Skip to main content

Functional proteomics can define prognosis and predict pathologic complete response in patients with breast cancer

Abstract

Purpose

To determine whether functional proteomics improves breast cancer classification and prognostication and can predict pathological complete response (pCR) in patients receiving neoadjuvant taxane and anthracycline-taxane-based systemic therapy (NST).

Methods

Reverse phase protein array (RPPA) using 146 antibodies to proteins relevant to breast cancer was applied to three independent tumor sets. Supervised clustering to identify subgroups and prognosis in surgical excision specimens from a training set (n = 712) was validated on a test set (n = 168) in two cohorts of patients with primary breast cancer. A score was constructed using ordinal logistic regression to quantify the probability of recurrence in the training set and tested in the test set. The score was then evaluated on 132 FNA biopsies of patients treated with NST to determine ability to predict pCR.

Results

Six breast cancer subgroups were identified by a 10-protein biomarker panel in the 712 tumor training set. They were associated with different recurrence-free survival (RFS) (log-rank p = 8.8 E-10). The structure and ability of the six subgroups to predict RFS was confirmed in the test set (log-rank p = 0.0013). A prognosis score constructed using the 10 proteins in the training set was associated with RFS in both training and test sets (p = 3.2E-13, for test set). There was a significant association between the prognostic score and likelihood of pCR to NST in the FNA set (p = 0.0021).

Conclusion

We developed a 10-protein biomarker panel that classifies breast cancer into prognostic groups that may have potential utility in the management of patients who receive anthracycline-taxane-based NST.

Introduction

To inform decisions about therapy, it is necessary to have a better understanding of the molecular mechanisms underlying the heterogeneity of breast cancer. Transcriptional profiling revealed that breast cancer represents at least six molecular subtypes associated with different clinical features [1–3]. However, comprehensive analysis of breast cancer transcriptomes does not capture all levels of biological complexity; important additional information may reside in the proteome [4–7].

Proteins are the direct effectors of cellular function. Protein levels and function depend on translation as well as on post-translational modifications [6], which influence protein stability and activity [7]. Although many proteins have been studied as prognostic and predictive factors in breast cancer, only three alter current practice: estrogen receptor (ER), progesterone receptor (PR) and HER2. Thus, a systematic study of expression and activation of multiple proteins and signaling pathways may facilitate more accurate classification and prediction in breast cancer.

Neoadjuvant systemic therapy (NST) allows for in vivo assessment of chemosensitivity. Attaining a pathologic complete response (pCR) following NST provides a surrogate marker for improved long-term outcome. Conversely, patients with residual breast cancer after NST are at increased risk for recurrence and may have therapy-resistant disease [8–12].

The objective of this study was to apply functional proteomics to breast cancer classification and prognosis, and to develop a predictor of pCR in a group of primary tumor samples obtained by fine needle aspirations (FNA) from patients who subsequently received NST.

Material and Methods

Tumor tissues

Three sets of frozen breast cancer tissues were used: Training set (n = 712) was collected at M. D. Anderson Cancer Center (MDACC), Hospital Clinico Universitario de Valencia, Spain, University of British Columbia, Vancouver, BC, and Baylor College of Medicine, Houston, TX. Complete clinical information was available for 541 patients. Test set (n = 168) was obtained from an independent group of patients enrolled in the Danish DBCG 82 b and c breast cancer studies [13, 14]. All tumors in the training and test sets were collected by excision during their primary surgery. Tumor content was verified by histopathology. The third set consisted of 256 FNAs obtained from primary breast cancers prior to NST of which 132 belonged to patients who subsequently received uniform taxane and anthracycline-based NST at MDACC (12 cycles of weekly paclitaxel or 4 cycles of every 3-week docetaxel, followed by 4 cycles of FAC or FEC100). All tissues were collected under Institutional Review Board-approved laboratory protocols.

Tumors were characterized for ER and PR status by immunohistochemistry (IHC), ligand-binding dextran-coated charcoal assay or reverse phase protein lysate array (RPPA). ER/PR positivity was designated when nuclear staining occurred in ≥10% of tumor cells, with ligand binding of ≥ 10 fmol/mg, or with a log2 mean centered cutoff of -1.48(ER) or +0.52(PR) by RPPA. Hormone receptor (HR) positivity was designated when either ER or PR was positive. HER2 status was assessed by IHC, fluorescent in situ hybridization (FISH) or RPPA. HER2 positivity was designated when 3+ membranous staining occurred in ≥10% of tumor cells, with a HER2/CEP17 ratio of > 2.0 or with a log2 mean centered cutoff of +0.82 by RPPA [15].

Reverse phase protein lysate microarray (RPPA)

RPPA was completed independently and at different time points for training and tests sets using individual arrays. Protein was extracted from human tumors and RPPA was performed as described previously [16–19]. Lysis buffer was used to lyse frozen tumors by homogenization (excised tumors) or sonication (FNAs). Tumor lysates were normalized to 1 μg/μL concentration as assessed by bicinchoninic acid assay (BCA) and boiled with 1% SDS. Supernatants were manually diluted in five-fold serial dilutions with lysis buffer. An Aushon Biosystems 2470 arrayer (Burlington, MA) created 1,056 sample arrays on nitrocellulose-coated FAST slides (Schleicher & Schuell BioScience, Inc.). Slides were probed with 146 validated primary antibodies (Additional File 1, Table S1) and signal amplified using a DakoCytomation-catalyzed system. Secondary antibodies were used as a starting point for amplification. Slides were scanned, analyzed, and quantified using Microvigene software (VigeneTech Inc., Carlisle, MA) to generate spot signal intensities, which were processed by the R package SuperCurve (version 1.01) [18], available at "http://bioinformatics.mdanderson.org/OOMPA". A fitted curve ("supercurve") was plotted with the signal intensities on the Y-axis and the relative log2 concentration of each protein on the X-axis using the non-parametric, monotone increasing B-spline model [18]. Protein concentrations were derived from the supercurve for each lysate by curve-fitting and normalized by median polish. Protein measurements were corrected for loading as described [15–17, 19]. For the selection of the 146 antibody set, we focused on markers currently used for breast cancer classification due to their value in treatment decisions (ER, PR, HER2). We then added additional antibodies to targets implicated in breast cancer pathophysiology, followed by antibodies to targets implicated in the pathophysiology of other cancer lineages. Final selection of antibodies was also driven by the availability of their high quality that could pass a strict validation process as previously described [20].

Statistical Methods

Detailed statistical methods are described in Additional File 2.

Identification of Prognostic Groups

To develop a set of markers for breast cancer classification and outcomes prediction, we used a hypothesis-driven approach, selecting markers according to their functional assignments and subsequently performing supervised proteomic clustering analysis to optimize the selection of groups with the most distinct recurrence-free survival (RFS) outcomes. We hypothesized that three functions would strongly affect the behavior and therapy responsiveness in breast cancer: ER function, grade/proliferation, and receptor tyrosine kinase activity. From the initial 146 antibodies, we selected markers within these three functional categories. We tested multiple combinations requiring that a minimum of one marker per functional category remain in each model. Unsupervised clustering analysis, using the uncentered correlation distance metric [21] and Ward's linkage rule [22], was applied to the training set to define groups and allow correlation with previously defined breast cancer subtypes. We then visualized the RFS curves to select the marker set that was associated with the clearest differences in RFS between the groups identified in the training set. Because of multiple testing and the possibility of false discovery, this model was locked and then applied to an independent test set to which the statistical analysis team was kept blinded. The selected protein groups were as follows: ER function (ER, ERpS118, ERpS167, PR, AR, EIG121, Bcl2, GATA3, IGF1R, and IGFBP2), grade/proliferation (CCNB1, CCND1, CCNE1, CCNE2, and PCNA), and receptor tyrosine kinase activity (cKit, EGFR, EGFRp1045, EGFRp922, HER2, HER2p1248, FGFR1, FGFR2, IGF1R, IGFRpY1135/Y1136).

RFS was estimated according to the Kaplan-Meier method and compared between groups using the log-rank statistic. Cox proportional Hazard Models were fitted using proteomic subgroups, selected markers and clinical variables.

Decision trees

We constructed a statistical model to predict the classes discovered by hierarchical clustering using a binary decision tree with a logistic regression model at each node. The split at each node was a union of two of the classes. Protein-by-protein two-sample t-tests between the two halves of the split were computed. The proteins were ordered by p-value and then added one at a time into a logistic regression model until the desired prediction accuracy was achieved. In order to avoid overfitting data, a default precision accuracy of 95% was set for each node. Finally, the Akaike Information Criterion (AIC) was used to eliminate redundant terms from the logistic regression model [23].

Validation of Prognostic Groups for RFS

The coefficients of the model, which used logistic regression at each node of a decision tree to place samples in one of six classes (or prognostic groups) were finalized and locked. An implementation of the model was provided to an independent analyst, along with the class predictions. The independent analyst was provided with the unblinded clinical data after implementation of the model. Cox proportional hazards models were then constructed using the predicted classes as covariates to test their association with RFS.

Validation of Prognostic Groups for pCR

We applied the algorithm to the last sample set (132 FNAs) and correlated the groups with response to NST. We clustered the samples as above and compared these clusters to the class labels predicted by the decision tree model with Cohen's kappa statistic [24, 25]. Using the predicted prognostic groups, we developed a Bayesian model to estimate the posterior probability of pCR in each group. We modeled the pCR rates as coming from a beta-binomial distribution [26].

Development of a Prognostic Score and its Application to Prediction of pCR

We next converted the six prognostic groups into a continuous prognostic score (PS) by fitting an ordinal regression model on the training set [27]. PS is a weighted linear combination of the relative protein concentration of the markers:

PS = -0.2841*ER - 1.3038*PR + 0.0826*Bcl2 -0.6876*GATA3 + 0.5169*CCNB1 + 0.1000*CCNE1 + 0.4321*EGFR + 0.5564*HER2 + 0.8284*HER2p1248 +

0.2424*EIG121.

We used this formula to compute PS on the test set; PS was associated with RFS estimates by the Cox proportional hazards model. We also used the same formula to compute PS on the NST treated FNA set. We fitted a logistic regression model using the NST response as the binary response variable (pCR vs. residual disease) and PS as a predictor. The prediction of response was evaluated by a receiver operating characteristics (ROC) curve.

Models for Recurrence-Free Survival and Likelihood of Pathologic Complete Response

A Cox proportional hazards model to estimate association with RFS was fit using each of the following covariates: prognostic group, tumor size, histologic grade, node status, each of the 10 protein markers, and PS. Using the same covariates, a logistic regression model was fit to estimate the association of each covariate with pCR. Stepwise multivariate model selection [28, 29] was used to determine the combination of covariates for the multivariate models.

All statistical analysis was performed in R 2.8.1. (R Development Core Team (2008). R: A language and environment for statistical computing (R Foundation for Statistical Computing, Vienna, Austria). http://www.R-project.org.

Results

Unsupervised Proteomic Clustering

Table 1 summarizes the clinical characteristics of each set. Training set (n = 712) was analyzed for 146 proteins (Additional File 1, Table S1) using RPPA. Proteins were chosen based on a literature search of important targets and proteomic processes in breast cancer for which robust antibodies binding to a single or dominant band on western blotting could be identified and validated for RPPA as described [1–3, 30–32]. Unsupervised clustering of the proteomic profiles is shown in Additional file 1: Figure S1. The 146 proteins stratified breast cancers into six major groups with different RFS outcomes (Additional file 1: Figure S2). The six groups included a predominantly HER2-positive group, a HR-negative and HER2-negative (triple receptor-negative) group with poor outcomes, a HR-positive group with a good outcome and three groups with intermediate outcome: an HR group with overexpression of proteins including cyclins B1 and E1 as well as components of the protein synthesis machinery including phosphorylated S6 ribosomal protein and 4EBP1, a group with overexpression of stromal markers including collagen VI, CD31 and caveolin1, and a group defined by up-regulation of a large number of proteins and phospho-proteins in several mechanistic pathways.

Table 1 Clinical characteristics of all sets

Supervised Proteomic Clustering

The hypothesis-driven approach described in Methods was applied to the training set and identified 10 markers in three functional groups known to be important to breast cancer behavior: ER function (ER, PR, Bcl2, GATA3, EIG121), tyrosine kinase receptor function (EGFR, HER2, HER2p1248), and cell proliferation (CCNB1, CCNE1). These markers separated the breast cancers into six subgroups (PG1 to 6) with markedly different RFS outcomes, (Log-rank p = 8.8 E-10), (Figures 1A and 1D). A decision tree model was developed (Figure 1C) that recovered the six subgroups of breast tumors identified by clustering with the 10 markers with an overall accuracy of 89%. Full description of the model is presented in Additional File 3. We then confirmed the presence of the six subgroups as well as their RFS in an independent test set, (Log-rank p = 0.0013), (Figures 1B and 1E). Table 2 summarizes the 5-year RFS estimates for each of the prognostic groups in the training and test sets.

Table 2 Five-year DFS estimates for each of the prognostic groups in both the training and test sets
Figure 1
figure 1

Supervised clustering of breast cancers with quantification data for 10 proteins derived using reverse phase protein arrays. The 712 breast tumor samples (Training set, 1A) were clustered with the 10 markers using an "uncentered correlation" distance metric along with the Ward linkage rule. This analysis yielded six subgroups (BG1-6). The 168 breast tumor samples (Test set, 1B) were subgrouped into one of 6 groups (PG1-6) using the decision tree (1C) that was derived from the training set. Patients in the six subgroups differed significantly in their recurrence-free survival in both training (1D) and test (1E) sets.

We applied this classification approach to 256 FNAs from MDACC. In order to confirm that the same clusters were present, we compared the patient groups obtained by direct hierarchical clustering of the 256 FNA samples to the prognostic groups predicted in the FNA samples by the decision tree model derived from the training set (Cohen's κ = 0.70, p < 1E-20). The decision tree predictions were also applied to the subset of 132 FNAs from patients who received uniform anthracycline and taxane-based NST, and the same six clusters were found (Cohen's κ = 0.66, p value < 1E-20, Figure 2A). The association between pCR rates and the (predicted) prognostic groups did not quite reach statistical significance (χ2 = 10.3076 on 5 degrees of freedom; p = 0.067). However, a Bayesian analysis of the pCR rates indicated that there was at least a 70% posterior probability that groups PG2 and PG3 have pCR rates at least 5% lower than those in PG4 or PG6 (Figure 2B).

Figure 2
figure 2

The 132 fine needle aspirates from patients who received anthracycline and taxane-based neoadjuvant systemic therapy were subgrouped into one of the 6 groups using the decision tree from the training set. Six true patient groups were obtained (2A), Cohen's kappa score = 0.66. Beta-binomial distribution and computed joint posterior probabilities were used to evaluate the association of the prognostic groups with pCR, the posterior distribution estimates of pCR by prognostic group are shown in 2B.

Prognostic Score Predicts pCR

As described in Methods, we computed a continuous prognostic score (PS) based on the grouping defined in the training set. A Cox proportional hazards model on the training set (CoxTrain) using PS to predict RFS was significant (Wald test; coefficient = 0.128, p = 3.2E-13). A second Cox model, fit on the test set (CoxTest), was also significant (Wald test; coefficient = 0.084, p = 1.1E-05) (Figure 3A). Of 132 patients who received anthracycline-taxane-based NST, 32 (24%) had a pCR. We computed the prognostic score PS for each FNA sample; the values ranged from -8.16 to 10.16. A logistic regression model showed that PS was also significantly associated with pCR (p = 0.0021, Figure 3B). Further, an unequal variance t-test comparing the prognostic scores between patients with pCR and residual disease also revealed a significant difference between mean scores (p = 0.00024 Figure 3C). The area under the curve (AUC) in a ROC curve analysis was 0.7 with a specificity of 98% and a negative predictive value of 76% (Figure 3D).

Figure 3
figure 3

A ten-protein prognosis score by ordinal regression modeling was derived from the training set. 3A. Probability of recurrence as a continuous function of the score. The rug plot shows the prognosis score for individual patients in the study. Dashed curves indicate the 95 percent confidence intervals. 3B. Probability of pCR as a function of the prognostic score. 3C. Stripcharts showing the level of prognostic score by response to anthracycline and taxane-based neoadjuvant systemic therapy. 3D. Receiver operating characteristics curves for the performance of the prediction of pCR versus residual disease by the logistic model using the prognostic score. AUC: area under the curve.

Models for Recurrence-Free Survival and Likelihood of Pathologic Complete Response

Univariate models for RFS (Cox proportional hazards on the test set; CoxTest) and pCR (logistic regression on the uniformly treated FNA dataset; LR-FNA) are summarized in Table 3. All clinical and molecular variables, except for EGFR, were significantly associated with RFS. The addition of the prognostic score to the model with clinical covariates reduced the residual deviance with a X21 = 2.96, p = 0.09. Stepwise model selection using AIC retained all clinical covariates and the prognostic score for the final model:

Table 3 Models for Recurrence-Free Survival and likelihood of pathological complete response

log(h(t)/h 0 (t)) = 0.414Size + 1.34Node + 0.803Grade + 0.070PrognosticScore.

For response (pCR vs. residual disease), grade was the only clinical covariate significantly associated with response. All protein markers except EGFR, HER2, pHER21248 and EIG121 were significantly associated with response. The addition of the prognostic score to grade reduced residual deviance with a X21 = 5.39, p = 0.02. Stepwise model selection using AIC showed that both grade and prognostic score were retained in the final model:

logit(pCR) = -2.61 + 0.902Grade + 0.2210PrognosticScore.

We compared ROC curves for predicting pCR by the prognostic scores and the stepwise selected model and found that AUC, as well as the specificity and negative predictive values were the same (0.7, 98% and 76% respectively), suggesting that the prognostic score may be a more powerful predictor than clinical information.

Discussion

We have identified and validated a 10-protein panel that accurately and reproducibly classifies patients with breast cancer into six subgroups with significantly different 5-year RFS times. These six groups included two HR positive groups differentiated primarily by PR levels with the PR high group having the best outcome, a HER2, pHER2 and EGFR positive group with the worst outcome (pre-trastuzumab treatment) and three triple negative groups, one with high cyclins and two groups without well defined selectors. Further, in an independent set of FNAs from patients who underwent NST, we were able to reproduce this classification and to use it to predict response to neoadjuvant anthracycline and taxane-based therapy. Further, in three independent sets, the 10-protein signature had a higher predictive value than clinical variables including tumor size, nodal status and grade in Cox models for RFS and in a logistic regression model to predict pCR.

Several studies using transcriptional profiling have classified breast cancer into different subtypes with implications in patient prognosis [1, 30–32], frequency of genomic alterations [33, 34], and therapy response [31, 35, 36]. Since proteins are the immediate effectors of cellular behavior, interrogation of the functional proteome is likely to complement data derived from transcriptional profiling. Thus, the integrated study of the expression and activation of multiple proteins and signaling pathways has the potential to provide powerful classifiers and predictors in breast cancer. As protein levels and function depend not only on translation but also on post-translational modifications, functional proteomic profiling may theoretically yield more direct answers to functional and pharmacological questions than transcriptional profiling alone. However, practical, high-throughput approaches to the study of the functional proteome have not been available until recently. RPPA is a useful tool to identify and validate protein and phospho-proteins [19–23]. Our data suggest that RPPA has the potential to advance our understanding of breast cancer biology and to aid in the identification and validation of useful biomarkers. Our findings validate the importance of ER, PR and HER2. However, seven additional markers including other tyrosine kinase receptors and proliferation markers involved in therapy resistance (EGFR, CCNB1, CCNE1) are part of the 10-protein panel. The combination of 10 markers and the power of the 10 markers as compared to ER/PR and HER2 is novel. The ER, PR and HER2 and the proliferation markers correspond to other breast cancer classifiers such us the intrinsic subtypes or the Oncotype DX Recurrence Score which have also shown that ER, HER2 and proliferation are the most important classifiers, prognostic and predictive markers in breast cancer [1, 31]. This demonstrates that RPPA can capture prognostic and predictive differences associated with breast cancer subtypes.

Several factors are important in selection and validation of biomarkers: The analysis platform must be sufficiently robust to detect subtle changes between tumors. Sample sets must be robust enough to reduce pre-analytical data biases and must reflect the intended use of the marker or marker set. Independent sample sets must be used to validate the prognostic and predictive power of biomarkers particularly when many biomarkers are assessed on small sample sets. Lastly, bioinformatics support is essential at all steps in any project. The current study has satisfied all of the requirements mentioned above. RPPA is a robust platform able to detect minimal changes in protein levels [15]. Three large independent sample sets with adequate clinical and outcome information were used for training and testing. Bioinformaticians were closely involved in study design as well as data analyses.

Our findings also have limitations. Patient cohorts received diverse types of systemic treatments and limiting the ability to dissect effects on prognosis from variables that predict endocrine and/or chemotherapy sensitivity. When looking at pCR predictors, all prognostic signatures can reasonably predict pCR, however patients predicted to obtain pCR may have significantly worse survival than those predicted not to respond due to different prognostic variables i.e. HR positivity. So, if our signature is primarily prognostic, its potential utility for selecting chemotherapy sensitivity would be limited; for this reason, validation studies in independent cohorts are needed.

The issues of tumor heterogeneity and the utility of laser captured microdissection were considered in our previous work focusing on the technical assessment of the utility of RPPA for the study of the functional proteome in non-microdissected human breast cancers [20]. This approach used captures information contained in the tumor cells, the stroma and in particular the tumor stroma interaction. The approach of using the complete tumor including interactions of tumor and stroma to classify patients and predict outcomes is the basis for the current transcriptional profiling approaches such as Oncotype Dx or Mammaprint. We have attempted to develop and implement RPPA approaches on microdissected tumors. However, due to a number of technical challenges, this approach is not as robust as study of complete tumors which captures information from the tumor and the stroma as well as tumor stroma interactions.

In summary, we have developed a 10-protein biomarker panel that may have potential utility in the management of patients with breast cancer. Today, it is clear that we should view breast cancer as several distinct diseases. Thus, further work is needed to identify predictors of response to individual therapies that target different clinical and molecular subgroups of breast cancer.

Abbreviations

AIC:

Akaike Information Criterion

BCA:

Bicinchoninic acid assay

ER:

estrogen receptor

FISH:

Flourescent in-situ hybridization

FNA:

Fine needle aspirate

HR:

hormone receptor

IHC:

Immunohistochemistry

MDACC:

MD Anderson Cancer Center

NST:

Neoadjuvant systemic therapy

pCR:

Pathologic complete response

PR:

Progesterone receptor

PS:

Prognostic Score

RFS:

Recurrence-free survival

ROC:

Receiving operating curve

RPPA:

Reverse phase protein array

References

  1. Sorlie T, Perou CM, Tibshirani R: Gene expression patterns of breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci USA. 2001, 98: 10869-74. 10.1073/pnas.191367098

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  2. Pusztai L, Ayers M, Stec J: Gene expression profiles obtained from fine-needle aspirations of breast cancer reliably identify routine prognostic markers and reveal large-scale molecular differences between estrogen-negative and estrogen-positive tumors. Clin Cancer Res. 2003, 9: 2406-15.

    CAS  PubMed  Google Scholar 

  3. van de Vijver MJ, He YD, van't Veer LJ: A gene-expression signature as a predictor of survival in breast cancer. N Engl J Med. 2002, 347: 1999-09. 10.1056/NEJMoa021967

    Article  CAS  PubMed  Google Scholar 

  4. Gygi SP, Rochon Y, Franza BR, Aebersold R: Correlation between protein and mRNA abundance in yeast. Mol Cell Biol. 1999, 19: 1720-30.

    PubMed Central  CAS  PubMed  Google Scholar 

  5. Diks SH, Peppelenbosch MP: Single cell proteomics for personalised medicine. Trends Mol Med. 2004, 10: 574-7. 10.1016/j.molmed.2004.10.005

    Article  CAS  PubMed  Google Scholar 

  6. Yeh E, Cunningham M, Arnold H: A signalling pathway controlling c-Myc degradation that impacts oncogenic transformation of human cells. Nat Cell Biol. 2004, 6: 308-18. 10.1038/ncb1110

    Article  CAS  PubMed  Google Scholar 

  7. Amerik AY, Hochstrasser M: Mechanism and function of deubiquitinating enzymes. Biochem Biophys Acta. 2004, 1695: 189-207. 10.1016/j.bbamcr.2004.10.003

    Article  CAS  PubMed  Google Scholar 

  8. Rastogi P, Anderson SJ, Bear HD: Pathologic assessment of response to induction chemotherapy in breast cancer. Preoperative chemotherapy: updates of National Surgical Adjuvant Breast and Bowel Project Protocols B-18 and B-27. J Clin Oncol. 2008, 10 (26): 78-85.

    Google Scholar 

  9. Kuerer HM, Newman LA, Smith TL: Clinical course of breast cancer patients with complete pathologic primary tumor and axillary lymph node response to doxorubicin-based neoadjuvant chemotherapy. J Clin Oncol. 1999, 17: 460-9.

    CAS  PubMed  Google Scholar 

  10. Guarneri V, Broglio K, Kau S: Prognostic value of pathologic complete response after primary chemotherapy in relation to hormone receptor status and other factors. J Clin Oncol. 2006, 24: 1037-44. 10.1200/JCO.2005.02.6914

    Article  PubMed  Google Scholar 

  11. Symmans WS, Peintinger F, Hatzis C: Measurement of residual breast cancer burden to predict survival after neoadjuvant chemotherapy. J Clin Oncol. 2007, 25: 4414-22. 10.1200/JCO.2007.10.6823

    Article  PubMed  Google Scholar 

  12. Honkoop AH, Pinedo HM, De Jong JS: Effects of chemotherapy on pathologic and biologic characteristics of locally advanced breast cancer. Am J Clin Pathol. 1997, 107: 211-8.

    CAS  PubMed  Google Scholar 

  13. Overgaard M, Hansen PS, Overgaard J: Postoperative radiotherapy in high-risk premenopausal women with breast cancer who receive adjuvant chemotherapy. Danish Breast Cancer Cooperative Group 82b Trial. N Engl J Med. 1997, 337: 949-55. 10.1056/NEJM199710023371401

    Article  CAS  PubMed  Google Scholar 

  14. Overgaard M, Hansen PS, Overgaard J: Postoperative radiotherapy in high-risk postmenopausal breast-cancer patients given adjuvant tamoxifen: Danish Breast Cancer Cooperative Group DBCG 82c randomised trial. Lancet. 1999, 353: 1641-8. 10.1016/S0140-6736(98)09201-0

    Article  CAS  PubMed  Google Scholar 

  15. Stemke-Hale K, Gonzalez-Angulo AM, Lluch A: An integrative genomic and proteomic analysis of PIK3CA, PTEN and AKT mutations in breast cancer. Cancer Res. 2008, 68: 6084-91. 10.1158/0008-5472.CAN-07-6854

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  16. Tibes R, Qiu Y, Lu Y, Hennessy B, Mills GB, Kornblau S, : Validation of a novel proteomic technology and utility for analysis of primary leukemia specimens and hematopoietic stem cells. Mol Cancer Ther. 2006, 2512-21.

    Google Scholar 

  17. Liang J, Shao SH, Xu ZX, Hennessy B, Ding Z, Larrea M: The energy sensing LKB1-AMPK pathway regulates p27kip1 phosphorylation mediating the decision to enter autophagy or apoptosis. Nat Cell Biol. 2007, 9: 218-24. 10.1038/ncb1537

    Article  CAS  PubMed  Google Scholar 

  18. Hu J, He X, Baggerly KA, Coombes KR, Hennessy BT, Mills GB: Non-parametric quantification of protein lysate arrays. Bioinformatics. 2007, 23: 1986-94. 10.1093/bioinformatics/btm283

    Article  CAS  PubMed  Google Scholar 

  19. Hennessy BT, Lu Y, Poradosu E, Liu Q, Yu S, Hall H: Quantified pathway inhibition as a pharmacodynamic marker facilitating optimal targeted therapy dosing: Proof of principle with the AKT inhibitor perifosine. Clin Cancer Res. 2007, 13: 7421-31. 10.1158/1078-0432.CCR-07-0760

    Article  CAS  PubMed  Google Scholar 

  20. Hennessy BT, Lu Y, Gonzalez-Angulo AM, Carey MS, Myhre S, Ju Z: A technical assessment of the utility of reverse phase protein arrays for the study of the functional proteome in non-microdissected human breast cancers. Clin Proteomics. 2010, 6: 129-151. 10.1007/s12014-010-9055-y

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  21. Eisen MB, Spellman PT, Brown PO, Botstein D: Cluster analysis and display of genome-wide expression patterns. Proc Natl Acad Sci USA. 1998, 95: 14863-8. 10.1073/pnas.95.25.14863

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  22. Ward JH: Hierachical grouping to optimize an objective function. J Am Statist Assoc. 1963, 58: 236-44. 10.2307/2282967. 10.2307/2282967

    Article  Google Scholar 

  23. Akaike H: A new look at the statistical model identification. IEEE Transactions on Automatic Control. 1974, 19: 716-23. 10.1109/TAC.1974.1100705. 10.1109/TAC.1974.1100705

    Article  Google Scholar 

  24. Wolfrum C: Appearance of quasiequivalent solutions, in a generalization of the Kruskal scale technic, to metric spaces using a Minkowski metric. Arch Psychol. 1976, 128: 96-111.

    CAS  Google Scholar 

  25. Landis JR, Koch GG: The measurement of observer agreement for categorical data. Biometrics. 1977, 33: 159-74. 10.2307/2529310

    Article  CAS  PubMed  Google Scholar 

  26. Gelman A, Carlin JB, Stern HS, Rubin DB: Bayesian Data Analysis. 2004, 125-31. Boca Raton, FL. Chapman & Hall, 2,

    Google Scholar 

  27. Verweij PJM, Van Houwelingen JC: Penalized likelihood in Cox regression. Stat in Med. 1994, 13: 2427-36. 10.1002/sim.4780132307. 10.1002/sim.4780132307

    Article  CAS  Google Scholar 

  28. Hastie TJ, Pregibon D: Generalized linear models. Statistical Models in S. Wadsworth & Brooks/Cole Computer Science Series. Edited by: Chambers JM and Hastie TJ. 1991, 195-248. Boca Raton, Fla. Chapman & Hall/CRC,

    Google Scholar 

  29. Venables WN, Ripley BD: Generalized Linear Models. Modern Applied Statistics with S. Edited by: Venables WN, Ripley BD. 2002, 183-208. New York, NY. Springer-Verlag, 4,

    Chapter  Google Scholar 

  30. Ayers M, Symmans WF, Stec J: Gene expression profiles predict complete pathologic response to neoadjuvant paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide chemotherapy in breast cancer. J Clin Oncol. 2004, 22: 2284-93. 10.1200/JCO.2004.05.166

    Article  CAS  PubMed  Google Scholar 

  31. Paik S, Shak S, Tang G: A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004, 351: 2817-26. 10.1056/NEJMoa041588

    Article  CAS  PubMed  Google Scholar 

  32. Hu Z, Fan C, Oh DS: The molecular portraits of breast tumors are conserved across microarray platforms. BMC Genomics. 2006, 7: 96- 10.1186/1471-2164-7-96

    Article  PubMed Central  PubMed  Google Scholar 

  33. Fan C, Oh DS, Wessels L: Concordance among gene-expression-based predictors for breast cancer. N Engl J Med. 2006, 355: 560-9. 10.1056/NEJMoa052933

    Article  CAS  PubMed  Google Scholar 

  34. Bergamaschi A, Kim YH, Wang P: Distinct patterns of DNA copy number alteration are associated with different clinicopathological features and gene-expression subtypes of breast cancer. Genes Chromosomes Cancer. 2006, 45: 1033-40. 10.1002/gcc.20366

    Article  CAS  PubMed  Google Scholar 

  35. Neve RM, Chin K, Fridlyand J: A collection of breast cancer cell lines for the study of functionally distinct cancer subtypes. Cancer Cell. 2006, 10: 515-27. 10.1016/j.ccr.2006.10.008

    Article  PubMed Central  CAS  PubMed  Google Scholar 

  36. Hess KR, Anderson K, Symmans WF: Pharmacogenomic predictor of sensitivity to preoperative chemotherapy with paclitaxel and fluorouracil, doxorubicin, and cyclophosphamide in breast cancer. J Clin Oncol. 2006, 24: 4236-44. 10.1200/JCO.2006.05.6861

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements and funding

The authors are thankful to Xuemei Wang for outstanding statistical help and review of the manuscript.

This work was supported in part by the Kleberg Center for Molecular Markers at M. D. Anderson Cancer Center, ASCO Career Development Award, NCI 1K23CA121994-01 and NCI 1R21CA120248-01 (to A.M.G.), The Susan G. Komen Foundation FAS0703849 (to A. M. G., B. T. H., G. B. M.), NCI 1R01CA112199 and NCRR Grant 3UL1RR024148 (to F.M.B.), the Research Council of Norway grant 175240/S10 (to A.L.B.), and the Danish Cancer Society, Danish Council for Strategic Research, and CIRRO, The Lundbeck Foundation Centre for Interventional Research in Radiation Oncology (to J.A., J.O.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ana M Gonzalez-Angulo.

Additional information

Competing interests

Authors declare that they have no competing interests.

Authors' contributions

AMG-A: Contributed samples, Performed all experiments, Analyzed the data, Wrote the manuscript, Funded the experiments. BTH: Contributed samples, Performed all experiments, Analyzed the data, Wrote the manuscript, Funded the experiments. FMB: Contributed samples, Analyzed the data, Wrote the manuscript. AS: Contributed samples, Approved final manuscript. WL: Analyzed the data, Approved final manuscript. ZJ: Analyzed the data, Approved final manuscript. MSC: Contributed samples, Performed experiments. SM: Contributed samples, Approved final manuscript. CS: Contributed samples, Approved final manuscript. LD: Contributed antibodies, Approved final manuscript. RB: Contributed antibodies, Approved final manuscript. AL: Contributed samples, Approved final manuscript SA: Contributed samples, Approved final manuscript. PB: Contributed samples, Approved final manuscript. LP: Contributed samples, Approved final manuscript. WFS: Contributed samples, Approved final manuscript. JA: Contributed samples, Approved final manuscript. JO: Contributed samples, Approved final manuscript. A-LB-D: Contributed samples, Approved final manuscript. GNH: Contributed data, Approved final manuscript. KRC: Analyzed the data, Approved final manuscript. GBM: Contributed samples, Analyzed the data, Wrote the manuscript, Funded the experiments, Approved final manuscript. All authors read and approved the final manuscript.

Electronic supplementary material

12014_2011_10_MOESM1_ESM.DOC

Additional file 1: Supplemental Data. Table S1. Monospecific antibodies used in this study, Figure S1. Unsupervised clustering of 712 breast cancers (Training Set) using quantification data for 146 proteins derived using reverse phase protein arrays. Figure S2. Kaplan Meier Survival Curves for RFS of the 541 patients according to their subgroup classification. Table S2. Distribution of tumors by breast cancer subtype and prognostic group (PG) according to the 10 marker signature Figure S3. A plot of the deviance residuals from the cox PH model (using the prognostic score and grade as the predictors to model relapse free survival in the training data set) against the prognostic score. Figure S4. A plot of the deviance residuals from the logistic model (using the prognostic score and grade to predict the probability of pCR in the FNA test data set) against the predicted probability of pCR. (DOC 628 KB)

12014_2011_10_MOESM2_ESM.DOCX

Additional file 2: Expansion of the Statistical methods. More detailed description of the statistical methods with the corresponding references. (DOCX 31 KB)

12014_2011_10_MOESM3_ESM.PDF

Additional file 3: Breast cancer classifier via a logistic-regression decision tree. Locked logistic-regression tree used for validation (PDF 89 KB)

Authors’ original submitted files for images

Rights and permissions

This article is published under license to BioMed Central Ltd. This is an Open Access article distributed under the terms of the Creative Commons Attribution License (http://creativecommons.org/licenses/by/2.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Reprints and permissions

About this article

Cite this article

Gonzalez-Angulo, A.M., Hennessy, B.T., Meric-Bernstam, F. et al. Functional proteomics can define prognosis and predict pathologic complete response in patients with breast cancer. Clin Proteom 8, 11 (2011). https://doi.org/10.1186/1559-0275-8-11

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/1559-0275-8-11

Keywords