’ Childhood Leukemia and Primary Prevention Todd P. Whitehead, PhD,a,b Catherine Metayer, MD, PhD,a,b Joseph L. Wiemels, PhD,b,c Amanda W. Singer, PhD,a and Mark D. Miller, MD, MPHb,d Leukemia is the most common pediatric cancer, affecting 3800 children per year in the United States. Its annual incidence has increased over the last decades, especially among Latinos. Although most children diagnosed with leukemia are now cured, many suffer long-term complications, and primary prevention efforts are urgently needed. The early onset of leukemia—usually before 5 years of age—and the presence at birth of “pre-leukemic” genetic signatures indicate that pre- and postnatal events are critical to the development of the disease. In contrast to most pediatric cancers, there is a growing body of literature—in the United States and internationally—that has implicated several environmental, infectious, and dietary risk factors in the etiology of childhood leukemia, mainly for acute lymphoblastic leukemia, the most common subtype. For example, exposures to pesticides, tobacco smoke, solvents, and traffic emissions have consistently demonstrated positive associations with the risk of developing childhood leukemia. In contrast, intake of vitamins and folate supplementation during the preconception period or pregnancy, breastfeeding, and exposure to routine childhood infections have been shown to reduce the risk of childhood leukemia. Some children may be especially vulnerable to these risk factors, as demonstrated by a disproportionate burden of childhood leukemia in the Latino population of California. The evidence supporting the associations between childhood leukemia and its risk factors—including pooled analyses from around the world and systematic reviews— is strong; however, the dissemination of this knowledge to clinicians has been limited. To protect children’s health, it is prudent to initiate programs designed to alter exposure to wellestablished leukemia risk factors rather than to suspend judgment until no uncertainty remains. Primary prevention programs for childhood leukemia would also result in the significant co-benefits of reductions in other adverse health outcomes that are common in children, such as detriments to neurocognitive development. Introduction leukemia (ALL) or acute myeloblastic leukemia (AML) in the United States (U.S.).1 A small but steady annual increase from 1975 and 2012 in the ageadjusted incidence rate of childhood leukemia in the U.S. has resulted in an overall rise of 55% in the annual number of cases during the past three and a half decades. Modern treatment protocols cure 80–90% of children with leukemia with fewer sequelae than ancer is the second most common cause of death in children 0–14 years of age, after accidents. Leukemia is the most common cancer in children, representing approximately onethird of pediatric cancers. Approximately 3800 children are diagnosed annually with acute lymphoblastic C Curr Probl Pediatr Adolesc Health Care 2016;46:317-352 From the aDepartment of Epidemiology, School of Public Health, University of California, Berkeley, CA; bCenter for Integrative Research on Childhood Leukemia and the Environment, University of California, Berkeley, CA; cDepartment of Epidemiology and Biostatistics, School of Medicine, University of California, San Francisco, CA; and dWestern States Pediatric Environmental Health Specialty Unit, University of California, San Francisco, CA. This publication was supported by the cooperative agreement award number 1 U61TS000237-02 from the Agency for Toxic Substances and Disease Registry (ATSDR), United States. The U.S. Environmental Protection Agency (EPA), United States, supports the Pediatric Environmental Health Specialty Units (PEHSU) by providing partial funding to ATSDR under Inter-Agency Agreement number DW-75-95877701. This work also was supported in part by the US National Institute of Environmental Health Sciences (NIEHS), United States, (Grants P01 ES018172 and P50ES018172) and the USEPA (Grants RD83451101 and RD83615901), as part of the Center for Integrative Research on Childhood Leukemia and the Environment (CIRCLE). The California Childhood Leukemia Study was also supported in part by NIEHS, United States (Grant R01ES009137). The ideas and opinions expressed herein are those of the authors and do not necessarily represent the official views of the ATSDR, EPA, or NIEHS. Endorsement of any product or service mentioned by ATSDR, EPA, or NIEHS is not intended nor should it be inferred. Curr Probl Pediatr Adolesc Health Care 2016;46:317-352 1538-5442/$ - see front matter & 2016 The Authors. Published by Elsevier Inc. All rights reserved. This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/). http://dx.doi.org/10.1016/j.cppeds.2016.08.004 Curr Probl PediatrAdolesc Health Care, October 2016 317 FIG. Incidence of childhood leukemia in California by race-ethnicity, 1988–2012. Adapted with permission from Giddings et al.7 previous regimens. Still, even with improved treatments, the immediate and long-term consequences of childhood leukemia continue to exact a heavy toll.2,3 The impacts and costs of childhood leukemia extend beyond the care of the sick child; affecting family, friends, and the community. Long-term and late-appearing secondary effects include detriments to neurocognitive development, mental health, endocrine system function, and general health.4 To avoid these risks completely, it would be beneficial to prevent the disease altogether. Though new genetic risk factors are likely still to be discovered, to date only a small fraction (less than 10%) of childhood leukemia cases can be attributed to the influence of genetics, including to genetic syndromes such as Down’s.5,6 Moreover, the aforementioned increase in childhood leukemia incidence—which is not fully explained by diagnostic trends—indicates that causal factors for childhood leukemia have become more prevalent in the last few decades. Since genetic factors do not change on this time scale, it is probable that environmental factors play a significant role in the etiology of childhood leukemias and their recent upward trends.6 These facts underscore the importance 318 of developing an approach to primary prevention of childhood leukemia focused on reducing exposure to environmental risk factors for the disease. Children of Latino (also referred to as Hispanic) descent have a higher incidence of leukemia than whites, African-Americans or Asian-Americans nationally and in California, a highly-populated and ethnically diverse State. Moreover, over the past 25 years, childhood leukemia incidence has been rising in California at a faster pace in Latino children, compared to white children (Fig),7 suggesting that Latino children (or parents) are even more vulnerable to and/or more exposed to harmful environmental factors than others. Exposure to carcinogens has generally been considered a non-threshold event and modeled in a linear fashion. However, there are chemicals for which a supra-linear dose–response curve has been observed, indicating that the risk from low-dose exposure may actually be proportionally greater than would be expected with a linear dose–response curve.8 One such example is the known leukemogen, benzene, which is metabolized to active intermediary compounds. This finding is consistent with other exposures of concern to pediatric environmental health, such as lead, where a Curr Probl PediatrAdolesc Health Care, October 2016 non-linear dose–response relationship has resulted in worse-than-anticipated health effects from low-dose environmental exposures that were presumed to be safe.9 As such, findings from observational studies— where participants are exposed to chemicals at environmental levels as opposed to higher dose occupational levels—are critical to avoid underestimating risk. Research into environmental, infectious, and dietary causes of childhood leukemia has been limited, amounting to only a small fraction of the total Federal funding for childhood leukemia research (e.g., 3–7% of National Institutes of Health and 1% of National Cancer Institute total funding in 2010–2011).10 Funding for treatment/outcome-related studies has been far greater than for those studies examining etiology. The dramatic increases in children cured clearly justifies the expenditures made to improve treatment, but research on etiologic factors that may lead to prevention is also important. Despite limited funding, evidence demonstrating associations between environmental, infectious, and dietary exposures and the risk of developing childhood leukemia is accumulating. This issue provides an overview of key biological concepts in childhood leukemia research and summarizes the environmental, infectious, and dietary risk factors for childhood leukemia that have been identified in epidemiologic studies from around the world. Our purpose is not to conduct a systematic review of the field of childhood leukemia epidemiology—an endeavor which could fill an entire book—but rather to provide a concise perspective of the subject for the benefit of clinicians and policy makers. The literature cited in this issue spans a wide gamut. Whenever possible, the authors emphasize relevant findings from well-designed meta-analyses of published data and from large international collaborations that have pooled and analyzed individual-level data from multiple studies. In some instances, both pooled analyses and meta-analyses (including unpublished data) were conducted in parallel. Because most of these comprehensive studies fulfill criteria for systematic reviews, they provide the strongest evidence of associations between childhood leukemia and its risk factors. However, when warranted by the novelty of the research, the authors also discuss “one-off” findings of particular interest. The authors are associated with the California Childhood Leukemia Study (CCLS) and the Center for Integrative Research on Childhood Leukemia and the Environment (CIRCLE) at the University of California, Berkeley. This research group, begun by Patricia Buffler, PhD, has studied environmental causes of childhood leukemia for Curr Probl PediatrAdolesc Health Care, October 2016 25 years. In this article, we identify many of the major epidemiologic investigations of childhood leukemia worldwide and we rely on our collective expertise to highlight the literature that we believe will be most instructive to clinicians and policy makers. However, we acknowledge that particular emphasis has been paid to the work of our research group and its collaborators in the Childhood Leukemia International Consortium (CLIC). Natural History of Leukemia as a Disease Development of Blood Cells and the Formation of Leukemia Hematopoiesis is a process involving extreme cellular blast growth, which produces 1011 blood cells per day throughout life. The activity of certain cell differentiation stages (defined by cell surface markers and the potential of the progeny) varies throughout life due to different needs of the organism. These changes in the activity of blood cell types over the course of development are reflected in the distinct types of blood cells with the propensity for cancer in childhood and adulthood. Although the maternal immune system protects the child before and shortly after birth (with maternal antibodies), the child’s immune system must begin its education immediately after birth. This education of the adaptive immune system requires exposures to immune stimuli (e.g., infections) leading to the formation of billions of naïve pre-B and pre-T cells. Leukemias in children arise from these naïve, preantigen stages, typically leading to primitive precursor blast cell leukemic populations. That is, the majority of childhood leukemias are of the pre-B cell or, to a lesser extent, pre-T-cell phenotype (exhibiting cell surface markers of normal pre-B or pre-T cells) and appear as clonal outgrowths of normal pre-B or pre-T cells “frozen” at a particular stage of differentiation. Through the adaptive formation of antibodies and T-cell receptors that respond to specific antigens, a stable long-lived repertoire of cells is developed by the time a child reaches young adulthood. The pre-B and pre-T stages decrease in frequency after adolescence, exemplified by the involution of the thymus and the reduction in size of the pre-B cell population in the bone marrow. As a result, adults typically contract hematopoietic cancers (lymphomas and leukemias) in post-antigen-stimulated B and T-cells and the myeloid 319 lineage predominates in adult tumors. Distinct from common translocations for ALL and AML, respecadult leukemias, the unique properties of childhood tively. Studies using cord blood from healthy children leukemia cells are likely to be important for underwithout cancer indicate that these translocations may standing the environmental sensitivities for leukemooccur at a rate of 1% or more in the general genesis in this age group. population,20,21 while the disease is much more rare. Childhood leukemia, like all cancers, is a product of two or more molecular changes in a stem-like cell that Mutational Mechanisms in Leukemia has the ability to divide while maintaining an immature state. Because they are formed from blood cells, Recent investigations into the precise molecular leukemias have an inherent capacity for mobilization attributes of leukemia point largely to three pathways: in the bloodstream and extravasation. Precursor blood (1) aberrations in a small number of lineage-specific cells also have an enormous capacity for “blast-like” transcription factors such as ETV6, RUNX1, IKZF1, and growth, and an ability to travel throughout the body PAX5 (2) defects in receptor protein tyrosine kinases without restriction. These attributes are among the six and their downstream pathways (i.e., RAS/MEK/ERK), 11 “hallmarks of cancer,” and the fact that hematopoiand (3) epigenetic modifiers. Mutations may involve translocations that comprise fusion genes, copy number etic precursors harbor these “cancer-like” attributes alterations (most common are deletions), single nucleomay explain why leukemias seem to need far fewer tide mutations, and broad changes in epigenetic features genetic aberrations for tumor progression compared to solid tumors, which need to evolve additional metasuch as DNA methylation aberrations. The molecular formation of the initiating transstatic capacities via genetic locations may provide some mutations. This simplicity may also help explain the short The development of childhood clues as to their causes. In the past several years, many translatency of childhood cancers. The genetic simplicity of leu- leukemia is a multi-step process; location breakpoints were shown kemia combined with the young initiation occurs in utero for most to be induced by enzymatic age of onset has allowed leukemias, whereas progression mutagenic processes—normally researchers to delineate—within into acute disease usually takes utilized by the immune system to diversify antibody and T-cell the lifetime of the child—the place after birth. repertoire—which were abertiming of the formation of charrantly targeted to oncogenes acteristic genetic aberrations.12 and genomic enhancers.22 These types of translocations These changes appear to occur within two time frames —prenatal initiating events that induce some cellular are common in hematopoietic cancers that form after changes, and postnatal genetic and epigenetic events birth in response to antigenic stimulation. Interestingly, that allow the emergence of acute disease. This leukemia translocations including the most common in research was made possible by the availability of children such as ETV6-RUNX1 do not show signs of archived newborn blood spots that are routinely such activity and their origin remains a mystery. The collected at birth by heel prick from neonates (e.g., lack of a clear endogenous path toward translocation by the State of California Newborn Screening Proformation points to exogenous causes such as in utero gram), including from children with leukemia. Several environmental exposures, and points again to the translocations commonly found in leukemias that were oncologic immaturity of childhood leukemia blasts. assessed using archived newborn blood spots, includEnvironmental causes may also interact with endogeing t(12;21) ETV6-RUNX1, t(8;21)RUNX1-MTG8, inv nous mutagenic mechanisms in children after birth. (16)CBFB-MYH11, have indicated a clear presence of Many of these postnatal secondary rearrangements the mutations in neonatal blood at birth in children who show the clear involvement of the activity of the contract leukemia later in life.13–16 Several other enzymes that create antibody diversity, that is, recombinase activating gene (RAG) and adenosine deaminase mutations, including t(1;19)TCF3-PBX1, FLT3, and 17–19 (AID) in the formation of secondary deletions and RAS mutations clearly occur postnatally. The mutations associated with leukemia are generally mutations.23 Illegitimate targeting of these endogenous enzymes has long been known to be stimulated by insufficient to cause disease by themselves. This is the exposures as diverse as pesticides and smoking. As case for ETV6-RUNX1 and RUNX1-MTG8, the most 320 Curr Probl PediatrAdolesc Health Care, October 2016 such, there is evidence that postnatal environmental exposures may initiate childhood leukemia via the stimulation of endogenous mutagenic mechanisms including recombinase activating gene activity.24,25 Apart from the chromosome breakage events mentioned above, additional mutations include point mutations and epigenetic modifications. Point mutations are caused by electrophilic chemicals that are either exposed to bone marrow cells directly or else they are metabolically activated, and their metabolites are exposed to bone marrow cells. Many oxidized reactive metabolites are produced from chemicals circulating in the bone marrow due to the presence of highly metabolically active cells such as neutrophils (containing myeloperoxidase, for instance) and the high levels of heme iron. Metabolites that adduct to DNA can directly cause miscoding mutations if unrepaired, or can result in repair-induced strand breaks. There is little current evidence that point mutations such as those found in RAS are caused by environmental chemicals26; however, the most common translocation in ALL, ETV6-RUNX1, appears to be strongly and specifically associated with parental smoking27 and home paint use,28,29 which provides some credence to viability of this pathway. Epigenetic modifications are a normal part of a blood cell’s development from a stem cell to a mature cell. Changes to the usual epigenetic programming can occur, however, as cells adapt to new environments. This type of adaptation is the essence of the “developmental origins of health and disease” initiatives that seek to understand when such adaptations early in life can lead to later disease risk, such as the demonstration that starvation in utero can lead to obesity and cardiac disease risk later in life.30 Adaptations can occur as the result of exposures to specific environmental factors—for instance the specific demethylation of CpG loci in the gene AHRR has been exquisitely related to cigarette smoke.31 Likewise, methylation changes have also been observed in response to folic acid exposures.32 Our understanding of the specific effects of environmental exposures on epigenetic features is just in its infancy, and it is unknown whether these perturbations could impact leukemogenesis. The Role of Immune Factors in a Cancer of the Immune System Infection is a direct cause of viral-induced cancers, such as human papillomavirus-induced cervical cancer Curr Probl PediatrAdolesc Health Care, October 2016 and Merkel cell virus-induced skin cancer. Inflammation from infection is also a risk factor for many cancers, resulting from the collateral damage of tissue disorganization, tissue remodeling, and chronic exposure to reactive oxygen species following inflammatory processes. Childhood ALL is also a disease of the immune system, which begs the question can infection directly stimulate leukemia or stimulate leukemia via inflammatory processes? Viruses that directly integrate into the genome have not been reported in childhood ALL. Epidemiological studies have, however, demonstrated clear effects of immune factors on leukemogenesis, most overtly in the form of a consistently observed reduced risk of childhood ALL associated with more childhood contacts in daycare [odds ratio (OR) ¼ 0.77; 95% confidence interval (95% CI): 0.71─0.84; N ¼ 7399 cases] and other markers of early exposure to immune stimulus such as breastfeeding for at least 6 months (OR ¼ 0.86, 95% CI: 0.79─0.94), having an older sibling (OR ¼ 0.94, 95% CI: 0.88─1.00), or a history of four or more common infections in the first year of life (OR ¼ 0.88, 95% CI: 0.79─0.98).33 Just as regular immune stimulation appears to reduce risk for allergies and asthma, the same immune exercise can reduce the risk of leukemia. In the absence of these priming exposures, children may respond too strongly to the myriad of infections subsequently encountered in school, resulting in a cytokine “storm” and excess cell stimulation, secondary mutations, and in some cases, leukemia. This putative pathology is referred to as the “Greaves hypotheses” after its first proponent.34 Other ancillary data on immune education seem to fit this pattern as well—normal childhood vaccines, the presence of older siblings, and breastfeeding were all associated with reduced risks of childhood leukemia. Population mixing, in which large numbers of a “new” population are mixed into a standing community, seems to transiently increase the risk of leukemia and also fit with the notion of an infectious stimulation for childhood ALL.35 A corollary hypothesis to Greaves' was suggested by Kinlen who noticed that leukemia space– time clusters often followed recent population mixing events, such as the creation of new towns or population movements during warfare. He proposed that such mixing facilitated the transmission of a specific leukemia-initiating virus which, while plausible, was not followed up with biological validation as noted above. More likely, population mixing permits the transmission of common viruses to populations 321 without herd immunity rather than spreading specific leukemia-initiating viruses. Recent studies have noted that the child’s medical records confuse the infection issue somewhat—ALL patients visit their general practitioner for infections in the first year of life much more commonly than children who do not grow up to get leukemia.36,37 This suggests that the damage wrought by fulminant infections may occur much earlier than previously thought, and also that children who get leukemia may respond to infection differently, that is, more strongly to typical childhood infections. Part of this sensitivity may be a lack of immunomodulation from suppressive cytokines like IL-10 that tend to be present at lower levels in newborns who go on to develop leukemia.38 Chemicals as Leukemogens The ability of a chemical to act as carcinogen was originally thought to hinge on its capacity as mutagen, but it is likely that many other more biologically relevant activities are important in leukemogenesis as well. Some chemicals exhibit properties that allow them to target the bone marrow specifically, due to its metabolic activity (benzene for an example). Other chemicals may impact the immune system indirectly, setting up the individual for aberrant responses to infection. The role of exogenous factors such as chemicals, many of which are immunosuppressive, in this process is unknown and likely to be a major research field in the future. Other activities of chemicals are summarized in a recent review of chemicals as carcinogens.39 Below, we discuss chemical exposures that may cause leukemia and weigh the evidence for causal relationships. Environmental Risk Factors for Childhood Leukemia Exposure Science in Studies of Childhood Leukemia Risk As mentioned earlier, childhood leukemia is the most common form of pediatric cancer; but, for the purposes of epidemiological study, it is a rare disease. As such, the vast majority of epidemiological investigations into the causes of childhood leukemia are forced to employ a case–control study design. In 322 many instances, case–control studies of childhood leukemia are designed to assess children’s exposure to disease risk factors retrospectively. That is, a child is first diagnosed with leukemia, then s/he is enrolled in the case–control study, and only afterwards can investigators begin to assess the agents to which s/he has been exposed. This design imposes limitations on the epidemiologist, as etiologically relevant specimens—biological and environmental—are not necessarily available for collection by the time the child is under study. As discussed above, childhood leukemia can be initiated during the prenatal period, but most cases are not diagnosed until the child is between 2 and 4 years old. This leaves a long time interval between the first windows of susceptibility to leukemogenic agents and the time period when investigators can start measuring a child’s exposure to those agents. To overcome these challenges, childhood leukemia investigators have employed a variety of strategies to assess children’s exposures to potentially carcinogenic agents. Using Parent Interviews to Assess Children’s Exposures to Chemicals One simple way to circumvent the need for the collection of biological or environmental samples during etiologically relevant time periods is to interview participating parents to obtain information about their child’s history of exposure to specific agents. Such interviews can be used to pinpoint historical exposures during critical windows of a child’s development (e.g., the second trimester of pregnancy), they can be wide-ranging in scope, and they can be especially effective in assessing parents' conscious behaviors (e.g., smoking habits, residential pesticide use, occupational histories). On the other hand, interview-based exposure assessments have inherent limitations; they are imprecise measures of chemical exposure, they provide little information about any of a child’s exposures that go unrecognized by the parents, and they are potentially subject to recall and reporting biases if parents of case and control children remember or report their children’s exposures in different ways. In practice, though, reproducibility studies have suggested that, for some of the environmental exposures that are of interest in childhood leukemia research— ionizing radiation,40–42 pesticides,43 and smoking27— there is minimal evidence of differential recall between cases and controls. Curr Probl PediatrAdolesc Health Care, October 2016 Measuring Chemicals in Settled Dust research from the Center for Integrative Research on Another strategy to obtain information about a Childhood Leukemia and the Environment also indicates child’s exposure to chemicals during etiologically a relationship between levels of persistent chemicals in relevant time periods is to measure levels of chemicals matched samples of settled dust and biospecimens.53,54 in stable environmental matrices. For example, persisTaken together, the observation that chemical levels can be tent chemicals accumulate on settled dust particles, correlated in matched samples of settled dust and human which can become trapped deep within a carpet, and serum as well as the observation that chemical levels in this settled dust acts as a long-term reservoir for these settled dust are relatively stable over time support the use chemicals.44 With limited exposure to direct sunlight of chemical levels measured in dust collected after diagnosis as surrogates for chemical exposures that and microbial action, persistent chemicals, like polychildren received during etiologically relevant time periods chlorinated biphenyls (PCBs), are extremely slow to (before diagnosis). degrade on dust particles that settle indoors. As a result, collecting samples of settled dust from carpets or other household surfaces and measuring levels of persistent chemicals in these samples provides an Estimating Ambient Environmental Exposures integrated measure of chemical contamination over a Using Geographic Information Systems long period of time. An alternative strategy for assessing a child’s history of As part of the California Childhood Leukemia Study, we environmental exposures is to estimate ambient pollution have collected multiple dust samples from a large group of using Geographic Information Systems (GIS) and geohomes at time intervals of several years between sampling spatial modeling and to use these estimates of ambient rounds. We found that while there was substantial conditions as surrogates for the child’s total exposure to variability in chemical levels between sampling rounds, chemicals. Many governing bodies record a child’s home there was also moderate correlation in the relative ranking address on the birth certificate and this information can be of exposures (i.e., rankings from highest to lowest obtained for research purposes contingent on appropriate exposures) among homes over time.45–47 These findings ethical review and approval. Exposure models based on support the hypothesis that chemical levels in dust samples GIS data and geocoded birth addresses could provide collected after diagnosis may be informative surrogates for useful information about childchemical contamination that was ren’s exposure to chemicals at present in the home during important developmental periods of a Chemicals measured in samples the time of birth and, potentially, child’s life. of settled dust collected from a throughout the prenatal period as well (if participating mothers did Accidental ingestion of settled child’s home after diagnosis are not change residence during dust is an important route of human exposure to chemicals useful surrogates for the chemical pregnancy). Moreover, complete along with the consumption of exposures s/he received during residential histories can be obtained from parents via intercontaminated food and the inhalaetiologically-relevant periods view, which allows for a more tion of contaminated air. For exambefore and after birth. comprehensive model of a ple, it has been suggested that dust child’s historic exposures to ingestion is the major route of ambient chemicals. Agricultural pesticide applicaexposure to the flame retardant chemicals, polybrominated tion,55,56 traffic-related air pollution,57 and electromagdiphenyl ethers (PBDEs) in North America48 and positive netic fields58 have been estimated using GIS in the relationships have been observed between PBDE levels in context of epidemiological studies of childhood leukematched samples of dust and serum among U.S. 49,50 mia. One limitation of using estimates of ambient adults. Due to their tendency to make hand-to-mouth pollution as surrogates for total exposures is the inability contact and their proximity to the floor, young children are to account for chemical exposures that occur indoors. expected to receive a relatively large proportion of their 51 This is a substantial drawback, because children tend to total chemical intake via settled dust compared to adults, spend the vast majority of their time indoors, there are and a positive relationship has been observed between distinct chemical sources indoors, and chemical expoPBDE levels in matched samples of dust and serum in one sures tend to be higher indoors than outdoors.59,60 investigation of toddlers from North Carolina.52 Likewise, Curr Probl PediatrAdolesc Health Care, October 2016 323 Measuring Chemicals in Archived Pre-diagnostic Biospecimens Using these strategies for assessing children’s exposure to environmental agents, epidemiologists have identified several suspected environmental risk factors for childhood leukemia, including pesticides, parental smoking, paint, petroleum solvents, traffic emissions, persistent organic pollutants, and radiation, as summarized in Table 1. Perhaps the most straightforward way to obtain prediagnostic biospecimens would be to establish a prospective birth cohort and follow leukemia incidence in the participating children into adulthood. However, as discussed above, this study design is generally not feasible for childhood leukemia and will generally Pesticides be limited by a small number of available cases. Several studies have suggested that home pesticide The International Childhood Cancer Cohort Consorexposure before birth and during a child’s early years tium seeks to combine a number of large infant/ may increase the risk of childhood leukemia. Indeed, child prospective studies (on the order of 100,000 exposure to pesticides is one of the participants per study)— most frequently investigated chemiwhich were originally cal risk factors for childhood leukedesigned to examine enviEpidemiological studies have mia. A causal link between exposure ronmental and genetic identified several environmental to pesticides and childhood leukemia determinants of common risk factors for childhood is supported by many studies, includchildhood diseases—for ing the California Childhood Leukepooled analyses of childleukemia and those findings hood leukemia and other have been confirmed with large mia Study, which demonstrated a relationship between exposure to childhood cancers.61 meta-analyses and pooled insecticides—as a general class— Alternatively, it may be analyses that combined data and childhood leukemia.67 Existing possible to utilize archived from thousands of children with studies have generally used interbiospecimens—collected before diagnosis as part of leukemia and healthy controls. views with parents to assess children’s exposure to pesticides; as such, routine medical testing—to 62 no specific pesticide, or class of pesticides, has been measure prenatal chemical exposures. For example, in implicated as the causal agent underlying these many countries, blood spots are collected from each observations. newborn infant by heel-stick for the purposes of genetic screening. In the State of California, blood spots left over Pooled Analyses of Home Pesticide Use and from genetic screening are archived in a State laboratory Childhood Leukemia and made available for research contingent upon approInvestigators from the Childhood Leukemia Internapriate ethical review and approval. These archived tional Consortium pooled individual parents' responses neonatal blood spots provide a useful resource for to interview questions about children’s exposure to childhood leukemia research, offering insight into the pesticides from 12 case–control studies.29 Each conchemicals to which the developing fetus was exposed tributing study used a unique set of questions, often during the prenatal period, including folate,63 cotinine,64 65 written in different languages, so exposure data were and PBDEs. Using current technology for exposure harmonized into compatible formats before pooled biology, investigators can characterize prenatal exposure analyses could be conducted using multivariable logisto thousands of different chemicals with as little as a few tic regression. ALL was associated with any pesticide drops of archived neonatal blood. Some important exposure shortly before conception (OR ¼ 1.39; 95% technical considerations include the potential instability CI: 1.25–1.55; N ¼ 2785 cases and 3635 controls), of chemical analytes during long-term storage, the during pregnancy (OR ¼ 1.43; 95% CI: 1.32–1.54; possibility of chemical contamination during storage, N ¼ 5055 cases and 7370 controls), and after birth (OR and the complex dynamics of newborn metabolite levels 66 ¼ 1.36; 95% CI: 1.23–1.51; N ¼ 4162 cases and 5179 immediately after birth. Despite these limitations, the controls). Corresponding odds ratios for risk of AML use of archived pre-diagnostic biospecimens for expowere 1.49 (95% CI: 1.02─2.16, N ¼ 173 cases and sure assessment in epidemiological studies of childhood 1789 controls), 1.55 (95% CI: 1.21–1.99; N ¼ 344 leukemia is very promising. 324 Curr Probl PediatrAdolesc Health Care, October 2016 TABLE 1. Environmental risk factors for childhood leukemia Childhood leukemia risk factor Exposure measure Level of consensus Subtype specificity Exposure details Odds ratio (95% CI) Home pesticide use Parental interview Pooled analysis of 12 studies from CLIC.29 ALL ALL ALL AML AML AML Before conception During pregnancy After birth Before conception During pregnancy After birth 1.39 1.43 1.36 1.49 1.55 1.08 Parental occupational exposure to pesticides Parental interview Pooled analysis of 13 studies from CLIC & meta-analysis of CLIC þ non-CLIC studies.74 ALL Maternal during pregnancy Paternal at conception Paternal at conception Paternal at conception Maternal during pregnancy Paternal at conception 1.01 (0.78, 1.30) ALL T-cell ALL ALL after 5y AML AML Proximity to agricultural GIS Model pesticide application Novel finding.55 ALL ALL ALL ALL ALL Chlorthal, an herbicide Settled dust levels Novel finding.80 ALL ALL ALL Parental smoking Parental interview Pooled analysis of 14 studies from CLIC & meta-analysis of CLIC þ non-CLIC studies.83 AML AML AMMoL Home paint use Parental interview Pooled analysis of 8 studies from CLIC.86 ALL ALL ALL ALL ALL—t(12;21) Parental occupational exposure to paint Parental interview Pooled analysis of 13 studies from CLIC.89 ALL ALL AML AML Parental occupational exposure to solvents Parental interview Meta-analysis.88 ALL Meta-analysis.88,90 ALL ALL ALL Curr Probl PediatrAdolesc Health Care, October 2016 Insecticides, moderate, lifetime Fumigants, moderate, lifetime Organophosphates, moderate, lifetime Chlorinated phenols, moderate, lifetime Triazines, moderate, lifetime 1.20 1.42 1.38 1.94 (1.25, (1.32, (1.23, (1.02, (1.21, (0.76, (1.06, (1.04, (1.13, (1.19, 1.55) 1.54) 1.51) 2.16) 1.99) 1.53) 1.38) 1.94) 1.67) 3.18) 0.91 (0.66, 1.24) 1.5 (0.9, 2.4) 1.7 (1.0, 3.1) 1.6 (1.0, 2.7) 2.0 (1.0, 3.8) 1.9 (1.0, 3.7) Detected, 1st Tertile vs. 1.49 (0.82, 2.72) Not Detected Detected, 2nd Tertile vs. 1.49 (0.83, 2.67) Not Detected Detected, 3rd Tertile vs. 1.57 (0.90, 2.73) Not Detected Paternal at any time Maternal during pregnancy, Latinas Paternal at any time 1.34 (1.11, 1.62) 2.08 (1.20, 3.61) 1–3 months before conception During pregnancy After birth Professional painter, during pregnancy During pregnancy 1.54 (1.28, 1.85) 1.87 (1.08, 3.25) 1.14 (1.04, 1.25) 1.22 (1.07, 1.39) 1.66 (1.21, 2.28) 1.51 (1.08, 2.11) Paternal at conception Maternal during pregnancy Paternal at conception Maternal during pregnancy Maternal during pregnancy 0.93 (0.76, 1.14) 0.81 (0.39, 1.68) All solvents, maternal during pregnancy Petroleum, maternal during pregnancy 1.25 (1.09, 1.45) 0.96 (0.65, 1.41) 1.31 (0.38, 4.47) 1.23 (1.02, 1.47) 1.42 (1.10, 1.84) 1.71 (0.91, 3.24)a 325 TABLE 1. (Continued ) Childhood leukemia risk factor Exposure measure Level of consensus Replicated.91 Subtype specificity ALL ALL Traffic-related air pollution GIS model Two independent metaanalyses.92,93 ALL Any ALL Exposure details Odds ratio (95% CI) Benzene, maternal during pregnancy Org. solvents, paternal 1.48 (1.01, 2.16) at conception Cl-hydrocarbon, paternal 2.28 (0.97, 5.37) at conception 1.25 (0.92, 1.69) 1.53 (1.12, 2.10) 1.21 (1.04, 1.41) AML Traffic density Traffic density Nitrogen dioxide estimates Benzene estimates 2.28 (1.09, 4.75) PAHs Settled dust levels, vacuum cleaners Novel finding.94 ALL ALL ALL ALL ALL Benzo[a]pyrene Dibenzo[a,h]anthracene Benzo[k]fluoranthene Indeno[1,2,3-c,d]pyrene PAH toxic equivalence 1.42 1.98 1.71 1.81 2.35 PCBs Settled dust levels Novel finding.81 ALL Any PCB detected vs. none detected Top quartile Σ6PCB vs. bottom quartile 1.97 (1.22, 3.17) ALL PBDEs Settled dust levels Novel finding.95 ALL ALL ALL ALL ALL ALL ALL Top quartile ΣPentaBDEs vs. bottom Top quartile ΣOcta-BDEs vs. bottom Top quartile ΣDecaBDEs vs. bottom BDE-196 concentrations BDE-203 concentrations BDE-206 concentrations BDE-207 concentrations (0.95, (1.11, (0.91, (1.04, (1.18, 2.12) 3.55) 3.22) 3.16) 4.69) 2.78 (1.41, 5.48) 0.7 (0.4, 1.3) 1.3 (0.7, 2.3) 1.0 (0.6, 1.8) 2.1 2.0 2.1 2.0 (1.1, 3.8) (1.1, 3.6) (1.1, 3.9) (1.03, 3.8) ALL ¼ acute lymphoblastic leukemia; AML ¼ acute myeloblastic leukemia; AMMoL ¼ acute myelomonocytic leukemia; CI ¼ confidence interval; CLIC ¼ Childhood Leukemia International Consortium; GIS ¼ geographic information systems; PAHs ¼ polycyclic aromatic hydrocarbons; PCBs ¼ polychlorinated biphenyls; PBDEs ¼ polybrominated diphenyl ethers. a Relative risk reported. cases and 4666 controls) and 1.08 (95% CI: 0.76–1.53, N ¼ 198 cases and 2655 controls), respectively. The Childhood Leukemia International Consortium investigators29 confirmed the observed association between home pesticide exposure during pregnancy and childhood leukemia using meta-analyses as well. Other investigators have reported similar findings in independent meta-analyses,68–70 generally observing the strongest associations for indoor insecticide use. Pooled Analyses of Parental Occupational Exposure to Pesticides and Childhood Leukemia Given the consistently observed association between home pesticide use during early childhood and 326 leukemia risk, a logical extension of this line of research has been to examine the effect of parental occupational exposure to pesticides on childhood leukemia risk. There is evidence that adults exposed to pesticides at work can track these chemicals back to their homes on their shoes, clothing, and skin, potentially exposing their families.71,72 Moreover, paternal exposure to pesticides before conception could result in germ cell damage, whereas maternal exposure to pesticides during pregnancy could also expose the fetus to these chemicals.73 The Childhood Leukemia International Consortium investigators pooled individual parents' responses to interview questions about job histories and the data were harmonized to a compatible format that characterized parents' pesticide exposures at work.74 ALL was associated with paternal exposure Curr Probl PediatrAdolesc Health Care, October 2016 to pesticides at work around the time of conception (OR ¼ 1.20; 95% CI: 1.06─1.38; N ¼ 8169 fathers of cases and 14,201 fathers of controls), but was not associated with maternal exposure during pregnancy (OR ¼ 1.01; 95% CI: 0.78─1.30; N ¼ 8236 case and 14,850 control mothers). In contrast, AML was associated with maternal exposure to pesticides at work during pregnancy (OR ¼ 1.94; 95% CI: 1.19─3.18; N ¼ 1329 case and 12,141 control mothers), but was not associated with paternal exposure around the time of conception (OR ¼ 0.91; 95% CI: 0.66─1.24; N ¼ 1231 case and 11,383 control fathers). The modest association between paternal exposure to pesticides around the time of conception and ALL risk in the offspring was more evident in children diagnosed at an older age (5þ years old) and in children with the T-cell ALL subtype. The Childhood Leukemia International Consortium’s findings of a significant association between maternal exposure to pesticides during pregnancy and AML risk in the offspring is consistent with previous reports.75–77 In a meta-analysis that accompanied the abovereferenced pooled analysis, the Childhood Leukemia International Consortium investigators74 found a positive association between maternal occupational exposures to pesticides during pregnancy and AML as well as between paternal occupational exposures around conception and T-cell ALL. This meta-analysis followed a systematic review and it included studies participating in the Childhood Leukemia International Consortium as well as nonparticipating, independent studies. Other investigators78,79 have reported similar findings in independent meta-analyses, observing an association between prenatal maternal exposure to pesticides at work and childhood leukemia. GIS-Estimated Ambient Pesticide Levels and Childhood Leukemia Investigators from the California Childhood Leukemia Study have also evaluated the association between residential proximity to agricultural pesticide applications and childhood ALL.55 For the families of 213 ALL cases and 268 matched controls, the authors linked residential histories together with agricultural pesticide use reports from the California Department of Pesticide Regulation, to assess whether living within a half-mile (0.8 km) of pesticide applications was associated with childhood leukemia risk. Elevated ALL Curr Probl PediatrAdolesc Health Care, October 2016 risk was associated with lifetime moderate exposure, but not high exposure, to certain physicochemical categories of pesticides, including organophosphates, chlorinated phenols, and triazines, and with pesticides classified as insecticides or fumigants. Exposure to Herbicides and Childhood Leukemia Epidemiological studies of childhood leukemia that use environmental or biological samples are relatively scarce. In one such study, investigators from the California Childhood Leukemia Study evaluated the relationship between childhood ALL and herbicide concentrations in settled dust as surrogates of herbicide exposures.80 The herbicide analysis included 269 ALL cases 0–7 years of age and 333 healthy controls matched on date of birth, sex, and race/ethnicity. Dust samples were collected from carpets using a high-volume small-surface sampler or from participant vacuum cleaners. Concentrations of agricultural or professional herbicides (alachlor, metolachlor, bromoxynil, bromoxynil octanoate, pebulate, butylate, prometryn, simazine, ethalfluralin, and pendimethalin) and residential herbicides (cyanazine, trifluralin, 2-methyl-4-chlorophenoxyacetic acid, mecoprop, 2,4-dichlorophenoxyacetic acid, chlorthal, and dicamba) were used in logistic regression adjusting for age, sex, race/ethnicity, household income, year and season of dust sampling, neighborhood type, and residence type. The risk of childhood ALL was associated with dust levels of chlorthal; compared to homes with a measurement below the analytical limit of detection, odds ratios for the first, second, and third tertiles were 1.49 (95% CI: 0.82─2.72), 1.49 (95% CI: 0.83─2.67), and 1.57 (95% CI: 0.90─2.73), respectively (p value for linear trend ¼ 0.05). No other herbicides were identified as risk factors of childhood ALL. Metayer et al.80 postulated that 2,3,7,8-tetrachlorodibenzo-p-dioxin —a potent carcinogen and an impurity found in chlorthal—might be the causal agent underlying the observed association. Limitations of Existing Research on Pesticides and Childhood Leukemia Previous studies of the relationship between pesticide exposure and childhood leukemia, including pooled analyses conducted by the Childhood Leukemia International Consortium have utilized interviews to assess pesticide exposures to children and their parents. 327 Unfortunately, this study design has precluded investigators from identifying specific chemicals that may be causal agents underlying the observed associations. Findings from the California Childhood Leukemia Study seem to rule out organochlorine pesticides, such as DDT and chlordane, as the culpable pesticides underlying the observed association with childhood leukemia.81 Summary of Existing Research on Pesticides and Childhood Leukemia Pooled analyses of data from studies around the world show a relationship between home pesticide use—especially of insecticides indoors—and the risk of childhood leukemia, which is confirmed by independent systematic reviews and meta-analyses. Pooled analyses of data from the Childhood Leukemia International Consortium demonstrate a relationship between prenatal maternal exposure to pesticides at work and the risk of childhood AML. Likewise, these findings were supported by independent systematic reviews and meta-analyses. Future studies will continue to exam relationships between pesticide exposures and the risk of specific leukemia subtypes and will also identify the specific pesticides which act as causal agents. Parental Smoking Parental tobacco use is another suspected risk factor for childhood leukemia that has received a lot of attention from researchers. Cigarettes contain numerous harmful constituents and tobacco use is well known to cause a variety of cancers in adults, including leukemia, via both direct and secondhand means of exposure. Likewise, there is evidence that parental cigarette smoking may also be associated with childhood cancer risk. Investigators from the California Childhood Leukemia Study, for example, examined the association between parental smoking and childhood leukemia among 281 ALL cases, 46 AML cases, and 416 controls matched on age, sex, maternal race, and Latino ethnicity.82 Maternal smoking was not associated with an increased risk of either ALL or AML. Paternal preconception smoking was significantly associated with an increased risk of AML (OR ¼ 3.84, 95% CI: 1.04─14.17) and marginally associated with an increased risk of ALL (OR ¼ 1.32, 95% CI: 0.86─2.04). 328 Pooled Analyses of Parental Cigarette Smoking and Childhood AML The Childhood Leukemia International Consortium pooled individual parents' responses to interview questions about tobacco use from 14 case–control studies, representing 1300 AML and 15,000 controls.83 Individual studies ascertained information about parental cigarette smoking at a number of stages of the child’s development with varying degrees of specificity, including maternal smoking during pregnancy and paternal smoking during the three months before conception. The findings from the pooled analyses strengthened the existing evidence of modest associations between paternal cigarette smoking at any time and childhood AML, with dose–response relationships (p o 0.05). Maternal smoking during pregnancy was associated with an increased risk of AML for Latino children only. Meta-analyses of Parental Cigarette Smoking and Childhood ALL In 2009, a review of studies which evaluated the association between parental smoking and childhood leukemia revealed that 6 of 13 studies which had examined the relationship between paternal smoking and childhood leukemia reported significant positive associations.84 Subsequently, Liu et al.85 conducted a meta-analysis, which suggested that childhood ALL was associated with paternal smoking during preconception (OR ¼ 1.25, 95% CI: 1.08─1.46) during pregnancy (OR ¼ 1.24, 95% CI: 1.07─1.43), and after birth (OR ¼ 1.24, 95% CI: 0.96─1.60), with a dose– response relationships observed between childhood ALL and paternal smoking before conception or after birth. Parental Cigarette Smoking and Childhood Leukemia Subtypes There is some evidence that the strength of the association between parental cigarette smoking and childhood leukemia varies by the cytogenetic subtype of the tumor. For example, Metayer and colleagues27 reported that children with a history of paternal prenatal smoking combined with postnatal passive smoking had a 1.5-fold increased risk of ALL (95% CI: 1.01─2.23), compared to those without smoking history; but this joint effect was seen for B-cell precursor ALL with t (12;21) only (OR ¼ 2.08, 95% CI: 1.04─4.16), not for high hyperdiploid B-cell ALL. Similarly, the Curr Probl PediatrAdolesc Health Care, October 2016 aforementioned pooled AML analysis conducted by the Childhood Leukemia International Consortium found that the highest smoking-related risk was seen for the myelomonocytic leukemia, a subtype common in treatment-related AML. Childhood leukemia comprises many subtypes and these findings demonstrate that each subtype may have a distinct set of characteristic risk factors corresponding to its unique etiology. As such, studies that evaluate subtype-specific chemical risk factors are the most likely to identify true relationships. Tellingly, when risk factors for each leukemia subtype are considered separately, higher odds ratios tend to be revealed. Limitations of Existing Research on Tobacco Use and Childhood Leukemia As was the case for pesticides above, previous studies of the relationship between parental tobacco use and childhood leukemia, including pooled analyses conducted by the Childhood Leukemia International Consortium, have utilized interviews to characterize parental smoking histories. This method of exposure assessment is useful, because it allows investigators to examine the effect of parental smoking at critical windows of a child’s development and because it enables investigators to untangle the separate effects of cigarette smoking done by the mother, father, or other family members. Moreover, in contrast to other environmental exposures that are of interest to leukemia researchers, parents are conscious of the number of cigarettes they tend to smoke each day and they can help quantify their own exposures to tobacco. However, recall and reporting biases are still concerns, as parents (especially parents of children with leukemia) may not accurately remember or may not feel comfortable discussing their past tobacco use history during the interview. The lack of an observed association between maternal smoking during pregnancy and childhood leukemia may be related to this potential for bias when using interview data to assess exposure. Alternatively, the lack of an observed association may also be the result of smoking-induced adverse birth outcomes (e.g., fetal loss, still birth) that preclude the subsequent development of childhood leukemia, thereby biasing epidemiological findings. Summary of Existing Research on Tobacco Use and Childhood Leukemia Pooled analyses of data from studies around the world show a relationship between paternal smoking Curr Probl PediatrAdolesc Health Care, October 2016 before conception and AML risk. Likewise, these findings were supported by an independent systematic review and meta-analysis. Future studies will provide an increased focus on the role of prenatal maternal smoking on leukemia risk in Latino children and will pool data from the Childhood Leukemia International Consortium for an analysis of smoking-related ALL risk. Chemicals Found in Paints, Petroleum Solvents, and Traffic Emissions A collection of studies have investigated the associations between childhood leukemia and a looselyrelated group of environmental exposures including paint, petroleum solvents, and vehicle traffic. These general exposure categories share characteristic chemical signatures, including the leukemogenic agent, benzene. Investigators from the California Childhood Leukemia Study examined the association between childhood leukemia and the use of paint or petroleum solvents in the home before birth and in early childhood. The analysis included 550 ALL cases, 100 AML cases, and one or two controls per case individually matched for sex, age, Latino ethnicity, and race. Conditional logistic regression techniques were used to adjust for income. Home paint exposure was associated with ALL risk (OR¼1.65; 95% CI: 1.26─2.15). The association was restricted to ALL with t(12;21) (OR ¼ 4.16, 95% CI: 1.66─10.4). Home use of petroleum solvents was associated with an increased risk for AML (OR ¼ 2.54, 95% CI: 1.19─5.42) but not ALL. Pooled Analyses of Home Exposure to Paint and Childhood Leukemia The Childhood Leukemia International Consortium pooled individual responses to questions about home paint exposures from eight case–control studies.86 Data were harmonized to account for inter-study differences in reported paint types, time periods of exposure, and leukemia subtypes and a compatible format was used in logistic regression. ALL risk was associated with home paint exposure in the 1–3 months before conception (OR ¼ 1.54; 95% CI: 1.28─1.85; N ¼ 3002 cases and 3836 controls), during pregnancy (OR ¼ 1.14; 95% CI: 1.04─1.25; N ¼ 4382 cases and 5747 controls), and after birth (OR ¼ 1.22; 95% CI: 1.07─1.39; N ¼ 1962 cases and 2973 controls). The 329 risk was greater if someone other than the parents did the painting, for example, a professional painter, which is indirect evidence of a dose–response relationship. The paint–leukemia association was stronger for ALL with t(12;21) than for other cytogenetic subtypes of leukemia. Meta-analyses of Parental Occupational Exposures to Paint, Solvents, Vehicle Exhaust, and Childhood Leukemia As an extension of the research that has identified an association between exposure to paint in the home and childhood leukemia, meta-analyses have demonstrated associations between childhood leukemia risk and parental exposures in related occupational settings. For example, a meta-analysis summarizing the existing literature on parental occupational exposures and childhood cancer found that relationships between childhood leukemia and paternal exposure to paints, solvents (e.g., benzene and trichloroethylene), and employment in motor vehicle-related occupations were among the strongest of the more than 1000 occupation–childhood cancer combinations that were evaluated.87 The studies that comprised this meta-analysis had several limitations related to the quality of the exposure assessment including the small numbers of exposed cases studied, the likelihood of false positives due to multiple comparisons, and possibility of publication bias. Despite these limitations, this metaanalysis provides evidence that when parents are exposed to certain chemicals at work, the effects are harmful to their offspring. Another meta-analysis summarized findings from 28 case–control studies and one cohort study that investigated the relationship between maternal occupational exposures and childhood leukemia in the offspring using 16,695 participating cases and 1,472,786 controls.88 ALL risk was associated with maternal paint exposures at work during pregnancy (OR ¼ 1.23, 95% CI: 1.02─1.47), with maternal solvent exposures at work during pregnancy (OR ¼ 1.25, 95% CI: 1.09─1.45), and with maternal petroleum exposure at work during pregnancy (OR ¼ 1.42, 95% CI: 1.10─1.84). No publication bias was found in this meta-analysis and consistent results were observed for subgroup and sensitivity analyses. However, an analysis of parental occupational paint exposures pooled from 13 case–controls studies revealed no increased risk for childhood ALL or AML.89 330 Carlos-Wallace et al.90 conducted meta-analyses to evaluate the risk of childhood leukemia associated with parental occupational exposure to benzene and solvents, as well as household use of products containing benzene and solvents. Maternal occupational exposure to benzene was associated with increased risk of childhood leukemia, yielding a summary relative risk of 1.71 (95% CI: 0.91─3.24). Use of household products containing benzene, aromatic hydrocarbons, solvents, or petroleum, was also associated with childhood leukemia risk, with a summary relative risk of 1.67 (95% CI: 1.01─2.78). The above associations were stronger for AML than for ALL and strongest for women who were exposed during pregnancy. The California Childhood Leukemia Study examined the relationship between occupational exposure to organic solvents and the risk of childhood leukemia.91 Occupational histories were obtained via interview using 19 task-based job modules from parents of children with ALL (N ¼ 670), children with AML (N ¼ 104), and healthy control children (N ¼ 1021). Logistic regressions were used to estimate odds ratios adjusted for socio-demographic factors. Among children with non-Latino fathers, none of the exposures evaluated were associated with risks of ALL and AML. In contrast, exposure to any organic solvents in Latino fathers was associated with an increased risk of childhood ALL (OR ¼ 1.48, 95% CI: 1.01─2.16); in multivariable analyses and the odds ratio for exposure to chlorinated hydrocarbons, in particular, was elevated (OR ¼ 2.28, 95% CI: 0.97─5.37), whereas risk estimates for other exposures—aromatic hydrocarbons, glycol ethers, and other hydrocarbon mixtures—were close to one. One common industrial chlorinated hydrocarbon, which might be the causal agent underlying the observed association, is trichloroethylene (TCE). As with other analyses that rely on interview-derived exposure data, the specific chlorinated hydrocarbon or mixture of chlorinated hydrocarbons responsible for the associations observed in these studies is not known. A large majority of mothers were not exposed to chemicals at work, and no associations were reported for risk of childhood ALL and AML. Traffic-Related Air Pollution and Childhood Leukemia Three recent meta-analyses independently demonstrated an association between childhood leukemia and postnatal traffic exposure.90,92,93 Boothe et al.92 Curr Probl PediatrAdolesc Health Care, October 2016 combined findings from seven studies to estimate that childhood leukemia was positively associated with postnatal traffic density near the residence (OR ¼ 1.53, 95% CI: 1.12─2.10). There was no association between childhood leukemia and prenatal traffic exposures. Filippini et al.93 combined 6 ecologic and 20 case– control studies that had assessed home exposure to traffic-related pollution by estimating traffic density in the neighboring roads, by estimating the vicinity to gas stations, or by modeling ambient nitrogen dioxide and benzene concentrations with GIS. Among high-quality studies that used traffic density to assign exposure, no significant increase in childhood leukemia risk was observed, even in the highest exposure category (OR ¼ 1.07, 95% CI: 0.93─1.24). Among studies that used NO2 estimates as the measure of exposure, there was a marginally significant association with childhood leukemia (OR ¼ 1.21, 95% CI: 0.97─1.52), which was stronger for ALL (OR ¼ 1.21, 95% CI: 1.04─1.41) than for AML (OR ¼ 1.06, 95% CI: 0.51─2.21). Among studies that used benzene estimates as the measure of exposure, there was a stronger association with AML (OR ¼ 2.28, 95% CI: 1.09─4.75) than ALL (OR ¼ 1.09, 95% CI: 0.67─1.77). Observed associations between childhood leukemia and exposure to traffic pollution were generally stronger for exposures in the postnatal period compared to the prenatal period. Carlos-Wallace et al.90 conducted meta-analyses to evaluate the risk of childhood leukemia associated with traffic density and traffic-related air pollution. Both measures of traffic were associated with childhood leukemia; the summary relative risk was 1.48 (95% CI: 1.10─1.99). The relationship was stronger for AML than for ALL and stronger in studies that involved detailed models of traffic pollution than in those that estimated traffic density. The findings from these three meta-analyses support a link between ambient exposure to traffic pollution and childhood leukemia risk, particularly due to benzene. Benzene as the Potential Causal Agent Once again, most of the studies that have evaluated the risk of childhood leukemia associated with paint, petroleum solvents, or traffic density are based on parent interviews. As such, it is challenging to identify the specific causal agent underlying the observed associations in these existing studies. Benzene is one potential culprit as it is a well-known leukemogen that Curr Probl PediatrAdolesc Health Care, October 2016 is present in oil-based paints, petroleum solvents used in occupational and residential settings (e.g., in paint thinner), and vehicle exhaust. However, given that childhood leukemia subtype analyses have yielded disparate results depending on the specific exposure measure that was used in the meta/pooled analysis, it is also possible that this loose grouping of chemical exposures actually comprises several distinct chemical risk factors for leukemia. For example, in addition to benzene, other chemicals, such as 1,3-butadiane, styrene, xylene, and polycyclic aromatic hydrocarbons (PAHs) might also play a role in some of these observed relationships. The recent development of a mouse model for childhood leukemia will enable investigators to evaluate the role of specific chemical risk factors in the etiology of childhood leukemia. Summary of Existing Research on Paint, Solvents, Traffic and Childhood Leukemia Pooled analyses of data from studies around the world show a relationship between home exposure to paint and ALL risk. These findings were indirectly supported by a variety of systematic reviews and metaanalyses, which showed evidence of relationships between childhood leukemia and exposure to petroleum solvents (at home and at the mother’s work) and traffic (as measured by surrounding traffic density and modeled concentrations of traffic-related air pollutants). Several possible chemical risk factors may explain the observed association, including the wellknown leukemogen, benzene. Persistent Organic Pollutants Epidemiological studies of childhood leukemia that use environmental or biological samples are relatively scarce. A series of analyses conducted as part of the California Childhood Leukemia Study have evaluated the relationship between childhood ALL and chemical concentrations in settled dust collected from participating homes as surrogates for chemical exposures. Polycyclic Aromatic Hydrocarbons (PAHs) and Childhood Leukemia The California Childhood Leukemia Study evaluated the relationship between childhood ALL and PAH concentrations in settled dust.94 As part of this population-based case–control study, dust samples were collected from 251 ALL cases and 306 birthcertificate controls using a high-volume small-surface 331 sampler (N ¼ 185 cases, 212 controls) or directly from participants' household vacuum cleaner bags (N ¼ 66 cases, 94 controls). Logistic regression was used to evaluate the relationship between ALL risk and logtransformed concentrations of 9 individual PAHs, the summed PAHs, and the summed PAHs weighted by their carcinogenic potency (the toxic equivalence) while adjusting for demographic characteristics and duration between diagnosis/reference date and dust collection. Among participants with dust samples collected by high-volume small-surface sampler, risk of ALL was not associated with increasing concentration of any PAHs. However, among participants with dust samples collected by participants' vacuum cleaners, a positive association was observed between ALL risk and increasing concentrations of benzo[a] pyrene (OR ¼ 1.42, 95% CI: 0.95─2.12), dibenzo[a,h] anthracene (OR ¼ 1.98, 95% CI: 1.11─3.55), benzo[k] fluoranthene (OR ¼ 1.71, 95% CI: 0.91─3.22), indeno [1,2,3-cd]pyrene (OR ¼ 1.81, 95% CI: 1.04─3.16), and the toxic equivalents (OR ¼ 2.35, 95% CI: 1.18─4.69). The observed association between ALL risk and PAH concentrations among participants with dust collected by vacuum suggests that PAH exposure may increase the risk of childhood ALL; however, understanding the reasons for the different results by sample type requires further scrutiny. PAHs are byproducts of incomplete combustion that are found at high concentrations in cigarette smoke and vehicle exhaust. PAHs, especially dibenzo[a,h]anthracene, are potent human carcinogens. As such, it is possible that one PAH or a combination of PAHs may be the causal agent(s) responsible for the observed associations between parental smoking and childhood leukemia or between traffic density and childhood leukemia. Polychlorinated Biphenyls (PCBs) and Childhood Leukemia The California Childhood Leukemia Study has also evaluated the relationship between childhood ALL and levels of six PCBs—industrial chemicals that are probable human carcinogens and immune system disruptors—in settled dust.81 The PCB analysis included 184 ALL cases 0–7 years of age and 212 birth certificate controls matched to cases by birth date, sex, race, and Latino ethnicity. Dust samples were collected from the room where the child spent the most time using the high-volume small-surface sampler. In 332 homes where any PCB was detected in the dust, there was a 2-fold increased risk of ALL (OR ¼ 1.97, 95% CI: 1.22─3.17). When considering the sum of the six PCBs analytes, compared to those in the lowest quartile of Σ6PCBs, the highest quartile was associated with about a 3-fold risk of ALL (OR ¼ 2.78, 95% CI: 1.41─5.48). The risk of ALL was positively associated with increasing concentrations of PCB congeners 118, 138, and 153 in dust. The associations with PCBs were stronger among non-Latino whites than among Latinos despite the presence of a similar distribution of PCB levels among controls in each racial/ethnic groups. Polybrominated Diphenyl Ethers (PBDEs) and Childhood Leukemia Along the same lines, the California Childhood Leukemia Study evaluated the relationship between childhood ALL and levels of PBDEs—chemical flame retardants—in settled dust.95 PBDEs are structural analogs to PCBs that also cause immune system perturbations. The PBDE analysis included 167 ALL cases 0–7 years of age and 214 birth certificate controls matched on date of birth, sex, and race/ethnicity. Dust samples were collected from carpets in the room where the child spent the most time while awake using a highvolume small-surface sampler or by sampling from participants' household vacuum cleaners. Concentrations of 14 PBDE congeners were measured including constituents of the Penta- (28, 47, 99, 100, 153, 154), Octa- (183, 196, 197, 203), and Deca-BDEs commercial mixtures (206–209). Odds ratios were calculated using logistic regression, adjusting for demographics, income, year of dust collection, and sampling method. Comparing the highest to lowest quartile showed no association with ALL for summed Penta- (OR ¼ 0.7, 95% CI: 0.4─1.3), Octa- (OR ¼ 1.3, 95% CI: 0.7─2.3), or Deca-BDEs (OR ¼ 1.0, 95% CI: 0.6─1.8). Comparing homes in the highest tertile to those below the analytical limit of detection, revealed a significant positive association with ALL risk for BDE-196 (OR ¼ 2.1, 95% CI: 1.1─3.8), BDE-203 (OR ¼ 2.0, 95% CI: 1.1─3.6), BDE-206 (OR ¼ 2.1; 95% CI: 1.1─3.9), and BDE-207 (OR ¼ 2.0, 95% CI: 1.03─3.8). Interestingly, the significant associations with ALL risk observed in this analysis were for minor PBDE congeners that are found in dust at relatively low concentrations; whereas the most abundant PBDE congeners (e.g., BDEs 47, 99, and 209) were not associated with ALL risk. These low-concentration Curr Probl PediatrAdolesc Health Care, October 2016 PBDE congeners were measured with less analytical precision than their more common analogs, because the measured values were relatively close to the analytical limit of detection. In other words, the low-level PBDE congeners that were associated with ALL risk in this analysis were the ones measured with the least precision and, therefore, the ones with the greatest potential for a spurious finding. Still, there may be a plausible biological mechanism to explain the inconsistency of the risk estimates between PBDE congeners, as toxic and carcinogenic effects are expected to differ by congener.48,96 The fact that PCBs and PBDEs have a similar chemical structure lends credence to the hypothesis that these chemicals may be acting via the same mechanism of action, for example, immune dysregulation. Strengths of Existing Research on Persistent Organic Pollutants and Childhood Leukemia Unlike much of the research described in this section, one strength of the existing research on persistent organic pollutants and childhood leukemia is the use of objective environmental measurements to assess chemical exposures, rather than interviews. Not only does this reduce the likelihood of recall bias, but it also allows investigators to identify specific chemicals as causal agents in the etiology of leukemia. Recognizing causal agents can be the first step in planning a successful intervention that will reduce future incidence of leukemia. Limitations of Existing Research on Persistent Organic Pollutants and Childhood Leukemia The relative stability of persistent organic pollutants in settled dust allows for exposure measurements that have limited temporal variability. This stability is a benefit of the sampling technique, because the resulting measurements represent long-term average levels of chemical contamination, which can be useful when trying to estimate past chemical exposures. However, this stability also obscures short-term fluctuations in chemical levels that might be important to investigators who want to identify critical windows of a child’s development when chemical exposures are especially harmful. That is, measuring chemical levels in settled dust will not enable a researcher to distinguish the leukemogenic effect of prenatal vs. postnatal chemical exposures, for example. Curr Probl PediatrAdolesc Health Care, October 2016 Moreover, children are exposed to chemicals through several other pathways in addition to the ingestion of contaminated settled dust. In particular, the ingestion of settled dust plays a relatively minor role in children’s exposure to PCBs compared to the ingestion of PCB-contaminated food. This is owing to the bioaccumulative nature of PCBs and the fact that they have been banned from production in the U.S. for several decades. Indeed, it is a testament to the persistence of these hazardous chemicals that they can still be so readily measured inside homes. In these analyses, the design of the California Childhood Leukemia Study did not account for chemical exposures received via the inhalation of contaminated air, the ingestion of contaminated food, or any other pathways. As such, the dust measurements are limited surrogates for total chemical exposure. To date, the California Childhood Leukemia Study has only utilized dust samples to identify risk factors for ALL, the most common leukemia subtype. There is an insufficient number participants with dust samples available to analyze the risk of AML or to stratify by cytogenetic subtype. Also of some concern is the fact that the subset of California Childhood Leukemia Study participants who were eligible for and consented to dust sampling had higher socioeconomic status than the full California Childhood Leukemia Study population and its underlying source population, the State of California. As such, the findings from these studies may not be representative of the general population. Fortunately, data from the California Childhood Leukemia Study indicates that both the case families and the control families participating in dust sampling had elevated socioeconomic status, which limits the risk of differential selection bias. Perhaps the most important limitation of these studies is that they have not been substantiated by independent investigators. Whereas many members of Childhood Leukemia International Consortium and other studies have collected interview data about pesticides, smoking, and paint; very few have measured chemicals in environmental samples. As such, there are no meta-analyses or pooled analyses published showing the risk of ALL associated with exposure to persistent organic pollutants, as there have been previously for the other, more well-studied, ALL risk factors that were discussed above. 333 Summary of Existing Research on Persistent Organic Pollutants and Childhood Leukemia Three novel findings from the California Childhood Leukemia Study suggest that home exposures to persistent organic pollutants, such as PAHs, PCBs, and PBDEs, are associated with an increased risk of ALL. Future studies should attempt to interrogate these associations to replicate or refute their veracity. Radiation In utero exposure to low-dose radiation delivered from medical x-rays is one of the few widelyrecognized risk factors for childhood leukemia.97 While the prevalence of fetal exposure to x-rays in utero has decreased markedly following radiation protection standards, the use of medical imaging procedures, including computerized tomography (CT) scans,98 has increased drastically during the past 30 years. In fact, CT scans are now the largest medical sources of radiation in economically developed countries.99 In addition, CT scans deliver an effective dose of radiation that is up to several hundred times stronger than conventional x-rays (depending on the target organ), which has led to major increases in the per capita radiation dose from medical sources (up 600% in the US since 1980).99 The carcinogenic effects of CT scans have not been established, but exposures to children are especially concerning because they are more sensitive to radiation-induced cell damage.97 Findings on children’s postnatal exposure to lowdose medical radiation and the risk of childhood leukemia are inconsistent, with modest positive associations reported in some studies, but not all.97,99 Risk prediction models have anticipated increased risks of childhood leukemia following CT scans, but these models were criticized for extrapolating from the effects of much higher levels of radiation that were observed in the Life Span Study of atomic-bomb survivors.100–102 Only one case–control study to date has published results on self-reported history of CT scans showing no increased risk of childhood ALL, based on small numbers of exposed children.103 Recent cohort studies with access to medical data in Europe,101,104–106 Australia,107 and the US,102 have reported small to moderate increases in the risk of leukemia in children exposed to CT scans; findings, however, were based on small number of excess cases ranging from 6 to 74. Pearce et al.106 conducted a retrospective cohort study of 178,604 UK children 334 and young adults with CT scans from 1985 to 2002. Patients were followed up through 2008 and linked with 74 leukemia diagnoses and 135 brain cancer diagnoses. A cumulative dose of 50 milligray was estimated to triple the risk of childhood leukemia within 10 years of the first CT scan; likewise, a cumulative dose of 60 milligray tripled the risk of brain cancer.106 The main criticism of the study was the lack of an unexposed control group. Other cohort studies are underway108 including a pooled European study (EPI-CT)104 of about one million subjects (age 0–21 years). Although the number of excess childhood leukemia cases is still likely to be low (estimated to be approximately 60–100), this study will obtain more precise exposure information than previously possible, including individual doses of radiation for each organ. Confounding by indication (reverse causation) is also a concern in studies of the relationship between medical radiation and childhood leukemia. In a French cohort study, the analyses accounting for child’s cancerpredisposing factors (mostly rare genetic conditions in less than 2% of children) showed a modest impact on the risk of childhood leukemia associated with CT scans.109 This was consistent with a case–control study of prenatal x-rays showing an overall 13% reduction in relative risk of childhood cancers, after adjusting for maternal illnesses during pregnancy.110 Others have argued that cancer-predisposing conditions may instead act as effect modifiers.111–113 Overall, despite methodological challenges, epidemiologic studies so far mostly support an association between postnatal exposure to CT scans and childhood leukemia, while results for lower dose x-rays are less consistent. Pooled analyses reported that exposure to high levels of extremely low-frequency-electromagnetic fields over 0.3 or 0.4 mT is associated with an increased risk of childhood leukemia,114,115 which was the basis to classify extremely low-frequency-electromagnetic fields as possibly carcinogenic to humans (Group 2B).116 Methodological issues including possible confounding, selection bias, and measurement errors have been put forward as an alternate explanation for the observed association, and animal studies are ongoing to identify possible biological mechanisms.117,118 If the association between extremely low-frequencyelectromagnetic fields and childhood leukemia is causal, the overall population attributable risk has been estimated to be 1.9% (1–4% depending on the countries).117,118 Curr Probl PediatrAdolesc Health Care, October 2016 Future Steps in Identifying Environmental Causes of Childhood Leukemia examining relationships between exposure and disease that have not been considered, to date. A multitude of studies on the environmental causes of childhood leukemia, including those conducted by our Dietary Risk Factors for Childhood research team and others that are described above, have Leukemia led to progress in identifying the etiological roles of environmental exposures in childhood leukemia. HowIntroduction ever, much work remains to be done. As discussed, Diet has been linked to several cancers in adults and several of the observed associations between chemical children.119 These observations may be explained by exposures and childhood leukemia are derived from various biological mechanisms such as exposure to interview data, an approach which can limit an invesdietary mutagens, mutagenesis due to nutrient defitigator’s ability to identify specific chemical risk factors ciencies, and intake of micronutrients and other dietary for childhood leukemia. As such one goal of future components that may protect against the development research will be to understand which specific causal of cancer by supporting cellular integrity, reducing agents underlie previously observed associations. For inflammation and improving immune response.120–122 example, in collaboration with the National Cancer A growing body of research also suggests that the Institute, the California Childhood Leukemia Study is influence of particular nutrients on epigenetic procconducting a study to measure glyophosate in particesses may contribute to carcinogenesis.123 ipating homes. This study will follow-up on a previThe role of the intrauterine environment is crucial in ously observed association between reported residential determining risk of disease later in life, and the herbicide use and childhood ALL. The California “developmental origins of health and disease” hypothChildhood Leukemia Study has observed associations esis posits that nutritional and between childhood leukemia risk environmental exposures in and levels of certain PCBs, PBDEs, PAHs, and herbicides Diet quality is a comprehensive utero permanently alter gene in settled dust. These unique measure of nutritional status, expression and the physical development of the fetus findings need to be confirmed and healthy maternal diet at the through a process called “proin independent studies, preferatime of conception and during gramming.”124,125 This hypothbly in ones that use a distinct method for assessing children’s pregnancy is linked to a reduced esis is likely central to the exposure to chemicals. risk of leukemia in the offspring. development of childhood leukemia, as well. Indeed, materAs an alternative approach for nal nutrition during pregnancy identifying the causal agents in may be related to both the occurrence of primary and childhood leukemia, the Center for Integrative Research secondary oncogenic events that lead to leukemia in on Childhood Leukemia and the Environment will the offspring. Subsequently, a child’s diet during the employ a mouse model of ALL with t(12;21) to test early years of life is likely to also play an important the carcinogenic potential of a variety of chemicals, role in leukemogenesis. Breastfeeding has been conincluding, for example, compounds that comprise the sistently found to reduce leukemia risk in children, via broadly-defined groups of pesticides, solvents, or traffic intake of essential nutrients not readily available in pollutants. The mouse model will also be helpful in newborns and due to the resulting beneficial effects of identifying mechanisms of action for the chemical risk immune system priming. In contrast, the impact of diet factors of childhood leukemia. In fact, this is a central —both the mother’s and the child’s—on childhood goal of the Center, as immunological factors and leukemia has been less studied. epigenetics will both be interrogated as possible cancer Here we provide an overview of the current knowlmechanisms. Moreover, state-of-the-science untargeted edge about the associations between diet and childhood analytical approaches will allow investigators to identify leukemia. novel chemical risk factors for childhood leukemia, by Curr Probl PediatrAdolesc Health Care, October 2016 335 Maternal Diet demonstrated to influence DNA methylation in children.131 Reduced risks of childhood Epidemiologic studies examfolic acid supplemenleukemia are associated with Maternal ining the relationship between tation also protects against some adequate folate before maternal diet during pregnancy childhood diseases, such as neuconception and early in and the risk of childhood leural tube defects.132 High folate kemia, have been mostly conintake has been associated with pregnancy, breastfeeding, ducted in developed countries. of breast133 and and early exposure to routine reduced risk 134 Exposures of interest varied to colorectal cancer and childhood infections. include food groups (e.g., fruits, increased risk of prostate canvegetables, proteins), micronucer135 among adults, whereas trients from dietary and supplement sources (e.g., meta-analyses have indicated no effect of folic acid folate and vitamins), food sources of topoisomerase supplementation on adult cancer incidence.135–137 II inhibitors (known risk factors for treatment-related Maternal intake of folate and other nutrients involved leukemia), consumption of coffee, tea, and alcohol, in one-carbon metabolism may influence childhood and holistic measures of a healthy diet. leukemia risk due to the importance of these nutrients for DNA synthesis and repair, chromosomal integrity, and epigenetic processes that determine gene expresFood Groups sion and influence cancer risk, including histone Fruits and vegetables contain a variety of vitamins and modification, levels of non-coding RNAs, and DNA minerals that have anti-cancer, anti-proliferative, and antimethylation.123,130 123 inflammatory effects, and the consumption of fruits Practices of prenatal folic acid supplementation have and vegetables has been associated with a reduced risk of varied substantially across countries and over time, and various types of cancer.119 Some food groups, in individual epidemiologic studies that have evaluated particular fruits and vegetables, have been associated the relationship between maternal folate intake through with childhood leukemia risk in several studies. Research supplements and the risk of childhood ALL have has found statistically significant negative associations yielded mixed findings. However, a recent Childhood between maternal consumption of fruits and vegetables Leukemia International Consortium analysis, the larg126–128 and risk of childhood ALL, and one study found est to date, pooled original interview data from 12 significant or near-significant inverse linear trends studies (representing 6963 ALL, 585 AML, and between the risk of infant leukemia and maternal con11,635 healthy controls) to observe that folic acid sumption of fresh fruits and vegetables, especially for supplementation was protective against childhood 129 specific ALL subtypes. Negative associations have leukemia.138 Folic acid taken before conception or also been observed for childhood leukemia and maternal during pregnancy—with or without intake of other consumption of other food groups, specifically protein vitamins—was associated with a reduced risk of sources such as fish and seafood128 as well as beans and childhood ALL (OR ¼ 0.80, 95% CI: 0.71─0.89) 126,127 One study demonstrated an increased risk of beef. and AML (OR ¼ 0.68, 95% CI: 0.48─0.96), after ALL with increased maternal consumption of meat or adjustment for study center.138 Interestingly, the meat products and sugars or sirups.128 reduced risks were seen only among women with low and medium education levels—a surrogate marker Folate and Other One-Carbon Metabolism for low socio-demographic status and possibly for a low-quality (low-folate) diets—suggesting that women Nutrients who enter the peri-conception period with inadequate The one-carbon metabolism cycle is critical for the folate/vitamin levels would benefit the most from synthesis of DNA and RNA, the conversion of prenatal folate and vitamin supplementation. homocysteine to methionine, and the formation of Only a small number of studies, however, have s-adenosylmethionine (SAM), the primary methyl 130 examined the role of folate intake from food or the donor for DNA, RNA, proteins, and lipids. Folate role of any other nutrients involved in the one-carbon and other B vitamins are important cofactors in the metabolism cycle in the development of childhood one-carbon metabolism cycle,130 and maternal folic leukemia. In addition, there has been only limited acid supplementation during pregnancy has been 336 Curr Probl PediatrAdolesc Health Care, October 2016 consideration of the role of maternal diet on risk of AML. A case–control study in Australia found some evidence that higher dietary intakes of folate and B12 from food in the last 6 months of pregnancy were associated with a decreased risk of ALL, whereas higher dietary intakes of vitamin B6 were unexpectedly associated with an increased risk of ALL.139 Previous analyses from a subset of the California Childhood Leukemia Study population examined the total intake of folate, vitamin B6 or vitamin B12 from both diet and supplements, and found no associations with ALL.126,127,140 However, in a recent and expanded California Childhood Leukemia Study analysis (681 ALL cases, 103 AML cases, and 1076 controls) that employed principal components analysis to account for the high correlations between nutrients, higher maternal intake of one-carbon metabolism nutrients from food and supplements was associated with a reduced risk of ALL (OR ¼ 0.91, 95% CI: 0.84─0.99) and possibly AML (OR ¼ 0.83, 95% CI: 0.66─1.04).141 The association of ALL with nutrient intake exclusively from food (excluding supplements) was similar to the association of total nutrient intake from food and supplements, both in the study population overall and within racial/ethnic groups. However, high intake of B vitamins from supplements (versus none) was associated with a statistically significant reduced risk of ALL in children of Latinas (OR ¼ 0.36 95% CI: 0.17─0.74), but not in children of non-Latina white women (OR ¼ 0.76, 95% CI: 0.50─1.16) or Asian women (OR ¼ 1.51, 95% CI: 0.47─4.89).141 Racial/ethnic differences in nutrient intake and in genetic polymorphisms in the one-carbon (folate) pathway may partly explain the observed difference in leukemia risk. In a biomarker analysis conducted as part of the California Childhood Leukemia Study, folate concentration measured in neonatal blood samples were similar between children with leukemia (313 ALL cases and 44 AML cases) and 405 controls, suggesting that folate levels at the end of pregnancy did not affect leukemia risk.63 The study was conducted in California where most pregnant women used prenatal vitamin supplementation, therefore limiting the ability to detect an association. Moreover, late pregnancy may not be the period of a child’s development during which a beneficial effect of folic acid and multivitamins, is critical to leukemia risk. However, this explanation would not be consistent with observations from the large pooled analyses of the Childhood Leukemia International Consortium that showed reduced risks Curr Probl PediatrAdolesc Health Care, October 2016 of ALL and AML associated with self-reported supplementation during each trimester of the pregnancy.138 Newborn and child serum-nutrient levels are influenced by many factors, including maternal and child genetic polymorphisms.142 Many studies have found significant associations between single nucleotide polymorphisms (SNPs) in folate-related genes, such as MTHFR variants, and childhood leukemia; but, there are inconsistencies in the specific SNPs that have been identified across studies.140,143–147 A study examining the largest number of genes and SNPs in the folate pathway found statistically significant associations between SNPs in genes CBS, MTRR, and TYMS/ ENOFS (but not MTHFR) and childhood ALL.140 Levels of maternal folate intake during pregnancy, and child’s Latino ethnicity were found to modify some of these associations.140 Other studies, however, did not report such interactions.144,148,149 Key findings regarding folate intake and other vitamins are summarized in Table 2. Topoisomerase II Inhibitors The use of topoisomerase II inhibitors (a nuclear enzyme involved in DNA replication) in cancer chemotherapy has long been known to be associated with common MLL gene translocations that are characteristic of therapy-related AML.150 Consequently, it was hypothesized that topoisomerase inhibitors may be involved in the etiology of infant leukemia, because this subtype commonly involves the same MLL gene translocation that has been identified in these therapyrelated leukemias.151 Aside from chemotherapeutic agents, topoisomerase inhibitors are found in diverse sources, including herbal medicines, quinolone antibiotics, certain types of laxatives, and pesticides.152 Certain dietary sources also contain topoisomerase inhibitors including, but not limited to, tea, coffee, wine, and certain fruits and vegetables.152 Recent laboratory studies, however, suggest that tea, wine, and cocoa do not inhibit topoisomerase activity in vitro and thus are unlikely to increase the risk of MLL translocations.153 A small exploratory study examining maternal exposure to topoisomerase inhibitors during pregnancy and the risk of childhood leukemia found an increased risk of AML with increasing topoisomerase II inhibitor exposure (OR ¼ 10.2; 95% CI: 1.1─96.4; N ¼ 29 cases for high exposure), but no increased risk for ALL 337 TABLE 2. Association between folate and other vitamins and risk of childhood leukemia: selected case–control studies Population No. of cases Source of vitamins Period of interest OR (95% CI) Childhood Leukemia 6963 ALL International Consortium.138 585 AML Maternal supplement Prenatal Folate only ALL: OR ¼ 0.80 (0.78, 0.92) AML: OR ¼ 0.68 (0.48–0.96) Any vitamins ALL: OR ¼ 0.85 (0.80, 0.94) AML: OR ¼ 0.92 (0.75–1.14) Australia.139 Maternal diet Pregnancy Folate ORQ2 ¼ 0.68 (0.44, ORQ3 ¼ 0.58 (0.37, ORQ4 ¼ 0.44 (0.27, ORQ5 ¼ 0.70 (0.44, p-trend ¼ 0.05 B12 vitamin ORQ2 ¼ 0.72 (0.47, ORQ3 ¼ 0.79 (0.71, ORQ4 ¼ 0.85 (0.71, ORQ5 ¼ 0.49 (0.71, p-trend ¼ 0.02 B6 vitamin ORQ2 ¼ 1.04 (0.67, ORQ3 ¼ 1.15 (0.74, ORQ4 ¼ 1.28 (0.82, ORQ5 ¼ 1.60 (1.02, p-trend ¼ 0.03 333 ALL 1.06) 0.91) 0.71) 1.12) 1.10) 1.21) 1.31) 0.77) 1.62) 1.81) 2.00) 2.51) California.141 681 ALL 103 AML Maternal diet & supplements Peri-conception One-carbon metabolism nutrients (i.e., folate, B vitamins) ALL: ORPC ¼ 0.91 (0.84, 0.99) AML: ORPC ¼ 0.91 (0.66, 1.04) California.63 317ALL 44 AML Blood levels Hemoglobin concentration of folate at birth No difference in between ALL, AML, and controls Neonatal ALL ¼ acute lymphoblastic leukemia; AML ¼ acute myeloblastic leukemia; Q ¼ quintile; PC ¼ principal component for one-carbon metabolism nutrients. (OR ¼ 1.1; 95% CI: 0.5─2.3; N ¼ 82).151 Subsequent studies confirmed a positive relationship between maternal dietary intake of topoisomerase II inhibitors and risk of infant AML with a MLL gene translocation, but have found no association between dietary intake of topoisomerase II inhibitors and risk of other subtypes of infant AML or any type of ALL.126,129 Coffee, Cola, and Tea An early case–control study of 280 cases and 288 hospitalized controls found an increased risk of ALL among children of mothers reporting coffee consumption more than four cups a day during pregnancy (OR ¼ 2.4; 95% CI: 1.3─4.7 for 4–8 cups; and OR ¼ 3.1, 95% CI: 1.0─9.5 for 48 cups), with similar ORs observed for acute non-lymphoblastic leukemia that did not reach statistical significance.154 Subsequent case–control studies have found maternal 338 consumption of coffee during pregnancy to be associated with an increased risk of ALL, AML, and possibly infant leukemia, while others have failed to find an association, as summarized in a recent metaanalysis.155 There is some evidence from these studies that the increased risk of leukemia with maternal coffee consumption may be more pronounced among children born to non-smoking mothers.156,157 Similarly, cola-based drinks have been associated with increased risk of childhood ALL (summary OR ¼ 1.31, 95% CI: 1.09─2.47), while reduced risks have been reported with maternal tea consumption during pregnancy (summary OR ¼ 0.85, 95% CI: 0.75─0.97).155 A general limitation of those studies is the lack of information on the type of drinks (e.g., caffeinated or not, green or black tea), which contain different nutrients and other compounds with either anti- or pro-carcinogenetic properties. Curr Probl PediatrAdolesc Health Care, October 2016 Alcohol also been associated with birth outcomes, such as Maternal alcohol intake before or during pregnancy neural tube and congenital heart defects.168 In a recent study169 overall maternal diet quality, as has been hypothesized to influence childhood leukemia summarized by a diet quality index using a modified risk by altering immune function or by teratogenic version of the 2010 Healthy Eating Index, was effects on cell differentiation.158 Alcohol is also an associated with a reduced risk of childhood ALL antagonist to folate metabolism and methionine syn(OR for each five point increase on the index ¼ thase and may modify DNA methylation status in 0.88, 95% CI: 0.78─0.98). A more pronounced reducinteraction with folate levels.159 A systematic review tion in risk was observed among younger children and and meta-analysis of 21 case–control studies found that children of women who did not use vitamin supplealcohol intake during pregnancy was associated with ments before pregnancy. There was a similar reduced AML (summary OR ¼ 1.56, 95% CI: 1.13─2.15 risk of AML with increasing produced from 9 studies maternal diet quality score, comprising 731 cases) but not with ALL (summary OR Breastfeeding for six months or although this assosciation was not statistically significant (OR ¼ ¼ 1.10, 95% CI: 0.93─1.29 longer, is associated with a 0.76, 95% CI: 0.52─1.11). No produced from 11 studies reduced risk of childhood single diet quality index compocomprising 5108 cases).159 leukemia, whereas early nent (i.e., food group or nutrient) Heterogeneity between studintroduction to milk formula appeared to account for the results, ies was explained in part by suggesting that the quality of the some studies160,161 that demmay increase leukemia risk. whole diet and the cumulative onstrated a negative associaeffects of many dietary components may be important tion of childhood leukemia with maternal alcohol in influencing childhood leukemia risk.169 consumption during pregnancy. Repeating the metaanalysis by subgroup of alcohol indicated an increased risk of AML associated with reported consumption of Paternal Diet wine but not beer or spirits, providing some additional 159 support for the topoisomerase II hypothesis. For In contrast to maternal diet, very few studies have ALL, there was an association between maternal examined the relationship between paternal diet before consumption of spirits during pregnancy, but not beer conception and childhood leukemia. One study sugor wine.159 One subsequent study supported the findgested that the risk of childhood leukemia increased with increasing paternal consumption of hot dogs ing of an increased risk of AML with maternal alcohol consumption during pregnancy162 while another did (sources of carcinogenic compounds from N-nitroso precursors).170 Also, there is no strong indication that not find an association156; neither found a relationship between maternal alcohol consumption and ALL. In paternal intake of folate and other vitamins from diet contrast, two other recent studies found that maternal and supplements before the child’s conception reduced consumption of alcohol during pregnancy was assothe risk of leukemia in the offspring.171,172 163 ciated with a decreased risk of ALL and of infant leukemia.164 Child’s Diet Healthy Diet Index In contrast to studies that have evaluated the role of a limited number of specific nutrients or food components, measures of overall diet quality may better represent nutritional status and the complex biological interaction of multiple nutrients.165 Diet quality indices are often positively correlated with biological markers of micronutrient intake and have been associated with reduced risk of all-cause mortality, including cancer risk.166,167 Maternal dietary patterns and quality have Curr Probl PediatrAdolesc Health Care, October 2016 As mentioned earlier, breastfeeding and duration of breastfeeding (6 months of more) have been associated with the risk of childhood ALL, as summarized in recent pooled173 and meta-analyses174 conducted by the Childhood Leukemia International Consortium. Besides breastfeeding, little is known about the influence of child’s early diet. Feeding with formula as early as 14 days after birth,175 alone or in combination with breast milk, was associated with an increased risk of childhood ALL and dose–response relationships were reported for the duration of formula 339 feeding.175,176 These studies contrasted previous null recognized that health care providers should act as findings.177 Infants and children fed with milk formula resources for information on environmental health for have been found to have higher serum levels of IGF-1 their patients; unfortunately, U.S. medical education is than those breastfed, and fetal growth pathway has largely void of training in Environmental Medicine.181 176 been hypothesized to play a role in leukemogenesis. Pediatricians and other providers report low selfAssociations between a child’s consumption of various efficacy in basic skills of Environmental Medicine food groups and the risk for childhood ALL or for all such as taking a history of environmental expoleukemias combined are inconsistent, especially sures.10,182,183 A survey of members of the American regarding the consumption of fruits and fruit juice, as College of Obstetrics and Gynecology reported that well as the consumption of meat.170,175,177–179 Older although three quarters of those surveyed agreed that counseling patients could reduce age at introduction to solid food exposure to environmental hazin general,176 and possibly ards, fewer than 20% routinely older age at introduction to Increased risks of childhood vegetables in particular,175 was leukemia have been consistently discussed environmental exposures—even known developassociated with an increased associated with exposures to mental toxicants—with their risk of childhood ALL, while pesticides, tobacco smoke, patients. Despite a lack of traina reduced risk was reported for ing in environmental health, late introduction to eggs.175 solvents, and traffic-related clinicians report a high interest Child’s consumption of colapollution. in learning more about current based drinks does not appear environmental health research as it may apply to their to be associated with leukemia, in contrast to maternal practice.10,182 consumption during pregnancy.155 Environmental Health Literacy is an evolving discipline that “combines key principles and procedural Conclusions elements from the fields of risk communication, health literacy, environmental health sciences, communicaMost of the evidence to date supports the role of tions' research and safety culture.”184 A basic level of prenatal folate supplementation, alone or with other environmental health literacy will help clinicians to vitamins, in reducing childhood leukemia risk. Apart have a sense of self-efficacy about environmental from the possible beneficial intake of fruits and health and to fulfill roles as alert clinicians, educators, vegetables both during pregnancy and in the early and advocates for children’s health. years of life, studies have led to mixed or isolated The key studies on environmental risk factors for findings regarding the potential leukemogenic effect of childhood leukemia are rarely published in clinical other food groups, likely due to small sample sizes and journals and presentations on new environmental challenges of collecting accurate descriptions of parepidemiologic research are rare at clinical meetings. ticipants' diets. In addition, there has been relatively These circumstances require clinicians to make a limited consideration of the role of maternal diet on special effort to stay informed about the impact of risk of AML, a less common subtype than ALL. the environment on health. Fortunately, many online Finally, measures of overall diet quality may represent resources are becoming available to make this easier nutritional status and the complex biological interac(Text Box 1). tion of multiple nutrients better than single-nutrient assessment, and should be used in future studies. A Clinical Perspective on Environmental Health Literacy Nurses, doctors, and other health care providers are highly respected as sources for health information; as such they play an important role in translating new scientific findings to the public.180 It has long been 340 Childhood Leukemia: Is it Time for Primary Prevention? As presented in the preceding sections of this article, there is a large and growing body of literature that demonstrates the role of environmental agents in determining the risk for childhood leukemias. This is in contrast to other pediatric cancers that are more rare and for which there have been far fewer environmental Curr Probl PediatrAdolesc Health Care, October 2016 TEXT BOX 1–Online resources on environmental health literacy for clinicians 1) Pediatric Environmental Health Toolkit online training. An introduction to the basics of children's environmental health and approaches to anticipatory guidance. Free continuing medical education credits (http://www.atsdr.cdc.gov/emes/health_pro fessionals/pediatrics.html). 2) A Story of Health. Multimedia e-book explores how environments interact with genes to influence health across the lifespan; includes a chapter on childhood leukemia. Free continuing medical education credits available from the CDC (http://wspehsu.ucsf.edu/forclinical-professionals/training/a-story-of-health-a-multimedia-ebook/). 3) Little Things Matter. Video illustrates key concepts in children’s environmental health (in multiple languages) (https://www.youtube.com/channel/UCbl p9EePwfR8doGm9JbJOjA/videos). 4) Webinars, fact sheets, and other resources are available from the Pediatric Environmental Health Specialty Units, a network of experts in reproductive and children's environmental health (http://www. pehsu.net/health_professionals.html). epidemiologic studies conducted. The evidence implicating environmental causes of childhood leukemia comes from a variety of individual studies worldwide and includes meta- and pooled analyses from the Childhood Leukemia International Consortium, as discussed in the Environmental Risk Factors for Childhood Leukemia section. Exposures to agents such as pesticides, tobacco smoke, solvents, and trafficrelated pollution have been consistently linked to an increased risk of developing childhood leukemia. On the other hand, intake of vitamins and folate supplementation during the preconception period or pregnancy has been associated with a reduced risk of childhood leukemia as have breastfeeding and early exposure to infection, as characterized by attendance at large daycare and pre-school settings. Despite the fact that many studies have identified modifiable risk factors (increased or decreased risk) for childhood leukemia we are aware of no current prevention program that specifically addresses childhood leukemia, anywhere in the world. The American Cancer Society does support programs that discourage tobacco use and promote healthy nutrition and exercise for children, with the understanding that addressing these factors early in life will reduce future cancer burden.1 Why is it that activities to reduce childhood leukemia have not been incorporated into cancer prevention programs when potentially modifiable risk factors have Curr Probl PediatrAdolesc Health Care, October 2016 been identified? Recently, our research group has suggested that the time is right to develop activities focused on primary prevention of childhood leukemia.185,186 What Evidence Is Needed Before We Take Action? Historically, there have been many chemicals for which early warning signs of serious health impacts have been ignored due to a lack of scientific consensus. In some cases, protective efforts and corrective actions were not undertaken until many years or even decades after the first trouble was spotted.187 For example, long after initial concerns were raised about DDT, PCBs, and lead, these chemicals continued to be used in large quantities. While these chemicals were eventually banned or restricted in use, in the interim they continued accumulating in the environment, and left a long-term legacy of detrimental exposures that were unnecessary and avoidable. The European Community recognizes the precautionary principle, which provides justification for public policy actions in situations of scientific uncertainly in order to reduce health threats. In contrast, in the U.S., the regulatory framework generally requires scientific consensus of proof of harm before policy actions are carried out. The lack of public health campaigns specifically focused on primary prevention of childhood leukemia may, in part, result from this tendency to avoid taking precautionary actions. In clinical medicine, our mandate to “do no harm” often dictates that we are wary of false positives, for example, when interpreting results from a clinical trial. However, in the context of environmental epidemiology, it is also important to avoid making interpretations that will yield false negatives, because a failure to identify a hazard and take preventive actions poses a threat to the public health. At this time, despite steadily accumulating evidence that environmental exposures increase the risk of childhood leukemia, authoritative bodies, including the International Agency for Research on Cancer (IARC), consider only radiation and parents' active smoking as “causative” factors in the development of childhood leukemia. The Agency reviews individual chemicals only periodically and many of the suspected environmental risk factors for childhood leukemia have not been reviewed in the last decade; a period 341 TEXT BOX 2–The range of evidence suggested* as necessary to validate public health action 1. 2. 3. 4. 5. 6. 7. Animal studies and toxicologic profiles Human studies Systematic structured reviews and meta-analyses Prevalence of exposure to risk factor Severe/dreaded outcomes Risk benefit or cost benefit analysis Likelihood of unintended consequences of potential actions 8. Difficulty of sustaining intervention 9. Co-morbidities also associated with exposure 10. Mechanistic basis for health impact *Adapted with permission from Holman and Buchanan.185 of time during which the environmental epidemiology of childhood leukemia has developed substantially. Clinical medicine has embraced an evidence-based approach and uses systematic reviews (e.g., Cochrane, GRADE) as the gold standard for determining the quality of evidence. The highest quality of evidence (within the evidence-based paradigm) is obtained from double blinded, randomized trials, but this study design would be impossible and unethical in the context of childhood leukemia research on environmental exposures. Prospective cohort studies, which generally are thought to provide high quality of evidence, are prohibitively expensive for a disease like childhood leukemia with an incidence of less than 100 per million population. Even an international effort to combine existing mother–child cohorts leads to small numbers of children diagnosed with leukemia and other cancers. Studies assessing the role of environmental exposures in childhood leukemia would almost exclusively be given a lower quality of evidence rating, given that they are usually, by necessity, observational studies with case–control-designs. Adaptations of the systematic review methodology have been developed to respond to the needs of environmental health and may allow for more precautionary assessments in the future.188,189 Primary prevention of cancer includes reducing exposures to risk factors or changing the underlying conditions which result in disease. While the CDC has been exploring opportunities for early life prevention of child and adult cancers, there are diverse opinions about whether there is an adequate evidence base for primary prevention of cancer.185 In a summary of expert opinion on what evidence should be necessary 342 to support taking action, suggestions range from animal studies and toxicologic profiles to highquality systematic reviews (Text Box 2).185 Addressing Specific Evidence Needs for Action Many of the requirements suggested as a rationale for undertaking primary prevention programs listed in Text Box 2 have already been satisfied in the context of childhood leukemia research. For example, many of the risk factors identified in recent research are common, resulting in widespread exposure to the general population. Moreover, childhood leukemia could certainly be considered “a severe and dreaded outcome.” Though childhood leukemia treatment results in an 80–90% cure rate, the treatment has long-term health implications for those treated, as well as profound impacts on the families and the communities who endure it. While most items on the checklist for taking preventive action (Text Box 2) have been completed, one notable exception is that, to date, animal studies of leukemia have been limited, since it is only recently that animal models of human childhood leukemia have been developed.190 Structured systematic reviews and meta-analyses are viewed as key elements that support the validity of findings from individual studies. Multiple metaanalyses have been conducted on the most highlystudied risk factors for childhood leukemia: pesticide use, tobacco smoke, and traffic-related air pollution. As noted in the Environmental Risk Factors for Childhood Leukemia section, the Childhood Leukemia International Consortium has investigated many of these key risk factors for childhood leukemia using metaanalyses and pooled analysis of original data from case–control studies. These studies have been carefully conducted and can be considered systematic reviews of the epidemiologic literature. Each factor in this issue that has been associated with altered risk of childhood leukemia is also associated with altering the risk of other health outcomes in children as well as adults. In fact, these same exposures that are implicated in the risk for childhood leukemia have substantial documentation of their non-cancer health impacts, including neurobehavioral deficits and respiratory disorders. Thus, acting to prevent childhood leukemia would also reduce the incidence of other diseases, and vice versa. These potential co-benefits should lessen any concerns that an error in attribution Curr Probl PediatrAdolesc Health Care, October 2016 TABLE 3. Examples of exposures associated with altered risk (increased or decreased) for developing childhood leukemia and. co-benefits of improved health outcomes by clinical and public health actions Exposure Health impacts other than childhood leukemia Clinical recommendations Public health activities Integrated Pest Management American Academy of Pediatrics recommended by U.S. EPA and recommends Integrated Pest cooperative extension services Management, exposure reductions. Advocates clinicians become familiar with acute and chronic/subclinical effects and provide anticipatory guidance Tobacco Respiratory disease and asthma, adverse Smoking cessation, avoidance of National and local tobacco control birth outcomes, cardiovascular disease, secondhand smoke universally programs, cessation hotlines adult cancers, neurocognitive disorders, recommended sudden infant death syndrome Many programs for air pollution reduction. Includes preterm birth, decreased birth Less amenable to individual action. Air Pollution Local programs to encourage walking Recommendations to restrict outdoor wt., asthma and respiratory (including and biking. No idle zones, replacement activities during high air pollution days development, cardiovascular dis., traffic related) of old diesel vehicles. School siting (AirNow.gov). Avoid wood fires. neurobehavioral disorders regulations. Fortification of supplementation Inadequate folate early in pregnancy Preconception or prenatal folate Folate (risk recommended by U.S. Preventive associated with neural tube defects, supplementation recommended by reduction) Services Task Force increase in autism risk, other birth American College of Obstetrics and supplementadefects Gynecology and American Academy of tion/healthy Family Physicians and others diet Breastfeeding Sudden infant death syndrome, diarrhea, Promote breastfeeding as the norm, 2011 Surgeon General's Call to Action to (risk reduction) bacteremia, otitis media, childhood develop skills to assess and collaborate Support Breastfeeding: Actions for nonobesity, respiratory infection.204 with obstetrical community and certified governmental organizations, counselors, serve as advocates for government, employers, etc. breastfeeding All states have breastfeeding coalitions, programs to promote and support breastfeeding in minority communities.205 Pesticides Neurobehavioral disorders, asthma, adverse birth outcomes, adult cancer, reproductive toxicity would result in unwarranted actions with potential negative impacts on health or increased financial burdens without benefit to society. Moreover, some high-risk behaviors—such as parental smoking— already have clinical and/or public health recommendations that attempt to alter exposure patterns. Although these efforts are to be applauded, including an additional focus on the potential of reducing childhood leukemia in these existing public health campaigns may improve effectiveness. With the exception of tobacco control and lead poisoning prevention, efforts at wide-scale promotion of children’s environmental health activities have received limited funding and attention. When evaluating the costs and benefits of reducing exposure to an environmental hazard, one must consider the total social and economic impacts for risk reduction associated with all relevant health outcomes (such as cancer, neurobehavioral deficits, and respiratory disorders). Examples of risk factors and protective factors associated with childhood leukemia, other co-morbidities associated with these exposures, and current clinical or public health recommendations relevant to these exposures are presented in Table 3. Curr Probl PediatrAdolesc Health Care, October 2016 Moving Toward Prevention There have been rare instances when the conventional American wisdom—that a determination of causation is required before any action can be taken to protect the public health—has been bypassed to great benefit. For example, in 1964, the U.S. Surgeon General supported action to reduce tobacco use stating Although the causative role of cigarette smoking in deaths from coronary disease is not proven, the Committee considers it more prudent from the public health viewpoint to assume that the established association has causative meaning than to suspend judgment until no uncertainty remains.191 Similarly, the “Back to Sleep” campaign sponsored by the U.S. National Institute of Child Health and Human Development to reduce sudden infant death syndrome (SIDS) is another example of a highly successful public health measure that has saved many lives despite being adopted with less-than-uniform agreement on causation.192 In 1992, the American Academy of Pediatrics issued its policy statement suggesting that infants be placed to sleep on their backs or sides rather than prone.193 At that time there were several case–control studies, but no prospective 343 randomized clinical trials, to support the American Academy of Pediatrics recommendation. Indeed, the American Academy of Pediatrics statement acknowledged many limitations in the epidemiological literature that was available for SIDS at that time. As highlighted in the policy statement abstract This recommendation is made with the full recognition that the existing studies have methodologic limitations and were conducted in countries with infant care practices and other SIDS risk factors that differ from those in the United States (eg, maternal smoking, types of bedding, central heating, etc). taking public health action to reduce human exposure should be considered substantial. Under the American Academy of Pediatrics rubric noted above many of the risk factors in Table 3 should receive the highest rating for evidence quality. Examples of Primary Prevention in Clinical and Public Health Practice Folate As noted in the Dietary Risk Factors for Childhood In fact, the recommendation was in part informed by Leukemia section, adequate folate during the preconception and very early pregnancy periods is not only ecologic studies, which is the study design that associated with reductions in the incidence of childgenerally provides the least convincing support for hood leukemia but it also protects causality. These ecologic studagainst neural tube and other birth ies reported on SIDS inciand possibly dence following large-scale Risk factors for childhood leu- defects autism.195,196 In 1992, the U.S. changes in regional sleep positioning practice; observing kemia are also associated with Public Health Service recomthe risk of developing other that when parents started putmended that all women capable ting children to sleep in a cancers, neurobehavioral defi- of becoming pregnant take prone position the incidence cits, and respiratory disorders. 400 μg of folic acid daily. In 194 of SIDS increased. 1998, the United States introSubseduced fortification of enriched quently, in 1994, the “Back to Sleep” campaign was launched to discourage prone cereal grain products with folate. It has been estimated that folate fortification has resulted in approximately sleeping for infants. By 2000, the U.S. mortality rate 1300 fewer children being born with neural tube for SIDS had dropped to one-half of the rate in 1990. A defects, annually.197 Despite these dramatic results, set of additional recommendations aimed at decreasing SIDS was included in the 2011 American Academy of nearly one quarter of women of childbearing age in the Pediatrics policy statement based on findings of varyNational Health and Nutrition Examination Survey 192 ing scientific rigor. (2007–2012) were found to have suboptimal folateblood levels.198 In addition, Latinas were noted to have In making these policy statements, American Academy of Pediatrics adapted a strength of evidence significantly lower blood folate levels than white scheme from the U.S. Preventive Services Task Force women. Given that Latino children—those living in with a highest level of recommendation that requires Central or South American as well as those living in California—are recognized to be at greater risk of …good and consistent scientific evidence (i.e., there are consistent childhood leukemia than white children, a public findings from at least 2 well-designed, well-conducted case–control studies, a systematic review, or a meta-analysis). There is high certainty health campaign targeting folate supplementation in that the net benefit is substantial, and the conclusion is unlikely to be Latinas of childbearing age could be a particularly strongly affected by the results of future studies. important opportunity for childhood leukemia prevention. Many of the childhood leukemia risk factors listed in A caution would be a concern about the possibility of Table 1 have multiple studies demonstrating their some women exceeding the tolerable upper intake effect and at least one meta-analysis if not a systematic level for folic acid. The National Health and Nutrition review. Moreover, at least for the environmental agents Examination Survey estimated that 2.7% of particithat have been assessed via the comprehensive, pooled pants (primarily women) exceed these recommended analyses conducted by the Childhood Leukemia Interfolate values.199 Nonetheless, a careful emphasis on national Consortium, it is unlikely that future studies will contradict the current findings. Finally, when one improving nutritional status by ensuring fresh foods takes into account the multiple other diseases that are and folic acid supplementation for women of pregalso associated with these exposures, the benefit of nancy age seems warranted. 344 Curr Probl PediatrAdolesc Health Care, October 2016 Tobacco Although tobacco control programs and policies have significantly reduced smoking, there are still between 15% and 23% of young adult men and women who smoke in the U.S.200 It is likely that most are not aware of the link between paternal smoking around the time of conception and a child’s subsequent risk of developing leukemia. To prevent soon-to-be-dads from smoking before conception, fuller implementation of proven tobacco control strategies aimed at preventing smoking initiation and reducing smoking in teenagers and young adults is needed. Pesticides Pesticide exposure, both prenatal and postnatal, has been associated with leukemia risk. This supports introduction of programs focused on the use of safe pesticide practices during the preconception period. An example of materials that target an audience of people who are considering becoming, or currently are, pregnant is available from the Program for Reproductive Health and the Environment at the University of California, San Francisco. These include Pesticides Matter: Steps to Reduce Exposure and Protect Your Health (http://prhe.ucsf.edu/prhe/pesticidesmatter.html). Preconception Care Recently, reproductive health professionals involved in the care and support of women of childbearing age have begun to emphasize preconception care. These professionals are interested in including environmental factors as part of their message to their patients about health promotion before conception and during pregnancy.201 Including both men and women in health promotion provides an ideal opportunity to address environmental exposures prior to early critical windows of development, even before conception. Initial studies on the effectiveness of strategies to promote behavior change to reduce chemical hazard exposures preconception and during pregnancy indicate that perceived normative pressure (perception of what is common among peers and important to family, friends and doctors) is a key element.202 The U.S. Centers for Disease Control and Prevention and the American College of Obstetrics and Gynecology have begun programs to address preconception health (http://www.cdc.gov/preconcep tion/index.html, see Preconception Health and Health Care). An effort should be made to ensure that Curr Probl PediatrAdolesc Health Care, October 2016 environmental health literacy and childhood cancer prevention activities are addressed within the context of these evolving programs and included in a new standard of care. The cancer prevention activities suggested by our research on childhood leukemia and others203 largely reinforce programs already in existence by different public health entities (e.g., tobacco cessation, healthy diet during pregnancy and folate supplementation, avoidance of exposure to volatile organic compounds, pesticides, and paint). Public health agencies and health care systems can partner to extend current programs with similar goals. Innovative cross-agency, multidisciplinary program opportunities include Enhance the use of news media and add risk reduction for childhood leukemia to environmental health literacy education activities for the general public, as appropriate. Include environmental health messaging in preconception, prenatal, and child preventive health care and as part of programs such as “Text 4 Baby” and “Bright Futures.” Assure that preconception health programs include environmental health components and additional emphasis on healthy diet. Use emerging systematic review methodologies to evaluate environmental health risks with the goal of having environmental health programs recognized as evidence-based medicine. Improve health care provider counseling for women and couples to integrate concepts of environmental health—specifically the risk factors associated with childhood leukemia—into medical education at all levels (during schooling and post-graduate education) for nurses, physicians, and allied health professionals. Develop policy initiatives to reduce exposures to chemicals associated with childhood leukemia (and other negative health outcomes) during prepregnancy, pregnancy, and early childhood. The currently available toxicologic and observational epidemiologic studies (including meta and pooled analyses) provide a strong evidentiary basis for the presence of casual associations (of small to moderate sizes) between several environmental exposures and childhood leukemia. Awaiting a more complete evidentiary basis for decision-making, though ideal, will 345 result in significant delays in safeguarding children’s health. Education of clinicians and the public on primary prevention—actions that an individual can take to reduce their own family’s exposures to chemical risk factors for childhood leukemia as well as other disorders—is something that can occur now. Ultimately, regulatory actions based on the evolving science are needed to shift the burden from the individual to producers. References 1. American Cancer Society. Cancer Facts & Figures 2016. http://www.cancer.org/acs/groups/content/@research/docu ments/document/acspc-047079.pdf. 2016 Accessed 17.06.16. 2. Madhusoodhan PP, Carroll WL, Bhatla T. Progress and prospects in pediatric leukemia. Curr Probl Pediatr Adolesc Health Care 2016;46(7):229–41. 3. Nathan PC, Wasilewski-Masker K, Janzen LA. Long-term outcomes in survivors of childhood acute lymphoblastic leukemia. Hematol Oncol Clin North Am 2009;23(5): 1065–82:vi-vii. 4. Iyer NS, Balsamo LM, Bracken MB, Kadan-Lottick NS. Chemotherapy-only treatment effects on long-term neurocognitive functioning in childhood ALL survivors: a review and meta-analysis. Blood 2015;126(3):346–53. 5. Curtin K, Smith KR, Fraser A, Pimentel R, Kohlmann W, Schiffman JD. Familial risk of childhood cancer and tumors in the Li-Fraumeni spectrum in the Utah Population Database: implications for genetic evaluation in pediatric practice. Int J Cancer 2013;133(10):2444–53. 6. Wiemels J. Perspectives on the causes of childhood leukemia. Chem Biol Interact 2012;196(3):59–67. 7. Giddings B, Whitehead TP, Metayer C, Miller MD. Childhood leukemia incidence in California: high and rising in the Hispanic population. Cancer 2016:(In press). 8. Vlaanderen J, Portengen L, Rothman N, Lan Q, Kromhout H, Vermeulen R. Flexible meta-regression to assess the shape of the benzene-leukemia exposure–response curve. Environ Health Perspect 2010;118(4):526–32. 9. Lanphear BP, Hornung R, Khoury J, et al. Low-level environmental lead exposure and children’s intellectual function: an international pooled analysis. Environ Health Perspect 2005;113(7):894–9. 10. Zachek CM, Miller MD, Hsu C, et al. Children’s Cancer and Environmental Exposures: Professional Attitudes and Practices. J Pediatr Hematol Oncol 2015;37(7):491–7. 11. Hanahan D, Weinberg RA. The hallmarks of cancer. Cell 2000;100(1):57–70. 12. Wiemels JL. Chromosomal translocations in childhood leukemia: natural history, mechanisms, and epidemiology. J Natl Cancer Inst Monogr 2016:(In press). 13. McHale CM, Wiemels JL, Zhang L, et al. Prenatal origin of childhood acute myeloid leukemias harboring chromosomal rearrangements t(15;17) and inv(16). Blood 2003;101(11): 4640–1. 346 14. McHale CM, Wiemels JL, Zhang L, et al. Prenatal origin of TEL-AML1-positive acute lymphoblastic leukemia in children born in California. Genes Chromosomes Cancer 2003; 37(1):36–43. 15. Wiemels JL, Cazzaniga G, Daniotti M, et al. Prenatal origin of acute lymphoblastic leukaemia in children. Lancet 1999;354(9189):1499–503. 16. Wiemels JL, Xiao Z, Buffler PA, et al. In utero origin of t(8;21) AML1-ETO translocations in childhood acute myeloid leukemia. Blood 2002;99(10):3801–5. 17. Chang P, Kang M, Xiao A, et al. FLT3 mutation incidence and timing of origin in a population case series of pediatric leukemia. BMC Cancer 2010;10:513. 18. Wiemels JL, Kang M, Chang JS, et al. Backtracking RAS mutations in high hyperdiploid childhood acute lymphoblastic leukemia. Blood Cells Mol Dis. 2010;45(3):186–91. 19. Wiemels JL, Leonard BC, Wang Y, et al. Site-specific translocation and evidence of postnatal origin of the t(1;19) E2A-PBX1 fusion in childhood acute lymphoblastic leukemia. Proc Natl Acad Sci U S A 2002;99(23):15101–6. 20. Mori H, Colman SM, Xiao Z, et al. Chromosome translocations and covert leukemic clones are generated during normal fetal development. Proc Natl Acad Sci U S A 2002; 99(12):8242–7. 21. Zuna J, Ford AM, Peham M, et al. TEL deletion analysis supports a novel view of relapse in childhood acute lymphoblastic leukemia. Clin Cancer Res 2004;10(16):5355–60. 22. Tsai AG, Lieber MR. Mechanisms of chromosomal rearrangement in the human genome. BMC Genomics 2010; 11(suppl 1):S1. 23. Papaemmanuil E, Hosking FJ, Vijayakrishnan J, et al. Loci on 7p12.2, 10q21.2 and 14q11.2 are associated with risk of childhood acute lymphoblastic leukemia. Nat Genet 2009;41(9): 1006–1010. 24. Pluth JM, Nicklas JA, O’Neill JP, Albertini RJ. Increased frequency of specific genomic deletions resulting from in vitro malathion exposure. Cancer Res 1996;56(10):2393–9. 25. Smith MT, Skibola CF, Allan JM, Morgan GJ. Causal models of leukaemia and lymphoma. IARC Sci Publ 2004;157: 373–92. 26. Kaur M, de Smith AJ, Selvin S, et al. Tobacco smoke and Ras mutations among Latino and non-Latino children with acute lymphoblastic leukemia. Arch Med Res 2016:(Invited submission, in preparation). 27. Metayer C, Zhang L, Wiemels JL, et al. Tobacco smoke exposure and the risk of childhood acute lymphoblastic and myeloid leukemias by cytogenetic subtype. Cancer Epidemiol Biomarkers Prev 2013;22(9):1600–11. 28. Scelo G, Metayer C, Zhang L, et al. Household exposure to paint and petroleum solvents, chromosomal translocations, and the risk of childhood leukemia. Environ Health Perspect 2009;117(1):133–9. 29. Bailey HD, Infante-Rivard C, Metayer C, et al. Home pesticide exposures and risk of childhood leukemia: findings from the childhood leukemia international consortium. Int J Cancer 2015;137(11):2644–63. 30. Barker DJ. The origins of the developmental origins theory. J Intern Med 2007;261(5):412–7. Curr Probl PediatrAdolesc Health Care, October 2016 31. Joubert BR, Felix JF, Yousefi P, et al. DNA methylation in newborns and maternal smoking in pregnancy: genome-wide consortium meta-analysis. Am J Hum Genet 2016;98(4): 680–96. 32. Gonseth S, Roy R, Houseman EA, et al. Periconceptional folate consumption is associated with neonatal DNA methylation modifications in neural crest regulatory and cancer development genes. Epigenetics 2015;10(12):1166–76. 33. Rudant J, Lightfoot T, Urayama K, et al. Childhood acute lymphoblastic leukemia and indicators of early immune stimulation: a Childhood Leukemia International Consortium (CLIC) Study. Am J Epidemiol 2016: (In press). 34. Greaves MF, Alexander FE. An infectious etiology for common acute lymphoblastic leukemia in childhood? Leukemia 1993;7(3):349–60. 35. Kinlen LJ. Epidemiological evidence for an infective basis in childhood leukaemia [editorial]. Br J Cancer 1995;71(1):1–5. 36. Crouch S, Lightfoot T, Simpson J, Smith A, Ansell P, Roman E. Infectious illness in children subsequently diagnosed with acute lymphoblastic leukemia: modeling the trends from birth to diagnosis. Am J Epidemiol 2012;176(5):402–8. 37. Chang JS, Tsai CR, Tsai YW, Wiemels JL. Medically diagnosed infections and risk of childhood leukaemia: a population-based case–control study. Int J Epidemiol 2012;41(4):1050–9. 38. Chang JS, Zhou M, Buffler PA, Chokkalingam AP, Metayer C, Wiemels JL. Profound deficit of IL10 at birth in children who develop childhood acute lymphoblastic leukemia. Cancer Epidemiol Biomarkers Prev 2011;20(8):1736–40. 39. Smith MT, Guyton KZ, Gibbons CF, et al. Key Characteristics of Carcinogens as a Basis for Organizing Data on Mechanisms of Carcinogenesis. Environ Health Perspect 2016;124(6):713–21. 40. Infante-Rivard C, Jacques L. Empirical study of parental recall bias. Am J Epidemiol 2000;152(5):480–6. 41. Preston-Martin S, Bernstein L, Maldonado AA, Henderson BE, White SC. A dental x-ray validation study. Comparison of information from patient interviews and dental charts. Am J Epidemiol 1985;121(3):430–9. 42. Berrington de Gonzalez A, Ekbom A, Glass AG, et al. Comparison of documented and recalled histories of exposure to diagnostic x-rays in case–control studies of thyroid cancer. Am J Epidemiol 2003;157(7):652–63. 43. Slusky DA, Metayer C, Aldrich MC, et al. Reliability of maternal-reports regarding the use of household pesticides: experience from a case–control study of childhood leukemia. Cancer Epidemiol 2012;36(4):375–80. 44. Roberts JW, Wallace LA, Camann DE, et al. Monitoring and reducing exposure of infants to pollutants in house dust. Rev Environ Contam Toxicol 2009;201:1–39. 45. Whitehead TP, Brown FR, Metayer C, et al. Polychlorinated biphenyls in residential dust: sources of variability. Environ Sci Technol 2014;48(1):157–64. 46. Whitehead TP, Brown FR, Metayer C, et al. Polybrominated diphenyl ethers in residential dust: sources of variability. Environ Int 2013;57–58:11–24. Curr Probl PediatrAdolesc Health Care, October 2016 47. Whitehead TP, Metayer C, Petreas M, Does M, Buffler PA, Rappaport SM. Polycyclic aromatic hydrocarbons in residential dust: sources of variability. Environ Health Perspect 2013;121(5):543–50. 48. Lorber M. Exposure of Americans to polybrominated diphenyl ethers. J Expo Sci Environ Epidemiol 2008;18(1):2–19. 49. Johnson PI, Stapleton HM, Sjodin A, Meeker JD. Relationships between polybrominated diphenyl ether concentrations in house dust and serum. Environ Sci Technol 2010;44(14): 5627–5632. 50. Watkins DJ, McClean MD, Fraser AJ, et al. Impact of dust from multiple microenvironments and diet on PentaBDE body burden. Environ Sci Techol 2012;46(2):1192–200. 51. Johnson-Restrepo B, Kannan K. An assessment of sources and pathways of human exposure to polybrominated diphenyl ethers in the United States. Chemosphere 2009;76(4):542–8. 52. Stapleton HM, Eagle S, Sjodin A, Webster TF. Serum PBDEs in a North Carolina toddler cohort: associations with handwipes, house dust, and socioeconomic variables. Environ Health Perspect 2012;120(7):1049–54. 53. Whitehead TP, Crispo-Smith SM, Park JS, Petreas M, Rappaport SM, Metayer C. Concentrations of persistent organic pollutants in California women’s serum and vacuum dust. Environ Res 2014;136:57–66. 54. Whitehead T, Crispo Smith SM, Park JS, Petreas M, Rappaport S, Metayer C. Concentrations of persistent organic pollutants in California children’s whole blood and residential dust. Chemosphere 2016:(Submitted for publication). 55. Rull RP, Gunier R, Von Behren J, et al. Residential proximity to agricultural pesticide applications and childhood acute lymphoblastic leukemia. Environ Res 2009;109(7) 891–9. 56. Gunier RB, Ward MH, Airola M, et al. Determinants of agricultural pesticide concentrations in carpet dust. Environ Health Perspect 2011;119(7):970–6. 57. Von Behren J, Reynolds P, Gunier RB, et al. Residential traffic density and childhood leukemia risk. Cancer Epidemiol Biomarkers Prev 2008;17(9):2298–301. 58. Slusky DA, Does M, Metayer C, Mezei G, Selvin S, Buffler PA. Potential role of selection bias in the association between childhood leukemia and residential magnetic fields exposure: a population-based assessment. Cancer Epidemiol 2014 307–13. 59. Wallace LA, Pellizzari ED, Hartwell TD, et al. The TEAM (Total Exposure Assessment Methodology) Study: personal exposures to toxic substances in air, drinking water, and breath of 400 residents of New Jersey, North Carolina, and North Dakota. Environ Res 1987;43(2):290–307. 60. Adgate JL, Eberly LE, Stroebel C, Pellizzari ED, Sexton K. Personal, indoor, and outdoor VOC exposures in a probability sample of children. J Expo Anal Environ Epidemiol 2004; 14(suppl 1):S4–13. 61. Brown RC, Dwyer T, Kasten C, et al. Cohort profile: the International Childhood Cancer Cohort Consortium (I4C). Int J Epidemiol 2007;36(4):724–30. 62. Funk WE, Waidyanatha S, Chaing SH, Rappaport SM. Hemoglobin adducts of benzene oxide in neonatal and adult 347 63. 64. 65. 66. 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 348 dried blood spots. Cancer Epidemiol Biomarkers Prev 2008;17(8):1896–901. Chokkalingam AP, Chun DS, Noonan EJ, et al. Blood levels of folate at birth and risk of childhood leukemia. Cancer Epidemiol Biomarkers Prev 2013;22(6):1088–94. Spector LG, Murphy SE, Wickham KM, Lindgren B, Joseph AM. Prenatal tobacco exposure and cotinine in newborn dried blood spots. Pediatrics 2014;133(6):e1632–8. Ma WL, Yun S, Bell EM, et al. Temporal trends of polybrominated diphenyl ethers (PBDEs) in the blood of newborns from New York State during 1997 through 2011: analysis of dried blood spots from the newborn screening program. Environ Sci Technol 2013;47(14):8015–21. Wagner M, Tonoli D, Varesio E, Hopfgartner G. The use of mass spectrometry to analyze dried blood spots. Mass Spectrom Rev 2016;35(3):361–438. Ma X, Buffler PA, Gunier RB, et al. Critical windows of exposure to household pesticides and risk of childhood leukemia. Environ Health Perspect 2002;110(9):955–60. Turner MC, Wigle DT, Krewski D. Residential pesticides and childhood leukemia: a systematic review and meta-analysis. Environ Health Perspect 2010;118(1):33–41. Van Maele-Fabry G, Lantin AC, Hoet P, Lison D. Residential exposure to pesticides and childhood leukaemia: a systematic review and meta-analysis. Environ Int 2011;37(1):280–91. Chen M, Chang CH, Tao L, Lu C. Residential exposure to pesticide during childhood and childhood cancers: a metaanalysis. Pediatrics 2015;136(4):719–29. Curl CL, Fenske RA, Kissel JC, et al. Evaluation of takehome organophosphorus pesticide exposure among agricultural workers and their children. Environ Health Perspect 2002;110(12):A787–92. Coronado GD, Vigoren EM, Thompson B, Griffith WC, Faustman EM. Organophosphate pesticide exposure and work in pome fruit: evidence for the take-home pesticide pathway. Environ Health Perspect 2006;114(7):999–1006. Ostrea EM Jr, Bielawski DM, Posecion NC Jr, et al. Combined analysis of prenatal (maternal hair and blood) and neonatal (infant hair, cord blood and meconium) matrices to detect fetal exposure to environmental pesticides. Environ Res 2009;109(1):116–22. Bailey HD, Fritschi L, Infante-Rivard C, et al. Parental occupational pesticide exposure and the risk of childhood leukemia in the offspring: findings from the childhood leukemia international consortium. Int J Cancer 2014;135(9): 2157–72. Alexander FE, Patheal SL, Biondi A, et al. Transplacental chemical exposure and risk of infant leukemia with MLL gene fusion. Cancer Res 2001;61(6):2542–6. Buckley JD, Robison LL, Swotinsky R, et al. Occupational exposures of parents of children with acute nonlymphocytic leukemia: a report from the Childrens Cancer Study Group. Cancer Res 1989;49(14):4030–7. Ferreira JD, Couto AC, Pombo-de-Oliveira MS, Koifman S, Brazilian Collaborative Study Group of Infant Acute Leukemia. . In utero pesticide exposure and leukemia in Brazilian children o 2 years of age. Environ Health Perspect 2013; 121(2):269–75. 78. Wigle DT, Turner MC, Krewski D. A systematic review and meta-analysis of childhood leukemia and parental occupational pesticide exposure. Environ Health Perspect 2009;117 (10):1505–13. 79. Van Maele-Fabry G, Lantin AC, Hoet P, Lison D. Childhood leukaemia and parental occupational exposure to pesticides: a systematic review and meta-analysis. Cancer Causes Control 2010;21(6):787–809. 80. Metayer C, Colt JS, Buffler PA, et al. Exposure to herbicides in house dust and risk of childhood acute lymphoblastic leukemia. J Expo Sci Environ Epidemiol 2013;23(4): 363–70. 81. Ward MH, Colt JS, Metayer C, et al. Residential exposure to polychlorinated biphenyls and organochlorine pesticides and risk of childhood leukemia. Environ Health Perspect 2009;117(6):1007–13. 82. Chang JS, Selvin S, Metayer C, Crouse V, Golembesky A, Buffler PA. Parental smoking and the risk of childhood leukemia. Am J Epidemiol 2006;163(12):1091–100. 83. Metayer C, Petridou E, Mejia Arangure JM, et al. Parental Tobacco Smoking and Acute Myeloid Leukemia in Children: Pooled and Meta-analyses from the Childhood Leukemia International Consortium. Am J Epidemiol 2016:(In press). 84. Chang JS. Parental smoking and childhood leukemia. Methods Mol Biol 2009;472:103–37. 85. Liu R, Zhang L, McHale CM, Hammond SK. Paternal smoking and risk of childhood acute lymphoblastic leukemia: systematic review and meta-analysis. J Oncol 2011;2011:854584. 86. Bailey H, Metayer C, Milne E, et al. Home paint exposures and risk of childhood acute lymphoblastic leukemia: findings from the Childhood Leukemia International Consortium. Cancer Causes Control 2015;26(9):1257–70. 87. Colt JS, Blair A. Parental occupational exposures and risk of childhood cancer. Environ Health Perspect 1998;106(suppl 3):909–25. 88. Zhou Y, Zhang S, Li Z, et al. Maternal benzene exposure during pregnancy and risk of childhood acute lymphoblastic leukemia: a meta-analysis of epidemiologic studies. PloS One 2014;9(10):e110466. 89. Bailey HD, Fritschi L, Metayer C, et al. Parental occupational paint exposure and risk of childhood leukemia in the offspring: findings from the Childhood Leukemia International Consortium. Cancer Causes Control 2014;25(10):1351–67. 90. Carlos-Wallace FM, Zhang L, Smith MT, Rader G, Steinmaus C. Parental, in utero, and early-life exposure to benzene and the risk of childhood leukemia: a meta-analysis. Am J Epidemiol 2016;183(1):1–14. 91. Metayer C, Scelo G, Kang A, et al. A task-based assessment of parental occupational exposure to organic solvents and other compounds and the risk of childhood leukemias in California. Environ Res 2016:(In press). 92. Boothe VL, Boehmer TK, Wendel AM, Yip FY. Residential traffic exposure and childhood leukemia: a systematic review and meta-analysis. Am J Prev Med 2014;46(4):413–22. 93. Filippini T, Heck JE, Malagoli C, Del Giovane C, Vinceti M. A review and meta-analysis of outdoor air pollution and risk of childhood leukemia. J Environ Sci Health C Environ Carcinog Ecotoxicol Rev 2015;33(1):36–66. Curr Probl PediatrAdolesc Health Care, October 2016 94. Deziel NC, Rull RP, Colt JS, et al. Polycyclic aromatic hydrocarbons in residential dust and risk of childhood acute lymphoblastic leukemia. Environ Res 2014;133:388–95. 95. Ward MH, Colt JS, Deziel NC, et al. Residential levels of polybrominated diphenyl ethers and risk of childhood acute lymphoblastic leukemia in california. Environ Health Perspect 2014;122(10):1110–6. 96. Birnbaum LS, Staskal DF. Brominated flame retardants: cause for concern? Environ Health Perspect 2004;112(1): 9–17. 97. Linet MS, Kim KP, Rajaraman P. Children’s exposure to diagnostic medical radiation and cancer risk: epidemiologic and dosimetric considerations. Pediatric Radiol 2009;39 (suppl 1):S4–26. 98. Smith-Bindman R, Miglioretti DL, Johnson E, et al. Use of diagnostic imaging studies and associated radiation exposure for patients enrolled in large integrated health care systems, 1996–2010. J Am Med Assoc 2012;307(22): 2400–2409. 99. Linet MS, Slovis TL, Miller DL, et al. Cancer risks associated with external radiation from diagnostic imaging procedures. CA Cancer J Clin 2012. 100. Brenner DJ, Hall EJ. Computed tomography—an increasing source of radiation exposure. N Engl J Med 2007;357(22): 2277–84. 101. Journy N, Ancelet S, Rehel JL, et al. Predicted cancer risks induced by computed tomography examinations during childhood, by a quantitative risk assessment approach. Radiat Environ Biophys 2014;53(1):39–54. 102. Miglioretti DL, Johnson E, Williams A, et al. The use of computed tomography in pediatrics and the associated radiation exposure and estimated cancer risk. JAMA Pediatr 2013;167(8):700–7. 103. Bailey HD, Armstrong BK, de Klerk NH, et al. Exposure to diagnostic radiological procedures and the risk of childhood acute lymphoblastic leukemia. Cancer Epidemiol Biomarkers Prev 2010;19(11):2897–909. 104. Krille L, Jahnen A, Mildenberger P, et al. Computed tomography in children: multicenter cohort study design for the evaluation of cancer risk. Eur J Epidemiol 2011;26 (3):249–50. 105. Meulepas JM, Ronckers CM, Smets AM, et al. Leukemia and brain tumors among children after radiation exposure from CT scans: design and methodological opportunities of the Dutch Pediatric CT Study. Eur J Epidemiol 2014;29(4): 293–301. 106. Pearce MS, Salotti JA, Little MP, et al. Radiation exposure from CT scans in childhood and subsequent risk of leukaemia and brain tumours: a retrospective cohort study. Lancet 2012;380(9840):499–505. 107. Mathews JD, Forsythe AV, Brady Z, et al. Cancer risk in 680,000 people exposed to computed tomography scans in childhood or adolescence: data linkage study of 11 million Australians. Br Med J 2013;346:f2360. 108. Einstein AJ. Beyond the bombs: cancer risks of low-dose medical radiation. Lancet 2012;380(9840):455–7. 109. Journy N, Rehel JL, Ducou Le Pointe H, et al. Are the studies on cancer risk from CT scans biased by indication? Elements Curr Probl PediatrAdolesc Health Care, October 2016 110. 111. 112. 113. 114. 115. 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. of answer from a large-scale cohort study in France Br J Cancer 2015;112(1):185–93. Knox EG, Stewart AM, Kneale GW, Gilman EA. Prenatal irradiation and childhood cancer. J Soc Radiol 1987;7(4): 177–87. Cardis E, de Basea MB. Comment on ‘Are the studies on cancer risk from CT scans biased by indication? Elements of answer from a large-scale cohort study in France’—evidence of confounding by predisposing factors unclear Br J Cancer 2015;112(11):1842–3. Journy N, Laurier D, Bernier MO. Comment on: are the studies on cancer risk from CT scans biased by indication? Elements of answer from a large-scale cohort study in France Br J Cancer 2015;112(11):1843–4. Muirhead CR. Response to ‘Are the studies on cancer risk from CT scans biased by indication? Elements of answer from a large-scale cohort study in France’ Br J Cancer. 2015;112(11):1841–2. Ahlbom A, Day N, Feychting M, et al. A pooled analysis of magnetic fields and childhood leukaemia. Br J Cancer 2000;83(5):692–8. Greenland S, Sheppard AR, Kaune WT, Poole C, Kelsh MA. A pooled analysis of magnetic fields, wire codes, and childhood leukemia. Childhood Leukemia-EMF Study Group. Epidemiology 2000;11(6):624–34. IARC. Non-Ionizing Radiation: I, Static and Extremely LowFrequency (ELF) Elctric and Magnetic Fields. Lyon, France: International Agency for Research on Cancer, 2002. Schuz J, Lagorio S, Bersani F. Electromagnetic fields and epidemiology: an overview inspired by the fourth course at the International School of Bioelectromagnetics. Bioelectromagnetics 2009;30(7):511–24. Teepen JC, van Dijck JA. Impact of high electromagnetic field levels on childhood leukemia incidence. Int J Cancer 2012;131(4):769–78. Mosby TT, Cosgrove M, Sarkardei S, Platt KL, Kaina B. Nutrition in adult and childhood cancer: role of carcinogens and anti-carcinogens. Anticancer Res 2012;32(10):4171–92. Ferguson LR, Philpott M. Nutrition and mutagenesis. Annu Rev Nutr 2008;28:313–29. Forster SE, Powers HJ, Foulds GA, et al. Improvement in nutritional status reduces the clinical impact of infections in older adults. J Am Geriatr Soc 2012;60(9):1645–54. Gibson A, Edgar JD, Neville CE, et al. Effect of fruit and vegetable consumption on immune function in older people: a randomized controlled trial. Am J Clinl Nutr 2012;96(6): 1429–36. Stefanska B, Karlic H, Varga F, Fabianowska-Majewska K, Haslberger A. Epigenetic mechanisms in anti-cancer actions of bioactive food components—the implications in cancer prevention. Br J Pharmacol 2012;167(2):279–97. Barker DJ. Sir Richard Doll Lecture. Developmental origins of chronic disease. Public Health 2012;126(3):185–9. Waterland RA, Michels KB. Epigenetic epidemiology of the developmental origins hypothesis. Annual Rev Nutr 2007; 27:363–88. Jensen CD, Block G, Buffler P, Ma X, Selvin S, Month S. Maternal dietary risk factors in childhood acute 349 127. 128. 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. 142. 350 lymphoblastic leukemia (United States). Cancer Causes Control 2004;15(6):559–70. Kwan ML, Jensen CD, Block G, Hudes ML, Chu LW, Buffler PA. Maternal diet and risk of childhood acute lymphoblastic leukemia. Public Health Rep 2009;124(4): 503–14. Petridou E, Ntouvelis E, Dessypris N, Terzidis A, Trichopoulos D, Childhood Hematology-Oncology Group. Maternal diet and acute lymphoblastic leukemia in young children. Cancer Epidemiol Biomarkers Prev 2005;14(8):1935–9. Spector LG, Xie Y, Robison LL, et al. Maternal diet and infant leukemia: the DNA topoisomerase II inhibitor hypothesis: a report from the children’s oncology group. Cancer Epidemiol Biomarkers Prev 2005;14(3):651–5. Locasale JW. Serine, glycine and one-carbon units: cancer metabolism in full circle. Nat Rev Cancer 2013;13(8): 572–83. Lim U, Song MA. Dietary and lifestyle factors of DNA methylation. Methods Mol Biol 2012;863:359–76. Blom HJ, Shaw GM, den Heijer M, Finnell RH. Neural tube defects and folate: case far from closed. Nat Rev Neurosci 2006;7(9):724–31. Chen P, Li C, Li X, Li J, Chu R, Wang H. Higher dietary folate intake reduces the breast cancer risk: a systematic review and meta-analysis. Br J Cancer 2014;110(9):2327–38. Kennedy DA, Stern SJ, Moretti M, et al. Folate intake and the risk of colorectal cancer: a systematic review and metaanalysis. Cancer Epidemiol 2011;35(1):2–10. Wien TN, Pike E, Wisloff T, Staff A, Smeland S, Klemp M. Cancer risk with folic acid supplements: a systematic review and meta-analysis. BMJ Open 2012;2(1):e000653. Clarke R, Halsey J, Lewington S, et al. Effects of lowering homocysteine levels with B vitamins on cardiovascular disease, cancer, and cause-specific mortality: meta-analysis of 8 randomized trials involving 37 485 individuals. Arch Intern Med 2010;170(18):1622–31. Vollset SE, Clarke R, Lewington S, et al. Effects of folic acid supplementation on overall and site-specific cancer incidence during the randomised trials: meta-analyses of data on 50,000 individuals. Lancet 2013;381(9871):1029–36. Metayer C, Milne E, Dockerty JD, et al. Maternal supplementation with folic acid and other vitamins and risk of leukemia in offspring: a childhood leukemia international consortium study. Epidemiology 2014;25(6):811–22. Bailey HD, Miller M, Langridge A, et al. Maternal dietary intake of folate and vitamins b6 and B12 during pregnancy and the risk of childhood acute lymphoblastic leukemia. Nutr Cancer 2012;64(7):1122–30. Metayer C, Scelo G, Chokkalingam AP, et al. Genetic variants in the folate pathway and risk of childhood acute lymphoblastic leukemia. Cancer Causes Control 2011;22(9):1243–58. Singer AW, Selvin S, Block G, Golden C, Carmichael SL, Metayer C. Maternal prenatal intake of one-carbon metabolism nutrients and risk of childhood leukemia. Cancer Causes Control 2016;27(7):929–40. Hay G, Clausen T, Whitelaw A, et al. Maternal folate and cobalamin status predicts vitamin status in newborns and 6-month-old infants. J Nutr 2010;140(3):557–64. 143. Ajrouche R, Rudant J, Orsi L, et al. Childhood acute lymphoblastic leukaemia and indicators of early immune stimulation: the Estelle study (SFCE). Br J Cancer 2015; 112(6):1017–106. 144. Amigou A, Rudant J, Orsi L, et al. Folic acid supplementation, MTHFR and MTRR polymorphisms, and the risk of childhood leukemia: the ESCALE study (SFCE). Cancer Causes Control 2012;23(8):1265–77. 145. Koppen IJ, Hermans FJ, Kaspers GJ. Folate related gene polymorphisms and susceptibility to develop childhood acute lymphoblastic leukaemia. Br J Haematol 2010;148(1):3–14. 146. Lightfoot TJ, Johnston WT, Painter D, et al. Genetic variation in the folate metabolic pathway and risk of childhood leukemia. Blood 2010;115(19):3923–9. 147. Wong NC, Ashley D, Chatterton Z, et al. A distinct DNA methylation signature defines pediatric pre-B cell acute lymphoblastic leukemia. Epigenetics 2012;7(6):535–41. 148. Lupo PJ, Dietz DJ, Kamdar KY, Scheurer ME. Geneenvironment interactions and the risk of childhood acute lymphoblastic leukemia: exploring the role of maternal folate genes and folic Acid fortification. Pediatr Hematol Oncol 2014;31(2):160–8. 149. Milne E, Greenop KR, Scott RJ, et al. Folate pathway gene polymorphisms, maternal folic acid use, and risk of childhood acute lymphoblastic leukemia. Cancer Epidemiol Biomarkers Prev 2015;24(1):48–56. 150. Mosad E, Abdou M, Zaky AH. Rearrangement of the myeloid/lymphoid leukemia gene in therapy-related myelodysplastic syndrome in patients previously treated with agents targeting DNA topoisomerase II. Oncology 2012;83(3): 128–34. 151. Ross JA, Potter JD, Reaman GH, Pendergrass TW, Robison LL. Maternal exposure to potential inhibitors of DNA topoisomerase II and infant leukemia (United States): a report from the Children’s Cancer Group. Cancer Causes Control 1996;7(6):581–90. 152. Lightfoot TJ, Roman E. Causes of childhood leukaemia and lymphoma. Toxicol Appl Pharmacol 2004;199(2):104–17. 153. Lanoue L, Green KK, Kwik-Uribe C, Keen CL. Dietary factors and the risk for acute infant leukemia: evaluating the effects of cocoa-derived flavanols on DNA topoisomerase activity. Exp Biol Med 2010;235(1):77–89. 154. Menegaux F, Steffen C, Bellec S, et al. Maternal coffee and alcohol consumption during pregnancy, parental smoking and risk of childhood acute leukaemia. Cancer Detect Prev 2005; 29(6):487–93. 155. Thomopoulos TP, Ntouvelis E, Diamantaras AA, et al. Maternal and childhood consumption of coffee, tea and cola beverages in association with childhood leukemia: a metaanalysis. Cancer Epidemiol 2015;39(6):1047–59. 156. Bonaventure A, Rudant J, Goujon-Bellec S, et al. Childhood acute leukemia, maternal beverage intake during pregnancy, and metabolic polymorphisms. Cancer Causes Control 2013; 24(4):783–93. 157. Milne E, Royle JA, Bennett LC, et al. Maternal consumption of coffee and tea during pregnancy and risk of childhood ALL: results from an Australian case–control study. Cancer Causes Control 2011;22(2)207–18. Curr Probl PediatrAdolesc Health Care, October 2016 158. Shu XO, Ross JA, Pendergrass TW, Reaman GH, Lampkin B, Robison LL. Parental alcohol consumption, cigarette smoking, and risk of infant leukemia: a Childrens Cancer Group study. J Natl Cancer Inst 1996;88(1):24–31. 159. Latino-Martel P, Chan DS, Druesne-Pecollo N, Barrandon E, Hercberg S, Norat T. Maternal alcohol consumption during pregnancy and risk of childhood leukemia: systematic review and meta-analysis. Cancer Epidemiol Biomarkers Prev 2010;19(5):1238–60. 160. Infante-Rivard C, Krajinovic M, Labuda D, Sinnett D. Childhood acute lymphoblastic leukemia associated with parental alcohol consumption and polymorphisms of carcinogen-metabolizing genes. Epidemiology 2002;13(3): 277–81. 161. Petridou E, Trichopoulos D, Kalapothaki V, et al. The risk profile of childhood leukaemia in Greece: a nationwide case– control study. Br J Cancer 1997;76(9):1241–7. 162. Orsi L, Rudant J, Ajrouche R, et al. Parental smoking, maternal alcohol, coffee and tea consumption during pregnancy, and childhood acute leukemia: the ESTELLE study. Cancer Causes Control 2015;26(7):1003–17. 163. Milne E, Greenop KR, Scott RJ, et al. Parental alcohol consumption and risk of childhood acute lymphoblastic leukemia and brain tumors. Cancer Causes Control 2013; 24(2):391–402. 164. Slater ME, Linabery AM, Blair CK, et al. Maternal prenatal cigarette, alcohol and illicit drug use and risk of infant leukaemia: a report from the Children’s Oncology Group. Paediatr Perinat Epidemiol 2011;25(6):559–65. 165. Hu FB. Dietary pattern analysis: a new direction in nutritional epidemiology. Curr Opin Lipidol 2002;13(1):3–9. 166. Harnack L, Nicodemus K, Jacobs D.R. Jr., Folsom AR. An evaluation of the dietary guidelines for Americans in relation to cancer occurrence. Am J Clin Nutr 2002;76(4):889–96. 167. Kant AK. Dietary patterns and health outcomes. J Am Diet Assoc 2004;104(4):615–35. 168. Carmichael SL, Yang W, Feldkamp ML, et al. Reduced risks of neural tube defects and orofacial clefts with higher diet quality. Arch Pediatr Adolesc Med 2012;166(2): 121–126. 169. Singer AW, Carmichael SL, Selvin S, Fu C, Block G, Metayer C. Maternal diet quality before pregnancy and risk of childhood leukemia. Br J Nutr (In press). 170. Peters JM, Preston-Martin S, London SJ, Bowman JD, Buckley JD, Thomas DC. Processed meats and risk of childhood leukemia (California, USA). Cancer Causes Control 1994;5(2):195–202. 171. Bailey HD, Miller M, Greenop KR, et al. Paternal intake of folate and vitamins B6 and B12 before conception and risk of childhood acute lymphoblastic leukemia. Cancer Causes Control 2014;25(12):1615–25. 172. Wen W, Shu XO, Potter JD, et al. Parental medication use and risk of childhood acute lymphoblastic leukemia. Cancer 2002;95(8):1786–94. 173. Rudant J, Amigou A, Orsi L, et al. Fertility treatments, congenital malformations, fetal loss, and childhood acute leukemia: The ESCALE study (SFCE). Pediatr Blood Cancer 2013;60(2):301–8. Curr Probl PediatrAdolesc Health Care, October 2016 174. Amitay EL, Keinan-Boker L. Breastfeeding and Childhood Leukemia Incidence: A Meta-analysis and Systematic Review. JAMA Pediatr 2015;169(6):e151025. 175. Greenop KR, Bailey HD, Miller M, et al. Breastfeeding and nutrition to 2 years of age and risk of childhood acute lymphoblastic leukemia and brain tumors. Nutr Cancer 2015;67(3):431–41. 176. Schraw JM, Dong YQ, Okcu MF, Scheurer ME, Forman MR. Do longer formula feeding and later introduction of solids increase risk for pediatric acute lymphoblastic leukemia? Cancer Causes Control 2014;25(1):73–80. 177. Kwan ML, Block G, Selvin S, Month S, Buffler PA. Food consumption by children and the risk of childhood acute leukemia. Am J Epidemiol 2004;160(11):1098–107. 178. Diamantaras AA, Dessypris N, Sergentanis TN, et al. Nutrition in early life and risk of childhood leukemia: a case– control study in Greece. Cancer Causes Control, 24; 2013. p. 117–24. 179. Sarasua S, Savitz DA. Cured and broiled meat consumption in relation to childhood cancer: Denver, Colorado (United States). Cancer Causes Control 1994;5(2):141–8. 180. Rifkin R. American rate nurses highest on honesty, ethical standards. Gallup Poll, Social Issues 2014; http://www.gallup. com/poll/180260/americansrate-nurses-highest-honesty-ethicalstandards.aspx; 2016 Accessed 09.06.16. 181. Institute of Medicine. Role of the Primary Care Physicians in Occupational and Environmental Medicine. National Academies Press, Washington, DC; 1988. 182. Kilpatrick N, Frumkin H, Trowbridge J, et al. The environmental history in pediatric practice: a study of pediatricians' attitudes, beliefs, and practices. Environ Health Perspect 2002;110(8):823–7. 183. Trasande L, Boscarino J, Graber N, et al. The environment in pediatric practice: a study of New York pediatricians' attitudes, beliefs, and practices towards children’s environmental health. J Urban Health 2006;83(4):760–72. 184. Finn S, O’Fallon L. The Emergence of Environmental Health Literacy-From Its Roots to Its Future Potential. Environ Health Perspect 2015; http://dx.doi.org/10.1289/ehp.1409337. 185. Holman DM, Buchanan N, Cancer Prevention During Early Life Expert Group. Opportunities during early life for cancer prevention: 2016. Highlights from a series of virtual meetings with experts. Pediatrics 2016:(In press). 186. Metayer C, Dahl G, Wiemels J, Miller M. Childhood leukemia: a preventable disease. Pediatrics 2016:(In press). 187. Gee D. Late lessons from early warnings: toward realism and precaution with endocrine-disrupting substances. Environ Health Perspect 2006;114(suppl 1):152–60. 188. Rooney AA, Boyles AL, Wolfe MS, Bucher JR, Thayer KA. Systematic review and evidence integration for literaturebased environmental health science assessments. Environ Health Perspect 2014;122(7):711–8. 189. Woodruff TJ, Sutton P. The navigation guide systematic review methodology: a rigorous and transparent method for translating environmental health science into better health outcomes. Environ Health Perspect 2014;122(10): 1007–14. 351 190. Bergerson RJ, Collier LS, Sarver AL, et al. An insertional mutagenesis screen identifies genes that cooperate with MllAF9 in a murine leukemogenesis model. Blood 2012;119(19) 4512–23. 191. US Department of Health E, and Welfare. Report of the Advisory Committee to the Surgeon General of the Public Health Service. Washington, DC: US Department of Health, Education, and Welfare, Public Health Service, 1964. 192. Task Force on Sudden Infant Death Syndrome, Moon RY. SIDS and other sleep-related infant deaths: expansion of recommendations for a safe infant sleeping environment. Pediatrics 2011;128(5):e1341–67. 193. American Academy of Pediatrics AAP Task Force on Infant Positioning and SIDS: positioning and SIDS. Pediatrics 1992;89(6 Pt 1):1120–6. 194. Engelberts AC, de Jonge GA. Choice of sleeping position for infants: possible association with cot death. Arch Dis Child 1990;65(4):462–7. 195. Schmidt RJ, Tancredi DJ, Ozonoff S, et al. Maternal periconceptional folic acid intake and risk of autism spectrum disorders and developmental delay in the CHARGE (CHildhood Autism Risks from Genetics and Environment) case– control study. Am J Clin Nutr 2012;96(1):80–9. 196. Suren P, Roth C, Bresnahan M, et al. Association between maternal use of folic acid supplements and risk of autism spectrum disorders in children. J Am Med Assoc 2013;309(6): 570–577. 197. Williams J, Mai CT, Mulinare J, et al. Updated estimates of neural tube defects prevented by mandatory folic Acid fortification—United States, 1995–2011. MMWR Morb Mortal Wkly Rep 2015;64(1):1–5. 198. Tinker SC, Hamner HC, Qi YP, Crider KS. U.S. women of childbearing age who are at possible increased risk of a neural 352 199. 200. 201. 202. 203. 204. 205. tube defect-affected pregnancy due to suboptimal red blood cell folate concentrations, National Health and Nutrition Examination Survey 2007 to 2012. Birth Defects Res A Clin Mol Teratol 2015;103(6):517–26. Orozco AM, Yeung LF, Guo J, Carriquiry A, Berry RJ. Characteristics of U.S. adults with usual daily folic acid intake above the tolerable upper intake level: National Health and Nutrition Examination Survey, 2003–2010. Nutrients 2016;8(4). Jamal A, Homa DM, O'Connor E, et al. Current cigarette smoking among adults—United States, 2005–2014. MMWR Morb Mortal Wkly Rep 2015;64(44):1233–40. Sutton P, Woodruff TJ, Perron J, et al. Toxic environmental chemicals: the role of reproductive health professionals in preventing harmful exposures. Am J Obstet Gynecol 2012;207(3):164–73. Mello S, Hovick SR. Predicting behaviors to reduce toxic chemical exposures among new and expectant mothers: the role of distal variables within the integrative model of behavioral prediction. Health Educ Behav 2016; http://dx. doi.org/10.1177/1090198116637600. Grason HA, Misra DP. Reducing exposure to environmental toxicants before birth: moving from risk perception to risk reduction. Public Health Rep 2009;124(5):629–41. Gartner LM, Morton J, Lawrence RA, et al. Breastfeeding and the use of human milk. Pediatrics 2005;115(2): 496–506. Office of the Surgeon General (US), Centers for Disease Control and Prevention (US), Office on Women’s Health (US). The Surgeon General’s Call to Action to Support Breastfeeding. Office of the Surgeon General, Rockville, MD, 2011. Curr Probl PediatrAdolesc Health Care, October 2016