Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2022 Sep 1.
Published in final edited form as: Bioessays. 2021 Jun 9;43(9):e2100030. doi: 10.1002/bies.202100030

Using Primary Teeth and Archived Dried Spots for Exposomic Studies in Children: Exploring New Paths in the Environmental Epidemiology of Pediatric Cancer

Philip J Lupo 1,*, Lauren M Petrick 2, Thanh T Hoang 3, Amanda E Janitz 4, Erin L Marcotte 5, Jeremy M Schraw 1, Manish Arora 2, Michael E Scheurer 1
PMCID: PMC8390438  NIHMSID: NIHMS1715805  PMID: 34106479

Abstract

It is estimated that 300,000 children 0–14 years of age are diagnosed with cancer worldwide each year. While the absolute risk of cancer in children is low, it is the leading cause of death due to disease in children in high-income countries. In spite of this, the etiologies of pediatric cancer are largely unknown. Environmental exposures have long been thought to play an etiologic role. However, to date, there are few well-established environmental risk factors for pediatric malignancies, likely due to technical barriers in collecting biological samples prospectively in pediatric populations for direct measurements. In this review, we propose the use of novel or underutilized biospecimens (dried blood spots and teeth) and molecular approaches for exposure assessment (epigenetics, metabolomics, and somatic mutational profiles). Future epidemiologic studies of pediatric cancer should incorporate novel exposure assessment methodologies, data on molecular features of tumors, and a more complete assessment of gene-environment interactions.

Keywords: Environmental exposures, Biomarkers, Blood spots, Omics, Epidemiology, Pediatric Cancer

1. Introduction

It is estimated that 300,000 children 0–14 years of age are diagnosed with cancer worldwide each year.[13] However, the absolute risk of cancer in children is low compared to adults; incidence is 183 cases per million in the United States.[63] While relatively rare, these conditions continue to pose a serious public health problem. For instance, in high-income countries, cancer is the leading cause of death due to disease in children after the first year of life.[13] Furthermore, while survival has continued to improve due to advances in treatment, there is still a high likelihood of relapse, and this young population with long life expectancy often have an increased risk of second cancers, an excess burden of multiple chronic health conditions as a consequence of their therapy, and diminished quality of life.[31] This is notable, as there are currently 500,000 survivors of pediatric cancer living in the United States.[39]

Pediatric cancers are heterogeneous and display a markedly different range of tumor types than in adults, including several classes which are largely exclusive to children. Based on data from the Surveillance, Epidemiology, and End Results (SEER) program in the United States,[63] for children 0 to 14 years, acute lymphoblastic leukemia (ALL) is the most common cancer, accounting for 25.5% of all cancer diagnoses. Acute myeloid leukemia is the next most common type of leukemia in this age group, occurring at a rate one-fifth that for ALL. Central nervous system cancers, primarily occurring in the brain, accounts for 20.8% of cancer diagnoses, and together with acute lymphoblastic leukemia and acute myeloid leukemia make up one-half of cancer diagnoses among children younger than 15 years of age. The most common non-central nervous system solid tumor in the 0- to 14-year age group is neuroblastoma (6.8%), followed by non-Hodgkin lymphoma (6.1%) and Wilms tumor (5.3%). Other diagnoses that individually represented 2% to 4% of cancer diagnoses in this age group include Hodgkin lymphoma, rhabdomyosarcoma, non-rhabdomyosarcoma soft tissue sarcomas, germ-cell tumors, retinoblastoma, and osteosarcoma. Notably, these distributions vary globally, suggesting differences in etiologic factors by geography.[51]

While key environmental exposures have been identified for adult cancers (e.g., smoking, benzene), much less is known in relation to pediatric cancer. A notable difference between adult cancers and pediatric cancers is the latency period associated with these conditions. For instance, smoking usually starts during adolescence or young adulthood, but associated malignancies do not become apparent until many decades later. However, several pediatric cancers are predominantly diagnosed in infancy (e.g., embryonal tumors such as neuroblastoma) or in early childhood (2 to 5 years of age), such as acute lymphoblastic leukemia. Therefore, the disruptions in molecular processes that may lead to pediatric cancer are likely different from those of adult cancers; at the least, the carcinogenic process in children is necessarily much shorter in time. There is also an emerging consensus that pediatric cancer is a disease that arises from dysregulated development.[25] This is supported by recent large-scale germline genetic studies of pediatric cancer indicating approximately 8% of cases are due to pathogenic variants in well-established cancer predisposition genes, some of which are known to play a key role in development (e.g., HRAS).[71] Additionally, genome-wide association studies of pediatric malignancies have pointed to the role of other important developmental genes on cancer risk (e.g., IKZF1).[51]

In spite of this, there are few well-established environmental determinants of pediatric cancer or modifiers of pediatric cancer risk in the context of underlying genetic susceptibility. While there is extensive evidence that high doses of ionizing radiation are associated with pediatric cancer, the prevalence of this exposure is very low.[44] Relatively common environmental exposures, including pesticides[18] and air pollution[26] have also been explored. While individual studies and meta-analyses have indicated associations between these exposures and some pediatric cancers, effect sizes are relatively modest. One problem with previous studies of environmental exposures includes the limitations of using questionnaire data to estimate exposure or the use of proxies for exposure assessment (e.g., residential information). Additionally, few studies have evaluated how environmental factors might influence genetic susceptibility to pediatric cancer. In this review, we propose the use of novel or underutilized biospecimens and molecular exposure assessment strategies for evaluating the environmental determinants and modifiers of pediatric cancer. Therefore, our objective is to consider novel and underutilized biospecimens (dried blood spots and primary teeth) for molecular-based exposure assessment strategies (e.g., epigenetics, metabolomics) in studies of pediatric cancer.

2. Novel or Underutilized Biospecimens in Environmental Studies of Pediatric Cancer

2.1. Using Dried Blood Spots to Estimate Perinatal Exposures

Newborn screening is a mandatory public health program in most developed countries worldwide that tests newborn babies for selected genetic, endocrine, and metabolic disorders. Under this program, dried blood spots (DBS) are routinely collected within 24–48 hours of birth for >98% of the nearly 4 million infants born in the United States each year.[56] After screening is complete, residual DBS are stored for confirmation of positive results or reanalysis, if needed. In addition, some states also retain residual DBS for research purpose. Although the duration of retention and storage practice varies widely by state, from 30 days to indefinite, and policies regarding their retention and use for research have rapidly evolved over the past decade,[48] DBS biobanks offer a valuable biospecimen for direct measurement of early life exposures.[70]

Etiologic research into the environmental causes of pediatric cancer is hindered by many factors, including the relative rarity of pediatric cancer subtypes, selection bias in case-control studies, and recall bias in questionnaire-based exposure assessments. Additionally, accurate exposure assessment in the absence of biospecimens is notoriously difficult to achieve.[75] Self-reported exposure data, sometimes collected many years after critical exposure windows, such as pregnancy, gives rise to the potential for exposure misclassification. Alternatively, biospecimens can provide accurate, unbiased estimates of exposures through the use of biomarkers. However, they must be collected prior to the onset of disease for use in etiologic research, because when collected contemporaneously with diagnosis, the disease process may alter exposure, biomarker status, or both (e.g., “reverse causality”). Given the low incidence of pediatric cancers, cohort studies for prospective collection of data and biologic specimens are not economically or practically feasible requiring follow-up of millions of children to accrue sufficient cases to produce statistically meaningful results. Therefore, DBS constitute the only population-based source of pre-diagnostic biospecimens for case-control studies.

The potential uses of DBS in environmental studies of pediatric cancer are extensive. Analytes of environmental exposures detectable in DBS reported to-date include benzene oxide,[29] lead,[6] mercury,[61] arsenic,[28] cadmium,[17] cotinine,[78,87] viral DNA,[27] organochlorine[53] and organophosphorus pesticides,[84] perfluorinated compounds,[88] polychlorinated biphenyls,[53] polybrominated diphenyl ethers,[52] bisphenol A,[12,30,88] phosphatidylethanol,[9] and polyfluoroalkyl chemicals.[43,54] Additionally, there is increasing awareness that epigenetic signatures of environmental exposures may provide effective biomarkers of past exposure.[46] Recent studies have shown that DNA methylation profiling, including epigenome-wide association studies, is possible using residual DBS as the source material.[36,38,58,81,89]

Residual DBS offer a unique and extremely valuable resource to assess environmental exposures present at birth, with possible extrapolation to earlier in utero exposures. Neonatal biobanks are population-based, can supply samples from representative controls, and can provide basic demographic data through linkage to birth certificates. Further utilization of DBS to elucidate causal environmental agents and biological response associated with pediatric cancer risk could enable primary prevention for modifiable environmental factors.

2.2. Using Primary Teeth to Reconstruct Early-Life Exposures

There is growing evidence that deciduous teeth (i.e., “baby teeth”) serve as a stable repository for exposures occurring during the prenatal period and early childhood. This arises from the developmental biology of teeth—deciduous teeth begin mineralization prenatally towards the end of the first trimester and then continue to develop in an incremental manner forming daily (and even sub-daily) markings.[66] Thus, naturally shed deciduous teeth are perhaps the only stable fetal tissue that can be attained in childhood non-invasively, which can then be used to reconstruct exposure history at fine-scale temporal resolution.

For decades teeth have been used to estimate long-term cumulative exposure to metals; however, these methods did not allow for longitudinal exposure assessment.[60] Recently, high-dimensional analytics combining sophisticated histological and chemical analyses have been developed to reconstruct the early-life exposome (e.g., metals, organic chemicals, nutrients, infection, inflammation) by measuring tooth layers corresponding to specific life stages.[57] While not yet applied to pediatric cancer, emerging autism and neurodevelopment studies are utilizing primary teeth to reconstruct the early-life exposome to measure the etiologic role of environmental exposures on disease risk.[57,59] Notably, even if shed after the initiation of cancer therapy, primary teeth can be used to reconstruct exposures prior to cancer diagnosis.[7]

Compared to using blood or other biological tissues, primary teeth provide a more accurate assessment of 1) exposure dose, 2) the developmental period during the time of exposure, and 3) exogenous exposures during earliest periods of fetal development that often have short half-lives.[8] Moreover, teeth are easy to transport and store being stable at room temperature. When taken together, these factors point to the importance of utilizing teeth as novel biospecimens in environmental studies of pediatric cancer.

3. Novel or Underutilized Molecular Approaches for Assessing Exposures in Environmental Studies of Pediatric Cancer

3.1. Using Epigenetics as Biomarkers of Environmental Exposures

Epigenetics refer to changes in gene expression that are not due to the DNA sequence. One type of epigenetic process is DNA methylation – the addition of a methyl group at the fifth carbon of a cytosine that is typically followed by a guanine. DNA methylation is the best-studied epigenetic mechanism in humans. The development of DNA methylation arrays has allowed researchers to systematically measure methylation at individual C-phosphate-G sites (CpGs) throughout the genome for a relatively inexpensive cost. Thus, there has been a prolific number of epigenome-wide methylation studies published in the past decade, including those in relation to environmental exposures.[32,38,40,46,67,73,77,81,85]

Because environmental factors can alter DNA methylation at specific CpGs, these specific CpGs can be used as a robust biomarker of exposure in adults[77,90] and can capture in utero exposures in newborns.[73] There is evidence that some altered DNA methylation in newborns can persist into childhood and adolescence.[41,74] There are also studies that have successfully evaluated the impact of air pollution on DNA methylation in children.[40] Leveraging DNA methylation as a biomarker of exposure may overcome the exposure assessment challenges in previous studies of pediatric cancer, as it can objectively capture exposures in utero and after birth. A limitation of this approach is that large epigenome-wide studies of environmental exposures are needed to identify robust CpGs related to the exposure of interest. Alternatively, epigenome-wide meta-analyses of several smaller studies can overcome this limitation, as meta-analyses of smoking,[41] alcohol,[49] and air pollution[3435] have identified robust CpGs in relation to the exposure.

DNA methylation may be a novel, cost-effective molecular tool that can be utilized to assess environmental exposures for pediatric cancer research. One DNA sample can provide information on many different environmental factors that may contribute to pediatric cancer. While it is ideal to collect samples prior to the time of diagnosis, ongoing assessments are evaluating if there are metastable epialleles related to pediatric cancer that can evaluated with samples collected at the time of diagnosis.[23] Because DNA methylation is tissue specific and is influenced by both genetic variation and environmental factors, alterations in DNA methylation may also provide mechanistic insight into treatment response and survivorship, as have been reported,[14,50] to improve clinical outcomes through epigenetic therapies in children with cancer. As noted, while a few studies have utilized DBS for epigenome-wide assessments, to our knowledge, there have been no assessment exploring the utility of primary teeth to evaluate DNA methylation patterns, providing territory for novel explorations of pediatric cancer risk.

3.2. Using Untargeted Metabolomics as Measure of the Exposome

Metabolomics, the global measurement of small molecules in a biological specimen that correlate with cellular function and dysregulated physiology, holds promise for understanding the environmental determinants of pediatric cancer incidence and survival.[19,68] Metabolomics experiments most commonly utilize nuclear magnetic resonance (NMR), liquid chromatography-mass spectrometry (LC-MS), or gas chromatography-mass spectrometry (GC-MS) methodologies.[11,20,24,86] The type of technology also determines the class of small molecules that can be measured, while there is some overlap in methodologies. Therefore, the choice of method in a particular study depends on practical considerations such as cost and sample type, as well as the investigator’s objectives. Each may contribute valuable data to studies of pediatric cancer.

Metabolomics may be useful for comprehensively characterizing an individual’s “exposome,” as the metabolome is a biochemical read-out of genetic predisposition, diet, environmental exposures, general metabolic health, and other traits.[62,72] Thus, when applied appropriately, metabolomics may allow for the simultaneous assessment of diverse exposures using a single experimental approach. This is particularly relevant to environmental health research, because humans are not exposed to one chemical at a time or even in small groups, but to complex mixtures. Further, by measuring dysregulated biological pathways, metabolomics enables the measurement of biological response to transient exposures that occurred weeks, months, or years ago and may be long gone from the body.[21] Thus, metabolomics has potential to accelerate the discovery of exposures associated with pediatric cancer, which has proven a difficult undertaking.

Of particular note to investigators interested in the etiologies of cancer and other childhood diseases, mass spectrometry-based metabolomics have been applied to samples such as archived DBS, cord blood, and deciduous teeth.[4,42,69] These samples may provide information on exposures during critical developmental periods, without concern for recall bias and, sometimes, with high temporal resolution.

Applications for metabolomics exist along the continuum of pediatric cancer research from etiology to survivorship. In the context of acute lymphoblastic leukemia, metabolomics has been applied to study the tumor microenvironment, treatment response, vincristine-induced neuropathy, and treatment-related fatigue.[10,15,22,76,80,82] In addition, as long-term survival for children with cancer continues to improve, the community has increasingly recognized the critical importance of survivorship research. Two-thirds of survivors report at least one chronic health condition in adulthood, including second malignant neoplasms, cardiovascular disease, and frailty.[16,37,64] In adult populations, recent studies have demonstrated associations between metabolomic profiles, future cardiovascular disease risk, and all-cause mortality;[47,65] this suggests the intriguing possibility that metabolomic profiling of pediatric cancer survivors may be useful for predicting outcomes and risk of late effects, particularly cardiovascular events.

Barriers to the broader application of metabolomics include cost; sample type and storage; lack of large, shareable spectral libraries contributing to difficulty in feature identification; heterogeneity in sample and data analysis pipelines; and data dimensionality.[20,86] However, the field is under active development, and it is anticipated that metabolomics approaches will become more comprehensive and powerful with time as these limitations are addressed. In combination with robust epidemiologic data and additional ‘-omics’ data such as from genomic studies, metabolomics has the potential to be a powerful tool for elucidating the developmental origins of pediatric cancer, understanding treatment response, and managing the health of long-term survivors.

3.3. Using Somatic Mutational Signatures in Pediatric Tumors to Understanding the Carcinogenic Role of Environmental Exposures

Somatic mutations found in cancer genomes may be the consequence of the intrinsic infidelity of the DNA replication machinery, exogenous or endogenous mutagen exposures, enzymatic modification of DNA, or defective DNA repair.[3] Biological processes that cause mutations in somatic cells leave a mutational signature.[3] To date, 30 distinct mutational signatures have been identified in 40 different cancer types using whole-genome and whole-exome sequencing, which include associations with age, smoking, and ultraviolet light in addition to other exogenous and endogenous exposures.[12] A previous analysis of smoking-related cancers revealed a mutational signature of tobacco exposure which was present in cancers derived from cells directly exposed to tobacco smoke.[1] Specifically for pediatric cancers, researchers have identified overlap with previously identified mutational signatures.[33,55] Additionally, unique signatures have been identified in atypical teratoid rhabdoid tumors (ATRT), ependymoma, B-lineage acute lymphoblastic leukemia, acute myeloid leukemia, neuroblastoma, and Wilms tumor.[33,55]

Understanding mutational signatures of pediatric cancer can help identify potential endogenous and exogenous exposures that are important in the etiology of pediatric cancers.[2] Gröbner et al.[33] noted that approximately 50% of pediatric cancers studied may have a targetable genetic mutation that can be used to improve precision therapeutics in children. Current resources to study mutational signatures in cancer include COSMIC (Catalogue Of Somatic Mutations in Cancer)[83] and, specifically for pediatric cancers, the PedPanCan[45] and PeCan Variant Knowledge Base.[79] Future studies would benefit from focusing on expanding cohorts to include additional types of pediatric cancer to better understand the impacts of these mutational signatures on etiology across cancer types.

4. Conclusion

While there have been tremendous strides in improving outcomes for children with cancer, there is still a great deal of work related to disentangling the etiologic origins of these conditions. Future studies should incorporate novel exposure assessment methodologies using unique biospecimens, such as those proposed here. Additionally, there are several well-established germline genetic variants that have been identified for pediatric cancers, which have yet to be leveraged in studies of environmental exposures in the context of genetic susceptibility.[71] Through these efforts, it is hoped that our understanding of the causes of pediatric cancer can be better ascertained, leading to novel surveillance or prevention strategies.

Acknowledgements

This manuscript was funding in part by the Cancer Prevention and Research Institute of Texas (CPRIT) RP180755.

REFERENCES

  • [1].Alexandrov LB, Ju YS, Haase K, Van Loo P, Martincorena I, Nik-Zainal S, . . . Stratton MR (2016). Mutational signatures associated with tobacco smoking in human cancer. Science, 354(6312), 618–622. doi: 10.1126/science.aag0299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Alexandrov LB, Nik-Zainal S, Wedge DC, Aparicio SA, Behjati S, Biankin AV, . . . Stratton MR (2013). Signatures of mutational processes in human cancer. Nature, 500(7463), 415–421. doi: 10.1038/nature12477 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [3].Alexandrov LB, & Stratton MR (2014). Mutational signatures: the patterns of somatic mutations hidden in cancer genomes. Current Opinion in Genetics & Development, 24(Supplement C), 52–60. doi: 10.1016/j.gde.2013.11.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Andra SS, Austin C, & Arora M. (2016). The tooth exposome in children’s health research. Curr Opin Pediatr, 28(2), 221–227. doi: 10.1097/mop.0000000000000327 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Andra SS, Austin C, Wright RO, & Arora M. (2015). Reconstructing pre-natal and early childhood exposure to multi-class organic chemicals using teeth: Towards a retrospective temporal exposome. Environ Int, 83, 137–145. doi: 10.1016/j.envint.2015.05.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Archer NP, Bradford CM, Klein DM, Barnes J, Smith LJ, & Villanacci JF (2012). Relationship between prenatal lead exposure and infant blood lead levels. Matern Child Health J, 16(7), 1518–1524. doi: 10.1007/s10995-011-0917-3 [DOI] [PubMed] [Google Scholar]
  • [7].Arora M, & Austin C. (2013). Teeth as a biomarker of past chemical exposure. Curr Opin Pediatr, 25(2), 261–267. doi: 10.1097/MOP.0b013e32835e9084 [DOI] [PubMed] [Google Scholar]
  • [8].Arora M, Austin C, Sarrafpour B, Hernandez-Avila M, Hu H, Wright RO, & Tellez-Rojo MM (2014). Determining prenatal, early childhood and cumulative long-term lead exposure using micro-spatial deciduous dentine levels. PLoS One, 9(5), e97805. doi: 10.1371/journal.pone.0097805 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [9].Bakhireva LN, Leeman L, Savich RD, Cano S, Gutierrez H, Savage DD, & Rayburn WF (2014). The validity of phosphatidylethanol in dried blood spots of newborns for the identification of prenatal alcohol exposure. Alcohol Clin Exp Res, 38(4), 1078–1085. doi: 10.1111/acer.12349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Bannur Z, Teh LK, Hennesy T, Rosli WR, Mohamad N, Nasir A, . . . Salleh MZ (2014). The differential metabolite profiles of acute lymphoblastic leukaemic patients treated with 6-mercaptopurine using untargeted metabolomics approach. Clin Biochem, 47(6), 427–431. doi: 10.1016/j.clinbiochem.2014.02.013 [DOI] [PubMed] [Google Scholar]
  • [11].Beale DJ, Pinu FR, Kouremenos KA, Poojary MM, Narayana VK, Boughton BA, . . . Dias DA (2018). Review of recent developments in GC-MS approaches to metabolomics-based research. Metabolomics, 14(11), 152. doi: 10.1007/s11306-018-1449-2 [DOI] [PubMed] [Google Scholar]
  • [12].Bell EM, Yeung EH, Ma W, Kannan K, Sundaram R, Smarr MM, & Buck Louis GM (2018). Concentrations of endocrine disrupting chemicals in newborn blood spots and infant outcomes in the upstate KIDS study. Environ Int, 121(Pt 1), 232–239. doi: 10.1016/j.envint.2018.09.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Bhakta N, Force LM, Allemani C, Atun R, Bray F, Coleman MP, . . . Fitzmaurice C. (2019). Childhood cancer burden: a review of global estimates. Lancet Oncol, 20(1), e42–e53. doi: 10.1016/S1470-2045(18)30761-7 [DOI] [PubMed] [Google Scholar]
  • [14].Brown AL, Foster KL, Lupo PJ, Peckham-Gregory EC, Murray JC, Okcu MF, . . . Scheurer ME (2017). DNA methylation of a novel PAK4 locus influences ototoxicity susceptibility following cisplatin and radiation therapy for pediatric embryonal tumors. Neuro Oncol, 19(10), 1372–1379. doi: 10.1093/neuonc/nox076 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Brown AL, Sok P, Taylor O, Woodhouse JP, Bernhardt MB, Raghubar KP, . . . Scheurer ME (2020). Cerebrospinal Fluid Metabolomic Profiles Associated With Fatigue During Treatment for Pediatric Acute Lymphoblastic Leukemia. J Pain Symptom Manage. doi: 10.1016/j.jpainsymman.2020.08.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Chao C, Bhatia S, Xu L, Cannavale KL, Wong FL, Huang P-YS, . . . Armenian SH (2019). Incidence, Risk Factors, and Mortality Associated With Second Malignant Neoplasms Among Survivors of Adolescent and Young Adult CancerSecond Malignant Neoplasms Among Survivors of Adolescent and Young Adult CancerSecond Malignant Neoplasms Among Survivors of Adolescent and Young Adult Cancer. JAMA Network Open, 2(6), e195536-e195536. doi: 10.1001/jamanetworkopen.2019.5536 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Chaudhuri SN, Butala SJ, Ball RW, Braniff CT, & Rocky Mountain Biomonitoring C. (2009). Pilot study for utilization of dried blood spots for screening of lead, mercury and cadmium in newborns. J Expo Sci Environ Epidemiol, 19(3), 298–316. doi: 10.1038/jesee.2008.1910.1038/jes.2008.19 [DOI] [PubMed] [Google Scholar]
  • [18].Chen M, Chang C-H, Tao L, & Lu C. (2015). Residential Exposure to Pesticide During Childhood and Childhood Cancers: A Meta-Analysis. Pediatrics, 136(4), 719–729. doi: 10.1542/peds.2015-0006 [DOI] [PubMed] [Google Scholar]
  • [19].Clish CB (2015). Metabolomics: an emerging but powerful tool for precision medicine. Cold Spring Harb Mol Case Stud, 1(1), a000588. doi: 10.1101/mcs.a000588 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Cui L, Lu H, & Lee YH (2018). Challenges and emergent solutions for LC-MS/MS based untargeted metabolomics in diseases. Mass Spectrom Rev, 37(6), 772–792. doi: 10.1002/mas.21562 [DOI] [PubMed] [Google Scholar]
  • [21].Dennis KK, Auerbach SS, Balshaw DM, Cui Y, Fallin MD, Smith MT, . . . Miller GW (2016). The Importance of the Biological Impact of Exposure to the Concept of the Exposome. Environ Health Perspect, 124(10), 1504–1510. doi: 10.1289/EHP140 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Dhakshinamoorthy S, Dinh NT, Skolnick J, & Styczynski MP (2015). Metabolomics identifies the intersection of phosphoethanolamine with menaquinone-triggered apoptosis in an in vitro model of leukemia. Mol Biosyst, 11(9), 2406–2416. doi: 10.1039/c5mb00237k [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [23].Dolinoy DC, Das R, Weidman JR, & Jirtle RL (2007). Metastable epialleles, imprinting, and the fetal origins of adult diseases. Pediatr Res, 61(5 Pt 2), 30R–37R. doi: 10.1203/pdr.0b013e31804575f7 [DOI] [PubMed] [Google Scholar]
  • [24].Emwas AH, Roy R, McKay RT, Tenori L, Saccenti E, Gowda GAN, . . . Wishart DS (2019). NMR Spectroscopy for Metabolomics Research. Metabolites, 9(7). doi: 10.3390/metabo9070123 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Filbin M, & Monje M. (2019). Developmental origins and emerging therapeutic opportunities for childhood cancer. Nat Med, 25(3), 367–376. doi: 10.1038/s41591-019-0383-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [26].Filippini T, Hatch EE, Rothman KJ, Heck JE, Park AS, Crippa A, . . . Vinceti M. (2019). Association between Outdoor Air Pollution and Childhood Leukemia: A Systematic Review and Dose-Response Meta-Analysis. Environmental health perspectives, 127(4), 46002–46002. doi: 10.1289/EHP4381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Francis SS, Wallace AD, Wendt GA, Li L, Liu F, Riley LW, . . . Wiemels JL (2017). In utero cytomegalovirus infection and development of childhood acute lymphoblastic leukemia. Blood, 129(12), 1680–1684. doi: 10.1182/blood-2016-07-723148 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Funk WE, McGee JK, Olshan AF, & Ghio AJ (2013). Quantification of arsenic, lead, mercury and cadmium in newborn dried blood spots. Biomarkers, 18(2), 174–177. doi: 10.3109/1354750X.2012.750379 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Funk WE, Waidyanatha S, Chaing SH, & Rappaport SM (2008). Hemoglobin adducts of benzene oxide in neonatal and adult dried blood spots. Cancer Epidemiol Biomarkers Prev, 17(8), 1896–1901. doi: 10.1158/1055-9965.EPI-08-0356 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Ghassabian A, Bell EM, Ma WL, Sundaram R, Kannan K, Buck Louis GM, & Yeung E. (2018). Concentrations of perfluoroalkyl substances and bisphenol A in newborn dried blood spots and the association with child behavior. Environ Pollut, 243(Pt B), 1629–1636. doi: 10.1016/j.envpol.2018.09.107 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Gibson TM, Mostoufi-Moab S, Stratton KL, Leisenring WM, Barnea D, Chow EJ, . . . Oeffinger KC (2018). Temporal patterns in the risk of chronic health conditions in survivors of childhood cancer diagnosed 1970–99: a report from the Childhood Cancer Survivor Study cohort. Lancet Oncol, 19(12), 1590–1601. doi: 10.1016/S1470-2045(18)30537-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Gondalia R, Baldassari A, Holliday KM, Justice AE, Mendez-Giraldez R, Stewart JD, . . . Whitsel EA (2019). Methylome-wide association study provides evidence of particulate matter air pollution-associated DNA methylation. Environ Int, 104723. doi: 10.1016/j.envint.2019.03.071 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Gröbner SN, Worst BC, Weischenfeldt J, Buchhalter I, Kleinheinz K, Rudneva VA, . . . Pfister SM (2018). The landscape of genomic alterations across childhood cancers. Nature, 555(7696), 321–327. doi: 10.1038/nature25480 [DOI] [PubMed] [Google Scholar]
  • [34].Gruzieva O, Xu CJ, Breton CV, Annesi-Maesano I, Anto JM, Auffray C, . . . Melen E. (2017). Epigenome-Wide Meta-Analysis of Methylation in Children Related to Prenatal NO2 Air Pollution Exposure. Environ Health Perspect, 125(1), 104–110. doi: 10.1289/EHP36 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Gruzieva O, Xu CJ, Yousefi P, Relton C, Merid SK, Breton CV, . . . Melen E. (2019). Prenatal Particulate Air Pollution and DNA Methylation in Newborns: An Epigenome-Wide Meta-Analysis. Environ Health Perspect, 127(5), 57012. doi: 10.1289/EHP4522 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Hannon E, Schendel D, Ladd-Acosta C, Grove J, Hansen CS, Hougaard DM, . . . Mill J. (2019). Variable DNA methylation in neonates mediates the association between prenatal smoking and birth weight. Philos Trans R Soc Lond B Biol Sci, 374(1770), 20180120. doi: 10.1098/rstb.2018.0120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Hayek S, Gibson TM, Leisenring WM, Guida JL, Gramatges MM, Lupo PJ, . . . Ness KK (2020). Prevalence and Predictors of Frailty in Childhood Cancer Survivors and Siblings: A Report From the Childhood Cancer Survivor Study. J Clin Oncol, 38(3), 232–247. doi: 10.1200/jco.19.01226 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Hollegaard MV, Grauholm J, Norgaard-Pedersen B, & Hougaard DM (2013). DNA methylome profiling using neonatal dried blood spot samples: a proof-of-principle study. Mol Genet Metab, 108(4), 225–231. doi: 10.1016/j.ymgme.2013.01.016 [DOI] [PubMed] [Google Scholar]
  • [39].Hudson MM, Ness KK, Nolan VG, Armstrong GT, Green DM, Morris EB, . . . Robison LL (2011). Prospective medical assessment of adults surviving childhood cancer: study design, cohort characteristics, and feasibility of the St. Jude Lifetime Cohort study. Pediatr Blood Cancer, 56(5), 825–836. doi: 10.1002/pbc.22875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Isaevska E, Moccia C, Asta F, Cibella F, Gagliardi L, Ronfani L, . . . Richiardi L. (2020). Exposure to ambient air pollution in the first 1000 days of life and alterations in the DNA methylome and telomere length in children: a systematic review. Environ Res, 110504. doi: 10.1016/j.envres.2020.110504 [DOI] [PubMed] [Google Scholar]
  • [41].Joubert BR, Felix JF, Yousefi P, Bakulski KM, Just AC, Breton C, . . . London SJ (2016). DNA Methylation in Newborns and Maternal Smoking in Pregnancy: Genome-wide Consortium Meta-analysis. Am J Hum Genet, 98(4), 680–696. doi: 10.1016/j.ajhg.2016.02.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Kadakia R, Talbot O, Kuang A, Bain JR, Muehlbauer MJ, Stevens RD, . . . Lowe WL (2019). Cord Blood Metabolomics: Association With Newborn Anthropometrics and C-Peptide Across Ancestries. J Clin Endocrinol Metab, 104(10), 4459–4472. doi: 10.1210/jc.2019-00238 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Kato K, Wanigatunga AA, Needham LL, & Calafat AM (2009). Analysis of blood spots for polyfluoroalkyl chemicals. Anal Chim Acta, 656(1–2), 51–55. doi: 10.1016/j.aca.2009.10.007 [DOI] [PubMed] [Google Scholar]
  • [44].Kendall GM, Bithell JF, Bunch KJ, Draper GJ, Kroll ME, Murphy MFG, . . . Vincent TJ (2018). Childhood cancer research in oxford III: The work of CCRG on ionising radiation. British Journal of Cancer, 119(6), 771–778. doi: 10.1038/s41416-018-0182-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [45].KiTZ Heidelberg Hopp Children’s Cancer Center. (2020). PedPanCan. Retrieved 11/8/20, from https://www.kitz-heidelberg.de/en/research/research-projects/pedpancan/
  • [46].Ladd-Acosta C, & Fallin MD (2019). DNA methylation signatures as biomarkers of prior environmental exposures. Curr Epidemiol Rep, 6(1), 1–13. doi: 10.1007/s40471-019-0178-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Li J, Guasch-Ferré M, Chung W, Ruiz-Canela M, Toledo E, Corella D, . . . Liang L. (2020). The Mediterranean diet, plasma metabolome, and cardiovascular disease risk. Eur Heart J, 41(28), 2645–2656. doi: 10.1093/eurheartj/ehaa209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Linabery AM, Slater ME, Spector LG, Olshan AF, Stork SK, Roesler MA, . . . Ross JA (2011). Feasibility of neonatal dried blood spot retrieval amid evolving state policies (2009–2010): a Children’s Oncology Group study. Paediatr Perinat Epidemiol, 25(6), 549–558. doi: 10.1111/j.1365-3016.2011.01228.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [49].Liu C, Marioni RE, Hedman AK, Pfeiffer L, Tsai PC, Reynolds LM, . . . Levy D. (2018). A DNA methylation biomarker of alcohol consumption. Mol Psychiatry, 23(2), 422–433. doi: 10.1038/mp.2016.192 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Lupo PJ, Brown AL, Arroyo VM, Kamdar KY, Belmont JW, Scheurer ME, . . . Bhatia S. (2019). DNA methylation and obesity in survivors of pediatric acute lymphoblastic leukemia: A report from the Childhood Cancer Survivor Study. Genes Chromosomes Cancer, 58(1), 52–59. doi: 10.1002/gcc.22701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Lupo PJ, & Spector LG (2020). Cancer Progress and Priorities: Childhood Cancer. Cancer Epidemiol Biomarkers Prev, 29(6), 1081–1094. doi: 10.1158/1055-9965.EPI-19-0941 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [52].Ma W, Kannan K, Wu Q, Bell EM, Druschel CM, Caggana M, & Aldous KM (2013). Analysis of polyfluoroalkyl substances and bisphenol A in dried blood spots by liquid chromatography tandem mass spectrometry. Anal Bioanal Chem, 405(12), 4127–4138. doi: 10.1007/s00216-013-6787-3 [DOI] [PubMed] [Google Scholar]
  • [53].Ma WL, Gao C, Bell EM, Druschel CM, Caggana M, Aldous KM, . . . Kannan K. (2014). Analysis of polychlorinated biphenyls and organochlorine pesticides in archived dried blood spots and its application to track temporal trends of environmental chemicals in newborns. Environ Res, 133, 204–210. doi: 10.1016/j.envres.2014.05.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Ma WL, Yun S, Bell EM, Druschel CM, Caggana M, Aldous KM, . . . Kannan K. (2013). 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, 47(14), 8015–8021. doi: 10.1021/es401857v [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Ma X, Liu Y, Liu Y, Alexandrov LB, Edmonson MN, Gawad C, . . . Zhang J. (2018). Pan-cancer genome and transcriptome analyses of 1,699 paediatric leukaemias and solid tumours. Nature, 555(7696), 371–376. doi: 10.1038/nature25795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [56].Martin JA, Hamilton BE, & Osterman MJK (2020). Births in the United States, 2019. NCHS Data Brief(387), 1–8. [PubMed] [Google Scholar]
  • [57].Modabbernia A, Velthorst E, Gennings C, De Haan L, Austin C, Sutterland A, . . . Reichenberg A. (2016). Early-life metal exposure and schizophrenia: A proof-of-concept study using novel tooth-matrix biomarkers. Eur Psychiatry, 36, 1–6. doi: 10.1016/j.eurpsy.2016.03.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [58].Mohandas N, Bass-Stringer S, Maksimovic J, Crompton K, Loke YJ, Walstab J, . . . Craig JM (2018). Epigenome-wide analysis in newborn blood spots from monozygotic twins discordant for cerebral palsy reveals consistent regional differences in DNA methylation. Clin Epigenetics, 10, 25. doi: 10.1186/s13148-018-0457-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Mora AM, Arora M, Harley KG, Kogut K, Parra K, Hernandez-Bonilla D, . . . Eskenazi B. (2015). Prenatal and postnatal manganese teeth levels and neurodevelopment at 7, 9, and 10.5 years in the CHAMACOS cohort. Environ Int, 84, 39–54. doi: 10.1016/j.envint.2015.07.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Needleman HL, Gunnoe C, Leviton A, Reed R, Peresie H, Maher C, & Barrett P. (1979). Deficits in psychologic and classroom performance of children with elevated dentine lead levels. N Engl J Med, 300(13), 689–695. doi: 10.1056/nejm197903293001301 [DOI] [PubMed] [Google Scholar]
  • [61].Nelson JW, Edhlund BL, Johnson J, Rosebush CE, Holmquist ZS, Swan SH, & Nguyen RH (2016). Assessing a New Method for Measuring Fetal Exposure to Mercury: Newborn Bloodspots. Int J Environ Res Public Health, 13(7). doi: 10.3390/ijerph13070692 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Niedzwiecki MM, Walker DI, Vermeulen R, Chadeau-Hyam M, Jones DP, & Miller GW (2019). The Exposome: Molecules to Populations. Annu Rev Pharmacol Toxicol, 59, 107–127. doi: 10.1146/annurev-pharmtox-010818-021315 [DOI] [PubMed] [Google Scholar]
  • [63].Noone AM, Howlader N, Krapcho M, Miller D, Brest A, Yu M, . . . Cronin KA (2017). SEER Cancer Statistics Review, 1975–2015. Bethesda, MD: National Cancer Institute. [Google Scholar]
  • [64].Oeffinger KC, Mertens AC, Sklar CA, Kawashima T, Hudson MM, Meadows AT, . . . Robison LL (2006). Chronic health conditions in adult survivors of childhood cancer. N Engl J Med, 355(15), 1572–1582. doi: 10.1056/NEJMsa060185 [DOI] [PubMed] [Google Scholar]
  • [65].Ottosson F, Smith E, Fernandez C, & Melander O. (2020). Plasma Metabolites Associate with All-Cause Mortality in Individuals with Type 2 Diabetes. Metabolites, 10(8). doi: 10.3390/metabo10080315 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Papakyrikos AM, Arora M, Austin C, Boughner JC, Capellini TD, Dingwall HL, . . . Smith TM (2020). Biological clocks and incremental growth line formation in dentine. J Anat, 237(2), 367–378. doi: 10.1111/joa.13198 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Park B, Khanam R, Vinayachandran V, Baqui AH, London SJ, & Biswal S. (2020). Epigenetic biomarkers and preterm birth. Environ Epigenet, 6(1), dvaa005. doi: 10.1093/eep/dvaa005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Patti GJ, Yanes O, & Siuzdak G. (2012). Innovation: Metabolomics: the apogee of the omics trilogy. Nat Rev Mol Cell Biol, 13(4), 263–269. doi: 10.1038/nrm3314 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Petrick LM, Schiffman C, Edmands WMB, Yano Y, Perttula K, Whitehead T, . . . Rappaport SM (2019). Metabolomics of neonatal blood spots reveal distinct phenotypes of pediatric acute lymphoblastic leukemia and potential effects of early-life nutrition. Cancer Lett, 452, 71–78. doi: 10.1016/j.canlet.2019.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Petrick LM, Uppal K, & Funk WE (2020). Metabolomics and adductomics of newborn bloodspots to retrospectively assess the early-life exposome. Curr Opin Pediatr, 32(2), 300–307. doi: 10.1097/MOP.0000000000000875 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Plon SE, & Lupo PJ (2019). Genetic Predisposition to Childhood Cancer in the Genomic Era. Annu Rev Genom Hum Genet. [DOI] [PubMed] [Google Scholar]
  • [72].Rattray NJW, Deziel NC, Wallach JD, Khan SA, Vasiliou V, Ioannidis JPA, & Johnson CH (2018). Beyond genomics: understanding exposotypes through metabolomics. Hum Genomics, 12(1), 4. doi: 10.1186/s40246-018-0134-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Reese SE, Zhao S, Wu MC, Joubert BR, Parr CL, Haberg SE, . . . London SJ (2017). DNA Methylation Score as a Biomarker in Newborns for Sustained Maternal Smoking during Pregnancy. Environ Health Perspect, 125(4), 760–766. doi: 10.1289/ehp333 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Richmond RC, Simpkin AJ, Woodward G, Gaunt TR, Lyttleton O, McArdle WL, . . . Relton CL (2015). Prenatal exposure to maternal smoking and offspring DNA methylation across the lifecourse: findings from the Avon Longitudinal Study of Parents and Children (ALSPAC). Hum Mol Genet, 24(8), 2201–2217. doi: 10.1093/hmg/ddu739 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Rothman KJ (1993). Methodologic frontiers in environmental epidemiology. Environ Health Perspect, 101 Suppl 4, 19–21. doi: 10.1289/ehp.93101s419 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Schraw JM, Junco JJ, Brown AL, Scheurer ME, Rabin KR, & Lupo PJ (2019). Metabolomic profiling identifies pathways associated with minimal residual disease in childhood acute lymphoblastic leukaemia. EBioMedicine, 48, 49–57. doi: 10.1016/j.ebiom.2019.09.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [77].Shenker NS, Ueland PM, Polidoro S, van Veldhoven K, Ricceri F, Brown R, . . . Vineis P. (2013). DNA methylation as a long-term biomarker of exposure to tobacco smoke. Epidemiology, 24(5), 712–716. doi: 10.1097/EDE.0b013e31829d5cb3 [DOI] [PubMed] [Google Scholar]
  • [78].Spector LG, Murphy SE, Wickham KM, Lindgren B, & Joseph AM (2014). Prenatal tobacco exposure and cotinine in newborn dried blood spots. Pediatrics, 133(6), e1632–1638. doi: 10.1542/peds.2013-3118 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [79].St. Jude Children’s Research Hospital. (2020). St. Jude Cloud PeCan. Retrieved 11/8/20, from https://pecan.stjude.cloud/
  • [80].Tiziani S, Kang Y, Harjanto R, Axelrod J, Piermarocchi C, Roberts W, & Paternostro G. (2013). Metabolomics of the tumor microenvironment in pediatric acute lymphoblastic leukemia. PLoS One, 8(12), e82859. doi: 10.1371/journal.pone.0082859 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [81].van Dijk SJ, Peters TJ, Buckley M, Zhou J, Jones PA, Gibson RA, . . . Molloy PL (2018). DNA methylation in blood from neonatal screening cards and the association with BMI and insulin sensitivity in early childhood. Int J Obes (Lond), 42(1), 28–35. doi: 10.1038/ijo.2017.228 [DOI] [PubMed] [Google Scholar]
  • [82].Verma P, Devaraj J, Skiles JL, Sajdyk T, Ho RH, Hutchinson R, . . . Ramkrishna D. (2020). A Metabolomics Approach for Early Prediction of Vincristine-Induced Peripheral Neuropathy. Sci Rep, 10(1), 9659. doi: 10.1038/s41598-020-66815-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [83].Wellcome Sanger Institute. (2020). COSMIC: Catalogue Of Somatic Mutations In Cancer. Retrieved 11/8/20, from https://cancer.sanger.ac.uk/cosmic
  • [84].Wessels D, Barr DB, & Mendola P. (2003). Use of biomarkers to indicate exposure of children to organophosphate pesticides: implications for a longitudinal study of children’s environmental health. Environ Health Perspect, 111(16), 1939–1946. doi: 10.1289/ehp.6179 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Wilson SL, & Robinson WP (2018). Utility of DNA methylation to assess placental health. Placenta, 64 Suppl 1, S23–S28. doi: 10.1016/j.placenta.2017.12.013 [DOI] [PubMed] [Google Scholar]
  • [86].Wishart DS (2019). NMR metabolomics: A look ahead. J Magn Reson, 306, 155–161. doi: 10.1016/j.jmr.2019.07.013 [DOI] [PubMed] [Google Scholar]
  • [87].Yang J, Pearl M, Jacob P 3rd, DeLorenze GN, Benowitz NL, Yu L, . . . Kharrazi M. (2013). Levels of cotinine in dried blood specimens from newborns as a biomarker of maternal smoking close to the time of delivery. Am J Epidemiol, 178(11), 1648–1654. doi: 10.1093/aje/kwt182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Yeung EH, Bell EM, Sundaram R, Ghassabian A, Ma W, Kannan K, & Louis GM (2019). Examining Endocrine Disruptors Measured in Newborn Dried Blood Spots and Early Childhood Growth in a Prospective Cohort. Obesity (Silver Spring), 27(1), 145–151. doi: 10.1002/oby.22332 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Zar Kyaw T, Yamaguchi S, Imai C, Uematsu M, & Sato N. (2019). The utility of post-test newborn blood spot screening cards for epigenetic association analyses: association between HIF3A methylation and birth weight-for-gestational age. J Hum Genet, 64(8), 795–801. doi: 10.1038/s10038-019-0621-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Zhang Y, Schottker B, Florath I, Stock C, Butterbach K, Holleczek B, . . . Brenner H. (2016). Smoking-Associated DNA Methylation Biomarkers and Their Predictive Value for All-Cause and Cardiovascular Mortality. Environ Health Perspect, 124(1), 67–74. doi: 10.1289/ehp.1409020 [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES