central Italy. Microchem J. 107:178-181.

Guo Y, Kannan K. 2011. Comparative assessment of human exposure to phthalate esters from house dust in China and the United States. Environ Sci Technol. 45:3788-3794.

Guo Y, Zhang Z, Liu L, Li Y, Ren N, Kannan K. 2012. Occurrence and profiles of phthalates in foodstuffs from China and their implications for human exposure. J Agric Food Chem. 60:6913-6919.

Guo Y, Kannan K. 2013. A survey of phthalates and parabens in personal care products from the United States and its implications for human exposure. Environ Sci Technol. 47:14442-14449.

Guo Y, Wang L, Kannan K. 2013. Phthalates and parabens in personal care products from China: concentrations and human exposure. Arch Environ Contam Toxicol. 66(1):113-119. doi:10.1007/s00244-013-9937-x.

Hammond BG, Levinskas GJ, Robinson EC, Johannsen FR. 1987. A review of the subchronic toxicity of butyl benzyl phthalate. Toxicol Ind Health. 3:79-98 [as cited in AGDH 2008b, US CPSC 2010a].

Hansen E, Meyer O. 1989. No embryotoxic or teratogenic effect of dimethyl phthalate in rats after epicutaneous application. Pharmacol Toxicol. 64:237-238.

Harries JE, Runnalls T, Hill E, Harris CA, Maddix S, Sumpter JP, Tyler CR. 2000. Development of a reproductive performance test for endocrine disrupting chemicals using pair-breeding fathead minnows (Pimephales promelas). Environ Sci Technol. 34:3003-3011.

Hashizume K, Nanya J, Toda C, Yasui T, Nagano H, Kojima N. 2002. Phthalate esters detected in various water samples and biodegradation of the phthalates by microbes isolated from river water. Biol Pharm Bull. 25(2):209-214.

Haynes WM, Lide DR, editors. 2010. CRC handbook of chemistry and physics. 90th ed.[Internet version 2010]. Physical constants of organic compounds [Internet]. Section 3: 238. [accessed 2009 Dec]. [restricted access].

Hazleton Laboratories. 1968b. 13-week dietary administration - dogs plasticiser (DIDP) [cited in ECJRC 2003; AGDH 2008f; US CPSC 2010a].

He H, Hu G-j, Sun C, Chen S-l, Yang M-n, Li J, Zhao Y, Wang H. 2011. Trace analysis of persistent toxic substances in the main stream of Jiangsu section of the Yangtze River, China. Environ Sci Pollut Res Int. 18(4):638-648.

Health Canada. 1998. Exposure factors for assessing total daily intake of priority substances by the general population of Canada. Unpublished report. Ottawa (ON): Health Canada, Environmental Health Directorate.

Health Canada. 2007. Market Evaluation: Analysis of phthalate content in children's toys. Consumer Product Safety Bureau. Project #850950.

Health Canada. 2009. Survey-determination of phthalate in various children's toys. Consumer Product Safety Bureau. Project #2008-1090.

Health Canada. [modified 2011a]. Cosmetic ingredient hotlist: list of ingredients that are prohibited for use in cosmetic products. Ottawa (ON): Health Canada, Consumer Product Safety Directorate. [accessed 2015 Jul 13]..

Health Canada. 2011b. First Report on Human Biomonitoring of Environmental Chemicals in Canada. Results of the Canadian Health Measures Survey Cycle 1. [accessed 2014].

Health Canada. 2012. Phthalates in toys: cyclical enforcement 2011-2012. Consumer Product Safety Bureau. Project #2011-1387.

Health Canada. 2013. Second Report on Human Biomonitoring of Environmental Chemicals in Canada. Results of the Canadian Health Measures Survey Cycle 2 (2009-2011). ISBN: 978-1-100-22140-3. [accessed 2014].

Health Canada. 2014. Survey 2014-15: Determination of a Series of 34 Phthalates in Plastic Consumer Products. Consumer Product Safety Bureau. Project #2014-2047.

Health Canada. 2015. Stakeholder Technical Workshop Document: Approach for using chemical categories and read-across to address data gaps for effects on the developing male reproductive system: Phthalate Substance Grouping. August 2015. Ottawa (ON): Health Canada.

Health Canada. 2016a. Supporting Documentation: Evaluation of Epidemiologic Studies on Phthalate Compounds and Their Metabolites (Part II). Ottawa (ON): Health Canada.

Health Canada. 2016b. Supporting Documentation: A Summary of the Health Effects Associated with Exposure to Additional Phthalates. Ottawa (ON): Health Canada.

Higuchi TT, Palmer JS, Gray LE Jr., Veeramachaneni DNR. 2003. Effects of dibutyl phthalate in male rabbits following in utero, adolescent, or postpubertal exposure. Toxicol Sci. 72(2):301-313.

Högberg J, Hanberg A, Berglund M, Skerfving S, Remberger M, Calafat AM, Appelgren M. 2008. Phthalate diesters and their metabolites in human breast milk, blood or serum, and urine as biomarkers of exposure in vulnerable populations. Environ Health Perspect. 116(3):3343-339.

Hongjun Y, Wenjun X, Qing L, Jingtao L, Hongwen Y, Zhaohua L. 2014. Distribution of phthalate esters in topsoil: a case study in the Yellow River Delta, China. Environ Monit Assess. 185:8489-8500.

Hornung MW, Tapper MA, Denny JS, Kolanczyk RC, Sheedy BR, Hartig PC, Aladjov H, Henry TR, Schmieder PK. 2014. Effects-based chemical category approach for prioritization of low affinity estrogenic chemicals. SAR QSAR Environ Res. 25(4):289-323.

Hoshino N, Iwai M, Okazaki Y. 2005. A two-generation reproductive toxicity study of dicyclohexyl phthalate in rats. J Toxicol Sci. 30:79-96.

[HPD] Household Products Database [database]. 1993- . Bethesda (MD): US National Library of Medecine. [accessed 2014 Jul 3].

Hsu NY, Lee CC, Wang JY, Li YC, Chang HW, Chen CY, Bornehag CG, Wu PC, Sundell J, Su HJ. 2012. Predicted risk of childhood allergy, asthma, and reported symptoms using measured phthalate expsure exposure in dust and urine. Indoor Air. 22:186-199.

[HSDB] Hazardous Substances Data Bank [database]. 1983- . Bethesda (MD): National Library of Medicine (US). [accessed 2013 Dec].

Huang L-P, Lee C-C, Fan J-P, Kuo P-H, Shih-T-S, Hsu P-C. 2014. Urinary metabolites of di(2-ethylhexyl) phthalate relation to sperm motility, reactive oxygen species generation, and apoptosis in polyvinyl chloride workers. Int Arch Occup Environ Health. 87:635-646.

Hudson RA, Austerberry CF, Bagshaw JC. 1981. Phthalate ester hydrolases and phthalate ester toxicity in synchronously developing larvae of the brine shrimp (Artemia). Life Sci. 29(18):1865-1872.

Jeddi MZ, Rastkari N, Ahmadkhaniha R, Yunesian M. (2015) Concentrations of phthalates in bottled water under common storage conditions: Do they pose a health risk to children? Food Res Int. 69:256-265. doi: 10.1016/j.foodres.2014.11.057.

Jensen MS, Anand-Ivell R, Nørgaard-Pedersen B, Jönsson BAG, Bonde JP, Hougaard DM, Cohen A, Lindh CH, Ivell R, Toft G. 2015. Amniotic fluid phthalate levels and male fetal gonad function. Epidemiology. 26(1):91-99.

Joensen UN, Frederiksen H, Blomberg Jensen M, Lauritsen MP, Olesen IA, Lassen TH, Andersson AM, Jørgensen N. 2012. Phthalate excretion pattern and testicular function: a study of 881 healthy Danish men. Environ Health Perspect. 120(10):1397-1403.

Johnson S, Saikia N, Sahu R. 2011. Phthalates in toys available in Indian market. Bull Environ Contam Toxicol. 86:621-626.

Jurewicz J, Radwan M, Sobala W, Ligocka D, Radwan P, Bochenek M, Hawuła W, Jakubowski L, Hanke W. 2013. Human urinary phthalate metabolites level and main sem*n parameters, sperm chromatin structure, sperm aneuploidy and reproductive hormones. Reprod Toxicol. 42:232-241.

Kang Y, Man YB, Cheung KC, Wong MH. 2012. Risk assessment of human exposure to bioaccessible phthalate ethers via indoor dust around the Pear River Delta. Environ Sci Technol. 46:8422-8430. doi: 10.1021/es300379v.

Kato H, Furuhashi T, Tanaka M, Katsu Y, Watanabe H, Ohta Y, Iguchi T. 2006. Effects of bisphenol A given neonatally on reproductive functions of male rats. Reprod Toxicol. 22(1):20-29.

[KEMI] Swedish Chemicals Agency. 2015. An additional assessment factor (MAF) - A suitable approach for improving the regulatory risk assessment of chemical mixtures?.

Khosravi K, Price GW. 2015. Determination of phthalates in soils and biosolids using accelerated solvent extraction coupled with SPE cleanup and GC-MS quantification. Microchem J. 121:205-212.

Kickham P, Otton SV, Moore MM, Ikonomou MG, Gobas FAPC. 2012. Relationship between biodegradation and sorption of phthalate esters and their metabolites in natural sediments. Environ Toxicol Chem. 31(8):1730-1737.

Kim YH, Lee JW, Ahn JY, Gu MB, Moon SH. 2002a. Enhanced degradation of an endocrine-disrupting chemical, butyl benzyl phthalate, by Fusarium oxysporum f. sp. pisi cutinase. Appl Environ Microbiol. 68:4684-4688.

Kim EJ, Kim JW, Lee SK. 2002b. Inhibition of oocyte development in Japanese medaka (Oryzias latipes) exposed to di-2-ethylhexyl phthalate. Environ Int. 28:359-365.

Kim HS, Kim TS, Shin JH, Moon HJ, Kang IH, Kim IY, Oh JY, Han SY. 2004. Neonatal exposure to di(n-butyl) phthalate (dbp) alters male reproductive-tract development. J Toxicol Environ Health A. 67(23-24):2045-2060.

Kim YH, Lee J, Moon SH. 2003. Degradation of an endocrine disrupting chemical, DEHP [di-(2-ethylhexyl)-phthalate], by Fusarium oxysporum f. sp. pisi cutinase. Appl Microbiol Biotechnol. 63:75-80.

Kim YH, Lee J. 2005. Enzymatic degradation of dibutyl phthalate and toxicity of its degradation products. Biotechnol Lett. 27:635-639.

Kim YH, Seo HS, Min J, Kim YC, Ban YH, Han KY, Park JS, Bae KD, Gu MB, Lee J. 2007. Enhanced degradation and toxicity reduction of dihexyl phthalate by Fusarium oxysporum f. sp. pisi cutinase. J Appl Microbiol. 102:221-228.

Kim DS, Um H-J, Lim E-S, Min J, Kim YH. 2008. Degradation of diphenyl phthalate by Sphingomonas chungbukensis. Biotechnol Lett. 30:93-96.

Koch HM, Becker K, Wittassek M, Seiwert M, Angerer J, Kolossa-Gehring M. 2007 Di-n-butylphthalate and butylbenzylphthalate-urinary metabolite levels and estimated daily intakes: pilot study for the German Environmental Survey on children. J Expo Sci Environ Epidemiol. 17:378-387.

Koch HM, Calafat AM. 2009. Human body burdens of chemicals used in plastic manufacture. PhilosTrans R Soc Lond B Biol Sci. 364(1526):2063-2078.

Koch HM, Haller A, Weiss T, Käfferlein HU, Stork J, Brüning T. 2012. Phthalate exposure during cold plastisol application-a human biomonitoring study. Toxicol Lett. 213(1):100-106.

Kolarik B, Bornehag C-G, Naydenov K, Sundell J, Nielsen OF. 2008. The concentrations of phthalates in settled dust in Bulgarian homes in relation to building characteristic and cleaning habits in the family. Atmos Environ. 42:8553-8559.

Koniecki D, Wang R, Moody RP, Zhu J. 2011. Phthalates in cosmetic and personal care products: Concentrations and possible dermal exposure. Environ Res. 111:329-336.

Korfali SI, Sabra R, Jurdi M, Taleb RI. 2013. Assessment of toxic metals and phthalates in children's toys and clays. Arch Envrion Contam Toxicol. 65(3):368-381. doi: 10.1007/s00244-013-9925-1.

[KOWWIN] Octanol-Water Partition Coefficient Program for Microsoft Windows [estimation model]. 2010. Ver. 1.68. Washington (DC): US Environmental Protection Agency, Office of Pollution Prevention and Toxics; Syracuse (NY): Syracuse Research Corporation.

Kubwabo C, Rasmussen P, Fan X, Kosarac I, Wu F, Zidek A, Kuchta S. 2013. Analysis of selected phthalates in Canadian indoor dust collected using household vacuum and standardized sampling techniques. Indoor Air. 23:506-514.

Kurata Y, Kidachi F, Yokoyama M, Toyota N, Tsuchitani M, Katoh M. 1998. Subchronic toxicity of di(2-ethylhexyl)phthalate in common marmosets: lack of hepatic peroxisome proliferation, testicular atrophy, or pancreatic acinar cell hyperplasia. Toxicol Sci. 42(1):49-56.

Kwack SJ, Kim KB, Kim HS, Lee BM. 2009. Comparative toxicological evaluation of phthalate diesters and metabolites in Sprague-Dawley male rats for risk assessment. J Toxicol Environ Health. 72:1446-54.

Lake BG, Brantom PG, Gangolli SD, Butterworth KR, Grasso P. 1976. Studies on the effects of orally administered Di-(2-ethylhexyl) phthalate in the ferret. Toxicology. 6(3):341-356.

Lake BG, Foster JR, Collins MA, Stubberfield CR, Gangolli SD, Srivastava SP. 1982. Studies on the effects of orally administered dicyclohexyl phthalate in the rat. Acta Pharmacol Toxicol. 51:217-226.

Lamb JC, Chapin RE, Teague J, Lawton AD, Reel JR. 1987. Reproductive effects of four phthalic acid esters in the mouse. Toxicol Appl Pharmacol. 88:255-269.

Langer S, Weschler CJ, Fischer A, Beko G, Toftum J, Clausen G. 2010. Phthalate and PAH concentrations in dust collected from Danish homes and daycare centers. Atmos. Environ. 44:2294-2301.

Latini G, Wittassek M, Del Vecchio A, Presta G, De Felice C, Angerer J. 2009. Lactational exposure to phthalates in southern Italy. Environ Int. 35(2):236-239.

Lee KY, Shibutani M, Takagi H, Kato N, Takigami S, Uneyama C, Hirose M. 2004. Diverse developmental toxicity of di-n-butyl phthalate in both sexes of rat offspring after maternal exposure during the period from late gestation through lactation. Toxicology. 203:221-238.

Lee BM, Koo HJ. 2007. Hershberger assay for antiandrogenic effects of phthalates. J Toxicol Environ Health. 70:1365-1370.

Lee SM, Lee JW, Koo BW, Kim MK, Choi DH, Choi IG. 2007. Dibutyl phthalate biodegradation by the white rot fungus, Polyporus brumalis. Biotechnol Bioeng. 97:1516-1522.

Lehmann KP, Phillips S, Sar M, Foster PMD, Gaido KW. 2004. Dose-dependent alterations in gene expression and testosterone synthesis in the fetal testes of male rats exposed to di (n-butyl) phthalate. Toxicol Sci. 81:60-68.

Lenters V, Portengen L, Smit LAM, Jönsson BAG, Giwercman A, Rylander L, Lindh CH, Spanò M, Pedersen HS, Ludwicki JK, Chumak L, Piersma AH, Toft G, Bonde JP, Heederik D, Vermeulen R. 2015a. Phthalates, perfluoroalkyl acids, metals and organochlorines and reproductive function: a multipollutant assessment in Greenlandic, Polish and Ukrainian men. Occup Environ Med. 72:385-393.

Lenters V, Portengen L, Rignell-Hydbom A, Jönsson BAG, Lindh CH, Piersma AH, Toft G, Bonde JP, Heederik D, Rylander L, Vermeulen R. 2015b. Prenatal phthalate, perfluoroalkyl acid, and organochlorines exposures and term birth weight in three birth cohorts: multi-pollutant models based on elastic net regression. Environ Health Perspect. 124(3):365-372. doi: 10.1289/ehp.1408933.

Letinski DJ, Connelly MJ, Peterson DR, Parkerton TF. 2002. Slow-stir water solubility measurements of selected alcohols and diesters. Chemosphere. 48(3):257-265.

Lertsirisopon R, Soda S, Sei K, Ike M, Fujita M. 2006. Biodegradability of four phthalic acid esters under anaerobic condition assessed using natural sediment. J Environ Sci (China). 18(4):793-6.

Li B, Chi J, Wu WX, Wang ZK. 2007. Effect of nutrients and light on biodegradation of dibutyl phthalate and di-2-ethylhexyl phthalate in Haihe Estuary. Bull Environ ContamToxicol. 79:80-83.

Li B, Hu X, Liu R, Zeng P, Song Y. 2015a. Occurrence and distribution of phthalic acid esters and phenols in Hun River Watersheds. Environ Earth Sci. 73:5095-5106.

Li J, Wang G. 2015. Airborne particulate endocrine disrupting compounds in China: Compositions, size distributions and seasonal variations of phthalate esters and bisphenol A. Atmos Res. 154:138-145.

Li LX, Bu T, Su HN, Chen ZC, Liang YY, Zhang GL, Zhu DY, Shan YY, Xu RA, Hu YY, Li JW, Hu GX, Lian QQ, Ge RS. 2015b. In utero exposure to diisononyl phthalate caused testicular dysgenesis of rat fetal testis. Toxicol Lett. 232:466-474.

Li X, Chen X, Hu G, Li L, Su H, Wang Y, Chen D, Zhu Q, Li C, Li J, Wang M, Lian Q, Ge RS. 2016. Effects of in utero exposure to dicyclohexyl phthalate on rat fetal Leydig cells. Int J Environ Res Public Health. 13(3):246.

Liang D-W, Zhang T, Fang HHP, He J. 2008. Phthalates biodegradation in the environment. Appl Microbiol Biotechnol. 80:183-98.

Liang Z, Luo Q, Fan H, Lv S, Zeng Y. 2013. Determination of phthalate esters in the children's products using ultrasonic extraction and gas chromatography. Adv Mater Res. 807-809:124-129.

Lin X, Wang X, Shen T. 2014. Human exposure to phthalate esters from indoor air and dust in China. WIT Trans Eng Sci. 84:1139-1144.

Lington AW, Bird MG, Plutnick RT, Stubblefield WA, Scala RA. 1997. Chronic toxicity and carcinogenic evaluation of diisononyl phthalate in rats. Fundam Appl Toxicol. 36:79-89.

Liu Y, Guan YT, Yang ZH, Cai ZH, Mizuno T, Tsuno H, Zhu WP, Zhang XH. 2009. Toxicity of seven phthalate esters to embryonic development of the abalone Haliotis diversicolor supertexta. Ecotoxicology. 18(3):293-303.

Liu P, Tian T, Barreto J, Chou J. 2013. Assessment and analysis of phthalate esters, in Lake Pontchartrain, by SPME combining with GC-MS. Environ Technol. 34:453-462.

Liu X, Shi J, Bo T, Li H, Crittenden JC. 2015. Occurrence and risk assessment of selected phthalates in drinking water from waterworks in China. Environ Sci Pollut Res Int. 22(14):10690-10698. doi: 10.1007/s11356-015-4253-9.

[LNHPD] Licensed Natural Health Products Database [database]. [modified 2014 ]. Ottawa (ON): Health Canada. [accessed 2014 Sep 9].

Luongo G, Östman C. 2015. Organophosphate and phthalate esters in settled dust from apartment buildings in Stockholm. Indoor Air. 26(3):414-425. doi: 10.1111/ina.12217.

Lv Y-K, Zhang W, Guo M-M, Zhao F-F, Du X-X. 2015. Centrifugal microextraction tube-cloud point extraction coupled with gas chromatography for simultaneous determination of six phthalate esters in mineral water. Anal Methods. 7:560-565.

Ma J, Chen LL, Guo Y, Wu Q, Yang M, Wu MH, Kannan K. 2014. Phthalate in airborne PM2.5 and PM10 in a suburban area of Shanghai: seasonal distribution and risk assessment. Sci Total Environ. 497-498:467-474.

Ma P, Liu X, Wu J, Yan B, Zhang Y, Lu Y, Wu Y, Liu C, Guo J, Nanberg E, Bornehag CG, Yang X. 2015. Cognitive deficits and anxiety induced by diisononyl phthalate in mice and the neuroprotective effects of melatonin. Sci Rep. 5:14676.

Madsen PL, Thyme JB, Henrikenk K, Moldrup P, Roslev P. 1999. Kinetics of di-(2-ethylhexyl)phthalate mineralization in sludge-amended soil. Environ Sci Technol. 33:2601-2606.

Main K, Mortensen G, Kaleva M, Boisen K, Damgaard I, Chellakooty M, Schmidt I, Suomi A, Virtanen H, Petersen J, Andersson A, Toppari J, Skakkebaek N. 2006. Human breast milk contamination with phthalates and alterations of endogenous reproductive hormones in infants three months of age. Environ. Health Perspect. 114(2):270-276.

Mankidy R, Wiseman S, Ma H, Giesy JP. 2013. Biological impact of phthalates. Toxicol Lett. 217:50-58.

Marsman D. 1995. NTP technical report on the toxicity studies of dibutyl phthalate (CAS No. 84-74-2) administered in feed to F344/N rats and B6C3F1 mice. Toxic Rep Ser. 30:1-G5.

Mathieu-Denoncourt J, de Solla SR, Langlois VS. 2015. Chronic exposures to monomethyl phthalate in Western clawed frogs. Gen Comp Endocrinol. 219:53-63.

Mathieu-Denoncourt J, Martyniuk CJ, Loughery JR, Yargeau V, de Solla S, Langlois VS. 2016. Lethal and sublethal effects of phthalate diesters in Silurana tropicalis larvae. Environ Toxicol Chem. 35(10):2511-2522. doi: 10.1002/etc.3413.

Matsumoto M, Furuhashi T, Poncipe C, Ema M. 2008. Combined repeated dose and reproductive/developmental toxicity screening test of the nitrophenolic herbicide dinoseb, 2-sec-butyl-4,6-dinitrophenol, in rats. Environ Toxicol. 23(2):169-183.

Mayer FL, Ellersieck MR. 1986. Manual of acute toxicity: interpretation and data base for 410 chemicals and 66 species of freshwater animals. U.S. Fish and Wildlife Service Res. Pub. 160. Washington (DC): United States Department of the Interior, Fish and Wildlife Service [cited in ECJRC 2004].

Mayer P, Reichenberg F. 2006. Can highly hydrophobic organic substances cause aquatic baseline toxicity and can they contribute to mixture toxicity? Environ Toxicol Chem. 25(10):2639-2644.

McCarthy JF, Whitmore DK. 1985. Chronic toxicity of di-n-butyl and di-n-octyl phthalate to Daphnia magna and the fathead minnow. Environ Toxicol Chem. 4:167-179.

McCarty LS, Mackay D. 1993. Enhancing ecotoxicological modeling and assessment: body residues and modes of toxic action. Environ Sci Technol. 27(9):1719-1728.

McConnell ML. 2007. Distribution of phthalate monoesters in an aquatic food web. School of Resource and Environmental Management. Master of Resource Management Thesis Project Report No. 426. Spring 2007. Burnaby (BC): Simon Fraser University. pp. 78.

McKee RH, Pavkov KL, Trimmer GW, Keller LH, Stump DG. 2006. An assessment of the potential developmental and reproductive toxicity of diisoheptyl phthalate in rodents. Reprod Toxicol. 21:241-252.

Meek M, Boobis A, Crofton K, Heinemeyer G, VanRaaij M, Vickers C. 2011. Risk assessment of combined exposure to multiple chemicals: A WHO/IPCS framework. Regul Toxicol Pharmacol. 60:S1-S14.

Meeker JD, Hu H, Cantonwine DE, Lamadrid-Figueroa H, Calafat AM, Ettinger AS, Hernandez-Avila M, Loch-Caruso R, Téllez-Rojo MM. 2009. Urinary phthalate metabolites in relation to preterm birth in Mexico City. Environ Health Perspect. 117(10):1587-1592.

Meeker JD, Ferguson KK. 2014. Urinary phthalate metabolites are associated with decreased serum testosterone in men, women, and children from NHANES 2011-2012.J Clin Endocrinol Metab. 99(11):4346-4352.

Mendiola J, Jørgensen N, Andersson A-M, Calafat AM, Silva MJ, Redmon JB, Sparks A, Drobnis EZ, Wang C, Liu F, Swan SH. 2011. Associations between urinary metabolites of di(2-ethylhexyl) phthalate and reproductive hormones in fertile men. Int J Androl. 34(4):369-378.

Mendiola J, Meeker JD, Jørgensen N, Andersson AM, Liu F, Calafat AM, Redmon JB, Drobnis EZ, Sparks AE, Wang C, Hauser R, Swan SH. 2012. Urinary concentrations of di(2-ethylhexyl) phthalate metabolites and serum reproductive hormones: pooled analysis of fertile and infertile men. J Androl. 33(3):488-498.

Mlynarcíková A, Ficková M, Scsuková S. 2007. The effects of selected phenol and phthalate derivatives on steroid hormone production by cultured porcine granulosa cells. ATLA 35:71-77.

Moody S, Goh H, Bielanowiicz A, Rippon P, Loveland KL, Itman C. 2013. Prepubertal mouse testis growth and maturation and androgen production are acutely sensitive to di-n butyl phthalate. Endocrinology. 154:3460-3475.

Moore MR. 1998. Oncogenicity study in mice with di(isononyl) phthalate including ancillary hepatocellular proliferation and biochemical analyses. Covance Laboratories, Inc., Vienna, VA 22182. For Aristech Chemical Corporation, Pittsburgh, PA 15230. Covance 2598-105 [as cited in AGDH 2008c; US CPSC 2010b].

Morin A. 2003. Distribution of phthalate esters in marine mammal food chain from Canada's Eastern Arctic. Masters Thesis. Simon Fraser University, BC. Report No. 338, 1-89 pp.

Mortensen GK, Main KM, Andersson AM, Leffers H, Skakkebaek NE. 2005. Determination of phthalate monoesters in human milk, consumer milk, and infant formula by tandem mass spectrometry (LC-MS-MS). Anal Bioanal Chem. 382:1084-1092.

Mouritsen A, Frederiksen H, Sørensen K, Aksglaede L, Hagen C, Skakkebaek NE, Main KM, Andersson AM, Juul A. 2013. Urinary phthalates from 168 girls and boys measured twice a year during a 5-year period: associations with adrenal androgen levels and puberty. J Clin Endocrinol Metab. 98(9):3755-64.

Mousa A, Basheer C, Al-Arfaj AR. 2013. Determination of phthalate esters in bottled water using dispersive liquid-liquid microextraction coupled with GC-MS. J Sep Sci. 36(12):2003-2009.

Nagao T, Ohta R, Marumo H, Shindo T, Yoshimura S, Ono H. 2000. Effect of butyl benzyl phthalate in Sprague-Dawley rats after gavage administration: a two-generation reproductive study. Reprod Toxicol. 14:513-532.

Nair N. 2015. Dose-dependent short-term study of di-n-butyl phthalate on the testicular antioxidant system of Wistar rats. Environ Sci Pollut Res Int. 22(3):2196-2204.

[NAS] National Academies of Science. 2008. Phthalates and Cumulative Risk Assessment: The Task Ahead. Committee on the Health Risks of Phthalates, National Research Council. ISBN: 0-309-12842-0.

Net S, Rabodonirina S, Sghaier RB, Dumoulin D, Chbib C, Tlili I, Ouddane B. 2015. Distribution of phthalates, pesticides and drug residues in the dissolved, particulate and sedimentary phases from transboundary rivers (France-Belgium). Sci Total Environ. 521-522:152-159.

[New EQC] New Equilibrium Criterion Model. 2011. Ver. 1.00 (Beta). Peterborough (ON): Trent University, Canadian Centre for Environmental Modelling and Chemistry.

[NHPID] Natural Health Products Ingredients Database [database]. [modified 2014]. Ottawa (ON): Health Canada. [accessed 2014 Sep 9].

Niu L, Xu Y, Xu C, Yun L, Liu W. 2014. Status of phthalate esters contamination in agricultural soils across China and associated health risks. Environ Pollut. 195:16-23. doi: 10.1016/j.envpol.2014.08.014.

Nilsson NH, Malmgren-Hansen B, Bernth N, Pedersen E and Pommer K. 2006. Survey and health assessment of chemicals substances in sex toys. Survey of Chemical Substances in Consumer Products, No. 77. Danish Environmental Protection Agency, Copenhagen.

Norman A, Börjeson H, David F, Tienpont B, Norrgren L. 2007. Studies of uptake, elimination, and late effects in Atlantic salmon (Salmo salar) dietary exposed to Di-2-ethylhexyl phthalate (DEHP) during early life. Arch Environ Contam Toxicol. 52:235-242.

[NPRI] National Pollutant Release Inventory [database]. 2010-2014. Search results for CAS RNs 131-11-3, 84-66-2, 84-74-2, 85-68-7, 117-81-7 and 117-84-0. Gatineau (QC): Environment and Climate Change Canada. [updated 2016 Jan 4; accessed 2016 Apr 6]..

[NRC] National Research Council. 2012. Material Emissions Testing for Phthalates. Unpublished report.

[NTP] National Toxicology Program (US). 1995. Toxicology and carcinogenesis studies of diethylphthalate (CAS no. 84-66-2) in F344/N rats and B6C3F1 mice (dermal studies) with dermal initiation/promotion study of diethylphthalate and dimethylphthalate (CAS no. 131-11-3) in male Swiss (CD-1) mice. Technical Report Series No. 429:1-286.

[NTP] National Toxicology Program (US) . 1997. Toxicology and Carcinogenesis Studies of Butyl Benzyl Phthalate (CAS No. 85-68-7) in F344/N Rats (Feed Studies). Report No. NTP TR 458. NIH Publication No. 97-3374: US Department of Health and Human Services, Public Health Service, National Institute of Health [as cited in AGDH 2008b].

Oehlmann J, Schulte-Oehlmann U, Kloas W, Jagnytsch O, Lutz I, Kusk KO, Wollenberger L, Santos EM, Paull GC, Van Look KJW, Tyler CR. 2009. A critical analysis of the biological impacts of plasticizers on wildlife. Philos Trans R Soc Lond B Biol Sci. 364(1526):2047-2062.

Oishi S, Hiraga K. 1980a. Testicular atrophy induced by phthalic acid esters: effect on testosterone and zinc concentrations. Toxicol Appl Pharmacol. 53:35-41.

Oishi S, Hiraga K. 1980b. Effect of phthalic acid esters on mouse testes. Toxicol Lett. 5:413-416.

Orecchio S, Indelicato R, Barreca S. 2013. The distribution of phthalate esters in indoor dust of Palermo (Italy). Environ Geochem Health. 35:613-624. doi: 10.1007/s10653-013-9544-9.

Otton SV, Sura S, Blair J, Ikonomou MG, Gobas FAPC. 2008. Biodegradation of mono-alkyl phthalate esters in natural sediments. Chemosphere. 71:2011-2016.

Ortiz-Zarragitia M, Trant JM, Cajaraville MP. 2006. Effects of dibutylphthalate and ethynylestradiol on liver peroxisomes, reproduction, and development of zebrafish (Danio rerio). Environ Toxicol Chem. 25(9):2394-2404.

Page BD, Lacroix GM. 1992. Studies into the transfer and migration of phthalate esters from aluminium foil-paper laminates to butter and margarine. Food Addit Contam. 9(3):197-212.

Page BD, Lacroix GM. 1995. The occurrence of phthalate ester and di-2-ethylhexyl adipate plasticizers in Canadian packaging and food sampled in 1985-1989: a survey. Food Addit Contam. 12(1):129-151.

Pan G, Hanaoka T, Yoshimura M, Zhang S, Wang P, Tsukino H, Inoue K, Nakazawa H, Tsugane S, Takahashi K. 2006. Decreased serum free testosterone in workers exposed to high levels of Di-n-butyl phthalate (DBP) and di-2-ethylhexyl phthalate (DEHP): a cross-sectional study in China. Environ Health Perspect. 114(11):1643-1648.

Pan Y, Jing J, Dong F, Yao Q, Zhang W, Zhang H, Yao B, Dai J. 2015. Association between phthalate metabolites and biomarkers of reproductive function in 1066 Chinese men of reproductive age. J Hazard Mater. 300:729-736.

Pant N, Shukla M, Kumar Patel D, Shukla Y, Mathur N, Kumar Gupta Y, Saxena DK. 2008. Correlation of phthalate exposures with sem*n quality. Toxicol Appl Pharmacol. 23(1):112-116.

Pant N, Kumar G, Upadhyay AD, Patel DK, Gupta YK, Chaturvedi PK. 2014. Reproductive toxicity of lead, cadmium, and phthalate exposure in men. Environ Sci Pollut Res Int. 21(18):11066-11074.

Papadopoulos A, Vlachogiannis D, Maggos T, Sfetsos A, Karayiannis MI. 2013. A semi-quantitative approach for analysing low-volatile organic compounds in house dust using an SFE method: Significant common features and particular differences of the extracts. J Supercrit Fluids. 82:268-281.

Patyna PJ, Brown RP, Davi RA, Letinski DJ, Thomas PE, Cooper KR, Parkerton TF. 2006. Hazard evaluation of diisononyl phthalate and diisodecyl phthalate in a Japanese medaka multigenerational assay. Ecotoxicol Environ Saf. 65(1):36-47.

Pei XQ, Song M, Guo M, Mo FF, Shen XY. 2013. Concentration and risk assessment of phthalates present in indoor air from newly decorated apartments. Atmos Environ. 68:17-23.

Peng L. 2015. Mice brain tissue injury induced by diisononyl phthalate exposure and the protective application of vitamin E. J Biochem Mol Toxicol. 29(7):311-20.

Peterson DR, Staples CA. 2003. Degradation of phthalate esters in the environment. In: Staples, C.A. (Ed.), Phthalate esters. In: Handbook of Environmental Chemistry, V.3. Part Q. Springer-Verlag, Berlin-Heidelberg-New York-Hong Kong- London-Milan- Paris-Tokyo, pp. 85-124.

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

Phokha W, Kessler W, Csanady GA, Filser JG. 2002. Toxico*kinetics of di(2-ethylhexyl) phthalate and mono(2-ethylhexyl) phthalate in non-pregnant and pregnant rats. Naunyn Schmiedebergs Arch Pharmacol. 365(Suppl 1):R497, p. 128. [Abstract].

Poon R, Lecavalier P, Mueller R, Valli VE, Procter BG, Chu I. 1997. Subchronic oral toxicity of di-n-octyl phthalate and di(2-ethylhexyl) phthalate in the rat. Food Chem Toxicol. 35:225-239.

[RAPEX] Rapid Alert System for non-food dangerous products, 'RAPid EXchange'. 2015. Weekly Notification reports.

Remberger M, Kaj L, Hansson K, Andersson H, Brorström-Lundén E, Lunder H, Schlabach M. 2013. Selected plasticisers and additional sweeteners in the Nordic Environment. Nordic Council of Ministers. Copenhagen (DK). doi:.

Renberg LO, Sundström SG, Rosen-Olofsson AC. 1985. The determination of partition coefficients of organic compounds in technical products and waste waters for the estimation of their bioaccumulation potential using reverse-phase thin layer chromatography. Environ Toxicol Chem. 10:333-349.

Rhind SM, Kyle CE, Kerr C, Osprey M, Zhang ZL, Duff EI, Lilly A, Nolan A, Hudson G, Towers W, Bell J, Coull M, McKenzie C. 2013. Concentrations and geographic distribution of selected organic pollutants in Scottish surface soils. Environ Pollut. 182:15-27. doi: 10.1016/j.envpol.2013.06.041

Roslev P, Madsen PL, Thyme JB, Henriksen K. 1998. Degradation of phthalate and di-(2-ethylhexyl) phthalate by indigenous and inoculated microorganisms in sludge-amended soil. Appl Environ Microbiol. 64:4711-4719.

Rüdel H, Schmidt S, Kordel W, Klein W. 1993. Degradation of pesticides in soil: comparison of laboratory experiments in a biometer system and outdoor lysimeter experiments. Sci Total Environ. 132:181-200.

Rudel RA, Dodson RE, Perovich LJ, Morello-Frosch R, Camann DE, Zuniga MM, Yau AY, Just AC, Green Brody J. 2010. Semivolatile endocrine-disrupting compounds in paired indoor and outdoor air in two northern California communities. Environ Sci Technol. 44(17):6583-6590.

Ruzicková J, Raclavskáa H, Raclavsky K, Juchelková V. 2016. Phthalates in PM2.5 airborne particles in the Moravian-Silesian Region, Czech Republic. Perspect Sci. 7:178-183.

Saffarini CM, Heger NE, Yamasaki H, Liu T, Hall SJ, Boekelheidi K. 2012. Induction and persistence of abnormal testicular germ cells following gestational exposure to di-(n-butyl) phthalate in p53-null mice. J Androl. 33(3):505-513.

Saillenfait AM, Sabaté JP, Gallissot F. 2008. Diisobutyl phthalate impairs the androgen-dependent reproductive development of the male rat. Reprod Toxicol. 26(2):107-115.

Saillenfait AM, Sabate JP, Robert A, Rouiller-Fabre V, Roudot AC, Moison D, Denis F. 2013a. Dose-dependent alterations in gene expression and testosterone production in fetal rat testis after exposure to di-n-hexyl phthalate. J Appl Toxicol. 33(9):1027-1035.

Saillenfait AM, Sabate JP, Robert A, Cossec B, Roudot AC, Denis Flavien D, Burgart M. 2013b. Adverse effects of diisooctyl phthalate on the male rat reproductive development following prenatal exposure. Reprod Toxicol. 42:192-202.

[SCCNFP] Scientific Committee on Cosmetic Products and Non-food Products Intended for Consumers (scientific advisory body to the European Commission). 2002. Opinion of the scientific committee on cosmetic products and non-food products intended for consumers concerning diethyl phthalate (SCCNFP/0411/01). Adopted by the SCCNFP during the 20th Plenary meeting of 4 June 2002.

Schecter A, Lorber M, Guo Y, Wu Q, Yun SH, Kannan K, Birnbaum LS. 2013. Phthalate concentrations and dietary exposure from food purchased in New York State. Environ Health Perspect. 121(4):473-494. doi: 10.1289/ehp.1206367.

Scheunert I, Vockel D, Schmitzer J, Korte F. 1987. Biomineralization rates of 14C-labelled organic chemicals in aerobic and anaerobic suspended soil. Chemosphere. 16(5):1031-1041.

Schmieder P, Kolanczyk RC, Hornung MW, Tapper MA, Denny JS, Sheedy BR, Aladjov H. 2014. A rule-based expert system for chemical prioritization using effects-based chemical categories. SAR QSAR Environ Res. 25(4):253-287.

Selvaraj KK, Sundaramoorthy G, Ravichandran PK, Girijan GK, Sampath S, Ramaswamy BR. 2015. Phthalate esters in water and sediments of the Kaveri River, India: environmental levels and ecotoxicological evaluations. 37:83-96.

Shanker R, Ramakrishna C, Seth PK. 1985. Degradation of some phthalic acid esters in soil. Environ Pollut 39:1-7.

Shao X-L. 2013. Determination of phthalate acid esters in water and sediment samples by GC-MS. Adv Mater Res. 610-613:157-162.

Sivamurthy K, Swamy BM, Pujar BG. 1991. Transformation of dimethyl-terephthalate by the fungus Sclerotium rolfsii. FEMS Microbiol Lett. 79:37-40.

Sørensen LK. 2006. Determination of phthalates in milk and milk products by liquid chromatography/tandem mass spectrometry. Rapid Commun Mass Spectrom. 20(7):1135-1143.

Specht IO, Toft G, Houggard KS, Lindh CH, Lenters V, Jönsson BAG, Heederik D, Giwercman A, Bonde JPE. 2014. Associations between serum phthalates and biomarkers of reproductive function in 589 adult men. Environ Int. 66:146-156.

Srivastava SP, Srivastava S, Saxena DK, Chandra SV, Seth PK. 1990a. Testicular effects of di-n-butyl phthalate (DBP): biochemical and histopathological alterations. Arch Toxicol. 64:148-152.

Srivastava S, Singh GB, Srivastava SP, Seth PK. 1990b. Testicular toxicity of di-n-butyl phthalate in adult rats: Effect on marker enzymes of spermatogenesis. Indian J Exp Biol. 28:67-70.

Statistics Canada. 2004. Canadian Community Health Survey - Nutrition (CCHS). Detailed information for 2004 (Cycle 2.2). Ottawa (ON): Statistics Canada.

Stringer R, Labunska I, Santillo D, Johnston P, Siddorn J, Stephenson A. 2000. Concentrations of phthalate esters and identification of other additives in PVC children's toys. Environ Sci Pollut Res Int. 7(1):27-36.

Study Submission. 2014a. Unpublished confidential study submitted to Environment Canada under the Chemicals Management Plan initiative. Gatineau (QC): Environment Canada, Program Development and Engagement Division. Internal reference: 2008 study_85-68-7_Other.

Study Submission. 2014b. Unpublished confidential study submitted to Environment Canada under the Chemicals Management Plan initiative. Gatineau (QC): Environment Canada, Program Development and Engagement Division. Internal reference: 1986 study_85-68-7_Other_2.

Study Submission. 2014d. Unpublished confidential study submitted to Environment Canada under the Chemicals Management Plan initiative. Gatineau (QC): Environment Canada, Program Development and Engagement Division. Internal reference: 2008 study_85-68-7_Other.

Su P-H, Chen J-Y, Lin C-Y, Chen H-Y, Liao P-C, Ying T-H, Wang S-L. 2014. Sex steroid hormone levels and reproductive development of eight-year-old children following in utero and environmental exposure to phthalates. PLOS ONE. 9(9):e102788.

Takeuchi S, Kojima H, Saito I, Jin K, Kobayashi S, Tanaka-Kagawa T, Jinno H. 2014. Detection of 34 plasticizers and 25 flame retardants in indoor air from houses in Sapporo, Japan. Sci Total Environ. 491-492:28-33.

Teil M-J, Blanchard M, Dargnat C, Larcher-Tiphagne K, Chevreuil M. 2007. Occurrence of phthalate diesters in rivers of the Paris district (France). Hydrol Process. 21:2515-2525.

Thurston SW, Mendiola J,Bellamy AR, Levine H, Wang HC, Sparks A, Redmon JB, Drobnis EZ, Swan SH. 2016. Phthalate exposure and sem*n quality in fertile US men. Andrology 4:632-638.

[TIMES] TIssue MEtabolism Simulator [prediction module]. 2014. Ver. 2.27.15. Bourgas (BG): University "Prof. Dr. Assen Zlatarov", Laboratory of Mathematical Chemistry.

Timofieyskaya LA. 1976. In: Plyasunov AK, Pahkova GA, editors. Major Problems of Remote After-Effects of Exposure to Occupational Poisons. Collected scientific works. p. 40-43 [as cited in IUCLID 2000].

Tonk ESM, Verhoef A, Gremmer ER, van Loveren H, Piersma AH. 2012. Relative sensitivity of developmental and immune parameters in juvenile versus adult male rats after exposure to di(2-ethylhexyl) phthalate. Toxicol Appl Pharmacol. 260:48-57.

Tran TM, Kannan K. 2015. Occurrence of phthalate diesters in particulate and vapor phases in indoor air and implications for human exposure in Albany, New York, USA. Arch Environ Contam Toxicol. 68:489-499.

Tran BC, Teil M-J, Blanchard M, Alliott F, Chevreuil M. 2015. Fate of phthalates and BPA in agricultural and non-agricultural soils of the Paris area (France). Environ Sci Pollut Res Int. 22(14):11118-11126. doi: 10.1007/s11356-015-4178-3.

Türk G, Ateşşahin A, Sönmez M, Ceribaşi AO, Yüce A. 2008. Improvement of cisplatin-induced injuries to sperm quality, the oxidant-antioxidant system, and the histologic structure of the rat testis by ellagic acid. Fertil Steril. 89(5):1474-1481.

Tyl RW, Myers CB, Marr MC, Fail PA, Seely JC, Brine DR, Barter RA, Butala JH. 2004. Reproductive toxicity evaluation of dietary butyl benzyl phthalate (BBP) in rats. Reprod Toxicol. 18:241-264.

Tyl et al. 2012. Developmental toxicity evaluation of Santicizer® 261A (S-261A) administered in the diet to CD® (Sprague Dawley) rats. RTI Project Number 0212856.000.001. Unpublished information submitted to Environment Canada under the Chemicals Management Plan initiative. Gatineau (QC): Environment Canada, Program Development and Engagement Division. [restricted access].

Tyl et al. 2013. Two-generation reproductive toxicity evaluation of Santicizer® 261A (S-261A) administered in the diet to CD® (Sprague Dawley) rats. RTI Project Number 0212856.000.002. Unpublished information submitted to Environment Canada under the Chemicals Management Plan initiative. Gatineau (QC): Environment Canada, Program Development and Engagement Division. [restricted access].

[US CPSC] United States Consumer Product Safety Commission. 2010a. Toxicity Review of Benzyl-n-butyl Phthalate. Bethesda (MD).

[US CPSC] United States Consumer Product Safety Commission. 2010b. Toxicity review of diisononyl phthalate (DINP). Bethesda (MD).

[US CPSC CHAP] United States Consumer Product Safety Commission Chronic Hazard Advisory Panel. 2014. Chronic Hazard Advisory Panel on Phthalates and Phthalate Alternatives Final Report.

[US EPA] United States Environmental Protection Agency. 2010. Screening-level hazard characterization phthalate esters category. April 2010.

[US EPA] United States Environmental Protection Agency. 2011. Exposure factors handbook: 2011 edition. Washington (DC): US EPA, National Center for Environmental Assessment, Office of Research and Development.

Van den Belt K, Verheyen R, Witters H. 2003. Comparison of vitellogenin responses in zebrafish and rainbow trout following exposure to environmental estrogens. Ecotoxicol Environ Saf. 56(2):271-281.

van den Driesche S, Walker M, McKinnell C, Scott HM, Eddie SL, Mitchell RT. 2012. Proposed role for coup-tfii in regulating fetal Leydig cell steroidogenesis, perturbation of which leads to masculinization disorders in rodents. PLOS ONE. 7(5):e37064.

[VCCLab] Virtual Computational Chemistry Laboratory. ALOGPS [non-Java interface]. 2005. Ver. 2.1. Munich (DE): VCCLab. [Tetko IV, Gasteiger J, Todeschini R, Mauri A, Livingstone D, Ertl P, Palyulin VA, Radchenko EV, Zefirov NS, Makarenko AS, et al. 2005. Virtual computational chemistry laboratory - design and description. J Comput Aid Mol Des. 19:453-463.].

Veith GD, Broderius SJ. 1987. Structure-toxicity relationships for industrial chemicals causing type (II) narcosis syndrome. In: Kaiser KLE (ed). QSAR in environmental toxicology-II. Dordrecht (NL): D. Reidel Publishing Company. p. 385-391.

Veith G.D, Broderius SJ. 1990. Rules for distinguishing toxicants that cause type I and type II narcosis syndromes. Environ Health Perspect. 87:207-211.

Wang JL, Chen LJ, Shi HC, Qian Y. 2000. Microbial degradation of phthalic acid esters under anaerobic digestion of sludge. Chemosphere. 41(8):1245-1248.

Wang J, Bo L, Li L, Wang D, Chen G, Christie P, Teng Y. 2014. Occurrence of phthalate esters in river sediments in areas with different land use patterns. Sci Total Environ. 500-501:113-119.

Wang J, Chen G, Christie P, Zhang M, Luo Y, Teng Y. 2015a. Occurrence and risk assessment of phthalate esters (PAEs) in vegetables and soils of suburban plastic film greenhouses. Sci Total Environ. 523:129-137.

Wang Y-X, You L, Zeng Q, Sun Y, Huang Y-H, Wang C, Wang P, Cao W-C, Yang P, Li Y-F, Lu W-Q. 2015b. Phthalate exposure and human sem*n quality: results from an infertility clinic. Environ Res. 142:1-9.

Wang Y-X, Zeng Q, Sun Y, You L, Wang P, Li M, Yang P, Li J, Huang Z, Wang C, Li S, Dan Y, Li Y-F. 2016. Phthalate exposure in association with serum hormone levels, sperm DNA damage and spermatozoa apoptosis: a cross-sectional study in China. Environ Res. 150:557-565.

Waterman SJ, Keller LH, Trimmer GW, Freeman JJ, Nikiforov AI, Harris SB, Nicolich MJ, McKee RH. 2000. Two-generation reproduction study in rats given di-isononyl phthalate in the diet. Reprod Toxicol. 14:21-36.

Wilson R, Jones-Otazo H, Petrovic S, Mitchell I, Bonvalot Y, Williams D, Richardson GM. 2013. Revisiting dust and soil ingestion rates based on hand-to-mouth transfer. Hum Ecol Risk Assess. 19(1):158-188.

Wine RN, Li LH, Barnes LH, Gulati DK, Chapin RE. 1997. Reproductive toxicity of di-n-butylphthalate in a continuous breeding protocol in Sprague-Dawley rats. Environ Health Perspect. 105(1):102-107.

Wolfe GW, Layton KA. 2003. Multigeneration reproduction toxicity study in rats: Di (2- ethylhexyl) Phthalate: multigenerational reproductive assessment by continuous breeding when administered to Sprague-Dawley rats in the diet. Unaudited draft. Gaithersburg (MD): TherImmune Research Corporation. TRC Study No 7244-200.

[WSKOWWIN] Water Solubility for Organic Compounds Program for Microsoft Windows [estimation model]. 2010. Ver. 1.42. Washington (DC): US Environmental Protection Agency, Office of Pollution Prevention and Toxics; Syracuse (NY): Syracuse Research Corporation.

Xiao-feng Z, Nai-qiang Q, Jing Z, Zi L, Yang Z. 2009. Di (n-butyl) phthalate inhibits testosterone synthesis through a glucocorticoid-mediated pathway in rats. Int J Toxicol. 28(5):448-456.

Xie Z, Ebinghaus R, Temme C, Lohmann R, Caba A, Ruck W. 2007. Occurrence and air-sea exchange of phthalates in the arctic. Environ. Sci. Technol. 41:4555-4560.

Xie C, Zhao Y, Gao L, Chen J, Cai D, Zhang Y. 2015. Elevated phthalates' exposure in children with constitutional delay of growth and puberty. Mol Cell Endocrinol. 407:67-73.

Xu H, Shao X, Zhang Z, Zou Y, Wu X, Yang L. 2013. Oxidative stress and immune related gene expression following exposure to di-n-butyl phthalate and diethyl phthalate in zebrafish embryos. Ecotoxicol Environ Saf. 93:39-44.

Yan H, Pan G, Liang PL. 2002. Effect and mechanism of inorganic carbon on the biodegradation of dimethyl phthalate by Chlorella pyrenoidosa. J Environ Sci Health Part A 37:553-562.

Yan H, Pan G. 2004. Increase in biodegradation of dimethyl phthalate by Closterium lunula using inorganic carbon. Chemosphere. 55:1281-1285.

Yang ZH, Zhang XJ, Cai ZH. 2009. Toxic effects of several phthalate esters on the embryos and larvae of abalone Haliotis diversicolor supertexta. Chin J Oceanol Limnol. 27(2):395-399.

Yang GC, Yen CH, Wang CL. 2014. Monitoring and removal of residual phthalate esters and pharmaceuticals in the drinking water of Kaohsiung City, Taiwan. J Hazard Mater. 277:53-61. doi: 10.1016/j.jhazmat.2014.03.005.

Ye T, Kang M, Huang Q, Fang C, Chen Y, Shen H, Dong S. 2014. Exposure to DEHP and MEHP from hatching to adulthood causes reproductive dysfunction and endocrine disruption in marine medaka (Oryzias melastigma). Aquat Toxicol. 146:115-126.

Zeng F, Cui K, Li X, Fu J, Sheng G. 2004. Biodegradation kinetics of phthalate esters by Pseudomonas fluorescens FS1. Process Biochem. 39:1125-1129.

Zhang Y, Lee H. 2013. Low-density solvent-based vortex-assisted surfactant-enhanced-emulsification liquid-liquid microextraction combined with gas chromatography-mass spectrometry for the fast determination of phthalate esters in bottled water. J Chromatogr A. 1274:28-35.

Zhao LL, Xi YL, Huang L, Zha CW. 2009. Effects of three phthalate esters on the life-table demography of freshwater rotifer Brachionus calyciflorus Pallas. Aquat Ecol 43:395-402.

Zhou J, Cai ZH, Xing KZ. 2011a. Potential mechanisms of phthalate ester embryotoxicity in the abalone Haliotis diversicolor supertexta. Environ Pollut. 159:1114-1122.

Zhou J, Zhu XS, Cai ZH. 2011b. Influences of DMP on the fertilization process and subsequent embryogenesis of abalone (Haliotis diversicolor supertexta) by gametes exposure. PLOS ONE. 6(10).

Zhou D, Wang H, Zhang J. 2011c. Di-n-butyl phthalate (DBP) exposure induces oxidative stress in epididymis of adult rats. Toxicol Ind Health. 27(1):65-71.

Zhu J, Phillips S, Feng Y, Yang X. 2006. Phthalate esters in human milk: concentration variations over a 6-month postpartum time. Environ Sci Technol. 40:5276-5281.

Zhu J, Harner H, Kubwabo C, White P, Shoeib M, Wilford BH, Feng Y-L. 2007. Semi-volatile organic pollutants in indoor air and indoor dust in Ottawa residences and their implication for human exposure. Proceedings of the 6th International Conference on Indoor Air Quality, Ventilation & Energy Conservation in Buildings, Sendai, Japan, October 28-31, 2007. Vol 2, 115-120.

Zhu XB, Tay TW, Andriana BB, Alam MS, Choi EK, Tsunekawa N, Kanai Y, Kurohmaru M. 2010. Effects of di-iso-butyl phthalate on testes of prepubertal rats and mice. Okajimas Folia Anat Jpn. 86(4):129-136.

Page details

Date modified:
Draft Screening Assessment Phthalate Substance Grouping: Sections 9 and 10 (2024)

References

Top Articles
Latest Posts
Article information

Author: Duncan Muller

Last Updated:

Views: 6442

Rating: 4.9 / 5 (79 voted)

Reviews: 86% of readers found this page helpful

Author information

Name: Duncan Muller

Birthday: 1997-01-13

Address: Apt. 505 914 Phillip Crossroad, O'Konborough, NV 62411

Phone: +8555305800947

Job: Construction Agent

Hobby: Shopping, Table tennis, Snowboarding, Rafting, Motor sports, Homebrewing, Taxidermy

Introduction: My name is Duncan Muller, I am a enchanting, good, gentle, modern, tasty, nice, elegant person who loves writing and wants to share my knowledge and understanding with you.