National Academies Press: OpenBook

Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants (2005)

Chapter: 7 Reproductive and Developmental Outcomes

« Previous: 6 Cardiovascular Disease
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

7
REPRODUCTIVE AND DEVELOPMENTAL OUTCOMES

This chapter examines reproductive and developmental outcomes of exposure to fuel and combustion products. The outcomes of interest include infertility, preterm birth and low-birth rate, as well as birth defects and childhood cancers.

In previous reports, Institute of Medicine committees focused primarily on exposures that occurred in men and women before conception. Preconception exposure was considered the most relevant to the Gulf War veterans’ experience. Those committees assumed that pregnancies during Gulf War deployment would have been rare that pregnant women would have been immediately evacuated. A newly published study (Araneta et al. 2004), however, indicates that there were many more pregnancies than previously reported and that pregnancies were typically ascertained 2–6 weeks after conception. Thus, it is likely that most pregnant women would have been evacuated during the first 6–12 weeks of pregnancy; that inference led the present committee to expand its search of the epidemiologic literature to include reproductive outcomes, with relevant environmental exposures, during the first trimester of pregnancy.

STUDIES OF BIRTH DEFECTS IN GULF WAR VETERANS

Several studies investigated reproductive effects of service in the Gulf War. Most were of birth defects, and none analyzed whether effects were related to specific biologic or chemical compounds. Early studies did not find any adverse effects, but more recent ones have reported excess cardiac birth defects and other adverse reproductive outcomes in offspring of men and women who served in the Gulf War.

Early studies of Gulf War veterans failed to identify an excess of birth defects in offspring of deployed vs nondeployed veterans. A small study of two Mississippi National Guard units deployed to the Gulf War (n=282) found no excess rate of birth defects in National Guard members’ children compared with rates expected on the basis of surveillance systems and previous surveys (Penman et al. 1996). A much larger study of all live births in military hospitals (n=75,000) in 1991–1993, included a comparison population of births to nondeployed personnel. The risk of birth defects was the same in children of Gulf War personnel as in the control population (Cowan et al. 1997). The study was limited to military hospitals and thereby excluded persons not being cared for in those hospitals (for example, members of the National Guard, reservists, and those who left the military over the course of the study). National Guard

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

and reserve troops, as noted earlier, constituted a relatively high percentage of US troops deployed to the Gulf War. Anecdotal reports of an excess of Goldenhar syndrome, a rare congenital anomaly that affects the development of facial structures, prompted another study of birth defects. The syndrome is not specifically coded in the reporting of birth defects, so the authors reviewed medical records of birth-defect categories that would have subsumed the Goldenhar syndrome. Too few cases of the syndrome were found to support definitive conclusions (Araneta et al. 1997).

A large, population-based Department of Veterans Affairs study of 15,000 US Gulf War veterans vs 15,000 non-deployed veterans found that both male and female veterans self-reported higher rates of birth defects among liveborn infants, including “moderate to severe” defects (odds ratios [ORs] 1.8–2.8). The defects were grouped into broad categories; the largest (n=151) was described as “isolated anomaly”. Male veterans self-reported a higher rate of miscarriage among their partners (OR 1.62, 95% confidence interval [CI] 1.32–1.99) than did controls. Concerned about reporting bias, the investigators suggested that those observations be confirmed by a review of medical records (Kang et al. 2001).

A population-based study in several states captured births in all hospitals, both military and civilian, and matched birth certificates with military records during the period 1989–1993 (Araneta et al. 2003). The study measured the prevalence of birth defects among infants of Gulf War veterans and non-deployed veterans in states that conducted active case ascertainment of birth defects. Military record of 684,645 Gulf War veterans and 1,587,102 non-deployed veterans were electronically linked with 2,314,908 birth certificates from Arizona, Hawaii, Iowa, and selected counties of Arkansas, Califonia, and Georgia; 11,961 Gulf War veterans’ infants and 33,052 non-deployed veterans’ infants were identified. Of those, 450 infants had mothers who served in the Gulf War and 3966 had non-deployed veteran mothers. After examining 48 specific categories of birth defects, the study found a greater prevalence of three defects—tricuspid valve insufficiency (relative risk [RR] 2.7, 95% CI 1.1–6.6), aortic valve stenosis (RR 6.0, 95% CI 1.2–31.0), and renal agenesis (RR 2.4, 95% CI 0.7–8.3)—in infants conceived after the war by Gulf War-deployed men in comparison with non-Gulf War-deployed men. Aortic valve stenosis and renal agenesis had higher RRs among infants conceived after the war by Gulf War veteran men than among infants conceived before the war. The study also found a greater prevalence of hypospadias in male infants (RR 6.3, 95% CI 1.5–26.3) conceived during or after the war and born to female Gulf War veterans (in comparison with non-Gulf War-deployed females). The study was not designed to determine whether the excess risk was caused by environmental agents, and it should be interpreted as an exploratory study that investigated 48 categories of birth defects.

A recently published population-based UK study probed the prevalence of birth defects and fetal deaths in all UK Gulf War veterans and Gulf War-era controls (that is, veterans who were not deployed to the gulf war)—a total of 105,735 veterans—studied with a validated postal questionnaire (Doyle et al. 2004). The study period covered conception after the Gulf War and before November 1997. Male Gulf War veterans reported a higher risk of miscarriages in their partners than the comparison cohort (OR 1.4, 95% CI 1.3–1.5). They also reported a higher proportion of offspring with any type of malformation (OR 1.5, 95% CI 1.3–1.7). Examination by type of malformation revealed some evidence of an increased risk of malformation but it was weakened when the analyses were restricted to clinically confirmed conditions. Female veterans did not report an excess of miscarriage, and stillbirths and malformations were too few to be usefully analyzed.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Five reproductive outcomes (livebirth, stillbirth, spontaneous abortion, ectopic pregnancy, and induced abortion) were analyzed among female veterans during or after the Gulf War compared to nondeployed female veterans (Araneta et al. 2004). A postal survey was sent to every woman identified by the Department of Defense as having a pregnancy-related admission to a military hospital from August 2, 1990 to May 31, 1992. The responses were validated with medical records; it was observed that female veterans with post-Gulf War conceptions (n=292) were at increased risk for spontaneous abortion (OR 2.92, 95% CI 1.9–4.6) and ectopic pregnancy (OR 7.70, 95% CI 3.0–19.8) compared with nondeployed female veterans (n=427). No difference was found between pregnancies of Gulf War-deployed women and those of non-Gulf War-deployed women.

FUELS AND REPRODUCTIVE AND DEVELOPMENTAL OUTCOMES

In reviewing the literature on paternal and maternal exposure to fuels, the committee found several studies on adverse reproductive outcomes, including infertility, spontaneous abortion, childhood leukemia, neuroblastoma, and Prader-Willi syndrome—which may result from genetic alterations in either sperm or egg. The studies related to each of those outcomes are discussed below (Table 7.1).

TABLE 7.1 Selected Epidemiologic Studies—Reproductive Outcomes and Exposure to Fuel

Reference

Population

Exposed Cases

Estimated Relative Risk

Acute nonlymphocytic leukemia

Case-Control Study

Buckley et al. 1989

Paternal exposure to petroleum products

 

 

 

None

93

1.0

1–1,000 days duration

32

1.4 (0.8–2.5)

> 1,000 days duration

53

2.4 (1.3–4.1)

Before conception

NA

2.0 (p<0.05)

During gestation

NA

2.8 (p<0.05)

Leukemia

Case-Control study

Shu et al. 1988

Maternal occupational exposure during pregnancy

 

 

 

Gasoline

38

1.6 (0.8–3.1)

Diesel oil

16

1.4 (0.6–3.3)

Kerosene

16

1.4 (0.6–3.1)

Maternal occupational exposure during pregnancy by histopathologic cell type

 

 

Gasoline

 

 

ANLL

13

2.1 (1.1–4.3)

ALL

21

1.7 (1.0–3.0)

Kerosene

 

 

ANLL

6

2.3 (0.9–6.3)

ALL

9

1.5 (0.6–3.4)

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Reference

Population

Exposed Cases

Estimated Relative Risk

Neuroblastoma

Case-Control Studies

De Roos et al. 2001

Maternal occupational exposure

 

 

Nonvolatile hydrocarbons, self-reported exposure

26

1.2 (0.7–2.2)

 

Nonvolatile hydrocarbons, industrial hygienist reviewed exposure

12

1.1 (0.5–2.5)

Diesel fuel, self-reported exposure

12

1.3 (0.5–3.1)

Diesel fuel, industrial hygienist reviewed exposure

3

0.9 (0.2–4.4)

Gasoline, self-reported exposure

14

1.6 (0.6–3.8)

Gasoline, industrial hygienist reviewed exposure

3

0.8 (0.2–4.2)

Paternal occupational exposure

 

 

Nonvolatile hydrocarbons, self-reported exposure

130

1.3 (0.9–1.9)

Nonvolatile hydrocarbons, industrial hygienist reviewed exposure

91

1.5 (1.0–2.2)

Diesel fuel, self-reported exposure

72

1.2 (0.8–1.9)

Diesel fuel, industrial hygienist reviewed exposure

42

1.5 (0.8–2.6)

Kerosene, self-reported exposure

26

1.1 (0.6–2.0)

Kerosene, industrial hygienist reviewed exposure

16

1.0 (0.5–2.2)

Gasoline, self-reported exposure

77

0.8 (0.5–1.2)

Gasoline, industrial hygienist reviewed exposure

45

0.8 (0.5–1.3)

Adjusted for frequently co-occurring hydrocarbons and paints for paternal exposure to diesel fuel

NA

2.0 (1.0–4.3)

Kerr et al. 2000

Maternal exposure to petroleum

23

3.0 (1.5–6.1)

 

Paternal exposure to petroleum

53

1.8 (1.1–2.8)

Prader-Willi syndrome

Case-Control Study

Cassidy et al. 1989

Paternal occupational exposure to hydrocarbons

24

0.72 (0.28–1.81)

Infertility

Infertility is the inability to conceive after at least 12 months of unprotected intercourse (Rowe et al. 1993). It has been estimated that 10–15% of couples of reproductive age experience some form of infertility (Speroff et al. 1999). There are numerous risk factors for infertility including: advanced age, obesity in women, previous reproductive experiences, genetic factors, and such diseases as chlamydial infection in women and epididymitis in men (Templeton 2000). No studies of infertility in women and exposure to fuels met the committee’s inclusion criteria (see Chapter 2) but the committee found one study on semen characteristics.

In men, semen samples can be collected and used to assess sperm production, structure, and function as measures of the effect of exposure on the male reproductive system. A prospective study was conducted to evaluate the effects of mixed, low-level exposure to complex mixtures on reproductive potential in men (Lemasters et al. 1999). The study included 50 men

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

working on aircraft maintenance at an Air Force installation and eight nonexposed men at the same facility.

Semen quality was assessed at baseline, before entry into the exposed job, and at periods of 15 and 30 weeks post-baseline, during which the men were occupationally exposed to solvents or jet fuels (primarily JP4). Although those two periods were chosen to allow for two complete cycles of spermatogenesis, the distal regions of the epididymis contain sperm of different ages.

At baseline (before entry into the workplace) semen was collected but there was no exposure monitoring. For 15 and 30 weeks of occupational exposure, semen assays and exposure assessment—with industrial-hygiene monitoring and breath sampling—were conducted for the exposed group, and only semen assays were requested for the nonexposed office workers.

Overall, for most sperm measures, the mean values remained within normal range throughout the 30 weeks of exposure. The findings by cycle for all exposed subjects were compared with reference values, and all semen measures at all cycles were found to be similar to reference limits except for percent motile,1 which was consistently lower. The eight nonexposed subjects had sperm measures similar to the exposed group, and their percent motile was also lower at baseline than the reference. Thus, the findings indicate that exposure to jet fuel did not have an apparent effect on semen quality of aircraft-maintenance personnel. Two possible explanations for the findings are that the mixtures are not associated with spermatotoxic effects at low concentrations and that the exposure measured at one point may be inadequate for characterizing the true absorbed dose.

The strengths of the study were that analyses were adjusted for potential confounders, including alcohol, smoking, and caffeine. Limitations of the study include the appreciable differences in age among the men in the different job groups and the low statistical power.

Spontaneous Abortion

The most common cause of spontaneous abortion is a genetic abnormality of the embryo. Risk factors for spontaneous abortion include age, maternal illness, cigarette-smoking, alcohol use, use of some medications, and a previous spontaneous abortion. The risk of pregnancy loss is known to increase with maternal age, especially after the age of 30 or 35 years; and is also high in women under the age of 18 years. In a woman who has had one spontaneous abortion, the probability of a second is estimated to be 13–26%; and the probability of another increases with successive spontaneous abortions (Smith and Suess 1998). Several maternal occupational exposures have been associated with the risk of spontaneous abortion, including exposure to ethylene oxide, antineoplastic agents, and possibly anesthetic gases.

Spontaneous abortion was studied in women living near oil fields in the Amazon basin of Ecuador (San Sebastián et al. 2002). The water in the rivers and streams of that area is contaminated with oils and is used for drinking, cooking, and bathing. Women living in several communities downstream of oil fields (n=365) were compared with women living upstream and farther from the fields (n=283). The concentration of total petroleum hydrocarbons in drinking water were 0.02 to 2.88 ppm in exposed communities. After adjustment for confounders, women in exposed communities were more likely to report pregnancies ending in spontaneous abortion (OR 2.47, 95% CI 1.61–3.79, p<0.01). The multivariate analysis adjusted for age at interview, age at pregnancy, pregnancy order, year of pregnancy, and socioeconomic status (SES).

1  

Percent motile depends on time from ejaculation until sample analysis.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Limitations of the study include the assignment of the same exposure status, based on drinking-water measurements, to every woman in a given study area, and the potential for recall bias.

Childhood Cancers

The committee considered any health effect in the child of a veteran as a reproductive effect even if the manifestation was cancer. Childhood cancer is defined by the American Cancer Society as cancer diagnosed between birth and the age of 14 years. The causes of most childhood cancers are not well known, especially with regard to potential environmental risk factors. Some of the suggested risk factors are genetics, advanced maternal age, birthweight of more than 4000 g, prenatal viral exposure, and prenatal radiation exposure (Ross and Swensen 2000).

Developments in treatment and supportive care for some types of cancer have enabled 75% of afflicted children to survive 5 years or more, and mortality from all childhood cancers combined has declined by 50% since 1973. Although childhood cancer is rare, it is still the leading cause of death from disease in children up to 14 years old (ACS 1999).

Leukemia

Leukemia is the most common cancer in children and accounts for almost one-third of all cases of childhood cancer. Acute lymphocytic leukemia (ALL) is the most common leukemia in children, accounting for nearly 75% of all leukemia cases (ACS 2002b). Slightly more prevalent among white children and among boys, ALL generally occurs in early childhood, particularly at the age of 2–3 years. The 5-year survival rate of children who have ALL has increased to nearly 80%, primarily because of advances in treatment (ACS 2002b).

Most of the remaining childhood leukemia cases are categorized as acute myeloid leukemia (AML), which occurs most commonly in the first 2 years of life and less commonly among older children. Developments in treatment have improved the survival rate of children who have AML, who have 5-year survival rate of about 40% (ACS 2002b).

Prenatal exposure to radiation is known to be associated with the development of ALL (IARC 2000). Some genetic disorders—such as Li-Fraumeni syndrome, Down syndrome, and Klinefelter syndrome—also are associated with ALL, and chemotherapeutic agents are associated with secondary leukemia later in childhood or in adulthood.

A population-based case-control study of 204 children under the age of 18 years who had acute nonlymphocytic leukemia (ANLL) or AML examined occupational exposures of parents (Buckley et al. 1989). The cases were age-matched to population controls, and all the mothers were interviewed, as were the fathers if available. During the interviews, information was collected on demographics, occupational histories and exposures, household exposures, lifestyle factors, reproductive and medical histories, complications of delivery, and congenital abnormalities of the child and first-degree relatives. For each exposure, the nature of the exposure, duration, frequency, and timing in relation to the index pregnancy was asked of each parent. All exposures and related information were self-reported. During the interviews, the parents were asked to report occupational exposures to 52 agents, including petroleum products. Among fathers who had been exposed to petroleum products for more than 1,000 days, the OR for ANLL was 2.4 (95% CI 1.3–4.1; p trend=0.002) for exposure at any time before, during, or after the birth. The point estimate of the risk decreased to 2.0 for paternal exposure only before conception.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

A population-based case-control study of 309 children who had leukemia and 618 healthy population controls in urban Shanghai, China, was conducted (Shu et al. 1988). Controls were randomly selected from the Shanghai general population and matched on sex and age. Information on parental lifestyle, radiation exposure, medication use, birth characteristics, and parental occupation was collected through in-person interviews. Maternal and paternal occupations during pregnancy included working in the chemical industry or a related occupation, working in agriculture or metal refining and processing, being a physician or pharmacist, and working in pharmacy manufacturing. Paternal occupation during pregnancy had no influence on the occurrence of leukemia. There were increased adjusted ORs for maternal exposure to gasoline (OR 1.6, 95% CI 0.8–3.1), diesel oil (OR 1.4, 95% CI 0.6–3.3), and kerosene (OR 1.4, 95% CI 0.6–3.1). Risk by leukemia type showed an association between maternal gasoline exposure and ANLL (OR 2.1, 95% CI 1.1–4.3) and ALL (OR 1.7, 95% CI 1.0–3.0) and between exposure to kerosene and ANLL (OR 2.3, 95% CI 0.9–6.3) and ALL (OR 1.5, 95% CI 0.6–3.4). According to the authors, limitations of the study include recall bias (cases were diagnosed over a 12-year period, whereas controls were healthy children selected in 1985–1986) and interviewer bias (trained interviewers could not be blinded to case-control status). Furthermore, the control-selection procedures might have resulted in bias in the favoring of children from large residential areas. Finally, multiple comparisons in the study and lack of validation for parental x-ray, drug, or occupational exposure indicate the need for cautious interpretation.

Four additional studies examined the relationship between parental occupation and childhood leukemia (Gold et al. 1982; Hakulinen et al. 1976; Lowengart et al. 1987; van Steensel-Moll et al. 1985). They were limited by their inability to validate employment history and their lack of details on specific assessment of exposure as in the study by Shu et al. above (1988). The broad exposure group “hydrocarbon related” included many diverse occupations with exposures to chemicals in addition to hydrocarbons, therefore, it is difficult to draw conclusions on exposure to fuels. Three of those studies did not find increased risks of leukemia related to hydrocarbon- or petroleum-product-exposed parental occupations, either before or during pregnancy. The study of van Steensel-Moll (1985) found that maternal exposure to paint, petroleum products, and unspecified chemicals during pregnancy was related to childhood leukemia (RR 2.4, 95% CI 1.2–4.6). No particular periods of pregnancy were specified.

Central Nervous System Cancer

Central nervous system (CNS) cancers include malignant brain and spinal-cord tumors. Brain tumors are the second-most common group of cancers in children, accounting for about 20% of all childhood cancers. Common types of childhood brain tumors are astrocytomas (tumors originating in the brain cells), primitive neuroectodermal tumors (PNETs, tumors that develop from primitive stem cells), and germ-cell tumors. Neuroblastoma, a type of CNS tumor derived from embryonic neural crest cells, is a common form of cancer in children and accounts for 7–10% of all childhood cancers (ACS 2002a).

The etiology of childhood brain cancer appears to be multifactorial; there is no clear primary cause. Such genetic syndromes as Li-Fraumeni syndrome and von Recklinghausen disease are known to be associated with a modest fraction of these tumors. One well-established risk factor for the development of brain tumors is exposure to ionizing radiation, which can occur during the treatment of other cancers. Other factors—such as exposure to nitrates, aspartame, and electromagnetic fields—have been studied, but no conclusive evidence clearly implicates them

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

as causal factors. More than 50% of children with brain tumors (all types combined) survive over 5 years (ACS 2002a).

To study the effects of parental occupational exposure on the incidence of neuroblastoma among children, De Roos et al. (2001) conducted a case-control study of 538 incident cases of neuroblastoma in children under the age of 19 years who had a new and confirmed diagnosis of neuroblastoma and registered at 139 participating hospitals in the US and Canada. Cases were age-matched to controls 1:1 recruited with random-digit dialing. Telephone interviews were conducted with all the mothers, and fathers if available, to obtain information on demographic characteristics, occupational histories, and exposures to specific chemicals. For each job held during the 2-year period before a child’s date of birth, the mother and father were asked specifically about 65 chemicals, including the category of “nonvolatile hydrocarbons” and such specific agents as petroleum products, oils and lubricants, cutting oil, diesel fuel, kerosene, and lubricating oil or grease. All self-reported exposures were reviewed by an industrial hygienist in an attempt to reduce the number of incorrect reports of exposures.

Maternal exposure to most chemicals was not associated with neuroblastoma; however, paternal exposure to some hydrocarbons was associated with increased incidence of neuroblastoma. The OR for neuroblastoma related to self-reported paternal exposure to diesel fuels was 1.2 (CI 0.8–1.9), to kerosene 1.1 (CI 0.6–2.0), and to gasoline 0.8 (CI 0.5–1.2). The analyses were rerun using an industrial hygiene review to assign exposure; the OR for diesel fuel was 1.5 (CI 0.8–2.6), for kerosene 1.0 (CI 0.5–2.2), and for gasoline 0.8 (CI 0.5–1.3); adjustment did not indicate increased risk.

The authors noted that they were unable to explore effects of maternal and paternal exposure before conception or during pregnancy separately because exposures overlapped during those periods. The large number of chemicals considered and the retrospective assessment of exposure limit the strength of this study. Overall, the study found weak support for an association between paternal exposure to diesel fuel and neuroblastoma and no support for an association between maternal exposures to any of the chemicals and neuroblastoma.

In another case-control study, interviews were conducted with the mothers and fathers of 183 children 0–14 years old who had neuroblastoma and resided in New York state (excluding New York City) and with the parents of 372 controls selected from the New York state live-birth certificate registry and matched to cases on year of birth (Kerr et al. 2000). Occupational histories and exposure information on 25 specific physical and chemical compounds were collected for both parents. Information was collected on parental occupation during the entire pregnancy with the index child. Mothers reported both their own and the fathers’ occupational histories. An increased risk of neuroblastoma was associated with maternal and paternal exposure to petroleum (OR 3.0, 95% CI 1.5–6.1 and OR 1.8, 95% CI 1.1–2.8, respectively). According to the authors, limitations of the study include possible misclassification of job and exposure, the increased likelihood of chance findings due to the large numbers of multiple comparisons, potential interviewer bias because interviewers were not blinded to the disease status of the index child of the mother being interviewed, and collection of exposure information on both parents from mothers.

A case-control study of occupational exposures of parents with children who had astrocytoma (n=155) or PNET (n=166) was conducted (Bunin et al. 1994). Cases were compared with matched control children who were identified with random-digit dialing. Kerosene was the only exposure of relevance to this committee, but the study explored an array of occupational and lifestyle exposures. The study found that maternal exposure to kerosene

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

during pregnancy was associated with an increased risk of astrocytoma (OR 8.9, 95% CI 1.1–71.1, p=0.04). No specific information on the nature of the exposure was provided, and the study carries the potential for recall bias.

Prader-Willi Syndrome

Prader-Willi syndrome (PWS) is a neurogenetic disorder generally associated with an abnormality of chromosome 15. About 60–75% (Akefeldt et al. 1995) of patients with PWS have a cytogenetically visible deletion in the q11–q13 region of paternal chromosome 15. Because PWS is highly associated with the loss of paternally derived genetic material, some have speculated that environmental factors may lead to PWS in men’s offspring (Magenis 1988; Strakowski and Butler 1987).

The incidence of PWS is 1 in 12,000 affecting both sexes and various races equally. Children with PWS are often characterized as short and having mental retardation or learning disabilities, incomplete sexual development, behavior problems, low muscle tone, and an urge to eat constantly, which often leads to obesity (PWSA 2004).

Akefeldt et al (1995) conducted a case-control study of 15 PWS patients and 13 controls to assess the relationship between PWS and parental exposures to hydrocarbons, including gasoline. The PWS patients were referred by pediatric outpatient departments. The controls were children referred for obesity that had mental retardation but did not fulfill all the diagnostic criteria for PWS. The study did not include a “normal” control group. Both parents of each child completed a questionnaire about their exposure to environmental and occupational agents (for example, gasoline, petrochemicals, and pesticides). Seven of the 15 PWS fathers reported exposure to gasoline (p=0.01) compared with none in the control group. The fathers reported that they had been exposed for 5–17 years (mean, 9 years) before the children’s conceptions. Among the seven PWS children whose fathers had been exposed to gasoline, five had molecular genetic deletions. Four of the eight PWS children, whose fathers were not exposed to gasoline also had chromosomal deletions. One PWS mother and two control group mothers reported exposure to gasoline. The results of the study indicate that only gasoline, not hydrocarbons in general, is implicated in the etiology of PWS. However, the authors note that the small number of cases makes it difficult to obtain significant values. Given the difficulty in correctly recalling details about exposure, especially the duration of exposure, the possibility of errors cannot be excluded.

An earlier study found an association between PWS and paternal occupational hydrocarbon exposure at or about time of conception (Strakowski and Butler 1987). The study included 652 PWS patients identified from the National Prader-Willi Syndrome Association. To obtain controls with a genetic disease in which paternal environmental exposure was an unlikely causative factor, the authors chose fragile X syndrome (n=66) and Down syndrome (n=268). The average age of the fathers was 32 years, no different from the national average. Occupations were classified into 23 categories; 12 categories involved considerable occupational exposure to hydrocarbons, and five to lead. The percentage of fathers with unknown occupations was similar in the PWS group (7.1%) and in the controls (7.5%), and they were excluded from statistical calculations. Paternal occupational hydrocarbon exposure in the PWS group (20.8%) was greater than that in the controls (12.0%), (p-value <0.001).

Although the Akefeldt et al. and Strakowski and Butler studies found associations between PWS and paternal hydrocarbon or gasoline exposure, neither study collected information on potential confounders, such as paternal drug use, radiation exposure, or smoking

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

history. However, despite the lack of control for potential confounders, the consistent associations found in both studies, the presence of paternal chromosomal deletions in 60–75% of PWS patients, and studies showing associations between chromosomal aberrations and occupational exposure to gasoline (Carere et al. 1998; Khalil 1995; Santos-Mello and Cavalcante 1992) suggest that it is biologically reasonable that paternal exposure to hydrocarbons, such as gasoline, is a possible mechanism for paternally derived PWS. Studies have also shown that in nearly all cases involving an interstitial chromosomal deletion in PWS patients (Ledbetter and Cassidy 1988), parental chromosomes have been normal; thus the deletion in PWS patients might be a de novo event.

To explore the hypothesis that hydrocarbon exposure was associated with the chromosomal abnormality in PWS, Cassidy et al. (1989) conducted a study to determine whether there was a difference in the prevalence of occupational exposure to hydrocarbons at the time of conception between fathers of PWS children who have a 15q chromosomal deletion and fathers of PWS children who do not have such a deletion. Using job classification, they assigned exposure to hydrocarbons. Of the 53 fathers of children with 15q deletions, 24 (45%) were engaged in potentially hydrocarbon-exposed occupations compared with 15 (54%) of the 28 fathers of children with apparently normal chromosomes (OR 0.72, 95% CI 0.28–1.81). Although there were no differences in exposure history on the basis of the presence of 15q deletions, the authors recognize that they were unable to determine whether submicroscopic deletions may be present in the children without apparent chromosome abnormalities. They suggest that research on smaller, less visible microdeletions is needed to understand the relationship between hydrocarbon exposure and PWS.

Conclusion

Overall, it is difficult to reach conclusions on the epidemiologic studies of adverse reproductive outcomes and exposure to fuels. Assessment of findings is limited by the small number of studies available on each health outcome, the possibility of recall bias, and the lack of specificity of exposure to the agents of concern in this report.

The committee concludes, from its assessment of the epidemiologic literature, that there is inadequate/insufficient evidence to determine whether an association exists between exposure to fuels and adverse reproductive or developmental outcomes, including infertility, spontaneous abortion, childhood leukemia, central nervous system tumors, neuroblastoma, and Prader-Willi syndrome.

COMBUSTION PRODUCTS AND REPRODUCTIVE AND DEVELOPMENTAL OUTCOMES

This section covers the following outcomes that have been addressed in epidemiologic research in relation to combustion-product exposures: preterm birth, low birthweight (LBW), very low birthweight (VLBW), intrauterine growth retardation (IUGR), birth defects, and childhood cancers.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Adverse Pregnancy Outcomes

The committee sought information on whether maternal or paternal exposure to combustion products before conception or maternal exposure during the first 3 months (first trimester) of gestation affected pregnancy outcomes. A number of adverse outcomes of pregnancy have been studied for possible associations with exposure to combustion products in air pollution. Most studies examined adverse effects on live births including preterm birth, IUGR, and LBW, which have a pronounced influence on infant morbidity and mortality (Pschirrer and Monga 2000). In the studies covered in this chapter, IUGR and LBW were usually analyzed in relation to full-term births. Other studies examined stillbirths or birth defects. The studies reviewed in this section, conducted largely in heavily polluted cities worldwide, examined associations between adverse pregnancy outcomes and combustion products.

Preterm Births

Preterm birth (<37 weeks) is the second leading cause of infant mortality in the United States (MMWR 1999) and a major cause of infant morbidity. Known environmental risk factors include maternal cigarette-smoking and alcohol dependence; other risk factors are a history of preterm birth, maternal race, and multiple gestation (Pschirrer and Monga 2000). Most preterm births in the United States do not result in LBW (Ritz et al. 2000). At least seven studies, in different continents, have explored the effects of air pollution on preterm births (Table 7.2).

The effects of air pollution on preterm births were studied in a cohort of nearly 100,000 births in Los Angeles in 1989–1993 (Ritz et al. 2000). The exposure, based on collections at 17 air quality monitoring stations, was of averaged pollutant measures over distinct periods such as 1, 2, 4, 6, 8, 12, and 26 weeks before birth, and the entire pregnancy period. The authors also calculated average exposures in the first and second month of pregnancy. Pollutants of interest were carbon monoxide (CO), nitrogen dioxide (NO2), ozone (O3), and PM10 (particulate matter 10 μg in diameter). About 9% of infants in the sample were born preterm. For PM10, a 50-μg/m3 increase in concentration over the first month of pregnancy was associated with a 16% increase in the rate of preterm births (RR 1.16, 95% CI 1.06–1.26). For CO, a 3-ppm increase over the first month of pregnancy was associated with a 4% increase in the rate of preterm births (RR 1.04, 95% CI 1.01–1.09). This study was well designed, with analyses controlling for several known risk factors for preterm birth, including age, race, education, parity, access to prenatal care, and previous low-birthweight or preterm births. The authors adjusted for maternal smoking but reported that the adjustment was incomplete.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

TABLE 7.2 Preterm Birth and Combustion-Product Exposure

Reference

Population

Pollutants Measured and Exposure Period

Adjusted OR (95% CI or p) by Period and Pollutant

Adjustments

Limitations

Ritz et al. 2000

97,518 live births, Los Angeles, CA 1989–1993

PM10, CO, NO2, O3

PM10 per 50-μg increase

Month 1: RR 1.09, 95% CI 0.99–1.20 6 weeks before birth: RR 1.20, 95% CI 1.09–1.33

Maternal age, race, education, parity, access to prenatal care, previous low-weight or preterm births; season of birth or conception

Inadequate adjustment for maternal smoking

Month 1

Final 6 weeks

CO

Month 1: RR 1.04, 95% CI 0.99–1.09 6 weeks before birth: RR 1.12, 95% CI 1.08–1.7

NO2, O3

Nonsignificant

Bobak 2000

108,173 live births, Czech Republic 1991

SO2, TSP, NOx

SO2 per 50-μg increase:

Trimester 1: OR 1.27, 95% CI 1.16–1.39

Trimester 2: OR 1.25, 95% CI 1.14–1.38

Trimester 3: OR 1.24, 95% CI 1.13–1.36

Sex of child; parity, maternal age group, education, marital status, nationality; month of birth

No adjustment for maternal smoking

Trimesters 1–3

TSP per 50-μg increase

Trimester 1: OR 1.18, 95% CI 1.05–1.31

Trimester 2: OR 1.11, 95% CI 0.97–1.26

Trimester 3: OR 1.12, 95% CI 0.97–1.28

NOx per 50-μg increase

Trimester 1: OR 1.10, 95% CI 1.00–1.21

Trimester 2: OR 1.08, 95% CI 0.98–1.19

Trimester 3: OR 1.11, 95% CI 1.00–1.23

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Reference

Population

Pollutants Measured and Exposure Period

Adjusted OR (95% CI or p) by Period and Pollutant

Adjustments

Limitations

Liu et al. 2003

229,085 live births, Vancouver, Canada 1986–1998

SO2, NO2, CO, ozone

Month 1

Last month

SO2 per 5-ppb increase

Month 1: OR 0.95, 95% CI 0.88–1.03

Last month: OR 1.09, 95% CI 1.01–1.19

Maternal age, parity; infant sex, birth weight; birth season

No adjustment for maternal smoking

NO2

Month 1: OR 1.01, 95% CI 0.94–1.07

Last month: OR 1.08, 95% CI 0.99–1.17

CO per 1-ppm increase:

Month 1: OR 0.95, 95% CI 0.89–1.01

Last month: OR 1.08, 95% CI 1.01–1.15

O3

Month 1: OR 0.98, 95% CI 0.89–1.03

Last month: OR 0.93, 95% CI 0.86–1.00

Wihelm & Ritz 2003

Case-Control Los Angeles, CA 1994–1996

Distance-weighted traffic density (DWTD) Only third trimester specified

DWTD highest quintile Preterm RR 1.08, 95% CI 1.01–1.15

Infant sex; maternal age, race or ethnicity, education; birth season; year of analysis; SES, among other covariates

Could not adjust for active and passive smoking and diet; these are related to SES, which was included in models

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Bobak (2000) studied all singleton live births registered in 1991 by the Czech Republic (n=108,173). The effects of maternal exposure to sulfur dioxide (SO2), total suspended particulates (TSP), and nitrous oxides (NOx) were investigated. Daily measurements taken with air monitors in the district of each infant’s birth were collected by trimester. After controlling for sex of the child, parity, maternal age group, education, marital status, nationality, and month of birth, a slightly increased OR of premature birth was found for a 50-μg/m3 increase in mean concentration of each pollutant. For exposure during the first trimester, SO2 had an OR of 1.27 (95% CI 1.16–1.39); TSP had an OR of 1.18 (95% CI 1.05–1.31), and NOx an OR of 1.10 (95% CI 1.00–1.21). Associations with pollutants in the second and third trimester were similar to those found in the first trimester. The study did not adjust for maternal smoking, which is an important risk factor for preterm birth.

Liu et al (2003) studied the relationship between ambient air pollution and preterm birth, LBW, and IUGR among singleton live births in Vancouver, Canada. Ambient exposure to SO2, NO2, CO, and O3 were linked with data obtained from the live-birth database maintained by Statistics Canada. Maternal residence during pregnancy was used to explore exposure during several periods throughout pregnancy: the first, second, and third months of pregnancy and the last and next-to-last months of pregnancy. In addition, exposure during the three trimesters was calculated. They found increased adjusted ORs for preterm birth and SO2 (OR 1.09, 95% CI 1.01–1.19) and CO (1.08, 95% CI 1.01–1.15) during the last month of pregnancy but not during the first month. The association between preterm birth and SO2 and CO remained after adjustment for other copollutants. Although the data are not presented, the authors note that they did not find associations between PM10 and any of the outcomes. However, they had only 5 years of available data on PM10 because there were a small number of births during that period. Overall, the study was of high quality, but a potential limitation is the lack of control for maternal smoking, a risk factor for preterm birth.

A case-control study of preterm and LBW babies in relation to traffic density in Los Angeles County in 1994–1996 was conducted (Wilhelm and Ritz 2003). For each subject, the authors assigned an annual average exposure by using a model of the distance-weighted traffic density and the location of the subject’s residence. The traffic density was the same in each trimester. However, the effect was strongest for the last trimester. The magnitude of the preterm-birth effect was small, but there was an increase in preterm births, with an RR of 1.08 (95% CI 1.01–1.51) for the highest quintile of exposure. There was an increased risk of full-term LBW, but no exposure-response relationship was found. In these analyses, the authors adjusted for infant sex, maternal age, maternal race or ethnicity, maternal education, birth season, year of analysis, and other covariates but could not adjust for such potential covariates as active and passive smoking. Because maternal smoking is related to SES indicators, which were included in the models, smoking was probably at least partially accounted for in the multivariate analyses.

Xu et al. (1995) conducted a prospective cohort study of 25,370 pregnant women in Beijing, China, in 1988. Beijing has high ambient mean concentrations of SO2 (102 μg/m3) and TSP (375 μg/m3). Air-monitoring data on TSP and SO2 concentrations were collected, and their effect on preterm births was studied with a time-series analysis of lagged moving averages. After adjustment for several confounders—including season, maternal age, and residential area—increased risks of preterm birth with increased TSP and SO2 exposure were found. Although the study was well designed, it explored only acute effects of pollutant exposure up to 7 days before delivery.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

A case-control study of VLBW (<1500 grams at birth) was conducted in the state of Georgia (Rogers et al. 2000). VLBW babies were largely preterm, so the study was unable to examine the effects of air pollutants on fetal growth independently of the preterm effects. Comparison of 143 mothers of VLBW babies with 202 mothers of babies that weighed more than 2,500 g on the basis of birth records (1986–1988) and face-to-face interviews showed a dose-response relationship between VLBW and increasing maternal exposure to SO2 and TSP. Complex environmental-transport modeling was performed, and exposure at the birth home and annual average TSP and SO2 concentrations were estimated for each subject. Although associations were found between the risk of having a VLBW babies and exposures to SO2 and TSP above the 95th percentile, the study was unable to differentiate between exposures throughout pregnancy and those in the first trimester.

An ecologic design was used to compare Teplice, a highly polluted district in the Czech Republic (where high-sulfur coal is combusted, with Prachatice, a less polluted one (Sram et al. 1996). Teplice’s SO2 and TSP concentrations in winter 1993, for example, were comparable with those in the infamous London fog of 1952. The authors found a higher prevalence of preterm births (6.2% vs 3.4%, p<0.01) in Teplice, but they were not able to adjust for covariates. It was noted that preterm birth was highly related to maternal smoking status.

In summary, the well-designed study of Ritz et al. (2000) found evidence of a relationship between preterm birth and combustion-product exposure. Its analysis controlled for several known risk factors for preterm birth (such as maternal age, race, education, and access to prenatal care). Several other studies reviewed by the committee provide supportive evidence of a relationship (Bobak 2000; Liu et al. 2003; Wilhelm and Ritz 2003).

The committee concludes, from its assessment of the epidemiologic literature, that there is limited/suggestive evidence of an association between combustion-product exposure during pregnancy and preterm birth, but the data provided inadequate/insufficient evidence of an association between combustion-product exposure at specific periods during pregnancy (for example, first trimester) and preterm birth.

Low Birthweight and Intrauterine Growth Retardation

LBW (birthweight <2500 g) affects 5–8% of pregnancies in the United States (Ventura et al. 1998). Retarded fetal growth (IUGR) refers to birthweight falling below the 10th percentile of national standards. Its known risk factors include infant sex and race, maternal weight gain, cigarette-smoking, and alcohol consumption. LBW can be the result of either preterm birth or retarded fetal growth in a preterm or full-term birth. LBW is a measure of retarded fetal growth if a study’s analysis adjusts for gestational age or is restricted to full-term births. Most studies evaluated in this section examined full-term births for LBW and IUGR and excluded preterm births (Table 7.3).

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

TABLE 7.3 Low Birthweight or Intrauterine Growth Retardation and Combustion-Product Exposure

Reference

Population

Pollutant Measured and Exposure Period

Adjusted OR (95% CI or p) by Period and Pollutant

Adjustments

Limitations

Dejmek et al. 1999

Czech Republic 1994–1996

PM2.5 (low, <27 μg/m3; medium, 27 μg/m3 to <37 μg/m3; high, ≥37 μg/m3)

IUGR:

PM2.5

Month 1, medium level: OR 1.26, 95% CI 0.81–1.95

Month 1, high level: OR 2.11, 95% CI 1.20–3.70

Other pregnancy months nonsignificant

Year, season, smoking, maternal height, prepregnancy weight, completed high school

 

PM10 (low, <40 μg/m3; medium, 40 μg/m3 to <50 μg/m3; high, ≥50 μg/m3)

Each month of gestation

PM10

Month 1, medium level: OR 1.62, 95% CI 1.07–2.50

Month 1, high level: OR 2.64, 95% CI 1.48–4.71

Other pregnancy months nonsignificant

Dejmek et al. 2000 (same population as Dejmek et al 1999)

Czech Republic 1994–1998

PAH, PM10

IUGR:

PAH (Teplice)

Month 1, medium level: OR 1.59 95% CI 1.06–2.39

Month 1, high level: OR 2.15, 95% CI 1.27–3.63

Year, season, smoking, maternal height, prepregnancy weight, completed high school

PM10 (Teplice):

Month 1, medium level: OR 1.44 95% CI 1.03–2.02

Month 1, high level: OR 2.14 95% CI 1.42–3.23

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Reference

Population

Pollutant Measured and Exposure Period

Adjusted OR (95% CI or p) by Period and Pollutant

Adjustments

Limitations

Support Studies

Bobak 2000

126,752 live births, Czech Republic 1991

SO2, TSP, NO2

Trimesters 1–3

LBW:

Gestational age; sex of child, parity; maternal age group, education, marital status, nationality; month of birth

No adjustment for maternal smoking

SO2 per 50-μg increase

Trimester 1: OR 1.01, 95% CI 0.88–1.17

Trimester 2: OR 0.95, 95% CI 0.82–1.10

Trimester 3: OR 0.97, 95% CI 0.85–1.10

TSP per 50-μg increase

Trimester 1: OR 1.13, 95% CI 0.93–1.38

Trimester 2: OR 1.14, 95% CI 0.92–1.40

Trimester 3: OR 1.14, 95% CI 0.93–1.38

No association between pollutants and IUGR

Lee et al. 2003

388,105 live births, South Korea 1996–1998

CO, PM10

Trimesters 1–3

Months 1–5

Months 6–10

LBW:

Date, gestational age, infant sex, maternal age, parental education

No adjustment for maternal smoking

CO

Trimester 1: OR 1.04, 95% CI 1.01–1.07

Trimester 2: OR 1.04, 95% CI 1.00–1.06

Trimester 3: OR 0.96, 95% CI 0.93–0.99

Months 1–5: OR 1.06, 95% CI 0.98–1.14

Months 6–10: OR 0.88, 95% CI 0.79–0.99

PM10

Trimester 1: OR 1.03, 95% CI 1.00–1.07

Trimester 2: OR 1.04, 95% CI 1.00–1.08

Trimester 3: OR 1.00, 95% 0.95–1.04

Months 1–5: OR 1.04, 95% CI 1.01–1.08

Months 6–10: OR 0.94, 95% CI 0.85–1.05

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Reference

Population

Pollutant Measured and Exposure Period

Adjusted OR (95% CI or p) by Period and Pollutant

Adjustments

Limitations

Liu et al. 2003

229,095 live births, Vancouver, Canada 1986–1998

SO2, NO2, CO, Ozone

IUGR:

Maternal age, parity, infant sex, gestational age or birthweight, month of birth

 

Month 1

Last Month

SO2 per 5-ppb increase:

Month 1: OR 1.07, 95% CI 1.01–1.13

NO2 per 10-ppb increase:

Month 1: OR 1.05, 95% CI 1.01–1.11

CO per 1-ppm increase:

Month 1: OR 1.06, 95% CI 1.01–1.10

O3:

Month 1: OR 0.99, 95% CI 0.93–1.04

Maisonet et al. 2001

89,557 live births, northeastern United States 1994–1996

CO, PM10, SO2

LBW:

Gestational age; alcohol, smoking; maternal education, age, race or ethnicity, marital status, weight gain, previous terminations; infant sex; season; gestational age, and so on.

Trimesters 1–3

CO per 1-ppm increase

Trimester 1: OR 1.08, 95% CI 0.91–128

Trimester 2: OR 1.14, 95% CI 0.83–1.58

Trimester 3: OR 1.31, 95% CI 1.06–1.62

SO2 per 10-ppm increase

Trimester 1: OR 0.98, 95% CI 0.93–1.03

Trimester 2: OR 1.01, 95% CI 0.93–1.10

Trimester 3: OR 1.01, 95% CI 0.86–1.20

PM10 per 10-μg/m3 increase

Trimester 1: OR 0.93, 95% CI 0.85–1.00

Trimester 2: OR0.93, 95% CI 0.85–1.02

Trimester 3: OR0.96, 95% CI 0.88–1.06

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Reference

Population

Pollutant Measured and Exposure Period

Adjusted OR (95% CI or p) by Period and Pollutant

Adjustments

Limitations

Wang et al. 1997

74,671 live births, China 1988–1991

SO2, TSP

LBW:

Gestational age, residence, year of birth, maternal age, and infant sex.

No adjustment for maternal smoking, but few women in China during study period smoked, so smoking unlikely to be important confounder

Trimesters 1–3

SO2 per 100-μg/m3 increase

Trimesters 1, 2 NS after adjusting for trimester 3

Trimester 3: OR 1.11 95% CI 1.06–1.16

TSP per 100-μg/m3 increase

Trimesters 1, 2 NS after adjusting for Trimester 3

Trimester 3: OR 1.10 95% CI 1.05–1.14

Ritz & Yu 1999

125,573 live births, Los Angeles 1989–1993

CO

LBW:

Gestational age; female child; maternal race, education, age; no prenatal care; and so on

Did not examine trimesters 1–2

Trimester 3

CO:

2.2 to <5 ppm OR 1.04, 95% CI 0.96–1.13

≥5 ppm OR 1.22, 95% CI 1.03–1.44

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Two studies of full-term births were published in the Czech Republic (Dejmek et al. 1999, 2000). One of the primary sources of particles in those populations is coal combustion. The first study was of all live singleton births in the heavily polluted district of Teplice, Czech Republic, covering the period 1994–1996 (Dejmek et al. 1999), and the second study covered the years 1994–1998 (Dejmek et al. 2000). The first study examined the effects of PM10 exposure during each month of pregnancy. Of the 1,943 enrolled women who gave birth to full-term singleton infants (at 37–43 weeks of gestation), nearly 10% delivered a child who had IUGR. The study also collected information from questionnaires and medical records in addition to vital statistics and environmental monitoring. After adjustment for relevant confounders—including maternal smoking, year, and season—there were associations between IUGR and PM10 and PM2.5 exposure during the first month of pregnancy. In particular, the OR for medium exposure to PM10 was 1.62 (95% CI 1.07–2.50) and for high exposure 2.64 (95% CI 1.48–4.71); for PM2.5, the OR for medium exposure was 1.26 (95% CI 0.81–1.95) and for high exposure 2.11 (95% CI 1.20–3.70). ORs for exposures during the other 8 months of pregnancy were mostly close to 1.0. The study was well designed and statistically powerful, controlling for relevant risk factors and assessing first-trimester exposure separately from exposure in other periods.

Another study (Dejmek et al. 2000) examined whether exposure to polycyclic aromatic hydrocarbons (PAHs), usually bound to fine particles, was associated with IUGR in full-term births. The study included a comparison district (Prachatice) with lower pollution. This study was not an independent dataset from the earlier Dejmek et al. study discussed above. It expanded the period of study to 4 years and was designed to explore associations with components of the particles. In multivariate models that adjusted for confounders, higher PAH concentrations during the first month of gestation increased the risk of IUGR and the adjusted ORs were 1.59 (95% CI 1.06–2.39) for medium exposure and 2.15 (95% CI 1.27–3.63) for high exposure. The authors concluded that the risk of delivering a growth-retarded infant increased with the concentration of fine particles and PAHs in the first month of gestation.

Bobak (2000) conducted a study of all singleton live births registered in 1991 by the Czech Republic (n=126,752 singleton live births). The study linked Czech national birth-register data with area-based measures of air pollution in 67 districts where at least one pollutant was measured (n=108,173). Maternal exposures to outdoor SO2, TSP, and NO2 in each trimester of pregnancy were studied in relation to LBW, IUGR (<10th percentile of birthweight for gestational age and sex) and prematurity (results were given in the previous section). After adjustment for sex of infant, education, parity, age, marital status of mother, and month of birth, LBW was associated with SO2 and TSP (OR 1.2, 95% CI 1.11–1.30 and 1.15, 95% CI 1.07–1.24, respectively, for a 50-μg/m3 increase in mean SO2 and TSP concentrations in first trimester). When gestational age was also adjusted for, the relationship with SO2 was substantially weaker, but the TSP relationship remained strong although the confidence interval widened and included the null value. Prematurity was also associated with SO2, TSP, and NO2 concentrations during the first trimester (as described in the previous section). Associations in the second and third trimesters were similar to those found in the first trimester; this was expected because there were correlations between pollutants across trimesters. One potential limitation of the study is the lack of control for maternal smoking, a risk factor for LBW.

A study explored the relationship between LBW and exposure to air pollution in South Korea during different gestational phases (Lee et al. 2003). Birthweight was extracted from birth certificates for all full-term singletons born at 37–44 weeks gestational weeks (n=388,105). Air pollution monitoring data were used to assign exposure during each trimester and each month of

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

pregnancy. After adjustment for sex, birth order, maternal age, parental education level, and gestational age, first-trimester CO and PM10 exposure increased LBW risk (OR 1.04, 95% CI 1.01–1.07 and OR 1.03, 95% CI 1.00–1.07, respectively), as did second-trimester exposure to PM10, SO2, and NO2, for which ORs were generally between 1.03 and 1.06 with narrow confidence intervals reflecting the large sample size. There were positive (small) dose-response relationships between LBW and CO during the first trimester and LBW and PM10 and SO2 during the second trimester. Point estimates of effect in the third trimester ranged between 0.96 and 1.00. In the monthly analyses, the risk of LBW were higher for CO exposure in months 2–5 and for PM10 in months 2–4 than in later months. For SO2 and NO2, exposure in months 3–5 was associated with LBW. Further analyses, dividing exposure into the first 5 months and the last months also suggested that earlier exposure was associated with LBW. Exposure during the first 5 months was associated with LBW even if air pollution was low during the last 5 months. A potential limitation is the lack of control for maternal smoking, a risk factor for LBW. Overall, the study found evidence of an association between LBW and early-pregnancy exposure, primarily during the first 5 months.

Liu et al. (2003), as noted in the previous section, explored the relationship between ambient air pollution and preterm birth, LBW, and IUGR among singleton live births in Vancouver, Canada. IUGR was studied only in full-term births. The authors found an increased adjusted OR for LBW and maternal exposure to SO2 during the first month of pregnancy (OR 1.11, 95% CI 1.01–1.22 per 5-ppb increase). The LBW-SO2 association persisted even after adjustment for the other pollutants. IUGR was also associated with exposure to NO2, SO2, CO during the first month of pregnancy but not during the last month. IUGR was associated with SO2 and CO exposure during the first trimester. Overall, the study was of high quality. One potential limitation is the lack of control for maternal smoking, a risk factor for adverse pregnancy outcomes.

A study examined the relationship between ambient air pollution in six northeastern US cities and full-term LBW among 89,557 singleton live births (Maisonet et al. 2001). Average exposure during each trimester was estimated for each study subject on the basis of maternal residence from the birth certificate. In the multivariate analyses, an association was found between LBW and third-trimester CO (adjusted OR 1.31 per 1-ppm increase, 95% CI 1.06–1.62). The models adjusted for maternal smoking during pregnancy, alcohol consumption, maternal education, maternal age, maternal race or ethnicity, marital status, weight gain during pregnancy, previous terminations, infant sex, season of birth, prenatal care, gestational age, and other pollutants. No associations were found between LBW and first- or second-trimester CO exposure or between LBW and exposure to PM10 in any trimester. Second-trimester SO2 exposure was not associated with LBW when it was used as a continuous variable; there were some associations when second-trimester SO2 exposure was entered into the models in quintiles. This high-quality study was well designed and statistically powerful. It controlled for relevant risk factors and assessed first-trimester exposure separately from other periods, but found consistent associations only with third-trimester CO exposure. The ORs between LBW and first-trimester exposure to the three pollutants were consistently less than 1.0.

A community-based study of all pregnant women in four residential areas of Beijing, China, was conducted (Wang et al. 1997). First-parity single live full-term births (n=74,671) were studied in relation to average TSP and SO2 exposure at various times throughout pregnancy, means during each trimester, and lagged moving averages in weeks before birth. After adjustment, third-trimester exposure to TSP and SO2 was most strongly associated with

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

LBW: ORs increased by a factor of 1.11 for each 100-μg/m3 increase in SO2 (95% CI 1.06–1.16) and 1.10 (95% CI 1.05–1.14) for TSP. The other exposure variables, TSP and SO2 at other times, had little predictive value after adjustment for third-trimester exposure. Most analyses adjusted for gestational age, residence, year of birth, maternal age, and infant sex. Small negative associations between LBW and first- and second-trimester exposure suggested lower risk with higher exposure. Although maternal smoking was not adjusted for, the authors note that few women in China during the study period smoked. Residential exposure to combustion products from burning fuel or cooking are other potentially important confounders which were not adjusted for in this study.

Another study examined the effect of ambient CO on LBW among full-term infants in southern California (Ritz and Yu 1999). It limited exposure assessments to the third trimester because there was evidence that smoking-related effects are mediated by hypoxia during the last trimester. Although not directly informative on first-trimester exposures, the study supports associations between LBW and third trimester exposure. For CO exposure at more than 5.5 ppm, there was a 22% increase in LBW (OR 1.22, 95% CI 1.03–1.44).

Some published studies (for example, Bobak and Leon 1999; Sram et al. 1996; Vassilev et al. 2001a, 2001b) were cross-sectional or ecologic studies and did not differentiate between early-pregnancy exposure (first trimester) and exposure throughout pregnancy. One study (Perera et al. 2003) was only of third-trimester PAH exposure, and another (Wilhelm and Ritz 2003), although well designed, studied traffic patterns in relation to LBW but did not distinguish between first-trimester exposure and exposure throughout pregnancy.

It is not now possible to identify critical periods of gestation during which exposure is associated with increased risks of LBW and IUGR. The data among studies are not consistent: some studies found associations between LBW and early-pregnancy exposure to specific pollutants (for example, Dejmek et al. 1999), between LBW and late-pregnancy exposures to specific pollutants (for example, Maisonet et al. 2001; Ritz and Yu 1999; Wang et al. 1997), or between LBW and exposure to specific pollutants during both early and late pregnancy (for example, Dejmek et al. 2000); others failed to find associations between LBW or IUGR and either early or late exposure to specific pollutants (for example, Bobak 2000; Maisonet et al. 2001). Identifying a specific period of vulnerability is especially difficult because ambient exposures at different times are correlated, and most studies did not adjust for exposure at different points in pregnancy. One study (Lee et al. 2003) attempted to do that and found the most consistent associations with exposures during the first 5 months (that is, the first and second trimesters), a period that overlaps with but is longer than the period of Gulf War exposure. In conclusion, there is inadequate evidence to conclude that early-pregnancy exposure alone, independently of middle- and late-pregnancy exposure to air pollutants, is associated with full-term LBW and IUGR. Although the evidence on exposure during any time during pregnancy and adverse pregnancy end points is accumulating, further studies are needed to determine the relevant exposure time. Studies adjusting for exposure at different times during gestation would be especially informative.

The two studies conducted in the Czech Republic (Dejmek et al. 1999, 2000) were well designed studies and found evidence of a relationship between LBW or IUGR and combustion-product exposure. Their analyses controlled for several known risk factors, including maternal smoking. Several other studies reviewed by the committee provide supportive evidence of a relationship (Bobak 2000; Lee et al. 2003; Liu et al. 2003; Maisonet et al. 2001; Ritz and Yu 1999; Wang et al. 1997) but most were unable to adjust for maternal smoking.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

The committee concludes, from its assessment of the epidemiologic literature, that there is limited/suggestive evidence of an association between combustion product exposure during pregnancy and low birthweight or intrauterine growth retardation, but the data provided inadequate/insufficient evidence of an association between combustion-product exposure at specific periods during pregnancy (for example, the first trimester) and low birthweight and intrauterine growth retardation.

Birth Defects

The association between maternal exposure to air pollutants and the risk of birth defects in southern California was studied during 1987–1993 (Ritz et al. 2002). With a population-based, case-control method, average monthly exposure was assigned to birth-defect cases and control pregnancies (n=10,649) on the basis of ambient-air monitoring data related to maternal residence during gestation. Dose-response relationships were found between cardiac defects and increasing exposure to CO and O3 during the second month of gestation. In the case of CO, the risk of ventricular septal defect was increased (ORs per quartile of exposure 1.62–2.95). In the case of O3, the risk of aortic artery and valve defects, pulmonary artery and valve anomalies, and conotruncal defects was similarly increased.

Three studies examined the association between paternal employment as a firefighter and the risk of cardiac birth defects. A case-control study considered 20 birth defect groups in 22,192 live births in British Columbia (Olshan et al. 1990). After adjustment for paternal age and race and maternal age, increased ORs were reported for paternal firefighting and ventricular septal defects (OR 2.7, 95% CI 1.02–7.18) and atrial septal defects (OR 5.91, 95% CI 1.60–21.83), compared with all other occupations. Increased ORs for both types of cardiac defects were also observed when firemen were compared with policemen. The second comparison group was selected to reduce potential confounding due to lifestyle factors and SES. Because the study relied on limited occupational information from birth registers, it was not possible to identify whether the firefighter fathers were urban or rural. However, the investigators tested whether there was any relationship between the year of a child’s birth and paternal employment as firefighter, hypothesizing that the risk of birth defects might have increased with increasing use of synthetic materials over the study period of 1952–1973. The findings suggest that paternal employment as a firefighter increases a child’s risk of being born with a ventricular or atrial septal defect. No specific information was available about duration of paternal firefighting and potential confounding factors beyond paternal age and race and maternal age.

A cohort study of 836 infants of firefighters in Sweden was conducted (Kallen and Pradat 1992). There was no increased risk of cardiac defects (RR 0.93, 95% CI 0.38–1.92). Ventricular and atrial septal defects were examined separately, but no increased risk was found among infants fathered by firemen. The cohort, however, was relatively small and thus of limited statistical power.

In a study of maternal or paternal exposure, Bates et al. (1997) studied all types of birth defects (ICD codes 40–759) in the offspring of parents living in Rotorua, New Zealand, a city with high geothermal exposure to hydrogen sulfide. No excess birth defects were reported in comparison with residents in the rest of New Zealand. The Bates studies were the only epidemiologic studies of H2S found by the committee that examined long-term health outcomes. Due to the paucity of literature, the committee did not make a separate conclusion on H2S.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

The committee concludes, from its assessment of the epidemiologic literature, that there is inadequate/insufficient evidence of an association between maternal and paternal combustion product exposure prior to conception or maternal exposure during early pregnancy and specific birth defects, including cardiac effects.

Childhood Cancers

Several childhood cancers have been investigated in relation to combustion-product exposure before birth (that is, before or during gestation): brain cancer, leukemia, and neuroblastoma. Each of those can arise from embryonic cells or poorly differentiated cells. Most studies of childhood cancer used case-control designs and examined cancer in relation to parental exposure.

A California statewide, population-based cancer registry was used to identify cancers diagnosed in children who were less than 5 years old in 1988–1997 (Reynolds et al. 2004). The 4,369 cases were matched to 8,730 controls by birth date and sex. A mother’s residential address at the time of her child’s birth was used to calculate traffic density. For all cancer sites combined, the OR for the highest traffic-density exposure category compared with the lowest was 0.87 (95% CI 0.75–1.0). For all sites combined and for leukemia, the ORs were also below 1.0. For CNS tumors, the OR was 1.22 (CI 0.87–1.70). The authors found no increase in risk of cancer or leukemia among offspring of mothers living in high traffic density areas.

Raaschou-Nielsen et al. (2001) studied 1,989 children in the Danish Cancer Registry who had leukemia, CNS tumors, or malignant lymphoma diagnosed during 1968–1991 and 5,506 randomly selected control children from the entire childhood population. Residential histories of cases and controls were traced from 9 months before birth until the time of diagnosis. Traffic patterns and concentrations of benzene and NO2 were calculated for the relevant period, and exposures to air pollution during pregnancy and during childhood were calculated separately. The risks of leukemia, CNS tumors, and all selected cancers combined were not linked to benzene or NO2, but the risk of lymphomas (specifically Hodgkin’s disease) increased by 25% (p for trend=0.06) and 51% (p for trend=0.05) with a doubling of the concentration of benzene and NO2, respectively during pregnancy. The authors note that traffic-related air pollution at the residence does not appear to be associated with leukemias, CNS tumors, or NHL in children. The Hodgkin’s disease finding was of borderline significance in univariate models and may be a chance finding or due to multiple testing.

Results from the United Kingdom Childhood Cancer Study were analyzed (McKinney et al. 2003). Eligible cases were children who had a diagnosed malignant disease (including childhood leukemia, ALL, and brain cancer) or specified benign tumor. Cases were 0–14 years while a resident in England, Scotland, or Wales. Controls were randomly selected from the same family-health services in England and Wales and health boards in Scotland. There were 3,838 cases and 7,629 controls. Parents were interviewed to obtain an occupational history. Job titles and associated industries were coded according to the Standard Occupational Classification and Standard Industrial Classification of Economic Activities, respectively. The authors created 31 occupational groups by combining job titles considered to entail the same specific exposures. Timing of parental occupational exposure was also taken into account. When examining fathers occupationally exposed to combustion products at periconception, the authors found small increased risks for childhood leukemia and ALL. (Owing to substantial overlap between three occupational groups with “exhaust fumes” exposure, parents with some jobs may have been exposed to more than one of the agents and therefore included in more than one of the analyses.)

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

For ALL, paternal exposure to exhaust fumes at periconception had an OR of 1.26 (95% CI 1.02–1.56); exposure to inhaled particulate hydrocarbons, 1.41 (95% CI 1.11–1.79); and driving, 1.26 (95% CI 1.00–1.59). For leukemia, the ORs for paternal exposures were 1.33 (95% CI 1.09–1.61), 1.48 (95% CI 1.19–1.84), and 1.36 (95% CI 1.10–1.68), respectively. For CNS tumors, the ORs for paternal exposure to exhaust fumes and inhaled particulate hydrocarbons at periconception were 1.08 and 1.13, respectively, but the 95% CI included the null. For ALL, maternal exposure to exhaust fumes had an OR of 1.68 (95% CI 0.76–3.74); exposure to inhaled particulate hydrocarbons, 2.26 (95% CI 0.79–6.45); and driving, 1.74 (95% CI 0.63–4.85); but the 95% CIs included the null. The major limitations of this study are the nonspecific assessment of exposure based on occupation and the inability to account for other confounding exposures in the occupational groups. In addition, the creation of 31 occupational groups led to multiple comparisons and raised caution in interpreting the results. The strengths of the study include its size and population-based design.

Vianna et al. (1984) conducted a case-control study of acute leukemia and neuroblastoma. Trained interviewers questioned the mother of each patient and control. Several control groups were used, including children chosen from birth certificates and matched to cases on several risk factors. The neuroblastoma cases were also used as a control group to explore whether the quality of information obtained from parents of children with leukemia and the parents of controls without a malignancy might differ, assuming that neuroblastoma did not share the risk factors of interest with leukemia. Exposure to aromatic hydrocarbons from gasoline exhaust was defined as working full-time in a specified occupation for at least a year before the birth of the child. The authors did not find an association between exposure to aromatic hydrocarbons and increased risk of neuroblastoma, but did report a higher risk of leukemia in patients whose fathers worked in high-exposure occupations (p=0.007). The primary study limitation is reliance on occupational groups to assign measures of exposure to combustion products. However, that limitation would most likely bias results toward the null.

The relationship between parental occupation and leukemia and brain tumors in children was explored (Gold et al. 1982). A nonspecific measure of exposure to combustion products was used (motor-vehicle-related and including driver, motor-vehicle mechanic, service-station attendant, and railroad worker and engineer). There was an increased risk of leukemia in children whose fathers had vehicle-related occupations (p<0.05). No association was observed for brain cancer and vehicle-related occupations. The study is limited by the concurrent exposures to multiple agents in the fathers’ occupations.

Cordier et al. (1997) conducted a population-based, case-control study of childhood brain tumors (251 cases and 601 controls) in three European centers. A Job-Exposure Matrix (JEM) was used to estimate parental occupational exposure to PAHs during the 5-year period before birth. Paternal occupation described as motor-vehicle-related had an OR of 1.6 (95% CI 1.0–2.8). The subset of cases with primitive neuroectodermal tumors had an OR of 2.7 (95% CI 1.1–6.6). They also described increased risks, although no dose-response relationship, with paternal exposure specifically to PAHs; the OR for medium exposure 1.8 (95% CI 1.2–2.6), and the OR for high exposure was 1.3 (95% CI 0.8–2.0). Maternal occupation was not associated with increased risk. The study limitations include the use of maternal recall to measure maternal and paternal occupations and the nonspecificity of jobs categorized as PAH-exposed.

No association was found between childhood nervous system tumors and occupational groups broadly defined as hydrocarbon-related (Johnson et al. 1987). Another study (Hakulinen et al. 1976) found similar results in demonstrating no association between brain tumors,

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

leukemia, and lymphoma and paternal occupations described nonspecifically as hydrocarbon-related.

A case-control study of childhood leukemia and parental occupation did not find an increased risk with occupational exposure to exhaust gases (van Steensel-Moll et al. 1985), and another study (Zack et al. 1980) found no association between leukemia or lymphoma and paternal occupation as a motor-vehicle mechanic or service-station attendant. A study examining neuroblastoma and numerous paternal occupations, including firefighter and motor-vehicle operator, also found no increased risk (Olshan et al. 1999).

All the studies were limited by their inability to validate employment history. They also lacked details on specific assessment of exposure to combustion products. The broad exposure groups included many diverse occupations with exposure to other chemicals in addition to combustion products. In addition, the studies should be viewed cautiously because many of them conducted multiple comparisons. They are best used to generate hypotheses that require more precise assessment of exposure to combustion products.

The committee concludes, from its assessment of the epidemiologic literature, that there is inadequate/insufficient evidence of an association between parental combustion product exposure and all childhood cancers studied, including acute lymphocytic leukemia, leukemia, neuroblastoma, and brain cancer.

REFERENCES

ACS (American Cancer Society). 1999. Childhood Cancer—General Statement. [Online]. Available: http://www.cancer.org/downloads/CRI/Childhood%20Cancer%20%-20General%20Statement%20_PDF_.pdf [accessed February 21, 2002].

ACS. 2002a. ACS: Brain and Spinal Cord Tumors in Children. [Online]. Available: http://www.cancer.org/downloads/CRI/Brain%20and%20Spinal%20Cord%20Tumors%20in%20Children%20_PDF_.pdf [accessed February 21, 2002].

ACS. 2002b. Childhood Leukemia. [Online]. Available: http://www.cancer.org/downloads/CRI/Childhood%20Leukemia%20_PDF_.pdf [accessed February 21, 2002].

Akefeldt A, Anvret M, Grandell U, Nordlinder R, Gillberg C. 1995. Parental exposure to hydrocarbons in Prader-Willi syndrome. Developmental Medicine and Child Neurology 37(12):1101–1109.

Araneta MR, Kamens DR, Zau AC, Gastanaga VM, Schlangen KM, Hiliopoulos KM, Gray GC. 2004. Conception and pregnancy during the Persian Gulf War: The risk to women veterans. Annals of Epidemiology 14(2):109–116.

Araneta MR, Moore CA, Olney RS, Edmonds LD, Karcher JA, McDonough C, Hiliopoulos KM, Schlangen KM, Gray GC. 1997. Goldenhar syndrome among infants born in military hospitals to Gulf War veterans. Teratology 56(4):244–251.

Araneta MR, Schlangen KM, Edmonds LD, Destiche DA, Merz RD, Hobbs CA, Flood TJ, Harris JA, Krishnamurti D, Gray GC. 2003. Prevalence of birth defects among infants of Gulf War veterans in Arkansas, Arizona, California, Georgia, Hawaii, and Iowa, 1989–1993. Birth Defects Research Part A: Clinical and Molecular Teratology 67(4):246–260.


Bates MN, Garrett N, Graham B, Read D. 1997. Air pollution and mortality in the Rotorua geothermal area. Australian and New Zealand Journal of Public Health 21(6):581–586.

Bobak M, Leon DA. 1999. Pregnancy outcomes and outdoor air pollution: An ecological study in districts of the Czech Republic 1986–8. Occupational and Environmental Medicine 56(8):539–543.

Bobak M. 2000. Outdoor air pollution, low birth weight, and prematurity. Environmental Health Perspectives 108(2):173–176.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Buckley JD, Robison LL, Swotinsky R, Garabrant DH, LeBeau M, Manchester P, Nesbit ME, Odom L, Peters JM, Woods WG. 1989. Occupational exposures of parents of children with acute nonlymphocytic leukemia: A report from the Childrens Cancer Study Group. Cancer Research 49(14):4030–4037.

Bunin GR, Buckley JD, Boesel CP, Rorke LB, Meadows AT. 1994. Risk factors for astrocytic glioma and primitive neuroectodermal tumor of the brain in young children: A report from the Children’s Cancer Group. Cancer Epidemiology, Biomarkers & Prevention 3(3):197–204.


Carere A, Antoccia A, Cimini D, Crebelli R, Degrassi F, Leopardi P, Marcon F, Sgura A, Tanzarella C, Zijno A. 1998. Genetic effects of petroleum fuels: II. Analysis of chromosome loss and hyperploidy in peripheral lymphocytes of gasoline station attendants. Environmental and Molecular Mutagenesis 32(2):130–138.

Cassidy SB, Gainey AJ, Butler MG. 1989. Occupational hydrocarbon exposure among fathers of Prader-Willi syndrome patients with and without deletions of 15q. American Journal of Human Genetics 44(6):806–810.

Cordier S, Lefeuvre B, Filippini G, Peris-Bonet R, Farinotti M, Lovicu G, Mandereau L. 1997. Parental occupation, occupational exposure to solvents and polycyclic aromatic hydrocarbons and risk of childhood brain tumors (Italy, France, Spain). Cancer Causes & Control 8(5):688–697.

Cowan DN, DeFraites RF, Gray GC, Goldenbaum MB, Wishik SM. 1997. The risk of birth defects among children of Persian Gulf War veterans. New England Journal of Medicine 336(23):1650–1656.


De Roos AJ, Olshan AF, Teschke K, Poole C, Savitz DA, Blatt J, Bondy ML, Pollock BH. 2001. Parental occupational exposures to chemicals and incidence of neuroblastoma in offspring. American Journal of Epidemiology 154(2):106–114.

Dejmek J, Selevan SG, Benes I, Solansky I, Sram RJ. 1999. Fetal growth and maternal exposure to particulate matter during pregnancy. Environmental Health Perspectives 107(6):475–480.

Dejmek J, Solansky I, Benes I, Lenicek J, Sram RJ. 2000. The impact of polycyclic aromatic hydrocarbons and fine particles on pregnancy outcome. Environmental Health Perspectives 108(12):1159–1164.

Doyle P, Maconochie N, Davies G, Maconochie I, Pelerin M, Prior S, Lewis S. 2004. Miscarriage, stillbirth and congenital malformation in the offspring of UK veterans of the first Gulf war. International Journal of Epidemiology 33(1):74–86.


Gold EB, Diener MD, Szklo M. 1982. Parental occupations and cancer in children—a case-control study and review of the methodologic issues. Journal of Occupational Medicine 24(8):578–584.


Hakulinen T, Salonen T, Teppo L. 1976. Cancer in the offspring of fathers in hydrocarbon-related occupations. British Journal of Preventive and Social Medicine 30(2):138–140.


IARC (International Agency for Research on Cancer). 2000. Ionizing Radiation, Part 1: X- and Gamma (g)-Radiation, and Neutrons. 75. Lyon, France: IARC.


Johnson CC, Annegers JF, Frankowski RF, Spitz MR, Buffler PA. 1987. Childhood nervous system tumors—an evaluation of the association with paternal occupational exposure to hydrocarbons. American Journal of Epidemiology 126(4):605–613.


Kallen B, Pradat P. 1992. Re: “Birth defects among offspring of firemen”. American Journal of Epidemiology 135(11):1318–1320.

Kang H, Magee C, Mahan C, Lee K, Murphy F, Jackson L, Matanoski G. 2001. Pregnancy outcomes among U.S. Gulf War veterans: A population-based survey of 30,000 veterans. Annals of Epidemiology 11(7):504–511.

Kerr MA, Nasca PC, Mundt KA, Michalek AM, Baptiste MS, Mahoney MC. 2000. Parental occupational exposures and risk of neuroblastoma: A case-control study (United States). Cancer Causes and Control 11(7):635–643.

Khalil AM. 1995. Chromosome aberrations in blood lymphocytes from petroleum refinery workers. Archives of Environmental Contamination and Toxicology 28(2):236–239.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Ledbetter DH, Cassidy SB. 1988. Etiology of Prader-Willi Syndrome. In: Caldwell ML, Taylor FL, eds. Prader-Willi Syndrome: Selected Research and Management Issues. New York: Springer; as cited in Cassidy et al. 1989.

Lee BE, Ha EH, Park HS, Kim YJ, Hong YC, Kim H, Lee JT. 2003. Exposure to air pollution during different gestational phases contributes to risks of low birth weight. Human Reproduction 18(3):638–643.

Lemasters GK, Olsen DM, Yiin JH, Lockey JE, Shukla R, Selevan SG, Schrader SM, Toth GP, Evenson DP, Huszar GB. 1999. Male reproductive effects of solvent and fuel exposure during aircraft maintenance. Reproductive Toxicology 13(3):155–166.

Liu S, Krewski D, Shi Y, Chen Y, Burnett RT. 2003. Association between gaseous ambient air pollutants and adverse pregnancy outcomes in Vancouver, Canada. Environmental Health Perspectives 111(14):1773–1778.

Lowengart RA, Peters JM, Cicioni C, Buckley J, Bernstein L, Preston-Martin S, Rappaport E. 1987. Childhood leukemia and parents’ occupational and home exposures. Journal of the National Cancer Institute 79(1):39–46.


Magenis RE. 1988. On the origin of chromosome anomaly. American Journal of Human Genetics 42(4):529–533.

Maisonet M, Bush TJ, Correa A, Jaakkola JJ. 2001. Relation between ambient air pollution and low birth weight in the Northeastern United States. Environmental Health Perspectives 109(Suppl 3):351–356.

McKinney PA, Fear NT, Stockton D, UK Childhood Cancer Study Investigators. 2003. Parental occupation at periconception: Findings from the United Kingdom Childhood Cancer Study. Occupational and Environmental Medicine 60(12):901–909.

MMWR (Morbidity and Mortality Weekly Report). 1999. Preterm singleton births—United States, 1989–1996. Morbidity and Mortality Weekly Report 48(9):185–189.


Olshan AF, De Roos AJ, Teschke K, Neglia JP, Stram DO, Pollock BH, Castleberry RP. 1999. Neuroblastoma and parental occupation. Cancer Causes and Control 10(6):539–549.

Olshan AF, Teschke K, Baird PA. 1990. Birth defects among offspring of firemen. American Journal of Epidemiology 131(2):312–321.


Penman AD, Tarver RS, Currier MM. 1996. No evidence of increase in birth defects and health problems among children born to Persian Gulf War Veterans in Mississippi. Military Medicine 161(1):1–6.

Perera FP, Rauh V, Tsai WY, Kinney P, Camann D, Barr D, Bernert T, Garfinkel R, Tu YH, Diaz D, Dietrich J, Whyatt RM. 2003. Effects of transplacental exposure to environmental pollutants on birth outcomes in a multiethnic population. Envirnomental Health Perspectives 111(2):201–205.

Pschirrer ER, Monga M. 2000. Risk factors for preterm labor. Clinical Obstetrics and Gynecology 43(4):727–734.

PWSA (Prader-Willi Syndrome Association). 2004. Basic Facts About the Prader-Willi Syndrome. [Online]. Available: http://www.pwsausa.org/ [accessed September 13, 2004].


Raaschou-Nielsen O, Hertel O, Thomsen BL, Olsen JH. 2001. Air pollution from traffic at the residence of children with cancer. American Journal of Epidemiology 153(5):433–443.

Reynolds P, Von Behren J, Gunier RB, Goldberg DE, Hertz A. 2004. Residential exposure to traffic in California and childhood cancer. Epidemiology 15(1):6–12.

Ritz B, Yu F. 1999. The effect of ambient carbon monoxide on low birth weight among children born in Southern California between 1989 and 1993. Environmental Health Perspectives 107(1):17–25.

Ritz B, Yu F, Chapa G, Fruin S. 2000. Effect of air pollution on preterm birth among children born in Southern California between 1989 and 1993. Epidemiology 11(5):502–511.

Ritz B, Yu F, Fruin S, Chapa G, Shaw GM, Harris JA. 2002. Ambient air pollution and risk of birth defects in Southern California. American Journal of Epidemiology 155(1):17–25.

Rogers JF, Thompson SJ, Addy CL, McKeown RE, Cowen DJ, Decoufle P. 2000. Association of very low birth weight with exposures to environmental sulfur dioxide and total suspended particulates. American Journal of Epidemiology 151(6):602–613.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×

Ross JA, Swensen AR. 2000. Prenatal epidemiology of pediatric tumors. Current Oncology Reports 2(3):234–241.

Rowe PJ, Comhaire FH, Hargreave TB, Mellows HJ. 1993. WHO Manual for the Standardized Investigation and Diagnosis of the Infertile Couple. New York: Cambridge University Press.


San Sebastián M, Armstrong B, Stephens C. 2002. Outcomes of pregnancy among women living in the proximity of oil fields in the Amazon basin of Ecuador. International Journal of Occupational and Environmental Health 8(4):312–319.

Santos-Mello R, Cavalcante B. 1992. Cytogenetic studies on gas station attendants. Mutation Research 280(4):285–290.

Shu XO, Gao YT, Brinton LA, Linet MS, Tu JT, Zheng W, Fraumeni JF Jr. 1988. A population-based case-control study of childhood leukemia in Shanghai. Cancer 62(3):635–644.

Smith Ma, Suess JA. 1998. Obstetric complications during pregnancy. In: Taylor RB, ed. Family Medicine: Principles and Practice. 5th ed. New York: Springer. Pp. 106–121.

Speroff L, Glass RH, Kase NG. 1999. Clinical Gynecologic Endocrinology and Infertility. Philadelphia: Lippincott Williams & Wilkins.

Sram RJ, Benes I, Binkova B, Dejmek J, Horstman D, Kotesovec F, Otto D, Perreault SD, Rubes J, Selevan SG, Skalik I, Stevens RK, Lewtas J. 1996. Teplice Program—The impact of air pollution on human health. Environmental Health Perspectives 104(suppl. 4):699–714.

Strakowski SM, Butler MG. 1987. Paternal hydrocarbon exposure in Prader-Willi syndrome. Lancet 2(8573):1458.


Templeton A. 2000. Infertility and the establishment of pregnancy—overview. British Medical Bulletin 56(3):577–587.


van Steensel-Moll HA, Valkenburg HA, van Zanen GE. 1985. Childhood leukemia and parental occupation. A register-based case-control study. American Journal of Epidemiology 121(2):216–224.

Vassilev ZP, Robson MG, Klotz JB. 2001a. Outdoor exposure to airborne polycyclic organic matter and adverse reproductive outcomes: A pilot study. American Journal of Industrial Medicine 40(3):255–262.

Vassilev ZP, Robson MG, Klotz JB. 2001b. Associations of polycyclic organic matter in outdoor air with decreased birth weight: A pilot cross-sectional analysis. Journal of Toxicology and Environmental Health. Part A 64(8):595–605.

Ventura SJ, Martin JA, Curtin SC, Mathews TJ. 1998. Report of final natality statistics, 1996. Monthly vital statistics report. Hyattsville, MD: National Center for Health Statistics: Provisional statistics from the National Center for Health Statistics; as cited in Lee et al. 2003.

Vianna NJ, Kovasznay B, Polan A, Ju C. 1984. Infant leukemia and paternal exposure to motor vehicle exhaust fumes. Journal of Occupational Medicine 26(9):679–682.


Wang X, Ding H, Ryan L, Xu X. 1997. Association between air pollution and low birth weight: A community-based study. Environmental Health Perspectives 105(5):514–520.

Wilhelm M, Ritz B. 2003. Residential proximity to traffic and adverse birth outcomes in Los Angeles County, California, 1994–1996. Environmental Health Perspectives 111(2):207–216.


Xu X, Ding H, Wang X. 1995. Acute effects of total suspended particles and sulfur dioxides on preterm delivery: A community-based cohort study. Archives of Environmental Health 50(6):407–415.


Zack M, Cannon S, Loyd D, Heath CW Jr, Falletta JM, Jones B, Housworth J, Crowley S. 1980. Cancer in children of parents exposed to hydrocarbon-related industries and occupations. American Journal of Epidemiology 111(3):329–336.

Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 288
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 289
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 290
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 291
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 292
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 293
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 294
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 295
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 296
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 297
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 298
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 299
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 300
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 301
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 302
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 303
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 304
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 305
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 306
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 307
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 308
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 309
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 310
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 311
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 312
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 313
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 314
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 315
Suggested Citation:"7 Reproductive and Developmental Outcomes." Institute of Medicine. 2005. Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants. Washington, DC: The National Academies Press. doi: 10.17226/11180.
×
Page 316
Next: 8 Other Health Outcomes »
Gulf War and Health: Volume 3: Fuels, Combustion Products, and Propellants Get This Book
×
Buy Hardback | $85.00 Buy Ebook | $69.99
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

The third in a series of congressionally mandated reports on Gulf War veterans’ health, this volume evaluates the long-term, human health effects associated with exposure to selected environmental agents, pollutants, and synthetic chemical compounds believed to have been present during the Gulf War. The committee specifically evaluated the literature on hydrogen sulfide, combustion products, hydrazine and red fuming nitric acid. Both the epidemiologic and toxicologic literature were reviewed.

  1. ×

    Welcome to OpenBook!

    You're looking at OpenBook, NAP.edu's online reading room since 1999. Based on feedback from you, our users, we've made some improvements that make it easier than ever to read thousands of publications on our website.

    Do you want to take a quick tour of the OpenBook's features?

    No Thanks Take a Tour »
  2. ×

    Show this book's table of contents, where you can jump to any chapter by name.

    « Back Next »
  3. ×

    ...or use these buttons to go back to the previous chapter or skip to the next one.

    « Back Next »
  4. ×

    Jump up to the previous page or down to the next one. Also, you can type in a page number and press Enter to go directly to that page in the book.

    « Back Next »
  5. ×

    Switch between the Original Pages, where you can read the report as it appeared in print, and Text Pages for the web version, where you can highlight and search the text.

    « Back Next »
  6. ×

    To search the entire text of this book, type in your search term here and press Enter.

    « Back Next »
  7. ×

    Share a link to this book page on your preferred social network or via email.

    « Back Next »
  8. ×

    View our suggested citation for this chapter.

    « Back Next »
  9. ×

    Ready to take your reading offline? Click here to buy this book in print or download it as a free PDF, if available.

    « Back Next »
Stay Connected!