Environ Health PerspectEnvironmental Health Perspectives0091-6765National Institue of Environmental Health Sciences15238287124738710.1289/ehp.6789ehp0112-001116Children's HealthArticlesAssociation of in Utero Organophosphate Pesticide Exposure and Fetal Growth and Length of Gestation in an Agricultural PopulationEskenaziBrenda1HarleyKim1BradmanAsa1WeltzienErin1JewellNicholas P.1BarrDana B.2FurlongClement E.3HollandNina T.11Center for Children’s Environmental Health Research, School of Public Health, University of California Berkeley, Berkeley, California, USA2National Center for Environmental Health, Centers for Disease Control and Prevention, Atlanta, Georgia, USA3Department of Genome Sciences and Medicine, Division of Medical Genetics, University of Washington Seattle, Seattle, Washington, USAAddress correspondence to B. Eskenazi, Center for Children’s Environmental Health Research, School of Public Health, UC Berkeley, 2150 Shattuck Ave., Suite 600, Berkeley, CA 94720-7380 USA. Telephone: (510) 642-3496. Fax: (510) 642-9083. E-mail: eskenazi@uclink.berkeley.edu

We gratefully acknowledge L. Fenster, R. Richter, the Center for the Health Assessment of Mothers and Children of Salinas (CHAMACOS) staff, students, and community partners, and especially the CHAMA-COS participants and their families, without whom this study would not be possible.

This research was supported by grants R82679-01-0 from the U.S. Environmental Protection Agency, PO1ES09605-02 from the National Institute of Environmental Health Sciences, and RO1 OH07400-01 from the National Institute of Occupational Safety and Health.

The authors declare they have no competing financial interests.

720041132004112101116112471020031132004This is an Open Access article: verbatim copying and redistribution of this article are permitted in all media for any purpose, provided this notice is preserved along with the article's original DOI.

Although pesticide use is widespread, little is known about potential adverse health effects of in utero exposure. We investigated the effects of organophosphate pesticide exposure during pregnancy on fetal growth and gestational duration in a cohort of low-income, Latina women living in an agricultural community in the Salinas Valley, California. We measured nonspecific metabolites of organophosphate pesticides (dimethyl and diethyl phosphates) and metabolites specific to malathion (malathion dicarboxylic acid), chlorpyrifos [O,O-diethyl O-(3,5,6-trichloro-2-pyridinyl) phosphoro-thioate], and parathion (4-nitrophenol) in maternal urine collected twice during pregnancy. We also measured levels of cholinesterase in whole blood and butyryl cholinesterase in plasma in maternal and umbilical cord blood. We failed to demonstrate an adverse relationship between fetal growth and any measure of in utero organophosphate pesticide exposure. In fact, we found increases in body length and head circumference associated with some exposure measures. However, we did find decreases in gestational duration associated with two measures of in utero pesticide exposure: urinary dimethyl phosphate metabolites [βadjusted = −0.41 weeks per log10 unit increase; 95% confidence interval (CI), 0.75–0.02; p = 0.02], which reflect exposure to dimethyl organophosphate compounds such as malathion, and umbilical cord cholinesterase (βadjusted = 0.34 weeks per unit increase; 95% CI, 0.13–0.55; p = 0.001). Shortened gestational duration was most clearly related to increasing exposure levels in the latter part of pregnancy. These associations with gestational age may be biologically plausible given that organophosphate pesticides depress cholinesterase and acetylcholine stimulates contraction of the uterus. However, despite these observed associations, the rate of preterm delivery in this population (6.4%) was lower than in a U.S. reference population.

birth outcomesbirth weightcholinesterasedialkyl phosphatesfetal growthgestational ageorganophosphatespesticidesurinary metabolites

More than one billion pounds of pesticides are used each year in the United States, with more than 700 million pounds used annually in agriculture (Donaldson et al. 2002). Recent studies have demonstrated widespread pesticide exposures for the U.S. population, including pregnant women and children (Adgate et al. 2001; Berkowitz et al. 2003; Bradman et al. 1997 ,2003; Hill et al. 1995; Loewenherz et al. 1997; Lu et al. 2001; National Center for Environmental Health 2003; Whyatt et al. 2002), with several studies suggesting that resident farm families and farm workers have higher exposures than do other populations (Curl et al. 2002; Fenske et al. 2002; Lu et al. 2000; McCauley et al. 2001; O’Rourke et al. 2000; Simcox et al. 1999).

In 1993, a National Academy of Sciences report (National Research Council 1993) stated that current tolerances for pesticide levels in food may not adequately protect fetuses and children and that the U.S. Environmental Protection Agency (EPA) needs to consider both dietary and nondietary sources of exposures in setting pesticide tolerances. The National Academy of Sciences called for research to fill the gaps of information on exposures and health consequences of pesticide exposures to the fetus and child.

Exposure of rodent dams during pregnancy to certain organophosphate pesticides, such as chlorpyrifos (Chanda et al. 1995; Muto et al. 1992), quinalphos (Srivastava et al. 1992), and dimethoate (Srivastava and Raizada 1996), has been associated with decrements in fetal growth in some studies. Other studies of the same pesticides (Institoris et al. 1995) and other organophosphates (Clemens et al. 1990; Institoris et al. 1995; Spyker and Avery 1977) have shown no association with fetal growth. To our knowledge, no animal studies have examined the relationship of length of gestation and organophosphate pesticide exposure.

The few studies that have examined the association of prenatal pesticide exposure and fetal growth or gestational duration in humans have also shown conflicting results (Fenster and Coye 1990; Grether et al. 1987; Kristensen et al. 1997; Perera et al. 2003; Restrepo et al. 1990; Savitz et al. 1989; Thomas et al. 1992; Willis et al. 1993; Xiang et al. 2000). Only three of these studies used biomarkers to measure pesticide exposure. Perera et al. (2003) found that, in residents of upper Manhattan, New York, increasing levels of the organophosphate pesticide chlorpyrifos in umbilical cord blood were associated with decreased birth weight and birth length but not with head circumference. Gestational duration was not examined. Berkowitz et al. (2004) reported that, in residents of east Harlem, levels of the urinary metabolite of chlorpyrifos, 3,5,6-trichloro-2-pyridinol (TCPy), were not associated with decreased birth weight, birth length, or head circumference or shortened gestation. However, head circumference was diminished in the children of women with low expression of paraoxonase 1 (PON1), an esterase involved in the detoxification of organophosphates. Willis et al. (1993) estimated pesticide exposure using plasma cholinesterase levels but failed to find an association with birth weight or preterm delivery. An additional eight studies have estimated prenatal pesticide exposure based either on the mother’s occupation or location of mother’s residence in relation to areas where pesticides were sprayed. Of these, five studies (Dabrowski et al. 2003; Kristensen et al. 1997; Restrepo et al. 1990; Savitz et al. 1989; Xiang et al. 2000) found that potential exposure of women to pesticides during pregnancy was associated with an increased risk of low birth weight, small for gestational age (SGA), preterm delivery, or shortened gestation, whereas three studies (Fenster and Coye 1990; Grether et al. 1987; Thomas et al. 1992) found no association. In general, most studies have been hampered by their inability to accurately classify pesticide exposure.

The purpose of the present analysis is to determine whether organophosphate pesticide exposure, as assessed by biologic markers, is associated with poorer fetal growth and shortened length of gestation in a cohort of pregnant women living in an agricultural community in the Salinas Valley of California (Eskenazi et al. 2003). The Salinas Valley is located southeast of San Francisco and runs approximately 60 miles within Monterey County. This area is often referred to as the “nation’s salad bowl,” growing primarily lettuce, broccoli, other cole crops, strawberries, artichokes, and grapes. Approximately 500,000 lb of organophosphate pesticides are applied annually in the Salinas Valley (California EPA 2002).

Materials and MethodsParticipants and recruitment.

The CHAMA-COS (Center for the Health Assessment of Mothers and Children of Salinas) project, a component of the Center for Children’s Environmental Health Research at the University of California, Berkeley, is a longitudinal birth cohort study of the effects of pesticides and other environmental exposures on the health of pregnant women and their children living in the Salinas Valley. Pregnant women entering prenatal care at Natividad Medical Center, a county hospital located in the town of Salinas, or at one of five centers of Clinica de Salud del Valle de Salinas (located in Castroville, Salinas, Soledad, and Greenfield) were screened for eligibility over 1 year between October 1999 and October 2000. Clinica de Salud del Valle de Salinas is a network of community clinics located throughout the Salinas Valley and serving a low-income population, many of whom are farm workers.

Eligible women were ≥ 18 years of age, < 20 weeks gestation at enrollment, English or Spanish speaking, Medi-Cal eligible, and planning to deliver at the Natividad Medical Center. Of 1,130 eligible women, 601 (53.2%) agreed to participate in this multiyear study. Women who declined to participate were similar to study subjects in age and parity but were more likely to be English speaking and born in the United States and less likely to be living with agricultural field workers. After losses due to miscarriage, moving, or dropping from the study before delivery, birth weight information was available for 538 women. We excluded from these analyses women with gestational or preexisting diabetes (n = 26), hypertension (n = 15), twin births (n = 5), or stillbirths (n = 3). We also excluded one woman for whom birth weight information was out of range (< 500 g). Eleven infants diagnosed with congenital anomalies at birth [International Classification of Diseases, 9th Revision (ICD-9; 1989) codes 740–759] were included in the final sample because their exclusion did not materially affect the results. The final sample size was 488. Written informed consent was obtained from all participants, and the study was approved by the institutional review boards.

Interview and medical record abstraction.

Women were interviewed twice during pregnancy and again shortly after delivery. The baseline interview occurred at a mean of 13 weeks gestation (range, 4–29 weeks), and the second interview occurred at a mean of 26 weeks gestation (range, 18–39 weeks). Interviews were conducted in English or Spanish by bilingual, bicultural interviewers.

Demographic information obtained during the baseline interview included maternal age, family income, the number of people supported by this income, country of birth, and number of years lived in the United States. Information on alcohol, tobacco, drug and caffeine use, and agricultural work was obtained at each interview. Information about previous pregnancies and any medical conditions, medications, or pregnancy complications was obtained by interview and confirmed by medical records. Medical records from prenatal visits and delivery were abstracted by a registered nurse.

Pesticide exposure measurement.

Exposure to organophosphate pesticides was assessed in three ways: a) by measuring organophosphate dialkyl phosphate metabolites in maternal urine during pregnancy; b) by measuring seven different pesticide-specific metabolites in maternal urine during pregnancy; and c) by measuring cholinesterase (ChE) in whole blood and butyryl cholinesterase (BChE) in plasma collected from mothers during pregnancy and at delivery and from the umbilical cord.

For dialkyl phosphate metabolites, spot urine samples were collected from the pregnant women at the time of the two pregnancy interviews. Urine specimens were aliquoted and stored at −80°C until shipment to the Centers for Disease Control and Prevention (CDC; Atlanta, GA) for analysis of dialkyl phosphate and pesticide-specific metabolite levels.

Six dialkyl phosphate metabolites were measured in the urine samples using gas chromatography and mass spectrometry and quantified using isotope dilution calibration (Bravo et al. 2002). The dialkyl phosphates measured were dimethylphosphate, dimethyldithiophosphate, dimethylthiophosphate, diethylphosphate, diethyldithiophosphate, and diethylthiophosphate. Approximately 80% of the organophosphate pesticides used in the Salinas Valley devolve to one or more of these metabolites, which are excreted in urine. The most commonly used pesticides in this region that devolve to dialkyl phosphates are presented in Table 1.

Quality control (QC) procedures were conducted on laboratory and field samples. Laboratory QC was established by repeat analysis of two in-house urine pools enriched with known amounts of pesticide residues whose target values and confidence limits were previously determined. An analytical run was considered “out-of control” if the QC value failed to meet the requirements of the Westgard QC multirules (Westgard 2002). Data were not reported from runs considered “out-of-control.” Mean recoveries for laboratory QC samples ranged from 98 to 105%, and the coefficients of variation (CV) ranged from 11 to 15%. Field QC was conducted by blindly inserting QC samples among the study samples in the field. These QC materials were thawed in the field, aliquoted into regular sample vials, and shipped with the samples on dry ice to CDC. The concentrations of the field QC materials, which were blinded to CDC analysts, generally agreed well with the spike concentrations (recovery and CV ranged from 94 to 103% and from 4.3 to 8.7%, respectively), indicating little contamination and/or degradation during the sampling procedures and further establishing the validity of the analytic measurements (Bravo et al., in press).

Because dialkyl phosphates originate from more than one organophosphate pesticide, quantities of the six metabolites were converted to molar concentration (nanomoles per liter) and summed to obtain the total concentrations of dialkyl phosphate metabolites for each woman. This provided an estimate of total organophosphate exposure for each individual at each of the two measurement times. The three dimethyl phosphate metabolites and three diethyl phosphate metabolites were also summed to obtain total concentrations of dimethyl and diethyl phosphate metabolites. For eight women, the level for one of the six metabolites was not readable because of analytic interference. Because metabolites within each group (i.e., diethyl or dimethyl phosphates) were highly correlated, the missing values were imputed using regression analysis to predict the missing metabolite level based on the other metabolites levels for that woman at that time point. Metabolite levels below the limit of detection (LOD) were given the value of the LOD divided by the square root of two (Hornung and Reed 1990).

Creatinine concentrations in urine were determined using a commercially available diagnostic enzyme method (Vitros CREA slides; Ortho Clinical Diagnostics, Raritan, NJ). Urine samples with creatinine levels < 10 mg/dL were considered too dilute for accurate analysis, and one measurement for one woman was excluded because of low creatinine levels.

Total dialkyl phosphate and dimethyl phosphate metabolite levels were available for 485 women, and diethyl phosphate metabolite levels were available for 486 women.

For pesticide-specific metabolites, the same spot urine samples were analyzed using analytic and QC procedures similar to those used for the dialkyl phosphate metabolites (Olsson et al. 2003). The metabolites measured were malathion dicarboxylic acid (MDA; derived from malathion); 4-nitrophenol (PNP; derived from methyl parathion, parathion, and other nonpesticide chemicals); TCPy (from chlorpyrifos and chlorpyrifos methyl); 2-diethylamino-4-hydroxy-6-methylpyrimidine (DEAMPY; from pirimiphos methyl); 2-isopropyl-4-methyl-6-hydroxypyrimidine (IMPY; from diazinon); 3-chloro-4-methyl-7-hydroxycoumarin (CMHC; from coumaphos and coumaphos methyl); and 5-chloro-1-isopropyl-3-hydroxytriazole (CIT; from isazophos and isazophos methyl). With the exception of PNP, all of these metabolites derive from parent organophosphate pesticide compounds alone (Table 1). As with dialkyl phosphates, metabolite levels below the LOD were given the value of the LOD divided by the square root of two.

Specific metabolite levels were available for 482 women for six metabolites and, because of technical problems, for 382 women for the metabolite MDA.

For cholinesterase, because organophosphate pesticides at high doses are known to depress acetylcholinesterase, we measured ChE (or whole blood ChE) and BChE (or plasma ChE) in maternal and umbilical cord blood. Blood was collected from mothers at the time of the second pregnancy interview and in the hospital before delivery. Umbilical cord blood was collected by delivery room staff.

Blood samples were analyzed for ChE and BChE using a modification of the Wilson et al. (2002) microtiter-plate–adapted assay based on the original Ellman procedure (Ellman et al. 1961). Blood samples were stabilized immediately at the time of collection by diluting them 1:10 with 0.1 M NaPO4 buffer (pH 8.0) containing 1% Triton X-100. Processed samples were stored at −80°C before being shipped on dry ice to the University of Washington, Seattle, for analysis. At the time of assay, the samples were thawed, mixed thoroughly, and diluted 1:25 with 0.1 M NaPO4 buffer (pH 8.0) without Triton X-100 for a final 1:250 dilution of sample. One hundred micro-liters of 1:250 diluted samples were distributed to the wells of a microtiter plate. The reaction was initiated with 100 μL of a 2× assay mix of either ChE or BChE substrate, resulting in 1 mM final substrate. Assays were followed continuously at ambient temperature (23°C) at 412 nm for 10 min in a SpectraMax PLUS Microplate Reader (Molecular Devices Corp, Sunnyvale, CA). The initial linear rates of hydrolysis were obtained in units of optical density per minute. The path length for each well of the plate was measured immediately after the reaction, and rates were converted to rates of change in A412 per minute, and then to units of enzyme activity per milliliter. Each assay was run in triplicate. If the rate values varied by more than 5%, the sample was reanalyzed. A standard ChE sample supplied by the laboratory of B. Wilson (Departments of Environmental Toxicology and Avian Sciences, University of California, Davis, CA) was used to assure interlaboratory standardization.

ChE and BChE analysis was performed on all samples available and stabilized for this analysis, for a total of 292 women during pregnancy, 357 women at delivery, and 340 umbilical cord bloods.

Definition of outcomes.

Infant birth weight, crown–heel length, and head circumference were obtained from hospital delivery logs and medical records. Infant ponderal index, a measure of proportionality of growth, was calculated as (birth weight in grams × 100)/(length in centimeters)3. Gestational age was obtained from medical records and was based on ultrasound procedures for 25% of women. Because ultrasound estimates of gestational age may mask intrauterine growth retardation, we also estimated gestational age based on the woman’s self-reported date of last menstrual period. Results were similar using both methods, and the medical record gestational age is reported, except where noted.

Low birth weight was defined as < 2,500 g. Preterm delivery was defined as birth at less than 37 completed weeks of gestation. An SGA birth was defined as birth weight < 10th percentile for gestational age according to ethnicity (Mexican American or non-Hispanic white), parity, and infant sex from national data (Overpeck et al. 1999).

Data analysis.

Linear regression models were used to test for associations between exposure measurements and length of gestation, birth weight, length, head circumference, and ponderal index. Logistic regression was used to test for associations between exposure measurements and low birth weight, preterm delivery, and SGA births.

ChE and BChE were analyzed as continuous variables. Dialkyl phosphate metabolites were analyzed as continuous variables on a log10 scale. The pesticide-specific metabolite levels were analyzed as categorical variables because of the large proportion of women with nondetectable levels. For each pesticide-specific metabolite, women were assigned to one of three groups: no detectable levels (referent group), detectable levels below the median of the detectable levels, and detectable levels above the median. Associations with all urinary metabolites (dialkyl phosphates and pesticide-specific metabolites) were analyzed both adjusted and unadjusted for creatinine.

For analysis, the two pregnancy measurements of each urinary metabolite were averaged for each woman. This method was justified because there was no evidence of trend over time in metabolite levels and because there was a large within person variability in the metabolite measures, which was reduced by using the average. In addition, average metabolite level can be viewed as an approximate measure of cumulative pesticide dose over pregnancy. For specific metabolites, if a woman was below the LOD for both measurements, she was classified as below LOD for the average. If she was above the LOD at either or both measurements, her measurements were averaged and classified in relation to the median.

To identify “critical windows” of fetal development when exposure may have a greater impact, we analyzed the associations of outcomes and metabolite levels measured during moving 6-week windows of pregnancy (e.g., 5–10 weeks, 6–11 weeks, 7–12 weeks) using a series of multiple regression analyses. Six weeks was chosen because this time frame ensured a sample size of at least 100 women in each interval. When a woman had two measurements within a single interval, one measurement was randomly selected.

For analyses using gestational age as an outcome, metabolite levels were dropped for 19 women whose measurements occurred after 30 weeks gestation (the gestational duration of the earliest birth) to prevent a “survival” bias associated with a late metabolite measurement.

All models of birth weight, length, head circumference, and ponderal index were adjusted for gestational age and gestational age squared. We selected potential confounders for the multivariate models based on associations reported in the literature, and we included in the models those that changed the coefficient of exposure by 10% or more. The models included continuous variables for maternal age, pregnancy weight gain, and week of initiating prenatal care and categorical variables for parity, infant sex, mother’s country of birth, body mass index (BMI), and family income. Poverty level was calculated by dividing household income by the number of people supported by that income and comparing it with federal poverty thresholds (U.S. Census Bureau 2000). Smoking, alcohol, and illicit drug use were not included in the models because very few women reported use and controlling for these variables did not alter the results. The more commonly reported exposure to environmental tobacco smoke and caffeinated beverages also did not alter the relationship of pesticide metabolites and birth outcome and were not included. Additional analyses were conducted including history of low birth weight and history of preterm delivery, but these covariates were dropped from the final analyses because they did not affect the results. All analyses were conducted using Stata, version 8.0 (Stata Corporation, College Station, TX).

Results

Table 2 describes the sociodemographic characteristics of the population. The women averaged 25 years of age (SD = 5); two-thirds were multiparous, 80% were married, 79% had not graduated from high school, 58% were overweight or obese, 88% preferred to speak Spanish, and 84% were born in Mexico, with more than half residing in the United States for < 5 years (data not shown). Almost all of the women were living within 200% of the poverty level. Very few women reported smoking (6%), drug use (2%), or alcohol consumption (1%) during pregnancy. Approximately 28% of the women had worked in the fields during the pregnancy, and another 14% had worked at other jobs in agriculture, including packing shed, nursery, and greenhouse work. Overall, 85% of the women had agricultural workers living in their homes during their pregnancy (data not shown).

Table 1 shows the percentage of women with detectable levels of each urinary metabolite during pregnancy as well as the median values and ranges. The median dimethyl, diethyl, and total dialkyl phosphate metabolite levels for the study population were 101 nmol/L, 22 nmol/L, and 136 nmol/L, respectively. Only one woman had no detectable levels of dialkyl phosphate metabolites in urine during pregnancy. The percentages of women with detectable levels of MDA, TCPy, and PNP during pregnancy were 30, 77, and 54%, respectively. The median level for MDA was 0.2 μg/L, for TCPy was 3.3 μg/L, and for PNP was 0.5 μg/L. Because only a small percentage of women had levels above the LOD for the pesticide-specific metabolites DEAMPY, IMPY, CMHC, and CIT, we did not analyze their associations with birth outcomes.

The mean levels of ChE were 5.2 μmol/min/mL in maternal blood during pregnancy, 5.7 μmol/min/mL in maternal blood immediately before delivery, but somewhat lower at 3.8 μmol/min/mL in umbilical cord blood. For BChE, the mean levels were similar for all three. Maternal dialkyl phosphate metabolite levels and ChE levels collected concurrently at the second pregnancy interview were not correlated (Pearson r = 0.02, p = 0.71), and a small positive rather than the expected negative correlation was seen between average dialkyl phosphate metabolite levels during pregnancy and predelivery maternal blood (Pearson r = 0.11, p = 0.04) and umbilical cord ChE levels (Pearson r = 0.13, p = 0.02).

The mean (± SD) duration of gestation was 38.9 ± 1.7 weeks; mean birth weight was 3,449 ± 516 g; mean body (crown–heel) length was 50.2 ± 2.7 cm; mean head circumference was 34.1 ± 1.5 cm; and mean ponderal index was 2.7 ± 0.3 g/m3. A total of 3.7% (n = 18) of children were born of low birth weight; 4.8% (n = 23) were SGA births, and 6.6% (n = 32) were preterm.

Table 3 presents the adjusted regression results for dialkyl phosphate metabolite levels and pesticide-specific metabolite levels with measures of fetal growth and length of gestation. After adjusting for covariates, a 10-fold increase (i.e., one log-unit increase) in average dialkyl phosphate metabolite concentration was associated with an increase in infant’s body length of 0.52 cm (p = 0.06), and in head circumference of 0.32 cm (p = 0.03). Figure 1A–B indicates that these positive associations of dialkyl phosphate metabolite levels are related to exposure throughout the gestation period, although rarely significantly at any point for body length. Similar increases in body length and head circumference were seen when dimethyl and diethyl phosphate metabolites were examined separately, although these increases were not statistically significant (Table 3).

As shown in Table 3, a 10-fold increase in average dimethyl but not diethyl phosphate metabolites was associated with a decrease of 3 days in gestational duration (p = 0.02). Figure 1C shows that only after 22 weeks of gestation do increasing levels of dimethyl phosphate metabolites have a significant adverse association with gestational duration.

Dimethyl, diethyl, and total dialkyl phosphate metabolite levels were not associated with birth weight or infant ponderal index and were also unrelated to risk of preterm delivery, low birth weight, and SGA births. The findings of increased head circumference and decreased gestational duration persisted when metabolite levels were controlled for creatinine, but the finding of increased body length did not (data not shown).

No adverse associations were found between MDA or TCPy and parameters of fetal growth or gestational age. Increased head circumference was seen in infants of women with PNP levels above the median (β = 0.29 cm, p = 0.06) when compared with women with no detectable levels. Women with levels of PNP below the median also showed increased length (β = 0.60 cm, p = 0.03) compared with women with no detectable levels, but this effect was not observed in women with levels above the median. Borderline significant associations were seen between PNP and decreasing gestational age (β = −0.37 weeks, p = 0.06) and ponderal index (β = −0.08 g/cm3, p = 0.06), but only in the group below the median. However, associations seen with PNP should be viewed with caution because PNP may derive from compounds other than parathion (Table 1).

The associations between ChE levels in umbilical cord blood and parameters of fetal growth and length of gestation are shown in Table 4. Lower levels of ChE in umbilical cord blood were associated with significantly shorter length of gestation, averaging 0.34 weeks (p = 0.001) for each unit decrease in ChE (in micro-moles per minute per milliter; range of ChE in cord blood is 4.4 units). Decreasing levels of ChE in umbilical cord blood were also associated with increased risk of preterm delivery [adjusted odds ratio (OR) = 2.3; 95% confidence interval (CI), 1.1–4.8; p = 0.02] and low birth weight (adjusted OR = 4.3; 95% CI, 1.1–17.5; p = 0.04); however, 6 of the 11 low birth weight infants were also preterm (data not shown). Lower levels of ChE in maternal blood at delivery were associated with decreased gestational duration, but only when the estimate based on last menstrual period was used (β = 1.1 days, p = 0.04; data not shown). Lower ChE levels in maternal blood collected earlier in pregnancy were not associated with gestational duration but were somewhat associated with an increased risk of preterm delivery (OR = 1.6; 95% CI, 1.0–2.5; p = 0.06; data not shown). Neither maternal nor umbilical cord ChE levels were associated with any other parameters of fetal growth. BChE levels in maternal and umbilical cord blood were not associated with any birth outcome.

Discussion

We found clear decreases in gestational duration associated with two measures of in utero pesticide exposure: levels of metabolites of dimethyl phosphate pesticide compounds and whole blood ChE. Shortened gestational duration was most clearly related to increasing exposure levels in the latter part of pregnancy. However, the results of this study failed to demonstrate an adverse relationship between fetal growth and in utero organophosphate pesticide exposure as assessed by multiple measures of exposure, including plasma and whole blood cholinesterase, urinary dialkyl phosphate metabolites, and pesticide-specific metabolites of parathion, malathion, and chlorpyrifos. In fact, we found increases in length and head circumference associated with some of these measures.

Our results are consistent with those of Berkowitz et al. (2004), who found no adverse relationship between any measures of fetal growth or length of gestation and maternal urinary levels of TCPy, the metabolite of the diethyl organophosphate pesticide chlorpyrifos. Our results are also consistent with those of Willis et al. (1993), who reported no association of either fetal growth or length of gestation and maternal plasma BChE levels. However, our results differ from those of Perera et al. (2003), who reported decreased birth weight and length in association with blood measurements of the parent compound chlorpyrifos in pregnant residents of New York City.

Disparities among study results may be due, in part, to differences in exposure measurements. For example, in our study and in the study by Willis et al. (1993), there was no association between fetal outcome measures and plasma BChE levels, but we did observe clear associations with shortened gestation using whole blood ChE as the measure of exposure. Whole blood ChE reflects exposure to organophosphate and n-methyl carbamate pesticides over a few months, whereas plasma BChE reflects more immediate exposure (Karlsen et al. 1981; Lessenger and Reese 1999; Sanz et al. 1991; U.S. EPA 2000; Yuknavage et al. 1997). In our study, as in the study by Willis et al. (1993), we used the absolute level of cholinesterase as an indicator of exposure, rather than the change from a preexposure baseline level, as is typically used in occupational monitoring of pesticide poisoning. There is some evidence that absolute ChE measurements can be effective in establishing effects of organophosphate pesticide exposures. For example, this approach was used to show a significant inhibition of ChE in children exposed to rain water runoff from a large crop-dusting airport (McConnell et al. 1999).

Both the present study and the study by Berkowitz et al. (2004) found no association of the metabolite TCPy and fetal growth or length of gestation, although our methods of chemical analysis may have differed. Berkowitz et al. (2004) reported a detection frequency of 42% versus our report of 77%. Their lower detection frequency is not surprising given that our LOD was nearly 50 times lower. However, the median TCPy level in these New York City residents was more than twice (7.5 vs. 3.3 μg/L) that of our residents who lived in an agricultural community where > 50,000 lb of chlorpyrifos were applied annually to agricultural fields. The median level in both studies was considerably higher than the median (1.7 μg/L) from a stratified random sample of the U.S. population participating in the National Health and Nutrition Examination Survey (NHANES; National Center for Environmental Health 2003) and analyzed by the same CDC laboratory as samples in our study. In addition, the present study and both New York City studies (Berkowitz et al. 2004; Perera et al. 2003) straddled the time frame of the U.S. EPA ban on chlorpyrifos use in the home, beginning 1 January 2001 (U.S. EPA 2000). Although chlorpyrifos exposure from home use is likely to have had an important contribution to exposure in New York City, very few of the home pesticides found in the homes of CHAMACOS participants contained chlorpyrifos. Possible sources of chlorpyrifos exposure to our population include diet, residues in the home from past home pesticide use, or agricultural use, which was largely unaffecated by the U.S. EPA regulation.

Measurements of pesticides in blood, as in the study by Perera et al. (2003), is a direct measure of exposure to the parent compound and may more accurately reflect the dose to the target organ than measurements of metabolites in urine (Barr et al. 1999; Needham et al. 1995).

Although blood measurements may be preferable in certain cases, estimating organophosphate pesticide exposure with urinary levels of dialkyl phosphate metabolites has an important advantage beyond the ease of specimen collection. The dialkyl phosphate metabolites reflect exposure to about 80% of the organophosphate pesticides used in the Salinas Valley (California EPA 2000), although a number of highly used organophosphate pesticides (e.g., acephate) do not devolve into these urinary metabolites. Although dialkyl phosphate metabolites measurements do not allow differentiation between exposures that result from more or from less toxic pesticides [e.g., oxydemeton-methyl is orders of magnitude more acutely neurotoxic than is malathion; both may devolve to dimethyl metabolites (Olsson et al. 2003; Wessels et al. 2003)], they are an excellent nonspecific but integrated measure of exposure to a class of pesticides. We have partially addressed this limitation by complementing measurements of dialkyl phosphate metabolites with available analyses of pesticide-specific organophosphate metabolites. However, currently, there are no analytical methods for measurement of specific exposure to many important organophosphate pesticides (e.g., oxydemeton-methyl) in urine or in blood, and even some of the ones we can measure (e.g., PNP) may derive from other sources in addition to the pesticide of interest. Thus, measurement of dialkyl phosphate metabolites may be the only biologic measure currently available to characterize and integrate exposure to multiple organophosphate pesticides that may originate from different sources.

Current biomarkers used to assess pesticide exposure, whether in blood or urine, can result in exposure misclassification. Organophosphate pesticides and their metabolites have a short residence time in the body [Abu-Qare et al. 2001; Garfitt et al. 2002; Griffin et al. 1999; World Health Organization (WHO) 1996]. Exposures may be transient and highly variable. In fact, we found that the within-person standard deviation for the two urinary dialkyl phosphate metabolite measurements was approximately three times larger than the between-person standard deviation. Thus, measurements conducted on one [as in the studies of Berkowitz et al. (2004) and Perera et al. (2003)] or two (as in the present study) blood or urine specimens during pregnancy may not accurately reflect exposures over the entire pregnancy. Furthermore, the interrelationship of these different exposure measurements has not been well studied and may differ in pregnancy.

Understanding the mechanism of pesticide exposure and shortened gestational duration will require further examination of the interrelationship of different exposure measurements. For example, we found no correlation between concurrent measurements of dialkyl phosphate metabolites in urine and ChE in blood, and we found an unexpected small positive relationship between ChE and average dialkyl phosphate levels over pregnancy. The absence of a negative correlation between dialkyl phosphate metabolites and ChE is perhaps partially caused by substantial measurement error in both measures. The fact that decreases in gestational age were seen with both dimethyl phosphates and cord ChE may reflect a true association of organophosphate exposure and length of gestation, even though measurement error prevents these two markers of exposure from being negatively correlated with each other. An additional explanation for the lack of expected correlation between ChE and dialkyl phosphate metabolites is that dialkyl phosphate metabolites are specific to organophosphate compounds, whereas ChE levels may reflect exposure to both organophosphate and n-methyl carbamate pesticides. n-Methyl carbamate use in Monterey County in the year this study was conducted exceeded 100,000 lb (California EPA 2002) and may be a major contributor to ChE levels in this population. We note, however, that at least among the women who delivered prematurely, we found the anticipated negative correlation between dialkyl phosphate metabolites and ChE levels (r = −0.45, p = 0.05). Thus, population correlations may be masking associations in high-risk groups.

Despite difficulties in exposure assessment, the associations between pesticide exposure measures and gestational duration are quite compelling. The relation of ChE and shortened gestation may be biologically plausible. Cholinergic nerves play a significant role in the control of the uterine musculature and myometrium. Acetylcholine stimulates contraction of the uterus and dilates its arterial supply (Papka et al. 1999). Thus, an inhibition of acetyl ChE could produce an accumulation of acetylcholine in the neuronal junctions and hence the overstimulation of cholinergic fibers resulting in premature initiation of labor. Our results suggest that exposure to dimethyl organophosphate pesticides in the latter part of pregnancy may be particularly suspect. This is further supported by the observation that the urinary MDA levels (parent compound is malathion, a dimethyl organophosphate) during the latter part of pregnancy were also associated with significant increased risks for preterm delivery (OR = 5.2, 95% CI, 1.2–22.1, p = 0.03 for levels below the median; OR = 3.5, 95% CI, 0.9–13.3 p = 0.07 for levels above the median), although the numbers were small (data not shown). Nevertheless, the CHAMACOS cohort has a substantially lower rate of preterm delivery (6.4%) than that reported for Mexican-born women in the United States (10%) (Singh and Yu 1996). This suggests that although our findings are biologically intriguing, the potential effects of pesticides have had little clinical impact at the population level.

We have no ready explanation for an increase in some measures of fetal growth (i.e., head circumference and crown–heel length) in relation to pesticide exposure measures. These results were apparent only after controlling for gestational age. Berkowitz et al. (2004) reported a decrease in head circumference only in those infants whose mothers had low expression of PON1. Future investigations will determine whether PON1 status (genotype and phenotype) modifies the association of birth outcomes and pesticide exposure in the CHAMACOS cohort.

In summary, we have found no adverse association of in utero organophosphate pesticide exposure and measures of fetal growth, but a fairly consistent adverse association with gestational duration. The strengths of this study include the use of multiple exposure biomarkers in a large population of women living in an agricultural community. However, as in all studies of the effects of pesticide exposure, we are limited in our ability to accurately characterize exposure to multiple pesticides and multiple classes of pesticides over the course of pregnancy. Nevertheless, given the importance of premature delivery on the viability and health of the fetus, these findings warrant further evaluation of the risks associated with pesticide exposure, especially as new measures of exposure are developed. Furthermore, additional research should determine whether certain subpopulations are more biologically susceptible to the potential hazards of organophosphate exposure.

ReferencesAbu-QareAWAbdel-RahmanAAAhmadHKishkAMAbou-DoniaMB2001Absorption, distribution, metabolism and excretion of daily oral doses of [14C]methyl parathion in hensToxicol Lett12511011701217Abu-QareAWAbou-DoniaMB2000Urinary excretion of metabolites following a single dermal dose of [14C]methyl parathion in pregnant ratsToxicology15011912710996668AdgateJLBarrDBClaytonCAEberlyLEFreemanNCLioyPJ2001Measurement of children’s exposure to pesticides: analysis of urinary metabolite levels in a probability-based sampleEnviron Health Perspect10958359011445512BarrJRDriskellWJHillRHNeedhamLLHeadSL1999Strategies for biological monitoring of exposure for contemporary-use pesticidesToxicol Ind Health1516817910188199BerkowitzGSObelJDeychELapinskiRGodboldJLiuZ2003Exposure to indoor pesticides during pregnancy in a multiethnic, urban cohortEnviron Health Perspect111798412515682BerkowitzGSWetmurJBirman-DeychEObelJLapinskiRGodboldJ2004In utero pesticide exposure, maternal paraoxonase activity, and head circumferenceEnviron Health Perspect11238839114998758BoenigerMFLowryLKRosenbergJ1993Interpretation of urine results used to assess chemical exposure with emphasis on creatinine adjustments: a reviewAm Ind Hyg Assoc J546156278237794BradmanABarrDBClaus HennBGDrumhellerTCurryCEskenaziB2003Measurement of pesticides and other toxicants in amniotic fluid as a potential biomarker of prenatal exposure: a validation studyEnviron Health Perspect1111779178214594631BradmanMAHarnlyMEDraperWSeidelSTeranSWakehamD1997Pesticide exposures to children from California’s Central Valley: results of a pilot studyJ Expo Anal Environ Epidemiol72172349185013BradwayDEShafikTM1977Malathion exposure studies. Determination of mono- and dicarboxylic acids and alkyl phosphates in urineJ Agric Food Chem2513421344915135BravoRCaltabianoLWeerasekeraGWhiteheadRDFernandezCNeedhamLL In press. Measurement of dialkyl phosphate metabolites of organophosphorus pesticides in human urine using lyophilization with gas chromatography-tandem mass spectrometry and isotope dilution quantification. J Expo Anal Environ Epidemiol.BravoRDriskellWJWhiteheadRDJrNeedhamLLBarrDB2002Quantitation of dialkyl phosphate metabolites of organophosphate pesticides in human urine using GC-MS-MS with isotopic internal standardsJ Anal Toxicol2624525212166810California EPA 2002. Pesticide Use Reporting 2001 Summary Data. Sacramento, CA:Department of Pesticide Regulation, California Environmental Protection Agency. Available: http://www.cdpr.ca.gov/docs/pur/pur01rep/01_pur.htm [accessed 11 February 2004].ChandaSMHarpPLiuJPopeCN1995Comparative developmental and maternal neurotoxicity following acute gestational exposure to chlorpyrifos in ratsJ Toxicol Environ Health441892027531776ClemensGRHartnagelREBareJJThyssenJH1990Teratological, neurochemical, and postnatal neurobehavioral assessment of METASYSTOX-R, an organophosphate pesticide in the ratFundam Appl Toxicol141311432307312CurlCLFenskeRAKisselJCShiraiJHMoateTFGriffithW2002Evaluation of take-home organophosphorus pesticide exposure among agricultural workers and their childrenEnviron Health Perspect110A787A79212460819DabrowskiSHankeWPolanskaKMakowiec-DabrowskaTSobalaW2003Pesticide exposure and birthweight: an epidemiological study in Central PolandInt J Occup Med Environ Health16313912705715DonaldsonDKielyTGrubeA 2002. Pesticides Industry Sales and Usage 1998 and 1999 Market Estimates. Washington, DC:U.S. Environmental Protection Agency, Office of Prevention, Pesticides, and Toxic Substances, Office of Pesticide Programs.EllmanGLCourtneyKDAndresVJrFeather-StoneRM1961A new and rapid colorimetric determination of acetyl-cholinesterase activityBiochem Pharmacol7889513726518EskenaziEBradmanAGladstoneEJaramilloSBirchKHollandN2003CHAMACOS, a longitudinal birth cohort study: lessons from the fieldsJ Children’s Health1327FenskeRALuCBarrDNeedhamL2002Children’s exposure to chlorpyrifos and parathion in an agricultural community in central Washington StateEnviron Health Perspect11054955312003762FensterLCoyeMJ1990Birthweight of infants born to Hispanic women employed in agricultureArch Environ Health4546522317089GarfittSJJonesKMasonHJCockerJ2002Exposure to the organophosphate diazinon: data from a human volunteer study with oral and dermal dosesToxicol Lett13410511312191867GretherJKHarrisJANeutraRKizerKW1987Exposure to aerial malathion application and the occurrence of congenital anomalies and low birthweightAm J Public Health77100910103605467GriffinPMasonHHeywoodKCockerJ1999Oral and dermal absorption of chlorpyrifos: a human volunteer studyOccup Environ Med56101310341740HillRHJrHeadSLBakerSGreggMShealyDBBaileySL1995Pesticide residues in urine of adults living in the United States: reference range concentrationsEnviron Res71991088977618HornungRWReedLD1990Estimation of average concentration in the presence of nondetectable valuesAppl Occup Environ Hyg54651InstitorisLSirokiODesiI1995Immunotoxicity study of repeated small doses of dimethoate and methylparathion administered to rats over three generationsHum Exp Toxicol148798838588948International Classification of Diseases, 9th Revision, Clinical Modification 1989. Washington, DC:U.S. Department of Health and Human Services.KarlsenRLSterriSLyngaasSFonnumF1981Reference values for erythrocyte acetylcholinesterase and plasma cholinesterase activities in children, implications for organophosphate intoxicationScand J Clin Lab Invest413013027313514KristensenPIrgensLMAndersenAByeASSundheimL1997Gestational age, birth weight, and perinatal death among births to Norwegian farmers, 1967–1991Am J Epidemiol1463293389270412LessengerJEReeseBE1999Rational use of cholinesterase activity testing in pesticide poisoningJ Am Board Fam Pract1230731410477195LoewenherzCFenskeRASimcoxNJBellamyGKalmanD1997Biological monitoring of organophosphorus pesticide exposure among children of agricultural workers in central Washington StateEnviron Health Perspect105134413539405329LuCFenskeRASimcoxNJKalmanD2000Pesticide exposure of children in an agricultural community: evidence of household proximity to farmland and take home exposure pathwaysEnviron Res8429030211097803LuCKnutsonDEFisker-AndersenJFenskeRA2001Biological monitoring survey of organophosphorus pesticide exposure among preschool children in the Seattle metropolitan areaEnviron Health Perspect10929930311333193McCauleyLALasarevMRHigginsGRothleinJMunizJEbbertC2001Work characteristics and pesticide exposures among migrant agricultural families: a community-based research approachEnviron Health Perspect10953353811401767McConnellRPachecoFWahlbergKKleinWMalespinOMagnottiR1999Subclinical health effects of environmental pesticide contamination in a developing country: cholinesterase depression in childrenEnviron Res81879110433839MutoMALobelleFJrBidansetJHWurpelJN1992Embryotoxicity and neurotoxicity in rats associated with prenatal exposure to DURSBANVet Hum Toxicol344985011283795National Center for Environmental Health 2003. Second National Report on Human Exposure to Environmental Chemicals. Atlanta, GA:Centers for Disease Control and Prevention.National Research Council 1993. Pesticides in the Diets of Infants and Children. Washington, DC:National Academy of Sciences.NeedhamLLAshleyDLPattersonDGJr.1995Case studies of the use of biomarkers to assess exposuresToxicol Lett82–83373378NolanRJRickDLFreshourNLSaundersJH1984Chlorpyrifos: pharmacokinetics in human volunteersToxicol Appl Pharmacol738156200956OlssonAONguyenJVSadowskiMABarrDB2003A liquid chromatography/electrospray ionization-tandem mass spectrometry method for quantification of specific organophosphorus pesticide biomarkers in human urineAnal Bioanal Chem37680881512811448O’RourkeMKLizardiPSRoganSPFreemanNCAguirreASaintCG2000Pesticide exposure and creatinine variation among young childrenJ Expo Anal Environ Epidemiol1067268111138659OverpeckMDHedigerMLZhangJTrumbleACKlebanoffMA1999Birth weight for gestational age of Mexican American infants born in the United StatesObstet Gynecol9394394710362159PapkaRETraurigHHSchemannMCollinsJCopelinTWilsonK1999Cholinergic neurons of the pelvic autonomic ganglia and uterus of the female rat: distribution of axons and presence of muscarinic receptorsCell Tissue Res29629330510382273PereraFPRauhVTsaiWYKinneyPCamannDBarrD2003Effects of transplacental exposure to environmental pollutants on birth outcomes in a multiethnic populationEnviron Health Perspect11120120612573906RestrepoMMunozNDayNEParraJEde RomeroLNguyen-DinhX1990Prevalence of adverse reproductive outcomes in a population occupationally exposed to pesticides in ColombiaScand J Work Environ Health162322382389129SanzPRodriguez-VicenteMCDiazDRepettoJRepettoM1991Red blood cell and total blood acetylcholinesterase and plasma pseudocholinesterase in humans: observed variancesJ Toxicol Clin Toxicol2981902005669SavitzDAWhelanEAKlecknerRC1989Self-reported exposure to pesticides and radiation related to pregnancy outcome—results from National Natality and Fetal Mortality SurveysPublic Health Rep1044734772508175SimcoxNJCampJKalmanDStebbinsABellamyGLeeIC1999Farmworker exposure to organophosphorus pesticide residues during apple thinning in central Washington StateAm Ind Hyg Assoc J6075276110635541SinghGKYuSM1996Adverse pregnancy outcomes: differences between US- and foreign-born women in major US racial and ethnic groupsAm J Public Health868378438659659SpykerJMAveryDL1977Neurobehavioral effects of prenatal exposure to the organophosphate diazinon in miceJ Toxicol Environ Health39891002599594SrivastavaMKRaizadaRB1996Development effect of technical dimethoate in rats: maternal and fetal toxicity evaluationIndian J Exp Biol343293338698421SrivastavaMKRaizadaRBDikshithTS1992Fetotoxic response of technical quinalphos in ratsVet Hum Toxicol341311331509672ThomasDCPetittiDBGoldhaberMSwanSHRappaportEBHertz-PicciottoI1992Reproductive outcomes in relation to malathion spraying in the San Francisco Bay Area, 1981–1982Epidemiology332391554808U.S. Census Bureau 2000. Poverty Thresholds 2000, Current Population Survey. Washington, DC:U.S. Census Bureau. Available: http://www.census.gov/hhes/poverty/poverty00/pv00thrs.html [accessed 5 March 2003].U.S. EPA 2000. Science Policy on the Use of Data on Cholinesterase Inhibition for Risk Assessments of Organophosphorous and Carbamate Pesticides. Washington, DC:Office of Pesticide Programs. Available: http://www.epa.gov/pesticides/trac/science/cholin.pdf [accessed 12 November 2002].WesselsDBarrDBMendolaP2003Use of biomarkers to indicate exposure of children to organophosphate pesticides: implications for a longitudinal study of children’s environmental healthEnviron Health Perspect1111939194614644670WestgardJO 2002. Basic QC Practices: Training in Statistical Quality Control for Health Care Laboratories. Madison, WI:Westgard QC, Inc.WHO 1996. Biological Monitoring of Chemical Exposures in the Workplace. Vol. 1. Geneva:World Health Organization.WhyattRMCamannDEKinneyPLReyesARamirezJDietrichJ2002Residential pesticide use during pregnancy among a cohort of urban minority womenEnviron Health Perspect11050751412003754WillisWOde PeysterAMolgaardCAWalkerCMacKendrickT1993Pregnancy outcome among women exposed to pesticides through work or residence in an agricultural areaJ Occup Med359439498229348WilsonBWHendersonJDRamirezAO’MalleyMA2002Standardization of clinical cholinesterase measurementsInt J Toxicol2138538812396684WilsonNKChuangJCLyuCMentonRMorganMK2003Aggregate exposures of nine preschool children to persistent organic pollutants at day care and at homeJ Expo Anal Environ Epidemiol1318720212743613XiangHNuckolsJRStallonesL2000A geographic information assessment of birth weight and crop production patterns around mother’s residenceEnviron Res8216016710662530YuknavageKLFenskeRAKalmanDAKeiferMCFurlongCE1997Simulated dermal contamination with capillary samples and field cholinesterase biomonitoringJ Toxicol Environ Health5135559169060Figures and TablesAdjusted regression coefficients (solid lines) and 95% CIs (dashed lines) for the association of urinary dialkyl phosphate metabolites (log<sub>10</sub> scale) and (<italic>A</italic>) crown–heel length, (<italic>B</italic>) head circumference, and (<italic>C</italic>) the association of dimethyl phosphates and length of gestation according to timing of exposure during pregnancy. Regression coefficients were adjusted to control for timing of urine collection, week of entry to prenatal care, maternal age, parity, infant sex, maternal BMI, maternal weight gain, country of birth, and poverty level.

Number of women with measurements and percentage detectable, median values, and ranges (average of two measurements) of various organophosphate exposure measures during pregnancy: CHAMACOS study, Salinas Valley, California, 2000–2001.

Marker of exposureParent compounds or classNo.LODPercent > LODaMedian (range)bSample measured
Dialkyl phosphate metabolites (nmol/L)
 Dimethyl phosphatesMalathion, oxydemeton-methyl, dimethoate, naled, methidathionc4860.08–1.299.8101 (5–6,587)Urine
 Diethyl phosphatesDiazinon, chlorpyrifos, disulfoton4850.05–0.899.822 (2–680)Urine
 Total dialkyl phosphatesAll of above4850.05–1.299.8136 (10–6,854)Urine
Pesticide-specific metabolites (μg/L)
 MDAMalathion3820.2930.10.2 (0.2–28.9)Urine
 TCPyChlorpyrifos, chlorpyrifos methyl4820.2676.33.3 (0.2–56.1)Urine
 PNPMethyl parathion, parathion, EPNd4820.1454.40.5 (0.1–34.7)Urine
 DEAMPYPirimiphos methyl4820.21–0.225.00.2 (0.1–14.9)Urine
 IMPYDiazinon4820.692.40.5 (0.5–7.1)Urine
 CMHCCoumaphos, coumaphos methyl4820.180.70.1 (0.1–0.3)Urine
 CITIsazophos, isazophos methyl4821.5010.91.1 (1.1–36.0)Urine
Cholinesterase (μmol/min/mL)
 ChEOrganophosphates and n-methyl carbamates340NANA3.8 (1.7–6.1)Whole blood (cord)
357NANA5.1 (0.7–10.2)Whole blood (maternal delivery)
292NANA5.7 (2.2–10.9)Whole blood (maternal pregnancy)
 BChEOrganophosphates and n-methyl carbamates340NANA1.2 (0.6–2.7)Plasma (cord)
357NANA1.4 (0.6–3.9)Plasma (maternal delivery)
292NANA1.4 (0.3–2.7)Plasma (maternal pregnancy)

NA, Not applicable.

Percentage of women with metabolite levels above the LOD for at least one measurement during pregnancy.

Urinary metabolites: average of two pregnancy measurements, not adjusted for creatinine.

Only parent compounds with annual use in Salinas Valley > 10,000 lb are listed.

PNP may also derive from nonpesticide chemicals used in industrial processes.

Demographic characteristics of CHAMA-COS study population, Salinas Valley, California, 2000–2001 (n = 488).

CharacteristicsNo. (%)
Age (years)
 18–24237 (48.6)
 25–29151 (30.9)
 30–3471 (14.5)
 ≥ 3529 (5.9)
Parity
 0162 (32.5)
 ≥ 1336 (67.5)
Education
 < 6th grade205 (42.0)
 7th–12th grade180 (36.9)
 Completed high school103 (21.1)
Marital status
 Married/living as married391 (80.1)
 Single97 (19.9)
Preferred language
 Spanish429 (87.9)
 English30 (6.1)
 Both24 (4.9)
 Other5 (1.0)
Country of birth
 Mexico409 (83.8)
 United States68 (13.9)
 Other11 (2.3)
Family income
 ≤ Poverty level282 (61.3)
 Within 200% of poverty level161 (35.0)
 > 200% of poverty level17 (3.7)
Body mass index (kg/m2)
 Underweight (< 18.5)176 (37.6)
 Normal (18.5–24.9)194 (41.5)
 Overweight (25–29.9)2 (0.4)
 Obese (> 30)96 (20.5)
Pregnancy weight gain (lbs)
 < 25159 (32.6)
 25–35173 (35.5)
 > 35156 (32.0)
Smoked during pregnancy
 Yes30 (6.1)
 No458 (93.9)
History of preterm/low-birth-weight delivery
 Not applicable162 (37.6)
 Neither217 (50.3)
 Preterm only14 (3.2)
 Low birth weight only17 (3.9)
 Both21 (4.9)
Work status during pregnancy
 Not working170 (35.9)
 Working in fields133 (28.1)
 Other agricultural work65 (13.7)
 Other work (not agricultural)105 (22.2)

Association of average urinary metabolites of organophosphate pesticides measured at two points during pregnancya with length of gestation and fetal growth: CHAMACOS study, Salinas Valley, California, 2000–2001.

Length of gestation (weeks)c
Birth weight (g)d
Length (cm)d
Head circumference (cm)d
Ponderal index (g/cm3)d
MetaboliteNo.bβ (95% CI)p-Valueβ (95% CI)p-Valueβ (95% CI)p-Valueβ (95% CI)p-Valueβ (95% CI)p-Value
Dialkyl phosphate metabolites (nmol/L, log10 scale)
 Dimethyl phosphates485−0.41 (0.75–0.07)0.02**41 (40–122)0.320.42 (0.07–0.91)0.09*0.25 (0.02–0.52)0.07*−0.03 (0.10–0.04)0.45
 Diethyl phosphates486−0.16 (0.53–0.22)0.4152 (40–144)0.260.40 (0.15–0.94)0.160.28 (0.02–0.59)0.07*−0.01 (0.09–0.07)0.74
 Total dialkyl phosphates485−0.20 (0.55–0.15)0.2742 (46–131)0.350.52 (0.01–1.05)0.06*0.32 (0.03–0.62)0.03**−0.04 (0.12–0.04)0.28
Pesticide-specific metabolites (μg/L)
 MDA
  No detectable levels233ReferentReferentReferentReferentReferent
  Detectable levels < median74−0.13 (0.55–0.30)0.55−45 (154–63)0.41−0.53 (1.18–0.11)0.11−0.16 (0.52–0.19)0.370.05 (0.05–0.14)0.33
  Detectable levels ≥ median75−0.21 (0.62–0.20)0.3256 (49–161)0.290.14 (0.48–0.76)0.660.11 (0.24–0.46)0.530.02 (0.07–0.12)0.60
 TCPy
  No detectable levels41ReferentReferentReferentReferentReferent
  Detectable levels < median220−0.17 (0.74–0.40)0.55−6 (138–126)0.930.09 (0.70–0.87)0.830.06 (0.37–0.49)0.78−0.01 (0.12–0.11)0.89
  Detectable levels ≥ median221−0.06 (0.63–0.51)0.8427 (106–159)0.690.44 (0.35–1.22)0.270.04 (0.39–0.47)0.85−0.04 (0.16–0.08)0.50
 PNP
  No detectable levels124ReferentReferentReferentReferentReferent
  Detectable levels < median179−0.37 (0.76–0.02)0.06*34 (57–125)0.460.60 (0.06–1.13)0.03**0.18 (0.12–0.48)0.23−0.08 (0.16–0.0)0.06
  Detectable levels ≥ median1790.18 (0.21–0.57)0.3649 (42–140)0.290.41 (0.13–94)0.140.29 (0.01–0.58)0.06*−0.03 (0.11–0.05)0.48

Urinary metabolite levels are not adjusted for urinary creatinine concentration.

Numbers vary slightly for different outcomes due to missing data.

Models adjusted for timing of urine collection, timing of entry into prenatal care, maternal age, parity, country of birth, and poverty level.

Models adjusted for timing of urine collection, timing of entry into prenatal care, maternal age, parity, infant sex, country of birth, weight gain, BMI, poverty level, gestational age, and (gestational age)2.

p < 0.10;

p < 0.05.

Association of ChE and BChE in maternal blood during pregnancy and at delivery and in umbilical cord blood with length of gestation and fetal growth: CHAMACOS study, Salinas Valley, California, 2000–2001.

Length of gestation (weeks)b
Birth weight (g)c
Length (cm) c
Head circumference (cm)c
Ponderal index (g/cm3)c
No.aβ(95% CI)p-Valueβ (95% CI)p-Valueβ (95% CI)p-Valueβ (95% CI)p -Valueβ (95% CI)p -Value
ChE (μmol/min/mL)
 Maternal blood, pregnancy3400.01 (0.15–0.17)0.878 (35–52)0.710.05 (0.20–0.29)0.720.06 (0.09–0.21)0.450.00 (0.03–0.03)0.90
 Maternal blood, delivery3570.09 (0.04–0.23)0.166 (30–43)0.730.05 (0.17–0.27)0.67−0.07 (0.19–0.05)0.270.00 (0.03–0.03)0.95
 Cord blood2920.34 (0.13–0.55)0.001**12 (46–70)0.68−0.01 (0.36–0.34)0.95−0.04 (0.23–0.14)0.650.02 (0.03–0.07)0.43
BChE (μmol/min/mL)
 Maternal blood, pregnancy340−0.2 (0.64–0.27)0.4256 (67–179)0.370.07 (0.63–0.78)0.830.12 (0.31–0.56)0.580.03 (0.06–0.12)0.51
 Maternal blood, delivery357−0.1 (0.48–0.36)0.78−90 (206–25)0.130.05 (0.65–0.75)0.89−0.07 (0.45–0.31)0.73−0.07 (0.16–0.03)0.16
 Cord blood292−0.2 (0.78–0.32)0.41111 (35–257)0.140.23 (0.65–1.12)0.6−0.03 (0.50–0.45)0.910.05 (0.07–0.17)0.45

Numbers vary slightly for different outcomes due to missing data.

Models adjusted for timing of urine collection, timing of entry into prenatal care, maternal age, parity, country of birth, and poverty level.

Models adjusted for timing of urine collection, timing of entry into prenatal care, maternal age, parity, infant sex, country of birth, weight gain, BMI, poverty level, gestational age, and (gestational age)2.

p < 0.10;

p < 0.05.