National Academies Press: OpenBook

Veterans and Agent Orange: Update 2006 (2007)

Chapter: 7 Reproductive and Developmental Effects

« Previous: 6 Cancer
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 517
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 518
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 519
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 520
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 521
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 522
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 523
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 524
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 525
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 526
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 527
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 528
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 529
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 530
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 531
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 532
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 533
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 534
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 535
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 536
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 537
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 538
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 539
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 540
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 541
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 542
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 543
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 544
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 545
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 546
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 547
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 548
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 549
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 550
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 551
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 552
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 553
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 554
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 555
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 556
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 557
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 558
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 559
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 560
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 561
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 562
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 563
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 564
Suggested Citation:"7 Reproductive and Developmental Effects." Institute of Medicine. 2007. Veterans and Agent Orange: Update 2006. Washington, DC: The National Academies Press. doi: 10.17226/11906.
×
Page 565

Below is the uncorrected machine-read text of this chapter, intended to provide our own search engines and external engines with highly rich, chapter-representative searchable text of each book. Because it is UNCORRECTED material, please consider the following text as a useful but insufficient proxy for the authoritative book pages.

7 Reproductive and Developmental Effects This chapter summarizes the scientific literature published since Veterans and Agent Orange: Update 2004, hereafter referred to as Update 2004 (IOM, 2005), on the association between exposure to herbicides and adverse reproduc- tive or developmental effects. (Analogous shortened names are used to refer to the updates for 1996, 1998, 2000, and 2002 [IOM, 1996, 1999, 2001, 2003].) The categories of association and the approach to categorizing the health outcomes are discussed in Chapters 1 and 2. The literature discussed includes papers that describe environmental, occupational, and Vietnam-veteran studies that evalu- ate herbicide exposure and the risk of birth defects, declines in sperm quality and fertility, spontaneous abortion, stillbirth, neonatal and infant mortality, low birth weight and preterm birth, and childhood cancer. In addition to studies of herbicides and 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD), studies of popula- tions exposed to polychlorinated biphenyls (PCBs) were reviewed when relevant because TCDD is sometimes a contaminant of PCBs. For new studies that report only a single reproductive health outcome and that are not revisiting a previously studied population, design information is summarized here with the results; de- sign information on all other new studies can be found in Chapter 4. This chapter’s primary emphasis is on the potential adverse reproductive effects of herbicide exposure in men because the vast majority of Vietnam veter- ans are men. About 8,000 women served in Vietnam (H. Kang, US Department of Veterans Affairs, personal communication, December 14, 2000), so findings relevant to female reproductive health are also included. Studies that investigated the potential reproductive consequences of exposure of either parent were consid- ered; whenever the information was available, an attempt was made to evaluate the effects of maternal and paternal exposure separately. 517

518 VETERANS AND AGENT ORANGE: UPDATE 2006 FERTILITY Male reproductive function is under the control of several components whose proper coordination is important for normal fertility. Several of the components and some endpoints related to male fertility, including reproductive hormones and sperm characteristics, can be studied as indicators of fertility. The reproductive neuroendocrine axis involves the central nervous system, the anterior pituitary gland, and the testis. The hypothalamus integrates neural inputs from the cen- tral and peripheral nervous systems and regulates the gonadotropins luteinizing hormone (LH) and follicle-stimulating hormone (FSH). Both are secreted into the circulation in episodic bursts by the anterior pituitary gland and are neces- sary for normal spermatogenesis. In the testis, LH interacts with receptors on Leydig cells, where it stimulates increased testosterone synthesis. FSH and the testosterone from the Leydig cells interact with the Sertoli cells in the seminifer- ous tubule epithelium to regulate spermatogenesis. More detailed reviews of the male reproductive hormones can be found elsewhere (Knobil et al., 1994; Yen and Jaffe, 1991). Several agents, such as lead and dibromochloropropane, affect the neuroendocrine system and spermatogenesis (for reviews, see Bonde and Giwercman, 1995; Tas et al., 1996). Whereas many studies have investigated the relationship between chemi- cals and male fertility, studies among women are sparse. Some chemicals may disrupt the female hormonal balance necessary for proper functioning. Normal menstrual-cycle functioning is also important in the risk of hormonally related diseases, such as osteopenia, breast cancer, and cardiovascular disease. Chemicals can have multiple effects on the female system, including modulation of hormone concentrations, such menstrual- or ovarian-cycle irregularities as changes in men- arche and menopause, and impairment of fertility (Bretveld et al., 2006a,b). In this chapter, we discuss studies that have focused on menstrual-cycle characteris- tics and age of menarche or age of menopause. Studies of the association between the chemicals of interest and endometriosis are reviewed in Chapter 9. Conclusions from VAO and Updates The committee responsible for Veterans and Agent Orange, hereafter re- ferred to as VAO (IOM, 1994), concluded that there was inadequate or insuffi- cient evidence of an association between exposure to 2,4-dichlorophenoxyacetic acid (2,4-D), 2,4,5-trichlorophenoxyacetic acid (2,4,5-T), TCDD, picloram, or cacodylic acid and altered sperm characteristics or infertility. Additional in- formation available to the committees responsible for Update 1996, Update 1998, Update 2000, Update 2002, and Update 2004 did not change that finding. Reviews of the relevant studies are presented in the earlier reports. Table 7-1 summarizes the studies.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 519 TABLE 7-1 Selected Epidemiologic Studies—Fertility (altered hormone concentrations, decreased sperm counts or quality, subfertility, or infertility) Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a OCCUPATIONAL New Studies Farr et al., 2006 Age of menopause women who self-reported pesticide exposure 8,038 0.9 (0.8–1.0) Oh et al., 2005 Male fertility—dioxin exposure with air monitoring 31 1.4* Farr et al., 2004 Menstrual cycle characteristics of premenopausal women in AHS aged 21–40 1,754 Short menstrual cycle 0.8 (0.6–1.0) Long menstrual cycle 1.4 (0.9–2.1) Irregular 0.6 (0.4–0.8) Missed Period 1.6 (1.3–2.0) Intermenstrual bleeding 1.1 (0.9–1.4) Studies Reviewed in Update 2000 Abell et al., 2000 Female greenhouse workers in Denmark (maternal exposure) 20 hours manual contact per week 220 0.7 (0.5–1.0)b Never used gloves 156 0.7 (0.5–1.0)b High exposure 202 0.6 (0.5–0.9)b Larsen et al., 1998 Danish farmers who used any potentially spermatotoxic pesticides, including 2,4-D Farmers using pesticides vs. organic farmers 523 1.0 (0.8–1.4)b Used three or more pesticides 0.9 (0.7–1.2)b Used manual sprayer for pesticides 0.8 (0.6–1.1)b Studies Reviewed in Update 1998 Heacock et al., Workers at sawmills using chlorophenates 1998 Standardized fertility ratio 18,016 0.7 (0.7–0.8)c (births) Mantel-Haenszel rate ratio estimator 18,016 0.9 (0.8-0.9)c (births) Cumulative exposure (hours) 120–1,999 7,139 0.8 (0.8–0.9)c 2,000–3,999 4,582 0.9 (0.8–1.0)c 4,000–9,999 4,145 1.0 (0.9–1.1)c 10,000 1,300 1.1 (1.0–1.2)c continued

520 VETERANS AND AGENT ORANGE: UPDATE 2006 TABLE 7-1 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a Lerda and Rizzi, Argentinean farmers exposed to 2,4-D 32 1991 Sperm count (millions/ml) exposed: 49.0 vs. control: 101.6 Motility (%) exposed: 24.8 vs. control: 70.4 Sperm death (%) exposed: 82.9 vs. control: 37.1d Anomalies (%) exposed: 72.9 vs. control: 33.4 (p 0.01 overall) ENVIRONMENTAL New Studies Eskanazi et al., Seveso cohort-serum dioxin concentrations 2005 and age of menopause 616 Premenopause 260 43.6 (0.2–0.9) Natural Menopause 169 45.8 (0.3–1.0) Surgical menopause 83 43.4 (0.3–1.0) Impending menopause 13 43.8 (0.2–0.9) Perimenopause 33 36.5 (0.2–0.9) Other 58 39.6 (0.2–0.9) Warner et al., 2004 Age of menarche at time of exposure 282 1.0 (0.8–1.1) Greenlee et al., Women from Wisconsin, US infertility 2003 (maternal exposure) Mixed or applied herbicides 21 2.3 (0.9–6.1) Used 2,4,5-T 9 9 cases (2.7%) 11controls (3.4%) Used 2,4-D 4 4 cases (1.2%) 4 controls (1.2%) Swan et al., 2003 Men from Missouri, US low sperm quality Elevated urinary metabolite marker for 2,4-D 5 0.8 (0.2–3.0) Studies Reviewed in Update 2002 Staessen et al., Adolescents in communities close to industrial 2001 sources of heavy metals, PCBs, VOCs, and PAHs—delays in sexual maturity In Hoboken, Belgium 8 4.0 (*) In Wilrik, Belgium 15 1.7 (*)

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 521 TABLE 7-1 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a VIETNAM VETERANS Studies Reviewed in Update 1996 Henriksen et al., Effects on specific hormone levels or sperm 1996 count in Ranch Hands Low testosterone High dioxin (1992) 18 1.6 (0.9–2.7) High dioxin (1987) 3 0.7 (0.2–2.3) Low dioxin (1992) 10 0.9 (0.5–1.8) Low dioxin (1987) 10 2.3 (1.1–4.9) Background (1992) 9 0.5 (0.3–1.1) High FSH High dioxin (1992) 8 1.0 (0.5–2.1) Low dioxin (1992) 12 1.6 (0.8–3.0) Background (1992) 16 1.3 (0.7–2.4) High LH High dioxin (1992) 5 0.8 (0.3–1.9) Low dioxin (1992) 5 0.8 (0.5–3.3) Background (1992) 8 0.8 (0.4–1.8) Low sperm count High dioxin 49 0.9 (0.7–1.2) Low dioxin 43 0.8 (0.6–1.0) Background 66 0.9 (0.7–1.2) Studies Reviewed in VAO CDC, 1989b Vietnam Experience Study Lower sperm concentration 42 2.3 (1.2–4.3) Proportion of abnormal sperm 51 1.6 (0.9–2.8) Reduced sperm motility 83 1.2 (0.8–1.8) Stellman et al., American Legionnaires who served in 1988 Southeast Asia Difficulty having children 349 1.3 (p 0.01) Unless otherwise indicated, studies show paternal exposure. a Given when available. b For this study, relative risk has been replaced with the fecundability ratio, for which a value less than 1.0 indicates an adverse effect. c For this study, relative risk has been replaced with the standardized fertility ratio, for which a value less than 1.0 indicates an adverse effect. d Table 1 in the reference reverses these figures—control: 82.9%; exposed: 37.1%—but the text (“The percentages of asthenospermia, mobility, necrosperma and teratospermia were greater in the exposed group than in controls…”) suggests that this is a typographic error. * Information not provided by study authors.

522 VETERANS AND AGENT ORANGE: UPDATE 2006 Update of the Epidemiologic Literature Occupational Studies After exclusion of women who were pregnant, were nursing, were taking oral contraceptives, had extreme body-mass indexes, or had missing values, Farr et al. (2004) reported on the menstrual-cycle characteristics of 3,103 premeno- pausal women in the Agricultural Health Study (AHS) who were 21–40 years old when they completed a female health and family health questionnaire. They examined the association between pesticide mixing or applying and menstrual characteristics of short cycles, long cycles, irregular cycles, missed periods, and bleeding or spotting between periods in the preceding 12 months. Women who had never mixed or applied pesticides were considered the control group. The investigators reported a significant relationship between increased cycle length and ever mixing or applying any type of pesticide (p 0.02) and increased reports of missed periods (OR 1.6). There was a trend toward increased odds of long cycles (p 0.08) and missed periods (p 0.001) with increasing days of pesticide exposure. Although using hormonally active pesticides was found to be associ- ated with increased cycle length and increased frequency of missed cycles, the pesticides with this observed association did not include any of the chemicals of interest to the present review committee. The study used self-reported informa- tion on menstrual cycle that may have been unreliable, and no hormonal confir- mation of menstrual dysfunction was available. Overall, there was no indication of an association with menstrual-cycle characteristics and the specific chemicals of interest in this review. There also has been a report from the AHS (Farr et al., 2006) concerning age at menopause in 8,038 women who were 35–55 years old at the time of enroll- ment. Women were classified according to their self-reported pesticide exposure. Overall, women who ever mixed or applied pesticides were found to have a higher age at menopause (hazard ratio [HR] by Cox proportional hazard analysis 0.87, 95% CI 0.78–0.97) that translates into a delay of about 3 months. The estimate did not vary much when restricted to herbicides (HR 0.88, 95% CI 0.74–1.05) or to phenoxy herbicides (HR 0.85, 95% CI 0.65–1.11). One study of male fertility outcomes has been published since the last update. Oh et al. (2005) studied a group of 31 male incinerator workers and 84 controls in Seoul, South Korea. They measured dioxin exposure with air monitor- ing in the facility and found that levels were 100 times higher than those reported for the general Seoul area (31.17 ng TEQ/m3 compared with 0.32 ng TEQ/m3). Sperm characteristics were analyzed for eight controls and six workers. No sta- tistically significant differences were observed in the number of sperm (p = 0.05) or sperm mobility (p 0.35). The fractions of sperm with DNA damage in waste- incineration workers and control subjects were measured at 1.40% 0.08% and 1.26% 0.03%, respectively (p 0.001).

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 523 Environmental Studies The committee reviewed two reports from the Seveso Women’s Health Study (SWHS) published since the last update that focused on age at menarche and age at menopause in the Seveso population, which was exposed to high concentrations of TCDD as the result of an industrial explosion in 1976. Warner et al. (2004) examined age at menarche in 282 women who were premenarcheal at the time of the explosion. TCDD was measured in archived blood samples. Subjects had a mean age of 6.9 years at the time of the explosion. The median serum TCDD concentration was 140.3 ppt for all premenarcheal women. Serum TCDD did not vary with self-reported age at menarche in all subjects or in a group that were less than 8 years old at the time of the explosion. A major limi- tation of the study was that age at menarche was based on recall, and the time between onset of menarche and study interview ranged from 5 to 19 years. The finding of no association between age at menarche and exposure of young girls to TCDD may be related to the possibility that susceptibility is greater in utero than during childhood. The committee reviewed a second SWHS paper by Eskenazi et al. (2005) on serum dioxin concentrations and age at menopause in the Seveso cohort. The study included 616 women who were premenopausal at the time of the explo- sion and were older than 35 years at the time of the interview. The median lipid- adjusted serum TCCD concentration was 43.7 ppt and did not vary significantly among the menopausal categories of premenstrual, natural menopause, surgical menopause, impending menopause, and perimenopause. The HRs of the serum TCDD quintiles (1.0, 1.1, 1.4, 1.6, and 1.1) suggested a trend between TCDD exposure (up to about 100 ppt) and earlier onset of natural menopause but also suggested that women with the highest serum TCDD did not have the earliest onset of menopause. Age at which the subjects of this study were exposed rep- resents an appropriate match for the experience of female Vietnam veterans. The literature suggests, however, that ovarian follicles are most susceptible to effects in the prepubertal period. A publication by Swan (2006) only reiterated the findings in Swan et al. (2003), which were considered in Update 2004. Vietnam-Veteran Studies No new Vietnam-veteran studies concerning exposure to the compounds of interest and fertility were published since Update 2004. Biologic Plausibility There is little evidence that 2,4-D or 2,4,5-T has substantial effects on re- productive organs or fertility. One recent study has demonstrated that 2,4,5-T

524 VETERANS AND AGENT ORANGE: UPDATE 2006 competes with 17 -estradiol for binding to estrogen receptor and can function as an antiestrogen in cell culture (Lemaire et al., 2006), suggesting 2,4,5-T may have the potential to disrupt female reproductive function. In contrast with the lack of evidence on 2,4-D and 2,4,5-T, many diverse laboratory studies provide evidence that TCDD can affect reproductive organ function and reduce fertility in both men and women. TCDD exposure can re- duce fertility in male rats and is associated with histologic changes in the testis (Chahoud et al., 1989). More recent studies of TCDD’s effects on the testis have shown that it can induce significant changes in gene expression (Kuroda et al., 2005; Lai et al., 2005a; Volz et al., 2005; Yamano et al., 2005), leading to modification of steroidogenesis in particular (Lai et al., 2005b). Those changes are associated with disruption or complete inhibition of spermatogenesis (Fisher et al., 2005; Simanainen et al., 2004; Volz et al., 2005). Furthermore, the TCDD- induced reduction in spermatogenesis has been associated with reduced erectile function in one study (Moon et al., 2004) and reduced serum testosterone in another (Simanainen et al., 2004). In women, TCDD has been shown to reduce reproductive success, and this reduction could be mediated by alterations in the ovaries, uterus, and placenta. TCDD has been shown to disrupt ovarian steroidogenesis, impair ovulation, re- duce circulating progesterone and estradiol, and decrease fertility (Li et al., 2006; Petroff and Mizinga, 2003; Ushinohama et al., 2001). Recent studies demonstrate that TCDD at low concentrations suppresses gene expression essential to ovarian function and downregulates estrogen-dependent signaling (Hombach-Klonisch et al., 2006; Miyamoto, 2004). TCDD-induced reduction in fertility in women could also be mediated by changes in the uterus. TCDD has antiestrogenic activ- ity on the uterus, causing impairment of uterine epithelial function (Buchanan et al., 2000) that may contribute to TCDD-induced reduction in the survival of implanted embryos early in gestation (Kitajima et al., 2004). TCDD-induced reduction in reproductive success may also be mediated by altered placental function, which can lead to fetal death. TCDD alters gene expression in the pla- centa, suppresses placental vascular remodeling, and induces placental hypoxia (Ishimura et al., 2002, 2006; Mizutani et al., 2004). The biologic plausibility of reproductive effects in general arising from ex- posure to the chemicals of interest is discussed at the end of this chapter. Synthesis Although there is much evidence of the biologic plausibility of disruption of male and female fertility by exposure to the chemicals of interest, there continues to be a lack of substantive epidemiologic data that demonstrate any association in human populations.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 525 Conclusions On the basis of its evaluation of the evidence reviewed here and in previous VAO reports, the committee concludes that there is inadequate or insufficient evidence of an association between exposure to the compounds of interest and altered hormone concentrations, menstrual-cycle abnormalities, decreased sperm counts or sperm quality, subfertility, or infertility. SPONTANEOUS ABORTION Spontaneous abortion is the expulsion of a nonviable fetus, generally before 20 weeks of gestation, that is not induced by physical or pharmacologic means. The background risk of recognized spontaneous abortion is generally 7–15 per- cent (Hertz-Picciotto and Samuels, 1988), but it is established that many more pregnancies terminate before women become aware of them (Wilcox et al., 1988)—these terminations are known as subclinical pregnancy losses and gener- ally are not included in studies of spontaneous abortion. Estimates of the risk of recognized spontaneous abortion vary with the design and method of analysis. Study designs include cohorts of women asked retrospectively about pregnancy history, cohorts of pregnant women (usually those receiving prenatal care), and cohorts of women who are monitored for future pregnancies. Retrospective re- ports can be limited by memory loss, particularly of spontaneous abortions that took place a long time before. Studies that enroll women who appear for prenatal care require the use of life tables and specialized statistical techniques to account for differences in the times at which women seek medical care during pregnancy. Enrollment of women before pregnancy provides the theoretically most valid estimate of risk, but it can attract non-representative study groups because pro- tocols are demanding. Conclusions from VAO and Updates The committee responsible for VAO concluded that there was inadequate or insufficient evidence of an association between exposure to 2,4-D, 2,4,5-T, TCDD, picloram, or cacodylic acid and spontaneous abortion. Additional infor- mation available to the committees responsible for Update 1996, Update 1998, and Update 2000 did not change that conclusion. Information available to the committee responsible for Update 2002, however, led to the conclusion that there was suggestive evidence that paternal exposure to TCDD is not associated with the risk of spontaneous abortion, but that there was insufficient information to determine whether an association exists between maternal exposure to TCDD and the risk of spontaneous abortion or between maternal or paternal exposure to 2,4-D, 2,4,5-T, picloram, or cacodylic acid and the risk of spontaneous abortion. The additional information reviewed by the committee responsible for Update

526 VETERANS AND AGENT ORANGE: UPDATE 2006 2004 did not change this conclusion. The relevant studies are reviewed in the earlier reports. Table 7-2 summarizes them. Update of the Epidemiologic Literature Environmental Studies Tango et al. (2004) studied the distribution of several birth outcomes around Japanese municipal-waste incinerators with elevated dioxin emissions. They found fetal death after the 12th week of gestation (with or without congenital malformations) was not associated with the distance the mother lived from an incinerator at the time of birth or whether her residence was in the area known to have the highest dioxin soil concentrations. No new occupational or Vietnam-veteran studies concerning exposure to the compounds of interest and spontaneous abortion were published since Update 2004. Biologic Plausibility Laboratory animal studies demonstrate that TCDD exposure during preg- nancy can alter circulating steroid hormone concentrations (Simanainen et al., 2004) and disrupt placental development and function (Ishimura et al., 2006; Mizutani et al., 2004) and thus contribute to a reduction in survival of implanted embryos and to fetal death (Kitajima et al., 2004). However, the reproductive significance of those effects and the risk of recognized pregnancy loss before 20 weeks of gestation in humans are not clear. There is no evidence of a relationship between paternal or maternal exposure to TCDD and spontaneous abortion. Ex- posure to 2,4-D or 2,4,5-T causes fetal toxicity and death after maternal exposure in experimental animals. However, that effect occurs only at high doses and in the presence of maternal toxicity. No fetal toxicity or death has been reported to occur after paternal exposure to 2,4-D. The biologic plausibility of reproductive effects in general arising from ex- posure to the chemicals of interest is discussed at the end of this chapter. Synthesis No additional information was available to the committee responsible for Update 2006 to motivate changing the assessment of the last two committees. Given the age of the Vietnam-veteran cohort, it is highly unlikely that additional information on this outcome among the population will appear.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 527 TABLE 7-2 Selected Epidemiologic Studies—Spontaneous Abortion Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a OCCUPATIONAL Studies Reviewed in Update 2002 Schnorr et al., Wives and partners of men in the NIOSH 2001 cohort Estimated paternal TCDD serum level at the time of conception 20 ppt 29 0.8 (0.5–1.2) 20 to 255 ppt 11 0.8 (0.4–1.6) 255 to 1,120 11 0.7 (0.3–1.6) 1,120 ppt 8 1.0 (0.4–2.2) Studies Reviewed in Update 2000 Driscoll et al., Women employed by the US Forest 1998 Service—miscarriages (maternal exposure) 141 2.0 (1.1–3.5) Studies Reviewed in VAO Moses et al., 1984 Follow-up of 2,4,5-T production workers 14 0.9 (0.4–1.8) Suskind and Hertzberg, 1984 Follow-up of 2,4,5-T production workers 69 0.9 (0.6–1.2) Smith et al., Follow-up of 2,4,5-T sprayers vs 1982 non-sprayers 43 0.9 (0.6–1.3)** Townsend et al., Wives of men employed involved in 1982 chlorophenol processing at Dow Chemical Company 85 1.0 (0.8–1.4) Carmelli et al., Wives of men occupationally exposed to 1981 2,4-D All reported work exposure to herbicides (high and medium) 63 0.8 (0.6–1.1)** Farm exposure 32 0.7 (0.4–1.5) Forest and commercial exposure 31 0.9 (0.6–1.4) Exposure during conception period Farm exposure 15 1.0 (0.5–1.8) Forest and commercial exposure 16 1.6 (0.9–1.8) All exposures, father aged 18–25 years Forest and commercial exposure 8 3.1 (1.2–7.8) Exposure during conception period Father aged 31–35 years, farm exposure 10 2.9 (0.8–10.9) ENVIRONMENTAL New Studies Eskenazi et al., Seveso (Italy) Women’s Health Study 2003 participants living in exposure Zones A and B in 1976 (maternal exposure) Pregnancies 1976–1998 97 0.8 (0.6–1.2) Pregnancies 1976–1984 44 1.0 (0.6–1.6) continued

528 VETERANS AND AGENT ORANGE: UPDATE 2006 TABLE 7-2 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a Studies Reviewed in Update 2002 Arbuckle et al., Ontario farm families (maternal and paternal 2001 exposure) Phenoxyacetic acid herbicide exposure in the preconception period & spontaneous abortion risk 48 1.5 (1.1–2.1) Revich et al., Residents of the Samara Region, Russia 2001 (maternal and paternal exposure) Chapaevsk 24.4% (20.0–29.5%)b Samara * 15.2% (14.3–16.1%)b Toliatti * 10.6% (9.8–11.5%)b Syzran * 15.6% (13.4–18.1%)b Novokuibyshevsk * 16.9% (14.0–20.3%)b Other small towns * 11.3% (9.4–13.8%)b Tuyet and Vietnamese women who were or whose (*) [anecdotal reports Johansson, husbands were exposed to herbicides sprayed of miscarriage in 2001 during the Vietnam war * pilot study] Studies Reviewed in Update 2000 Axmon et al., Wives of Swedish fishermen 2000 Before gestation week 12 0.5 (0.3–1.0) East coast residents 12 West coast residents 54 Petrelli et al., Wives of pesticide appliers 26 3.8 (1.2–12.0) 2000 VIETNAM VETERANS Studies Reviewed in Update 2002 Kang et al., Female Vietnam-era veterans (maternal 2000 exposure) 1.0 (0.82–1.21) Vietnam veterans (1,665 pregnancies) 278 (*) Vietnam-era veterans who did not serve in Vietnam (1,912 pregnancies) 317 (*) Studies Reviewed in Update 2000 Schwartz, 1998 Female Vietnam veterans (maternal exposure) Women who served in Vietnam 113 (*) Women who did not serve in the war zone 124 (*) Civilian women 86 (*) Studies Reviewed in Update 1996 Wolfe et al., Air Force Ranch Hand veterans 157 1995 Background 57 1.1 (0.8–1.5) Low-level exposure 56 1.3 (1.0–1.7) High-level exposure 44 1.0 (0.7–1.3) Studies Reviewed in VAO Aschengrau and Wives of Vietnam veterans presenting at Monson, 1989 Boston Hospital for Women 27 weeks gestation 10 0.9 (0.4–1.9) 13 weeks gestation * 1.2 (0.6–2.8)

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 529 TABLE 7-2 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a CDC, 1989b Vietnam Experience Study Overall 1,566 1.3 (1.2–1.4) Self-reported low exposure 489 1.2 (1.0–1.4) Self-reported medium exposure 406 1.4 (1.2–1.6) Self-reported high exposure 113 1.7 (1.3–2.1) Field and Kerr, Follow-up of Australian Vietnam veterans 199 1.6 (1.3–2.0) 1988 Stellman et al., American Legionnaires with service 1988 1961–1975 Vietnam-era veterans vs Vietnam veterans All Vietnam veterans 231 1.4 (1.1–1.6) Low exposure 72 1.3 (1.0–1.7) Medium exposure 53 1.5 (1.1–2.1) High exposure 58 1.7 (1.2–2.4) Vietnam-era veterans vs herbicide handlers 9 1.6 (0.7–3.3) Vietnam veterans with medium or high exposure vs Vietnam veterans with low exposure Medium exposure 53 1.2 (0.8–1.7) High exposure 58 1.4 (0.9–1.9) ABBREVIATIONS: CDC, Centers for Disease Control and Prevention; CI, confidence interval; NIOSH, National Institute for Occupational Safety and Health. Unless otherwise indicated, studies show paternal exposure. a Given when available. b Spontaneous abortion rate per 100 full-term pregnancies for the years 1991–1997. * Information not provided by study authors. ** 90% confidence interval. Conclusions The present committee concurs with the overall conclusion of the previ- ous committees that paternal exposure to TCDD is not associated with risk of spontaneous abortion and that insufficient information is available to determine whether an association exists between the risk of spontaneous abortion and mater- nal exposure to TCDD or either maternal or paternal exposure to 2,4-D, 2,4,5-T, picloram, or cacodylic acid. STILLBIRTH, NEONATAL DEATH, AND INFANT DEATH Stillbirth or late fetal death typically refers to the delivery at or after 20 weeks of gestation of a fetus that shows no signs of life, including fetuses that

530 VETERANS AND AGENT ORANGE: UPDATE 2006 weigh more than 500 g regardless of gestational age (Kline et al., 1989). Neonatal death refers to the death of a liveborn infant within 28 days of birth, while infant death includes deaths occurring before the first birthday. Because the causes of stillbirth and early neonatal death overlap considerably, they are commonly analyzed together in a category referred to as perinatal mor- tality (Kallen, 1988). Stillbirths make up less than 1 percent of all births (CDC, 2000). The most common causes of perinatal mortality (Kallen, 1988) among low-birth-weight (500 g to 2,500 g) liveborn and stillborn infants are placental and delivery complications—abruptio placenta, placenta previa, malpresentation, and umbilical-cord complications. Among infants weighing more than 2,500 g at birth, the most common causes of perinatal death are complications of the cord, placenta, and membranes and congenital malformations (Kallen, 1988). Conclusions from VAO and Updates The committee responsible for VAO concluded that there was inadequate or insufficient evidence of an association between exposure to 2,4-D, 2,4,5-T, TCDD, picloram, or cacodylic acid and stillbirth, neonatal death, or infant death. Additional information available to the committees responsible for Update 1996, Update 1998, Update 2000, Update 2002, and Update 2004 did not change that conclusion. Reviews of the relevant studies are presented in the earlier reports. Update of the Epidemiologic Literature The only relevant study published since the last update was a large study of multiple pregnancy outcomes in Japan (Tango et al., 2004). The study found no associations between proximity of the mother’s residence at the time of birth (defined in terms of 1-km bands around the incinerators) and rates infant deaths ( 1 yr, 1 mo, 1 wk with and without congenital malformations). Analyses were conducted for a “peak-decline” relationship with “peak” dioxin soil concentra- tions (known to occur about 2 km from an incinerator); only death within the first year of life, overall (p 0.023) or with congenital malformations (p 0.047), showed a significant result. This study had several methodologic weaknesses, including the lack of individual-level information on other risk factors and of in- dividual exposure data. In addition, the birth-outcome effects might be associated with socioeconomic differences in residents in the different zones. No new occupational or Vietnam-veteran studies concerning exposure to the compounds of interest and stillbirth, neonatal death, or infant death were published since Update 2004. Biologic Plausibility Laboratory studies of maternal TCDD exposure during pregnancy have demonstrated the induction of fetal death; neonatal death, however, is only rarely

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 531 observed and is usually the result of cleft palate leading to an inability to nurse. Studies addressing the potential for perinatal death as a result of paternal expo- sure to TCDD or herbicides are too limited to support conclusions. The biologic plausibility of reproductive effects in general arising from ex- posure to the chemicals of interest is discussed at the end of this chapter. Synthesis The study reviewed for this update did not find significant associations be- tween the relevant exposures and rates of infant or fetal deaths. The study was limited in that exposure was based on environmental concentrations of dioxin and individual exposure data were not obtained. Furthermore, several risk factors that could confound associations between exposure and outcome were not assessed. Conclusions On the basis of its evaluation of the evidence reviewed here and in previous VAO reports, the committee concludes that there is inadequate or insufficient evidence of an association between exposure to the compounds of interest and stillbirth, neonatal death, or infant death. BIRTH WEIGHT AND PRETERM DELIVERY The World Health Organization recommends 2,500 g as the threshold for designation of low birth weight (Alberman, 1984). Low infant weight at birth is among the important predictors of neonatal death and morbidity in the United States. The concept of low birth weight actually encompasses two causal path- ways, often treated as a single entity: low birth weight secondary to intrauterine growth retardation (IUGR), in which case a fetus or baby is referred to as small for gestational age, and low birth weight secondary to preterm delivery (PTD), which can have other long-term consequences. The concept of IUGR represents birth weight adjusted for gestational age. The current definition of PTD is de- livery at less than 259 days, or 37 completed weeks, of gestation, calculated on the basis of the date of the first day of the last menstrual period (Bryce, 1991). About 7 percent of live births are low birth weight. The incidence of IUGR is much more difficult to quantify because there are no standards for distribut- ing birth weight by gestational age. When no distinction is made between the causes of low birth weight (IUGR or PTD), the factors most strongly associated with it are maternal tobacco use during pregnancy, multiple births, and race or ethnicity. Other potential risk factors are socioeconomic status (SES), maternal weight, birth order, maternal complications during pregnancy (such as severe pre- eclampsia) and obstetric history, job stress, and cocaine or caffeine use during pregnancy (Kallen, 1988). Established risk factors for PTD include race (black), marital status (single), low SES, previous low birth weight or PTD, multiple ges-

532 VETERANS AND AGENT ORANGE: UPDATE 2006 tations, tobacco use, and cervical, uterine, or placental abnormalities (Berkowitz and Papiernik, 1993). Conclusions from VAO and Updates The committee responsible for VAO concluded that there was inadequate or insufficient evidence of an association between exposure to the compounds of interest and low birth weight or preterm delivery. Additional information avail- able to the committees responsible for Update 1996, Update 1998, Update 2000, Update 2002, and Update 2004 did not change that conclusion. Reviews of the relevant studies are presented in the earlier reports. Update of the Epidemiologic Literature Update 2000, Update 2002, and Update 2004 discussed change in the sex ratio at birth from the usual ratio of male-to-female newborns, which is approxi- mately 1.02 as a possible adverse reproductive outcome. If the herbicides used in Vietnam did alter this endpoint in humans, it would not represent an outcome for which an individual veteran could be compensated. Such an alteration would, however, be indicative of some impact on reproductive function, as mentioned in the section on biologic plausibility at the end of this chapter. Of the new articles reporting reproductive effects, two environmental studies that presented findings on birth weight and gestational age also assessed possible changes in the sex ratio at birth; their findings for this additional endpoint are included below. Occupational Studies Lawson et al. (2004) conducted a follow-on study of PTD and birth weight among the children of men in the National Institute for Occupational Safety and Health (NIOSH) cohort in comparison to age- and race-matched neighborhood referents. The analysis of birth weight considered 1,117 singleton, full-term births (at least 37 weeks gestation) to 217 referent wives (604 referent births) and 176 worker wives (513 births; 221 pre-exposure and 292 exposed). The suggestion of an increase in birth weight with high TCDD concentrations vanished when adjustment was made for mother’s education, parity, smoking during pregnancy, length of gestation, and the infant’s sex. In order to consider the possibility that the mothers might have experienced direct exposure from materials carried home by the fathers when they were engaged in their NIOSH employment during a pregnancy, another analysis was restricted to such pregnancies for the cohort members and pregnancies occurring while the NIOSH plants were in operation for the referents, or 334 referent births and 98 exposed births. In this case, the adjusted analysis found a statistically significant increase of 31 g in birth weight with each log increase in TCDD concentration (p 0.03).

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 533 Lawson et al. (2004) also conducted an analysis of preterm births based on a total of 1,153 births: 618 referent, 238 pre-exposed, and 297 during-exposure. With or without adjustment for age of mother and for the occurrence of an ac- cident, smoking, or medication use during pregnancy, no increase in risk of pre- maturity was observed (odds ratio [OR] 0.8, 95% CI 0.6–1.1). Environmental Studies Another study of the effect of dioxins on reproductive outcomes was con- ducted in an area surrounding a municipal-waste incinerator located 10 km from Taipei that began operating in 1992 (Lin et al., 2006). The incinerator’s average emission concentration of polychlorinated dibenzo-p-dioxins and dibenzofurans (PCDD/Fs) for 1997 (6.47 ng TEQ/m3) was used as the input for a dispersion model (EPA’s Industrial Source Complex Model—Short Term Model), which was used to estimate annual averages for each administrative district of Taipei. A cut point of 0.03 pg TEQ/m3, between the background concentrations for rural (0.02 pg TEQ/m3) and urban air (0.05 pg TEQ/m3), was selected. The dispersion model predicted concentrations greater than 0.03 pg TEQ/m3 for 40 districts; those were retained as the “exposed” districts, which were further subdivided at 0.05 pg TEQ/m3. Among the districts with predicted concentrations of zero, 40 “reference” districts were randomly selected; those were all in non-industrial areas at least 12 km from the incinerator. From Taiwan’s birth registry, Lin et al. (2006) assembled newborn gender, birth order, gestational age, and birth weight, plus maternal age, address, and educational level, for all singleton live births in the study districts for 1991 and 1997. After excluding those with birth weights less than 500 g, with gestational ages less than 20 weeks, or born to unwed women, 6,697 and 6,282 infants born in 1991 and 1997, respectively, were available for analysis. Analyses were adjusted for maternal age and education, sex of newborn, and birth order; information on other potential confounders (such as maternal smoking) was not available. Categorical comparisons of the predicted high and low exposed districts with the reference districts found no associations for low birth weight (under 2,500 g), PTD (less than 37 weeks of gestation), or sex ratio for 1991 (as might have been expected before exposure began) or for 1997 (after the incinerator had been in operation for 5 years). Regression on the continuous variables determined the difference between the exposed and reference districts in 1991 and in 1997 for mean birth weight or mean gestational age. In 1997, the adjusted decrease in birth weight of 5.87 g for the exposed districts was non- significant, but the relative reduction in gestational age of 0.09 week was sig- nificant (p 0.05). (For 1991, both these endpoints had non-significantly higher values in the “not yet” exposed districts than in the reference districts; therefore, a comprehensive before-and-after analysis might have revealed stronger effects in the districts that ultimately were exposed.) In light of no suggestion of an

534 VETERANS AND AGENT ORANGE: UPDATE 2006 increased risk of PTD, the small reported reduction in gestational age is of ques- tionable biological significance. The study’s large size gave it considerable sensitivity. On the whole it used appropriate methods, but apparently an analysis was not conducted to make full use of the 1991 data as self-control information to determined the relative changes between the exposed and reference districts from 1991 to 1997. The exposure assessment is only as reliable as the estimates generated by EPA’s dis- persion model for the districts of Taipei, so exposure misclassification could have contributed to biasing the generally negative results toward the null. In their study of birth outcomes for mothers living within about 10 km of Japanese municipal-waste incinerators emitting high concentrations of dioxin, Tango et al. (2004) found no relationship between proximity of the mother’s residence to the incinerator for low or extremely low (under 1,500 g) birth weight. Analyses for association with peak dioxin concentrations in soil (known to occur about 2 km from the incinerators) were also negative. The sex ratio at birth, evaluated in terms of deviation in the proportion of female live births from the national proportion, was also found not to be related to the distance of the maternal residence from an incinerator. Vietnam-Veteran Studies No new Vietnam veteran studies concerning exposure to the compounds of interest and birth weight or preterm delivery were published since Update 2004. Biologic Plausibility Laboratory studies of the potential male-mediated developmental toxicity of TCDD and herbicides as a result of exposure of adult male animals are too lim- ited to permit conclusions. TCDD and herbicides are known to cross the placenta leading to direct exposure of the fetus. Data from studies in experimental animals also suggest that the pre-implantation embryo and developing fetus are sensitive to the toxic effects of 2,4-D and TCDD after maternal exposure. However, the significance of those animal effects for humans is not clear. The biologic plausibility of reproductive effects in general arising from ex- posure to the chemicals of interest is discussed at the end of this chapter. Synthesis The three studies reviewed here (two environmental and one based on an occupational cohort) did not find an association between exposure to the com- pounds of interest and the risk of low birth weight or prematurity. The two new weakly significant findings may simply be spurious results arising among many

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 535 comparisons; a modest increase in average birth weight would not be construed as an adverse effect, and the small decrease in average gestation is of questionable biologic importance. Although the results overall suggest a lack of an association, they should be interpreted with caution because of some methodologic limita- tions, such as a long recall period in the cohort study and exposure misclassifica- tion in the environmental studies. Conclusions On the basis of its evaluation of the evidence reviewed here and in previous VAO reports, the committee concludes that there is inadequate or insufficient evidence of an association between exposure to the compounds of interest and low birth weight or preterm delivery. BIRTH DEFECTS The March of Dimes defines a birth defect as “an abnormality of structure, function or metabolism, whether genetically determined or as the result of an en- vironmental influence during embryonic or fetal life” (Bloom, 1981). Other terms, often used interchangeably, are congenital anomaly and congenital malformation. Major birth defects, which occur in 2–3 percent of live births, are abnormalities that are present at birth and are severe enough to interfere with viability or physi- cal well-being. Birth defects are detected in another 5 percent of babies through the first year of life. The causes of most birth defects are unknown. Genetic fac- tors, exposure to some medications, exposure to environmental contaminants, oc- cupational exposures, and lifestyle factors have been implicated in the etiology of birth defects (Kalter and Warkany, 1983). Most etiologic research has focused on the effects of maternal and fetal exposures, but some work has addressed paternal exposures. Paternally mediated exposures might occur by several routes and exert effects in various ways. One way is through direct genetic damage to the male germ cell transmitted to the offspring and dominantly expressed as a birth defect. A hypothesized route is the transfer of toxic compounds through a man’s body into his seminal fluid, resulting in intermittent fetal exposures throughout gesta- tion (Chia and Shi, 2002). Another, even more indirect route of paternally medi- ated exposure could be contact of family members with contamination brought into the home from the workplace, but this would not be applicable to offspring of Vietnam veterans conceived during the post-deployment period. Conclusions from VAO and Updates The committee responsible for VAO determined that there was inadequate or insufficient evidence of an association between exposure to 2,4-D, 2,4,5-T or its contaminant TCDD, picloram, or cacodylic acid and birth defects among

536 VETERANS AND AGENT ORANGE: UPDATE 2006 offspring. Additional information available to the committee responsible for Update 1996 led it to conclude that there was limited or suggestive evidence of an association between at least one of the compounds of interest and spina bifida in the children of veterans; there was no change in the conclusions regarding other birth defects. The committee for Update 2002, which reviewed the study of female Vietnam veterans (Kang et al., 2000) reporting significant increases in the occurrence of birth defects among their offspring, did not find those results adequate to modify prior conclusions. Later VAO committees have not encoun- tered additional data to merit changing the conclusion that the evidence is inad- equate to support an association between exposure to the chemicals of interest and birth defects (aside from spina bifida) among the offspring of either male or female veterans. Summaries of the results from studies of birth defects and specifically neural- tube defects that were reviewed here and in earlier reports can be found in the Tables 7-3 and 7-4, respectively. Update of the Epidemiologic Literature Occupational Studies Lawson et al. (2004) reported on the pregnancy outcomes among wives of men from the NIOSH cohort of workers who were highly exposed to chemicals contaminated with TCDD compared to those of matched neighborhood referents. The analysis of birth defects was performed on 1,153 liveborn and 13 stillborn infants. Because only a modest number of birth defects were reported, pre- exposed and referent pregnancies were combined when they were qualitatively contrasted to the approximately 300 exposed pregnancies. Confirmation from vital and medical records was attempted, but was successful for only about 50 percent of the reported birth defects. The reported categories included 6 major central nervous system defects (4 among the exposed pregnancies), plus clubfoot, hip and lower limb defects, genitourinary defects, cardiovascular defects, and cleft lip or palate. Although the numbers were too small for reliable conclusions and only descriptive data were presented, there did not appear to be any trend of increased incidence of birth defects among offspring of exposed workers. Environmental Studies The only relevant environmental study published since Update 2004 was Tango et al. (2004). The study found no associations between proximity to the incinerators and rates of fetal loss or infant deaths (before 1 week, 1 month, or 1 year) due to congenital malformation. In analyses using a “peak-decline” ap- proach based on soil concentrations that had been shown to peak approximately 2 km from the incinerators, a significant outcome was found for infant death in

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 537 TABLE 7-3 Selected Epidemiologic Studies—Birth Defects in the Offspring of Subjects Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a OCCUPATIONAL New Studies Lawson et al., Wives of workers with serum TCDD levels in 2004 NIOSH cohort 14 * (*) Studies Reviewed in Update 1998 Kristensen et al., Norwegian farmers (maternal and paternal 1997 exposure) 4,189 1.0 (1.0–1.1)b Dimich-Ward Sawmill workers et al., 1996 Cataracts 11c 5.7 (1.4–22.6) Genital organs 105c 1.3 (0.9–1.5) Garry et al., 1996 Private pesticide appliers Circulatory–respiratory 17 1.7 (1.0–2.8) Gastrointestinal 6 1.7 (0.8–3.8) Urogenital 20 1.7 (1.1–2.6) Musculoskeletal–integumental 30 Maternal age 30 11 0.9 (0.5–1.7) Maternal age 30 19 2.5 (1.6–4.0) Chromosomal 8 1.1 (0.5–2.1) Other 48 Maternal age 35 36 1.1 (0.8–1.6) Maternal age 35 12 3.0 (1.6–5.3) All births with anomalies 125 1.4 (1.2–1.7) Studies Reviewed in VAO Moses et al., 1984 Follow-up of 2,4,5-T male production workers 11 1.3 (0.5–3.4) Suskind and Follow-up of 2,4,5-T male production workers 18 1.1 (0.5–2.2) Hertzberg, 1984 Smith et al., 1982 Follow-up of 2,4,5-T sprayers—sprayers vs non-sprayers 13 1.2 (0.6–2.5)e Townsend et al., Follow-up of Dow Chemical plant workers 30 0.9 (0.5–1.4) 1982 ENVIRONMENTAL New Studies Cordier et al., Residents of the Rhône-Alpes region of France 2004 living near municipal solid waste incinerators (maternal and paternal exposure) Minor anomalies 518 0.9 (0.8–1.1) Chromosomal anomalies 204 1.0 (0.9–1.2) Monogenic anomalies 83 1.1 (0.8–1.4) Unknown or multifactoral etiology 964 1.1 (1.0–1.2) Tango et al., 2004 Investigated multiple pregnancy outcomes in Japan—infant deaths from congenital defects 42 (*) NS continued

538 VETERANS AND AGENT ORANGE: UPDATE 2006 TABLE 7-3 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a Schreinemachers, Rural or farm residents of Minnesota, Montana, 2003 North and South Dakota (maternal and paternal exposure) Any birth anomaly 213 1.1 (0.9–1.3) Central nervous system anomalies 12 0.8 (0.5–1.4) Circulatory or respiratory anomalies 39 1.7 (1.1–2.6) Digestive system anomalies 24 0.9 (0.6–1.5) Urogenital anomalies 44 1.0 (0.7–1.5) Musculoskeletal or integumental anomalies 70 1.5 (1.1–2.1) Chromosomal anomalies 17 0.9 (0.6–1.6) Studies Reviewed in Update 2002 Loffredo et al., Mothers in the Baltimore–Washington Infant 2001 Study exposed to herbicides during the first trimester (maternal exposure) 8 2.8 (1.2–6.9) Revich et al., Residents of Chapaevsk, Russia—congenital 2001 malformations * (*) NS ten Tusscher Infants born in Zeeburg, Amsterdam clinics et al., 2000 1963–1965 with orofacial cleft (maternal exposure) Births in 1963 5 (*) SS Births in 1964 7 (*) SS Studies Reviewed in Update 2000 García et al., 1998 Residents of agricultural areas in Spain— median score on chlorophenoxy herbicides exposure duration (months) index 14 3.1(0.6–16.9) Studies Reviewed in VAO Fitzgerald et al., Persons exposed to an electrical transformer 1989 fire—total birth defects (maternal or paternal exposure) 1 2.1 (0.05–11.85) Mastroiacovo Seveso residents (maternal, paternal, and in et al., 1988 utero exposure) Zones A and B total defects 27 1.2 (0.9–1.6)e Zones A, B, R total defects 137 1.0 (0.8–1.1)e Zones A and B mild defects 14 1.4 (0.9–2.2)e Stockbauer et al., Persons in Missouri with documented TCDD 1988 soil contamination near residence (maternal; paternal; in utero exposure) Total birth defects 17 0.8 (0.4–1.5) Major defects 15 0.8 (0.4–1.7) Midline defects 4 0.7 (0.2–2.3)

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 539 TABLE 7-3 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a Hanify et al., Residents of areas of Northland New Zealand 1981 subject to aerial 2,4,5-T sprayingd All birth malformations 164 1.7 (1.4–2.1)e All heart malformations 20 3.9 (2.1–7.4)e Hypospadias, epispadias 18 5.6 (2.7–11.7)e Talipes 52 1.7 (1.2–2.3)e Cleft lip 6 0.6 (0.3–1.3)e Isolated cleft palate 7 1.4 (0.6–3.2)e VIETNAM VETERANS Studies Reviewed in Update 2002 Kang et al., 2000 Female Vietnam-era veterans—deployed vs non- deployed (maternal exposure) “Likely” birth defects * 1.7 (1.2–2.2) “Moderate-to-severe” birth defects * 1.5 (1.1–2.0) Studies Reviewed in Update 2000 AIHW, 1999 Australian Vietnam veterans—Validation Study Down syndrome 67 92 expected (73–111) Tracheo-oesophageal fistula 10 23 expected (14–32) Anencephaly 13 16 expected (8–24) Cleft lip or palate 94 64 expected (48–80) Absent external body part 22 34 expected (23–45) Extra body part 74 74 expected (*) Michalek et al., Air Force Ranch Hand veterans 1998a Before service in Southeast Asia * 0.7 (*) After service in Southeast Asia * 1.5 (*) Studies Reviewed in Update 1996 Wolfe et al., 1995 High exposure Ranch Hands relative to comparisons All anomalies 57 1.0 (0.8–1.3) Nervous system 3 (*) Eye 3 1.6 (0.4–6.0) Ear, face, and neck 5 1.7 (0.6–4.7) Circulatory system and heart 4 0.9 (0.3–2.7) Respiratory system 2 (*) Digestive system 5 0.8 (0.3–2.0) Genital system 6 1.2 (0.5–3.0) Urinary system 7 2.1 (0.8–5.4) Musculoskeletal 31 0.9 (0.6–1.2) Skin 3 0.5 (0.2–1.7) Chromosomal anomalies 1 (*) continued

540 VETERANS AND AGENT ORANGE: UPDATE 2006 TABLE 7-3 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a Studies Reviewed in VAO AFHS, 1992 Air Force Operation Ranch Hand veterans— birth defects in conceptions following service in Southeast Asia Congenital anomalies 229 1.3 (1.1–1.6) Nervous system 5 1.9 (0.5–7.2) Respiratory system 5 2.6 (0.6–10.7) Circulatory system or heart 19 1.4 (0.7–2.6) Urinary system 21 2.5 (1.3–5.0) Chromosomal 6 1.8 (0.6–6.1) Other 5 2.6 (0.6–10.7) Aschengrau and Vietnam veterans whose children were born at Monson, 1990 Boston Hospital for Women All congenital anomalies (crude OR) Vietnam veterans vs men without known military service 55 1.3 (0.9–1.9) Vietnam veterans vs non-Vietnam veterans 55 1.2 (0.8–1.9) One or more major malformations (crude OR) Vietnam veterans vs men without known military service 18 1.8 (1.0–3.1) Vietnam veterans vs non-Vietnam veterans 18 1.3 (0.7–2.4) CDC, 1989a Vietnam Experience Study—interview data Total anomalies 826 1.3 (1.2–1.4) Nervous system defects 33 2.3 (1.2–4.5) Ear, face, neck defects 37 1.6 (0.9–2.8) Integument 41 2.2 (1.2–4.0) Musculoskeletal defects 426 1.2 (1.1–1.5) Hydrocephalus 11 5.1 (1.1–23.1) Spina bifida 9 1.7 (0.6–5.0) Hypospadias 10 3.1 (0.9–11.3) Multiple defects 71 1.6 (1.1–2.5) Children of veterans reporting high exposure 46 1.7 (1.2–2.4) CDC, 1989b General Birth Defects Study—hospital records Birth defects 130 1.0 (0.8–1.3) Major birth defects 51 1.2 (0.8–1.9) Digestive system defects 18 2.0 (0.9–4.6) Birth defects—black Vietnam veterans only 21 3.4 (1.5–7.6) Donovan et al., Australian Vietnam veterans 1984 Vietnam veterans vs all other men 127 1.02 (0.8–1.3) National Service veterans—Vietnam service vs no Vietnam service 69 1.3 (0.9–2.0)

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 541 TABLE 7-3 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a Erikson et al., Vietnam veterans identified through the CDC 1984a Metropolitan Atlanta Congenital Defects Program Any major birth defects 428 1.0 (0.8–1.1) Multiple birth defects with reported exposure 25 1.1 (0.7–1.7) EOI-5: spina bifida 1 2.7 (1.2–6.2) EOI-5: cleft lip with or without cleft palate 5 2.2 (1.0–4.9) ABBREVIATIONS: AFHS, Air Force Health Study; AIHW, Australian Institute of Health and Wel- fare; CDC, Centers for Disease Control and Prevention; CI, confidence interval; EOI, exposure op- portunity index; NS, not significant; SIR, standardized incidence ratio; SS, statistically significant. Unless otherwise indicated, studies show paternal exposure. a Given when available. b 95% confidence intervals contained one for all outcomes. Anencephaly and spina bifida included in this calculation. c Number of workers with maximal index of exposure (upper three quartiles) for any job held up to 3 months prior to conception. d Excludes stillbirths, neonatal death, or dislocated or dislocatable hip. e 90% confidence interval. * Information not provided by study authors. TABLE 7-4 Selected Epidemiologic Studies—Neural Tube Defects in the Offspring of Subjects Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a OCCUPATIONAL Studies Reviewed in Update 1998 Blatter et al., Dutch farmers 1997 Spina bifida—moderate/heavy exposure Pesticide use 8 1.7 (0.7–4.0) Herbicide use 7 1.6 (0.6–4.0)g Kristensen Norwegian farmers—spina bifida (maternal and paternal et al., 1997 exposure) Tractor spraying equipment 28 1.6 (0.9–2.7) Tractor spraying equipment and orchards or greenhouses 5 2.8 (1.1–7.1) Dimich- Sawmill workers Ward et al., Spina bifida or anencephaly 22b 2.4 (1.1–5.3) 1996 Spina bifida only 18b 1.8 (0.8–4.1) Garry et al., Private pesticide appliers—central nervous system 1996 defects 6 1.1 (0.5–2.4) continued

542 VETERANS AND AGENT ORANGE: UPDATE 2006 TABLE 7-4 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a ENVIRONMENTALc New Studies Cordier Residents of the Rhône-Alpes region of France et al., 2004 (maternal and paternal exposure) 49 0.9 (0.6–1.2) Studies Reviewed in VAO Stockbauer Persons in Missouri with documented TCDD soil et al., 1988 contamination—central nervous system defects (maternal; paternal; in utero exposure) 3 3.0 (0.3–35.9) Hanify et al., Spraying of 2,4,5-T in New Zealand (all exposures) 1981 Anencephaly 10 1.4 (0.7–2.9)d Spina bifida 13 1.1 (0.6–2.1)d VIETNAM VETERANS Studies Reviewed in Update 2000 AIHW, 1999 Australian Vietnam veterans—Validation Study Spina bifida—maxima 50 33 expected (22–44) Anencephaly 13 16 expected (8–24) Studies Reviewed in Update 1996 Wolfe et al., Air Force Operation Ranch Hand personnel—neural 1995 tube defectse 4 (*) Studies Reviewed in VAO CDC, 1989b Vietnam Experience Study Spina bifida Vietnam veterans’ children 9 1.7 (0.6–5.0) Non-Vietnam veterans’ children 5 (*) Anencephaly Vietnam Veterans’ children 3 (*) Non-Vietnam veterans’ children 0 (*) Erickson Birth Defects Study—Vietnam veterans et al., Spina Bifida 19 1.1 (0.6–1.7) 1984a,b Anencephaly 12 0.9 (0.5–1.7) EOI-5: spina bifida 20f 2.7 (1.2–6.2) EOI-5: anencephaly 7f 0.7 (0.2–2.8) ADVA, 1983 Australian Vietnam veterans—neural tube defects 16 0.9 ABBREVIATIONS: 2,4,5-T, 2,4,5-trichlorophenoxyacetic acid; AIHW, Australian Institute of Health and Welfare; CDC, Centers for Disease Control and Prevention; EOI, exposure opportunity index; NR, not reported. Unless otherwise indicated, studies show paternal exposure. a Given when available. b Number of workers with maximal index of exposure (upper three quartiles) for any job held up to 3 months prior to conception. c Either or both parents potentially exposed. d 90% confidence interval.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 543 TABLE 7-4 Continued e Of the four neural tube defects reported among Ranch Hand offspring there were two spina bifida (high dioxin level), one spina bifida (low dioxin), and one anencephaly (low dioxin). No neural tube defects were reported in the comparison cohort. 454 post-service births were studied in Ranch Hand veterans; 570 in comparison cohort. f Number of Vietnam veterans fathering a child with a neural tube defect given any exposure op- portunity index. g Calculated from data presented in the paper. * Information not provided by study authors. the first year of life due to any congenital defect (p 0.047), but the relation- ship was slightly more significant for all deaths in the first year (p 0.023); no relationship was found for the intervals closer to birth. This study had several methodologic weaknesses, including the lack of individual-level information on other risk factors and of individual exposure data. No data on specific types of congenital defects were included. Vietnam-Veteran Studies No new Vietnam veteran studies concerning exposure to the compounds of interest and birth defects were published since Update 2004. Biologic Plausibility Laboratory studies have established that maternal exposure to TCDD during pregnancy is associated with a wide variety of birth defects, which depend on the timing of exposure and the species being studied. However, laboratory stud- ies of potential male-mediated developmental toxicity attributable to exposure to TCDD and herbicides, specifically with regard to birth defects, are too limited to permit conclusions. It is notable that both the aryl hydrocarbon receptor (AhR) and the AhR nuclear translocator are expressed in the human testis and sperm (Khorram et al., 2004; Shultz et al., 2003), and studies in rodents have shown that TCDD exposure results in significant changes in gene expression in spermato- cytes and Sertoli cells; hence, cells of the testis are responsive to TCDD expo- sure (Kuroda et al., 2005; Yamano et al., 2005). Furthermore, laboratory studies have shown that TCDD inhibits spermatogenesis and reduces erectile function in animals (Moon et al., 2004; Simanainen et al., 2004). Thus, there is biologic potential for paternal exposure to contribute to TCDD-induced reproductive and developmental toxicity. The only study assessing paternal TCDD exposure in humans that was re- viewed for the present update failed to show any evidence that paternal TCDD

544 VETERANS AND AGENT ORANGE: UPDATE 2006 exposure of male chemical workers caused an increase in birth defects in their offspring. In a previous laboratory study, paternal exposure of rats to TCDD failed to result in any birth defects in the F1 generation (Chahoud et al., 1991). And an epidemiologic study of maternal serum TCDD failed to establish any link with maternal thyroid dysfunction, which might contribute to cognitive or motor impairment in offspring (Foster et al., 2005). The biologic plausibility of reproductive effects in general arising from ex- posure to the chemicals of interest is discussed at the end of this chapter. Synthesis For this update, the only new occupational study of birth defects and expo- sures to the chemicals of interest was a follow-on study of the children of men in the NIOSH cohort (Lawson et al., 2004). Because only 1,166 liveborn and stillborn infants were available for analyses, the information generated was too sparse to provide additional insights into the risks of birth defects. Birth defects were addressed indirectly by a new environmental study of residents in areas sur- rounding metropolitan waste-incineration facilities in Japan (Tango et al., 2004). On the basis of large numbers of births, the Japanese study found an association between residence in the areas with the highest soil dioxin concentrations and deaths before the first birthday due to any congenital abnormality, but this rela- tionship did not carry over to deaths occurring in the first month or in the first week of life. Ngo et al. (2006) conducted a systematic review and meta-analysis of the association between Agent Orange and birth defects. Relevant studies published from 1966 to 2002 were identified by search engines, and the authors also in- cluded unpublished conference proceedings and information from researchers on findings from unpublished studies. They reviewed 22 studies, including 13 Vietnamese studies (11 of them unpublished) and 9 of Vietnam veterans (2 unpublished). Secondary analyses of primary data reviewed by previous VAO committees fall outside the type of findings ordinarily factored into the evidence bases evaluated in the VAO reviews, but given the nature of the reported results, the present committee wanted to comment on the Ngo et al. review. Unpublished studies are outside VAO’s inclusion criteria, so the 13 unpublished studies con- sidered by Ngo et al. were not included in previous VAO reviews of the associa- tion between the chemicals of interest and birth defects. All the studies that Ngo et al. indicate as published were reviewed in the VAO series. The Vietnam-veteran study that provided the highest risk estimates for birth defects (Field and Kerr, 1988) was excluded from consideration by previous VAO committees because of serious methodologic issues (IOM, 1994). In limiting their review to Agent Orange, Ngo et al. also excluded the broader array of occupational and environ-

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 545 mental studies on the components of Agent Orange that previous VAO reviews have addressed. Ngo et al. (2006) reported a substantially greater strength of association between exposure to Agent Orange and birth defects in the studies of Vietnamese popula- tions (RR 3.00, 95% CI 2.19–4.12) than in those of non-Vietnamese populations (RR 1.29, 95% CI 1.04–1.59). The non-Vietnamese study populations consisted of Vietnam veterans, who were almost exclusively men, whereas the Vietnamese popu- lations had a much greater likelihood of maternal exposure. Only Kang et al. (2000) addressed birth defects among the offspring of female Vietnam War veterans, who overall constitute fewer than 10,000 of the roughly 3 million US Vietnam veterans. Ngo et al. (2006) also conducted subgroup meta-analyses based on presumed exposure intensity. They reported no significant increased risk of birth defects among non-Ranch Hand veterans (RR 1.04, 95% CI 0.93–1.16). Combining three sequential studies of Ranch Hand veterans, who were presumed to have been more highly exposed, risk was increased (RR 1.20, 95% CI 1.08–1.34), but they were unable to control for non-independent observations among the stud- ies of Ranch Hand veterans studies. Combined risks from studies of North Viet- namese veterans and of sprayed Vietnamese civilians were successively higher. The numbers of cases reported in the studies reviewed by Ngo et al. (2006) were too small to allow meta-analysis of specific types of birth defects. In general, the relatively small number of offspring among Vietnam veterans seriously restricts the ability to detect statistically significant increases in specific birth defects, and meta-analytic methods are the best approach to assessing the overall import of the studies of exposures to the chemicals of interest and the risk of specific birth defects. In addition, as the offspring of veterans become older, the risk of diseases stemming from congenitally transmitted defects that alter normal physiologic function, such as endocrine and reproductive function, merits increasing attention. Conclusions There were no new relevant studies on the association between parental exposure to 2,4-D, 2,4,5-T, TCDD, cacodylic acid, or picloram and spina bifida in offspring. The committee concludes that the evidence is still limited or sug- gestive of an association between exposure to the compounds of interest and spina bifida. Its evaluation of the evidence reviewed here and in previous VAO reports leads the committee to conclude that there is still inadequate or insufficient evi- dence of an association between exposure to the compounds of interest and all other birth defects.

546 VETERANS AND AGENT ORANGE: UPDATE 2006 CHILDHOOD CANCER The American Cancer Society estimated that 9,200 children under the age of 15 years would have a diagnosis of cancer in the United States in 2004 (ACS, 2004). Treatment and supportive care of children with cancer have improved greatly, and mortality has declined by 49 percent over the last 30 years. Despite those advances, cancer remains the leading cause of death from disease in chil- dren under the age of 15 years, and 1,510 deaths were projected for 2004 (ACS, 2004). Leukemia is the most common cancer in children. It accounts for about one-third of all childhood cancer cases; leukemia is expected to be diagnosed in nearly 2,760 children in 2004 (ACS, 2004). Of those, nearly 2,000 will have acute lymphocytic leukemia (ALL); most of the rest will have acute myelogenous leukemia (AML). AML (International Classification of Diseases, Ninth Edition [ICD-9] 205) is commonly referred to as acute myeloid leukemia and acute nonlymphocytic leukemia. For consistency, this report uses acute myelogenous leukemia, or simply AML, regardless of usage in the source materials. ALL is most common in early childhood, peaking between the ages of 2 and 3 years, and AML is most common during the first 2 years of life. ALL incidence is consistently higher in boys than in girls; AML incidence is similar in boys and girls (NCI, 2001). Through early adulthood, ALL rates are about twice as high in whites as in blacks; AML exhibits no consistent pattern in this respect. Chapter 6 contains additional information on leukemia as part of the discussion of adult cancer. The second-most common group of cancers in children are those of the central nervous system—the brain and the spinal cord. Other cancers in children include lymphomas, bone cancers, soft-tissue sarcomas, kidney cancers, eye can- cers, and adrenal gland cancers. Compared with adult cancers, relatively little is known about the etiology of most childhood cancers, especially about potential environmental risk factors and the effect of parental exposures. Conclusions from VAO and Updates The committee responsible for VAO concluded that there was inadequate or insufficient evidence of an association between exposure to 2,4-D, 2,4,5-T, TCDD, picloram, or cacodylic acid and childhood cancers. Additional informa- tion available to the committees responsible for Update 1996 and Update 1998 did not change that conclusion. The committee responsible for Update 2000 reviewed the material in earlier VAO reports and newly available published literature and determined there was limited or suggestive evidence of an association between exposure to at least one of the compounds of interest and AML. After the release of Update 2000, investigators involved in one study discovered an error in their published data. The committee reconvened to evaluate the previously reviewed

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 547 and new literature regarding AML, and Acute Myelogenous Leukemia (IOM, 2002) was produced. It reclassified AML from “limited/suggestive evidence of an association” to “inadequate evidence to determine whether an association ex- ists.” Table 7-5 summarizes the results of the relevant studies. The committees responsible for Update 2000, Update 2002, and Update 2004 reviewed the mate- rial in earlier VAO reports and in newly available published literature and agreed that there remained inadequate or insufficient evidence of an association between exposure and childhood cancers. Update of Epidemiologic Literature Occupational Studies Two related papers from the Children’s Oncology Group addressed exposures to pesticides and the risk of childhood germ-cell tumors (GCTs). One focused on residential pesticides and chemicals (Chen et al., 2006) and the other on parental occupational exposures (Chen et al., 2005), but they were based on the same overall case–control study. GCTs are very rare childhood cancers, so recruitment was done throughout the United States and included incident cases diagnosed from 1993 to 2001 in children under the age of 15 years. Controls were selected by random-digit dialing and frequency-matched on sex, year of birth, and state. Data were collected via two questionnaires (telephone and self-administered). Maternal exposure to herbicides at home from 6 months before pregnancy until termination of breastfeeding was associated with a slightly increased risk among female offspring (OR 1.4, 95% CI 1.0–2.0), but not among male offspring (OR 1.0, 95% CI 0.5–1.8). Similarly, Chen et al. (2005) reported on maternal and paternal exposures to pesticides before, during, and after pregnancy from occupational exposures. The data on analyses for exposures to herbicides, insecticides, or fungicides were not presented, but it was stated that the only positive association was for maternal exposure to herbicides at work during the postnatal period and GCTs in female offspring (OR 2.3, 95% CI 1.0–5.2); therefore, exposures with timing relevant to this review were not associated with increased risks of GCTs in the offspring. Both studies are limited by the questionable reliability of the self-reported ex- posures, reporting of the results only for the preconception through postnatal period, the small numbers of subjects, and the failure to consider residential and occupational herbicide exposures simultaneously. Environmental Studies Reynolds et al. (2005b) conducted a case–control study on childhood cancer and gestational exposure to pesticides. Compilation in California’s Pesticide Use Reporting Database (CPUR) of information of amount and location of application

548 VETERANS AND AGENT ORANGE: UPDATE 2006 TABLE 7-5 Selected Epidemiologic Studies—Childhood Cancers Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a OCCUPATIONAL New Studies Chen et al., Parental occupational exposure to pesticide and 2005 childhood GCTs Maternal 32 1.1 (0.7–1.6) Paternal 39 0.9 (0.6–1.3) Reynolds Maternal exposure to agricultural pesticide in class of et al., 2005b “probable human carcinogens” (including cacodylic acid) during 9 months prior to delivery All sites 223 1.0 (0.9–1.2) Leukemias 179 1.2 (0.9–1.5) Central nervous system tumors 31 0.9 (0.5–1.4) Studies Reviewed in Update 2004 Flower Offspring of male pesticide applicators in Iowa from et al., 2004 AHS Maternal exposure to chlorophenoxy herbicides 7 0.7 (0.3–1.5) Paternal exposure to chlorophenoxy herbicides 28 1.3 (0.6–2.6) Maternal exposure to 2,4-D 7 0.7 (0.3–1.6) Paternal exposure to 2,4-D 26 1.29 (0.7–2.4) Studies Reviewed in Update 2000 Heacock Offspring of sawmill workers exposed to fungicides et al., 2000 contaminated with PCDDs and PCDFs Leukemia All workers offspring—incidence 11 1.0 (0.5–1.8) Offspring of workers with high chlorophenate exposure 5 0.8 (0.2–3.6)b Brain cancer All workers offspring—incidence 9 1.3 (0.6–2.5) Offspring of workers with high chlorophenate exposure 5 1.5 (0.4–6.9)b Buckley Children’s Cancer Study Group—exposure to pesticides et al., 1989 and weed killers—AML Any paternal exposure 27 2.3 (p 0.5) Paternal exposure 1,000 days 17 2.7 (1.0–7.0) Maternal exposure 1,000 days 7 undefined

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 549 TABLE 7-5 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a ENVIRONMENTAL New Studies Chen et al., Childhood GCTs and residential exposure to herbicides 2006 6 months before conception, during gestation, or through breastfeeding period Maternal exposure 47 1.3 (0.9–1.7) Daughters 36 1.4 (1.0–2.0) Sons 11 1.0 (0.5–1.8) Paternal exposure 90 1.0 (0.7–1.3) Daughters 32 1.2 (0.7–2.0) Sons 58 1.0 (0.7–1.4) Studies Reviewed in Update 2002 Daniels Neuroblastoma risk in children (case–control study) (as et al., 2001 reported by both parents) Pesticides in the home (used ever) * 1.6 (1.0–2.3) Herbicides in the garden * 1.9 (1.1–3.2) Pesticides in the garden * 2.2 (1.3–3.6) Buckley NHL diagnosed at the age of 20 years in children with et al., 2000 potential prenatal exposure to herbicides * (*)c Kerr et al., Neuroblastoma risk in children 2000 Maternal occupational exposure to insecticides 40 2.3 (1.4–3.7) Paternal exposure to dioxin 7 6.9 (1.3–68.4) Studies Reviewed in Herbicide/Dioxin Exposure and AML in the Children of Veterans Kristensen Children of agricultural workers in Norway et al., 1996 Children with AML whose parents purchased pesticides 12 1.4 (0.6–2.9) Studies Reviewed in Update 2000 Meinert Childhood cancer—population-based case–control study et al., 2000 Leukemia Paternal exposure; year before pregnancy 62 1.5 (1.1–2.2) Paternal exposure; during pregnancy 57 1.6 (1.1–2.3) Maternal exposure; year before pregnancy 19 2.1 (1.1–4.2) Maternal exposure, during pregnancy 15 3.6 (1.5–8.8) Lymphomas Paternal exposure, year before pregnancy 11 1.5 (0.7–3.1) Paternal exposure, during pregnancy 10 1.6 (0.7–3.6) Maternal exposure, year before pregnancy 3 2.9 (0.7–13) Maternal exposure, during pregnancy 4 11.8 (2.2–64) Pearce and Kidney cancer in subjects (1–15 yrs) with paternal Parker, 2000 occupation in agriculture 21 0.9 (0.2–3.8) continued

550 VETERANS AND AGENT ORANGE: UPDATE 2006 TABLE 7-5 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a Infante- Childhood ALL in households using herbicides— Rivard population-based case–control study et al., 1999 Exposure during pregnancy 118 1.8 (1.3–2.6) Exposure during childhood 178 1.4 (1.1–1.9) Studies Reviewed in Update 1996 Pesatori Seveso residents aged 0–19 years—10-year follow-up, et al., 1993 morbidity, all exposure zones All cancers 17 1.2 (0.7–2.1) Ovary and uterine adnexa 2 — (0 expected) Brain 3 1.1 (0.3–4.1) Thyroid 2 4.6 (0.6–32.7) Hodgkin’s lymphoma 3 2.0 (0.5–7.6) Lymphatic leukemia 2 1.3 (0.3–6.2) Myeloid leukemia 3 2.7 (0.7–11.4) Bertazzi Seveso residents aged 0–19 years—10-year follow-up, et al., 1992 mortality, all exposure zones All cancers 10 7.9 (3.8–13.6) Leukemias 5 3.9 (1.2–1.8) Lymphatic leukemia 2 1.6 (0.1–4.5) Myeloid leukemia 1 0.8 (0.0–3.1) Leukemia, others 2 1.6 (0.1–4.6) Central nervous system tumors 2 1.6 (0.1–4.6) VIETNAM VETERANS Studies Reviewed in Herbicide/Dioxin Exposure and AML in the Children of Veterans AIHW, Australian Vietnam veterans’ children—Revised 2001 Validation Study—AML 12d 1.3 (0.8–4.0) Studies Reviewed in Update 2000 AIHW, 2000 Australian Vietnam veterans’ children—Validation Study–AML This study, which incorrectly calculated the expected number of AML cases, is superseded by AIHW, 2001 above. Wen et al., Case–control study of children’s leukemia 2000 AML and ALL Father ever served in Vietnam or Cambodia 117 1.2 (0.9–1.6) 1 year in Vietnam or Cambodia 61 1.4 (0.9–2.0) 1 year in Vietnam or Cambodia 49 1.2 (0.8–1.7) AML only Father ever served in Vietnam or Cambodia 40 1.7 (1.0–2.9) 1 year in Vietnam or Cambodia 13 2.4 (1.1–5.4) 1 year in Vietnam or Cambodia 16 1.5 (0.7–3.2)

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 551 TABLE 7-5 Continued Estimated Exposed Relative Risk Reference Study Population Casesa (95% CI)a Studies Reviewed in VAO CDC, 1989b Vietnam Experience Study—outcomes in the offspring of veterans Cancer 25 1.5 (0.7–2.8) Leukemia 12 1.6 (0.6–4.0) Field and Cancer in children of Australian Vietnam veterans 4 (*) Kerr, 1988 Erickson CDC Birth Defects Study—children of Vietnam veterans et al., 1984b “Other” neoplasms 87 1.8 (1.0–3.3) ABBREVIATIONS: AFHS, Air Force Health Study; AHS, Agricultural Health Study, AIHW, Aus- tralian Institute of Health and Welfare; ALL, acute lymphocytic leukemia; AML, acute myeloid leukemia; CDC, Centers for Disease Control and Prevention; CI, confidence interval; EOI, exposure opportunity index; GCT, germ-cell tumor; NS, not significant; SS, statistically significant. a Given when available. b OR estimated using low exposure subjects as the comparison cohort. c No information on herbicides as a class, distinct from insecticides or other pesticides, was available; exposures before conception were not singled out, and no distinction between maternal and paternal exposure was made. d Of the 12 cases, 9 were observed and 3 additional cases were estimated to have occurred in the portion of the cohort whose data were not validated. * Information not provided by study authors. Unless otherwise indicated, studies show paternal exposure. by specific product began in 1990. Considering children born after September 1990 and diagnosed with cancer before the age of 5, they identified 2,189 children entered in California’s population-based cancer registry by the end of 1997. A total of 4,335 live controls were randomly selected from California birth certifi- cates and matched on date of birth and sex. A geographic information system approach was 99 percent successful in geocoding residence at time of birth for 2,189 case mothers and 4,388 control mothers, permitting linkage to the CPUR. Considering the amount of pesticide applied and the distance of the treated areas from the mother’s residence, cumulative exposures for the 9 months before birth were determined for several different classifications of pesticides. California’s category of pesticides that are “probable human carcinogens,” which includes cacodylic acid, is the only classification of possible relevance for this review. Cases of all types of cancer were no more likely than controls to have experienced high exposure during gestation to “probable human carcinogens” (OR 1.01, 95% CI 0.85–1.20); the same was true for the subset of leukemias (OR 1.17, 95% CI 0.90–1.54) or central nervous system cancers (OR 0.86, 95% CI 0.54–1.36). No individual-level data were obtained for the study participants or

552 VETERANS AND AGENT ORANGE: UPDATE 2006 their parents regarding lifestyle or other personal characteristics to permit adjust- ment for possible confounder. The exposure assessment assumes that residential proximity is a good indicator of exposure. Distance to pesticide exposure sources was based on the children’s residences at birth, which would lead to at least some misclassification due to residential mobility. Finally, although the study included many subjects, most of them lived in areas that experienced little or no agricul- tural pesticide use, so the risk estimates for the high exposure categories were based on small numbers. Vietnam-Veteran Studies No new Vietnam veteran studies concerning exposure to the compounds of interest and childhood cancer were published since Update 2004. Biologic Plausibility Paternal or maternal exposure to xenobiotics potentially could increase the susceptibility of offspring to developing cancer through multiple mechanisms. Susceptibility could be increased by inheriting a genetic predisposition, which by itself could increase the development of cancer or the likelihood of developing cancer after future exposure to a carcinogen; the mother or father would transmit either an acquired genetic defect or an epigenetic alteration that predisposed the child to cancer. Alternatively, a maternally mediated increased susceptibility to childhood cancer could result from direct exposure of a child in utero or via lactation to a xenobiotic that induces epigenetic alterations that increase cancer susceptibility or is itself a carcinogen. It has been shown that prenatal TCDD exposure of rats is associated with al- tered mammary gland differentiation and an increase in the number of mammary adenocarcinomas (Brown et al., 1998). A recent study’s demonstration that early postnatal TCDD exposure does not increase mammary-cancer risk (Desaulniers et al., 2004) suggests that TCDD-induced changes in utero mediate the increase in cancer susceptibility. Developmental epigenetic alterations may be involved in those prenatal effects. TCDD has been shown to suppress the expression of two tumor-suppressor genes, p16Ink4a and p53, via an epigenetic mechanism that ap- pears to involve DNA methylation (Ray and Swanson, 2004). Similarly, it was re- ported that prenatal TCDD exposure increases methylation of two growth-related imprinted genes, H19 and Igf2, in the developing fetus (Wu et al., 2004). Although there is no direct evidence from animal models that TCDD in- creases the risk of childhood cancers, such as acute leukemia or GCTs, emerging research suggests that prenatal TCDD exposure can disrupt epigenetic imprinting patterns and alter organ differentiation, and this could contribute to an increased susceptibility to cancer later in life. A recent study has shown that chromosomal rearrangements associated with childhood ALL are evident in the neonatal blood

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 553 spots; this suggests that childhood leukemias begin before birth and that mater- nal and perinatal exposures to xenobiotics may contribute to genetic mutations (Smith et al., 2005). Synthesis The studies reviewed for this update did not find significant associations between the relevant exposures and childhood cancers. As with other outcomes in the offspring of Vietnam veterans, the small number of these rare childhood cancers expected among the circumscribed number of Vietnam veterans would seriously hinder detection of any actual increases; meta-analytic methods may be the best approach for assessing the overall significance of the association between exposures to the chemicals of interest and the risk of specific childhood cancers. Conclusions On the basis of its evaluation of the evidence reviewed here and in previous VAO reports, the committee concludes that there is inadequate or insufficient evidence of an association between exposure to the compounds of interest and childhood cancers. SUMMARY Overall Biologic Plausibility of Reproductive and Developmental Effects This section summarizes the general biologic plausibility of a connection be- tween exposure to the compounds of interest and reproductive and developmental effects on the basis of data from animal and cellular studies. Chapter 3 presents details of the committee’s evaluation of data from the recent studies. TCDD induces a variety of adverse reproductive and developmental effects in laboratory animals. The administration of TCDD to male animals elicits re- productive toxicity by affecting testicular and seminal vesicle weight and func- tion and by decreasing the rate of sperm production. The mechanisms of those effects are not known, but a primary hypothesis is that they are mediated through dysregulation of testicular steroidogenesis. Exposure to TCDD is associated with increased estradiol secretion and decreased testosterone secretion; both hormones regulate sperm production. However, high doses of TCDD were required to elicit those effects, and TCDD-exposed male rats were able to sire viable fetuses. Thus, there is little evidence from laboratory studies that TCDD exposure of men con- tributes to reduced fertility. Many studies also have examined the effects of TCDD on the female re- productive system. Two primary mechanisms that probably contribute to abnor-

554 VETERANS AND AGENT ORANGE: UPDATE 2006 mal follicle development and decreased numbers of ova after TCDD exposure are crosstalk of the AhR with the estrogen receptor and dysregulation of the hypothalamic-pituitary-gonadal axis. Interaction of the activated AhR leads to inhibition of estradiol-induced gene expression and to enhancement of estrogen- receptor protein degradation; both activities may contribute to TCDD’s anties- trogenic effects. TCDD also dysregulates the secretion pattern of preovulatory gonadotropin hormones, and this leads to abnormal and reduced follicle devel- opment. In addition, oocytes are directly responsive to TCDD. Thus, TCDD’s effects on hormone concentrations, hormone-receptor signaling, and ovarian responsiveness to hormones all probably contribute to TCDD-induced female reproductive toxicity. On the basis of animal data, male and female reproductive effects of TCDD in humans are biologically plausible. Furthermore, a recent study has shown that dioxins and dibenzofurans significantly correlate with dysregulation of estrogen metabolism in pregnant women (Wang et al., 2006)— evidence of a biologically plausible mechanism of TCDD’s adverse effects on human reproduction. In animal studies, TCDD crosses the placenta and is transferred via breast milk. In humans, TCDD has been measured in circulating maternal blood, cord blood, placenta, and breast milk (Suzuki et al., 2005), and it is estimated that an infant breast fed for 1 year accumulates a dose of TCDD that is 6 times higher than an infant not breast fed (Lorber and Phillips., 2002). Thus, the exposure of human infants in utero and via lactation has been demonstrated. In animal studies, TCDD exposure during pregnancy can reduce body weight at birth, but this typically occurs at high doses. Thus, there is little evidence that TCDD exposure is associated with reduction in birth weight. TCDD is a potent teratogen in all laboratory species that have been studied, although the pattern of induced birth defects is often species-specific. For exam- ple, fish, mice, and avian embryos exhibit substantial alterations in craniofacial development—shortened jaw in piscine species, cleft palate in mice, and beak malformations in birds. The developing cardiovascular system is also a com- mon target for TCDD-induced teratogenicity, and it has been shown that cardiac myocytes and the endothelial lining of the heart and blood vessels are primary target sites of TCDD’s effects. Cytochrome P450 1A1 induction or alterations in pathways controlled by vascular endothelial growth factor might mediate the early lesions that result in TCDD-related vascular derangements. That an- tioxidant treatment provides protection against TCDD-induced embryotoxicity in some systems suggests that reactive oxygen species might be involved in the teratogenic effects of exposure to TCDD. Several reports of studies in exposed animals and humans suggest that low perinatal exposure to TCDD and 2,4-D could impair brain development. Outcomes can be subtle, ranging from altered learning and memory to modified sex-related behavior. The mechanisms of those effects are unclear. Studies in rodents show that a single maternal dose of TCDD produces

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 555 malformations of the external genitalia and functional reproductive alterations in female progeny, including decreased fertility rate, reduced fecundity, cystic en- dometrial hyperplasia, and disrupted estrous cycles. Those effects depend on the timing of exposure. Similarly, male progeny exhibit alterations in reproductive- organ development and function. Maternal exposure to TCDD impairs prostate growth and seminal vesicle weight and branching and decreases sperm production and caudal epididymal sperm number in offspring. Little research has been conducted on the offspring of male animals exposed to herbicides. Feeding of simulated Agent Orange mixtures to male mice pro- duced no adverse effects in offspring; a statistically significant excess of fused sternebrae in the offspring of the two most highly exposed groups was attributed to an anomalously low rate of this defect in the controls (Lamb et al., 1981). Altered sex ratio might reflect the effects of exposure to the chemicals of interest on reproductive capability. It has been hypothesized that concentrations of parental hormones at conception or induction of lethal mutations before birth could affect sex ratio. There has been no work with experimental animals that specifically examined the effects of TCDD on sex ratios of offspring, nor have any alterations in sex ratio been reported in animal studies that examined devel- opmental effects of TCDD on offspring. The mechanisms by which TCDD induces birth defects have not been es- tablished and are probably species- and organ-specific. Nonetheless, studies have consistently demonstrated that TCDD-induced developmental toxicity required the AhR. That has been definitively established in mice that lack AhR expression. When pregnant AhR-null mice are exposed to TCDD, the fetuses fail to exhibit any of the typical developmental malformations associated with TCDD exposure. The activated AhR mediates changes in gene transcription, so the inappropriate and sustained activation of AhR by TCDD during development appears to be a key first step in mediating TCDD’s developmental toxicity. Although structural differences in the AhR have been identified among species, it functions similarly in animals and humans. Therefore, a common mechanism probably underlies the reproductive and developmental toxicity of TCDD in humans and animals, and data on animals support a biologic basis of TCDD’s toxic effects. Little information is available on the reproductive and developmental effects of exposure to the herbicides discussed in this report. Studies indicate that 2,4-D does not affect male or female fertility and does not produce fetal abnormalities. Offspring of pregnant rodents exposed to 4-(2,4-dichlorophenoxy)butyric acid (2,4- DB) exhibit a reduced growth rate and increased mortality (Charles et al., 1999), but only after very high doses. Exposure to 2,4-D also alters the concentration and function of reproductive hormones and prostaglandins. One study reported an increased incidence of malformed offspring of male mice exposed to a mixture of 2,4-D and picloram in drinking water; however, paternal toxicity was observed in the high-dose group, and there was no clear dose–response relationship (Blakley et al., 1989). Picloram alone produced fetal abnormalities in rabbits at doses that

556 VETERANS AND AGENT ORANGE: UPDATE 2006 are also toxic to the pregnant animals (John-Greene et al., 1985), but that effect has not been seen in many studies. 2,4,5-T was toxic to fetuses when administered to pregnant rats, mice, and hamsters; its ability to interfere with calcium homeostasis in vitro has been documented and linked to its teratogenic effects on the early de- velopment of sea urchin eggs. Cacodylic acid is toxic to rat, mouse, and hamster fetuses at high doses that are also toxic to the pregnant mother. There is growing evidence from laboratory animal and human studies that exposures during fetal or postnatal development can lead to adverse effects later in life that are not immediately apparent as structural malformations or functional deficits. For example, exposure of humans and rats to TCDD in early postnatal life induces dental aberrations and reduces enamel maturation of teeth (Alaluusua et al., 2004; Gao et al., 2004). A study of human exposure to background con- centrations of dioxins, furans, and PCBs during prenatal development (Nakajima et al., 2006) suggests possibly more relationship with reduced motor development in 6-month-old infants than with their mental development; however, the few sig- nificant correlations found among dozens of comparisons made were for specific congeners with low relative potency (TEFs), so the study is essentially negative for developmental effects arising from prenatal exposure to TCDD. The foregoing suggests that a connection between TCDD exposure and hu- man reproductive and developmental effects is, in general, biologically plausible. However, more definitive conclusions about the potential for such TCDD toxic- ity in humans are complicated by differences in sensitivity and susceptibility among individual animals, strains, and species; by the lack of strong evidence of organ-specific effects among species; by differences in route, dose, duration, and timing of exposure; and by substantial differences in the toxicokinetics of TCDD between laboratory animals and humans. Experiments with 2,4-D and 2,4,5-T indicate that they have subcellular effects that could constitute a biologi- cally plausible mechanism for reproductive and developmental effects. Evidence from animals, however, indicates that they do not have reproductive effects and that they have developmental effects only at very high doses. There is insufficient information on picloram and cacodylic acid to assess the biologic plausibility of those compounds’ reproductive or developmental effects. Considerable uncertainty remains about how to apply toxicologic informa- tion to the evaluation of potential health effects of herbicide or TCDD exposure on the offspring of Vietnam veterans. Scientists disagree over the extent to which information derived from animal and cellular studies can be used to predict hu- man health outcomes and over the extent to which the health effects of high-dose exposure can be extrapolated to low-dose exposure. The biologic mechanisms that underlie TCDD’s toxic effects continue to be a subject of active research, and future VAO updates are likely to have more and better information on which to base conclusions, at least for TCDD.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 557 Synthesis The studies reviewed for this update did not find any significant associa- tions between the relevant exposures and reproductive outcomes. The scientific evidence supports the biologic plausibility of a connection between exposure to the chemicals of interest and reproductive effects, but the epidemiologic studies of occupational cohorts, exposed communities, and Vietnam veterans have not provided conclusive evidence of any additional associations between exposures and an array of reproductive outcomes and conditions among the offspring of exposed parents. The mechanisms by which the chemicals exert their biologic effects are still subjects of scientific investigation. With the aging of the Vietnam- veteran population, additional studies on fertility, spontaneous abortion, and sex ratio cannot be expected, although there may be additional studies of reproductive outcomes in other populations with exposure to the chemicals of interest. The possibility of structural or functional abnormalities in the offspring of exposed people will continue to be of interest. Conclusions There is inadequate or insufficient evidence of an association between expo- sure to 2,4-D, 2,4,5-T, TCDD, picloram, or cacodylic acid and altered hormone concentrations; semen quality; infertility; spontaneous abortion; late fetal, neo- natal, or infant death; low birth weight or preterm delivery; birth defects other than spina bifida; and childhood cancers. There is limited or suggestive evidence of an association between exposure to the compounds of interest and spina bifida. There is limited or suggestive evidence that the specific combination of pa- ternal exposure to TCDD is not associated with risk of spontaneous abortion. REFERENCES1 Abell A, Juul S, Bonde JP. 2000. Time to pregnancy among female greenhouse workers. Scandinavian Journal of Work, Environment and Health 26(2):131–136. ACS. 2004. Cancer Facts and Figures 2004. http://www.cancer.org/downloads/STT/CAFF_finalP- WSecured.pdf (Accessed September 13). ADVA (Australia Department of Veterans Affairs). 1983. Case–Control Study of Congenital Anoma- lies and Vietnam Service. Canberra. AFHS (Air Force Health Study). 1992. An Epidemiologic Investigation of Health Effects in Air Force Personnel Following Exposure to Herbicides. Reproductive Outcomes. Brooks AFB, TX: USAF School of Aerospace Medicine. AL-TR-1992-0090. 602 pp. 1Throughout the report the same alphabetic indicator following year of publication is used con- sistently for the same article when there were multiple citations by the same first author in a given year. The convention of assigning the alphabetic indicator in order of citation in a given chapter is not followed.

558 VETERANS AND AGENT ORANGE: UPDATE 2006 AIHW (Australian Institute of Health and Welfare). 1999. Morbidity of Vietnam Veterans: A Study of the Health of Australia’s Vietnam Veteran Community: Volume 3: Validation Study. Canberra, Australia: AIHW. AIHW. 2000. Morbidity of Vietnam Veterans. Adrenal Gland Cancer, Leukaemia and Non-Hodgkin’s Lymphoma: Supplementary Report No. 2. (AIHW cat. no. PHE 28). Canberra, Australia: AIHW. AIHW. 2001. Morbidity of Vietnam Veterans. Adrenal Gland Cancer, Leukaemia and Non-Hodgkin’s Lymphoma: Supplementary Report No. 2. Revised edition (AIHW cat. No. PHE 34). Canberra, Australia: AIHW. Alaluusua S, Calderara P, Gerthoux PM, Lukinmaa PL, Kovero O, Needham L, Patterson DG Jr, Tuomisto J, Mocarelli P. 2004. Developmental dental aberrations after the dioxin accident in Seveso. Environmental Health Perspectives 112(13):1313–1318. Alberman E. 1984. Low birth weight. In: Bracken MB, ed. Perinatal Epidemiology. New York: Oxford University Press. Pp. 86–98. Arbuckle TE, Lin Z, Mery LS. 2001. An exploratory analysis of the effect of pesticide exposure on the risk of spontaneous abortion in an Ontario farm population. Environmental Health Perspec- tives 109(8):851–857. Aschengrau A, Monson RR. 1989. Paternal military service in Vietnam and risk of spontaneous abor- tion. Journal of Occupational Medicine 31(7):618–623. Aschengrau A, Monson RR. 1990. Paternal military service in Vietnam and the risk of late adverse pregnancy outcomes. American Journal of Public Health 80(10):1218–1224. Axmon A, Rylander L, Strömberg U, Hagmar L. 2000. Miscarriages and stillbirths in women with a high intake of fish contaminated with persistent organochlorine compounds. International Archives of Occupational and Environmental Health 73(3):204–208. Berkowitz GS, Papiernik E. 1993. Epidemiology of preterm delivery. Epidemiologic Reviews 15:414–443. Bertazzi PA, Zocchetti C, Pesatori AC, Guercilena S, Consonni D, Tironi A, Landi MT. 1992. Mortality of a young population after accidental exposure to 2,3,7,8-tetrachlorodibenzodioxin. International Journal of Epidemiology 21(1):118–123. Blakley PM, Kim JS, Firneisz GD. 1989. Effects of paternal subacute exposure to Tordon 202c on fetal growth and development in CD-1 mice. Teratology 39(3):237–241. Blatter BM, Hermens R, Bakker M, Roeleveld N, Verbeek AL, Zielhuis GA. 1997. Paternal occupa- tional exposure around conception and spina bifida in offspring. American Journal of Industrial Medicine 32(3):283–291. Bloom AD, ed. 1981. Guidelines for Studies of Human Populations Exposed to Mutagenic and Re- productive Hazards. White Plains, NY: March of Dimes Foundation. Bonde JP, Giwercman A. 1995. Occupational hazards to male fecundity. Reproductive Medicine Review 4:59–73. Bretveld RW, Thomas CMG, Scheepers PTJ, Zielhuis GA, Roeleveld N. 2006a. Pesticide exposure: The hormonal function of the female reproductive system disrupted? Reproductive Biology and Endocrinology 4:20. Bretveld RW, Thomas CM, Scheepers PT, Zielhuis GA, Roeleveld N 2006b. Pesticide exposure: The hormonal function of the female reproductive system disrupted? [Review] Reproductive Biology and Endocrinology 4:30. Brown NM, Manzolillo PA, Zhang JX, Wang J, Lamartiniere CA. 1998. Prenatal TCDD and predis- position to mammary cancer in the rat. Carcinogenesis 19(9):1623–1629. Bryce R. 1991. The epidemiology of preterm birth. In: Kiely M, ed. Reproductive and Perinatal Epidemiology. Boca Raton, FL: CRC Press. Pp. 437–444. Buchanan DL, Sato T, Peterson RE, Cooke PS. 2000. Antiestrogenic effects of 2,3,7,8-tetrachloro- dibenzo-p-dioxin in mouse uterus: Critical role of the aryl hydrocarbon receptor in stromal tissue. Toxicological Sciences 57(2):302–311.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 559 Buckley JD, Robison LL, Swotinsky R, Garabrant DH, LeBeau M, Manchester P, Nesbit ME, Odom L, Peters JM, Woods WG, Hammond GD. 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. Buckley JD, Meadows AT, Kadin ME, Le Beau MM, Siegel S, Robison LL. 2000. Pesticide exposures in children with non-Hodgkin lymphoma. Cancer 89(11):2315–2321. Carmelli D, Hofherr L, Tomsic J, Morgan RW. 1981. A Case–Control Study of the Relationship Be- tween Exposure to 2,4-D and Spontaneous Abortions in Humans. SRI International. Prepared for the National Forest Products Association and the US Department of Agriculture, Forest Service. CDC (Centers for Disease Control and Prevention). 1989a. Health Status of Vietnam Veterans Vols. I–V, Supplements A–C. Vietnam Experience Study. Atlanta, GA: US Department of Health and Human Services. CDC. 1989b. Health Status of Vietnam Veterans. Vietnam Experience Study, Vol. V, Reproductive Outcomes and Child Health. Atlanta, GA: US Department of Health and Human Services. CDC. 2000. National Center for Health Statistics. National Vital Statistics System. Vital Statistics of the United States, Vol. II, Mortality, Part A, for Data Years 1950–1993. Washington, DC: US Government Printing Office. Data for 1994 to 1998, data are available on the NCHS Web site at www.cdc.gov/nchs/datawh/statab/unpubd/mortabs.htm. Chahoud I, Krowke R, Schimmel A, Merker HJ, Neubert D. 1989. Reproductive toxicity and phar- macokinetics of 2,3,7,8-tetrachlorodibenzo-p-dioxin. 1. Effects of high doses on the fertility of male rats. Archives of Toxicology 63:432–439. Chahoud I, Krowke R, Bochert G, Burkle B, Neubert D. 1991. Reproductive toxicity and toxicokinet- ics of 2,3,7,8-tetrachlorodibenzo-p-dioxin. 2. Problem of paternal-mediated abnormalities in the progeny of rat. Archives of Toxicology 65(1):27–31. Charles JM, Henwood SM, Leeming NM. 1999. Developmental toxicity studies in rats and rabbits and two-generation reproduction study in rats on 4-(2,4-dichlorophenoxy)butyric acid. International Journal of Toxicology 18:177–189. Chen Z, Stewart PA, Davies S, Giller R, Krailo M, Davis M, Robison L, Shu XO. 2005. Parental occupational exposure to pesticides and childhood germ-cell tumors. American Journal of Epidemiology 162(9):858–867. Chen Z, Robison L, Giller R, Krailo M, Davis M, Davies S, Shu XO. 2006. Environmental exposure to residential pesticides, chemicals, dusts, fumes, and metals, and risk of childhood germ cell tumors. International Journal of Hygiene and Environmental Health 209(1):31–40. Chia SE, Shi LM. 2002. Review of recent epidemiological studies on paternal occupations and birth defects. Occupational and Environmental Medicine 59(3):149–155. Cordier S, Chevrier C, Robert-Gnansia E, Lorente C, Brula P, Hours M. 2004. Risk of congenital anomalies in the vicinity of municipal solid waste incinerators. Occupational and Environmental Medicine 61(1):8–15. Daniels JL, Olshan AF, Teschke K, Herz-Picciotto I, Savitz DA, Blatt J, Bondy ML, Neglia JP, Pollock BH, Cohn SL, Look AT, Seeger RC, Castleberry RP. 2001. Residential pesticide exposure and neuroblastoma. Epidemiology 12:20–27. Desaulniers D, Leingartner K, Musicki B, Cole J, Li M, Charboneau M, Tsang BK. 2004. Lack of effects of postnatal exposure to a mixture of aryl hydrocarbon-receptor agonists on the devel- opment of methylnitrosourea-induced mammary tumors in Sprague-Dawley rats. Journal of Toxicology and Environmental Health Part A 67(18):1457–1475. Dimich-Ward H, Hertzman C, Teschke K, Hershler R, Marion SA, Ostry A, Kelly S. 1996. Reproduc- tive effects of paternal exposure to chlorophenate wood preservatives in the sawmill industry. Scandinavian Journal of Work, Environment and Health 22(4):267–273. Donovan JW, MacLennan R, Adena M. 1984. Vietnam service and the risk of cogenital anomalies: A case–control study. Medical Journal of Australia 140(7):394–397.

560 VETERANS AND AGENT ORANGE: UPDATE 2006 Driscoll R, Donovan B, Esswein E, Mattorano D. 1998. Health Hazard Evaluation Report. US De- partment of Agriculture, Pp. 1–72. Erickson J, Mulinare J, Mcclain P, Fitch T, James L, McClearn A, Adams M. 1984a. Vietnam Veter- ans’ Risks for Fathering Babies with Birth Defects. Atlanta, GA: US Department of Health and Human Services, Centers for Disease Control. Erickson JD, Mulinare J, McClain PW, Fitch TG, James LM, McClearn AB, Adams MJ. 1984b. Vietnam veterans’ risks for fathering babies with birth defects. Journal of the American Medical Association 252(7):903–912. Eskenazi B, Mocarelli P, Warner M, Chee WY, Gerthoux PM, Samuels S, Needham LL, Patterson DG Jr. 2003. Maternal serum dioxin levels and birth outcomes in women of Seveso, Italy. En- vironmental Health Perspectives 111(7):947–953. Eskenazi B, Warner M, Marks AR, Samuels S, Gerthoux PM, Vercellini P, Olive DL, Needham L, Patterson D Jr, Mocarelli P. 2005. Serum dioxin concentrations and age at menopause. Environ- mental Health Perspectives 113(7):858–862. Farr SL, Cooper GS, Cai J, Savitz DA, Sandler DP. 2004. Pesticide use and menstrual cycle charac- teristics among premenopausal women in the Agricultural Health Study. American Journal of Epidemiology 160(12):1194–1204. Farr SL, Cai J, Savitz DA, Sandler DP, Hoppin JA, Cooper GS. 2006. Pesticide exposure and tim- ing of menopause: the Agricultural Health Study. American Journal of Epidemiology 163(8): 731–742. Field B, Kerr C. 1988. Reproductive behaviour and consistent patterns of abnormality in offspring of Vietnam veterans. Journal of Medical Genetics 25:819–826. Fisher MT, Nagarkatti M, Nagarkatti PS. 2005. Aryl hydrocarbon receptor-dependent induction of loss of mitochondrial membrane potential in epididydimal spermatozoa by 2,3,7,8-tetrachloro- dibenzo-p-dioxin (TCDD). Toxicology Letters 157(2):99–107. Fitzgerald EF, Weinstein AL, Youngblood LG, Standfast SJ, Melius JM. 1989. Health effects three years after potential exposure to the toxic contaminants of an electrical transformer fire. Archives of Environmental Health 44:214–221. Flower KB, Hoppin JA, Lynch CF, Blair A, Knott C, Shore DL, Sandler DP. 2004. Cancer risk and parental pesticide application in children of Agricultural Health Study participants. Environmen- tal Health Perspectives 112(5):631–635. Foster WG, Holloway AC, Hughes CL Jr. 2005. Dioxin-like activity and maternal thyroid hormone levels in second trimester maternal serum. American Journal of Obstetrics and Gynecology 193(6):1900–1907. Gao Y, Sahlberg C, Kiukkonen A, Alaluusua W, Pohjanvirta R, Tuomisto J, Lukinmaa PL. 2004. Lacta- tional exposure of Han/Wistar rats to 2,3,7,8-tetrachlorodibenzo-p-dioxin interferes with enamel maturation and retards dentin mineralization. Journal of Dental Research 83(2):139–144. García AM, Benavides FG, Fletcher T, Orts E. 1998. Paternal exposure to pesticides and congenital malformations. Scandinavian Journal of Work, Environment and Health 24(6):473–480. Garry VF, Schreinemachers D, Harkins ME, Griffith J. 1996. Pesticide appliers, biocides, and birth defects in rural Minnesota. Environmental Health Perspectives 104(4):394–399. Greenlee AR, Arbuckle TE, Chyou PH. 2003. Risk factors for female infertility in an agricultural region. Epidemiology 14(4):429–436. Hanify JA, Metcalf P, Nobbs CL, Worsley KJ. 1981. Aerial spraying of 2,4,5-T and human birth malformations: An epidemiological investigation. Science 212:349–351. Heacock H, Hogg R, Marion SA, Hershler R, Teschke K, Dimich-Ward H, Demers P, Kelly S, Ostry A, Hertzman C. 1998. Fertility among a cohort of male sawmill workers exposed to chloro- phenate fungicides. Epidemiology 9(1):56–60.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 561 Heacock H, Hertzman C, Demers PA, Gallagher R, Hogg RS, Teschke K, Hershler R, Bajdik CD, Dimich-Ward H, Marion SA, Ostry A, Kelly S. 2000. Childhood cancer in the offspring of male sawmill workers occupationally exposed to chlorophenate fungicides. Environmental Health Perspectives 108(6):499–503. Henriksen GL, Michalek JE, Swaby JA, Rahe AJ. 1996. Serum dioxin, testosterone, and gonadotro- pins in veterans of Operation Ranch Hand. Epidemiology 7(4):352–357. Hertz-Picciotto I, Samuels SJ. 1988. Incidence of early loss of pregnancy. New England Journal of Medicine 319(22):483–484. Hombach-Klonisch S, Pocar P, Kauffold J, Klonisch T. 2006. Dioxin exerts anti-estrogenic actions in a novel dioxin-responsive telomerase-immortalized epithelial cell line of the porcine oviduct (TERT-OPEC). Toxicological Sciences 90(2):519–528. Infante-Rivard C, Labuda D, Krajinovic M, Sinnett D. 1999. Risk of childhood leukemia associated with exposure to pesticides and with gene polymorphisms. Epidemiology 10:481–487. IOM (Institute of Medicine). 1994. Veterans and Agent Orange Health Effects of Herbicides Used in Vietnam. Washington DC: National Academy Press. IOM. 1996. Veterans and Agent Orange: Update 1996. Washington, DC: National Academy Press. IOM. 1999. Veterans and Agent Orange: Update 1998. Washington, DC: National Academy Press. IOM. 2001. Veterans and Agent Orange: Update 2000. Washington, DC: National Academy Press. IOM. 2002. Veterans and Agent Orange: Herbicide/Dioxin Exposure and Acute Myelogenous Leuke- mia in the Children of Vietnam Veterans. Washington, DC: The National Academies Press. IOM. 2003. Veterans and Agent Orange: Update 2002. Washington, DC: The National Academies Press. IOM. 2005. Veterans and Agent Orange: Update 2004. Washington, DC: The National Academies Press. Ishimura R, Ohsako S, Kawakami T, Sakaue M, Aoki Y, Tohyama C. 2002. Altered protein profile and possible hypoxia in the placenta of 2,3,7,8-tetrachlorodibenzo-p-dioxin-exposed rats. Toxicology and Applied Pharmacology 185(3):197–206. Ishimura R, Kawakami T, Ohsako S, Nohara K, Tohyama C. 2006. Suppressive effect of 2,3,7,8- tetrachlorodibenzo-p-dioxin on vascular remodeling that takes place in the normal labyrinth zone of rat placenta during late gestation. Toxicological Sciences 91(1):265–274. John-Greene JA, Ouellette JH, Jeffries TK, Johnson KA, Rao KS. 1985. Teratological evaluation of picloram potassium salt in rabbits. Food and Chemical Toxicology 23(8):753–756. Kallen B. 1988. Epidemiology of Human Reproduction. Boca Raton, FL: CRC Press. Kalter H, Warkany J. 1983. Congenital malformations. Etiologic factors and their role in prevention (first of two parts). New England Journal of Medicine 308:424–431. Kang HK, Mahan CM, Lee KY, Magee CA, Mather SH, Matanoski G. 2000. Pregnancy out- comes among US women Vietnam veterans. American Journal of Industrial Medicine 38(4): 447–454. Kerr M, Nasca PC, Mundt KA, Michalek AM, Baptiste MS, Mahoney MC. 2000. Parental occu- pational exposures and risk of neuroblastoma: A case–control study (United States). Cancer Causes and Control 11:635–643. Khorram O, Garthwaite M, Jones J, Golos T. 2004. Expression of aryl hydrocarbon receptor (AHR) and aryl hydrocarbon receptor nuclear translocator (ARNT) mRNA expression in human sper- matozoa. Medical Science Monitor 10(5):BR135–BR138. Kitajima M, Khan KN, Fujishita A, Masuzaki H, Ishimaru T. 2004. Histomorphometric alteration and cell-type specific modulation of arylhydrocarbon receptor and estrogen receptor expression by 2,3,7,8-tetrachlorodibenzo-p-dioxin and 17beta-estradiol in mouse experimental model of endometriosis. Reproductive Toxicology 18(6):793–801. Kline J, Stein Z, Susser M. 1989. Conception to Birth: Epidemiology of Prenatal Development. New York: Oxford University Press.

562 VETERANS AND AGENT ORANGE: UPDATE 2006 Knobil E, Neill JD, Greenwald GS, Markert CL, Pfaff DW, eds. 1994. The Physiology of Reproduc- tion. New York: Raven Press. Kristensen P, Andersen A, Irgens LM, Bye AS, Sundheim L. 1996. Cancer in offspring of parents engaged in agricultural activities in Norway: Incidence and risk factors in the farm environment. International Journal of Cancer 65(1):39–50. Kristensen P, Irgens LM, Andersen A, Bye AS, Sundheim L. 1997. Birth defects among offspring of Norwegian farmers, 1967–1991. Epidemiology 8(5):537–544. Kuroda M, Oikawa K, Ohbayashi T, Yoshida K, Yamada K, Mimura J, Matsuda Y, Fujii-Kuriyama Y, Mukai K. 2005. A dioxin sensitive gene, mammalian WAPL, is implicated in spermatogenesis. FEBS Letters 579(1):167–172. Lai KP, Wong MH, Wong CKC. 2005a. Effects of TCDD in modulating the expression of Sertoli cell secretory products and markers for cell-cell interaction. Toxicology 206(1):111–123. Lai KP, Wong MH, Wong CKC. 2005b. Inhibition of CYP450scc expression in dioxin-exposed rat Leydig cells. Journal of Endocrinology 185:519–527. Lamb JC 4th, Moore JA, Marks TA, Haseman JK. 1981. Development and viability of offspring of male mice treated with chlorinated phenoxy acids and 2,3,7,8-tetrachlorodibenzo-p-dioxin. Journal of Toxicology and Environmental Health 8(5-6):835–844. Larsen SB, Joffe M, Bonde JP. 1998. Time to pregnancy and exposure to pesticides in Danish farmers. Occupational and Environmental Medicine 55(4):278–283. Lawson CC, Schnorr TM, Whelan EA, Deddens JA, Dankovic DA, Piacitelli LA, Sweeney MH, Connally LB. 2004. Paternal occupational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin and birth outcomes of offspring: Birth weight, preterm delivery, and birth defects. Environmental Health Perspectives 112(14):1403–1408. Lemaire G, Mnif W, Pascussi J-M, Pillon A, Rabenoelina F, Fenet H, Gomez E, Casellas C, Nicolas J-C, Cavailles V, Duchesne M-J, Balaguer P. 2006. Identification of new human pregnane X receptor ligands among pesticides using a stable reporter cell system. Toxicological Sciences 91(2):501–509. Lerda D, Rizzi R. 1991. Study of reproductive function in persons occupationally exposed to 2,4- dichlorophenoxyacetic acid (2,4-D). Mutation Research 262(1):47–50. Li B, Liu H-Y, Dai L-J, Lu J-C, Yang Z-M, Huang L. 2006. The early embryo loss caused by 2,3,7,8- tetrachlorodibenzo-p-dioxin may be related to the accumulation of this compound in the uterus. Reproductive Toxicology 21(3):301–306. Lin C-M, Li C-Y, Mao I-F. 2006. Birth outcomes of infants born in areas with elevated ambient ex- posure to incinerator generated PCDD/Fs. Environment International 32(5):624–629. Loffredo CA, Silbergeld EK, Ferencz C, Zhang J. 2001. Association of transposition of the great arteries in infants with maternal exposures to herbicides and rodenticides. American Journal of Epidemiology 153(6):529–536. Lorber M, Phillips L. 2002. Infant exposure to dioxin-like compounds in breast milk. Environmental Health Perspectives 110(6):A325–A332. Mastroiacovo P, Spagnolo A, Marni E, Meazza L, Betrollini R, Segni G, Brogna-Pignatti C. 1988. Birth deffects in Seveso area after TCDD contamination. Journal of the American Medical As- sociation 259:1668–1672 (published erratum appears in JAMA 1988, 260:792). Meinert R, Schüz J, Kaletsch U, Kaatsch P, Michaelis J. 2000. Leukemia and non-Hodgkin’s lym- phoma in childhood and exposure to pesticides: Results of a register-based case–control study in Germany. American Journal of Epidemiology 151(7):639–646. Michalek JE, Albanese RA, Wolfe WH. 1998a. Project Ranch Hand II: An Epidemiologic Investiga- tion of Health Effects in Air Force Personnel Following Exposure to Herbicides—Reproductive Outcome Update. US Department of Commerce: National Technical Information Service. Report number AFRL-HE-BR-TR-1998-0073. Michalek JE, Rahe AJ, Boyle CA. 1998b. Paternal dioxin, preterm birth, intrauterine growth retarda- tion, and infant death. Epidemiology 9(2):161–167.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 563 Miyamoto K. 2004. Effects of dioxin on gene expression in female reproductive system in the rat. Environmental Sciences 11(1):47–55. Mizutani T, Yoshino M, Satake T, Nakagawa M, Ishimura R, Tohyama C, Kokame K, Kangawa K, Miyamoto K. 2004. Identification of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-inducible and -suppressive genes in the rat placenta: Induction of interferon-regulated genes with possible inhibitory roles for angiogenesis in the placenta. Endocrine Journal 51(6):569–577. Moon DG, Lee KC, Kim YW, Park HS, Cho HY, Kim JJ. 2004. Effect of TCDD on corpus caver- nosum histology and smooth muscle physiology. International Journal of Impotence Research 16(3):224–230. Moses M, Lilis R, Crow KD, Thornton J, Fischbein A, Anderson HA, Selikoff IJ. 1984. Health status of workers with past exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin in the manufacture of 2,4,5-trichlorophenoxyacetic acid: comparison of findings with and without chloracne. Ameri- can Journal of Industrial Medicine 5(3):161–182. Nakajima S, Saijo Y, Kato S, Sasaki S, Uno A, Kanagami N, Hirakawa H, Hori T, Tobiishi K, Todaka T, Nakamura Y, Yanagiya S, Sengoku Y, Iida T, Sata F, Kishi R. 2006. Effects of prenatal ex- posure to polychlorinated biphenyls and dioxins on mental and motor development in Japanese children at 6 months of age. Environmental Health Perspectives 114(5):773–778. NCI (National Cancer Institute). 2001. Surveillance, Epidemiology, and End Results (SEER) database. http://seer.cancer.gov/ScientificSystems/CanQues (Accessed March 19). Ngo AD, Taylor R, Roberts CL, Nguyen TV. 2006. Association between Agent Orange and birth defects: Systematic review and meta-analysis. International Journal of Epidemiology 35(5):1220–1230. Oh E, Lee E, Im H, Kang HS, Jung WW, Won NH, Kim EM, Sul D. 2005. Evaluation of immuno- and reproductive toxicities and association between immunotoxicological and genotoxicological parameters in waste incineration workers. Toxicology 2(1):65–80. Pearce MS, Parker L. 2000. Paternal employment in agriculture and childhood kidney cancer. Pedi- atric Hematology and Oncology 17(3):223–230. Pesatori AC, Consonni D, Tironi A, Zocchetti C, Fini A, Bertazzi PA. 1993. Cancer in a young popula- tion in a dioxin-contaminated area. International Journal of Epidemiology 22(6):1010–1013. Petrelli G, Figa-Talamanca I, Tropeano R, Tangucci M, Cini C, Aquilini S, Gasperini L, Meli P. 2000. Reproductive male-mediated risk: Spontaneous abortion among wives of pesticide applicators. European Journal of Epidemiology 16(4):391–393. Petroff BK, Mizinga KM. 2003. Pharmacokinetics of ovarian steroids in Sprague-Dawley rats after acute exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD). Reproductive Biology 3(2):131–141. Ray SS, Swanson HI. 2004. Dioxin-induced immortalization of normal human keratinocytes and silencing of p53 and p16INK4a. Journal of Biological Chemistry 279(26):27187–27193. Revich B, Aksel E, Ushakova T, Ivanova I, Zuchenko N, Lyuev N, Brodsky B, Sotsov Y. 2001. Dioxin exposure and public health in Chapaevsk, Russia. Chemosphere 43(4–7):951–966. Reynolds P, Von Behren J, Gunier RB, Goldberg DE, Harnly M, Hertz A. 2005b. Agricultural pesti- cide use and childhood cancer in California. Epidemiology 16(1):93–100. Schnorr TM, Lawson CC, Whelan EA, Dankovic DA, Deddens JA, Piacitelli LA, Reefhuis J, Sweeney MH, Connally LB, Fingerhut MA. 2001. Spontaneous abortion, sex ratio, and paternal occu- pational exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Environmental Health Perspectives 109(11):1127–1132. Schreinemachers DM. 2003. Birth malformations and other adverse perinatal outcomes in four US wheat-producing states. Environmental Health Perspectives 111(9):1259–1264. Schultz R, Suominen J, Varre T, Hakovirta H, Parvinen M, Toppari J, Pelto-Huikko M. 2003. Expres- sion of aryl hydrocarbon receptor and aryl hydrocarbon receptor nuclear translocator messenger ribonucleic acids and proteins in rat and human testis. Endocrinology 144(3):767–776.

564 VETERANS AND AGENT ORANGE: UPDATE 2006 Schwartz LS. 1998. Health Problems of Women Veterans of the Vietnam War. Doctoral dissertation, Yale University. Simanainen U, Adamsson A, Tuomisto JT, Miettinen HM, Toppari J, Tuomisto J, Viluksela M. 2004. Adult 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) exposure and effects on male re- productive organs in three differentially TCDD-susceptible rat lines. Toxicological Sciences 81(2):401–407. Smith AH, Fisher DO, Pearce N, Chapman CJ. 1982. Congenital defects and miscarriages among New Zealand 2,4,5-T sprayers. Archives of Environmental Health 37:197–200. Smith MT, McHale CM, Wiemels JL, Zhang L, Wiencke JK, Zheng S, Gunn L, Skibola CF, Ma X, Buffler PA. 2005. Molecular biomarkers for the study of childhood leukemia. Toxicology and Applied Pharmacology 206(2):237–245. Staessen JA, Nawrot T, Hond ED, Thijs L, Fagard R, Hoppenbrouwers K, Koppen G, Nelen V, Schoeters G, Vanderschueren D, Van Hecke E, Verschaeve L, Vlietinck R, Roels HA. 2001. Renal function, cytogenetic measurements, and sexual development in adolescents in relation to environmental pollutants: A feasibility study of biomarkers. Lancet 357(9269):1660–1669. [Comment in Lancet. 2001. 358(9295):1816–1817.] Stellman SD, Stellman JM, Sommer JF Jr. 1988. Health and reproductive outcomes among American Legionnaires in relation to combat and herbicide exposure in Vietnam. Environmental Research 47:150–174. Stockbauer JW, Hoffman RE, Schramm WF, Edmonds LD. 1988. Reproductive outcomes of mothers with potential exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin. American Journal of Epidemiol- ogy 128:410–419. Suskind RR, Hertzberg VS. 1984. Human health effects of 2,4,5-T and its toxic contaminants. Journal of the American Medical Association 251:2372–2380. Suzuki G, Nakano M, Nakano S. 2005. Distribution of PCDDs/PCDFs and Co-PCBs in human maternal blood, cord blood, placenta, milk, and adipose tissue: Dioxins showing high toxic equivalency factor accumulate in the placenta. Bioscience, Biotechnology and Biochemistry 69(10):1836–1847. Swan SH. 2006. Semen quality in fertile US men in relation to geographical area and pesticide expo- sure. International Journal of Andrology 29(1):62–68; discussion 105–108. Swan SH, Kruse RL, Liu F, Barr DB, Drobnis EZ, Redmon JB, Wang C, Brazil C, Overstreet JW; Study for Future Families Research Group. 2003. Semen quality in relation to biomarkers of pesticide exposure. Environmental Health Perspectives 111(12):1478–1484. Tango T, Fujita T, Tanihata T, Minowa M, Doi Y, Kato N, Kunikane S, Uchiyama I, Tanaka M, Uehata T. 2004. Risk of adverse reproductive outcomes associated with proximity to municipal solid waste incinerators with high dioxin emission levels in Japan. Journal of Epidemiology 14(3):83–93. Tas S, Lauwerys R, Lison D. 1996. Occupational hazards for the male reproductive system. Critical Reviews in Toxicology 26(3):261–307. ten Tusscher GW, Stam GA, Koppe JG. 2000. Open chemical combustions resulting in a local in- creased incidence of orofacial clefts. Chemosphere 40(9-11):1263–1270. Townsend JC, Bodner KM, Van Peenen PFD, Olson RD, Cook RR. 1982. Survey of reproductive events of wives of employees exposed to chlorinated dioxins. American Journal of Epidemiol- ogy 115:695–713. Tuyet LTN, Johansson A. 2001. Impact of chemical warfare with Agent Orange on women’s reproduc- tive lives in Vietnam: A pilot study. Reproductive Health Matters 9(18):156–164. Ushinohama K, Son D, Roby KF, Rozman KK, Terranova PF. 2001. Impaired ovulation by 2,3,7,8 tetrachlorodibenzo-p-dioxin (TCDD) in immature rats treated with equine chorionic gonadotro- pin. Reproductive Toxicology 15(3):275–280.

REPRODUCTIVE AND DEVELOPMENTAL EFFECTS 565 Volz DC, Bencic DC, Hinton DE, Law MJ, Kullman SW. 2005. 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces organ-specific differential gene expression in male Japanese medaka (Oryzias latipes). Toxicological Sciences 85(1):572–584. Wang S-L, Chang Y-C, Chao H-R, Li C-M, Li L-A, Lin L-Y, Papke O. 2006. Body burdens of poly- chlorinated dibenzo-p-dioxins, dibenzofurans, and biphenyls and their relations to estrogen metabolism in pregnant women. Environmental Health Perspectives 114(5):740–745. Warner M, Samuels S, Mocarelli P, Gerthoux PM, Needham L, Patterson DG Jr, Eskenazi B. 2004. Serum dioxin concentrations and age at menarche. Environmental Health Perspectives 112(13):1289–1292. Wen WQ, Shu XO, Steinbuch M, Severson RK, Reaman GH, Buckley JD, Robison LL. 2000. Paternal military service and risk for childhood leukemia in offspring. American Journal of Epidemiol- ogy 151(3):231–240. Wilcox AJ, Weinberg CR, O’Connor JF, Baird DD, Schlatterer JP, Canfield RE, Armstrong EG, Nisula BC. 1988. Incidence of early pregnancy loss. New England Journal of Medicine 319:189–194. Wolfe WH, Michalek JE, Miner JC, Rahe AJ, Moore CA, Needham LL, Patterson DG Jr. 1995. Paternal serum dioxin and reproductive outcomes among veterans of Operation Ranch Hand. Epidemiology 6:17–22. Wu Q, Ohsako S, Ishimura R, Suzuki JS, Tohyama C. 2004. Exposure of mouse preimplantation em- bryos to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) alters the methylation status of imprinted genes H19 and Igf2. Biological Reproduction 70(6):1790–1797. Yamano Y, Ohyama K, Ohta M, Sano T, Ritani A, Shimada J, Ashida N, Yoshida E, Ikehara K, Morishima I. 2005. A novel spermatogenesis related factor-2 (SRF-2) gene expression affected by TCDD treatment. Endocrine Journal 52(1):75–81. Yen SC, Jaffe RB. 1991. Reproductive Endocrinology. Philadelphia, PA: W.B. Saunders Company.

Next: 8 Neurologic Disorders »
Veterans and Agent Orange: Update 2006 Get This Book
×
Buy Paperback | $225.00
MyNAP members save 10% online.
Login or Register to save!
Download Free PDF

From 1962 to 1971, the U.S. military sprayed herbicides over Vietnam to strip the thick jungle canopy that could conceal opposition forces, to destroy crops that those forces might depend on, and to clear tall grasses and bushes from the perimeters of U.S. base camps and outlying fire-support bases.

In response to concerns and continuing uncertainty about the long-term health effects of the sprayed herbicides on Vietnam veterans, Veterans and Agent Orange provides a comprehensive evaluation of scientific and medical information regarding the health effects of exposure to Agent Orange and other herbicides used in Vietnam. The 2006 report is the seventh volume in this series of biennial updates. It will be of interest to policy makers and physicians in the federal government, veterans and their families, veterans' organizations, researchers, and health professionals.

  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. ×

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

    « Back Next »
  6. ×

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

    « Back Next »
  7. ×

    View our suggested citation for this chapter.

    « Back Next »
  8. ×

    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!