Identification of a metabolic and canonical biomarker signature in Mexican HR+/HER2-, triple positive and triple-negative breast cancer patients

  • Authors:
    • Silvia Cecilia Pacheco-Velázquez
    • Juan Carlos Gallardo-Pérez
    • José Luis Aguilar-Ponce
    • Patricia Villarreal
    • Luz Ruiz-Godoy
    • Manuel Pérez-Sánchez
    • Alvaro Marín-Hernández
    • Erika Ruiz-García
    • Abelardo Meneses-García
    • Rafael Moreno-Sánchez
    • Sara Rodríguez-Enríquez
  • View Affiliations

  • Published online on: September 25, 2014     https://doi.org/10.3892/ijo.2014.2676
  • Pages: 2549-2559
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Infiltrating ductal breast cancer (IDC) is the principal tumor associated-malignancy in Mexican women. In IDC, the development of intermittent hypoxia leads to an adaptive response coordinated by the transcriptional factor HIF-1α. In the present pilot, retrospective/cross-sectional study, the HIF-1α expression was analyzed in 102 tru-cut biopsies from female patients (51±12 years) without previous clinical treatment and compared to 31 normal breast biopsies. The 102 IDC samples corresponded to 56% of HER2-/HR+; 8% of HER2+/HR-; 22% of triple positive (HER2+/HR+); and 14% of triple negative (TN, HER2-/HR-) subtypes. To assess HIF-1α functionality, proteomic and kinetic analysis of glycolytic as well as mitochondrial enzymes, were determined. Validation of HIF-1α as cancer biomarker was assessed by determining the contents of the commonly used biomarkers c-MYC, Ki67, and H- and K-RAS, as well as metastatic and autophagy proteins. Proteomic analysis revealed that HIF-1α, c-MYC, HER2 and COXIV contents were significantly increased in all IDC subtypes vs. normal tissue. The contents and activities of glycolytic proteins were similar between normal and IDC samples, except for HER2-/HR+ where a substantial increase of HKII was observed. Significant increase in 2OGDH and E-cadherin was detected for TN samples vs. other IDC subtypes and for normal samples. These results clearly indicated that HIF-1α + COXIV + c-MYC (+ HER2 for HER2+ subtype) may be useful to depict a breast cancer metabolic marker pattern for diagnosis, whereas the contents of HIF-1α + c-MYC + 2OGDH + E-cadherin may be an alternative useful and reliable signature for TN subtype cancer prognosis.

Introduction

Breast cancer involves a wide range of genetic and biochemical alterations and different clinical behavior and outcomes. As a consequence, there is an imperative clinical need to accurately identify the different classes of breast cancer to better define specific therapies. Molecular characterization may allow for a more precise classification of breast cancer by identifying new functional and more specific markers and which may represent potential therapeutic targets and/or indicators of prognosis (1). Currently, routine clinical management of breast cancer involves the use of presumably sensitive and specific molecular markers such as the hormone receptors (HR) for estrogen (ER) and progesterone (PR), and the epidermal growth factor receptor 2 (HER2). Thus, ER, PR and HER2 immunohistochemistry (IHC) has become a routine and standard clinical task for diagnosis and treatment of breast cancer subtypes.

However, some relevant disadvantages have emerged from using these standard clinical procedures for diagnosis such as: i) the elevated number of false negatives (20% vs. total cases) and false positives (7% vs. total cases), i.e., increased levels of the ER, HER2 and PR proteins are also attained in non-tumor but proliferative tissues such as hematopoietic cells (2) and adipocytes of post-menopausal women (3) stimulated by proliferation-related cytokines (4); ii) the undefined and trial/error based treatment for the TN breast cancer patients (which account for approximately 20% of total cases) (4); iii) the acquired tumor resistance to prolonged trastuzumab initial treatments (35% vs. total cases) leading to metastatic progression despite the significant diminution detected in HER2 levels (70% vs. total cases) (5,6); and iv) the development of chemotherapy-resistant tumors after anti-hormonal treatments (7).

These disadvantages prompt the need for developing more reliable biomarkers with lower percentages of false positive and false negative responses.

Cancer is a multi-factorial disease (8). Therefore, the identification of several key proteins constituting different altered pathways (i.e., signaling, metabolism and proliferation), and whose contents and activities are very likely altered in tumor cells vs. normal cells, seems a more rational strategy, than that based on the identification of one single protein or gene (9,10), for precise tumor subtype diagnosis and improvement in the design of treatments for primary, refractory and metastatic tumors.

It has been proposed that the changes observed in several tumor signal transduction and metabolic pathways vs. normal ones may provide a molecular signature (8,10). The abnormal activation of the glycolytic pathway even under high oxygen availability is considered to be one of the most important metabolic hallmarks of cancer (reviewed in refs. 11,12). This observation takes relevance under normoxic and hypoxic conditions as well as normoglycemia and hypoglycemia, because glycolysis provides both ATP and glycolytic intermediaries for DNA, protein and lipid synthesis required for tumor proliferation and survival (12). In this regard, it has been determined that overexpression and secretion of glycolytic enzymes may improve diagnosis of ER+ breast cancer (13).

The molecular mechanism of the glycolytic activation involves several transcription factors and oncogenes (14,15). In particular, the hypoxia-inducible factor 1α (HIF-1α) increases the transcription of genes coding for specific glycolytic protein isoforms (GLUT1, GLUT 3, HK-I, HK-II, PFK-L, ALDO-A and -C, PGK1, ENO-α, PYK-M2, LDH-A and PFKFB-3) found overexpressed in tumor cells and absent (or in low expression) in their normal counterparts (reviewed in ref. 15). It has been also documented in monolayer cultures of metastatic tumors that elevated HIF-1α contents correlate with accelerated glycolysis (15) which in turn, correlates with high malignancy (16). Notably, the expression of all these particular glycolytic isoforms is linked to other activated tumor pathways such as cellular survival, apoptosis resistance and cellular migration onset, which are indeed also regulated by HIF-1α as well as by other transcription factors (15).

The HIF1α-glycolysis-malignancy interrelationship has not been studied in human tumor biopsies, although a high content in HIF1-α protein has been found in advanced clinical stages (IIIA) and in highly metastatic breast tumor biopsies vs. non-metastatic stage tumors, suggesting that this transcription factor can be a reliable diagnosis/predictive/prognostic biomarker (17). Unfortunately, HIF-1α increased levels can also be associated with other hypoxic diseases (heart attack, preeclampsia, diabetes, inflammation, ischemia and psoriasis) which are not linked to cancer development (18), deterring its potential role as cancer biomarker. Notwithstanding, to examine the possibility of establishing HIF1-α as selective biomarker in Mexican breast cancer patients, its content and functionality (assessing the contents and activities of glycoytic protein isoforms targeted by HIF-1α) were determined in human infiltrating ductal breast carcinoma biopsies. In parallel, canonical diagnosis/prognosis biomarkers involved in altered signaling tumor pathways were also analyzed. Furthermore, the contents of several mitochondrial enzymes were also determined to establish a possible relationship between HIF-1α and mitochondrial metabolism in cancer biopsies. The design of a global biomarker panel, including proteins of the most altered pathways in tumors (metabolism, proliferation and signal transduction) in different breast cancer subtypes may help to improve cancer prognosis and hence its clinical treatment.

Materials and methods

Human breast tumor tissue collection and histopathology

The present study represents a pilot (19), prospective/cross-sectional research approved by the Ethics and Research Committees of the Instituto Nacional de Cancerología (INCAN), México. The study from 2008 to 2013 included the evaluation of 102 women (age from 30–86, mean 51±12 years) diagnosed with infiltrating ductal breast carcinoma (IDC) also called invasive ductal carcinoma at the INCAN, México. According to the Cochran’s categorical formula for determining sample sizes in pilot and prospective studies, 100 was the appropriate sample size based on i) the α level = 0.05; and ii) an acceptable margin of error <10% (20).

Normal tissue was surgically withdrawn from selected areas of normal breast tissue of 31 cancer patients (age from 27 to 89 years, mean 58±17 years) and stored in liquid nitrogen in the INCAN Tumor Bank for 12–24 months (21). Internal control of sample stability revealed that the contents of several proteins (HIF-1α, GLUT, LDH and ATPase) from fresh samples were similar to those stored in the INCAN Tumor Bank (data not shown). Normal samples were further validated as non-tumorigenic by assessing immunohistochemistry (IHC) negativity towards HER2 and hormone receptor. Patients diagnosed with IDC and no previous clinical treatments were used as the inclusion criterion for the present study, whereas insufficient material for biochemical assays was used as the exclusion criterion.

Tumor samples were obtained following the medical proceedings for tissue handling and patient care approved by the Institutional Ethics Committee supported by a patient’s informed consent according to the Declaration of Helsinki. Patients were punctured with a tru-cut biopsy needle in the absence of local anesthesia. Histopathology was performed on hematoxylin and eosin stained biopsy slides. Each specimen was analyzed by visual inspection using standard light microscope by experimented pathologists as described (22) without knowledge of IHC results.

IHC analysis of biopsies

Human tissue was fixed in 10% neutral buffered formalin for 24 h. The sample was embedded in paraffin, cut with a microtome at 3 μm thick and placed on microscope slide. Immunostaining was performed with a BenchMark Ultra automated immunostainer (Ventana Medical Systems, Tucson, AZ, USA); estrogen receptor, progesterone receptor, HER2 and Ki67 protein antibodies were used at 1:100–250 dilutions. All antibodies were from Ventana Medical Systems, except for Ki67 (Bio SB Inc., Santa Barbara, CA, USA).

For estrogen and progesterone receptor identification, H-SCORE method was employed as described (23). For HER2 identification, negative score indicated <10% of stained cells; positive score indicated >30% of cellular staining.

The IHC analysis revealed that from 102 IDC biopsies, 23 corresponded to triple positive; 57 to HER2/HR+; 8 to HER2+/HR; and 14 to TN.

Western blot analysis

Samples from biopsies (0.4–1.8 mg total cellular protein, n=102) were placed in liquid nitrogen and kept at −70°C until used. For western blotting processing, frozen tumor and normal samples were powdered, re-suspended and homogenized in 0.6 ml 25 mM Tris-HCl buffer, pH 7.4, plus 1 mM PMSF (phenylmethanesulfonyl fluoride), 1 mM EDTA and 5 mM DTT, and centrifuged at 2,000 × g for 30 min at 4°C. Supernatants were recollected and protein content was determined by using the Lowry assay. IDC and normal breast samples (50 μg cellular protein) were re-suspended in loading buffer (10% glycerol; 2% SDS and 5% β-mercaptoethanol) and loaded onto 12.5% SDS-PAGE under denaturalizing conditions. The proteins were blotted to PVDF membranes (Bio-Rad Laboratories, Hercules, CA, USA) and protein identification was performed by overnight incubation with anti-HIF-1α, -GLUT1, -HKI, -HKII, -LDHA, -COXIV, -ATPase, -ANT, -GA, -2OGDH, -HER2, -c-MYC, -Ki67, -α-tubulin and -HRAS (Santa Cruz Biotechnology, Santa Cruz, CA, USA) specific monoclonal antibodies (1:500–1:1,000 dilution). Detection of the hybridization bands was performed with the horseradish peroxidase reaction in photographic plates as previously described. Densitometry analysis was carried out using Scion Image software (Scion Corp., Walkersville, MD, USA). Double normalization of tumor sample signal was first performed against its respective load control (α-tubulin) and then considering the normal tissue as 100% (24).

Enzyme activities

The supernatants from the frozen-thawed biopsy samples, prepared as described above, were stored at −20°C in the presence of 10% (v/v) glycerol until determination of enzyme activities. Hexokinase (HK) and lactate dehydrogenase (LDH) activities were spectrophotometrically determined at 340 nm and 37°C as described elsewhere (25). Briefly, HK activity was assayed in 50 mM MOPS buffer, pH 7.0 plus 2 U glucose-6-phosphate dehydrogenase, 1 mM NADP+, 15 mM MgCl2, 10 mM ATP and 20–60 μg cell extract protein/ml. The reaction was started by adding 3 mM glucose after 3-min pre-incubation and generation of NADPH was measured at 340 nm. Lactate dehydrogenase (LDH) was assayed in 50 mM MOPS, pH 7.0, 0.15 mM NADH and 10–20 μg cell extract protein/ml; after 3-min pre-incubation; the reaction was started with 1 mM pyruvate and NADH consumption was registered at 340 nm.

Statistical analyses

To identify significant differences in protein contents between non-tumor and tumor samples as well as among the different tumor subtypes, parametric and non-parametric statistical analyses were performed. The Kolmogorov-Smirnov and Levene tests (26,27) were applied to demonstrate the protein normal distribution and homogeneity of variance of samples. Analysis of Kolmogorov-Smirnov and Levene tests data indicated that non-parametric analysis (NPA) should be used for appropriate statistical assessment between non-tumor and tumor samples. The NPA analysis and graphical data were carried out by using the Microsoft SPSS v.20 (SPSS Inc., Chicago, IL, USA) and Microsoft OriginPro 8 (OriginLab Corp., Northampton, MA, USA) software, respectively. To validate the results obtained with NPA test, samples were re-analyzed by the Mann-Withney U test with a P<0.01 (28).

To assess differences of the analyzed proteins among the tumor subtypes HER2+/HR; HER2/HR+; TP and TN the NPA Kruskal-Wallis test and the parametric analysis ANOVA were applied. To validate the results of the Kruskal-Wallis and ANOVA tests, the Mann-Whitney U test (28) corrected by the Holm-Bonferroni method and Scheffé post hoc test was used. Receiver operative characteristic (ROC) curves were also built for further validation as well as identification of the cut-off values for each protein. All statistical tests were performed at significance level of at least 0.05 as reported for the majority of human biopsy studies (29).

Results

Patient characteristics

Infiltrating ductal breast cancer (IDC) is the most dominant type of breast carcinoma found in the Mexican female population (30). From a total of 116 cases analyzed, 88% (102 biopsies) corresponded to IDC. IDC incidence was identified by standard IHC analyses inspecting hematoxylineosin stains for apparent cellular abnormalities (atypias) and cellular mitosis (Fig. 1A). For HER2 positive subtypes, all samples showed HER2 intensity on tumor cell plasma membrane with 3+ score (i.e., >30% of stained cells) (Fig. 1B), whereas the score of all HER2 negative samples was 0 and 1+ (i.e., 100% non-stained cells) (31).

The clinical characteristics of the IDC subtypes are described in Table I. The highest number of IDC patients (57 samples or 56%) corresponded to HER2/HR+ followed by 23 samples (22%) of HER2+/HR+ (triple-positive, TP), 14 samples (14%) of HER2/HR (triple-negative, TN) and 8 samples (8%) of HER2+/HR. The percentages found in the present study for all IDC subtypes correlated with those reported in other clinical studies (32). The TN patients arguably have survival advantages as a result of the absence of HER2 overexpression. However, they lack the benefit of both routinely available targeted therapy and specific biomarkers. It is worth noting that the present prospective study analyzes a small number of IDC samples, as it has been reported for pilot clinical studies (19), and in contrast to other non-pilot epidemiological studies the IDC sample number is higher (>450 patients) (32); in consequence, in the present study the number of samples analyzed for each IDC subtype was smaller. Therefore, to avoid erroneous interpretations derived from a relative small number of samples, the analyses of the metabolic protein contents and activities were performed without considering the patient’s age or menopausal status. Future investigations will be oriented to increase the sample size to further validate the results found in the present study with other analytical methods such as RT-PCR and microarray approaches.

Table I

Clinical characteristics of infiltrating ductal breast carcinoma (IDC) patients.

Table I

Clinical characteristics of infiltrating ductal breast carcinoma (IDC) patients.

PatientsTotal n=97 (%)Ki67 score (%)TP (%) Her2/HR+ (%) Her2+/HR (%)TN (%)
Age (years)
 ≤454022111649
 >456020124035
IDC disease stage
 I480400
 II362082404
 III55211327510
 IV5202030
Tumor size
 T13151200
 T2322152025
 T3302381615
 T4342091654
Histological grade SBR
 37141600
 44150220
 51193800
 6202031511
 719137822
 82285737
 9183041104

[i] TN, triple negative; TP, triple positive; SBR, Scarff-Bloom-Richardson scale. Stage I corresponds to small tumors (<2 cm); stage II corresponds to tumors <5 cm which spread to axillary lymph nodes; stage III corresponds to cancer of any size that has spread to axillary lymph nodes, to lymph nodes near the breastbone or to the chest wall and/or skin; stage IV corresponds to metastatic cancer. The tumor size is based on the size of the tumor and the extent to which it has grown into neighboring breast tissue. The rating scale Scarff-Bloom-Richardson used in histopathological analysis considers the formation of breast tubule formation, nuclear pleomorphism and the number of cells entering mitosis.

Contents and activity of HIF-1α glycolytic and mitochondrial proteins in the IDC subtypes

The content of the transcription factor HIF-1α and its glycolytic targets GLUT1, HK (I and II) and LDH-A of tumor and non-tumor biopsies were normalized against α-tubulin (Fig. 2). The HIF-1α contents in tumor samples were significantly higher than those in non-tumor biopsies following the Mann-Whitney U test (Table II) and ROC analyses (Fig. 3A). The last test also revealed a cut-off point of 27 (i.e., 27% of the HIF1-α band intensity respect to the α-tubulin signal) for tumor vs. non tumor samples, where the highest sensibility (>80%) and specificity (>90%) were attained (Table II). These data indicated that, at least, a HIF-1α band intensity of a 27% is required to ensure that, in the IDC biopsy, <20% of false negatives and 10% of false positives can be found.

Table II

Statistical analysis revealing significant differences (showed in bold letters) in tumor vs. non-tumor breast tissue samples.

Table II

Statistical analysis revealing significant differences (showed in bold letters) in tumor vs. non-tumor breast tissue samples.

ProteinMann-Withney U test P-value (P<0.01)ROC analysisResult vs. non-tumor samples

AUCP-value
HIF-1α <0.0010.9050.000Statistically different
GlycolyticGLUT10.793Non-statistically different
HKI0.578Non-statistically different
HKII0.420Non-statistically different
LDH-A0.874Non-statistically different
OxPhosGA0.698Non-statistically different
2OGDH0.193Non-statistically different
ANT0.0040.420.471Non-statistically different
COX <0.0010.7060.001Statistically different
ATPsynthase0.656Non-statistically different
CanonicalKi67<0.0010.6100.321Non-statistically different
HER2 <0.0010.8250.000Statistically different
C-MYC <0.0010.7700.000Statistically different
OncogenesHRAS0.593Non-statistically different
KRASs0.224Non-statistically different
MetastasisVimentin0.283Non-statistically different
E-cadherin0.871Non-statistically different
AutophagyBNIP30.0080.7010.058Non-statistically different
LAMP0.0350.7000.071Non-statistically different

The increased HIF-1α content detected in tumor samples was accompanied by a slight, but non-significant increase in the protein content of its glycolytic target GLUT1. On the contrary, HK (I and II) and LDH-A contents were without change compared to normal breast tissue (Fig. 2). The functional determination of HK and LDH supported the observed western blot analysis. Activity of HK and LDH from tumor biopsies (22±5 mU/mg protein for total HK; n=23; and 307±78 mU/mg protein for total LDH; n=23) was similar to those from breast normal tissue (16±5 mU/mg protein for total HK; n=5; and 474±196 mU/mg protein for total LDH; n=5).

The contents of HIF-1α, GLUT1, HKI and LDH-A were similar among the different IDC subtypes (Fig. 4). However, for HKII a significant change, indicated by ANOVA and Scheffé post hoc tests, was determined in HER2+/HR vs. TP, vs. HER2/HR+ and vs. TN (Fig. 4; Table III). The ROC for HKII indicated a cut-off of 73.5 with sensitivity and specificity of 86 and 31%, respectively.

Table III

ROC parameters revealing cut-off points, sensitivity and specificity percentages of different biomarkers in tumor vs. non-tumor samples.

Table III

ROC parameters revealing cut-off points, sensitivity and specificity percentages of different biomarkers in tumor vs. non-tumor samples.

ProteinAUCP-valueCut-offSensitivity %Specificity %RRPPV %NPV %
HIF1-α0.9050.0000.588.777.42.9592.568.6
4.587.683.92.8893.467.6
27.0585.690.32.7995.465.9
HER20.8250.0003.5651003.2110069
13.8601002.9410066
26.2551002.7210063.3
C-MYC0.7700.0000.3576.774.21.8889.652.27
2.174.477.41.8590.551.1
30.7861.180.61.5990.343.1
COX0.7060.0010.543.396.81.5197.735.3
15.0438.11001.5310034.8

[i] AUC, area under ROC; RR, relative risk; PPV, positive predictive value; NPV, negative predictive value.

For mitochondrial proteins, no apparent changes were observed in the contents of 2OGDH, GA isoforms K and L, and ATP synthase between tumor breast biopsies vs. non-tumor tissue (Fig. 2). On the contrary, COXIV and adenine nucleotide translocase (ANT) levels were significantly higher and lower, respectively, in tumor vs. non-tumor samples by using Mann-Withney U test (Fig. 2; Table II). However, the ROC tests (Fig. 3A) revealed that only COXIV, but not ANT, was significantly different vs. non-tumor tissue. For this case, the cut-off point was 15 with 100% specificity but low (38%) sensitivity, indicating that COXIV could be considered as a good tumor biomarker but with high probability (>50%) to detect also false positives (Table III). The content of 2OGDH was similar between TP and HER2/HR+. On the contrary, in TN samples, the levels of 2OGDH were significantly higher vs. the pooled subpopulation of the other IDC subtypes (Fig. 4 and Table IV), whereas for HER2+/HR the content of 2OGDH was lower although non-significantly different vs. TP and HER2/HR+ subtypes, by applying ANOVA and Scheffé post hoc tests (Fig. 4). For 2OGDH, data of cut-off value, sensitivity and specificity (Table V) showed that it is required at least 70% of band intensity (respect to that of tubulin as the loading control) in the biopsy to diminish to 20 and 40% the probability to have false positives or negatives, respectively. Also, a substantial but not significant increase in the ANT content was observed in TN compared to pooled subpopulation of the other IDC subtypes (Fig. 4), in which the ANT content was similar.

Table IV

Statistical analysis revealing significant differences among IDC subtypes.

Table IV

Statistical analysis revealing significant differences among IDC subtypes.

Protein1st Statistical analysisP-value2nd Statistical analysisP-valueStatistically different between subtypes
HKIIANOVA test0.024Scheffé post hoc test0.046Her 2 vs. TP
Scheffé post hoc test0.033Her 2 vs. HR
2OGDHANOVA test0.001Scheffé post hoc test0.001TN vs. TP
Scheffé post hoc test0.008TN vs. HR
Scheffé post hoc test0.045TN vs. Her 2
HER2ANOVA test<0.001Scheffé post hoc test0.025Her2 vs. TP
Scheffé post hoc test0.00Her2 vs. HR
Scheffé post hoc test0.003Her2 vs. TN
KRASKruskal-Wallis test0.023Mann Whitney U corrected by the Holm-Bonferroni methodNo differences
E-cadherinKruskal-Wallis test0.001Mann Whitney U corrected by the Holm-Bonferroni method0.000249TN vs. HR
0.009TN vs. TP

[i] Her2 (Her2 positive), Her2+/HR; HR, Her2/HR+ (hormone receptor positive); TP, triple positive; TN, triple negative.

Table V

ROC parameters revealing cut-off points, sensitivity and specificity percentages of 2OGDH and E-cadherin in triple negative samples.

Table V

ROC parameters revealing cut-off points, sensitivity and specificity percentages of 2OGDH and E-cadherin in triple negative samples.

ProteinAUCP-valueCut-offSensitivity %Specificity%
2OGDH0.8690.0047.9510023.4
43.4510040.4
76.2383.364.8
E-cadherin0.8920.0022.883.370.2
36.983.385.1
102.366.7100

[i] AUC, area under ROC.

Contents of the proliferation, oncogenes, metastasis and autophagy proteins in IDC subtypes

In tumor breast biopsies, the proliferation protein Ki67, the oncogene c-MYC and the growth factor HER2 (in TP and HER2+/HR subtypes) were fully apparent, whereas no presence of these proteins was detected in normal breast tissue (Fig. 5), in agreement with previous reports (33). The Mann-Withney U test (Table II) showed that Ki67, c-MYC and HER2 were significantly different in TP and HER2+/HR vs. normal biopsies. ROC analysis showed that HER2 and c-MYC exhibited higher area under curve (AUC) than Ki67 (Table III) indicating that HER2 and c-MYC were indeed significantly different in TP and HER2+/HR vs. normal biopsies (Fig. 3A). For these two proteins, similar cut-off points, sensitivity and specificity were determined (Table III), indicating that only 26–30% of protein signal (respect to the tubulin content) in TP and HER2+/HR samples is required to achieve: i) high probability (80–100%) to discard negative diagnosis (i.e., values lower than 26–30% indicate the absolute certainty of absence of disease), although ii) moderate probability (55–60%) for true positive identification (vs. false positive, i.e., the sample derived from a non-cancer patient having some other health problem).

HER2 presence (TP, HER2+/HR) or absence (TN, HER2/HR+), as well as HR positivity or negativity, was confirmed for the majority of the samples by western blotting (Fig. 6), further validating the standard IHC clinical approach (Fig. 1B; Table I) currently used at the Instituto Nacional de Cancerología de México. However, 12 out of 71 HER2 samples (17%), and 3 out of 22 HR samples (14%), as indicated by the IHC assay, yielded positive signal by western blot assay (data not shown). These results clearly indicated that additional support for breast cancer subtype diagnosis such as western blotting should be routinely implemented to decrease the emergence of false negatives and hence to establish the appropriate therapy.

Surprisingly, H- and K-RAS proteins were also found in normal tissue (Fig. 5). This result could not be confirmed by literature data regarding the presence or absence of H- or K-RAS in normal cells. Assessment of Ki67, oncogenes and transcription factor contents among the different IDC subtypes by the Krustal-Wallis test only revealed significant difference for the HER2 content in TP vs. HER2+/HR. However, the Holm-Bonferroni/Mann-Whitney U corrected test showed no significant difference for such datasets.

Proteins involved in the metastatic response of tumor cells such as vimentin and E-cadherin were determined in tumor vs. non-tumor samples to assess the migration and invasion profiles of tumor biopsies. Both proteins were not statistically different between assayed groups (Fig. 5; Table II). Although, both BNIP3 (a key regulator of hypoxia-induced autophagy) as well as LAMP1 (lysosome biogenesis-induced autophagy) were significantly higher in IDC samples vs. non-tumor samples (Fig. 5), according to the Mann-Whitney test, ROC analysis showed no significant differences. Among IDC subtypes, metastasis and autophagy protein contents were similar, except for TN whose E-cadherin content was significantly higher (Fig. 3B; Table IV) to those determined for HER2/HR+ and TP. In this regard, an E-cadherin cut-off value of 3–37 was determined (Table V), indicating that this is the required protein expression (respect to the tubulin signal) to reach a high probability (80–100%) to identify true positive samples and discard false positive and false negative results.

Identification of new metabolic biomarkers in IDC subtypes

Identification of significant differences among the contents of all proteins assayed (metabolic proteins, oncogenes, transcription factors, as well as metastatic and autophagic proteins) between tumor and non-tumor tissue was determined by using the stringent non-parametric statistical Mann-Whitney U test validated by the ROC graphs (Fig. 3A; Tables II, III and V). With these rigorous statistical analyses, significant differences between non-tumor and tumor samples were observed for HIF-1α > HER2 > c-MYC > COXIV (Fig. 3A; Table II), whereas non-significant changes were detected for the rest of the mitochondrial and glycolytic proteins analyzed. In contrast, for the identification of significant differences on the proteins assayed among the different IDC subtypes, non-parametric statistical Kruskal-Wallis test and the parametric ANOVA validated by the ROC graphs (Figs. 1B and 3B; Tables IV and V) were used. These last analyses showed significant changes in 2OGDH and E-cadherin protein contents in TN vs. TP, HER2+/HR and HER2/HR+, thus, providing a differentiated and selective panel of biomarkers constituted by HIF-1α > c-MYC > 2OGDH > E-cadherin for TN subtype IDC biopsies.

Discussion

Accurate and early detection, and successful treatment of cancer are at present still unsolved challenging clinical problems. This situation most likely derives from the variety of genetic and biochemical strategies a tumor is able to develop to survive under a wide range of environmental stresses such as hypoxia and normoxia, hypoglucemia and normoglucemia, and immune host response. This cellular robustness allows a tumor to efficiently deal with the inhibition of a single kinase, transcription factor or oncogene, as usually thought, designed, and applied in the treatment of cancer in experimental animal models (9,34). Under this context, it is clearly understandable and expected the eventual and frequent emergence of tumor chemo-resistance from these mono-therapy regimes.

Tumor protective mechanisms include the activation of multiple and redundant kinases, transcription factors and oncogenes that readily circumvent the initial therapy directed to a single target (34). Therefore, as a suitable alternative strategy, multisite or combinatory therapy should be considered (9,34,35). Some clinical treatments empirically have employed multisite therapy against well-known over-expressed proteins in tumor cells with moderately higher success rates.

In breast tumor biopsies two overexpressed proteins (HER2 and HR) have frequently been used as biomarkers (36,37). Therefore, a clinical combination treatment of breast cancer with tamoxifen against HR plus trastuzumab against HER2 is commonly used. However, i) positive responses against HER2 or HER2+/HR+ treatment are not always obtained. For instance, trastuzumab alone or combined with taxol or cisplatin or 5-fluorouracil plus surgery plus hormone therapy showed no effect in 50–75% of cases with metastatic breast cancer (reviewed in ref. 9); ii) the content of this marker is low (15–20%) compared with other highly expressed proteins such as mTOR (40%) and cyclin D1 (50%) in HER2-positive cancer patients (36,38); and iii) HER2 and HR predictive power severely diminishes when they are individually analyzed (39).

It has been demonstrated that the HER2 and HR protein contents change over time affecting the selected clinical treatment (40). One study revealed a severe cancer recurrence after anti-HER2 or anti-HR treatment in more than 1000 women diagnosed with early-stage breast cancer. A puzzling observation is that tumor recurrence correlates with a diminution in the levels of HER2 (33%) and HR (15%) contents in all biopsies analyzed compared with the initial diagnoses validated by a high HER2 and HR overexpression (40). In other breast cancer patients the status of the HER2 receptor changes from positive to negative or vice versa (40). The inconsistencies described for these canonical markers have led to the search for other molecular biomarkers which may reliably improve cancer diagnosis and prognosis.

HIF-1α as a suitable cancer biomarker in breast cancer

High HIF-1α content and glycolysis are indicative of increased malignancy and poor prognosis (41). Therefore, HIF-1α has been proposed as a biomarker for different metastasic carcinomas (colon, breast, gastric, lung, skin, prostate, ovarian, pancreatic, brain, glioblastoma and renal) (reviewed in refs. 42,43). In all these studies, the HIF-1α content in cancer samples, solely evaluated by IHC, showed an increase of 30–83% vs. normal tissue (43). Our present data using western blot analyses revealed a substantial increment of HIF-1α in all cancer subtypes compared with their respective normal tissue (Fig. 2), indicating that this transcription factor can be a reliable biomarker of breast cancer biopsies. The high sensitivity and specificity of the HIF-1α detection by western blotting (i.e., the antibody selectively detects a target protein in a mixture of several thousand different proteins coming from heterogeneous tissue) compared to the IHC assay makes the former the method of choice for routine clinical use, despite the longer processing time for western blotting (days) than for IHC.

On the other hand, HIF-1α functional status has not been systematically evaluated to establish a correlation between protein content and transcriptional activity (42,44). In the present study the transcriptional functionality of HIF-1α was assessed in all IDC biopsies. The data indicated that although a substantial increment in the HIF-1α levels was observed compared to normal tissue (Fig. 1), all IDC subtypes maintain similar GLUT1, HKII and LDH-A contents and activities to those determined in non-tumor samples. Unfortunately, there are no studies in which the content and activity of these glycolytic proteins have been determined, except for LDH whose activity, determined here, was within the range reported for breast cancer patients (45). Nevertheless, the present data indicated that HIF-1α, but not the glycolytic proteins can be a striking reliable marker of Mexican breast carcinoma as has been already suggested for human squamous cancer cervix epithelium (46). In other studies performed in breast tumor perinecrotic area and cervical cancer a strong correlation between HIF-1α and GLUT1 has been found (44,46).

Regarding mitochondrial OxPhos proteins, only the ANT content was detected to be significantly decreased in the cancer biopsies. In PC12 tumor cells, short-term hypoxia (30 min) downregulates the transcription of genes encoding mitochondrial complex I/NADH dehydrogenase as an adaptive response mechanism for adjusting the OxPhos rate to the low O2 availability (47). A putative regulatory site in the ANT gene for HIF-1α has not been reported.

Clinical implications in the search of new biomarkers for triple negative breast cancer

Epidemiological studies of breast cancer in the Mexican female population revealed that approximately 20% of patients develop the TN phenotype (36). Unfortunately until now, the treatment with anti-hormone drugs (tamoxifen) or monoclonal anti-HER2 (trastuzumab) has been ineffective in the majority of these diagnosed cases (48). Therefore, it appears relevant to develop research focused on the identification of specific TN biomarkers. Recently it has been documented that the MAG13-AKT3 protein may fulfill such as role (49). However, its content is only overexpressed in a scarce number of TN patients (7% or 5/72), disabling its use for all TN patients (49). In the present study, significant changes in some mitochondrial (2OGDH) and metastatic (E-cadherin) proteins were observed in TN samples vs. other IDC subtypes (Figs. 4 and 6). Therefore, an alternative therapeutic strategy may be the use of combined treatment including the usual first line of treatment (tamoxifen or 5-fluorouracil or doxorubicin or cyclophosphamide or trastuzumab) plus some mitochondrially-targeted inhibitor (casiopeina II-gly and vitamin E analogues) (50,51).

In conclusion, all analyzed breast cancer subtypes exhibited high HIF-1α levels. Therefore, anti-HIF therapy (i.e., echinomycin and bortezomib) combined with canonical drugs (and/or energy metabolism drugs for TN cases) could be a promising alternative treatment against breast cancer. In this regard, it has been documented that HIF-1α overexpression in cancer cells is linked to a substantial augment in EGFR levels. However, mono-therapy with cetuximab, a monoclonal anti-EGFR antibody or gefitinib yields low efficacy (52), suggesting that combinatory therapy with canonical drugs plus anti-HIF1α therapy (plus energy inhibitors for TN cases) may be required for effective tumor abolishment.

Acknowledgements

The present study was partially supported by grants from CONACyT-México (nos. 80534, 107183,123636 and 180322) and the Instituto de Ciencia y Tecnología del Distrito Federal (no. PICS08-5) to S.R.E., R.M.S. and A.M.H. S.C.P.V. was supported by a CONACyT-México fellowship (no. 269212). The present study is part of the PhD thesis of S.C.P.V. in the Doctorado en Ciencias Biomédicas program at the Universidad Nacional Autónoma de México. The authors thank Dr I. Pérez-Neri for his help in the use of the different statistical methods.

References

1 

Rakha EA and Ellis IO: Triple-negative/basal-like breast cancer: review. Pathology. 41:40–47. 2009. View Article : Google Scholar

2 

Leone F, Perissinotto E, Cavalloni G, et al: Expression of the c-ErbB-2/HER2 proto-oncogene in normal hematopoietic cells. J Leukoc Biol. 74:593–601. 2003. View Article : Google Scholar : PubMed/NCBI

3 

McInnes KJ, Andersson TC, Simonytė K, Söderström I, Mattsson C, Seckl JR and Olsson T: Association of 11β-hydroxysteroid dehydrogenase type I expression and activity with estrogen receptor β in adipose tissue from postmenopausal women. Menopause. 19:1347–1352. 2012.

4 

Regil-Hallmann RS, Ibarra-del-Río M and Flores-Hernández L: Correlación citohistológica en el Instituto Nacional de Cancerología en el año 2006. Patología. 46:309–314. 2008.(In Spanish).

5 

Mittendorf EA, Wu Y, Scaltriti M, et al: Loss of HER2 amplification following trastuzumab-based neoadjuvant systemic therapy and survival outcomes. Clin Cancer Res. 15:7381–7388. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Vu T and Claret FX: Trastuzumab: updated mechanisms of action and resistance in breast cancer. Front Oncol. 2:622012.PubMed/NCBI

7 

González-Angulo AM, Morales-Vasquez F and Hortobagyi GN: Overview of resistance to systemic therapy in patients with breast cancer. Adv Exp Med Biol. 608:1–22. 2007.PubMed/NCBI

8 

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

9 

Moreno-Sánchez R, Saavedra E, Rodríguez-Enríquez S, Gallardo-Pérez JC, Quezada H and Westerhoff HV: Metabolic control analysis indicates a change of strategy in the treatment of cancer. Mitochondrion. 10:626–639. 2010.PubMed/NCBI

10 

Rodríguez-Enríquez S, Pacheco-Velázquez SC, Gallardo-Pérez JC, et al: Multi-biomarker pattern for tumor identification and prognosis. J Cell Biochem. 112:2703–2715. 2011.PubMed/NCBI

11 

Moreno-Sánchez R, Rodríguez-Enríquez S, Marín-Hernández A and Saavedra E: Energy metabolism in tumor cells. FEBS J. 274:1393–1418. 2007.

12 

Moreno-Sánchez R, Rodríguez-Enríquez S, Saavedra E, Marín-Hernández A and Gallardo-Pérez JC: The bioenergetics of cancer: is glycolys the main ATP supplier in all tumor cells? Biofactors. 35:209–225. 2009.PubMed/NCBI

13 

Amon LM, Pitteri SJ, Li CI, et al: Concordant release of glycolysis proteins into the plasma preceding a diagnosis of ER+ breast cancer. Cancer Res. 72:1935–1942. 2012. View Article : Google Scholar : PubMed/NCBI

14 

Yeung SJ, Pan J and Lee MH: Roles of p53, MYC and HIF-1 in regulating glycolysis: the seventh hallmark of cancer. Cell Mol Life Sci. 65:3981–3999. 2008. View Article : Google Scholar : PubMed/NCBI

15 

Marín-Hernández A, Gallardo-Pérez JC, Ralph SJ, Rodríguez-Enríquez S and Moreno-Sánchez R: HIF-1alpha modulates energy metabolism in cancer cells by inducing over-expression of specific glycolytic isoforms. Mini Rev Med Chem. 9:1084–1101. 2009.PubMed/NCBI

16 

Gillies RJ and Gatenby RA: Adaptive landscapes and emergent phenotypes: why do cancers have high glycolysis? J Bioenerg Biomembr. 39:251–257. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Xueguan L, Xiaoshen W, Yongsheng Z, Chaosu H, Chunying S and Yan F: Hypoxia inducible factor-1 alpha and vascular endothelial growth factor expression are associated with a poor prognosis in patients with nasopharyngeal carcinoma receiving radiotherapy with carbogen and nicotinamide. Clin Oncol. 20:606–612. 2008. View Article : Google Scholar

18 

Brahimi-Horn M and Pouysségur J: HIF at a glance. J Cell Sci. 122:1055–1057. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Leon AC, Davis LL and Kraemer HC: The role and interpretation of pilot studies in clinical research. J Psychiatr Res. 45:626–629. 2011. View Article : Google Scholar : PubMed/NCBI

20 

Bartlett JE II, Kotrlik JW and Higginsm CC: Organizational research: Determining appropriate sample size in survey. Off Syst Res J. 19:43–50. 2001.

21 

Ruíz-Godoy L, Meneses-García A, Suárez-Roa L, Enriquez V, Lechuga-Rojas R and Reyes-Lira E: Organization of a tumor bank: the experience of the National Cancer Institute of Mexico. Pathobiology. 77:147–154. 2010.

22 

Holland R, Peterse JL, Millis RR, Eusebi V, Faverly D, van de Vijver MJ and Zafrani B: Ductal carcinoma in situ: a proposal for a new classification. Semin Diagn Pathol. 11:167–180. 1994.PubMed/NCBI

23 

Cheang MC, Chia SK, Voduc D, et al: Ki67 index, HER2 status, and prognosis of patients with luminal B breast cancer. J Natl Cancer Inst. 101:736–750. 2009. View Article : Google Scholar

24 

Rodríguez-Enríquez S, Carreño-Fuentes L, Gallardo-Pérez JC, et al: Oxidative phosphorylation is impaired by prolonged hypoxia in breast and possibly in cervix carcinoma. Int J Biochem Cell Biol. 42:1744–1751. 2010.PubMed/NCBI

25 

Marín-Hernández A, Rodríguez-Enríquez S, Vital-González PA, Flores-Rodríguez FL, Macías-Silva M, Sosa-Garrocho M and Moreno-Sánchez R: Determining and understanding the control of glycolysis in fast-growth tumor cells. Flux control by an over-expressed but strongly product-inhibited hexokinase. FEBS J. 273:1975–1988. 2006.PubMed/NCBI

26 

Lilliefors HW: On the Kolmogorov-Smirnov Test for normality with mean and variance unknown. J Am Statistical Assoc. 62:399–402. 1967. View Article : Google Scholar

27 

Young DA, Zerbe GO and Hay WW Jr: Fieller’s theorem, Scheffé simultaneous confidence intervals, and ratios of parameters of linear and nonlinear mixed-effects models. Biometrics. 53:838–847. 1997.

28 

Krauth J: The interpretation of significance tests for independent and dependent samples. J Neurosci Methods. 9:269–281. 1983. View Article : Google Scholar : PubMed/NCBI

29 

Abba MC, Lacunza E, Butti M and Aldaz CM: Breast cancer biomarker discovery in the functional genomic age: a systematic review of 42 gene expression signatures. Biomark Insights. 5:103–118. 2010. View Article : Google Scholar : PubMed/NCBI

30 

Salazar EL, Calzada L and Pedron N: Infiltrating ductal/lobular carcinoma: an evaluation of prognostic factors in primary breast cancer. Arch AIDS Res. 10:73–82. 1996.PubMed/NCBI

31 

Gown AM: Current issues in ER and HER2 testing by IHC in breast cancer. Mod Pathol. 21:S8–S15. 2008. View Article : Google Scholar : PubMed/NCBI

32 

Carey LA, Perou CM, Livasy CA, et al: Race, breast cancer subtypes, and survival in the Carolina Breast Cancer Study. JAMA. 295:2492–2502. 2006. View Article : Google Scholar : PubMed/NCBI

33 

Mitas M, Mikhitarian K, Walters C, et al: Quantitative real-time RT-PCR detection of breast cancer micrometastasis using a multigene marker panel. Int J Cancer. 93:162–171. 2001. View Article : Google Scholar : PubMed/NCBI

34 

Xu AM and Huang PH: Receptor tyrosine kinase coactivation networks in cancer. Cancer Res. 70:3857–3860. 2010. View Article : Google Scholar : PubMed/NCBI

35 

Al-Lazikani B, Banerji U and Workman P: Combinatorial drug therapy for cancer in the post-genomic era. Nat Biotechnol. 30:679–692. 2012. View Article : Google Scholar : PubMed/NCBI

36 

Pérez-Sánchez VM, Vela-Chávez TA and Mora-Tiscareño MA: Diagnóstico histopatológico y factores pronósticos en cáncer infiltrante de glándula mamaria. Cancerología. 3:7–17. 2008.(In Spanish).

37 

Maughan KL, Lutterbie MA and Ham PS: Treatment of breast cancer. Am Fam Physician. 81:1339–1346. 2010.PubMed/NCBI

38 

Weigel MT and Dowsett M: Current and emerging biomarkers in breast cancer: prognosis and prediction. Endocr Relat Cancer. 17:R245–R262. 2010. View Article : Google Scholar : PubMed/NCBI

39 

Lari S and Kuerer H: Biological markers in DCIS and risk of breast recurrence: a systematic review. J Cancer. 2:232–261. 2011. View Article : Google Scholar : PubMed/NCBI

40 

Lindström LS, Karlsson E, Wilking UM, et al: Clinically used breast cancer markers such as estrogen receptor, progesterone receptor, and human epidermal growth factor receptor 2 are unstable throughout tumor progression. J Clin Oncol. 30:2601–2608. 2012.

41 

Porporato PE, Dhup S, Dadhich RK, Copetti T and Sonveaux P: Anticancer targets in the glycolytic metabolism of tumors: a comprehensive review. Front Pharmacol. 2:492011. View Article : Google Scholar : PubMed/NCBI

42 

Talks KL, Turley H, Gatter KC, Maxwell PH, Pugh CW, Ratcliffe PJ and Harris AL: The expression and distribution of the hypoxia-inducible factors HIF-1alpha and HIF-2alpha in normal human tissues, cancers, and tumor-associated macrophages. Am J Pathol. 157:411–421. 2000. View Article : Google Scholar : PubMed/NCBI

43 

Zhong H, De Marzo AM, Laughner E, et al: Overexpression of hypoxia-inducible factor 1alpha in common human cancers and their metastases. Cancer Res. 59:5830–5835. 1999.PubMed/NCBI

44 

Vleugel MM, Greijer AE, van der Wall E and van Diest PJ: Mutation analysis of the HIF-1alpha oxygen-dependent degradation domain in invasive breast cancer. Cancer Genet Cytogenet. 163:168–172. 2005. View Article : Google Scholar : PubMed/NCBI

45 

Balinsky D, Platz CE and Lewis JW: Isozyme patterns of normal, benign, and malignant human breast tissues. Cancer Res. 43:5895–5901. 1983.PubMed/NCBI

46 

Lee WY, Huang SC, Hsu KF, Tzeng CC and Shen WL: Roles for hypoxia-regulated genes during cervical carcinogenesis: somatic evolution during the hypoxia-glycolysis-acidosis sequence. Gynecol Oncol. 108:377–384. 2008. View Article : Google Scholar

47 

Piruat JI and López-Barneo J: Oxygen tension regulates mitochondrial DNA-encoded complex I gene expression. J Biol Chem. 280:42676–42684. 2005. View Article : Google Scholar : PubMed/NCBI

48 

Rexer BN and Arteaga CL: Intrinsic and acquired resistance to HER2-targeted therapies in HER2 gene-amplified breast cancer: mechanisms and clinical implications. Crit Rev Oncog. 17:1–16. 2012. View Article : Google Scholar : PubMed/NCBI

49 

Banerji S, Cibulskis K, Rangel-Escareno C, et al: Sequence analysis of mutations and translocations across breast cancer subtypes. Nature. 486:405–409. 2012. View Article : Google Scholar : PubMed/NCBI

50 

Rodríguez-Enríquez S, Marín-Hernández A, Gallardo-Pérez JC, Carreño-Fuentes L and Moreno-Sánchez R: Targeting of cancer energy metabolism. Mol Nutr Food Res. 53:29–48. 2009.

51 

Rodríguez-Enríquez S, Hernández-Esquivel L, Marín-Hernández A, Dong LF, Akporiaye ET, Neuzil J, Ralph SJ and Moreno-Sánchez R: Molecular mechanism for the selective impairment of cancer mitochondrial function by a mitochondrially targeted vitamin E analogue. Biochim Biophys Acta. 1817:1597–1607. 2012.PubMed/NCBI

52 

el Guerrab A, Zegrour R, Nemlin CC, et al: Differential impact of EGFR-targeted therapies on hypoxia responses: implications for treatment sensitivity in triple-negative metastatic breast cancer. PLoS One. 6:e250802001.PubMed/NCBI

Related Articles

Journal Cover

December-2014
Volume 45 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
Pacheco-Velázquez SC, Gallardo-Pérez JC, Aguilar-Ponce JL, Villarreal P, Ruiz-Godoy L, Pérez-Sánchez M, Marín-Hernández A, Ruiz-García E, Meneses-García A, Moreno-Sánchez R, Moreno-Sánchez R, et al: Identification of a metabolic and canonical biomarker signature in Mexican HR+/HER2-, triple positive and triple-negative breast cancer patients. Int J Oncol 45: 2549-2559, 2014
APA
Pacheco-Velázquez, S.C., Gallardo-Pérez, J.C., Aguilar-Ponce, J.L., Villarreal, P., Ruiz-Godoy, L., Pérez-Sánchez, M. ... Rodríguez-Enríquez, S. (2014). Identification of a metabolic and canonical biomarker signature in Mexican HR+/HER2-, triple positive and triple-negative breast cancer patients. International Journal of Oncology, 45, 2549-2559. https://doi.org/10.3892/ijo.2014.2676
MLA
Pacheco-Velázquez, S. C., Gallardo-Pérez, J. C., Aguilar-Ponce, J. L., Villarreal, P., Ruiz-Godoy, L., Pérez-Sánchez, M., Marín-Hernández, A., Ruiz-García, E., Meneses-García, A., Moreno-Sánchez, R., Rodríguez-Enríquez, S."Identification of a metabolic and canonical biomarker signature in Mexican HR+/HER2-, triple positive and triple-negative breast cancer patients". International Journal of Oncology 45.6 (2014): 2549-2559.
Chicago
Pacheco-Velázquez, S. C., Gallardo-Pérez, J. C., Aguilar-Ponce, J. L., Villarreal, P., Ruiz-Godoy, L., Pérez-Sánchez, M., Marín-Hernández, A., Ruiz-García, E., Meneses-García, A., Moreno-Sánchez, R., Rodríguez-Enríquez, S."Identification of a metabolic and canonical biomarker signature in Mexican HR+/HER2-, triple positive and triple-negative breast cancer patients". International Journal of Oncology 45, no. 6 (2014): 2549-2559. https://doi.org/10.3892/ijo.2014.2676