Patterns of copy number alterations in primary breast tumors of South African patients and their impact on functional cellular pathways

  • Authors:
    • Kamil Lupicki
    • Selene Elifio-Esposito
    • Aline S. Fonseca
    • Saulo H. Weber
    • Bruna Sugita
    • Bridget C. Langa
    • Silma R.F. Pereira
    • Dhirendra Govender
    • Eugene Panieri
    • Donavon C. Hiss
    • Sahar Abdul-Rasool
    • Luciane R. Cavalli
  • View Affiliations

  • Published online on: October 10, 2018     https://doi.org/10.3892/ijo.2018.4589
  • Pages: 2745-2757
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Breast cancer is the most common and the leading cause of female mortality among South African (SA) women. Several non‑biological and biological risk factors may be attributed to their observed high mortality rate; however, the molecular profiles associated with their breast tumors are poorly characterized. The present study examined the patterns of genome-wide copy number alterations (CNAs) and their potential impact on functional cellular pathways targeted by cancer driver genes in patients with breast cancer from the Western Cape region of SA. Array-comparative genomic hybridization analysis, performed in 28 cases of invasive breast cancer, revealed a mean number of 8.68±6.18 CNAs per case, affecting primarily the Xp22.3 and 6p21-p25 cytobands (57.14% of the cases), followed by 19p13.3-p13.11 (35.7%), 2p25.3-p24.3, 4p16.3-p15.3, 8q11.1-q24.3 and 16 p13.3-p11.2 (32.14%). Functional enrichment analysis of genes and microRNA targets mapped in these affected cytobands revealed critical cancer-associated pathways, including fatty acid biosynthesis and metabolism, extracellular matrix-receptor interaction, hippo and tumor protein p53 signaling pathways, which are regulated by known cancer genes, including CCND1, CDKN1A, MAPK1, MDM2, TP53 and SMAD2. An inverse correlation was observed among the number of CNAs and tumor size and grade; CNAs on the 4p and 6p cytobands were also inversely correlated with tumor grade. No association was observed in the number of CNAs and/or the affected cytobands and the different ethnic groups of the SA patients, indicating that their tumor genome is affected by CNAs, irrespectively of their genetic descent. Additional genomic tumor profiling in SA and other Sub-Saharan African patients with breast cancer is required to determine the associations of the CNAs observed with prognosis and clinical outcome.

Introduction

The incidence of breast cancer continues to increase globally. According to the most recent Globocan estimates (2012), breast cancer ranks as the fifth highest cause of mortality from cancer overall, and is the most frequent cause of cancer mortalities in women in developing countries (1). In Africa, there is a substantial variation in the estimated breast cancer incidence across different regions, which may be attributed to the exposure to different environmental factors (2). South Africa (SA) presents the highest incidence of breast cancer, affecting primarily young women (≤50 years old), who are diagnosed at a mean age of 10 years younger compared with women in Western countries. However, the mortality rates in SA remain high (~17%) among female populations of all ages, potentially due to the late stage of the disease at diagnosis (1). The high rates of incidence and mortality are due to multiple and complex social, cultural and economic factors that directly impact breast cancer diagnosis and response to treatment. The health care system in SA and other Sub-Saharan countries, in general, face inadequate health personnel, offer poor health insurance programs and limited facilities for cancer screening and treatment, and are compounded by prohibitive costs for breast biopsies and pathological exams (3). Other factors involve lifestyle-associated risks including smoking, alcohol consumption and high fat diets (4). In addition, several studies have demonstrated that in the Sub-Saharan region, including SA, there is an over-representation of breast cancer subtypes that denote poorer prognosis, most notably among pre-menopausal women (5-9). Although the standard classification of breast cancer, based on three surrogate immunohistochemistry (IHC) markers, the estrogen (ER), progesterone (PR) and human epidermal growth factor receptor 2 (HER2) receptors, is not routinely performed in the majority of SA cancer centers, but studies have demonstrated the prevalence of hormonal negative tumor subtypes, including a high rate of triple negative breast cancer (TNBC) and HER2-positive tumor subtypes. These two subtypes are known to present a clinically aggressive phenotype, including resistance to treatment and high rates of disease recurrence (7-9). Although the determinants for the high prevalence of these unfavorable tumor subtypes is unclear, several studies implicate intrinsic genetic differences, such as the presence of founder mutations in breast cancer susceptibility genes, including the BRCA1 gene (10-12) or unknown genomic variants identified by genomic wide association studies (13-16).

The extensive characterization of the molecular profiling in breast cancer with the use of multiomics platforms, have allowed for extraordinary progress in the understanding of the biological factors and their clinical impact on each breast cancer subtype (17,18). In African countries, however, they remain poorly characterized, mainly due to the unavailability and the high costs of such platforms in addition to the poor annotation of patients’ clinical data (19). Among the African countries, SA has the highest number of published genomic research studies (assessed between 2004-2013), however only 6.1% of these studies are associated with cancer (20). To the best of our knowledge, few breast cancer studies have characterized patient genomic profiles and/or specific molecular alterations in their populations (14,21-25). As a consequence, there is limited knowledge on their tumor biology and the corresponding lack of clinically relevant biomarkers that would be particularly beneficial for the prognosis and treatment of patients with breast cancer representative of the diverse racial groups of SA.

The integrity of the genome is crucial for tumor suppression. Genomic instability, one of the hallmarks of cancer, can be evidenced by the presence of DNA copy number changes (26,27). The presence of gains and losses in specific cytobands of chromosomes is a clear evidence of loss of the genomic integrity control cancers (28). The recurrence of these specific alterations, which may affect the function of cancer driver genes mapped in these regions, including oncogenes and tumor suppressors, occurs during tumor progression and may also facilitate this process (29).

In the present study, the main aim was to determine the patterns of the genome-wide copy number alterations (CNAs) and their potential impact on functional cellular pathways targeted by cancer driver genes in patients with breast cancer from the Western Cape region of SA. This study further assessed the correlations between the genomic results with the patients clinicopathological data. In addition, considering the extensive genetic diversity of the SA population, composed of groups originating from Europe, Asia and various African countries, the present study also aimed to determine whether the main copy number patterns observed were associated with patient’s ethnicity.

Materials and methods

Sample population

Formalin-fixed paraffin-embedded (FFPE) breast tumor specimens were obtained from 28 female patients from the archives of the Division of Anatomical Pathology, National Health Laboratory Services, Groote Schuur Hospital (Cape Town, SA). These cases were obtained retrospectively between 2006 and 2010 with ethical approval from the Groote Schuur Hospital (Human Research Ethics Committee protocol no. 454/2010); patient identities were kept anonymous at all stages of the study. The specimens were collected at diagnosis and prior to any cancer treatment. The clinical data pertaining to age, ethnicity, tumor stage and receptor status were obtained for each patient. The mean age of the study population at diagnosis was 48.2±12.9 years, ranging from 22 to 80 years old. All cases were of infiltrating ductal carcinoma and the mean tumor size was 40.6±18.9 mm. Tumor types were classified according to the Tumor Node Metastasis staging system (30) and were of grade 1, 2 and 3 in 16%, 44%, and 40% of the cases, respectively. Stages I, II and III were observed in 7.4%, 22.2%, and 70.4% of the cases, respectively. The cases were evaluated for ER, PR and HER2 receptor status by IHC, as previously described (31,32). ER and HER2 positivity was observed in 57.1% and 53.8% of cases, respectively. PR status was available for only 21.4% of the patients, out of which 33% (2/6) were positive. Three patients were of the TNBC subtype.

Information regarding ethnicity was obtained from self-reported records. The majority of the patients were of the Colored group (57.1%), while 21.4% were White, 17.9% were Black and 3.6% were Asian. Based on the South African census nomenclature and definitions, these categories refer to people with common characteristics in terms of history and descent, which were mostly defined in the Apartheid period (31). The Colored group is composed of highly admixed individuals who present ancestry from Europe and Asia and various indigenous tribes of SA. They form the majority population of the Western Cape region, and genetic studies have demonstrated that they are mainly descendants of the Khoisan tribe (31,32). White refers to people of European descent, mainly from the Netherlands, Germany, France and England. Black refers to individuals of African tribe origin including Zulu, Xhosa and Basotho tribes (31). The samples were not selected based on any of the clinical information and ethnicity. The main inclusion criteria, subsequent to institutional review board local approval, included patients with invasive breast cancer and with known hormonal and ERBB2 status and clinicopathological data. The distribution of the clinicopathological data and ethnic groups in the studied population is presented in Table I.

Table I

Clinicopathological and ethnicity information and corresponding CNAs observed in the South African patients with breast cancer used in the present study.

Table I

Clinicopathological and ethnicity information and corresponding CNAs observed in the South African patients with breast cancer used in the present study.

Patient no.AgeEthnicityTu.SizeaTu.GradeTu.StageERPRHER2CNA number2p25.34p16.3-p156p21-p25 8q1116p1319p13Xp22
156White402IIPOSNAPOS14No CNANo CNANo CNA GainGainNo CNANo CNA
280Colored1102IIIPOSNANEG3No CNANo CNANo CNA No CNANo CNANo CNANo CNA
358Black332IIIPOSNAPOS9LossLossLoss No CNANo CNANo CNAGain
444Colored403IINEGNAPOS14No CNANo CNANo CNA No CNANo CNANo CNANo CNA
538Black503IIINEGNEGNEG2No CNANo CNANo CNA No CNANo CNANo CNANo CNA
657Colored192INEGNAPOS12No CNANo CNAGain GainNo CNAGainGain
757Colored253IIINEGNAPOS5No CNANo CNALoss GainNo CNAampl.No CNA
860Colored551IIIPOSNANEG5DeletionLossDeletion No CNANo CNANo CNAGain
959Black50NAIIIPOSNANEG10No CNANo CNAGain No CNAGainGainGain
1055White652IIIPOSNAPOS5DeletionLossDeletion No CNANo CNANo CNAGain
1136White302IIIPOSNANEG25No CNANo CNAampl. Gainampl.GainNo CNA
1243Asian353IINEGNEGNEG4LossNo CNANo CNA No CNANo CNANo CNANo CNA
1344White582IIIPOSNANEG5LossNo CNANo CNA GainNo CNANo CNANo CNA
1446Colored20NANANANANA15No CNANo CNAGain, loss GainGainGainGain
1545Black352IIIPOSNANEG10No CNANo CNAampl., loss No CNANo CNAGainNo CNA
1635Colored30NAIIIPOSNAPOS3No CNANo CNANo CNA GainNo CNANo CNANo CNA
1740Black453IIINEGNANEG7No CNANo CNANo CNA GainNo CNANo CNAGain
1829Colored451IIIPOSPOSPOS15LossLossampl., loss No CNAGainGainGain
1978Colored552IIIPOSNANEG8No CNANo CNAampl. No CNAampl.GainGain
2055Colored603IIINEGNAPOS1No CNANo CNANo CNA No CNANo CNANo CNANo CNA
2139Colored252IIINEGNEGPOS15LossLossampl., loss No CNAGainGainGain
2238White181IPOSPOSNEG12DeletionLossDeletion No CNANo CNAGainampl.
2351White302IIPOSNANEG2No CNANo CNANo CNA No CNANo CNANo CNAGain
2449Colored453IIINEGNEGNEG3No CNALossNo CNA No CNANo CNANo CNAGain
2548Colored253IIPOSNAPOS1No CNANo CNANo CNA No CNANo CNANo CNANo CNA
2636Colored243IIINEGNAPOS8DeletionLossDeletion No CNANo CNANo CNAampl.
2722Colored403IIIPOSNAPOS8No CNANo CNAampl. No CNAampl.ampl.Gain
2853Colored301IIPOSNAPOS22No CNALossGain GainGainno CNAGain

a mm. NA, not available; POS, positive; NEG, negative; ampl., amplification; CNA, copy number alteration; Tu, tumor; ER, estrogen receptor; PR, progesterone receptor; HER2, human epidermal growth factor receptor 2.

Array-comparative genomic hybridization (array-CGH) analysis

DNA copy number analysis was performed using an oligonucleotide array-CGH platform (SurePrint G3 Human CGH Microarray 8×60K; Agilent Technologies Inc., Santa Clara, CA, USA), according to the protocol for FFPE samples that were established as previously described (33,34). Prior to DNA isolation, 5-μm sections of FFPE tissue from each case were evaluated by a pathologist from the Division of Anatomical Pathology, University of Cape Town (National Health Laboratory Service, Groote Schuur Hospital, Cape Town), to confirm the tumor histopathology and the presence of breast epithelial cancer cells. These cells were identified and microdissected in tissue sections that were 5 μm thick from consecutive unstained slides to ensure the array-CGH analysis of a pure tumor cell population. DNA was isolated using the standard phenol-chlorophorm method as was previously described (32). Normal (reference) DNA was prepared from the peripheral blood of a pool of multiple healthy female donors, as previously described (35,36). Equal amounts of tumor and reference genomic DNA (300-500 ng) were digested and enzymatically labeled using the SureTag Complete DNA Labeling kit (Agilent Technologies, Inc.) and hybridized to the arrays, according to the manufacture’s protocols. The array data was analyzed using the Feature Extraction v.10.10 and Agilent Cytogenomics v.3.0 software (Agilent Technologies Inc.), (ADM) after aberration detection method-2 algorithm, a threshold of 6.0 and an aberration filter with a minimum number of >3 probes. Copy number gains and losses were defined as previously described (34): Minimum mean absolute log2 ratio [intensity of the Cy5 dye (reference DNA)/intensity of the Cy3 dye (test DNA) value] of ≥0.25 and ≤-0.25, respectively, as per the array-CGH analytics analysis. Amplifications and deletions were determined using values of log2≥2 and log2≤-2, respectively.

Functional enriched pathways

The identification of the genes and microRNAs (miRNAs) mapped in the cytobands that presented CNAs was obtained from the Agilent Cytogenomics v.7.0 (Agilent Technologies, Inc.) interval base reports (based on the analysis parameters described above). DIANA-miRPath v.3 (37) was used to perform the pathway enrichment analysis, based on the Kyoto Encyclopedia of Genes and Genomes database (37). miRNA targets were identified using TarBase v.7.0 (38). Only miRNA/mRNA targets that presented a miRNA Target Gene (miTG) score >0.7 based on the microT-CDS (39) interactions were included.

Correlation of CNAs with clinicopathological data and ethnicity

Spearman bivariate correlation analysis was performed to determine the association among the number of CNAs and the most frequent affected cytobands with patient clinicopathological data (age <50 or ≥50 years, mean tumor size + and - standard deviation, tumor grades 1-3, tumor stages I-III, ER and HER2 expression status) and ethnicity. Data is presented as Spearman’s correlation coefficient (r), and P<0.05 was considered to indicate a statistically significant difference. Statistical analysis was performed using SPSS Statistics 20 (IBM Corp., Armonk, NY, USA).

Results

DNA copy number changes

Array-CGH analysis was performed for all cases. The CNAs ranged from 1 to 25, with a mean number of 8.68±6.18 CNAs per case. Twelve cases presented CNAs above or equal the mean and 16 cases below. The combined genomic profile of the cases analyzed are presented in Fig. 1. The most frequent affected chromosome cytobands were Xp22.3 and 6p21-p25 (57.14% of the cases), 19p13.3-p13.11 (35.7%), followed by 2p25.3-p24.3, 4p16.3-p15.3, 8q11.1-q24.3 and 16 p13.3-p11.2 (32.14%) (Table I). Chromosomes 2p and 4p were mostly affected by losses and deletions of copy number, whereas chromosomes 8q, 16p and Xp by gains and amplifications, and chromosome 6p by gains and amplifications (mainly 6p22.2-p21) and losses/deletions (mainly 6p25-p24).

A higher number of CNAs (CNAs ≥8.68) were significantly correlated with the cases that present with CNAs in the 6p (r=0.458; P<0.05), 16p (r=0.486; P<0.01) and 19p (r=0.486; P<0.01) cytobands. CNAs on 2p were significantly correlated with CNAs on 4p (r=0.580; P<0.001); CNAs on 4p were significantly correlated with CNAs on 6p (r=0.442; P<0.05) and Xp (r=0.596; P<0.001); CNAs on 6p were significantly correlated with CNAs on 19p (r=0.697; P<0.001) and Xp (r=0.563; P<0.01) and CNAs on 16p were significantly correlated with CNAs on 19p (r=0.542; P<0.01).

The identification of the genes mapped in these cytobands, using the Agilent Cytogenomics interval base reports, revealed a total number of 2,719 genes and 93 miRNAs (Table II). To investigate the function of these specific genes and miRNAs that may be affected by the presence of CNAs in these cytobands, pathway enrichment analysis (DIANA-miRPath v.3) was used. Among the top 15 pathways identified, based on P-value, were pathways associated with prion diseases, fatty acid biosynthesis and metabolism, adherens junction, extracellular matrix (ECM)-receptor interaction, hippo and tumor protein p53 (TP53) signaling pathways (Fig. 2; Table III). Among these top pathways, the ones that were affected by the largest number of miRNAs were the adherens junctions (14 miRNAs), fatty acid metabolism and the viral carcinogenesis pathway (12 miRNAs each) (Table III).

Table II

Main cytobands affected by copy number alterations and corresponding genes and miRNAs observed in the South African patients with breast cancer.

Table II

Main cytobands affected by copy number alterations and corresponding genes and miRNAs observed in the South African patients with breast cancer.

ChrCytobandStartStopSize (kb)Number of genesaNumber of miRNAsmiRNA names
chr 2p25.3-p24.3424441065513110,61460-
chr 4p16.3-p15.2715522059330720,521226miR-218-1, miR-3138, miR-4274, miR-548i2, miR-572, miR-943
chr 6p25.3-p21.12553503328481833,036479miR-1266, miR-1275, miR-219-1, miR-3143, miR-3691, miR-3925, miR-548a1, miR-586, miR-879
chr 8p23.3-q24.34768133514623096798,5445420miR-1204, miR-1205, miR-1206, miR-1207, miR-1208, miR-1234, miR-142-2, miR-2052, miR-2053, miR-30b, miR-30d, miR-3150, miR-3150b, miR-3151, miR-3610, miR-599, miR-661, miR-875, miR-937, miR-939
chr 16p13.3-p11.21062713422630034,1249422miR-1225, miR-193b, miR-3176, miR-3177, miR-3178, miR-3179-1, miR-3179, miR-3179-3, miR-3180-1,-2,-3,-4, miR-3180, miR-365, miR-3670, miR-3766, miR-3680, miR-484, miR-548aa2, miR-66, miR-762, miR-94
chr 19p13.3-q13.436510281929053618,63957021miR-1181, miR-1227, miR-1238, miR-1470, miR-181c, miR-181d, miR-1909, miR-199A1, miR-23a, miR-24-2, miR-27a, miR-3187, miR-3188, miR-3189, miR-3940, miR-4321, miR-4322, miR-637, miR-638, miR-639, miR-7
chr Xp22.33-p21.1610913620475236,1438615miR-188, miR-221, miR-22, miR-23c, miR-362, miR-3690, miR-500a, miR-500b, miR-501, miR-502, miR-532, miR-651, miR-660, miR-98, miR-let7f

a Number of genes does not reflect the total number of genes located in the entire chromosome region, but only the genes affected by copy number alterations as presented in the array-comparative genomic hybridization interval base reports (Cytogenomics v.3.0, Agilent Inc.). miRNA/miR, microRNA; Chr, chromosome number.

Table III

Top 15 KEGG pathways identified by miRNAs located in the main cytobands affected by CNAs.

Table III

Top 15 KEGG pathways identified by miRNAs located in the main cytobands affected by CNAs.

KEGG pathwayaP-valueNumber of genesNumber of miRNAsmiRNA names
Prion diseases <1.00×10-33134miR-193b-3p, miR-221-5p, miR-365a-3p, miR-502-3p
Fatty acid biosynthesis <1.00×10-3358miR-193b-3p, miR-199a-3p, miR-30b-5p, miR-30d-5p, miR-3176, miR-484, miR-877-3p, miR-940
Fatty acid metabolism <1.00×10-332212miR-193b-3p, miR-199a-3p, miR-27a-5p, miR-30b-5p, miR-30d-5p, miR-3176, miR-484, miR-500a-5p, miR-548i, miR-660-5p, miR-877-3p, miR-940
Adherens junction 1.72×10-144714miR-142-3p, miR-188-5p, miR-193b-3p, miR-1207-5p, miR-27a-5p, miR-221-5p, miR-484, miR-502-3p, miR-548×-3p, miR-638, miR-661, miR-877-3p, miR-940, miR-99b-3p
Viral carcinogenesis 1.62×10-119812miR-142-3p, miR-188-5p, miR-193b-3p, miR-23c, miR-24-2-5p, miR-30d-5p, miR-364a-3p, miR-532-5p, miR-548×-3p, miR-572, miR-877-3p, miR-940
Proteoglycans in cancer 1.14×10-8907miR-142-3p, miR-193b-3p, miR-199a-3p, miR-27a-5p, miR-3176, miR-484, miR- 877-3p
Extracellular matrix-receptor interaction 4.37×10-8308miR-188-5p, miR-23a-5p, miR-27a-5p, miR-221-5p, miR-484, miR-532-5p, miR-638, miR-875-3p
Lysine degradation 1.56×10-6308miR-142-3p, miR-193b-3p, miR-30b-5p, miR-30d-5p, miR-3176, miR-484, miR-500a-5p, miR-660-5p
Hippo signaling pathway 1.94×10-5709miR-193b-3p, miR-27a-5p, miR-221-5p, miR-30d-5p, miR-362-3p, miR-484, miR548×-3p, miR-638, miR-99b-3p
Cell cycle 7.53×10-5618miR-193b-3p, miR-30d-5p, miR-484, miR-501-3p, miR-532-5p, miR-572, miR-875-3p, miR-99b-3p
Glioma 1.07×10-5296miR-193b-3p, miR-24-2-5p, miR-27a-5p, miR-362-3p, miR-365a-3p, miR-572
Protein processing in endoplasmic reticulum 1.77×10-2798miR-193b-3p, miR-1909-3p, miR-23c, miR-221-5p, miR-30b-5p, miR-30d-5p, miR-3179, miR-877-3p
Steroid biosynthesis 1.05×10-266miR-199a-3p, miR-23a-5p, miR-221-5p, miR-365a-3p, miR-484, miR-532-5p
p53 signaling pathway 2.80×10-2347miR-193b-3p, miR-30b-5p, miR-30d-5p, miR-362-3p, miR-365a-3p, miR-660-5p, miR-940
Pathways in cancer 4.25×10-21133miR-193b-3p, miR-27a-5p, miR-30d-5p

a Pathways union analysis (DIANA miRPath v.3.0). KEGG, Kyoto Encylopedia of Genes and Genomes.

Using miRNA target prediction analysis (using TarBase v.7.0), 512 targets predicted to be regulated by these miRNAs were identified (miTG score >0.7- microT-CDS interactions). A number of common miRNA targets were observed within these 15 pathways, including the cancer driver genes CCND1 in 46.7% of the pathways, CDKN1A, MAPK1 and MDM2 in 40%, SMAD2 in 33.3% and CCND2, IGF1R, KRAS, SMAD3 and TP53 genes in 26.7% (data not shown).

The integration of these miRNAs gene targets with the 2,719 genes that were mapped in the cytobands with CNAs, as generated by the Cytogenomic interval base reports (Table III), revealed a number of 65 common genes (Fig. 3). From these common genes, that may be potentially affected by copy number and miRNA expression regulation, 35.4% were mapped at the 6p25.3-p21.1 cytoband, 18.5% at 8q11.1-q24.3, 17% at 16p13.3-p11.2 and 19p13.3-p13.11, 7.7% at Xp22.33-p21.1, 3% at 4p16.3-p15.2 and 1.5% at 2p25.3-p24.3. The majority of these 65 common genes were involved in viral carcinogenesis (33.8%) and cell cycle pathways (16.9%) (Table IV). Among these genes are known cancer driver genes, including CDKN1A, MAPKs, MYC and VEGFA.

Table IV

Common genes targeted by microRNAs and copy number alterations in the main affected cytobands and their corresponding involvement in signaling pathways.

Table IV

Common genes targeted by microRNAs and copy number alterations in the main affected cytobands and their corresponding involvement in signaling pathways.

Genes nameChromosomeKyoto Encylopedia of Genes and Genomes Pathway
YWHAQ2p25.3-p24.3Viral carcinogenesis; Hippo Signaling; Cell Cycle
WFS14p16.3-p15.2Protein processing RE
WHSC14p16.3-p15.2Lysine degradation
CCND36p25.3-p21.1TP53 signaling pathway; Cell Cycle; Hippo signaling
CDKN1A6p25.3-p21.1Proteoglycans in cancer; Viral Carcinogenesis; Gliomas; Cell cycle; TP53 signaling pathway
E2F36p25.3-p21.1Gliomas; Pathways in Cancer; Cell cycle
HIST1H2BD6p25.3-p21.1Viral carcinogenesis
HIST1H2BH6p25.3-p21.1Viral carcinogenesis
HIST1H4C6p25.3-p21.1Viral carcinogenesis
HIST1H4D6p25.3-p21.1Viral carcinogenesis
HLA-A6p25.3-p21.1Viral carcinogenesis
HLA-B6p25.3-p21.1Viral carcinogenesis
HLA-C6p25.3-p21.1Viral carcinogenesis
HSP90AB16p25.3-p21.1Pathways in Cancer; Protein processing RE
HSPA1A6p25.3-p21.1Protein processing RE; Prion disease
HSPA1B6p25.3-p21.1Protein processing RE
HSPA1L6p25.3-p21.1Protein processing RE
ITPR36p25.3-p21.1Proteoglycans in cancer
MAPK136p25.3-p21.1Proteoglycans in cancer
MRPS18B6p25.3-p21.1Viral carcinogenesis
PPARD6p25.3-p21.1Pathways in cancer
PPT26p25.3-p21.1Fatty Acid Metabolism
SRF6p25.3-p21.1Viral carcinogenesis
SSR16p25.3-p21.1Protein processing RE
TEAD36p25.3-p21.1Hippo signaling
VEGFA6p25.3-p21.1Pathways in Cancer; Proteoglycans in cancer
CCNE28q11.1-q24.3Viral carcinogenesis; Cell cycle; TP53 signaling pathway; Pathways in cancer
FZD68q11.1-q24.3Proteoglycans in cancer; Hippo Signaling
LYN8q11.1-q24.3Viral carcinogenesis
MCM48q11.1-q24.3Cell cycle
MYC8q11.1-q24.3Hippo Signaling; Pathways in cancer; Cell cycle
PRKDC8q11.1-q24.3Cell cycle
PTK28q11.1-q24.3Proteoglycans in cancer; Pathways in cancer
RAD218q11.1-q24.3Cell cycle
RRM2B8q11.1-q24.3TP53 signaling pathway;
SCRIB8q11.1-q24.3Viral carcinogenesis; Hippo Signaling
SQLE8q11.1-q24.3Steroid Biosynthesis
YWHAZ8q11.1-q24.3Viral carcinogenesis; Hippo Signaling
ADCY916p13.3-p11.2Pathways in cancer
AXIN116p13.3-p11.2Hippo signaling
CREBBP16p13.3-p11.2Adherens in junction; Viral carcinogenesis
DNAJA316p13.3-p11.2Viral carcinogenesis
MAPK316p13.3-p11.2Prion disease; Adherens in junction; Proteoglycans in cancer; Gliomas; Pathways in cancer
PKMYT116p13.3-p11.2Cell cycle
PLK116p13.3-p11.2Cell cycle
PRKCB16p13.3-p11.2Proteoglycans in cancer; Gliomas
SETD1A16p13.3-p11.2Lysine degradation
TFAP416p13.3-p11.2Proteoglycans in cancer
USP716p13.3-p11.2Viral carcinogenesis
DNAJB119p13.3-p13.11Protein processing RE
DOT1L19p13.3-p13.11Lysine degradation
FZR119p13.3-p13.11Cell cycle
INSR19p13.3-p13.11Adherens in junction
JAK319p13.3-p13.11Viral carcinogenesis
MAP2K219p13.3-p13.11Gliomas
MAP2K719p13.3-p13.11Protein processing RE
PIK3R219p13.3-p13.11Proteoglycans in cancer; Viral Carcinogenesis
PRKACA19p13.3-p13.11Prion Disease; Viral Carcinogenesis
PRKCSH19p13.3-p13.11Protein processing RE
TECR19p13.3-p13.11Fatty acid metabolism
DDX3XXp22.33-p21.1Viral carcinogenesis
MBTPS2Xp22.33-p21.1Protein processing RE
PRKXXp22.33-p21.1Prion disease
SMC1AXp22.33-p21.1Viral carcinogenesis; Hippo Signaling
UBQLN2Xp22.33-p21.1Protein processing RE

[i] RE, reticulum endoplasmic; TP53, tumor protein p53.

Correlation of CNAs with clinicopathological parameters and ethnicity

Prior to the association between the clinicopathological data and ethnicity (Table I) with the copy number data, a significant correlation between tumor size and tumor stage (r=0.532; P<0.01) was observed. Inverse correlations were observed between ER status and tumor grade (ER positivity and lower tumor grade r=-0.664; P<0.001) and HER2 status and tumor size (r=-0.408; P<0.05). No correlation was observed among the other clinicopathological parameters and/ or the diverse ethnic groups.

For the association with the number of CNAs, the cases were divided into two groups; one with the number of CNAs higher and one lower than the mean number of CNAs observed among the cases (≥8.68 and <8.68, respectively). Unpredictably, the cases in the group with a lower number of CNAs were significantly associated with higher tumor size (r=-0.401; P<0.05) and tumor grade (r=-0.516; P<0.01). The association of the most frequent cytobands affected with the clinicopathological parameters above, revealed correlation of CNAs on 4p (r=-0.452; P<0.05) and 6p (r=-0.491; P<0.05) with tumor grade. No significant association was observed among CNAs and/or specific cytobands with any ethnic group.

Discussion

Genome-wide molecular signatures are well-defined and established in breast cancer, characterizing the distinct intrinsic subtypes and their impact on prognosis and clinical outcome (18,19). However, there are limited genomic signatures characterizing breast cancer based on the ancestry of a patient, particularly associated with their tumor somatic profiles. In patients of African descent, the majority of the studies available are based on genetic variants, including single nucleotide polymorphism and copy number variation (CNV) (13,15,16). While they are relevant in determining cancer risk and susceptibility in specific populations, they fail to characterize the tumor genomic signatures and corresponding imbalances in gene dosages and mutations that are prevalent and may affect cancer genes.

CNAs in the tumor genome may result in the gain/amplification of oncogenes and/or loss/deletion of tumor suppressor genes and are major drivers of tumor development (27,40). A genome-wide analysis of CNAs conducted in different types of cancer revealed that a specific tumor type presents 17% of regions with amplifications and 16% of deletions, compared with <0.5% in normal samples (40). The patterns of these somatic tumor alterations, as CNVs, may also vary within ethnic groups (35,41). The majority of the studies assessing these alterations, in addition to their gene and epigenetic expression profiles, are in White European and African American patients with breast cancer (35,40-45). Few studies have been performed in patients from Sub-Saharan countries (24,25). Thus, the consequences of CNAs due to the genetic instability of their tumor types remain largely unknown.

In the present study, genome-wide copy number profiling in patients with breast cancer patients from the Western Cape region of SA was performed. Array-CGH analysis revealed recurrent CNAs in the cases analyzed, with the most frequent affecting the cytobands 2p25.3-p24.3, 4p16.3-p15.3, 6p25.3-p22.3, 6p22.3-p.21.1, 8q11.1-q24.3, 16p13.3-p11.2, 19p13.3-p13.11 and Xp22.3. A number of these CNAs, including gains on 8q and 16p, are commonly reported in breast cancer, and are not necessarily associated with specific tumor subtypes and/or ethnicity (18). A number of reports, however, have demonstrated that patients with TNBC and other BRCA1-associated tumor types, which are frequently identified in women of African descent (46,47), present a significantly higher frequency of focal amplifications on 8q24, where C-MYC is located (48). This amplification was, however, not a distinguishing characteristic associated with ancestry, as it was observed both in patients of European and African-American descent (18).

Interestingly, losses on 2p, 4p and gains on 6p, 8q, 16p and 19p were previously reported in the array-CGH analysis of a set of 28 breast cancer cases from patients from Sub-Saharan Africa (24). Among these alterations, gains on 6p presented a significant higher level of copy number increase when compared to American patients with breast cancer, previously analyzed (49). In a previous study in African-American (AA) and Non-Hispanic White patients (NHW) patients with breast cancer (35), it was observed that in the AA group there was a higher frequency of copy number gains, affecting 6p and 8q, when compared with the NHW group. However, in these cases, a significant higher level of genome-wide CNAs was observed in the AA group, which may justify the observed increased in the copy number in these chromosome regions. In fact, CNAs were the only significantly different variable between AA and NHW patients with TNBC tumor subtypes (P<0.01) when clinicopathological parameters were taken into account (35).

A direct correlation was observed among the total number of CNAs and the main cytobands observed; a higher number of CNAs were significantly correlated with the cases that presented with CNAs in the 6p, 16p and 19p cytobands. Direct correlations were observed among CNAs on 2p, 4p, 6p, 16p, 16p and Xp. No inverse correlations among the main cytobands affected by CNAs were observed.

The mean number of CNAs and the specific cytobands affected were correlated with clinicopathological parameters and the diverse SA ethnic groups. Surprisingly, a negative correlation was observed between the mean number of CNAs and tumor size and grade. CNAs on the 4p, 6p and Xp cytobands were also negatively correlated with tumor grade. In general, the increase in genomic instability is associated with poor prognostic parameters (50). However, a number of observations have demonstrated that there are clonal subpopulations of tumor types, including tumor types of epithelial origin, with high levels of somatic CNAs that are associated with a potentially better prognosis (and overall survival time) compared with tumor types with intermediate levels of CNAs (50-53). This interesting observation reflects the well-known intra-tumor heterogeneity marked by the presence of coexisting clones with diverse genomic compositions (52,53). This may explain the unexpected results of the present study, as it was observed that there were higher levels of CNAs in the aforementioned cytobands in tumor types with lower tumor grade and smaller size.

Alterations on 8p were significantly correlated with PR status; however, this correlation may be spurious, considering that only 6 cases presented PR status information. CNAs on chromosome 21p were the only ones associated with ethnicity; no other significant differences were observed between the mean number of CNAs and/or the other most commonly affected cytobands (2p, 4p, 6p, 8q, 16p, 19p and Xp) and ethnicity, indicating that these alterations occur in SA patients irrespective of their genetic descent. Furthermore, multivariate analyses revealed that no clinical or pathological variable, including age, tumor size, grade and stage, and ER, PR and ERBB2/HER2 status were significantly correlated with ethnicity of the patients. However, the comparison of the mean number of CNAs of these SA patients to the mean number of CNAs present in the AA and NHW patients of our previous study (35) revealed significant differences between the Black SA and AA patients (P<0.05), but interestingly no differences between the White SA and NHW patients (P>0.05), which may indicate a common European ancestry. However, when the Colored SA group of patients were compared with the AA and NHW groups, significant differences were observed (P<0.0001 and P<0.001, respectively), which may be resultant of the large variation in ethnicity in this particular group of patients (from Europe and Asia and various tribes of SA).

Finally, by performing functional enrichment analysis of genes and miRNA targets mapped in the cytobands most affected by CNAs, it was observed that they affected a number of critical cancer-associated pathways, including fatty acid biosynthesis and metabolism, adherens junctions, ECM-receptor interaction, hippo and TP53 signaling pathways. These pathways include a number of cancer driver genes associated with breast cancer (54), including CCND1, CDKN1A, MAPK1, MDM2, TP53 and SMAD2. A number of the pathways observed in the present study were also identified in the AA patients with breast cancer with TNBC of a previous study (35), in which a direct integration of copy number and miRNA data conducted on the same samples was performed. Remarkably, CNAs are one of the mechanisms that may result in miRNA dysregulation (55-57). In fact, 15/26 miRNAs of the miRNAs that were identified in the AA patients (of the TNBC subtype) (35) were mapped in cytobands that are frequently affected by recurrent CNAs in breast cancer, including the 6p25.3-p21.1, 8q11.1-q24.3, 16p13.3-p11.2 and 19p13.3-p13.11 similar to the present study.

In conclusion, the present study presents the pattern of copy number changes in the genome of SA patients with breast cancer and demonstrates their potential impact on critical cancer pathways associated with their tumor genomes. Additional genomic tumor profiling in SA and other Sub-Saharan patients with breast cancer in independent and larger sample size cohorts are required in order to determine the association of these alterations with their prognosis and clinical outcome, including response to therapy.

Funding

The present study was supported by The Cancer Association of South Africa (CANSA) (to Professor Sahar Abdul-Rasool and Dr Bridget C. Langa). Scholarships to Dr Bruna Sugita and Professor Silma R.F. Pereira were provided by Coordenação de Aperfeiçoamento de Pessoal de Nível Superior, (CAPES) and the scholarship to Dr Aline S. Fonseca was provided by Conselho Nacional de Desenvolvimento Científico e Tecnológico (CNPq). Additionally, the Genomics and Epigenomics Shared Resource of the Lombardi Comprehensive Cancer Center partially supported the present study (grant no. P30-CA051008).

Availability of data and materials

The datasets used and/or analyzed during the current study are available from the corresponding author on reasonable request.

Authors’ contributions

KL performed the experiments. SEE performed data analysis and wrote the manuscript. ASF assisted in the data analysis. SHW verified the statistical analysis. BS assisted in the data analysis. BCL contributed to the initial design of the study. SRFP contributed to the initial design of the study. DG performed the pathological analysis of the cases. EP provided the specimens and clinical data. DH and SAR contributed to scientific discussions and revised the final copy of the manuscript for intellectual content. LRC designed the study and conducted the final editing of the manuscript. All authors read and approved the manuscript.

Ethics approval and consent to participate

The present study was approved by the Human Research Ethics Committee (HREC 454/2010) of the Groote Schuur Hospital (Cape Town, SA). The analysis of the human samples used in the present study was performed following the international and national regulations in accordance with the Declaration of Helsinki.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

The authors would like to thank the personnel from the Division of Anatomical Pathology, National Health Laboratory Services at the Groote Schuur Hospital (Cape Town, SA) for performing the tissue sections of the cases and the Genomics and Epigenomics Shared Resource of the Lombardi Comprehensive Cancer Center for performing the washes and scanning of the array slides.

Referenes

1 

Ferlay J, Soerjomataram I, Dikshit R, Eser S, Mathers C, Rebelo M, Parkin DM, Forman D and Bray F: Cancer incidence and mortality worldwide: Sources, methods and major patterns in GLOBOCAN 2012. Int J Cancer. 136:E359–E386. 2015. View Article : Google Scholar

2 

Brinton LA, Figueroa JD, Awuah B, Yarney J, Wiafe S, Wood SN, Ansong D, Nyarko K, Wiafe-Addai B and Clegg-Lamptey JN: Breast cancer in Sub-Saharan Africa: Opportunities for prevention. Breast Cancer Res Treat. 144:467–478. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Tetteh DA and Faulkner SL: Sociocultural factors and breast cancer in sub-Saharan Africa: Implications for diagnosis and management. Wom Health Lond. 12:147–156. 2016. View Article : Google Scholar

4 

Wu S, Powers S, Zhu W and Hannun YA: Substantial contribution of extrinsic risk factors to cancer development. Nature. 529:43–47. 2016. View Article : Google Scholar :

5 

Bird PA, Hill AG and Houssami N: Poor hormone receptor expression in East African breast cancer: Evidence of a biologically different disease? Ann Surg Oncol. 15:1983–1988. 2008. View Article : Google Scholar : PubMed/NCBI

6 

Huo D, Ikpatt F, Khramtsov A, Dangou JM, Nanda R, Dignam J, Zhang B, Grushko T, Zhang C, Oluwasola O, et al: Population differences in breast cancer: Survey in indigenous African women reveals over-representation of triple-negative breast cancer. J Clin Oncol. 27:4515–4521. 2009. View Article : Google Scholar : PubMed/NCBI

7 

Ly M, Antoine M, André F, Callard P, Bernaudin JF and Diallo DA: Breast cancer in Sub-Saharan African women: Review. Bull Cancer. 98:797–806. 2011.In French. PubMed/NCBI

8 

Ly M, Antoine M, Dembélé AK, Levy P, Rodenas A, Touré BA, Badiaga Y, Dembélé BK, Bagayogo DC, Diallo YL, et al: High incidence of triple-negative tumors in sub-saharan Africa: A prospective study of breast cancer characteristics and risk factors in Malian women seen in a Bamako university hospital. Oncology. 83:257–263. 2012. View Article : Google Scholar : PubMed/NCBI

9 

Galukande M, Wabinga H, Mirembe F, Karamagi C and Asea A: Molecular breast cancer subtypes prevalence in an indigenous Sub Saharan African population. Pan Afr Med J. 17:2492014. View Article : Google Scholar : PubMed/NCBI

10 

Reeves MD, Yawitch TM, van der Merwe NC, van den Berg HJ, Dreyer G and van Rensburg EJ: BRCA1 mutations in South African breast and/or ovarian cancer families: Evidence of a novel founder mutation in Afrikaner families. Int J Cancer. 110:677–682. 2004. View Article : Google Scholar : PubMed/NCBI

11 

van der Merwe NC, Hamel N, Schneider SR, Apffelstaedt JP, Wijnen JT and Foulkes WD: A founder BRCA2 mutation in non-Afrikaner breast cancer patients of the Western Cape of South Africa. Clin Genet. 81:179–184. 2012. View Article : Google Scholar

12 

Fackenthal JD, Zhang J, Zhang B, Zheng Y, Hagos F, Burrill DR, Niu Q, Huo D, Sveen WE, Ogundiran T, et al: High prevalence of BRCA1 and BRCA2 mutations in unselected Nigerian breast cancer patients. Int J Cancer. 131:1114–1123. 2012. View Article : Google Scholar

13 

Zheng Y, Ogundiran TO, Falusi AG, Nathanson KL, John EM, Hennis AJ, Ambs S, Domchek SM, Rebbeck TR, Simon MS, et al: Fine mapping of breast cancer genome-wide association studies loci in women of African ancestry identifies novel susceptibility markers. Carcinogenesis. 34:1520–1528. 2013. View Article : Google Scholar : PubMed/NCBI

14 

Francies FZ, Wainstein T, De Leeneer K, Cairns A, Murdoch M, Nietz S, Cubasch H, Poppe B, Van Maerken T, Crombez B, et al: BRCA1, BRCA2 and PALB2 mutations and CHEK2 c1100delC in different South African ethnic groups diagnosed with premenopausal and/or triple negative breast cancer. BMC Cancer. 15:9122015. View Article : Google Scholar

15 

Couch FJ, Kuchenbaecker KB, Michailidou K, Mendoza-Fandino GA, Nord S, Lilyquist J, Olswold C, Hallberg E, Agata S, Ahsan H, et al: Identification of four novel susceptibility loci for oestrogen receptor negative breast cancer. Nat Commun. 7:113752016. View Article : Google Scholar : PubMed/NCBI

16 

Huo D, Feng Y, Haddad S, Zheng Y, Yao S, Han YJ, Ogundiran TO, Adebamowo C, Ojengbede O, Falusi AG, 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. 25:4835–4846. 2016.

17 

Network CGA; Cancer Genome Atlas Network: Comprehensive molecular portraits of human breast tumours. Nature. 490:61–70. 2012. View Article : Google Scholar : PubMed/NCBI

18 

Ciriello G, Gatza ML, Beck AH, Wilkerson MD, Rhie SK, Pastore A, Zhang H, McLellan M, Yau C, Kandoth C, et al TCGA Research Network: Comprehensive molecular portraits of invasive lobular breast cancer. Cell. 163:506–519. 2015. View Article : Google Scholar : PubMed/NCBI

19 

Basro S and Apffelstaedt JP: Breast cancer in young women in a limited-resource environment. World J Surg. 34:1427–1433. 2010. View Article : Google Scholar

20 

Adedokun BO, Olopade CO and Olopade OI: Building local capacity for genomics research in Africa: Recommendations from analysis of publications in Sub-Saharan Africa from 2004 to 2013. Glob Health Action. 9:310262016. View Article : Google Scholar : PubMed/NCBI

21 

Sjögren S, Inganäs M, Norberg T, Lindgren A, Nordgren H, Holmberg L and Bergh J: The p53 gene in breast cancer: Prognostic value of complementary DNA sequencing versus immunohistochemistry. J Natl Cancer Inst. 88:173–182. 1996. View Article : Google Scholar : PubMed/NCBI

22 

Sluiter M, Mew S and van Rensburg EJ: PALB2 sequence variants in young South African breast cancer patients. Fam Cancer. 8:347–353. 2009. View Article : Google Scholar : PubMed/NCBI

23 

van Bogaert LJ: Breast cancer molecular subtypes as identified by immunohistochemistry in South African black women. Breast J. 19:210–211. 2013. View Article : Google Scholar : PubMed/NCBI

24 

Valent Ly M, Diallo A, Penault-Lorca G, Dumke F, Marty K, Viehl V, Lazar P, Job V, Richon BC, et al: Gene copy number variations in breast cancer of Sub-Saharan African women. Breast. 22:295–300. 2013. View Article : Google Scholar

25 

Langa BC, Oliveira MM, Pereira SR, Lupicki K, Marian C, Govender D, Panieri E, Hiss D, Cavalli IJ, Abdul-Rasool S, et al: Copy number analysis of the DLX4 and ERBB2 genes in South African breast cancer patients. Cytogenet Genome Res. 146:195–203. 2015. View Article : Google Scholar : PubMed/NCBI

26 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

27 

Hanahan D and Weinberg RA: Hallmarks of cancer: The next generation. Cell. 144:646–674. 2011. View Article : Google Scholar : PubMed/NCBI

28 

Negrini S, Gorgoulis VG and Halazonetis TD: Genomic instability - an evolving hallmark of cancer. Nat Rev Mol Cell Biol. 11:220–228. 2010. View Article : Google Scholar : PubMed/NCBI

29 

Korkola J and Gray JW: Breast cancer genomes - form and function. Curr Opin Genet Dev. 20:4–14. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Cancer A-AJCo: Cancer Staging Manual. Society AC: Springer; 2002

31 

de Wit E, Delport W, Rugamika CE, Meintjes A, Möller M, van Helden PD, Seoighe C and Hoal EG: Genome-wide analysis of the structure of the South African coloured population in the Western Cape. Hum Genet. 128:145–153. 2010. View Article : Google Scholar : PubMed/NCBI

32 

Quintana-Murci L, Harmant C, Quach H, Balanovsky O, Zaporozhchenko V, Bormans C, van Helden PD, Hoal EG and Behar DM: Strong maternal Khoisan contribution to the South African coloured population: A case of gender-biased admixture. Am J Hum Genet. 86:611–620. 2010. View Article : Google Scholar : PubMed/NCBI

33 

Santos SC, Cavalli IJ, Ribeiro EM, Urban CA, Lima RS, Bleggi-Torres LF, Rone JD, Haddad BR and Cavalli LR: Patterns of DNA copy number changes in sentinel lymph node breast cancer metastases. Cytogenet Genome Res. 122:16–21. 2008. View Article : Google Scholar : PubMed/NCBI

34 

Torresan C, Oliveira MM, Pereira SR, Ribeiro EM, Marian C, Gusev Y, Lima RS, Urban CA, Berg PE, Haddad BR, et al: Increased copy number of the DLX4 homeobox gene in breast axillary lymph node metastasis. Cancer Genet. 207:177–187. 2014. View Article : Google Scholar : PubMed/NCBI

35 

Sugita B, Gill M, Mahajan A, Duttargi A, Kirolikar S, Almeida R, Regis K, Oluwasanmi OL, Marchi F, Marian C, et al: Differentially expressed miRNAs in triple negative breast cancer between African-American and non-Hispanic white women. Oncotarget. 7:79274–79291. 2016. View Article : Google Scholar : PubMed/NCBI

36 

Cavalli LR, Man YG, Schwartz AM, Rone JD, Zhang Y, Urban CA, Lima RS, Haddad BR and Berg PE: Amplification of the BP1 homeobox gene in breast cancer. Cancer Genet Cytogenet. 187:19–24. 2008. View Article : Google Scholar : PubMed/NCBI

37 

Vlachos IS, Kostoulas N, Vergoulis T, Georgakilas G, Reczko M, Maragkakis M, Paraskevopoulou MD, Prionidis K, Dalamagas T and Hatzigeorgiou AG: DIANA miRPath v2.0: Investigating the combinatorial effect of microRNAs in pathways. Nucleic Acids Res. 40W:W498–W504. 2012. View Article : Google Scholar

38 

Vlachos IS, Paraskevopoulou MD, Karagkouni D, Georgakilas G, Vergoulis T, Kanellos I, Anastasopoulos IL, Maniou S, Karathanou K, Kalfakakou D, et al: DIANA-TarBase v7.0: indexing more than half a million experimentally supported miRNA:mRNA interactions. Nucleic Acids Res. 43D:D153–D159. 2015. View Article : Google Scholar

39 

Paraskevopoulou MD, Georgakilas G, Kostoulas N, Vlachos IS, Vergoulis T, Reczko M, Filippidis C, Dalamagas T and Hatzigeorgiou AG: DIANA-microT web server v5.0: Service integration into miRNA functional analysis workflows. Nucleic Acids Res. 41W:W169–W173. 2013. View Article : Google Scholar

40 

Beroukhim R, Mermel CH, Porter D, Wei G, Raychaudhuri S, Donovan J, Barretina J, Boehm JS, Dobson J, Urashima M, et al: The landscape of somatic copy-number alteration across human cancers. Nature. 463:899–905. 2010. View Article : Google Scholar : PubMed/NCBI

41 

Loo LW, Wang Y, Flynn EM, Lund MJ, Bowles EJ, Buist DS, Liff JM, Flagg EW, Coates RJ, Eley JW, et al: Genome-wide copy number alterations in subtypes of invasive breast cancers in young white and African American women. Breast Cancer Res Treat. 127:297–308. 2011. View Article : Google Scholar : PubMed/NCBI

42 

Field LA, Love B, Deyarmin B, Hooke JA, Shriver CD and Ellsworth RE: Identification of differentially expressed genes in breast tumors from African American compared with Caucasian women. Cancer. 118:1334–1344. 2012. View Article : Google Scholar

43 

Sturtz LA, Melley J, Mamula K, Shriver CD and Ellsworth RE: Outcome disparities in African American women with triple negative breast cancer: A comparison of epidemiological and molecular factors between African American and Caucasian women with triple negative breast cancer. BMC Cancer. 14:622014. View Article : Google Scholar : PubMed/NCBI

44 

Keenan T, Moy B, Mroz EA, Ross K, Niemierko A, Rocco JW, Isakoff S, Ellisen LW and Bardia A: Comparison of the genomic landscape between primary breast cancer in African American versus white women and the association of racial differences with tumor recurrence. J Clin Oncol. 33:3621–3627. 2015. View Article : Google Scholar : PubMed/NCBI

45 

Ademuyiwa FO, Tao Y, Luo J, Weilbaecher K and Ma CX: Differences in the mutational landscape of triple-negative breast cancer in African Americans and Caucasians. Breast Cancer Res Treat. 161:491–499. 2017. View Article : Google Scholar

46 

Zhang J, Fackenthal JD, Zheng Y, Huo D, Hou N, Niu Q, Zvosec C, Ogundiran TO, Hennis AJ, Leske MC, et al: Recurrent BRCA1 and BRCA2 mutations in breast cancer patients of African ancestry. Breast Cancer Res Treat. 134:889–894. 2012. View Article : Google Scholar : PubMed/NCBI

47 

Churpek JE, Walsh T, Zheng Y, Moton Z, Thornton AM, Lee MK, Casadei S, Watts A, Neistadt B, Churpek MM, et al: Inherited predisposition to breast cancer among African American women. Breast Cancer Res Treat. 149:31–39. 2015. View Article : Google Scholar :

48 

Rodriguez-Pinilla SM, Jones RL, Lambros MB, Arriola E, Savage K, James M, Pinder SE and Reis-Filho JS: MYC amplification in breast cancer: A chromogenic in situ hybridisation study. J Clin Pathol. 60:1017–1023. 2007. View Article : Google Scholar

49 

Andre F, Job B, Dessen P, Tordai A, Michiels S, Liedtke C, Richon C, Yan K, Wang B, Vassal G, et al: Molecular characterization of breast cancer with high-resolution oligonucleotide comparative genomic hybridization array. Clin Cancer Res. 15:441–451. 2009. View Article : Google Scholar : PubMed/NCBI

50 

Andor N, Maley CC and Ji HP: Genomic instability in cancer: Teetering on the limit of tolerance. Cancer Res. 77:2179–2185. 2017. View Article : Google Scholar : PubMed/NCBI

51 

Birkbak NJ, Eklund AC, Li Q, McClelland SE, Endesfelder D, Tan P, Tan IB, Richardson AL, Szallasi Z and Swanton C: Paradoxical relationship between chromosomal instability and survival outcome in cancer. Cancer Res. 71:3447–3452. 2011. View Article : Google Scholar : PubMed/NCBI

52 

Mroz EA, Tward AD, Hammon RJ, Ren Y and Rocco JW: Intra-tumor genetic heterogeneity and mortality in head and neck cancer: Analysis of data from the Cancer Genome Atlas. PLoS Med. 12:e10017862015. View Article : Google Scholar : PubMed/NCBI

53 

Andor N, Graham TA, Jansen M, Xia LC, Aktipis CA, Petritsch C, Ji HP and Maley CC: Pan-cancer analysis of the extent and consequences of intratumor heterogeneity. Nat Med. 22:105–113. 2016. View Article : Google Scholar

54 

Rajendran BK and Deng CX: Characterization of potential driver mutations involved in human breast cancer by computational approaches. Oncotarget. 8:50252–50272. 2017. View Article : Google Scholar : PubMed/NCBI

55 

Calin GA, Sevignani C, Dumitru CD, Hyslop T, Noch E, Yendamuri S, Shimizu M, Rattan S, Bullrich F, Negrini M, et al: Human microRNA genes are frequently located at fragile sites and genomic regions involved in cancers. Proc Natl Acad Sci USA. 101:2999–3004. 2004. View Article : Google Scholar : PubMed/NCBI

56 

Calin GA and Croce CM: MicroRNAs and chromosomal abnormalities in cancer cells. Oncogene. 25:6202–6210. 2006. View Article : Google Scholar : PubMed/NCBI

57 

Zhang L, Huang J, Yang N, Greshock J, Megraw MS, Giannakakis A, Liang S, Naylor TL, Barchetti A, Ward MR, et al: microRNAs exhibit high frequency genomic alterations in human cancer. Proc Natl Acad Sci USA. 103:9136–9141. 2006. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

December-2018
Volume 53 Issue 6

Print ISSN: 1019-6439
Online ISSN:1791-2423

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Lupicki K, Elifio-Esposito S, Fonseca AS, Weber SH, Sugita B, Langa BC, Pereira SR, Govender D, Panieri E, Hiss DC, Hiss DC, et al: Patterns of copy number alterations in primary breast tumors of South African patients and their impact on functional cellular pathways. Int J Oncol 53: 2745-2757, 2018
APA
Lupicki, K., Elifio-Esposito, S., Fonseca, A.S., Weber, S.H., Sugita, B., Langa, B.C. ... Cavalli, L.R. (2018). Patterns of copy number alterations in primary breast tumors of South African patients and their impact on functional cellular pathways. International Journal of Oncology, 53, 2745-2757. https://doi.org/10.3892/ijo.2018.4589
MLA
Lupicki, K., Elifio-Esposito, S., Fonseca, A. S., Weber, S. H., Sugita, B., Langa, B. C., Pereira, S. R., Govender, D., Panieri, E., Hiss, D. C., Abdul-Rasool, S., Cavalli, L. R."Patterns of copy number alterations in primary breast tumors of South African patients and their impact on functional cellular pathways". International Journal of Oncology 53.6 (2018): 2745-2757.
Chicago
Lupicki, K., Elifio-Esposito, S., Fonseca, A. S., Weber, S. H., Sugita, B., Langa, B. C., Pereira, S. R., Govender, D., Panieri, E., Hiss, D. C., Abdul-Rasool, S., Cavalli, L. R."Patterns of copy number alterations in primary breast tumors of South African patients and their impact on functional cellular pathways". International Journal of Oncology 53, no. 6 (2018): 2745-2757. https://doi.org/10.3892/ijo.2018.4589