Next Article in Journal
PheWAS-Based Systems Genetics Methods for Anti-Breast Cancer Drug Discovery
Next Article in Special Issue
Genetic Epidemiology in Latin America: Identifying Strong Genetic Proxies for Complex Disease Risk Factors
Previous Article in Journal
Origin, Behaviour, and Transmission of B Chromosome with Special Reference to Plantago lagopus
Previous Article in Special Issue
Human Genetic Adaptation to High Altitude: Evidence from the Andes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Genetic Epidemiology of Breast Cancer in Latin America

by
Valentina A. Zavala
1,
Silvia J. Serrano-Gomez
2,
Julie Dutil
3 and
Laura Fejerman
1,*
1
Department of Medicine, Division of General Internal Medicine, University of California San Francisco, San Francisco, CA 94143-1793, USA
2
Grupo de investigación en biología del cáncer, Instituto Nacional de Cancerología, Bogotá 11001000, Colombia
3
Cancer Biology Division, Ponce Research Institute, Ponce Health Sciences University, Ponce, PR 00732, USA
*
Author to whom correspondence should be addressed.
Genes 2019, 10(2), 153; https://doi.org/10.3390/genes10020153
Submission received: 19 December 2018 / Revised: 13 February 2019 / Accepted: 14 February 2019 / Published: 18 February 2019
(This article belongs to the Special Issue Genetic Epidemiology of Complex Diseases in Latin America)

Abstract

:
The last 10 years witnessed an acceleration of our understanding of what genetic factors underpin the risk of breast cancer. Rare high- and moderate-penetrance variants such as those in the BRCA genes account for a small proportion of the familial risk of breast cancer. Low-penetrance alleles are expected to underlie the remaining heritability. By now, there are about 180 genetic polymorphisms that are associated with risk, most of them of modest effect. In combination, they can be used to identify women at the lowest or highest ends of the risk spectrum, which might lead to more efficient cancer prevention strategies. Most of these variants were discovered in populations of European descent. As a result, we might be failing to discover additional polymorphisms that could explain risk in other groups. This review highlights breast cancer genetic epidemiology studies conducted in Latin America, and summarizes the information that they provide, with special attention to similarities and differences with studies in other populations. It includes studies of common variants, as well as moderate- and high-penetrance variants. In addition, it addresses the gaps that need to be bridged in order to better understand breast cancer genetic risk in Latin America.

1. Introduction

Breast cancer is the most common cancer in women, and a major public health concern worldwide [1]. Improvements in screening, early detection, and treatment led to decreased mortality, a 12% decrease between 2002 and 2012 [2,3]. Breast cancer is a complex trait determined both by genetic and non-genetic factors and, in most cases, without a clear pattern of inheritance [4,5]. Twin studies have reported a heritability of 27–31% in Nordic populations [5,6,7], and recent analysis based on genome-wide association studies (GWAS) on individuals of American and European ancestry have reported a heritability between 9 to 13% [8,9].
Only a relatively small proportion of breast cancers can be explained by the presence of high-penetrance genetic mutations, such as those in the BRCA1 and BRCA2 genes [10,11,12,13,14,15,16,17,18,19]. These, together with mutations in genes of intermediate penetrance such as ATM, BARD1, PALB2, and CHECK2, explain ~20–25% of breast cancer risk [20], leaving a considerable proportion of heritability still to be explained by low-penetrance variants. Breast cancer GWAS were instrumental in the discovery of multiple single-nucleotide polymorphisms (SNPs), starting with relatively modest sample sizes in 2007 [21,22,23] and developing into major consortiums that now include more than 200,000 women [21,22,23,24,25,26,27,28,29,30,31,32,33,34,35,36,37,38,39,40,41,42,43,44,45,46,47,48,49,50,51]. These SNPs are not very informative on their own, but together may account for ~44% of the familial relative risk [34]. However, only a small proportion of samples included in these large efforts are from United States (US) minority populations (e.g., Hispanics/Latinas, African Americans).
Latin American populations are diverse, not only culturally, but also in their genetic ancestry composition [52,53]. This diversity is likely to have an impact on the distribution of genetic determinants of breast cancer risk in different regions. The aim of this review was to highlight breast cancer genetic epidemiology studies conducted in Latin America and to summarize the information that they provide, addressing gaps that need to be bridged in order to better understand breast cancer risk in Latin America.
For paper retrieval, we combined search terms such as “breast cancer” (breast cancer subtypes), “genetics” (genetic variant, genetic polymorphism, SNP, genetic mutation, BRCA), “case–control study” (cases/controls, cases–controls, cases, controls), and “Latinas” (Latinas, Hispanics, Latin America, South America) in the PubMed database (https://www.ncbi.nlm.nih.gov/pubmed/). We also added to these terms the name of each Latin American country. We complemented the search by reviewing manuscripts cited in the selected papers. We focused on studies with case series, cohorts, or case/control design, conducted in Latin America and that included genetic factors as main predictors. In some instances, we make reference to studies conducted in or including US Latinas. We only included studies published in the English language. We did not narrow the search by date; the selected studies were published between 2008 and 2018.

2. BRCA1 and BRCA2 Mutations in Latin America

The Hereditary Breast and Ovarian Cancer (HBOC) syndrome is caused by loss-of-function mutations in the BRCA1 and BRCA2 genes and explains approximately 16% of inherited breast cancers [13]. Despite being relatively rare, pathogenic variants in the BRCA genes are associated with substantial increases in risk of breast, ovarian, and other cancers [18,54,55,56,57,58,59,60]. The comparison of the prevalence of BRCA1/BRCA2 pathogenic variants across studies in Latin America is confounded by variation in the study design, such as the approach for patient accrual, inclusion criteria, and screening technology. Nevertheless, available evidence points to marked differences in the carrier prevalence in different populations. Latin American countries, for which prevalence was assessed in unselected breast cancer patient cohorts, report a similar prevalence to that of non-Hispanic Whites (NHW) in the US; this includes Brazil [61], Colombia [62], Cuba [63], Mexico [64,65], Peru [66], and Puerto Rico [67], for which the prevalence in unselected breast cancer cases was shown to be below 5%. Differences in the relative proportion of BRCA1 to BRCA2 carriers were also observed. While most countries report higher frequencies of BRCA1 mutations, in Costa Rica, Cuba, Puerto Rico, and Uruguay, BRCA2 mutations are predominant [68]. This is expected to impact the clinical presentation of HBOC in those countries, as each gene is associated with differences in the age of onset, molecular profile, and the magnitude of risk spectrum of associated cancers [18,69].

Founder Mutations and Regional Heterogeneity

Three founder mutations were shown to account for 79% of the BRCA1/BRCA2 carriers in the Ashkenazi Jewish population [70]. In Latin America, these known founder Jewish mutations were reported to be recurrent (35% of mutation carriers) in a Jewish population of Buenos Aires, illustrating the immigration history of this country [71]. The BRCA1 185delAG (c.68_69delAG, rs80357914) is the most common pathogenic variant with the largest geographical span as it was shown to be recurrent in Argentina, Brazil, Mexico, and Peru [68,72]. In Mexican Americans, haplotype analysis demonstrated that this mutation was on the same genetic background as the Jewish founder mutation [73]. Founder mutations were also reported in Brazil, Chile, Colombia, Mexico, and more recently in Puerto Rico [67,72,74,75]. In addition to founder mutations, several countries reported the presence of recurrent mutations, but the data regarding a common origin or ancestor were not assessed [74]. It was estimated that less than 10% of the BRCA pathogenic variants reported in the literature were observed in more than one country of Latin America and the Caribbean [68,76,77]. In a large cohort distributed across 11 Brazilian states, some recurrent mutations were restricted to defined regions of the country, while other areas displayed significant heterogeneity [78]. In Colombia, despite a more modest sample size, a recent study reported a very low frequency of known Colombian founder mutations, which was associated with variations in geographic origin of the cohort within the country [79]. The genetic heterogeneity of Latin America populations suggests that further sequencing of patients in different countries and regions would be required to properly characterize geographical variation in the BRCA mutation spectrum and to provide adequate recommendations and treatment in women from Latin America. A previous review by Dutil et al. provided additional information regarding specific BRCA mutations in Latin American women [68]. In addition to mutations in the BRCA genes, there are other high-penetrance mutations that are uncommon in the population (e.g., TP53, PTEN, and STK11), which are associated with well-known familial syndromes [80,81,82]. For example, the c.1010G>A (rs121912664) variant in the TP53 gene, best known as R337H, was found in 2–7% of Brazilian breast cancer patients [83,84,85].

3. Breast Cancer Candidate Gene Studies in Latin America

Polymorphisms in moderate- and low-penetrance genes may be relatively common in the population, and collectively confer a small to moderate increased relative risk for breast cancer [86]. Common polymorphisms located within genes in known cancer-related pathways are good candidates for investigation in the context of a case–control design and were explored in multiple Latin American studies (Table 1).

3.1. DNA Repair Genes

DNA repair is crucial for normal cell function, and common polymorphisms in DNA repair genes or genes that play a role in DNA repair pathways are potential candidates for association with breast cancer risk. Several studies in Latin America assessed the association between DNA repair gene variants and breast cancer risk [94,97,98,99,100,101,110,112,117,118,119,120,121,122,123]. Candidate genes that were most studied are those that code for proteins that interact with BRCA1/2 or that are involved in the same pathway. ATM is a protein kinase that finds double-strand breaks in DNA and controls the mechanism of DNA repair [124]. Three case–control studies assessed the effect of variants in the ATM gene in Chile and Mexico [99,100,112]. In both countries the variants c.3285-9delT (rs1799757), c.5497-8T>C (rs3092829), and c.5557G>A (rs1801516) were reported to be associated with breast cancer risk.
The BARD1 protein heterodimerizes with BRCA1 and induces translocation and retention of BRCA1 into the nucleus [125]. The c.1670G>C (rs28997576) variant was assessed in case–control studies from several populations [97,126,127,128,129,130,131,132]. In Chile, this variant was associated with an increased breast cancer risk in families with strong family history [97]. Two other variants were detected in a cohort of 124 Peruvian triple-negative breast cancer patients: the pathogenic c.334C>T (rs758972589) variant and the probably pathogenic c.1622C>A (rs777937955) variant [122].
Three other BRCA1 and BRCA2 functionally related genes were screened in Latin American populations. PALB2 binds BRCA2 and recruits it to BRCA1 foci on damaged DNA [133]. Leyton et al. screened this gene in 100 Chilean breast cancer patients negative for BRCA1/2 mutations and found two variants, c.1676A>G (rs152451) and c.2993C>T (rs45551636), that were associated with breast cancer risk in a subsequent case–control study [94]. CHEK2 protein is activated by ATM and phosphorylates BRCA1 [134]. The CHEK2 c.1100delC truncating mutation, which increases the risk of breast cancer twofold, was widely analyzed. This mutation has a frequency of 0.3–1.2% in European populations [135] and case-only studies in Brazil, and case–control studies in Colombia and Chile concluded that this variant is almost absent in these populations [119,123,136,137]. AKT1 is frequently mutated in mammary tumors [138]. Activated AKT1 phosphorylates a large number of downstream substrates that play a crucial role in tumor growth and survival [139]. Lopez-Cortes et al. [102] analysed six SNPs (c.49G>A (rs121434592), c.956A>G (rs12881616), c.1070T>C(rs11555432), c.1162C>A (rs11555431), c.1172+23A>G (rs2494732), and c.1172+69G>C (rs3803304)) in AKT1 and its association with breast cancer risk, as well as histopathological and immunohistochemical characteristics, in 276 Ecuadorian mestizo women. They found that the intronic SNP rs3803304 was associated with breast cancer risk. Moreover, they found that the heterozygous genotype CG of that SNP had a significantly different frequency between intrinsic subtypes (p ≤ 0.05), with luminal B showing the highest frequency (63.6%). This result suggests that the SNP rs3803304 may affect DNA transcription, acting as a risk factor for breast cancer in Ecuadorian mestizo population living in high altitudes.
Polymorphisms within two DNA double-strand break repair genes, XRCC3 c.722C>T (rs861539) and RAD51D c.698A>G (rs28363284), were assessed for their association with breast cancer risk in high-risk families in Chile without known BRCA1/2 mutations [98]. Only the variant in the XRCC3 gene yielded a statistically significant result [98]. In relation to DNA single-strand break repair genes, a case–control study in Puerto Rico tested associations between SNPs in the xeroderma pigmentosum (XPC and XPD) and ultraviolet (UV) excision repair protein (RAD23B) genes and breast cancer risk [117]. They hypothesized that genetic variation in the nucleotide excision repair pathway genes could modulate DNA repair capacity and contribute to breast cancer risk and found an association between RAD23B and breast cancer risk, and between XPC and RAD23B and DNA repair capacity in the cases [117]. Similarly, a Mexican study reported a significant association between the c.1196A>G (rs25487) variant of the XRCC1 gene and breast cancer risk [121]. Variants in TP53, which is an important gene in DNA-repair related pathways [140], were reported in Brazilian populations [83,84,85,114,141,142,143,144,145]. Four case-control studies assessed the rs1042522 (c.215C>A) variant, reporting negative results. The heterozygous frequency ranges between 40 and 60% and the frequency of the homozygous genotype is ~10%. These frequencies are also similar to the frequencies in the healthy population [141,142,143,144]. Even though the prevalence of this mutation is variable across different populations, these results are similar to the average frequencies observed worldwide [146]. Three other studies evaluated the c.1010G>A (rs121912664) variant, which was found in 2–7% of breast cancer patients and absent in controls [83,84,85].

3.2. Genes Involved in Amino-Acid and Nucleic-Acid Metabolism

The thymidylate synthase (TS) gene produces an enzyme that is involved in one-carbon metabolism and is crucial for DNA synthesis. A study in Mexico explored the association between polymorphisms in the TS gene and breast cancer risk and reported negative results [118]. Other case–control studies in Mexico, Ecuador, and Brazil [88,103,110,111,147] evaluated the association between polymorphisms in the methylenetetrahydrofolate reductase (MTHFR) and cystathionine β synthase (CBS) genes, which are involved in the processing of amino acids, and breast cancer risk. Four out of five studies found a positive association [88,103,110]. López-Cortés et al. [103] did not find statistically significant differences between breast cancer subtypes but reported an association between the heterozygous C/T and homozygous mutant T/T genotypes for the c.665C>T (rs1801133) polymorphism, and a lower MTHFR messenger RNA (mRNA) expression in triple-negative breast cancer (TNBC) tumors. The 5-methyltetrahydrofolate-homocysteine methyltransferase (MTR) gene, also involved in the amino-acid synthesis pathway, was tested in a Brazilian population with no significant results [147].

3.3. Genes Involved in Hormone Metabolism, and Hormone Receptor, Co-Activator, or Suppressor Genes

Sex hormones, their receptors, and other relevant components within their pathways, are thought to be involved in breast cancer development because of their role in cell growth and proliferation. Analyses of a limited number of candidate variants in hormone receptor genes (estrogen (ESR1), androgen (AR), and progesterone receptors (PGR)) yielded mostly negative results except in secondary stratified analyses (by body mass index or use of hormone replacement therapy) [148,149,150,151,152]. Studies conducted in Latin America focused on the ESR1, PGR, COMT, CYP19A1, and CYP1A1 genes [89,113,115,120,150,153]. One of these studies reported a positive association between an Alu insertion in the PGR gene and breast cancer risk [120], while other studies revealed statistically significant associations for polymorphisms within the CYP1A1 gene [113,115].

3.4. Extracellular Matrix Components

Matrix metalloproteinases (MMPs) are enzymes that can degrade the extracellular matrix and basement membrane and are thought to play an important role in cell proliferation, migration, differentiation, apoptosis, and angiogenesis [154]. A large study including US Hispanic/Latina women tested associations between polymorphisms in MMP-1, MMP-2, MMP-3, and MMP-9 genes and breast cancer risk [155]. Significant associations were found for intronic SNPs in the MMP-1 (c.625+332G>A, rs996999), MMP-3 (c.1229+495C>T, rs650108), and MMP-9 (c.1331-163G>T, rs3787268) genes among women with high Native-American ancestry [155]. A study conducted in Mexico reported a strong association between the MMP-2 c.-1586C>T polymorphism (rs243865) and breast cancer risk [156]. Other studies in Argentinian and Brazilian populations reported negative results for two SNPs (c.781G>A, rs11895564 and c.3841C>T, rs121912467) in the integrin subunit α6 (ITGA6) gene and the c.-1562 C>T (rs3918242) polymorphism in the MMP-9 gene, respectively [157,158].

3.5. Inflammation and Energy Balance

Different analyses conducted in the Breast Cancer Health Disparities Study samples analyzed tag SNPs in genes related to inflammation and energy balance, taking into account possible interactions with body mass index [159,160,161,162,163,164]. Few Latin American studies tested the association between genes in inflammation and energy balance pathways and breast cancer risk (TNF-α, RETN, CAP1, and PTGS2 genes). No association was detected for nine SNPs evaluated within the PTGS2 gene [165]. The TNF-α c.-308G>A (rs1800629) polymorphism was found to be associated with breast cancer risk in a study conducted in Mexico [166], and another Mexican study found associations between the RETN c.-225C>G (rs1862513) and CAP1 c.881G>A (rs35749351) polymorphisms and breast cancer risk [104].

3.6. Genes Associated with Tumoral Immunity

Two studies assessed polymorphisms related to tumor immune response in Brazilian populations, and both of them found statistically significant associations with breast cancer risk [87,90]. Tumor-infiltrating regulatory T cells (Tregs) possess relevant roles in tumor immunity. When activated by tumor-associated antigens, they can suppress specific antitumor immune responses [167]. Forkhead box P3 (FOXP3) is a transcription factor essential for the development and functions of Treg cells [87,168]. It was also reported as being expressed in breast cancer cells, and its expression is associated with a better prognosis after neoadjuvant chemotherapy in HER2-overexpressing breast cancer patients [168]. Banin Hirata et al. (2017) analyzed two FOXP3 SNPs (c.-22-902A>G (rs2232365) and c.-23+2882C>A (rs3761548)) and their possible association with susceptibility and clinical outcomes. They reported associations between genotypes and clinical characteristics for specific breast cancer subtypes [87], suggesting that FOXP3 may influence clinical outcome according to intrinsic subtype and might be a marker for aggressive disease. Another study reported associations between two SNPs in the promoter of the IL-18 gene, c.-838C>A(rs1946518) and c.-368G>T (rs187238), and breast cancer risk [90]. IL-18 is a pleiotropic cytokine that promotes antitumor pro-inflammatory responses and is highly expressed in breast cancer tumors in mice and in serum of breast cancer patients [169,170]. Predictive models showed that these SNPs might impair the binding of some transcription factors, decreasing promoter activity [171,172].

3.7. Metabolism of Xenobiotic Compounds and Oxidative Stress

Selenoproteins were shown to have a redox function and to decrease oxidative stress. One study tested the association between SNPs in different selenoprotein-related genes (GPX1, GPX2, GPX3, GPX4, SELS, SEP15, SEPN1, SEPP1, SEPW1, TXNRD1, and TXNRD2) and breast cancer risk in US Latinas, finding mostly negative results except when analyses were stratified by genetic ancestry and by tumor subtype [173]. Other studies were conducted in Mexico and Brazil and investigated multiple genes: CYP2W1, CYP4F11, CYP8A1, and CYP1B1 [174], which are important in the activation of carcinogenic compounds; ABCB1, which encodes the MDR1 protein, [105,106] and is involved in the elimination of xenotoxic agents; GSTM1 and GSTP1, members of the glutathione S-transferase (GST) gene family [91,108,109,113]; and eNOS, which can react with other free radicals and damage DNA [114]. Results for the CYP genes were mostly negative, except for CYP1B1 [107], while positive associations were reported for the GSTM1 deletion (tagged by rs366631), and ABCB1 c.3435C>T (rs1045642), GSTP1 c.313A>G (rs1695), and eNOS polymorphisms [91,105,106,108,109,113,114,174]. A study that included 275 Mexican women enrolled in the Ella Binational Breast Cancer Study reported that the T allele of the c.3435C>T polymorphism located in exon 26 of the ABCB1 gene was an important risk factor for breast cancer in premenopausal women and specifically for the TNBC subtype [106]. This polymorphism does not change the amino-acid sequence, but is associated with a decrease in ABCB1 function, and reduced mRNA and/or protein expression, which might impact the metabolism and posterior elimination of toxic and carcinogenic compounds inducing cellular accumulation leading to cancer development [106,175].

3.8. Non-Coding RNAs

Non-coding RNAs participate in complex networks involved in cancer pathways. MicroRNAs are short non-coding RNAs with relevant roles in the physiological control of cell homeostasis as they regulate the expression of hundreds of genes though direct binding to their mRNAs [176]. SNPs on microRNA precursors (pre-microRNAs) can influence the processing into their functional mature forms; moreover, SNPs in the mature forms may affect the strength of regulation of their target mRNAs. Two studies tested the association of SNPs within microRNAs in Latina patients [92,93]. The first study assessed the effect of the miR-196a2 rs11614913 SNP in a Brazilian population and revealed that the T allele was associated with increased breast cancer risk, while the CC genotype had a protective effect [92]. In a Chilean study, SNPs in pre-miR-27a, pre-miR-196a2, pre-miR-423, pre-miR-618, and miR-608 were tested for their association with breast cancer risk [93]. The authors found that the pre-miR-423 rs6505162 and pre-miR27a rs895819 SNPs were associated with breast cancer risk in individuals with a strong family history and with a moderate history of breast cancer, respectively, and that the pre-mir-618 rs2682818 was associated with an increased risk in non-familial early-onset breast cancer [93].

4. Genome-Wide Association Studies in Latin America

The first GWAS on Latina women was published in 2014 [40], reporting genome-wide statistically significant results for two linked SNPs 56 kb upstream of the ESR1 gene (rs140068132 and rs147157845). These SNPs have a frequency of between 5% and 23% in Latin American populations and are practically absent in all other groups. The minor allele was protective, with an associated odds ratio (OR) of 0.60 (95% confidence interval (CI): 0.53–0.67) and was more protective for estrogen-receptor-negative (ER) (OR: 0.34; 95% CI: 0.21–0.54) than for estrogen-receptor-positive disease (OR: 0.63; 95% CI: 0.49-0.80) [40]. An additional GWAs and fine mapping analysis of the 6q25 region in an extended sample identified three new SNPs associated with breast cancer risk [177].
Some studies conducted in Latin America replicated selected loci that were discovered in breast cancer GWAS conducted in samples of European origin: two in Chile and one in Mexico [95,96,116]. These studies tested the association between SNPs in FGFR2, TOX3, and MAP3K1 genes and the 2q35 and 8q24 loci, that were replicated in different populations [178,179,180,181,182,183,184]. FGFR2 and MAP3K1 are tyrosine kinases. FGFR2 mediates signaling of fibroblast growth factors, MAP3K1 is involved in apoptosis signaling, and TOX3 is a nuclear protein that acts as a chromatin remodeler. Altered expression of these proteins was described in breast cancer pathogenesis [185,186,187]. Two studies assessed the association between SNPs in FGFR2 (c.109+906T>C (rs2981582), c.109+1899A>G (rs2420946), 109+7033T>A (rs1219648), MAP3K1 (rs889312), TOX3 (rs3803662), 2q35 (rs13387042), and 8q24 (rs13281615) and breast cancer risk in Chilean patients [95,96]. They found associations for SNPs on FGFR2, MAP3K1, TOX3, and 2q35, but not for 8q24 [95,96]. Another analysis of the FGFR2 rs2981582 polymorphism in the Mexican population reported an interaction between the FGFR2 polymorphism and alcohol intake [116]. The first breast cancer GWAS in US Hispanic/Latinas also replicated previous associations, with most of the SNPs being concordant in terms of direction and magnitude of association with those reported in Europeans or Asians [40].

5. Genetic Ancestry and Breast Cancer Risk

There is a heterogeneous genetic background in Latin American populations, with regional variation in the relative proportions of the three main ancestral continental influences: Indigenous American, European, and African. A study conducted in US Hispanic/Latinas from the Bay Area first reported the association between genetic ancestry and breast cancer risk [188]. Women with higher European ancestry had an increased risk of breast cancer compared to women with higher Indigenous American ancestry. Subsequent studies in Latin American populations (two in Mexicans and one in Colombians) replicated this finding [189,190,191]. A study from Uruguay did not find a statistically significant difference in genetic ancestry between breast cancer cases and controls, possibly due to the lack of statistical power given the size of the sample and the relatively small proportion of Indigenous American ancestry in this population [192]. The original finding that genetic ancestry was associated with breast cancer risk in Latinas was followed by an admixture mapping [193] analysis and a breast cancer GWAS [40], which we previously mentioned, and led to the discovery of a protective genetic variant near the ESR1 gene mostly observed in women of Indigenous American ancestry.
A large proportion of the case/control studies described in Section 3 did not adjust for genetic ancestry, which is a known confounder in genetic epidemiology studies. Self-reported ethnicity is a useful way to control for population stratification on an ethnicity-matched case–control study design from comparable geographical regions [194]; however, this methodology is logistically challenging and costly. In addition, given the complex genetic substructure of Latin American populations, it is highly recommended to adjust for covariates that capture the ancestry composition of cases and controls using panels of ancestry informative markers or genome-wide genotype data [195].

6. Conclusions and Perspectives

In this review, we presented a summary of genetic association studies in Latin American women, including studies of relatively rare high-penetrance mutations and those discovered using candidate gene approaches or GWAS, which tend to be of moderate or small effects but are more common.
In recent years, a picture of the BRCA1- and BRCA2-associated hereditary breast cancers in Latin America emerged. While still incomplete, available evidence points to a unique genetic make-up characterized by remarkable diversity across and within countries [68,74,75,196]. Yet, a better understanding of the prevalence and types of mutations underlying hereditary breast cancers in Latin American women is a necessary step for informing the strategies for the detection and clinical management of carriers. The use of affordable targeted mutation panels presents an attractive solution in settings with limited resources, which are common in many Latin American countries, provided that larger studies representative of the population of interest are conducted during variant discovery. These populations also present unique opportunities to contribute to the classification of variants of unknown significance. In recent years, clinical testing for hereditary breast cancer transitioned from BRCA1 and BRCA2 analysis to extended panels of genes including high- and moderate-penetrance risk genes. Although this approach became standard of care in the US, the data on the use of these panels in Latin American populations are sparse. In parallel to emerging efforts aimed at describing the molecular characteristics of hereditary breast cancers in Latin America, there is a need to improve access to genetic testing and for the development of supporting infrastructures in cancer risk assessment, education, and genetic counseling [197,198].
Risk prediction models that combine non-genetic and genetic risk factors were developed and are constantly being improved [199,200,201,202]. Taken separately, variants of small effect sizes have limited impact on breast cancer risk prediction, but when combined into polygenic risk scores (PRS), the magnitude of their effect on risk may equal or surpass those of pathogenic variants in moderate-risk genes [203]. Prediction models that are usually applied are based on research conducted in European populations [204]. Because GWAS SNPs are selected for their tagging properties rather than their potential functional significance, known differences in linkage disequilibrium structures in populations of the world [205] are likely to confound replication studies and the clinical validity of the European GWAS SNPs. Recently, Wang et. al. demonstrated that 30–40% of the SNPs commonly used to estimate breast cancer PRS have inconsistent directionality in African American populations and, consequently, performed poorly in this group [206].
Progress in the discovery of population-specific breast cancer genetic risk variants in this region is limited by different factors, such as the slower incorporation of new genotyping and sequencing technologies, and the relatively small samples sizes partly due to the paucity of National Cancer Registries and the inconsistent collection of biospecimens (i.e., tumor blocks) for research (Table 2). On average, approximately half of the genome of Latina American women is of European origin and, therefore, much larger sample sizes would be required if similar results as for the European genome are to be expected for the Indigenous American component. We cannot assume that only overlapping variants between different ancestral genomes will be associated with risk and, therefore, we need to focus our efforts to reduce research disparities by expanding available resources to include large cohorts and case–control studies of diverse populations in and outside the US.
There are ongoing efforts to improve homogeneity of study design and increase sample size in studies in Latin America. In 2009, the Center for Global Health of the US National Cancer Institute (NCI) created the US-Latin America Cancer Research Network (LACRN), a collaborative management structure that includes Argentina, Brazil, Chile, Mexico, and Uruguay [207]. In 2011 the US–LACRN launched the “Molecular Profiling of Stage II and III Breast Cancer in Latin American Women Receiving Standard of Care Treatment (MPBC)” study. Future studies will involve genetic ancestry estimation, sequencing of high-penetrance genes, whole-exome sequencing, and identification of circulating tumor cells and cell-free DNA. The Molecular Subtypes of Premenopausal Breast Cancer in Latin American Women (PRECAMA) study is an effort led by the International Agency for Research on Cancer (IARC) involving data harmonization and standard protocols for recruitment of premenopausal breast cancer cases and controls, collection and storage of blood samples, tumor block fixation and handling, and pathology review in Chile, Colombia, Mexico, and Costa Rica [208]. Similarly, the EPIGEN-Brazil Initiative aims to study the association between genetic variants found in the Brazilian population and complex diseases, taking into account its complex admixture genetic background [209]. Many studies in Latin America are based on the recruitment of cases only and, therefore, cannot be included in larger studies based on case–control comparisons. However, efforts such as the Consortium for the Analysis of the Diversity and Evolution of Latin America, the Candela project, which aims to characterize the physical appearance and to examine the genetic and social background of 7342 subjects from Mexico, Colombia, Peru, Chile, and Brazil [210], could be leveraged to complement case-only designs. Another such resource is the Chile Genomico Project, aimed at describing the genomic structure of the Chilean population based on the contribution of the Indigenous Americans, European, and African populations. So far, they have genotyped 3500 individuals (chilegenomico.cl; Accessed February 10th 2019). Finally, the Mexico Biobank Project has created a nation-wide DNA biobank that facilitates research on complex diseases and aims to characterize the population genetics of native and admixed Mexicans (mxbiobankproject.org; Accessed February 10th, 2019).
To conclude, large collaborative international efforts that include knowledge and technology exchange and transfer will lead to studies with appropriate sample sizes and approaches to discover rare high-penetrance polymorphisms, as well as low-penetrance variants in Latin American women from different regions and with different ancestral proportions. Only then will we be able to provide appropriate genetic risk estimates and care to women of Latin American origin worldwide.

Author Contributions

V.A.Z. and L.F. wrote and conceived the structure of the manuscript. J.D. and S.J.S.-G. contributed to the writing of the article. All authors approved the final version of the manuscript.

Funding

This work was supported by the National Cancer Institute [R01CA204797 (LF)].

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Torre, L.A.; Islami, F.; Siegel, R.L.; Ward, E.M.; Jemal, A. Global cancer in women: Burden and trends. Cancer Epidemiol. Biomarkers Prev. 2017, 26, 444–457. [Google Scholar] [CrossRef] [PubMed]
  2. Carioli, G.; Malvezzi, M.; Rodriguez, T.; Bertuccio, P.; Negri, E.; La Vecchia, C. Trends and predictions to 2020 in breast cancer mortality: Americas and Australasia. Breast 2018, 37, 163–169. [Google Scholar] [CrossRef] [PubMed]
  3. Carioli, G.; Malvezzi, M.; Rodriguez, T.; Bertuccio, P.; Negri, E.; La Vecchia, C. Trends and predictions to 2020 in breast cancer mortality in Europe. Breast 2017, 36, 89–95. [Google Scholar] [CrossRef] [PubMed]
  4. Peto, J.; Mack, T.M. High constant incidence in twins and other relatives of women with breast cancer. Nat. Genet. 2000, 26, 411–414. [Google Scholar] [CrossRef] [PubMed]
  5. Lichtenstein, P.; Holm, N.V.; Verkasalo, P.K.; Iliadou, A.; Kaprio, J.; Koskenvuo, M.; Pukkala, E.; Skytthe, A.; Hemminki, K. Environmental and heritable factors in the causation of cancer--analyses of cohorts of twins from Sweden, Denmark, and Finland. N. Engl. J. Med. 2000, 43, 78–85. [Google Scholar] [CrossRef] [PubMed]
  6. Mucci, L.A.; Hjelmborg, J.B.; Harris, J.R.; Czene, K.; Havelick, D.J.; Scheike, T.; Graff, R.E.; Holst, K.; Möller, S.; Unger, R.H.; et al. Familial risk and heritability of cancer among twins in Nordic countries. JAMA 2016, 315, 68. [Google Scholar] [CrossRef]
  7. Möller, S.; Mucci, L.A.; Harris, J.R.; Scheike, T.; Holst, K.; Halekoh, U.; Adami, H.O.; Czene, K.; Christensen, K.; Holm, N.V. The heritability of breast cancer among women in the Nordic twin study of cancer. Cancer Epidemiol. Biomarkers Prev. 2016, 25, 145–150. [Google Scholar] [CrossRef]
  8. Jiang, X.; Finucane, H.K.; Schumacher, F.R.; Schmit, S.L.; Tyrer, J.P.; Han, Y.; Michailidou, K.; Lesseur, C.; Kuchenbaecker, K.B.; Dennis, J.; et al. Shared heritability and functional enrichment across six solid cancers. Nat. Commun. 2019, 10, 431. [Google Scholar] [CrossRef]
  9. Sampson, J.N.; Wheeler, W.A.; Yeager, M.; Panagiotou, O.; Wang, Z.; Berndt, S.I.; Lan, Q.; Abnet, C.C.; Amundadottir, L.T.; Figueroa, J.D.; et al. Analysis of heritability and shared heritability based on genome-wide association studies for thirteen cancer types. J. Natl. Cancer Inst. 2015, 107, djv279. [Google Scholar] [CrossRef]
  10. Miki, Y.; Swensen, J.; Shattuck-Eidens, D.; Futreal, P.A.; Harshman, K.; Tavtigian, S.; Liu, Q.; Cochran, C.; Bennett, L.M.; Ding, W.; et al. A strong candidate for the breast and ovarian cancer susceptibility gene BRCA1. Science 1994, 266, 66–71. [Google Scholar] [CrossRef] [PubMed]
  11. Wooster, R.; Bignell, G.; Lancaster, J.; Swift, S.; Seal, S.; Mangion, J.; Collins, N.; Gregory, S.; Gumbs, C.; Micklem, G.; et al. Identification of the breast cancer susceptibility gene BRCA2. Nature 1995, 378, 789–792. [Google Scholar] [CrossRef] [PubMed]
  12. Newman, B.; Mu, H.; Butler, L.M.; Millikan, R.C.; Moorman, P.G.; King, M.C. Frequency of breast cancer attributable to BRCA1 in a population-based series of American women. JAMA 1998, 279, 915–921. [Google Scholar] [CrossRef] [PubMed]
  13. Peto, J.; Collins, N.; Barfoot, R.; Seal, S.; Warren, W.; Rahman, N.; Easton, D.F.; Evans, C.; Deacon, J.; Stratton, M.R. Prevalence of BRCA1 and BRCA2 gene mutations in patients with early-onset breast cancer. J. Natl. Cancer Inst. 1999, 91, 943–949. [Google Scholar] [CrossRef] [PubMed]
  14. Antoniou, A.C.; Pharoah, P.D.; McMullan, G.; Day, N.E.; Ponder, B.A.; Easton, D. Evidence for further breast cancer susceptibility genes in addition to BRCA1 and BRCA2 in a population-based study. Genet. Epidemiol. 2001, 21, 1–18. [Google Scholar] [CrossRef] [PubMed]
  15. Cui, J.; Antoniou, A.C.; Dite, G.S.; Southey, M.C.; Venter, D.J.; Easton, D.F.; Giles, G.G.; McCredie, M.R.; Hopper, J.L. After BRCA1 and BRCA2—What next? Multifactorial segregation analyses of three-generation, population-based Australian families affected by female breast cancer. Am. J. Hum. Genet. 2001, 68, 420–431. [Google Scholar] [CrossRef] [PubMed]
  16. John, E.M.; Miron, A.; Gong, G.; Phipps, A.I.; Felberg, A.; Li, F.P.; West, D.W.; Whittemore, A.S. Prevalence of pathogenic BRCA1 mutation carriers in 5 US racial/ethnic groups. JAMA 2007, 298, 2869. [Google Scholar] [CrossRef] [PubMed]
  17. Easton, D.F. How many more breast cancer predisposition genes are there? Breast Cancer Res. 1999, 1, 14–17. [Google Scholar] [CrossRef] [PubMed]
  18. Antoniou, A.; Pharoah, P.D.; Narod, S.; Risch, H.A.; Eyfjord, J.E.; Hopper, J.L.; Loman, N.; Olsson, H.; Johannsson, O.; Borg, A.; et al. Average risks of breast and ovarian cancer associated with BRCA1 or BRCA2 mutations detected in case series unselected for family history: A combined analysis of 22 studies. Am. J. Hum. Genet. 2003, 72, 1117–1130. [Google Scholar] [CrossRef] [PubMed]
  19. Mavaddat, N.; Peock, S.; Frost, D.; Ellis, S.; Platte, R.; Fineberg, E.; Evans, D.G.; Izatt, L.; Eeles, R.A.; Adlard, J.; et al. Cancer risks for BRCA1 and BRCA2 mutation carriers: results from prospective analysis of EMBRACE. J. Natl. Cancer Inst. 2013, 105, 812–822. [Google Scholar] [CrossRef] [PubMed]
  20. Sapkota, Y. Germline DNA variations in breast cancer predisposition and prognosis: A systematic review of the literature. Cytogenet. Genome Res. 2014, 144, 77–91. [Google Scholar] [CrossRef] [PubMed]
  21. Easton, D.F.; Pooley, K.A.; Dunning, A.M.; Pharoah, P.D.; Thompson, D.; Ballinger, D.G.; Struewing, J.P.; Morrison, J.; Field, H.; Luben, R.; et al. Genome-wide association study identifies novel breast cancer susceptibility loci. Nature 2007, 447, 1087–1093. [Google Scholar] [CrossRef] [PubMed]
  22. Stacey, S.N.; Manolescu, A.; Sulem, P.; Rafnar, T.; Gudmundsson, J.; Gudjonsson, S.A.; Masson, G.; Jakobsdottir, M.; Thorlacius, S.; Helgason, A.; et al. Common variants on chromosomes 2q35 and 16q12 confer susceptibility to estrogen receptor-positive breast cancer. Nat. Genet. 2007, 39, 865–869. [Google Scholar] [CrossRef] [PubMed]
  23. Hunter, D.J.; Kraft, P.; Jacobs, K.B.; Cox, D.G.; Yeager, M.; Hankinson, S.E.; Wacholder, S.; Wang, Z.; Welch, R.; Hutchinson, A.; et al. A genome-wide association study identifies alleles in FGFR2 associated with risk of sporadic postmenopausal breast cancer. Nat. Genet. 2007, 39, 870–874. [Google Scholar] [CrossRef] [PubMed]
  24. Orr, N.; Dudbridge, F.; Dryden, N.; Maguire, S.; Novo, D.; Perrakis, E.; Johnson, N.; Ghoussaini, M.; Hopper, J.L.; Southey, M.C.; et al. Fine-mapping identifies two additional breast cancer susceptibility loci at 9q31.2. Hum. Mol. Genet. 2015, 24, 2966–2984. [Google Scholar] [CrossRef] [PubMed]
  25. Fletcher, O.; Johnson, N.; Orr, N.; Hosking, F.J.; Gibson, L.J.; Walker, K.; Zelenika, D.; Gut, I.; Heath, S.; Palles, C.; et al. Novel breast cancer susceptibility locus at 9q31.2: Results of a genome-wide association study. J. Natl. Cancer Inst. 2011, 103, 425–435. [Google Scholar] [CrossRef] [PubMed]
  26. Gold, B.; Kirchhoff, T.; Stefanov, S.; Lautenberger, J.; Viale, A.; Garber, J.; Friedman, E.; Narod, S.; Olshen, A.B.; Gregersen, P.; et al. Genome-wide association study provides evidence for a breast cancer risk locus at 6q22.33. Proc. Natl. Acad. Sci. USA 2008, 105, 4340–4345. [Google Scholar] [CrossRef] [PubMed]
  27. Turnbull, C.; Ahmed, S.; Morrison, J.; Pernet, D.; Renwick, A.; Maranian, M.; Seal, S.; Ghoussaini, M.; Hines, S.; Healey, C.S.; et al. Genome-wide association study identifies five new breast cancer susceptibility loci. Nat. Genet. 2010, 42, 504–507. [Google Scholar] [CrossRef] [PubMed]
  28. Gaudet, M.M.; Kuchenbaecker, K.B.; Vijai, J.; Klein, R.J.; Kirchhoff, T.; McGuffog, L.; Barrowdale, D.; Dunning, A.M.; Lee, A.; Dennis, J.; et al. Identification of a BRCA2-specific modifier locus at 6p24 related to breast cancer risk. PLoS Genet. 2013, 9, e1003173. [Google Scholar] [CrossRef] [PubMed]
  29. Long, J.; Cai, Q.; Sung, H.; Shi, J.; Zhang, B.; Choi, J.Y.; Wen, W.; Delahanty, R.J.; Lu, W.; Gao, Y.T.; et al. Genome-wide association study in East Asians identifies novel susceptibility loci for breast cancer. PLoS Genet. 2012, 8, e1002532. [Google Scholar] [CrossRef] [PubMed]
  30. Couch, F.J.; Wang, X.; McGuffog, L.; Lee, A.; Olswold, C.; Kuchenbaecker, K.B.; Soucy, P.; Fredericksen, Z.; Barrowdale, D.; Dennis, J.; et al. Genome-wide association study in BRCA1 mutation carriers identifies novel loci associated with breast and ovarian cancer risk. PLoS Genet. 2013, 9, e1003212. [Google Scholar] [CrossRef] [PubMed]
  31. Li, J.; Humphreys, K.; Darabi, H.; Rosin, G.; Hannelius, U.; Heikkinen, T.; Aittomäki, K.; Blomqvist, C.; Pharoah, P.D.; Dunning, A.M.; et al. A genome-wide association scan on estrogen receptor-negative breast cancer. Breast Cancer Res. 2010, 12, R93. [Google Scholar] [CrossRef] [PubMed]
  32. Murabito, J.M.; Rosenberg, C.L.; Finger, D.; Kreger, B.E.; Levy, D.; Splansky, G.L.; Antman, K.; Hwang, S.J. A genome-wide association study of breast and prostate cancer in the NHLBI’s Framingham Heart Study. BMC Med. Genet. 2007, 8, S6. [Google Scholar] [CrossRef] [PubMed]
  33. Antoniou, A.C.; Wang, X.; Fredericksen, Z.S.; McGuffog, L.; Tarrell, R.; Sinilnikova, O.M.; Healey, S.; Morrison, J.; Kartsonaki, C.; Lesnick, T.; et al. A locus on 19p13 modifies risk of breast cancer in BRCA1 mutation carriers and is associated with hormone receptor–negative breast cancer in the general population. Nat. Genet. 2010, 42, 885–892. [Google Scholar] [CrossRef] [PubMed]
  34. Michailidou, K.; Lindström, S.; Dennis, J.; Beesley, J.; Hui, S.; Kar, S.; Lemaçon, A.; Soucy, P.; Glubb, D.; Rostamianfar, A.; et al. Association analysis identifies 65 new breast cancer risk loci. Nature 2017, 551, 92–94. [Google Scholar] [CrossRef] [PubMed]
  35. Cai, Q.; Long, J.; Lu, W.; Qu, S.; Wen, W.; Kang, D.; Lee, J.Y.; Chen, K.; Shen, H.; Shen, C.Y.; et al. Genome-wide association study identifies breast cancer risk variant at 10q21.2: results from the Asia Breast Cancer Consortium. Hum. Mol. Genet. 2011, 20, 4991–4999. [Google Scholar] [CrossRef] [PubMed]
  36. Kim, H.C.; Lee, J.Y.; Sung, H.; Choi, J.Y.; Park, S.K.; Lee, K.M.; Kim, Y.J.; Go, M.J.; Li, L.; Cho, Y.S.; et al. A genome-wide association study identifies a breast cancer risk variant in ERBB4 at 2q34: results from the Seoul Breast Cancer Study. Breast Cancer Res. 2012, 14, R56. [Google Scholar] [CrossRef] [PubMed]
  37. Elgazzar, S.; Zembutsu, H.; Takahashi, A.; Kubo, M.; Aki, F.; Hirata, K.; Takatsuka, Y.; Okazaki, M.; Ohsumi, S.; Yamakawa, T.; et al. A genome-wide association study identifies a genetic variant in the SIAH2 locus associated with hormonal receptor-positive breast cancer in Japanese. J. Hum. Genet. 2012, 57, 766–771. [Google Scholar] [CrossRef] [PubMed]
  38. Long, J.; Cai, Q.; Shu, X.O.; Qu, S.; Li, C.; Zheng, Y.; Gu, K.; Wang, W.; Xiang, Y.B.; Cheng, J.; et al. Identification of a functional genetic variant at 16q12.1 for breast cancer risk: Results from the Asia breast cancer consortium. PLoS Genet. 2010, 6, e1001002. [Google Scholar] [CrossRef] [PubMed]
  39. Michailidou, K.; Beesley, J.; Lindstrom, S.; Canisius, S.; Dennis, J.; Lush, M.J.; Maranian, M.J.; Bolla, M.K.; Wang, Q.; Shah, M.; et al. Genome-wide association analysis of more than 120,000 individuals identifies 15 new susceptibility loci for breast cancer. Nat. Genet. 2015, 47, 373–380. [Google Scholar] [CrossRef] [PubMed]
  40. Fejerman, L.; Ahmadiyeh, N.; Hu, D.; Huntsman, S.; Beckman, K.B.; Caswell, J.L.; Tsung, K.; John, E.M.; Torres-Mejia, G.; Carvajal-Carmona, L.; et al. Genome-wide association study of breast cancer in Latinas identifies novel protective variants on 6q25. Nat. Commun. 2014, 5, 5260. [Google Scholar] [CrossRef] [PubMed]
  41. Milne, R.L.; Kuchenbaecker, K.B.; Michailidou, K.; Beesley, J.; Kar, S.; Lindström, S.; Hui, S.; Lemaçon, A.; Soucy, P.; Dennis, J.; et al. Identification of ten variants associated with risk of estrogen-receptor-negative breast cancer. Nat. Genet. 2017, 49, 1767–1778. [Google Scholar] [CrossRef] [PubMed]
  42. Huo, D.; Feng, Y.; Haddad, S.; Zheng, Y.; Yao, S.; Han, Y.J.; Ogundiran, T.O.; Adebamowo, C.; Ojengbede, O.; Falusi, A.G.; et al. Genome-wide association studies in women of African ancestry identified 3q26.21 as a novel susceptibility locus for oestrogen receptor negative breast cancer. Hum. Mol. Genet. 2016, 25, ddw305. [Google Scholar] [CrossRef] [PubMed]
  43. Cox, D.G.; Curtit, E.; Romieu, G.; Fumoleau, P.; Rios, M.; Bonnefoi, H.; Bachelot, T.; Soulié, P.; Jouannaud, C.; Bourgeois, H.; et al. GWAS in the SIGNAL/PHARE clinical cohort restricts the association between the FGFR2 locus and estrogen receptor status to HER2-negative breast cancer patients. Oncotarget 2016, 7, 77358–77364. [Google Scholar] [CrossRef] [PubMed]
  44. Thomas, G.; Jacobs, K.B.; Kraft, P.; Yeager, M.; Wacholder, S.; Cox, D.G.; Hankinson, S.E.; Hutchinson, A.; Wang, Z.; Yu, K.; et al. A multistage genome-wide association study in breast cancer identifies two new risk alleles at 1p11.2 and 14q24.1 (RAD51L1). Nat. Genet. 2009, 41, 579–584. [Google Scholar] [CrossRef] [PubMed]
  45. Han, M.-R.; Long, J.; Choi, J.-Y.; Low, S.-K.; Kweon, S.-S.; Zheng, Y.; Cai, Q.; Shi, J.; Guo, X.; Matsuo, K.; et al. Genome-wide association study in East Asians identifies two novel breast cancer susceptibility loci. Hum. Mol. Genet. 2016, 25, 3361–3371. [Google Scholar] [CrossRef] [PubMed]
  46. Siddiq, A.; Couch, F.J.; Chen, G.K.; Lindström, S.; Eccles, D.; Millikan, R.C.; Michailidou, K.; Stram, D.O.; Beckmann, L.; Rhie, S.K.; et al. A meta-analysis of genome-wide association studies of breast cancer identifies two novel susceptibility loci at 6q14 and 20q11. Hum. Mol. Genet. 2012, 21, 5373–5384. [Google Scholar] [CrossRef] [PubMed]
  47. Michailidou, K.; Hall, P.; Gonzalez-Neira, A.; Ghoussaini, M.; Dennis, J.; Milne, R.L.; Schmidt, M.K.; Chang-Claude, J.; Bojesen, S.E.; Bolla, M.K.; et al. Large-scale genotyping identifies 41 new loci associated with breast cancer risk. Nat. Genet. 2013, 45, 353–361. [Google Scholar] [CrossRef] [PubMed]
  48. Chen, F.; Chen, G.K.; Stram, D.O.; Millikan, R.C.; Ambrosone, C.B.; John, E.M.; Bernstein, L.; Zheng, W.; Palmer, J.R.; Hu, J.J.; et al. A genome-wide association study of breast cancer in women of African ancestry. Hum. Genet. 2013, 132, 39–48. [Google Scholar] [CrossRef] [PubMed]
  49. Haiman, C.A.; Chen, G.K.; Vachon, C.M.; Canzian, F.; Dunning, A.; Millikan, R.C.; Wang, X.; Ademuyiwa, F.; Ahmed, S.; Ambrosone, C.B.; et al. A common variant at the TERT-CLPTM1L locus is associated with estrogen receptor-negative breast cancer. Nat. Genet. 2011, 43, 1210–1214. [Google Scholar] [CrossRef] [PubMed]
  50. Zheng, W.; Long, J.; Gao, Y.-T.; Li, C.; Zheng, Y.; Xiang, Y.-B.; Wen, W.; Levy, S.; Deming, S.L.; Haines, J.L.; et al. Genome-wide association study identifies a new breast cancer susceptibility locus at 6q25.1. Nat. Genet. 2009, 41, 324–328. [Google Scholar] [CrossRef] [PubMed]
  51. Garcia-Closas, M.; Couch, F.J.; Lindstrom, S.; Michailidou, K.; Schmidt, M.K.; Brook, M.N.; Orr, N.; Rhie, S.K.; Riboli, E.; Feigelson, H.S.; et al. Genome-wide association studies identify four ER negative-specific breast cancer risk loci. Nat. Genet. 2013, 45, 392–398, 398e1-2. [Google Scholar] [CrossRef] [PubMed]
  52. Moreno-Estrada, A.; Gravel, S.; Zakharia, F.; McCauley, J.L.; Byrnes, J.K.; Gignoux, C.R.; Ortiz-Tello, P.A.; Martínez, R.J.; Hedges, D.J.; Morris, R.W.; et al. reconstructing the population genetic history of the Caribbean. PLoS Genet. 2013, 9, e1003925. [Google Scholar] [CrossRef] [PubMed]
  53. Homburger, J.R.; Moreno-Estrada, A.; Gignoux, C.R.; Nelson, D.; Sanchez, E.; Ortiz-Tello, P.; Pons-Estel, B.A.; Acevedo-Vasquez, E.; Miranda, P.; Langefeld, C.D.; et al. genomic insights into the ancestry and demographic history of South America. PLOS Genet. 2015, 11, e1005602. [Google Scholar] [CrossRef] [PubMed]
  54. Brose, M.S.; Rebbeck, T.R.; Calzone, K.A.; Stopfer, J.E.; Nathanson, K.L.; Weber, B.L. Cancer risk estimates for BRCA1 mutation carriers identified in a risk evaluation program. J. Natl. Cancer Inst. 2002, 94, 1365–1372. [Google Scholar] [CrossRef] [PubMed]
  55. Chen, S.; Parmigiani, G. Meta-analysis of BRCA1 and BRCA2 penetrance. J. Clin. Oncol. 2007, 25, 1329–1333. [Google Scholar] [CrossRef] [PubMed]
  56. Easton, D.F.; Ford, D.; Bishop, D.T. Breast and ovarian cancer incidence in BRCA1-mutation carriers. Breast cancer linkage consortium. Am. J. Hum. Genet. 1995, 56, 265–271. [Google Scholar] [PubMed]
  57. Ford, D.; Easton, D.F.; Bishop, D.T.; Narod, S.A.; Goldgar, D.E. Risks of cancer in BRCA1-mutation carriers. Breast cancer linkage consortium. Lancet (London, England) 1994, 343, 692–695. [Google Scholar] [CrossRef]
  58. King, M.-C.; Marks, J.H.; Mandell, J.B. New York breast cancer study group breast and ovarian cancer risks due to inherited mutations in BRCA1 and BRCA2. Science (New York, N.Y.) 2003, 302, 643–646. [Google Scholar] [CrossRef] [PubMed]
  59. Breast cancer linkage consortium cancer risks in BRCA2 mutation carriers. Natl. Cancer Inst. 1999, 91, 1310–1316. [CrossRef]
  60. Thompson, D.; Easton, D.F. Breast cancer linkage consortium cancer incidence in BRCA1 mutation carriers. J. Natl. Cancer Inst. 2002, 94, 1358–1365. [Google Scholar] [CrossRef] [PubMed]
  61. Gomes, M.C.B.; Costa, M.M.; Borojevic, R.; Monteiro, A.N.A.; Vieira, R.; Koifman, S.; Koifman, R.J.; Li, S.; Royer, R.; Zhang, S.; et al. Prevalence of BRCA1 and BRCA2 mutations in breast cancer patients from Brazil. Breast Cancer Res. 2007, 103, 349–353. [Google Scholar] [CrossRef] [PubMed]
  62. Hernandez, J.E.L.; Llacuachaqui, M.; Palacio, G.V.; Figueroa, J.D.; Madrid, J.; Lema, M.; Royer, R.; Li, S.; Larson, G.; Weitzel, J.N.; et al. Prevalence of BRCA1 and BRCA2 mutations in unselected breast cancer patients from Medellin, Colombia. Hered Cancer Clin. Pract. 2014, 12, 11. [Google Scholar] [CrossRef] [PubMed]
  63. Rodriguez, R.C.; Esperon, A.A.; Ropero, R.; Rubio, M.C.; Rodriguez, R.; Ortiz, R.M.; Anta, J.J.L.; los Rios, M.; Carnesolta, D.; Olivera, M.C.; et al. Prevalence of BRCA1 and BRCA2 mutations in breast cancer patients from Cuba. Fam. Cancer 2008, 7, 275–279. [Google Scholar] [CrossRef] [PubMed]
  64. Villarreal-Garza, C.; Alvarez-Gómez, R.M.; Pérez-Plasencia, C.; Herrera, L.A.; Herzog, J.; Castillo, D.; Mohar, A.; Castro, C.; Gallardo, L.N.; Gallardo, D.; et al. Significant clinical impact of recurrent BRCA1 and BRCA2 mutations in Mexico. Cancer 2015, 121, 372–378. [Google Scholar] [CrossRef] [PubMed]
  65. Torres-Mejía, G.; Royer, R.; Llacuachaqui, M.; Akbari, M.R.; Giuliano, A.R.; Martínez-Matsushita, L.; Angeles-Llerenas, A.; Ortega-Olvera, C.; Ziv, E.; Lazcano-Ponce, E.; et al. Recurrent BRCA1 and BRCA2 mutations in Mexican women with breast cancer. Cancer Epidemiol. Biomarkers Prev. 2015, 24, 498–505. [Google Scholar] [CrossRef] [PubMed]
  66. Abugattas, J.; Llacuachaqui, M.; Allende, Y.S.; Velásquez, A.A.; Velarde, R.; Cotrina, J.; Garcés, M.; León, M.; Calderón, G.; la Cruz, M.; et al. Prevalence of BRCA1 and BRCA2 mutations in unselected breast cancer patients from Peru. Clin. Genet. 2015, 88, 371–375. [Google Scholar] [CrossRef] [PubMed]
  67. Diaz-Zabala, H.J.; Ortiz, A.P.; Garland, L.; Jones, K.; Perez, C.M.; Mora, E.; Arroyo, N.; Oleksyk, T.K.; Echenique, M.; Matta, J.L.; et al. A recurrent BRCA2 mutation explains the majority of hereditary breast and ovarian cancer syndrome cases in Puerto Rico. Cancers 2018, 10, 419. [Google Scholar] [CrossRef] [PubMed]
  68. Dutil, J.; Golubeva, V.A.; Pacheco-Torres, A.L.; Diaz-Zabala, H.J.; Matta, J.L.; Monteiro, A.N. The spectrum of BRCA1 and BRCA2 alleles in Latin America and the Caribbean: a clinical perspective. Breast Cancer Res. 2015, 154, 441–453. [Google Scholar] [CrossRef] [PubMed]
  69. Hedenfalk, I.; Duggan, D.; Chen, Y.; Radmacher, M.; Bittner, M.; Simon, R.; Meltzer, P.; Gusterson, B.; Esteller, M.; Kallioniemi, O.P.; et al. Gene-expression profiles in hereditary breast cancer. N. Engl. J. Med. 2001, 344, 539–548. [Google Scholar] [CrossRef] [PubMed]
  70. Frank, T.S.; Deffenbaugh, A.M.; Reid, J.E.; Hulick, M.; Ward, B.E.; Lingenfelter, B.; Gumpper, K.L.; Scholl, T.; Tavtigian, S.; Pruss, D.R.; et al. Clinical characteristics of individuals with germline mutations in BRCA1 and BRCA2: analysis of 10,000 individuals. J. Clin. Oncol. 2002, 20, 1480–1490. [Google Scholar] [CrossRef] [PubMed]
  71. Solano, A.; Aceto, G.; Delettieres, D.; Veschi, S.; Neuman, M.; Alonso, E.; Chialina, S.; Chacón, R.; Renato, M.-C.; Podestá, E. BRCA1 and BRCA2 analysis of Argentinean breast/ovarian cancer patients selected for age and family history highlights a role for novel mutations of putative south-American origin. SpringerPlus 2012, 1, 20. [Google Scholar] [CrossRef] [PubMed]
  72. Alvarez, C.; Tapia, T.; Perez-Moreno, E.; Gajardo-Meneses, P.; Ruiz, C.; Rios, M.; Missarelli, C.; Silva, M.; Cruz, A.; Matamala, L.; et al. BRCA1 and BRCA2 founder mutations account for 78% of germline carriers among hereditary breast cancer families in Chile. Oncotarget 2017. [Google Scholar] [CrossRef] [PubMed]
  73. Weitzel, J.N.; Lagos, V.; Blazer, K.R.; Nelson, R.; Ricker, C.; Herzog, J.; McGuire, C.; Neuhausen, S. Prevalence of BRCA mutations and founder effect in high-risk Hispanic families. Cancer Epidemiol. Biomarkers Prev. 2005, 14, 1666–1671. [Google Scholar] [CrossRef] [PubMed]
  74. Ossa, C.A.; Torres, D. Founder and Recurrent Mutations in BRCA1 and BRCA2 Genes in Latin American Countries: State of the Art and Literature Review. Oncologist 2016, 21, 832–839. [Google Scholar] [CrossRef] [PubMed]
  75. Ashton-Prolla, P.; Vargas, F.R. Prevalence and impact of founder mutations in hereditary breast cancer in Latin America. Genet. Mol. Biol. 2014, 37, 234–240. [Google Scholar] [CrossRef] [PubMed]
  76. Solano, A.R.; Cardoso, F.C.; Romano, V.; Perazzo, F.; Bas, C.; Recondo, G.; Santillan, F.B.; Gonzalez, E.; Abalo, E.; Viniegra, M.; et al. Spectrum of BRCA1/2 variants in 940 patients from Argentina including novel, deleterious and recurrent germline mutations: impact on healthcare and clinical practice. Oncotarget 2017, 8, 60487–60495. [Google Scholar] [CrossRef] [PubMed]
  77. Alemar, B.; Herzog, J.; Brinckmann Oliveira Netto, C.; Artigalás, O.; Schwartz, I.V.D.; Matzenbacher Bittar, C.; Ashton-Prolla, P.; Weitzel, J.N. Prevalence of Hispanic BRCA1 and BRCA2 mutations among hereditary breast and ovarian cancer patients from Brazil reveals differences among Latin American populations. Cancer Genet. 2016, 209, 417–422. [Google Scholar] [CrossRef] [PubMed]
  78. Palmero, E.I.; Carraro, D.M.; Alemar, B.; Moreira, M.A.M.; Ribeiro-Dos-Santos, Â.; Abe-Sandes, K.; Galvão, H.C.R.; Reis, R.M.; Pádua Souza, C.; Campacci, N.; et al. The germline mutational landscape of BRCA1 and BRCA2 in Brazil. Sci. Rep. 2018, 8, 1–10. [Google Scholar] [CrossRef] [PubMed]
  79. Cock-Rada, A.M.; Ossa, C.A.; Garcia, H.I.; Gomez, L.R. A multi-gene panel study in hereditary breast and ovarian cancer in Colombia. Fam. Cancer 2018, 17, 23–30. [Google Scholar] [CrossRef] [PubMed]
  80. Walsh, T.; Casadei, S.; Coats, K.H.; Swisher, E.; Stray, S.M.; Higgins, J.; Roach, K.C.; Mandell, J.; Lee, M.K.; Ciernikova, S.; et al. Spectrum of mutations in BRCA1, BRCA2, CHEK2, and TP53 in families at high risk of breast cancer. JAMA 2006, 295, 1379. [Google Scholar] [CrossRef] [PubMed]
  81. Walsh, T.; King, M.-C. Ten genes for inherited breast cancer. Cancer Cell 2007, 11, 103–105. [Google Scholar] [CrossRef] [PubMed]
  82. Ripperger, T.; Gadzicki, D.; Meindl, A.; Schlegelberger, B. Breast cancer susceptibility: current knowledge and implications for genetic counselling. Eur. J. Hum. Genet. 2009, 17, 722–731. [Google Scholar] [CrossRef] [PubMed]
  83. Cury, N.M.; Ferraz, V.E.; Silva, W.A., Jr. TP53 p.R337H prevalence in a series of Brazilian hereditary breast cancer families. Hered Cancer Clin. 2014, 12, 8. [Google Scholar] [CrossRef] [PubMed]
  84. Gomes, M.C.B.; Kotsopoulos, J.; de Almeida, G.L.; Costa, M.M.; Vieira, R.; Filho, F.d.G.; Pitombo, M.B.; Leal, P.R.F.; Royer, R.; Zhang, P.; et al. The R337H mutation in TP53 and breast cancer in Brazil. Hered Cancer Clin. Pract. 2012, 10, 3. [Google Scholar] [CrossRef] [PubMed]
  85. Assumpção, J.G.; Seidinger, A.L.; Mastellaro, M.J.; Ribeiro, R.C.; Zambetti, G.P.; Ganti, R.; Srivastava, K.; Shurtleff, S.; Pei, D.; Zeferino, L.C.; et al. Association of the germline TP53 R337H mutation with breast cancer in southern Brazil. BMC Cancer 2008, 8, 357. [Google Scholar] [CrossRef] [PubMed]
  86. Weber, B.L.; Nathanson, K.L. Low penetrance genes associated with increased risk for breast cancer. Eur. J. Cancer 2000, 36, 1193–1199. [Google Scholar] [CrossRef]
  87. Banin Hirata, B.K.; Losi Guembarovski, R.; Vitiello, G.A.F.; Guembarovski, A.L.; de Oliveira, K.B.; Watanabe, M.A.E. FOXP3 allelic variants and haplotype structures are associated with aggressive breast cancer subtypes. Dis. Markers 2017, 2017, 6359603. [Google Scholar] [CrossRef] [PubMed]
  88. Zara-Lopes, T.; Gimenez-Martins, A.P.A.; Nascimento-Filho, C.H.; Castanhole-Nunes, M.M.U.; Galbiatti-Dias, A.L.S.; Padovani-Júnior, J.A.; Maniglia, J.; Francisco, J.L.E.; Pavarino, E.C.; Goloni-Bertollo, E.M. Role of MTHFR C677T and MTR A2756G polymorphisms in thyroid and breast cancer development. Genet. Mol. Res. 2016, 15. [Google Scholar] [CrossRef] [PubMed]
  89. Oliveira, C.B.M.; Cardoso-Filho, C.; Bossi, L.S.; Lourenço, G.J.; Costa-Gurgel, M.S.; Lima, C.S.P. Association of CYP1A1 A4889G and T6235C polymorphisms with the risk of sporadic breast cancer in Brazilian women. Clinics (Sao Paulo) 2015, 70, 680–685. [Google Scholar] [CrossRef]
  90. de Carlos Back, L.K.; Farias, T.D.J.; Cunha, P.A.; Muniz, Y.C.N.; Ribeiro, M.C.M.; Fernandes, B.L.; Fernandes, C.K.; Souza, I.R. Functional polymorphisms of interleukin-18 gene and risk of breast cancer in a Brazilian population. Tissue Antigens 2014, 84, 229–233. [Google Scholar] [CrossRef] [PubMed]
  91. Possuelo, L.G.; Peraça, C.F.; Eisenhardt, M.F.; Dotto, M.L.; Cappelletti, L.; Foletto, E.; de Moura Valim, A.R. Polymorphisms of GSTM1 and GSTT1 genes in breast cancer susceptibility: A case-control study. Rev. Bras. Ginecol e Obs. 2013, 35, 569–574. [Google Scholar] [CrossRef]
  92. Linhares, J.J.; Azevedo, M.; Siufi, A.A.; Carvalho, C.V.; Wolgien, M.D.C.G.M.; Noronha, E.C.; de Souza, B.T.C.; Silva, I.D.C.G. Evaluation of single nucleotide polymorphisms in microRNAs (hsa-miR-196a2 rs11614913 C/T) from Brazilian women with breast cancer. BMC Med. Genet. 2012, 13, 119. [Google Scholar] [CrossRef] [PubMed]
  93. Morales, S.; Gulppi, F.; Gonzalez-Hormazabal, P.; Fernandez-Ramires, R.; Bravo, T.; Reyes, J.M.; Gomez, F.; Waugh, E.; Jara, L. Association of single nucleotide polymorphisms in Pre-miR-27a, Pre-miR-196a2, Pre-miR-423, miR-608 and Pre-miR-618 with breast cancer susceptibility in a South American population. BMC Genet. 2016, 17, 109. [Google Scholar] [CrossRef] [PubMed]
  94. Leyton, Y.; Gonzalez-Hormazabal, P.; Blanco, R.; Bravo, T.; Fernandez-Ramires, R.; Morales, S.; Landeros, N.; Reyes, J.M.; Peralta, O.; Tapia, J.C.; et al. Association of PALB2 sequence variants with the risk of familial and early-onset breast cancer in a South-American population. BMC Cancer 2015, 15, 30. [Google Scholar] [CrossRef] [PubMed]
  95. Elematore, I.; Gonzalez-Hormazabal, P.; Reyes, J.M.; Blanco, R.; Bravo, T.; Peralta, O.; Gomez, F.; Waugh, E.; Margarit, S.; Ibañez, G.; et al. Association of genetic variants at TOX3, 2q35 and 8q24 with the risk of familial and early-onset breast cancer in a South-American population. Mol. Biol. Rep. 2014, 41, 3715–3722. [Google Scholar] [CrossRef] [PubMed]
  96. Jara, L.; Gonzalez-Hormazabal, P.; Cerceño, K.; Capua, G.A.; Reyes, J.M.; Blanco, R.; Bravo, T.; Peralta, O.; Gomez, F.; Waugh, E.; et al. Genetic variants in FGFR2 and MAP3K1 are associated with the risk of familial and early-onset breast cancer in a South-American population. Breast Cancer Res. Treat. 2013, 137, 559–569. [Google Scholar] [CrossRef] [PubMed]
  97. Gonzalez-Hormazabal, P.; Reyes, J.M.; Blanco, R.; Bravo, T.; Carrera, I.; Peralta, O.; Gomez, F.; Waugh, E.; Margarit, S.; Ibañez, G.; et al. The BARD1 Cys557Ser variant and risk of familial breast cancer in a South-American population. Mol. Biol. Rep. 2012, 39, 8091–8098. [Google Scholar] [CrossRef] [PubMed]
  98. Jara, L.; Dubois, K.; Gaete, D.; Mayo, T.; Ratkevicius, N.; Bravo, T.; Margarit, S.; Blanco, R.; Gómez, F.; Waugh, E.; et al. Variants in DNA double-strand break repair genes and risk of familial breast cancer in a South American population. Breast Cancer Res. Treat. 2010, 122, 813–822. [Google Scholar] [CrossRef] [PubMed]
  99. Tapia, T.; Sanchez, A.; Vallejos, M.; Alvarez, C.; Moraga, M.; Smalley, S.; Camus, M.; Alvarez, M.; Carvallo, P. ATM allelic variants associated to hereditary breast cancer in 94 Chilean women: Susceptibility or ethnic influences? Breast Cancer Res. Treat. 2008, 107, 281–288. [Google Scholar] [CrossRef] [PubMed]
  100. González-Hormazábal, P.; Bravo, T.; Blanco, R.; Valenzuela, C.Y.; Gómez, F.; Waugh, E.; Peralta, O.; Ortuzar, W.; Reyes, J.M.; Jara, L. Association of common ATM variants with familial breast cancer in a South American population. BMC Cancer 2008, 8, 1–8. [Google Scholar] [CrossRef] [PubMed]
  101. Jara, L.; Acevedo, M.L.; Blanco, R.; Castro, V.G.; Bravo, T.; Gómez, F.; Waugh, E.; Peralta, O.; Cabrera, E.; Reyes, J.M.; et al. RAD51 135G>C polymorphism and risk of familial breast cancer in a South American population. Cancer Genet. Cytogenet. 2007, 178, 65–69. [Google Scholar] [CrossRef] [PubMed]
  102. López-Cortés, A.; Leone, P.E.; Freire-Paspuel, B.; Arcos-Villacís, N.; Guevara-Ramírez, P.; Rosales, F.; Paz-Y-Miño, C. Mutational analysis of oncogenic AKT1 gene associated with breast cancer risk in the high-altitude Ecuadorian mestizo population. BioMed Res. Int. 2018, 2018. [Google Scholar] [CrossRef] [PubMed]
  103. López-Cortés, A.; Echeverría, C.; Oña-Cisneros, F.; Sánchez, M.E.; Herrera, C.; Cabrera-Andrade, A.; Rosales, F.; Ortiz, M.; Paz-y-Miño, C. Breast cancer risk associated with gene expression and genotype polymorphisms of the folate-metabolizing MTHFR gene: A case-control study in a high-altitude Ecuadorian mestizo population. Tumor Biol. 2015, 36, 6451–6461. [Google Scholar] [CrossRef] [PubMed]
  104. Muñoz-Palomeque, A.; Guerrero-Ramirez, M.A.; Rubio-Chavez, L.A.; Rosales-Gomez, R.C.; Lopez-Cardona, M.G.; Barajas-Avila, V.H.; Delgadillo-Barrera, A.; Canton-Romero, J.C.; Montoya-Fuentes, H.; Garcia-Cobian, T.A.; et al. Association of RETN and CAP1 SNPs, expression and serum resistin levels with breast cancer in Mexican women. Genet. Test. Mol. Biomarkers 2018, 22, 209–217. [Google Scholar] [CrossRef] [PubMed]
  105. Jaramillo-rangel, G.; Ortega-martínez, M.; Cerda-flores, R.M.; Barrera-saldaña, H.A. C3435T polymorphism in the MDR1 gene and breast cancer risk in northeastern Mexico. Int. J. Clin. Exp. Patho 2018, 11, 904–909. [Google Scholar]
  106. Gutierrez-Rubio, S.A.; Quintero-Ramos, A.; Durán-Cárdenas, A.; Franco-Topete, R.A.; Castro-Cervantes, J.M.; Oceguera-Villanueva, A.; Jiménez-Pérez, L.M.; Balderas-Peña, L.M.A.; Morgan-Villela, G.; Del-Toro-Arreola, A.; et al. 1236 C/T and 3435 C/T polymorphisms of the ABCB1 gene in Mexican breast cancer patients. Genet. Mol. Res. 2015, 14, 1250–1259. [Google Scholar] [CrossRef] [PubMed]
  107. García-Martínez, A.; Gamboa-Loira, B.; Tejero, M.E.; Sierra-Santoyo, A.; Cebrián, M.E.; López-Carrillo, L. CYP1A1, CYP1B1, GSTM1 and GSTT1 genetic variants and breast cancer risk in Mexican women. Salud Publica Mex. 2017, 59, 540. [Google Scholar] [CrossRef] [PubMed]
  108. Soto-Quintana, O.; Zúñiga-González, G.M.; Ramírez-Patiño, R.; Ramos-Silva, A.; Figuera, L.E.; Carrillo-Moreno, D.I.; Gutiérrez-Hurtado, I.A.; Puebla-Pérez, A.M.; Sánchez-Llamas, B.; Gallegos-Arreola, M.P. Association of the GSTM1 null polymorphism with breast cancer in a Mexican population. Genet. Mol. Res. 2015, 14, 13066–13075. [Google Scholar] [CrossRef] [PubMed]
  109. Jaramillo-Rangel, G.; Ortega-Martínez, M.; Cerda-Flores, R.M.; Barrera-Saldaña, H.A. Short Communication Polymorphisms in GSTM1, GSTT1, GSTP1, and GSTM3 genes and breast cancer risk in northeastern Mexico. Genet. Mol. Res. 2015, 14, 6465–6471. [Google Scholar] [CrossRef] [PubMed]
  110. Ramos-Silva, A.; Figuera, L.E.; Soto-Quintana, O.M.; Puebla-Pérez, A.M.; Ramírez-Patiño, R.; Gutiérrez-Hurtado, I.; Carrillo-Moreno, D.I.; Zúñiga-González, G.M.; Dávalos-Rodríguez, I.P.; Gallegos-Arreola, M.P. Association of the C677T polymorphism in the methylenetetrahydrofolate reductase gene with breast cancer in a Mexican population. Genet. Mol. Res. 2015, 14, 4015–4026. [Google Scholar] [CrossRef] [PubMed]
  111. Gallegos-Arreola, M.P.; Figuera-Villanueva, L.E.; Ramos-Silva, A.; Salas-González, E.; Puebla-Pérez, A.M.; Peralta-Leal, V.; García-Ortiz, J.E.; Dávalos-Rodríguez, I.P.; Zúńiga-González, G.M. The association between the 844ins68 polymorphism in the CBS gene and breast cancer. Arch. Med. Sci. 2014, 6, 1214–1224. [Google Scholar] [CrossRef] [PubMed]
  112. Calderón-Zúñiga, F.C.; Ocampo-Gómez, G.; López-Márquez, F.C.; Recio-Vega, R.; Serrano-Gallardo, L.B.; Ruiz-Flores, P. ATM polymorphisms IVS24-9delT, IVS38-8T > C, and 5557G > A in Mexican women with familial and/or early-onset breast cancer. Salud Publica Mex. 2014, 56, 206–212. [Google Scholar] [CrossRef]
  113. Martínez-Ramírez, O.C.; Pérez-Morales, R.; Castro, C.; Flores-Díaz, A.; Soto-Cruz, K.E.; Astorga-Ramos, A.; Gonsebatt, M.E.; Casas, L.; Valdés-Flores, M.; Rubio, J. Polymorphisms of catechol estrogens metabolism pathway genes and breast cancer risk in Mexican women. Breast 2013, 22, 335–343. [Google Scholar] [CrossRef] [PubMed]
  114. Ramírez-Patiño, R.; Figuera, L.E.; Puebla-Pérez, A.M.; Delgado-Saucedo, J.I.; Legazpí-Macias, M.M.; Mariaud-Schmidt, R.P.; Ramos-Silva, A.; Gutiérrez-Hurtado, I.A.; Gómez Flores-Ramos, L.; Zúñiga-González, G.M.; et al. Intron 4 VNTR (4a/b) Polymorphism of the Endothelial Nitric Oxide Synthase Gene Is Associated with Breast Cancer in Mexican Women. J. Korean Med. Sci. 2013, 28, 1587. [Google Scholar] [CrossRef] [PubMed]
  115. Moreno-Galván, M.; Herrera-González, N.E.; Robles-Pérez, V.; Velasco-Rodríguez, J.C.; Tapia-Conyer, R.; Sarti, E. Impact of CYP1A1 and COMT genotypes on breast cancer risk in Mexican women: A pilot study. Int. J. Biol. Markers 2010, 25, 157–163. [Google Scholar] [CrossRef] [PubMed]
  116. Murillo-Zamora, E.; Moreno-Macías, H.; Ziv, E.; Romieu, I.; Lazcano-Ponce, E.; Ángeles-Llerenas, A.; Pérez-Rodríguez, E.; Vidal-Millán, S.; Fejerman, L.; Torres-Mejía, G. Association between rs2981582 polymorphism in the FGFR2 gene and the risk of breast cancer in Mexican women. Arch. Med. Res. 2013, 44, 459–466. [Google Scholar] [CrossRef] [PubMed]
  117. Pérez-Mayoral, J.; Pacheco-Torres, A.L.; Morales, L.; Acosta-Rodríguez, H.; Matta, J.L.; Dutil, J. Genetic polymorphisms in RAD23B and XPC modulate DNA repair capacity and breast cancer risk in Puerto Rican women. Mol. Carcinog. 2013, 52 (Suppl. 1), E127–E138. [Google Scholar]
  118. Quintero-Ramos, A.; Gutiérrez-Rubio, S.A.; Toro-Arreola, A.; Franco-Topete, R.A.; Oceguera-Villanueva, A.; Jiménez-Pérez, L.M.; Castro-Cervantes, J.M.; Barragán-Ruiz, A.; Vázquez-Camacho, J.G.; Daneri-Navarro, A. Association between polymorphisms in the thymidylate synthase gene and risk of breast cancer in a Mexican population. Genet. Mol. Res. 2014, 13, 8749–8756. [Google Scholar] [CrossRef] [PubMed]
  119. Silva, F.C.; Lisboa, B.C.; Figueiredo, M.C.; Torrezan, G.T.; Santos, É.M.; Krepischi, A.C.; Rossi, B.M.; Achatz, M.I.; Carraro, D.M. Hereditary breast and ovarian cancer: assessment of point mutations and copy number variations in Brazilian patients. BMC Med. Genet. 2014, 15, 55. [Google Scholar] [CrossRef] [PubMed]
  120. Gallegos-Arreola, M.P.; Figuera, L.E.; Flores-Ramos, L.G.; Puebla-Pérez, A.M.; Zúñiga-González, G.M. Association of the Alu insertion polymorphism in the progesterone receptor gene with breast cancer in a Mexican population. Arch. Med. Sci. 2015, 11, 551–560. [Google Scholar] [CrossRef] [PubMed]
  121. Macías-Gómez, N.M.; Peralta-Leal, V.; Meza-Espinoza, J.P.; Gutiérrez-Angulo, M.; Durán-González, J.; Ramírez-González, J.M.; Gaspar-Del Toro, A.; Norberto-Rodríguez, A.; Leal-Ugarte, E. Polymorphisms of the XRCC1 gene and breast cancer risk in the Mexican population. Fam. Cancer 2015, 14, 349–354. [Google Scholar] [CrossRef] [PubMed]
  122. González-Rivera, M.; Lobo, M.; López-Tarruella, S.; Jerez, Y.; Monte-Millán, M.; Massarrah, T.; Ramos-Medina, R.; Ocaña, I.; Picornell, A.; Garzón, S.S.; et al. Frequency of germline DNA genetic findings in an unselected prospective cohort of triple-negative breast cancer patients participating in a platinum-based neoadjuvant chemotherapy trial. Breast Cancer Res. Treat. 2016, 156, 507–515. [Google Scholar] [CrossRef] [PubMed]
  123. González-Hormazábal, P.; Castro, V.G.; Blanco, R.; Gómez, F.; Peralta, O.; Waugh, E.; Bravo, T.; Reyes, J.M.; Jara, L. Absence of CHEK2 1100delC mutation in familial breast cancer cases from a South American population. Breast Cancer Res. Treat. 2008, 110, 543–545. [Google Scholar] [CrossRef] [PubMed]
  124. Shiloh, Y.; Ziv, Y. The ATM protein kinase: regulating the cellular response to genotoxic stress and more. Nat. Rev. Mol. Cell Biol. 2013, 14, 197–210. [Google Scholar] [CrossRef] [PubMed]
  125. Irminger-Finger, I.; Ratajska, M.; Pilyugin, M. New concepts on BARD1: Regulator of BRCA pathways and beyond. Int. J. Biochem. Cell Biol. 2016, 72, 1–17. [Google Scholar] [CrossRef] [PubMed]
  126. Thai, T.H.; Du, F.; Tsan, J.T.; Jin, Y.; Phung, A.; Spillman, M.A.; Massa, H.F.; Muller, C.Y.; Ashfaq, R.; Mathis, J.M.; et al. Mutations in the BRCA1-associated RING domain (BARD1) gene in primary breast, ovarian and uterine cancers. Hum. Mol. Genet. 1998, 7, 195–202. [Google Scholar] [CrossRef] [PubMed]
  127. Ishitobi, M.; Miyoshi, Y.; Hasegawa, S.; Egawa, C.; Tamaki, Y.; Monden, M.; Noguchi, S. Mutational analysis of BARD1 in familial breast cancer patients in Japan. Cancer Lett. 2003, 200, 1–7. [Google Scholar] [CrossRef]
  128. Stacey, S.N.; Sulem, P.; Johannsson, O.T.; Helgason, A.; Gudmundsson, J.; Kostic, J.P.; Kristjansson, K.; Jonsdottir, T.; Sigurdsson, H.; Hrafnkelsson, J.; et al. The BARD1 Cys557Ser variant and breast cancer risk in Iceland. PLoS Med. 2006, 3, e217. [Google Scholar] [CrossRef] [PubMed]
  129. Vahteristo, P.; Syrjäkoski, K.; Heikkinen, T.; Eerola, H.; Aittomäki, K.; Smitten, K.; Holli, K.; Blomqvist, C.; Kallioniemi, O.-P.; Nevanlinna, H. BARD1 variants Cys557Ser and Val507Met in breast cancer predisposition. Eur. J. Hum. Genet. 2006, 14, 167–172. [Google Scholar] [CrossRef] [PubMed]
  130. Jakubowska, A.; Cybulski, C.; Szymańska, A.; Huzarski, T.; Byrski, T.; Gronwald, J.; Dębniak, T.; Górski, B.; Kowalska, E.; Narod, S.A.; et al. BARD1 and breast cancer in Poland. Breast Cancer Res. Treat. 2007, 107, 119–122. [Google Scholar] [CrossRef] [PubMed]
  131. Gorringe, K.L.; Choong, D.Y.H.; Visvader, J.E.; Lindeman, G.J.; Campbell, I.G. BARD1 variants are not associated with breast cancer risk in Australian familial breast cancer. Breast Cancer Res. Treat. 2008, 111, 505–509. [Google Scholar] [CrossRef] [PubMed]
  132. Johnatty, S.E.; Beesley, J.; Chen, X.; Hopper, J.L.; Southey, M.C.; Giles, G.G.; Goldgar, D.E.; Chenevix-Trench, G.; Spurdle, A.B. Australian Ovarian Cancer Study Group; et al. The BARD1 Cys557Ser polymorphism and breast cancer risk: An Australian case–control and family analysis. Breast Cancer Res. Treat. 2009, 115, 145–150. [Google Scholar] [CrossRef] [PubMed]
  133. Zhang, F.; Ma, J.; Wu, J.; Ye, L.; Cai, H.; Xia, B.; Yu, X. PALB2 links BRCA1 and BRCA2 in the DNA-damage response. Curr Biol 2009, 19, 524–529. [Google Scholar] [CrossRef] [PubMed]
  134. Apostolou, P.; Papasotiriou, I. Current perspectives on CHEK2 mutations in breast cancer. Breast Cancer (Dove Med. Press) 2017, 9, 331–335. [Google Scholar] [CrossRef] [PubMed]
  135. Schmidt, M.K.; Hogervorst, F.; Hien, R.; Cornelissen, S.; Broeks, A.; Adank, M.A.; Meijers, H.; Waisfisz, Q.; Hollestelle, A.; Schutte, M.; et al. Age- and tumor subtype–specific breast cancer risk estimates for CHEK2*1100delC carriers. J. Clin. Oncol 2016, 34, 2750–2760. [Google Scholar] [CrossRef] [PubMed]
  136. Abud, J.; Koehler-Santos, P.; Ashton-Prolla, P.; Prolla, J.C.; Study Group on Hereditary Breast and Colorectal Cancer. CHEK2 1100DELC germline mutation: A frequency study in hereditary breast and colon cancer Brazilian families. Arq. Gastroenterol. 2012, 49, 273–278. [Google Scholar] [CrossRef] [PubMed]
  137. Rivera-Herrera, A.-L.; Cifuentes-C, L.; Gil-Vera, J.; Barreto, G. Absence of the CHEK2 c.1100delC mutation in familial breast and ovarian cancer in Colombia: A case-control study. F1000Research 2018, 7, 1032. [Google Scholar] [CrossRef]
  138. The Cancer Genome Atlas Network. Comprehensive molecular portraits of human breast tumours. Nature 2012, 490, 61–70. [Google Scholar] [CrossRef] [PubMed]
  139. Chen, J.; Shao, P.; Cao, Q.; Li, P.; Li, J.; Cai, H.; Zhu, J.; Wang, M.; Zhang, Z.; Qin, C.; et al. Genetic variations in a PTEN/AKT/mTOR axis and prostate cancer risk in a Chinese population. PloS ONE 2012, 7, e40817. [Google Scholar] [CrossRef] [PubMed]
  140. Williams, A.B.; Schumacher, B. p53 in the DNA-damage-repair process. Cold Spring Harb. Perspect. Med. 2016, 6, a026070. [Google Scholar] [CrossRef] [PubMed]
  141. Damin, A.P.S.; Frazzon, A.P.G.; Damin, D.C.; Roehe, A.; Hermes, V.; Zettler, C.; Alexandre, C.O.P. Evidence for an association of TP53 codon 72 polymorphism with breast cancer risk. Cancer Detect. Prev. 2006, 30, 523–529. [Google Scholar] [CrossRef] [PubMed]
  142. Aoki, M.N.; Silva do Amaral Herrera, A.C.; Amarante, M.K.; Val Carneiro, J.L.; Fungaro, M.H.P.; Watanabe, M.A.E. CCR5 and p53 codon 72 gene polymorphisms: implications in breast cancer development. Int. J. Mol. Med. 2009, 23, 429–435. [Google Scholar] [PubMed]
  143. Almeida, B.C.; Kleine, J.P.F.O.; Camargo-Kosugi, C.M.; Lisboa, M.R.; França, C.N.; França, J.P.; Silva, I.D.C.G. Analysis of polymorphisms in codons 11, 72 and 248 of TP53 in Brazilian women with breast cancer. Genet. Mol. Res. 2016, 15. [Google Scholar] [CrossRef] [PubMed]
  144. Ramalho, E.A.; Silva-Filho, J.L.; Cartaxo, M.F.; Cavalcanti, C.B.; Rêgo, M.J.; Oliveira, M.B.; Beltrão, E.I. Assessment of changes in the BRCA2 and P53 genes in breast invasive ductal carcinoma in northeast Brazil. Biol Res. 2014, 47, 3. [Google Scholar] [PubMed]
  145. Achatz, M.I.W.; Olivier, M.; Calvez, F.; Martel-Planche, G.; Lopes, A.; Rossi, B.M.; Ashton-Prolla, P.; Giugliani, R.; Palmero, E.I.; Vargas, F.R.; et al. The TP53 mutation, R337H, is associated with Li-Fraumeni and Li-Fraumeni-like syndromes in Brazilian families. Cancer Lett. 2007, 245, 96–102. [Google Scholar] [CrossRef] [PubMed]
  146. Hou, J.; Jiang, Y.; Tang, W.; Jia, S. p53 codon 72 polymorphism and breast cancer risk: A meta-analysis. Exp. Ther Med. 2013, 5, 1397–1402. [Google Scholar] [CrossRef] [PubMed]
  147. Conceição, L.L.; Pessoa, M.C.; Hermsdorff, H.H.M.; Freitas, R.N.; Do Carmo Gouveia Peluzio Peluzio, M. MTHFR and MTR Polymorphisms and Breast Cancer in Brazilian Women. World J. Res. Rev. 2016, 2, 29–32. [Google Scholar]
  148. Slattery, M.L.; Sweeney, C.; Herrick, J.; Wolff, R.; Baumgartner, K.; Giuliano, A.; Byers, T. ESR1, AR, body size, and breast cancer risk in Hispanic and non-Hispanic white women living in the Southwestern United States. Breast Cancer Res. Treat. 2007, 105, 327–335. [Google Scholar] [CrossRef] [PubMed]
  149. Haiman, C.A.; Garcia, R.R.; Hsu, C.; Xia, L.; Ha, H.; Sheng, X.; Marchand, L.; Kolonel, L.N.; Henderson, B.E.; Stallcup, M.R.; et al. Screening and association testing of common coding variation in steroid hormone receptor co-activator and co-repressor genes in relation to breast cancer risk: The multiethnic cohort. BMC Cancer 2009, 9, 43. [Google Scholar] [CrossRef] [PubMed]
  150. Boone, S.D.; Baumgartner, K.B.; Baumgartner, R.N.; Connor, A.E.; Pinkston, C.M.; Rai, S.N.; Riley, E.C.; Hines, L.M.; Giuliano, A.R.; John, E.M.; et al. Associations between CYP19A1 polymorphisms, native American ancestry, and breast cancer risk and mortality: The breast cancer health disparities study. Cancer Causes Control. 2014, 25, 1461–1471. [Google Scholar] [CrossRef] [PubMed]
  151. Haiman, C.A.; Stram, D.O.; Pike, M.C.; Kolonel, L.N.; Burtt, N.P.; Altshuler, D.; Hirschhorn, J.; Henderson, B.E. A comprehensive haplotype analysis of CYP19 and breast cancer risk: The Multiethnic Cohort. Hum. Mol. Genet. 2003, 12, 2679–2692. [Google Scholar] [CrossRef] [PubMed]
  152. Weston, A.; Pan, C.F.; Bleiweiss, I.J.; Ksieski, H.B.; Roy, N.; Maloney, N.; Wolff, M.S. CYP17 genotype and breast cancer risk. Cancer Epidemiol. Biomarkers Prev. 1998, 7, 941–944. [Google Scholar] [PubMed]
  153. Sierra-Martinez, M.; Hernández-Cadena, L.; García-Sánchez, J.R.; Acosta-Altamirano, G.; Palacios-Reyes, C.; Alonso-Themann, P.G.; García-Ortiz, L.; Quintas-Granados, L.I.; Reyes-Hernández, O.D. Predictive polymorphisms for breast cancer in postmenopausal Mexican women. J. Cancer Res. Ther 2018, 14, 640–646. [Google Scholar] [PubMed]
  154. Gialeli, C.; Theocharis, A.D.; Karamanos, N.K. Roles of matrix metalloproteinases in cancer progression and their pharmacological targeting. FEBS J. 2011, 278, 16–27. [Google Scholar] [CrossRef] [PubMed]
  155. Slattery, M.L.; John, E.; Torres-Mejia, G.; Stern, M.; Lundgreen, A.; Hines, L.; Giuliano, A.; Baumgartner, K.; Herrick, J.; Wolff, R.K. Matrix metalloproteinase genes are associated with breast cancer risk and survival: The breast cancer health disparities study. PLoS ONE 2013, 8, e63165. [Google Scholar] [CrossRef] [PubMed]
  156. Delgado-Enciso, I.; Cepeda-Lopez, F.R.; Monrroy-Guizar, E.A.; Bautista-Lam, J.R.; Andrade-Soto, M.; Jonguitud-Olguin, G.; Rodriguez-Hernandez, A.; Anaya-Ventura, A.; Baltazar-Rodriguez, L.M.; Orozco-Ruiz, M.; et al. Matrix metalloproteinase-2 promoter polymorphism is associated with breast cancer in a Mexican population. Gynecol Obstet Investig. 2008, 65, 68–72. [Google Scholar] [CrossRef] [PubMed]
  157. Acosta, K.B.; Lorenzini Campos, M.N.; Etcheverry, S.B.; Zapata, P.D. α6β4 integrin genetic variations (A380T and R1281W) and breast cancer risk in an Argentinian population. Int. J. Mol. Sci. 2016, 17, 1540. [Google Scholar] [CrossRef] [PubMed]
  158. Felizi, R.T.; Veiga, M.G.; Carelli-Filho, I.; Souto, R.P.D.; Fernandes, C.E.; Oliveira, E. Association between matrix metallopeptidase 9 polymorphism and breast cancer risk. Rev. Bras. Ginecol. Obstet 2018, 40, 620–624. [Google Scholar] [CrossRef] [PubMed]
  159. Slattery, M.L.; John, E.M.; Torres-Mejia, G.; Lundgreen, A.; Herrick, J.S.; Baumgartner, K.B.; Hines, L.M.; Stern, M.C.; Wolff, R.K. Genetic variation in genes involved in hormones, inflammation and energetic factors and breast cancer risk in an admixed population. Carcinogenesis 2012, 33, 1512–1521. [Google Scholar] [CrossRef] [PubMed]
  160. Slattery; Herrick, J.S.; Torres-Mejia, G.; John, E.M.; Giuliano, A.R.; Hines, L.M.; Stern, M.C.; Baumgartner, K.B.; Presson, A.P.; Wolff, R.K. Genetic variants in interleukin genes are associated with breast cancer risk and survival in a genetically admixed population: The breast cancer health disparities study. Carcinogenesis 2014, 35, 1750–1759. [Google Scholar] [CrossRef] [PubMed]
  161. Slattery, M.L.; Lundgreen, A.; Hines, L.; Wolff, R.K.; Torres-Mejia, G.; Baumgartner, K.N.; John, E.M. Energy homeostasis genes and breast cancer risk: The influence of ancestry, body size, and menopausal status, the breast cancer health disparities study. Cancer Epidemiol. 2015, 39, 1113–1122. [Google Scholar] [CrossRef] [PubMed]
  162. Connor, A.; Baumgartner, R.N.; Kerber, R.A.; O’Brien, E.; Rai, S.N.; Wolff, R.K.; Slattery, M.L.; Giuliano, A.R.; Risendal, B.C.; Byers, T.E.; et al. ADRB2 G–G haplotype associated with breast cancer risk among Hispanic and non-Hispanic white women: interaction with type 2 diabetes and obesity. Cancer Causes Control 2012, 23, 1653–1663. [Google Scholar] [CrossRef] [PubMed]
  163. Connor, A.E.; Baumgartner, R.N.; Baumgartner, K.B.; Pinkston, C.M.; Boone, S.D.; John, E.M.; Torres-Mejía, G.; Hines, L.M.; Giuliano, A.R.; Wolff, R.K.; et al. Associations between ALOX, COX, and CRP polymorphisms and breast cancer among Hispanic and non-Hispanic white women: The breast cancer health disparities study. Mol. Carcinog 2015, 54, 1541–1553. [Google Scholar] [CrossRef] [PubMed]
  164. Pellatt, A.J.; Lundgreen, A.; Wolff, R.K.; Hines, L.; John, E.M.; Slattery, M.L. Energy homeostasis genes and survival after breast cancer diagnosis: The breast cancer health disparities study. Cancer Causes Control 2016, 27, 47–57. [Google Scholar] [CrossRef] [PubMed]
  165. Piranda, D.N.; Festa-Vasconcellos, J.S.; Amaral, L.M.; Bergmann, A.; Vianna-Jorge, R. Polymorphisms in regulatory regions of Cyclooxygenase-2 gene and breast cancer risk in Brazilians: A case-control study. BMC Cancer 2010, 10, 613. [Google Scholar] [CrossRef] [PubMed]
  166. Gómez Flores-Ramos, L.; Escoto-De Dios, A.; Puebla-Pérez, A.M.; Figuera-Villanueva, L.E.; Ramos-Silva, A.; Ramírez-Patiño, R.; Delgado-Saucedo, J.I.; Salas-González, E.; Zúñiga-González, G.M.; Alonzo-Rojo, A.; et al. Association of the tumor necrosis factor α -308G>A polymorphism with breast cancer in Mexican women. Genet. Mol. Res. 2013, 12, 5680–5693. [Google Scholar] [CrossRef] [PubMed]
  167. Tanaka, A.; Sakaguchi, S. Regulatory T cells in cancer immunotherapy. Cell Res. 2017, 27, 109–118. [Google Scholar] [CrossRef] [PubMed]
  168. Ladoire, S.; Arnould, L.; Mignot, G.; Coudert, B.; Rébé, C.; Chalmin, F.; Vincent, J.; Bruchard, M.; Chauffert, B.; Martin, F.; et al. Presence of Foxp3 expression in tumor cells predicts better survival in HER2-overexpressing breast cancer patients treated with neoadjuvant chemotherapy. Breast Cancer Res. Treat. 2011, 125, 65–72. [Google Scholar] [CrossRef] [PubMed]
  169. Ni, J.; Miller, M.; Stojanovic, A.; Garbi, N.; Cerwenka, A. Sustained effector function of IL-12/15/18-preactivated NK cells against established tumors. J. Exp. Med. 2012, 209, 2351–2365. [Google Scholar] [CrossRef] [PubMed]
  170. Jung, M.Y.; Kim, S.H.; Cho, D.; Kim, T.S. Analysis of the expression profiles of cytokines and cytokine-related genes during the progression of breast cancer growth in mice. Oncol. Rep. 2009, 22, 1141–1147. [Google Scholar] [PubMed]
  171. Bai, F.; Smith, M.D.; Chan, H.L.; Pei, X.-H. Germline mutation of Brca1 alters the fate of mammary luminal cells and causes luminal-to-basal mammary tumor transformation. Oncogene 2012. [Google Scholar] [CrossRef] [PubMed]
  172. Giedraitis, V.; He, B.; Huang, W.X.; Hillert, J. Cloning and mutation analysis of the human IL-18 promoter: A possible role of polymorphisms in expression regulation. J. Neuroimmunol. 2001, 112, 146–152. [Google Scholar] [CrossRef]
  173. Pellatt, A.J.; Wolff, R.K.; John, E.M.; Torres-Mejia, G.; Hines, L.M.; Baumgartner, K.B.; Giuliano, A.R.; Lundgreen, A.; Slattery, M.L. SEPP1 influences breast cancer risk among women with greater native American ancestry: The breast cancer health disparities study. PloS ONE 2013, 8, e80554. [Google Scholar] [CrossRef] [PubMed]
  174. Cárdenas-Rodríguez, N.; Lara-Padilla, E.; Bandala, C.; López-Cruz, J.; Uscanga-Carmona, C.; Lucio-Monter, P.F.; Floriano-Sánchez, E. CYP2W1, CYP4F11 and CYP8A1 polymorphisms and interaction of CYP2W1 genotypes with risk factors in Mexican women with breast cancer. Asian Pac. J. Cancer Prev. 2012, 13, 837–846. [Google Scholar] [CrossRef] [PubMed]
  175. Wang, D.; Sadée, W. Searching for polymorphisms that affect gene expression and mRNA processing: example ABCB1 (MDR1). AAPS J. 2006, 8, E515–E520. [Google Scholar] [CrossRef] [PubMed]
  176. Fuchs-Wightman, F.; Giono, L.E.; Fededa, J.P.; de la Mata, M. Target RNAs strike back on microRNAs. Front. Genet. 2018, 9, 435. [Google Scholar] [CrossRef] [PubMed]
  177. Hoffman, J.; Fejerman, L.; Hu, D.; Huntsman, S.; Li, M.; John, E.M.; Torres-Mejia, G.; Kushi, L.; Ding, Y.C.; Weitzel, J.; et al. Identification of novel common breast cancer risk variants at the 6q25 locus among Latinas. Breast Cancer Res. 2019, 21, 3. [Google Scholar] [CrossRef] [PubMed]
  178. Slattery, M.L.; Baumgartner, K.B.; Giuliano, A.R.; Byers, T.; Herrick, J.S.; Wolff, R.K. Replication of five GWAS-identified loci and breast cancer risk among Hispanic and non-Hispanic white women living in the Southwestern United States. Breast Cancer Res. Treat. 2011, 129, 531–539. [Google Scholar] [CrossRef] [PubMed]
  179. Siddiqui, S.; Chattopadhyay, S.; Akhtar, M.S.; Najm, M.Z.; Deo, S.V.S.; Shukla, N.K.; Husain, S.A. A study on genetic variants of fibroblast growth factor receptor 2 (FGFR2) and the risk of breast cancer from North India. PLoS ONE 2014, 9, e110426. [Google Scholar] [CrossRef] [PubMed]
  180. Mazhar, A.; Jamil, F.; Bashir, Q.; Ahmad, M.S.; Masood, M.; Tanvir, I.; Rashid, N.; Waheed, A.; Afzal, M.N.; Tariq, M.A.; et al. Genetic variants in FGFR2 and TNRC9 genes are associated with breast cancer risk in Pakistani women. Mol. Med. Rep. 2016, 14, 3443–3451. [Google Scholar] [CrossRef] [PubMed]
  181. Barnholtz-Sloan, J.S.; Shetty, P.B.; Guan, X.; Nyante, S.J.; Luo, J.; Brennan, D.J.; Millikan, R.C. FGFR2 and other loci identified in genome-wide association studies are associated with breast cancer in African-American and younger women. Carcinogenesis 2010, 31, 1417–1423. [Google Scholar] [CrossRef] [PubMed]
  182. Danková, Z.; Žúbor, P.; Grendár, M.; Kapinová, A.; Kasajová, P.; Kalman, M.; Zelinová, K.; Štanclová, A.; Lasabová, Z.; Danko, J. Cumulative effect of low-penetrant SNPs in FGFR2, TOX3 and MAP3K1 genes on breast cancer risk in Slovak women. Breast 2017, 32, S91. [Google Scholar] [CrossRef]
  183. Yang, Y.; Wang, W.; Liu, G.; Yu, Y.; Liao, M. Association of single nucleotide polymorphism rs3803662 with the risk of breast cancer. Sci. Rep. 2016, 6, 29008. [Google Scholar] [CrossRef] [PubMed]
  184. Shan, J.; Mahfoudh, W.; Dsouza, S.P.; Hassen, E.; Bouaouina, N.; Abdelhak, S.; Benhadjayed, A.; Memmi, H.; Mathew, R.A.; Aigha, I.I.; et al. Genome-Wide Association Studies (GWAS) breast cancer susceptibility loci in Arabs: Susceptibility and prognostic implications in Tunisians. Breast Cancer Res. Treat. 2012, 135, 715–724. [Google Scholar] [CrossRef] [PubMed]
  185. Shan, J.; Dsouza, S.P.; Bakhru, S.; Al-Azwani, E.K.; Ascierto, M.L.; Sastry, K.S.; Bedri, S.; Kizhakayil, D.; Aigha, I.I.; Malek, J.; et al. TNRC9 downregulates BRCA1 expression and promotes breast cancer aggressiveness. Cancer Res. 2013, 73, 2840–2849. [Google Scholar] [CrossRef] [PubMed]
  186. Lei, H.; Deng, C.-X. Fibroblast growth factor receptor 2 signaling in breast cancer. Int. J. Biol. Sci. 2017, 13, 1163–1171. [Google Scholar] [CrossRef] [PubMed]
  187. Liu, C.; Wang, S.; Zhu, S.; Wang, H.; Gu, J.; Gui, Z.; Jing, J.; Hou, X.; Shao, Y. MAP3K1-targeting therapeutic artificial miRNA suppresses the growth and invasion of breast cancer in vivo and in vitro. SpringerPlus 2016, 5, 11. [Google Scholar] [CrossRef] [PubMed]
  188. Fejerman, L.; Hu, D.; Huntsman, S.; John, E.M.; Stern, M.C.; Haiman, C.A.; Pérez-Stable, E.J.; Ziv, E. Genetic ancestry and risk of mortality among U.S. latinas with breast cancer. Cancer Res. 2013, 73, 7243–7253. [Google Scholar] [CrossRef] [PubMed]
  189. Hines, L.M.; Sedjo, R.L.; Byers, T.; John, E.M.; Fejerman, L.; Stern, M.C.; Baumgartner, K.B.; Giuliano, A.R.; Torres-Mejia, G.; Wolff, R.K.; et al. The interaction between genetic ancestry and breast cancer risk factors among Hispanic women: The breast cancer health disparities study. Cancer Epidemiol. Biomarkers Prev. 2017, 26, 692–701. [Google Scholar] [CrossRef] [PubMed]
  190. Torres, D.; Lorenzo Bermejo, J.; Garcia Mesa, K.; Gilbert, M.; Briceño, I.; Pohl-Zeidler, S.; González Silos, R.; Boekstegers, F.; Plass, C.; Hamann, U. Interaction between genetic ancestry and common breast cancer susceptibility variants in Colombian women. Int. J. Cancer 2019. [Google Scholar] [CrossRef] [PubMed]
  191. Fejerman, L.; Romieu, I.; John, E.M.; Lazcano-Ponce, E.; Huntsman, S.; Beckman, K.B.; Pérez-Stable, E.J.; González Burchard, E.; Ziv, E.; Torres-Mejía, G. European ancestry is positively associated with breast cancer risk in Mexican women. Cancer Epidemiol. Biomarkers Prev. 2010, 19, 1074–1082. [Google Scholar] [CrossRef] [PubMed]
  192. Bonilla, C.; Bertoni, B.; Hidalgo, P.C.; Artagaveytia, N.; Ackermann, E.; Barreto, I.; Cancela, P.; Cappetta, M.; Egaña, A.; Figueiro, G.; et al. Breast cancer risk and genetic ancestry: A case-control study in Uruguay. BMC Womens Health 2015, 15, 11. [Google Scholar] [CrossRef] [PubMed]
  193. Fejerman, L.; Chen, G.K.; Eng, C.; Huntsman, S.; Hu, D.; Williams, A.; Pasaniuc, B.; John, E.M.; Via, M.; Gignoux, C.; et al. Admixture mapping identifies a locus on 6q25 associated with breast cancer risk in US Latinas. Hum. Mol. Genet. 2012, 21, 1907–1917. [Google Scholar] [CrossRef] [PubMed]
  194. Chokkalingam, A.P.; Aldrich, M.C.; Bartley, K.; Hsu, L.-I.; Metayer, C.; Barcellos, L.F.; Wiemels, J.L.; Wiencke, J.K.; Buffler, P.A.; Selvin, S. Matching on race and ethnicity in case-control studies as a means of control for population stratification. Epidemiology (Sunnyvale, Calif.) 2011, 1, 101. [Google Scholar] [CrossRef] [PubMed]
  195. Martin, E.R.; Tunc, I.; Liu, Z.; Slifer, S.H.; Beecham, A.H.; Beecham, G.W. Properties of global- and local-ancestry adjustments in genetic association tests in admixed populations. Genet. Epidemiol. 2018, 42, 214–229. [Google Scholar] [CrossRef] [PubMed]
  196. Jara, L.; Morales, S.; Mayo, T.; Gonzalez-Hormazabal, P.; Carrasco, V.; Godoy, R. Mutations in BRCA1, BRCA2 and other breast and ovarian cancer susceptibility genes in Central and South American populations. Biol. Res. 2017, 50, 35. [Google Scholar] [CrossRef] [PubMed]
  197. Chavarri-Guerra, Y.; Blazer, K.R.; Weitzel, J.N. Genetic cancer risk assessment for breast cancer in Latin America. Rev. Investig. Clinica 2017, 69, 94–102. [Google Scholar] [CrossRef]
  198. Cruz-Correa, M.; Pérez-Mayoral, J.; Dutil, J.; Echenique, M.; Mosquera, R.; Rivera-Román, K.; Umpierre, S.; Rodriguez-Quilichini, S.; Gonzalez-Pons, M.; Olivera, M.I.; et al. Clinical cancer genetics disparities among Latinos. J. Genet. Couns 2017, 26, 379–386. [Google Scholar] [CrossRef] [PubMed]
  199. Wacholder, S.; Hartge, P.; Prentice, R.; Garcia-Closas, M.; Feigelson, H.S.; Diver, W.R.; Thun, M.J.; Cox, D.G.; Hankinson, S.E.; Kraft, P.; et al. Performance of common genetic variants in breast-cancer risk models. N. Engl. J. Med. 2010, 362, 986–993. [Google Scholar] [CrossRef] [PubMed]
  200. Mealiffe, M.E.; Stokowski, R.P.; Rhees, B.K.; Prentice, R.L.; Pettinger, M.; Hinds, D.A. Assessment of clinical validity of a breast cancer risk model combining genetic and clinical information. J. Natl. Cancer Inst. 2010, 102, 1618–1627. [Google Scholar] [CrossRef] [PubMed]
  201. Pharoah, P.D.P.; Antoniou, A.C.; Easton, D.F.; Ponder, B.A.J. Polygenes, risk prediction, and targeted prevention of breast cancer. N. Engl. J. Med. 2008, 358, 2796–2803. [Google Scholar] [CrossRef] [PubMed]
  202. Vachon, C.M.; Pankratz, V.S.; Scott, C.G.; Haeberle, L.; Ziv, E.; Jensen, M.R.; Brandt, K.R.; Whaley, D.H.; Olson, J.E.; Heusinger, K.; et al. The contributions of breast density and common genetic variation to breast cancer risk. J. Natl. Cancer Inst. 2015, 107. [Google Scholar] [CrossRef] [PubMed]
  203. Khera, A.; Chaffin, M.; Aragam, K.G.; Haas, M.E.; Roselli, C.; Choi, S.H.; Natarajan, P.; Lander, E.S.; Lubitz, S.A.; Ellinor, P.T.; et al. Genome-wide polygenic scores for common diseases identify individuals with risk equivalent to monogenic mutations. Nat. Genet. 2018, 50, 1219–1224. [Google Scholar] [CrossRef] [PubMed]
  204. Guerrero, S.; López-Cortés, A.; Indacochea, A.; García-Cárdenas, J.M.; Zambrano, A.K.; Cabrera-Andrade, A.; Guevara-Ramírez, P.; González, D.A.; Leone, P.E.; Paz-y-Miño, C. Analysis of racial/ethnic representation in select basic and applied cancer research studies. Sci. Rep. 2018, 8, 13978. [Google Scholar] [CrossRef] [PubMed]
  205. The 1000 Genomes Project Consortium; Auton, A.; Brooks, L.D.; Durbin, R.M.; Garrison, E.P.; Kang, H.M.; Korbel, J.O.; Marchini, J.L.; McCarthy, S.; McVean, G.A.; et al. A global reference for human genetic variation. Nature 2015, 526, 68–74. [Google Scholar]
  206. Wang, S.; Qian, F.; Zheng, Y.; Ogundiran, T.; Ojengbede, O.; Zheng, W.; Blot, W.; Nathanson, K.L.; Hennis, A.; Nemesure, B.; et al. Genetic variants demonstrating flip-flop phenomenon and breast cancer risk prediction among women of African ancestry. Breast Cancer Res. Treat. 2018, 168, 703–712. [Google Scholar] [CrossRef] [PubMed]
  207. Investigators of the US–Latin America Cancer Research Network. Translational cancer research comes of age in Latin America. Sci. Transl. Med. 2015, 7, 319fs50. [Google Scholar] [CrossRef] [PubMed]
  208. Romieu, I.; Biessy, C.; Carayol, M.; His, M.; Torres-Mejía, G.; Ángeles-Llerenas, A.; Sánchez, G.I.; Jaramillo, R.; Navarro, E.; Porras, C.; et al. Reproductive factors and molecular subtypes of breast cancer among premenopausal women in Latin America: the PRECAMA study. Sci. Rep. 2018, 8, 13109. [Google Scholar] [CrossRef] [PubMed]
  209. Magalhães, W.C.S.; Araujo, N.M.; Leal, T.P.; Araujo, G.S.; Viriato, P.J.S.; Kehdy, F.S.; Costa, G.N.; Barreto, M.L.; Horta, B.L.; Lima-Costa, M.F.; Magalhães, W.C.S.; et al. EPIGEN-Brazil initiative resources: A Latin American imputation panel and the Scientific Workflow. Genome Res. 2018, 28, 1090–1095. [Google Scholar] [CrossRef] [PubMed]
  210. Ruiz-Linares, A.; Adhikari, K.; Acuña-Alonzo, V.; Quinto-Sanchez, M.; Jaramillo, C.; Arias, W.; Fuentes, M.; Pizarro, M.; Everardo, P.; Avila, F.; et al. Admixture in Latin America: Geographic structure, phenotypic diversity and self-perception of ancestry based on 7,342 individuals. PLoS Genet. 2014, 10, e1004572. [Google Scholar] [CrossRef] [PubMed]
Table 1. Summary results from breast cancer candidate gene studies in Latin America showing a positive association with cancer risk (p ≤ 0.05).
Table 1. Summary results from breast cancer candidate gene studies in Latin America showing a positive association with cancer risk (p ≤ 0.05).
CountryGeneVariantRisk Genotype vs. Common GenotypeOR (95% CIs)
p-Value
ModelDesignReference
FOXP3rs2232365A>G AA vs. GG1.93 (1.01–3.66)
p = 0 046
Controlled by age117 cases and 300 controlsBanin Hirata et al., 2017 [87]
BrazilMTHFRrs1801133C>TTT vs. (CC + CT)2.53 (1.08–5.93)
p = 0.03
Adjusted for age, alcohol and smoking consumption, and BMI100 cases and 144 controlsZara-Lopes et al., 2016 [88]
CYP1A1rs1048943A>G(AG + GG) vs. AA1.50 (1.14–1.97)
p = 0.004
Adjusted for age and ethnic origin742 cases and 742 controlsOliveira et al., 2015 [89]
IL-18rs1946518C>ACC vs AA2.78 (1.39–5.59)
p = 0.004
Adjusted for age and BMI154 cases and 118 controlsBack et al., 2014 [90]
rs187238G>C(GG + GC) vs. CC3.89 (1.43–10.64)
p = 0.008
GSTM1rs366631C>T−/− vs. (+/− and +/+)2.40 (1.1–5.6)
p =0.04
Not adjusted49 cases and 49 controlsPossuelo et al., 2013 [91]
miR-196a2rs11614913C>T(TC+ TT) vs. CC1.50 (CI not provided)
p = 0.014
Adjusted for age, race, menarche age, menopausal status, smoking habits, and first-degree breast cancer family history388 cases and 388 controlsLinhares et al., 2012 [92]
Chilepre-mir-423rs6505162C>AAA vs. CC1.70 (1.0–2.0)
p = 0.05
In families with at least three BC and/or OC cases440 cases a and 807 controlsMorales at al., 2016 [93]
pre-mir-618rs2682818C>ACA vs. CC1.60 (1.0 − 2.4)
p = 0.04
In families with a single case, diagnosed at ≤ 50 years of age
pre-mir-27ars895819A>GGG vs. AA0.30 (0.1–0.8)
p =  0.01
In families with 2 BC and/or OC cases
PALB2rs152451A>GAG vs. AA2.0 (1.2–3.1)
p < 0.01
In families with at least three BC and/or OC cases436 cases a and 809 controlsLeyton et al., 2015 [94]
rs45551636C>TCT vs. CC3.0 (1.2–6.8)
p < 0.01
TOX3rs3803662C>TTT vs. CC2.38 (1.44–3.90)
p = 0.0003
In families with at least 2 BC and/or OC cases344 cases a and 801 controlsElematore et al., 2014 [95]
2q35rs13387042 G>AAA vs. GG1.99 (1.25–3.14)
p = 0.0015
FGFR2rs2981582C>TCT vs. CC2.00 (1.26–3.23)
p = 0.002
In families with a single case, diagnosed at ≤ 50 years of age351 cases a and 802 controlsJara et al., 2013 [96]
rs2420946C>TTT vs. CC2.05 (1.16–3.63)
p = 0.009
rs1219648A>GGG vs. AA2.06 (1.16–3.66)p = 0.011
MAP3K1rs889312A>CCC vs. AA1.96 (1.13–3.37)
p = 0.016
BARD1rs28997576C>GCG vs.CC3.4 (1.2–10.2)
p = 0.04
In families with at least three BC and/or OC cases322 cases a and 570 controlsGonzalez-Hormazabal et al., 2012 [97]
XRCC3rs861539C>TTT vs. CC3.2 (1.40–1.72)
p = 0.006
In families with at least three BC and/or OC cases
TT vs. CC2.44 (1.34–4.43)
p = 0.003
Not adjusted267 cases a and 500 controlsJara et al., 2010 [98]
ATMrs1801516G>A(GA + AA) vs. GG2.52 (1.33–4.77)
p = 0.008
Not adjusted42 cases a and 200 controlsTapia et al., 2008 [99]
rs1801516G>AGA vs. GG1.74 (0.96–3.16)
p = 0.048
Not adjusted126 cases a and 200 controlsGonzalez-Hormazabal et al., 2008 [100]
IVS24-9delTT(-T) vs. TT1.74 (0.96–3.16)
p = 0.048
IVS38-8T>CTC vs. TT3.09 (1.11–8.59)
p = 0.024
RAD51rs1801320G>C(GC + CC) vs. GG2.17 (1.11–4.24)p = 0.020In patients <50 years at diagnosis143 cases a and 247 controlsJara et al., 2007 [101]
EcuadorAKT1rs3803304C>GGG vs. CC5.20 (1.3–20.9)
p ≤ 0.05
Not adjusted91 cases 185 controlsLopez-Cortes et al., 2018 [102]
MTHFRrs1801133C>TTT vs. CC2.9 (1.2–7.2)
p = 0.025
Not adjusted114 cases and 195 controlsLopez-Cortes et al., 2015 [103]
MexicoRETNrs1862513C>G(CG + GG) vs. CC1.62 (1.025–2.557)
p = 0.03791
Not adjusted100 cases and 308 controlsMuñoz-Palomeque et al., 2018 [104]
CAP1rs35749351G>A(GA + AA) vs. GG2.19 (1.075–4.475)
p = 0.0277
Not adjusted
ABCB1rs1045642C>TCC vs. TT2.91 (1.48–5.74)
p = 0.001
Not adjusted243 cases and 118 controlsJaramillo-Rangel et al., 2018 [105]
CT vs. CC2.27 (1.11–4.67)
p = 0.023
In premenopausal women248 cases and 180 controlsGutierrez-Rubio et al., 2015 [106]
CYP1B1rs1056827G>TTT vs. GG1.21 (0.85–1.72)
p = 0.04
Adjusted for age, years of education, first-degree relative with breast cancer, age at first full-term pregnancy, breastfeeding at first birth, consumption of alcohol and tobacco, and genetic ancestry (Native American, European, and African).952 cases and 998 controls *Garcia-Martinez et al., 2017 [107]
GSTM1rs366631C>TTT vs. (CC + CT)1.30 (1.02–1.8)
p = 0.04
Not adjusted558 cases and 276 controlsSoto-Quintana et al., 2015 [108]
TT vs. (CC + CT)2.19 (1.50–3.21)
p = 0.001
Not adjusted243 cases and 118 controlsJaramillo-Rangel et al., 2015 [109]
MTHFRrs1801133C>TTT vs. CC2.50 (1.6–3.8)
p = 0.0001
Not adjusted497 cases and 339 controlsRamos-Silva et al., 2015 [110]
CBS844ins68(−/+) vs. (−/−)2.2 (1.5–3.3)
p = 0.0001
Not adjusted323 cases and 371 controlsGallegos-Arreola et al., 2014 [111]
ATMIVS24-9delTT(-T) vs. TT3.02 (1.24–7.30)
p = 0.0122
Not adjusted94 cases and 97 controlsCalderón-Zúñiga et al., 2014 [112]
GSTP1rs1695A>GGG vs. AA3.28 (1.44–7.50)
p = 0.005
In premenopausal women150 cases and 382 controlsMartinez-Ramirez et al., 2013 [113]
eNOS4a/b polymorphismab vs. bb2.0 (1.3–3.1)
p = 0.001
Not adjusted429 cases and 281 controlsRamírez-Patiño et al., 2013 [114]
CYP1A1rs1048943A>GGG vs. AA2.77 (1.30–5.89)
p = 0.009
In premenopausal women150 cases and 382 controlsMartinez-Ramirez et al., 2013 [113]
rs4646903T>CCC vs. TT3.38 (1.05–10.87)
p = 0.041
In postmenopausal women91 cases and 94 controlsMoreno-Galvan et al., 2010 [115]
FGFR2rs2981582C>TTT vs. CC1.69 (1.25–2.27)
p = 0.001
Adjusted by design for place of residence, health service institution membership, and 5-year age interval687 cases and 907 controls *Murillo-Zamora et al., 2013 [116]
Puerto RicoRAD23Brs1805329C>TTT vs. (CT + CC)3.14 (1.65–5.97)
p < 0.001
Adjusted for age, civil status, education level, and contraceptive use228 cases and 418 controlsPerez-Mayoral et al., 2013 [117]
CI: confidence interval; BC: breast cancer; BMI: body mass index; OC: ovarian cancer; OR: odds ratio. a Patients were tested for BRCA1/2 mutations. * Population-based controls.
Table 2. Breast cancer genetic epidemiology in women of Latin American origin: Current limitations and possible solutions.
Table 2. Breast cancer genetic epidemiology in women of Latin American origin: Current limitations and possible solutions.
Current LimitationsPossible Solutions
Diverse genetic backgrounds and mutational frequencies among Latin American populations.
  • Foster collaborations between Latin American countries.
  • Account and stratify for ancestry proportions.
Small sample size in comparison with studies including European and European American individuals.
  • Promote access and exchange of information among researches to establish research partnerships within and across countries to generate large consortiums for joint data analysis.
  • Homogenize the design and data processing of studies from different countries to facilitate data sharing and sample pooling.
  • Extension of National Cancer Registries and quality improvement, including biospecimen collection.
Limited access to high-cost technologies for variant discovery in multiple Latin American countries.
  • Promote collaborative relationships with specialized multicenter initiatives to reduce costs and assure quality control.
  • Promote collaborations that allow Latin American countries to access technologies in the United States and other countries (e.g., European countries, Australia, Japan, etc.) with appropriate data sharing protocols that allow analyses to be conducted by scientists in Latin America.
Inequality in access to healthcare by genetic ancestry might limit the representation of highly Indigenous American women in genetic studies.
  • Foster active role of public hospitals in patient accrual.
  • Decentralization of institutions in charge of patient/individual accrual.

Share and Cite

MDPI and ACS Style

Zavala, V.A.; Serrano-Gomez, S.J.; Dutil, J.; Fejerman, L. Genetic Epidemiology of Breast Cancer in Latin America. Genes 2019, 10, 153. https://doi.org/10.3390/genes10020153

AMA Style

Zavala VA, Serrano-Gomez SJ, Dutil J, Fejerman L. Genetic Epidemiology of Breast Cancer in Latin America. Genes. 2019; 10(2):153. https://doi.org/10.3390/genes10020153

Chicago/Turabian Style

Zavala, Valentina A., Silvia J. Serrano-Gomez, Julie Dutil, and Laura Fejerman. 2019. "Genetic Epidemiology of Breast Cancer in Latin America" Genes 10, no. 2: 153. https://doi.org/10.3390/genes10020153

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop