1,320
Views
2
CrossRef citations to date
0
Altmetric
Review Article

Breast cancer and environmental contamination: A real connection?

& | (Reviewing editor)
Article: 1520470 | Received 07 Jul 2016, Accepted 03 Sep 2018, Published online: 12 Oct 2018

Abstract

Breast cancer is the most prevalent cancer among women and the second cause of cancer-related death in the world. This review describes the effects of bisphenol A, phthalates, and parabens, important environmental chemicals that have been associated with developing breast cancer. With more or less success, most of the studies have failed to establish a definitive correlation between cause and effect. The reason for these discrepancies and lack of consistency seems clear in some cases but is blurred in others. Here, we outline the facts reported in the literature and suggest that more studies should be done to clarify gene–environment interactions that could lead to breast cancer, and to identify groups of women that could be at higher risk according to their epigenome, since it seems that environmental chemicals are more harmful than previously thought.

PUBLIC INTEREST STATEMENT

Breast cancer is the most prevalent cancer among women and the second cause of cancer-related death in the world. Unfortunately, different environmental pollutants have been associated with developing breast cancer. Among those environmental toxins are a group of chemical consumer products that can be found in household items, personal care products, pharmaceuticals, insecticides, food and beverage cans and bottles, and drinking water. Several of them behave like estrogens, hormones that are required for the development of female secondary sexual characters that have also been linked to breast cancer. It has been suggested that they can increase the risk of breast cancer during periods of major susceptibility of the breast and even pass to infants through breast milk. Nowadays, the association between environmental toxic chemicals and breast cancer is still controversial and is imperative to determine if this association is real.

1. Introduction

Breast cancer is the most frequent type of cancer among women in the world and its incidence has increased in the past 25 years. The highest incidence is in non-Hispanic white and non-Hispanic black women, followed by American Indian/Alaska Native, Hispanic/Latina, and Asian/Pacific Islander. These incidence differences in breast cancer between race and ethnic categories are more probably related with lifestyle factors (Gray, Rasanayagam, Engel, & Rizzo, Citation2017; American Cancer Society, Citation2017–2018). In addition, environment, epigenetic modifications, miRNAs, and single nucleotide polymorphisms are associated with increased risk in developing breast cancer. Other mayor risk factors for breast cancer are age, dense breasts, longer time breast tissue exposure to estrogen (menarche before age 12; menopause after age 55), giving birth late in life or never given birth, no breastfeeding, hormone replacement therapy, breast/chest radiation therapy, obesity, sedentary lifestyle, stress, and shiftwork (Fletcher & Dudbridge, Citation2014; Gray et al., Citation2017; Loi et al., Citation2005; Montenegro et al., Citation2016; O’Brien et al., Citation2016; Samantarrai, Dash, Chhetri, & Mallick, Citation2013). In addition, only 5–10% of breast cancers are hereditary, and most of them are due to mutations in BRCA1 and BRCA2 genes, but other genes are also associated (Economopoulou, Dimitriadis, & Psyrri, Citation2015).

Since estrogens have been linked to breast cancer, it is important to consider that several synthetic chemicals behave like estrogens, increasing the risk of breast cancer during periods of major susceptibility of the breast, such as puberty and in utero. Additionally, some of those chemicals can pass to infants through breast milk (Fourth National Report on Human Exposure to Environmental Chemicals, Citation2017; Rudel, Gray, et al., Citation2011; Rudolph, Chang-Claude, & Schmidt, Citation2016). Thousands of different toxic environmental synthetic chemicals are used daily, and as the use of them has risen, so have been the rates of breast cancer. Hence, the research on the association among these new substances and breast cancer incidence is crucial. Importantly, nonindustrialized countries present lower rates of breast cancer compared to industrialized countries (Gray et al., Citation2017; Rodgers, Udesky, Rudel, & Green, Citation2018; Rudel, Ackerman, Attfield, & Brody, Citation2014; Witorsch & Thomas, Citation2010). However, it is difficult to determine how the exposure to toxic environmental chemicals affects the risk in developing breast cancer. The environment is a complex world and we are daily exposed to different toxic chemicals, and by the time breast cancer is diagnosed, a long period has passed, complicating the possible correlations.

The purpose of this report is to review the literature on some of the most common and still used environmental consumer product chemical toxins, bisphenol A (BPA), phthalates, and parabens, that have been associated with the development of breast cancer (Table ). An overview of research articles about these three compounds and their findings are summarized in Tables , respectively. Nowadays, the association between environmental toxic chemicals and breast cancer is still controversial and is imperative to determine if a particular chemical, or a combination of them, is causing breast cancer.

Table 1. Sources of exposure of bisphenol A, phthalates, and parabens

Table 2. Bisphenol A and breast cancer research findings

Table 3. Phthalates and breast cancer research findings

Table 4. Parabens and breast cancer research findings

2. BPA

BPA is found in dentistry composites, paints, plastics, and toys and is used to cover food and beverage cans, polycarbonate beverage bottles, and no long ago, baby bottles, and infant formula packaging. BPA has also been detected in air, house dust, rivers, and drinking water (Matsumoto, Adachi, & Suzuki, Citation2005; Rodriguez-Mozaz, de Alda, & Barceló, Citation2005) (Table ). Considering its broad use in food-related storage items, the exposure of humans to BFA is ubiquitous and has been detected in urine (Doerge, Twaddle, Vanlandingham, & Fisher, Citation2010; Hengstler et al., Citation2011; Lakind & Naiman, Citation2011; Rudel, Gray, et al., Citation2011; Rudel, Fenton, Ackerman, Euling, Makris, Citation2011; Vandenberg et al., Citation2010).

BPA is considered an estrogen-disruptor compound (EDC), since it binds to estrogen receptors (ERs), but its affinity is 1,000–10,000 lower than endogenous estrogen (Hengstler et al., Citation2011; Melzer et al., Citation2011). Following oral administration in live animals and humans, BPA is rapidly and almost completely metabolized in the gastrointestinal tract to an inactive form, and its primary metabolite does not bind to ER receptors, neither in vivo nor in vitro (Doerge et al., Citation2010; Frederick et al., Citation2014; Hengstler et al., Citation2011; Yang & Fisher, Citation2014). In humans, BPA derivatives are extensively excreted in urine, and after 3 days without eating packaged foods, BPA’s urine levels decrease about 65%. Other studies have demonstrated that BPA is mostly eliminated at higher concentrations through sweat (Frederick et al., Citation2014; Genuis, Beesoon, Birkholz, & Lobo, Citation2012; Rudel, Fenton, et al., Citation2011; Stahlhut, Welshons, & Swan, Citation2009; Yang & Fisher, Citation2014). BPA has also been detected in premature infants, breast milk, amniotic fluid, placental tissue, and umbilical cord (Calafat et al., Citation2009; Ikezuki, Tsutsumi, Takai, Kamei, & Taketani, Citation2002; Schönfelder et al., Citation2002; Sun et al., Citation2004). Another way of BPA exposure is through inhalation and skin absorption, routes of more concern, since unconjugated BPA may circulate longer periods in bloodstream compared to ingested BPA (Hengstler et al., Citation2011; Vom Saal & Welshons, Citation2014).

A study performed by Churchwell et al. (Citation2014) showed that the presence of BPA in food does not exert any pathological effects in the fetus, even though the doses of BPA orally exposed to the rat expecting dam and newborn pups were much higher than an expected human daily intake. Furthermore, studies performed by the FDA’s National Center for Toxicological Research researchers have shown that the BPA derived from food that could pass to the fetus is extremely low. Even the feeding of pregnant rodents with 100–1,000 times more BPA than what a women could be exposed do not lead to detectable active BPA in the fetus after mother’s exposure; in addition, it is considered that human infant exposure is as much as 92% less than earlier estimations (Churchwell et al., Citation2014; Delclos et al., Citation2014; Melnick et al., Citation2002).

On the other side, several studies suggest that BPA increase the risk of breast cancer, especially if the exposure is early in life. There are animal studies indicating that fetal environmental exposure, and also during lactation, can affect mammary gland development, even at low doses, and those changes were associated with development of breast cancer during adulthood (Acevedo, Davis, Schaeberle, Sonnenschein, & Soto, Citation2013; Betancourt, Eltourn, Desmond, Russo, & Lamartiniere, Citation2010; Mandrup, Boberg, Isling, Christiansen, & Hass, Citation2016; Paulose, Speroni, Sonnenschein, & Soto, Citation2015; Weber & Keri, Citation2011). In addition, rats exposed to BPA in womb presented abnormalities in adult mammary tissue and alterations in the milk protein content by the time they were feedings their pups (Kass, Altamirano, Bosquiazzo, Luque, & Muñoz-de-Toro, Citation2012). It has also been shown that mice prenatal exposure to BPA alters gene transcription of epithelial cellular genes and genes that are associated with the stroma of the mouse fetal mammary gland. Considering that BPA is an EDC, after birth, BFA exposure alters the sensitivity to estradiol-dependent mammary gland development and progesterone-dependent mammary cell proliferation. Additionally, BPA affects both ER-dependent and ER-independent pathways (Ibrahim, Elbakry, & Bayomy, Citation2016; Paulose et al., Citation2015; Zhang et al. Citation2015).

In the study by Jenkins, Wang, Eltoum, Desmond, and Lamartiniere (Citation2011), adult mice chronically exposed to low doses of BPA showed accelerated mammary tumorigenesis and metastasis, and only higher doses increased apoptosis. It has also been shown that the expression of several different genes involved in hormone-mediated pathways, cell proliferation, apoptosis, and cancer were altered in normal or cancerous human breast cells that were exposed to low levels of BPA. And the cells coming from the noncancerous breast also presented a gene expression profile similar to the pattern found in the development of aggressive tumors (Dairkee, Luciani-Torres, Moore, & Goodson III, Citation2013; Dairkee et al., Citation2008; Weng et al., Citation2010). In addition, Palacios-Arreola, Nava-Castro, Del Río-Araiza, Pérez-Sánchez, and Morales-Montor (Citation2017) induced the growth of a mammary adenocarcinoma tumor implanted in the mammary gland of mice and demonstrated that a single dose of BPA at postnatal day 3 affected the immune system and led to an increased tumor size during adulthood. The BPA exposure did not alter puberty onset but affected the expression of ERα in immune cells. These results suggest that early life BPA exposure can induce breast cancer development by altering the antitumoral immune response.

It has also been shown that BPA exposure alters the imprinting of particular human and rodent genes, such as LAMP3, important protein in invasive cancers (Dhimolea et al., Citation2014; Susiarjo, Sasson, Mesaros, & Bartolomei, Citation2013; Weng et al., Citation2010). In addition, studies performed in human MCF-7 breast cancer cell line, and in mice mammary glands exposed to BPA in utero, showed alterations in the expression of the histone methyltransferase EZH2 (Doherty, Bromer, Zhou, Aldad, & Taylor, Citation2010) and the transcription of the long noncoding RNA HOTAIR (Bhan et al., Citation2014), both linked to breast cancer. Thus, it is possible that BPA has other important targets affecting breast tumorigenesis.

3. Phthalates

Phthalates are a group of chemicals used to make polyvinyl chloride flexible and stable. They are found in different household items, medical devices, dentistry, vinyl flooring, automobiles, detergents, adhesives, insecticides, and toys. Phthalates are also used in personal care products, because they increment spreadability. Foods can be contaminated with phthalates by processing or packaging, and when food is heated in plastic containers in the microwave, they can be released into aqueous solution (Table ). Milk, medications and nutritional supplements, and ground and drinking water can contain phthalates. These chemicals can also be absorbed through the skin or breathed in from house dust or fumes (Breast Cancer & the Environment Research Centers, Citation2007; Center for Disease Control and Prevention, Citation2015).

Among the most common phthalates are butyl benzyl phthalate (BBP), di-n-butylphthalate (DBP), di-(2-ethylhexyl)phthalate (DEHP), diethylphthalate (DEP), di-isobutylphthalate (DiBP), di-isodecylphthalate, di-isononylphthalate, di-methylphthalate, di-n-hexylphthalate, and di-n-octylphthalate. The different phthalates are metabolized undergoing some specific biotransformations and are eliminated in urine and feces (Breast Cancer & the Environment Research Centers, Citation2007; Janjua, Frederiksen, Skakkebaek, Wulf, & Andersson, Citation2008; Meeker, Calafat, & Hauser, Citation2012); they have also been detected in human amniotic fluid and human breast milk (Hines, Calafat, Silva, Mendola, & Fenton, Citation2009; Silva et al., Citation2004). It has been demonstrated by Cirillo, Fasano, Castaldi, Montuori, and Amodio (Citation2011) that packed school meals contained DEHP and DBP with percentage values higher than the allowed tolerable daily intake.

BBP and DBP behave as weak estrogens in cell culture systems; DBP, DiBP, and BBP also bind weakly to androgen receptors (AR). Moreover, several phthalates simultaneously behave as agonists and/or antagonists of one or more hormone receptors, and others do not bind to either ER or AR (Czernych, Chraniuk, Zagożdżon, & Wolska, Citation2017; Okubo, Suzuki, Yokoyama, Kano, & Kano, Citation2003; Takeuchi et al., Citation2005). By binding to AR in utero and during lactation, phthalates alter rodent male sexual development, affecting the reproductive tract and fertility (Gray et al., Citation2000). The study by Gray, Laskey, and Ostby (Citation2006), in female rats, showed that oral administration of DBP during long periods, at doses that induce sexual alterations in male rat, triggers abortion in more than 50% female rats. Moyer and Hixon (Citation2012) showed that pregnant mice orally exposed to mono-(2-ethylhexyl) phthalate (MEHP), during gestational days 17–19, resulted in sexual alterations in F1 adult females, leading to a reduction in fertility and mammary gland hyperplasia. Other studies have shown that exposure to DEHP in utero, and prepubescent rats, retards the onset of puberty (Grande, Andrade, & Talsness, Citation2006; Ma, Kondo, & Ban, Citation2006), and BBP exposure in utero affects rat mammary gland morphology, retards puberty, and alters gene expression profile to a more carcinogenic type (Moral et al., Citation2011). Studies in humans have shown that girls going through thelarche at earlier years had elevated levels of phthalates compared to girls without premature breast development (Chou, Huang, Lee, Wu, & Lin, Citation2009; Colón, Caro, Bourdony, & Rosario, Citation2000). But a report by Lomenick et al. (Citation2010) suggested that phthalate exposure was not associated with girls premature puberty.

BBP also binds to aryl hydrocarbon receptor (AhR) and induces cancer stem-cell proliferation enhancing metastasis (Wang et al., Citation2016). A study by Hsieh, Tsai, Hsu, Kuo, and Lee, et al. (Citation2012), performed with MDA-MB-231 ER(−) breast cancer cell line, demonstrated that BBP and DBP treatments stimulated AhR, activating the downstream cAMP-PKA-CREB1 signaling pathway, inducing cell proliferation, migration, invasion, and tumor formation by non-estrogenic mechanisms. Additionally, it has been shown that phthalates activate PPAR-α (peroxisome proliferator-activated receptor) and can exert carcinogenic effects by other cellular pathways that do not involve ER and AR (Das, Singh, & Thakur, Citation2014; Venkata et al., Citation2006).

In order to clarify the effects of BBP, the most widely used phthalate, on genes transcriptions in human breast cancer cells, Hsieh, Tsai, Hsu, Kuo, Hsi, et al. (Citation2012) analyzed gene expression changes induced by BBP treatment to a whole human genome cDNA microarray. They focused in genes that changed their transcription pattern in more than 10-fold compared to the control and found upregulation and downregulation of particular genes associated with cell proliferation, epithelial–mesenchymal transition, and angiogenesis. It has also been shown that phthalates induce epigenetics alterations. Kang and Lee (Citation2005) demonstrated that phthalates can induce changes in the epigenome of normal (MCF10A) and breast cancer cells (MCF-7) after BBP exposure, leading to demethylation of ERα promoter. In addition, human studies on cord blood samples from newborns showed that in utero phthalate exposure modifies the DNA methylation pattern of BRCA1 gene and others genes involved in cancer, endocrine function, inflammatory response, and male fertility (Solomon et al., Citation2017).

The hospital-based case-control study by López-Carrillo et al. (Citation2010) evaluated the presence and concentration of phthalate metabolites in women’s urine samples and suggested an association between DEP and an increased risk in developing breast cancer. Additionally, the hospital-based case-control study by Holmes et al. (Citation2014) suggested an association between DEHP exposure (parent phthalate of MEHP metabolite) and breast cancer, even though the study was limited due to the small sample size.

Since it is known that estrogens induce drug resistance to the chemotherapeutic drug tamoxifen (Shiau et al., Citation1998), the action of phthalates was investigated in MCF-7 ER(+) breast cancer cells. It was demonstrated that BBP, DBP, and DEHP induce cell proliferation in MCF-7 cells, but not in ER(−) MDA-MB-231 breast cancer cells, and phthalates were also able to inhibit tamoxifen induced apoptosis in MCF-7 cells, demonstrating that phthalates mimicked estrogen effects. In addition, it has been demonstrated that BBP induced chemoresistance by decreasing doxorubicin/cyclophosphamide-induced apoptosis by altering the breast cancer microenvironment in mice (Hsu et al., Citation2015).

4. Parabens

The five most commonly used parabens are methylparaben, ethylparaben, n-propylparaben, n-butylparaben, and isobutylparaben. Parabens are found in personal care products, foods, beverages, and pharmaceuticals (Table ). They act as preservatives due to their bactericidal and fungicidal properties (Błędzka, Gromadzińska, & Wąsowicz, Citation2014; Shen, Jiang, Mao, Pan, & Cao, Citation2007).

Parabens have been measured in human breast milk, urine, amniotic fluid, serum, and seminal fluid. The concentrations of parabens in adult’s urine tend to be higher in women than in men, and lower in children than in adults, probably due to differences in the use of personal care products. Since parabens are quickly eliminated from the body, the exposure time is reduced. But, considering that they are constantly used, we have them continually present in our bodies (Calafat, Ye, Wong, Bishop, & Needham, Citation2010; Philippat et al., Citation2013; Sandager et al., Citation2011; Schlumpf et al., Citation2010). Parabens are considered weak EDCs, since they bound 10,000–100,000 times less strongly to ERs compared to estrogen (Routledge, Parker, Odum, Ashby, & Sumpter, Citation1998; Watanabe et al., Citation2013). But, since picogram concentrations of estradiol/gram of breast tissue and microgram concentrations of parabens/gram of breast tissue have been detected, the high concentration exposure could compensate the lower potency of parabens (Barr, Metaxas, Harbach, Savoy, & Darbre, Citation2012; Boberg, Taxvig, Christiansen, & Hass, Citation2010; Harvey & Everett, Citation2012). The more abundant parabens present in human primary breast cancer tissues have been n-propylparaben and methylparaben (Darbre, Citation2003; Sandager et al., Citation2011). In addition, the axilla was the prevalent region where n-propylparaben was detected and correlates with the fact that the incidence of breast cancer in the outer quadrant of the breast exceeds 50% of breast cancer cases in the United Kingdom. These results are consistent with the postulate that dermal applications of personal care products in underarm are coincident with this route of exposure, since oral route will implicate metabolic processes in the gut and liver that will produce p-hydroxybenzoic metabolite (Darbre, Citation2005; Darbre & Charles, Citation2010). However, the study by Barr et al. (Citation2012) also mentioned that the actual source of paraben cannot be identified, because 17.5% of the patients never used underarm cosmetics. The presence of parabens in the majority of the breast tissue samples does not mean they caused breast cancer but requires further investigation (Barr et al., Citation2012; Harvey & Everett, Citation2012).

Parabens have also been involved in cell growth and proliferation, and since combinations of them are present in human breast tissue at concentrations that can induce proliferation of MCF-7 breast cancer cell line, it implies that they could affect human breast tissue. In addition, certain parabens can induce migration and invasion of human breast cancer cells and affect apoptosis (Charles & Darbre, Citation2013; Darbre & Harvey, Citation2014; Gopalakrishan et al., Citation2017; Khanna, Dash, & Darbre, Citation2014; Wróbel & Gregoraszczuk, Citation2014). The study by Goodson et al. (Citation2011) showed that methylparaben induces cell cycling and favors apoptosis evasion in human breast epithelial cells, important phenomena for malignant transformation. In addition, DNA damage, chromosome aberrations, and gene expression profiles that resemble estrogen stimulation after exposure to methyl- and butylparabens have been reported (Pugazhendhi, Sadler, & Darbre, Citation2007).

Another relevant aspect to consider is that the human epidermal growth factor receptor HER2, overexpressed in ~25% breast tumors and considered a detrimental prognostic indicator, enhanced the potency of parabens by inducing oncogene expression and breast cancer cell proliferation in vitro, indicating that lower doses of parabens are required to induce breast cancer (Pan et al., Citation2016). Then, the reevaluation of xenoestrogens in the presence of HER ligands should be considered, since studies evaluating parabens alone can underestimate their proliferative effects.

5. Conclusions

Thus far, the relationship between environmental and toxic chemicals is still controversial. It constitutes a great challenge to determine if particular chemical, or a combination of them, is causing breast cancer. And since breast cancer incidence increases as countries move forward into industrialization, environmental toxic exposure constitutes a relevant public health concern.

An important aspect to be evaluated is the type of exposure that people usually faces, either at home, at work or just walking, or driving. And in view of the fact that exposures are usually a mixture of chemical compounds, with a wide range of concentrations and variable periods of exposure, the interpretation of the results gets harder. In that way, epidemiological studies should divide participants according to the type of exposure to get more reliable and convincing results. Furthermore, the direct measurement of the toxic chemical exposure during the years previous of breast cancer diagnosis is missing. This is an important aspect to consider, since some chemicals accumulate over time, and tumorigenic phenotype acquisition usually takes years. Moreover, given the multifactorial nature of the problem, it should be relevant to determine the genome haplotype of the participants in any study, to find whether polymorphisms show predisposition or resistance to the environmental insult being tested.

Additionally, it would be important to estimate how exposure during the most critical and vulnerable periods of breast development (in womb, newborn, puberty) correlates with the risk of developing breast cancer during adulthood. In that way, protocols should be standardized evaluating the range of doses of the environmental chemicals adjusted in regard to different stages of development. Also, from the point of view of predictive chemistry, it would be important to improve the tools to identify the potential chemicals that could induce cancer, as well as methods to measure them, avoiding possible contaminations. In this context, it would be interesting to know the methylation profile of the individual´s genome to evaluate whether particular changes in imprinting fingerprints correlate with cancer outcome upon chemical exposure.

In an effort to determine the breast cancer potential of environmental chemicals, the studies performed in animals are very helpful, but have to be more precise, considering the windows of vulnerability of the mammary tissue and the correlation between doses at which humans could be daily exposed. In other words, even though there are many examples of mechanistic explanations and discoveries done in mouse models that effectively mimic human diseases, significant differences remain and have to be considered when analyzing the data. These differences have all been shown to be, in various aspects, associated with cancer, such as mouse life span, differences in gut microbiota, the effect of the central nervous system on chronic inflammation and possibly cancer, and the immune system, central to the tumorigenesis and its progression, the epigenome regulatory differences, and lately, the systemic effects of tumors on the organism. However, in spite of this, it is still worth to rely on animals studies as bona fide approximations.

Some studies did not show positive epidemiological evidence. The epidemiological investigations should consist of single cohorts evaluating exposures early in life with continual assessments to be more precise and informative and considering unexposed populations to discard the baseline risk. Also, different but relevant aspects should be all together evaluated, such as alteration of mammary gland development and the activation of particular receptors, like ER and AhR, which impact different cellular pathways leading to breast cancer. Since endocrine disruption as well as genotoxicity play critical roles in cancer development, both biological activities have to be evaluated to determine how the cellular pathways are affected to find a way to counteract them. The alternative approaches should evaluate diverse and relevant biological activities transversely to multiple chemicals, such as estrogenic/antiestrogenic activity and DNA damage and epimutations. If we are able to comprehend the early events that could lead to genetic- or epimutations that could induce breast cancer, we will have more opportunities to avoid those risks and prevent the development of breast cancer later in life. Thus, in addition to previous suggestions outlined above, all available technology aimed to determine epigenetic modifications under various conditions should be attempted.

More studies should be done to search for the extensive number of toxic chemicals to understand the gene–environment interactions that could lead to breast cancer and identify groups of women that could be at higher risk. In addition, it is really important to understand how the environment impacts or affects breast cancer risk, and for that we need to count on better chemicals tests, because it seems that environmental chemicals are more harmful than previously thought.

Competing interests

The authors declare no competing interests.

Additional information

Funding

The authors received no direct funding for this research.

Notes on contributors

Fernando Delgado-López

Fernando Delgado-López, biochemist, worked at the Molecular Pharmacolgy Program at Memorial Sloan-Kettering Cancer Center (New York) from 1994 to 1997. He obtained his PhD from Albert Einstein College of Medicine, Bronx, New York, in 2006. Later, he made postdoctoral stays at the Pharmacology Department at Yale University, New Haven, Conneticut (2007) and at Montefiore Medical Center, Bronx, New York (2008-2010). He is Associate Professor at the Division of Medicine in Universidad Católica del Maule, since 2012. Dr. Delgado-López focuses on the study of adenoviral protein RID on inflammatory signaling at the tumor microenvironment.

S. Pilar Zamora-León

S. Pilar Zamora-León is a biochemist from Universidad Austral de Chile and obtained her Master of Science and PhD degree from Albert Einstein College of Medicine, New York, USA. She worked a couple of years at Memorial Sloan-Kettering Cancer Center (New York, USA). She returned to Chile, her country, in 2004, and since then she has been working at the Universidad Católica del Maule. Currently, she is Associate Professor at the Faculty of Medicine. Part of her research work has been focused on cancer, particularly in vitro studies with human breast cancer cell lines.

References

  • Acevedo, N., Davis, B., Schaeberle, C. M., Sonnenschein, C., & Soto, A. M. (2013). Perinatally administered bisphenol A as a potential mammary gland carcinogen in rats. Environment Health Perspectives, 121(9), 1040–1046. doi:10.1289/ehp.1306734
  • American Cancer Society. (2017). Breast cancer facts & figures 2017–2018. Atlanta: American Cancer Society, Inc.. https://www.cancer.org/content/dam/cancer-org/research/cancer-facts-and-statistics/breast-cancer-facts-and-figures/breast-cancer-facts-and-figures-2017-2018.pdf
  • Barr, L., Metaxas, G., Harbach, C. A., Savoy, L. A., & Darbre, P. D. (2012). Measurement of paraben concentrations in human breast tissue at serial locations across the breast from axilla to sternum. Journal Applications Toxicogical, 32(3), 219–232. doi:10.1002/jat.v32.3
  • Betancourt, A., Eltourn, I., Desmond, R., Russo, J., & Lamartiniere, C. (2010). In utero exposure to bisphenol A shifts the window of susceptibility for mammary carcinogenesis in the rat. Environment Health Perspectives, 118(11), 1614–1619. doi:10.1289/ehp.1002148
  • Bhan, A., Hussain, I., Ansari, K. I., Bobzean, S. A., Perrotti, L. I., & Mandal, S. S. (2014). Bisphenol-A and diethylstilbestrol exposure induces the expression of breast cancer associated long noncoding RNA HOTAIR in vitro and in vivo. Journal Steroid Biochemical Molecular Biologic, 141, 160–170. doi:10.1016/j.jsbmb.2014.02.002
  • Błędzka, D., Gromadzińska, J., & Wąsowicz, W. (2014). Parabens. From Environmental Studies to Human Health, 67, 27–42.
  • Boberg, J., Taxvig, C., Christiansen, S., & Hass, U. (2010). Possible endocrine disrupting effects of parabens and their metabolites. Reproduction Toxicogical, 30(2), 301–312. doi:10.1016/j.reprotox.2010.05.084
  • Breast Cancer & the Environment Research Centers. 2007. Early life exposure to phthalates and breast cancer risk in later years. Fact sheet on phthalates, https://www.google.cl/url?sa=t&rct=j&q=&esrc=s&source=web&cd=6&cad=rja&uact=8&ved=0ahUKEwiPw6GL2p3YAhXJgJAKHauHA-MQFghMMAU&url=https%3A%2F%2Fbcerp.org%2Fwp-content%2Fuploads%2F2017%2F01%2FFS4_BCERC.FactSheet_Phthalates.pdf&usg=AOvVaw2daCT_BYUG8QWvnvGDqCWl
  • Calafat, A. M., Weuve, J., Ye, X., Jia, L. T., Hu, H., & Ringer, S. (2009). Exposure to bisphenol A and other phenols in neonatal intensive care unit premature infants. Environment Health Perspectives, 117(4), 639–644. doi:10.1289/ehp.0800265
  • Calafat, A. M., Ye, X., Wong, L. Y., Bishop, A. M., & Needham, L. L. (2010). Urinary concentrations of four parabens in the U.S. population: NHANES 2005-2006. Environment Health Perspectives, 118(5), 679–685. doi:10.1289/ehp.0901560
  • Center for Disease Control and Prevention. National: biomonitoring program: Phthalates factsheet 2015, http://www.cdc.gov/biomonitoring/phthalates_factsheet.html.
  • Charles, A. K., & Darbre, P. D. (2013). Combinations of parabens at concentrations measured in human breast tissue can increase proliferation of MCF-7 human breast cancer cells. Journal Applications Toxicogical, 33(5), 390–398. doi:10.1002/jat.1792
  • Chou, Y. Y., Huang, P. C., Lee, C. C., Wu, M. H., & Lin, S. J. (2009). Phthalate exposure in girls during early puberty. Journal Pediatric Endocrinology Metabolic, 22(1), 69–77. doi:10.1515/JPEM.2009.22.1.69
  • Churchwell, M. I., Camacho, L., Vanlandingham, M. M., Twaddle, N. C., Sepehr, E., Delclos, K. B., … Doerge, D. R. (2014). Comparison of life-stage-dependent internal dosimetry for bisphenol A, ethinyl estradiol, a reference estrogen, and endogenous estradiol to test an estrogenic mode of action in Sprague Dawley rats. Toxicogical Sciences, 139(1), 4–20. doi:10.1093/toxsci/kfu021
  • Cirillo, T., Fasano, E., Castaldi, E., Montuori, P., & Amodio, C. R. (2011). Children’s exposure to di(2-ethylhexyl)phthalate and dibutylphthalate plasticizers from school meals. Journal Agricultural Food Chemical, 59(19), 10532–10538. doi:10.1021/jf2020446
  • Colón, I., Caro, D., Bourdony, C. J., & Rosario, O. (2000). Identification of phthalate esters in the serum of young Puerto Rican girls with premature breast development. Environment Health Perspectives, 108(9), 895–900.
  • Czernych, R., Chraniuk, M., Zagożdżon, P., & Wolska, L. (2017). Characterization of estrogenic and androgenic activity of phthalates by the XenoScreen YES/YAS in vitro assay. Environment Toxicogical Pharmacology, 53, 95–104. doi:10.1016/j.etap.2017.05.012
  • Dairkee, S. H., Luciani-Torres, G., Moore, D. H., & Goodson III, W. H. (2013). Bisphenol-A-induced inactivation of the p53 axis underlying deregulation of proliferation kinetics, and cell death in non malignant human breast epithelial cells. Carcinogenesis, 34(3), 703–712. doi:10.1093/carcin/bgs379
  • Dairkee, S. H., Seok, J., Champion, S., Sayeed, A., Mindrinos, M., Xiao, W., … Goodson, W. H. (2008). Bisphenol A induces a profile of tumor aggressiveness in high-risk cells from breast cancer patients. Cancer Research, 68(7), 2076–2080. doi:10.1158/0008-5472.CAN-07-6526
  • Darbre, P. D. (2003). Underarm cosmetics and breast cancer. Journal Applications Toxicogical, 23(2), 89–95. doi:10.1002/(ISSN)1099-1263
  • Darbre, P. D. (2005). Recorded quadrant incidence of female breast cancer in Great Britain suggests a disproportionate increase in the upper outer quadrant of the breast. Anticancer Researcher, 25(3c), 2543–2550.
  • Darbre, P. D., & Charles, A. K. (2010). Environmental oestrogens and breast cancer: Evidence for combined involvement of dietary, household and cosmetic xenoestrogens. Anticancer Research, 30(3), 815–828.
  • Darbre, P. D., & Harvey, P. W. (2014). Parabens can enable hallmarks and characteristics of cancer in human breast epithelial cells: A review of the literature with reference to new exposure data and regulatory status. Journal Applications Toxicogical, 34(9), 925–938. doi:10.1002/jat.v34.9
  • Das, M. T., Singh, M. K., & Thakur, I. S. (2014). Differential toxicological endpoints of di(2-ethylhexyl)phthalate (DEHP) exposure in MCF-7 and MDA-MB-231 cell lines: Possible estrogen receptorα (ERα) independent modulations. Indian Journal Experiments Biologic, 52(11), 1052–1061.
  • Delclos, K. B., Camacho, L., Lewis, S. M., Vanlandingham, M. M., Latendresse, J. R., Olson, G. R., … Thorn, B. T. (2014). Toxicity evaluation of bisphenol A administered by gavage to Sprague-Dawley rats from gestation day 6 through postnatal day 90. Toxicogical Sciences, 139(1), 174–197. doi:10.1093/toxsci/kfu022
  • Dhimolea, E., Wadia, P. R., Murray, T. J., Settles, M. L., Treitman, J. D., Sonnenschein, C., … Soto, A. M. (2014). Prenatal exposure to BPA alters the epigenome of the rat mammary gland and increases the propensity to neoplastic development. PLoS One, 9(7), e99800. doi:10.1371/journal.pone.0099800
  • Doerge, D. R., Twaddle, N. C., Vanlandingham, M., & Fisher, J. W. (2010). Pharmacokinetics of bisphenol A in neonatal and adult Sprague-Dawley rats. Toxicogical Applications Pharmacology, 247(2), 158–165. doi:10.1016/j.taap.2010.06.008
  • Doherty, L. F., Bromer, J. G., Zhou, Y., Aldad, T. S., & Taylor, H. S. (2010). In utero exposure to diethylstilbestrol (DES) or bisphenol-a (BPA) increases EZH2 expression in the mammary gland: An epigenetic mechanism linking endocrine disruptors to breast cancer. Hormone Cancer, 1(3), 146–155. doi:10.1007/s12672-010-0015-9
  • Economopoulou, P., Dimitriadis, G., & Psyrri, A. (2015). Beyond BRCA: New hereditary breast cancer susceptibility genes. Cancer Trent Reviews, 41(1), 1–8. doi:10.1016/j.ctrv.2014.10.008
  • Fletcher, O., & Dudbridge, F. (2014). Candidate gene-environment interactions in breast cancer. BMC Medica, 12, 195–198. doi:10.1186/s12916-014-0141-2
  • Fourth national report on human exposure to environmental chemicals updated tables. January 2017, volume one. U.S. department of health and human services. Center for disease Control and Prevention, https://www.cdc.gov/exposurereport/pdf/FourthReport_UpdatedTables_Volume1_Jan2017.pdf.
  • Frederick, S., Vom Saal, C. A., VandeVoort, C. A., Taylor, J. A., Welshons, W. V., Toutain, P.-L., & Hunt, P. A. (2014). Bisphenol A (BPA) pharmacokinetics with daily oral bolus or continuous exposure via silastic capsules in pregnant rhesus monkeys: Relevance for human exposures. Reproduction Toxicogical, 45, 105–116. doi:10.1016/j.reprotox.2014.01.007
  • Genuis, S. J., Beesoon, S., Birkholz, D., & Lobo, R. A. (2012). Human excretion of bisphenol A: Blood, urine, and sweat (BUS) study. Journal Environment Public Health, 2012, 185731. doi:10.1155/2012/185731
  • Goodson, W. H., Luciani, M. G., Sayeed, S. A., Jaffee, I. M., Moore, D. H., & Dairkee, S. H. (2011). Activation of the mTOR pathway by low levels of xenoestrogens in breast epithelial cells from high-risk women. Carcinogenesis, 32(11), 1724–1733. doi:10.1093/carcin/bgr196
  • Gopalakrishan, K., Teitelbaum, S. L., Lambertini, L., Wetmur, J., Manservisi, F., Luciani, M. G., … Chen, J. (2017). Changes in mammary histology and transcriptome profiles by low-dose exposure to environmental phenols at critical periods of development. Environment Researcher, 152, 232–243.
  • Grande, S. W., Andrade, A. J., & Talsness, C. E. (2006). A dose-response study following in utero and lactational exposure to di(2ethylhexyl)phthalate: Effects on female rat reproductive development. Toxicogical Sciences, 91(1), 247–254. doi:10.1093/toxsci/kfj128
  • Gray, J. M., Rasanayagam, S., Engel, C., & Rizzo, J. (2017). State of the evidence 2017: An update on the connection between breast cancer and the environment. Environment Health, 16(1), 94–155.
  • Gray, L. E., Jr, Laskey, J., & Ostby, J. (2006). Chronic di-n-butyl phthalate exposure in rats reduces fertility and alters ovarian function during pregnancy in female Long Evans hooded rats. Toxicogical Sciences, 93(1), 189–195. doi:10.1093/toxsci/kfl035
  • Gray, L. E., Jr, Ostby, J., Furr, J., Price, M., Veeramachaneni, D. N., & Parks, L. (2000). Perinatal exposure to the phthalates DEHP, BBP, and DINP, but not DEP, DMP, or DOTP, alters sexual differentiation of the male rat. Toxicogical Sciences, 58(2), 350–365. doi:10.1093/toxsci/58.2.350
  • Harvey, P. W., & Everett, D. J. (2012). Parabens detection in different zones of the human breast: Consideration of source and implications of findings. Journal Applications Toxicogical, 32(5), 305–309. doi:10.1002/jat.v32.5
  • Hengstler, J. G., Foth, H., Gebel, T., Kramer, P. J., Lilienblum, W., Schweinfurth, H., … Gundert-Remy, U. (2011). Critical evaluation of key evidence on the human health hazards of exposure to bisphenol A. Critical Reviews Toxicogical, 41(4), 263–291. doi:10.3109/10408444.2011.576008
  • Hines, E. P., Calafat, A. M., Silva, M. J., Mendola, P., & Fenton, S. E. (2009). Concentrations of phthalate metabolites in milk, urine, saliva, and serum of lactating North Carolina women. Environment Health Perspectives, 117(1), 86–92. doi:10.1289/ehp.11610
  • Holmes, A. K., Koller, K. R., Kieszak, S. M., Sjodin, A., Calafat, A. M., Sacco, F. D., … Rubin, C. H. (2014). Case-control study of breast cancer and exposure to synthetic environmental chemicals among Alaska Native women. International Journal Circumpolar Health, 73, 25760–25770. doi:10.3402/ijch.v73.25760
  • Hsieh, T. H., Tsai, C. F., Hsu, C. Y., Kuo, P. L., Hsi, E., Suen, J. L., … Tsai, E. M. (2012b). n-Butyl benzyl phthalate promotes breast cancer progression by inducing expression of lymphoid enhancer factor 1. PLoS One, 7(8), e42750. doi:10.1371/journal.pone.0042750
  • Hsieh, T. H., Tsai, C. F., Hsu, C. Y., Kuo, P. L., Lee, J. N., Chai, C. Y., … Tsai, E. M. (2012a). Phthalates induce proliferation and invasiveness of estrogen receptor-negative breast cancer through the AhR/HDAC6/c-Myc signaling pathway. FASEB Journal: Official Publication of the Federation of American Societies for Experimental Biology, 26(2), 778–787. doi:10.1096/fj.11-191742
  • Hsu, Y. L., Hung, J. Y., Tsai, E. M., Wu, C. Y., Ho, Y. W., Jian, S. F., … Kuo, P. L. (2015). Benzyl butyl phthalate increases the chemoresistance to doxorubicin/cyclophosphamide by increasing breast cancer-associated dendritic cell-derived CXCL1/GROα and S100A8/A9. Oncology Reports, 34(6), 2889–2900. doi:10.3892/or.2015.4307
  • Ibrahim, M. A. A., Elbakry, R. H., & Bayomy, N. A. (2016). Effect of bisphenol A on morphology, apoptosis and proliferation in the resting mammary gland of the adult albino rat. International Journal Experiments Pathologists, 97(1), 27–36. doi:10.1111/iep.12164
  • Ikezuki, Y., Tsutsumi, O., Takai, Y., Kamei, Y., & Taketani, Y. (2002). Determination of bisphenol A concentrations in human biological fluids reveals significant early prenatal exposure. Human Reproduction, 17(11), 2839–2841. doi:10.1093/humrep/17.11.2839
  • Janjua, N. R., Frederiksen, H., Skakkebaek, N. E., Wulf, H. C., & Andersson, A. M. (2008). Urinary excretion of phthalates and paraben after repeated whole-body topical application in humans. International Journal Andrology, 31(2), 118–130. doi:10.1111/j.1365-2605.2007.00784.x
  • Jenkins, S., Wang, J., Eltoum, I., Desmond, R., & Lamartiniere, C. A. (2011). Chronic oral exposure to bisphenol a results in a nonmonotonic dose response in mammary carcinogenesis and metastasis in MMTV-erbB2 mice. Environment Health Perspectives, 119(11), 1604–1609. doi:10.1289/ehp.119-a484
  • Kang, S. C., & Lee, B. M. (2005). DNA methylation of estrogen receptor alpha gene by phthalates. Journal Toxicogical Environment Health A, 68(23–24), 1995–2003. doi:10.1080/15287390491008913
  • Kass, L., Altamirano, G. A., Bosquiazzo, V. L., Luque, E. H., & Muñoz-de-Toro, M. (2012). Perinatal exposure to xenoestrogens impairs mammary gland differentiation and modifies milk composition in Wistar rats. Reproduction Toxicogical, 33(3), 390–400. doi:10.1016/j.reprotox.2011.04.005
  • Khanna, S., Dash, P. R., & Darbre, P. D. (2014). Exposure to parabens at the concentration of maximal proliferative response increases migratory and invasive activity of human breast cancer cells in vitro. Journal Applications Toxicogical, 34(9), 1051–1059.
  • Lakind, J. S., & Naiman, D. Q. (2011). Daily intake of bisphenol A and potential sources of exposure: 2005–2006 national health and nutrition examination survey. Journal Exposure Sciences Environment Epidemiology, 21(3), 272–279. doi:10.1038/jes.2010.9
  • Loi, S., Milne, R. L., Friedlander, M. L., McCredie, M. R. E., Giles, G. G., Hopper, J. L., & Phillips, K. A. (2005). Obesity and outcomes in premenopausal and postmenopausal breast cancer. Cancer Epidemiology Biomark Prevention, 14(7), 1686–1691. doi:10.1158/1055-9965.EPI-05-0042
  • Lomenick, J. P., Calafat, A. M., Melguizo Castro, M. S., Mier, R., Stenger, P., Foster, M. B., & Wintergerst, K. A. (2010). Phthalate exposure and precocious puberty in female. Journal Pediatrics, 156(2), 221–225.
  • López-Carrillo, L., Hernández-Ramírez, R. U., Calafat, A. M., Torres-Sánchez, L., Galván-Portillo, M., Needham, L. L., … Cebrián, M. E. (2010). Exposure to phthalates and breast cancer risk in northern Mexico. Environment Health Perspectives, 118(4), 539–544. doi:10.1289/ehp.0901091
  • Ma, M., Kondo, T., & Ban, S. (2006). Exposure of prepubertal female rats to inhaled di(2-ethylhexyl)phthalate affects the onset of puberty and postpubertal reproductive functions. Toxicogical Sciences, 93(1), 164–171. doi:10.1093/toxsci/kfl036
  • Mandrup, K., Boberg, J., Isling, L. K., Christiansen, S., & Hass, U. (2016). Low-dose effects of bisphenol A on mammary gland development in rats. Andrology, 4(4), 673–683. doi:10.1111/andr.12110
  • Matsumoto, H., Adachi, S., & Suzuki, Y. (2005). Bisphenol A in ambient air particulates responsible for the proliferation of MCF-7 human breast cancer cells and its concentration changes over 6 months. Archives Environment Contamination Toxicogical, 48(4), 459–466. doi:10.1007/s00244-003-0243-x
  • Meeker, J. D., Calafat, A. M., & Hauser, R. (2012). Urinary phthalate metabolites and their biotransformation products: Predictors and temporal variability among men and women. Journal Exposure Sciences Environment Epidemiology, 22(4), 376–385. doi:10.1038/jes.2012.7
  • Melnick, R., Lucier, G., Wolfe, M., Hall, R., Stancel, G., Prins, G., … Kohn, M. (2002). Summary of the national toxicology program’s report of the endocrine disruptors low-dose peer review. Environment Health Perspectives, 110(4), 427–431. doi:10.1289/ehp.02110s5883
  • Melzer, D. L., Harries, R., Cipelli, W., Henley, C., Money, P., McCormack, A., … Galloway, C. T. (2011). Bisphenol A exposure is associated with in vivo estrogenic gene expression in adults. Environment Health Perspectives, 119(12), 1788–1793. doi:10.1289/ehp.119-a484
  • Montenegro, M. F., González-Guerrero, R., Sánchez-del-Campo, L., Piñero-Madrona, A., Cabezas-Herrera, J., & Rodríguez-López, J. N. (2016). Targeting the epigenetics of the DNA damage response in breast cancer. Cell Death Disease, 7, e2180–12. doi:10.1038/cddis.2016.85
  • Moral, R., Santucci-Pereir, J., Wang, R., Russo, I. H., Lamartiniere, C. A., & Russo, J. (2011). In utero exposure to butyl benzyl phthalate induces modifications in the morphology and the gene expression profile of the mammary gland: An experimental study in rats. Environment Health, 10(1), 5–16. doi:10.1186/1476-069X-10-5
  • Moyer, B., & Hixon, M. L. (2012). Reproductive effects in F1 adult females exposed in utero to moderate to high doses of mono-2-ethylhexylphthalate (MEHP). Reproduction Toxicogical, 34(1), 43–50. doi:10.1016/j.reprotox.2012.02.006
  • O’Brien, K. M., Shi, M., Sandler, D. P., Taylor, J. A., Zaykin, D. V., Keller, J., … Weinberg, C. R. (2016). A family-based, genome-wide association study of young-onset breast cancer: Inherited variants and maternally mediated effects. European Journal Human Genetics, 24(9), 1316–1323. doi:10.1038/ejhg.2016.11
  • Okubo, T., Suzuki, T., Yokoyama, Y., Kano, K., & Kano, I. (2003). Estimation of estrogenic and anti-estrogenic activities of some phthalate diesters and monoesters by MCF-7 cell proliferation assay in vitro. Biologic Pharmaceutical Bulletin, 26(8), 1219–1224. doi:10.1248/bpb.26.1219
  • Palacios-Arreola, M. I., Nava-Castro, K. E., Del Río-Araiza, V. H., Pérez-Sánchez, N. Y., & Morales-Montor, J. (2017). A single neonatal administration of Bisphenol A induces higher tumour weight associated to changes in tumour microenvironment in the adulthood. Sciences Reports, 7(1), 10573. doi:10.1038/s41598-017-10135-1
  • Pan, S., Yuan, C., Tagmount, A., Rudel, R. A., Ackerman, J. M., Yaswen, P., … Leitman, D. C. (2016). Parabens and human epidermal growth factor receptor ligand cross-talk in breast cancer cells. Environment Health Perspectives, 124(5), 563–569. doi:10.1289/ehp.1409567
  • Paulose, T., Speroni, L., Sonnenschein, C., & Soto, A. M. (2015). Estrogens in the wrong place at the wrong time: Fetal BPA exposure and mammary cancer. Reproduction Toxicogical, 54, 58–65. doi:10.1016/j.reprotox.2014.09.012
  • Philippat, C., Wolff, M. S., Calafat, A. M., Ye, X., Bausell, R., Meadows, M., … Engel, S. M. (2013). Prenatal exposure to environmental phenols: Concentrations in amniotic fluid and variability in urinary concentrations during pregnancy. Environment Health Perspectives, 121(10), 1225–1231. doi:10.1289/ehp.1206335
  • Pugazhendhi, D., Sadler, A. J., & Darbre, P. D. (2007). Comparison of the global gene expression profiles produced by methylparaben, n-butylparaben and 17b-oestradiol in MCF7 human breast cancer cells. Journal Applications Toxicogical, 27(1), 67–77. doi:10.1002/jat.1200
  • Rodgers, K. M., Udesky, J. O., Rudel, R. A., & Green, B. (2018). J. environmental chemicals and breast cancer: An updated review of epidemiological literature informed by biological mechanisms. Environment Researcher, 160, 152–182. doi:10.1016/j.envres.2017.08.045
  • Rodriguez-Mozaz, S., de Alda, M. L., & Barceló, D. (2005). Analysis of bisphenol A in natural waters by means of an optical immunosensor. Water Research, 39(20), 5071–5079. doi:10.1016/j.watres.2005.02.007
  • Routledge, E. J., Parker, J., Odum, J., Ashby, J., & Sumpter, J. P. (1998). Some alkyl hydroxyl benzoate preservatives (parabens) are estrogenic. Toxicogical Applications Pharmacology, 153(1), 12–19. doi:10.1006/taap.1998.8544
  • Rudel, R. A., Ackerman, J. M., Attfield, K. R., & Brody, J. G. (2014). New exposure biomarkers as tools for breast cancer epidemiology, biomonitoring, and prevention: A systematic approach based on animal evidence. Environment Health Perspectives, 122(9), 881–895. doi:10.1289/ehp.1307455
  • Rudel, R. A., Fenton, S. E., Ackerman, J. M., Euling, S. Y., & Makris, S. I. (2011b). Environmental exposures and mammary gland development: State of the science, public health implications, and research recommendations. Environment Health Perspectives, 119(8), 1053–1061. doi:10.1289/ehp.1002864
  • Rudel, R. A., Gray, J. M., Engel, C. L., Rawsthorne, T. W., Dodson, R. E., Ackerman, J. M., … Brody, J. G. (2011a). Food packaging and bisphenol A and bis(2-ethyhexyl) phthalate exposure: Findings from a dietary intervention. Environment Health Perspectives, 119(7), 914–920. doi:10.1289/ehp.1003170
  • Rudolph, A., Chang-Claude, J., & Schmidt, M. K. (2016). Gene-environment interaction and risk of breast cancer. British Journal Cancer, 114(2), 125–133.
  • Samantarrai, D., Dash, S., Chhetri, B., & Mallick, B. (2013). Genomic and epigenomic cross-talks in the regulatory landscape of miRNAs in breast cancer. Molecular Cancer Researcher, 11(4), 315–328.
  • Sandager, T. M., Huber, S., Moe, M. K., Braathen, T., Leknes, H., & Lund, E. (2011). Plasma concentrations of parabens in postmenopausal women and self-reported use of personal care products: The NOWAC post genome study. Journal Experiments Sciences Environment Epidemiology, 21(6), 595–600.
  • Schlumpf, M., Kypke, K., Wittassek, M., Angerer, J., Mascher, H., Mascher, D., … Lichtensteiger, W. (2010). Exposure patterns of UV filters, fragrances, parabens, phthalates, organochlor pesticides, PBDEs, and PCBs in human milk: Correlation of UV filters with use of cosmetics. Chemosphere, 81(10), 1171–1183.
  • Schönfelder, G., Wittfoht, W., Hopp, H., Talsness, C. E., Paul, M., & Chahoud, I. (2002). Parent bisphenol A accumulation in the human maternal-fetal-placental unit. Environment Health Perspectives, 110(11), A703–707.
  • Shen, H. Y., Jiang, H. L., Mao, H. L., Pan, G., & Cao, Y. F. (2007). Simultaneous determination of seven phthalates and four parabens in cosmetic products using HPLC-DAD and GC-MS methods. Journal Sep Sciences, 30(1), 48–54.
  • Shiau, A. K., Barstad, D., Loria, P. M., Cheng, L., Kushner, P. J., Agard, D. A., & Greene, G. L. (1998). The structural basis of estrogen receptor/coactivator recognition and the antagonism of this interaction by tamoxifen. Cell, 95(7), 927–937.
  • Silva, M. J., Reidy, J. A., Herbert, A. R., Preau, J. L., Jr, Needham, L. L., & Calafat, A. M. (2004). Detection of phthalate metabolites in human amniotic fluid. Bulletin Environment Contamination Toxicogical, 72(6), 1226–1231.
  • Solomon, O., Yousefi, P., Huen, K., Gunier, R. B., Escudero-Fung, M., Barcellos, L. F., … Holland, N. (2017). Prenatal phthalate exposure and altered patterns of DNA methylation in cord blood. Environment Molecular Mutagen, 58(6), 398–410.
  • Stahlhut, R. W., Welshons, W. V., & Swan, S. H. (2009). Bisphenol A data in NHANES suggest longer than expected half-life, substantial nonfood exposure, or both. Environment Health Perspectives, 117(5), 784–789.
  • Sun, Y., Irie, M., Kishikawa, N., Wada, M., Kuroda, N., & Nakashima, K. (2004). Determination of bisphenol A in human breast milk by HPLC with column-switching and fluorescence detection. Biomedical Chromatographic, 18(8), 501–507.
  • Susiarjo, M., Sasson, I., Mesaros, C., & Bartolomei, M. S. (2013). Bisphenol A exposure disrupts genomic imprinting in the mouse. PLoS Genetics, 9(4), e100341.
  • Takeuchi, S., Lida, M., Kobayashi, S., Jin, K., Matsuda, T., & Kojima, H. (2005). Differential effects of phthalate esters on transcriptional activities via human estrogen receptors alpha and beta, and androgen receptor. Toxicology, 210(2–3), 223–233.
  • Vandenberg, L. N., Chahoud, I., Heindel, J. J., Padmanabhan, V., Paumgartten, F. J., & Schoenfelder, G. (2010). Urinary, circulating, and tissue biomonitoring studies indicate widespread exposure to bisphenol A. Environment Health Perspectives, 118(8), 1055–1070.
  • Venkata, N. G., Robinson, J. A., Cabot, P. J., Davis, B., Monteith, G. R., & Roberts-Thomson, S. J. (2006). Mono(2-ethylhexyl)phthalate and mono-n-butyl phthalate activation of peroxisome proliferator activated-receptors alpha and gamma in breast. Toxicogical Letters, 163(3), 224–234.
  • Vom Saal, F. S., & Welshons, W. V. (2014). Evidence that bisphenol a (BPA) can be accurately measured without contamination in human serum and urine and that BPA causes numerous hazards from multiple routes of exposure. Molecular Cellular Endocrinology, 398(1–2), 101–113.
  • Wang, Y. C., Tsai, C. F., Chuang, H. L., Chang, Y. C., Chen, H. S., Lee, J. N., & Tsai, E. M. (2016). Benzylbutyl phthalate promotes breast cancer stem cell expansion via SPHK1/S1P/S1PR3 signaling. Oncotarget, 7(20), 29563–29576.
  • Watanabe, Y., Kojima, H., Takeuchi, S., Uramaru, N., Ohta, S., & Kitamura, S. (2013). Comparative study on transcriptional activity of 17 parabens mediated by estrogen receptor α and β and androgen receptor. Food Chemical Toxicogical, 57, 227–234.
  • Weber, L., & Keri, R. (2011). Bisphenol A increases mammary cancer risk in two distinct mouse models of breast cancer. Biologic Reproduction, 85(3), 490–497.
  • Weng, Y. I., Hsu, P. Y., Liyanarachchi, S., Liu, J., Deatherage, D. E., Huang, Y. W., … Huang, T. H. (2010). Epigenetic influences of low-dose bisphenol a in primary human breast epithelial cells. Toxicogical Applications Pharmacology, 248(2), 111–121.
  • Witorsch, R., & Thomas, J. (2010). Personal care products and endocrine disruption: A critical review of the literature. Critical Reviews Toxicogical, 40(Suppl 3), 1–30.
  • Wróbel, A. M., & Gregoraszczuk, E. L. (2014). Differential effect of methyl-, butyl- and propylparaben and 17β-estradiol on selected cell cycle and apoptosis gene and protein expression in MCF-7 breast cancer cells and MCF-10A non-malignant cells. Journal Applications Toxicogical, 34(9), 1041–1050.
  • Yang, X., & Fisher, J. W. (2014). Unraveling bisphenol A pharmacokinetics using physiologically based pharmacokinetic modeling. Frontiers Pharmacology, 5, 292–299.
  • Zhang, X. L., Wang, H. S., Liu, N., & Ge, L. C. (2015). Bisphenol A stimulates the epithelial mesenchymal transition of estrogen negative breast cancer cells via FOXA1 signals. Archives Biochemical Biophysics, 585, 10–16.