Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Clinical Value of Prognosis Gene Expression Signatures in Colorectal Cancer: A Systematic Review

  • Rebeca Sanz-Pamplona ,

    Contributed equally to this work with: Rebeca Sanz-Pamplona, Antoni Berenguer

    Affiliation Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), and CIBERESP, L’Hospitalet de Llobregat, Barcelona, Spain

  • Antoni Berenguer ,

    Contributed equally to this work with: Rebeca Sanz-Pamplona, Antoni Berenguer

    Affiliation Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), and CIBERESP, L’Hospitalet de Llobregat, Barcelona, Spain

  • David Cordero,

    Affiliation Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), and CIBERESP, L’Hospitalet de Llobregat, Barcelona, Spain

  • Samantha Riccadonna,

    Affiliation Predictive Models for Biomedicine & Environment (PMBE), Fondazione Bruno Kessler (FBK), Trento, Italy

  • Xavier Solé,

    Affiliation Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), and CIBERESP, L’Hospitalet de Llobregat, Barcelona, Spain

  • Marta Crous-Bou,

    Affiliation Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), and CIBERESP, L’Hospitalet de Llobregat, Barcelona, Spain

  • Elisabet Guinó,

    Affiliation Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), and CIBERESP, L’Hospitalet de Llobregat, Barcelona, Spain

  • Xavier Sanjuan,

    Affiliation Pathology Service, University Hospital Bellvitge (HUB – IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain

  • Sebastiano Biondo,

    Affiliations General and Digestive Surgery Service, University Hospital Bellvitge (HUB – IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain, Department of Clinical Sciences, Faculty of Medicine, University of Barcelona (UB), Barcelona, Spain

  • Antonio Soriano,

    Affiliation Gastroenterology Service, University Hospital Bellvitge (HUB – IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain

  • Giuseppe Jurman,

    Affiliation Predictive Models for Biomedicine & Environment (PMBE), Fondazione Bruno Kessler (FBK), Trento, Italy

  • Gabriel Capella,

    Affiliation Hereditary Cancer Program, Catalan Institute of Oncology (ICO - IDIBELL), L’Hospitalet de Llobregat, Barcelona, Spain

  • Cesare Furlanello,

    Affiliation Predictive Models for Biomedicine & Environment (PMBE), Fondazione Bruno Kessler (FBK), Trento, Italy

  • Victor Moreno

    v.moreno@iconcologia.net

    Affiliations Unit of Biomarkers and Susceptibility (UBS), Catalan Institute of Oncology (ICO), Bellvitge Biomedical Research Institute (IDIBELL), and CIBERESP, L’Hospitalet de Llobregat, Barcelona, Spain, Department of Clinical Sciences, Faculty of Medicine, University of Barcelona (UB), Barcelona, Spain

Abstract

Introduction

The traditional staging system is inadequate to identify those patients with stage II colorectal cancer (CRC) at high risk of recurrence or with stage III CRC at low risk. A number of gene expression signatures to predict CRC prognosis have been proposed, but none is routinely used in the clinic. The aim of this work was to assess the prediction ability and potential clinical usefulness of these signatures in a series of independent datasets.

Methods

A literature review identified 31 gene expression signatures that used gene expression data to predict prognosis in CRC tissue. The search was based on the PubMed database and was restricted to papers published from January 2004 to December 2011. Eleven CRC gene expression datasets with outcome information were identified and downloaded from public repositories. Random Forest classifier was used to build predictors from the gene lists. Matthews correlation coefficient was chosen as a measure of classification accuracy and its associated p-value was used to assess association with prognosis. For clinical usefulness evaluation, positive and negative post-tests probabilities were computed in stage II and III samples.

Results

Five gene signatures showed significant association with prognosis and provided reasonable prediction accuracy in their own training datasets. Nevertheless, all signatures showed low reproducibility in independent data. Stratified analyses by stage or microsatellite instability status showed significant association but limited discrimination ability, especially in stage II tumors. From a clinical perspective, the most predictive signatures showed a minor but significant improvement over the classical staging system.

Conclusions

The published signatures show low prediction accuracy but moderate clinical usefulness. Although gene expression data may inform prognosis, better strategies for signature validation are needed to encourage their widespread use in the clinic.

Introduction

Colorectal cancer (CRC) is the third most common cancer worldwide and the second leading cause of cancer death. During the last decades, incidence has been increasing, while mortality has slowly been decreasing [1]. A remarkable feature of CRC is the difference in prognosis of the early and late stages of the disease: stage I and II have moderate risk of relapse after surgical resection, whereas patients with stage III have a higher chance of recurrence [2]. Recognized clinical risk factors for recurrence are emergency presentation, poorly differentiated tumor, depth of tumor invasion, and adjacent organ involvement (T4) [3][5]. However, these factors are insufficient to identify those patients with stage II CRC at high risk of recurrence and posterior metastasis or those patients with stage III CRC at low risk [6], leading to potential under-treatment or over-treatment [3].

Colon cancer metastasis is a tightly regulated process that requires aberrations in gene expression allowing cancer cells to progress through various steps until they colonize a distant organ [7]. Probably the alterations necessary for recurrence are already present in the primary colon carcinoma, which should allow identifying prognostic signatures [8][10]. Gene-expression profiling-based assays have been successful as prognostic tool in breast cancer [11], [12]. However, no signature has been adopted in routine clinical practice in CRC despite a large number of gene expression profiling studies on prognosis have been performed.

The aim of this work was to test the predictive ability of these published signatures as prognostic markers in a significant number of independent datasets, in order to understand their strengths and weakness and identify if any of them can be used clinically to guide decisions about adjuvant therapy for patients with stage II or III CRC.

Materials and Methods

Published Gene Expression Signatures and Validation Datasets

A systematic literature review was performed to identify studies that used gene expression data to predict prognosis in CRC [13]. The search was based on the PubMed database and was restricted to recent papers to increase validity (from January 2004 to December 2011). Files S1–S2, Table S1 and Figure 1 detail the selection protocol and PRISM checklist. Articles that provided a list of differentially expressed genes in primary tumor samples associated with CRC prognosis were included in our study. We indistinctly refer to these lists of genes as ‘signatures’ or ‘profiles’. Studies based on tissue microarray and those that exclusively were focused on differences between stages or between primary tumor and metastases were excluded. The studies finally included for analysis are described in Table 1 [14][43]. Publicly available datasets with whole-genome gene expression measures in CRC primary tumor samples were identified and downloaded from GEO [44] and ArrayExpress [45] microarray data repositories (Tables 2 and 3). Pre-processed series matrixes originally provided by the authors were used in our analysis.

thumbnail
Figure 1. PRISMA Diagram which depicts the flow of information through the different phases of the prognosis signatures studies systematic review.

https://doi.org/10.1371/journal.pone.0048877.g001

Because different platforms and feature identifiers were used in signatures and gene expression datasets, a translation into the official Gene Symbol was done in order to have a common annotation. This translation was performed using the Universal Protein Resource annotation database [46], the online repository of HUGO Gene Nomenclature Committee [47] and the chip annotation files from the Affymetrix web site [48]. Unavoidably, no match was found for some features in some datasets and they were lost for subsequent analysis (File S3).

Statistical Analysis

Since follow-up time was not available for most of the datasets, a binary outcome was defined as a prognosis status (Table 2). Whenever possible, a minimum of three years of follow up was required for patients without tumor recurrence. Nevertheless, two datasets with no follow up information were included (GSE5206 and GSE10402) to increase the sample size. Stage IV individuals were included in the analysis as recurrence events as it was expected that the specific expression changes in poor prognostic samples remain unaltered in the primary tumor once the metastases has occurred. When data was available, subgroup analysis were performed according to stage and microsatellite instability status (MSS/MSI).

Each signature’s ability to predict prognosis was independently tested in each dataset with a binary classification approach using the Random Forest ensemble classifier (RF) [49], [50]. Forests were grown with a high number of trees (5.000) in order to assure out-of-bag error convergence. The minimum size of terminal nodes was set to one. For signatures evaluation, accuracy measures were computed from a 10-fold cross-validation (10CV) process in which partitions were stratified based on outcome. Nested in this process, the number of candidate variables at each split was selected to minimize the out-of-bag error. As suggested in [51], sub-sampling was carried out without replacement and using the same number of observations in each prognosis group (0.632 times frequency of the smallest group). Due to lack of balance in outcome groups in some datasets, RF showed a trend to preferably classify into the most frequent group. To correct this artifact, the classification vote cutoff was modified according to the corresponding prognosis group frequencies. All these analyses were performed using the R package randomForest [52].

To confirm our results, a radial kernel Support Vector Machine (SVM) based classifier was also used [53], [54]. Due to unbalance, the same artifact described above was observed when applying the standard SVM classifier. So, we tried to correct it using an under-sampling strategy as follows [55]: i) select all samples from the less frequent group; ii) randomly select the same number of samples from the more frequent group; iii) repeat the process 25 times; iv) define the predicted labels using the outcome group frequencies as vote proportion cutoff for the classification rule. A 10CV process was carried out to compute accuracy measures with a nested 10CV for parameter tuning, both of them stratified by outcome groups. A wide range of values for cost and radial kernel parameters were evaluated during the tuning process (20 equidistant values from 0.001 to 1.000 in logarithm scale; 11 equidistant values ranging 0.05×p to 20×p, being p the number of features in each case). All these analysis were conducted with the R package svmpath [56].

The Matthews Correlation Coefficient (MCC) [57] was chosen as measure of classification accuracy [58]. This index combines test sensitivity and specificity. It ranges from −1 to 1 and its interpretation is similar to the Pearson’s correlation coefficient. In the context of a classification problem it is expected that MCC ranges from 0 (no prediction ability at all) to +1 (perfect prediction) with negative values near zero possibly occurring in random classifiers due to sample variability. MCC values lower than 0.3 can be considered as indicative of low predictive value as they correspond to less than 65% accuracy in balanced data. Sensitivity, specificity and overall accuracy rates were also computed for interpretation purposes.

The potential usefulness of the signatures on clinical practice was evaluated by means of the positive and negative likelihood ratios (LR+, LR-) and the predicted positive and negative post-test recurrence probabilities (PPTpr, NPTpr) in stage II and III samples separately [59].

To summarize signature’s global performance, each of the measures above was pooled across datasets to a unique index using weights proportional to each dataset sample size. In order to attenuate instability and bias in the cross-validation estimations, datasets with less than 10 samples per group and those used in the derivation of the profile in the original study were excluded from these computations [60].

Significance of MCC, accuracy, sensitivity, specificity, LR+, LR-, and differences of PPTpr and NPTpr were assessed using null distributions based on 100.000 permutations. Computations were done in the context of the theoretical framework for permutation tests [61] as implemented in the R package coin [62]. Intervals at 95% were built using the Bias Corrected and Accelerated bootstrap (BCa) method with 5.000 resamples stratified by prognosis group [63]. Empirical influence values were estimated by the usual jackknife method. These calculations were done using the R package boot [64]. In all cases, permutations and resampling were performed directly on the predicted values provided by the original models and no remodeling was done. Since this strategy doesn’t take into account the dependence in predictions implicitly imposed by the 10CV, it could potentially retain some bias towards refusing null hypothesis in the statistical tests when effects are small [65].

Results

Global Prognosis Performance of the Published Signatures

The literature search identified 29 papers reporting 31 signatures proposed as valid multi-gene tumor-outcome classification tool (Table 1 and File S4). Almost all signatures were based on microarray experiments with the exception of three signatures obtained from PCR experiments (OC10, PL10, SC09). The number of genes (signature size) ranged from 3 to 537.

Despite recommendations to provide raw data for microarray experiments, training datasets were publicly available only for five signatures: ST09, SM09, BD07, LN07, and VL10. Six additional gene expression datasets with information about recurrence were identified in GEO and ArrayExpress, for a total of 11 datasets available for analysis (Table 2).

For all combinations of signatures and datasets, the MCC and the corresponding p-value was computed. Figure 2 shows a color map of the MCC values (details provided in File S5 and Figure S1). As expected, the five signatures for which the training was available showed significant association and a reasonable predictive accuracy in their training datasets (black-highlighted cells at the top left quadrant in the plot in Figure 2). For all these signatures, MCC values were greater than 0.35 except for VL10 and dataset GSE17536 (MCC = 0.32). Nevertheless, in the independent datasets the performance was heterogeneous and none of these five signatures could reproduce the degree of predictive ability shown in the training datasets. When the remaining 26 signatures (those without training set available) were evaluated in the 11 datasets, similar results were obtained: some signatures showed a significant association with patient outcome but discrimination accuracy was low or moderate.

thumbnail
Figure 2. Heatmap showing Matthews Correlation Coefficient (MCC) values for each signature in each dataset as result of analyses with Random Forest.

Rows correspond to signatures and columns to datasets. Last column shows a pooled MCC across datasets using sample size as weights. Black lines delimit the first five signatures for which training datasets were available (cells highlighted in black). Cells representing signatures and datasets used to validate them are highlighted in blue. Color scale represents the MCC values: the darker the color, the higher MCC (see the legend). Negative values were collapsed to zero.

https://doi.org/10.1371/journal.pone.0048877.g002

A global MCC was computed for each signature to summarize their predictive ability across datasets (see Figure 2). Signatures BT04 and KN11 emerged as the most predictive, both with a MCC value of 0.25 (95% CI 0.19 to 0.31 and 0.19 to 0.30 respectively, p-values <105). Although nearly all the signatures reached the 5% significance level in this pool estimate that combines 396 events in 1077 patients, only three signatures exceed a 0.20 global MCC. The maximum value obtained for the proportion of correctly classified cases was only 63% (BT04, sensitivity = 65% and specificity = 61%) and it ranged from 52 to 61% for the remaining profiles (Table 4, File S6).

To assess influence of the statistical methodology in the results, a re-analyses was performed using an alternative method (SVM). Although some variations in the signatures ranking of performance were observed, similar results were obtained in terms of pooled MCCs (Figure S2 and File S7).

Subgroup Analysis: Prognosis Performance of Published Signatures Stratified by Stage or MSI Status

In order to assess the signatures’ performance in specific subgroups of tumors, a stratified analysis was done according to stage (stage II/stage III) and microsatellite instability status (MSS/MSI), when this information was available (see Table 3). Datasets contributing with less than 10 events were excluded.

thumbnail
Table 4. Global performance of top 10 signatures for all, stage II and stage III samples.

https://doi.org/10.1371/journal.pone.0048877.t004

Similar to the analysis including all samples, the performance of the signatures was heterogeneous when stage II and III tumor samples were analyzed separately (Figures S3 and S4). In the pooled MCC, 17 signatures in stage II and 22 signatures in stage III showed a significant association with prognosis (p-value <0.05). Six signatures ranked in the top ten in both sub-analyses. The MCC values obtained in stage II were much lower than those in stage III. In stage II, the best global MCC were achieved by YM06 (MCC = 0.21; 95%CI 0.11 to 0.31) and BT04 (MCC = 0.20; 95%CI 0.10 to 0.31). In stage III, the two best signatures were AJ08 (MCC = 0.42; 95%CI 0.28 to 0.55) and VL10 (MCC = 0.40; 95%CI 0.23 to 0.55). Table 4, Files S5, S6 and S7; and Figures S3, S4, S5, S6, S7 and S8 contain more details.

MSI status information was only available for two datasets (GSE13294 and GSE18088). In the analysis of MSS samples, those MCC values that reached significance were moderate (0.19 to 0.38) and only three signatures showed association in both datasets (p-value <0.1). Regarding the MSI subset, only signature HO09 provided a reasonably classification accuracy (MCC = 0.30) (File S5).

Potential Clinical Value of Signatures in Stage II and III Tumors

Despite the low discrimination ability (shown by their pooled MCC), the signatures could still have usefulness in clinical practice. Briefly, a useful clinical test typically shows large LR+ and low LR- which translate into more discriminant post-test event probabilities: high PPTpr and low NPTpr compared to the a priori expected event proportion. So, even tests with low discrimination ability according to pure statistic criteria could still be useful in clinical practice if PPTpr and NPTpr are significantly far enough from the probability expected in population when no test is performed. To explore this issue, positive and negative post-test probabilities of recurrence were calculated for stages II and III. The prior recurrence risk in patients with CRC was assumed to be 20% in stage II and 34% in stage III [4], [66].

For the best signature in stage II (YM06), the post-test recurrence probability for the high-risk group increased to 28%, and for the low-risk group the prediction was 12% probability of recurrence (16% absolute difference, Figure 3A). The best profile in stage III (AJ08) increased to 56% the post-test probability of recurrence for the high-risk group, while the post-test probability was 18% for the low-risk group (38% absolute difference, Figure 3B). Detailed results for all signatures are shown in Files S6 and S7.

thumbnail
Figure 3. Differences between positive and negative post-test probabilities of recurrence and their 95% confidence interval for stage II (A) and stage III (B).

Prevalence probability of recurrence for stage II and III were assumed to be 20 and 34% respectively. Signatures are listed in decreasing order of post-tests probability differences.

https://doi.org/10.1371/journal.pone.0048877.g003

Discussion

The identification of molecular prognostic tools to facilitate treatment decisions is an important step for individualized patient therapy [10]. Here we report an exhaustive analysis of published multi-gene prognostic classifiers in colorectal cancer, analyzing their external validity in a large number of independent datasets that total more than 1.000 patients. The present work is focused in two objectives which are addressed by the two main parts of the analysis: to evaluate the global performance of the signatures from a statistical point of view, in which all stages were included, and to assess their potential clinical usefulness, restricted to stage II and III CRC patients, by means of appropriate accuracy measures (post-test probabilities).

A meta-analysis of gene expression profiles in stage II CRC has been previously reported by Lu et al. [67]. In that study, promising results were reported regarding predictive accuracy, but the analysis was confined to the same datasets and predictions used in the original studies. To our knowledge, our study is the first meta-analysis in which prediction accuracy of many signatures is measured in a large number of independent CRC samples to assess external validity and their subsequently potential usefulness in clinics.

In terms of global performance, our results indicate that in their training dataset, most signatures showed a significant association with prognosis and could reasonably predict the outcome. However, none of the signatures performed satisfactorily when the prediction ability was assessed in independent datasets. The best pooled MCC was 0.25 (BT04), which should be considered a low classification value. As a reference, stage provides an MCC of 0.23 (data not shown).

Next, we focused in specific performance of signatures in stage II and III patients, who could benefit more of an accurate prognosis prediction since adjuvant chemotherapy could be tailored to their predicted recurrence risk. Although association with outcome was observed for 17 signatures in stage II, their predictive ability can only be considered poor from a statistical point of view. Otherwise, MCC values in stage III were observed to be near double those in stage II. Nevertheless, only eight signatures achieved a 0.30 pooled MCC value, considered as indicative of moderate predictive value.

Although poor results were observed in terms of classification accuracy, almost all profiles (30) showed a significant association with prognosis when tested in independent datasets (p-value <0.05). Notice however that significant association only means that a signature prediction is not completely random (MCC = 0). Association is not sufficient to be useful since, with enough sample size, small effects can be significant. Better indicators of potential usefulness than significance are magnitude of the sensitivity and specificity or derived measures like the MCC or likelihood ratios, which measure the ability to correctly classify patients by their outcome.

Despite these disappointing results according to pure statistic criteria of discrimination ability, signatures might still be useful in clinical practice if they provide additional risk stratification within known sub-populations defined by relevant clinical variables. The positive and negative post-test probabilities of recurrence were calculated stratified by stage to identify the degree of prognosis discrimination beyond stage. The results for the best signature in stage II samples (YM06) moderately modified the 20% a priory recurrence probability to a 28% and 12% for the positive and negative post-test result, respectively. This discriminating ability is not completely satisfactory given the large false negative and positive rates that it would induce, but might contribute to the identification of stage II patients at high risk for recurrence leading to a better indication of adjuvant chemotherapy [6]. The best signature for stage III patients (AJ08) resulted in a larger discrimination of risk groups, with a difference between positive and negative post-test probabilities of 38%. However, the low risk group still showed a relatively large recurrence probability of 18%, too high to recommend avoiding adjuvant chemotherapy as it is indicated nowadays.

Potential explanations for these modest results must be considered. From a statistical point of view, technical problems such as low sample size, the number of genes included in the classifier, translation between platforms or cohort heterogeneity, among others, have been reported as potential explanations for the lack of clinical translation of genomic classifiers (see references [68], [69], [70]). In our case and for some signatures, only association with prognosis was reported in the original work, thus the authors implicitly recognized poor classification ability. In those profiles that were reported to be highly discriminative, the reason could be a poor control of over-fitting in the training methodology, since external validation was performed only in three studies and the test samples sizes were small (JG08, WN10 and YM06, see Table 1). The need to map probes to genes for signatures that had used different platforms may also have affected the results, since it is known that even multiple probes of the same gene in the same platform may show important variability. We could not detect, however, that platform had a relevant effect in the MCC estimates.

Low availability of information and heterogeneity in clinical data is inherent to the use of public datasets and this is a major impediment for repeatability and integration of published microarray studies [71]. Datasets differ in patient characteristics, inclusion criteria and outcome definitions. A precise and homogeneous definition of the outcome across datasets would be desirable in order to obtain an accurate estimation of the signatures’ prognosis ability. Nevertheless, heterogeneity of datasets allows for a more pragmatic analysis and the estimates should reflect the expected results when profiles were used in real practice, since hospital settings are also heterogeneous. Since different outcomes are supposed to be highly correlated [72], we decided to prioritize a minimum sample size availability to get more precise estimates and avoid uncertainty introduced by datasets with less than 10 events [60]. The requirement of a three years minimum follow up also allowed maximizing sample size and was supported by the literature: it has been described that most of these relapses occur within 3 years after surgery and it is recommended to be used as endpoint in adjuvant clinical trials [73]. Therapy regimen followed by treated patients was not considered, as this information was not available for most of the analyzed datasets.

Patients diagnosed at stage IV were included and considered recurrent events to assess global performance. This implicitly assumes that the molecular changes playing a driver role to disease relapse remain unchanged in primary tumor after recurrence has occurred. Though this underlying hypothesis could be questionable, it was shared by many of the original studies analyzed that included stage IV subjects in their training and test datasets (AJ08, BT04, EC05, HO09, JS09, LN07G, LN07NZ, PL10, SC09, SL10, SM09, ST09, VL10 and YM06).

From a biological perspective, this moderate prognosis ability could be explained by heterogeneity in tumor cell populations that might dilute the prognosis molecular signal. It is well known that CRC tumors are composed not only by tumor epithelial cells but also by cancer-associated stromal fibroblasts (CAFs), endothelial cells or inflammatory cells, among others [74]. Moreover, those cancer cells at the invasive front are different from those in the main tumor mass [75]. The problem of the tumor bulk heterogeneity can be overcome by isolating specific cells populations by laser microdissection technology [76]. In this regard, one out of the eleven sample sets used in this study (GSE12945) used this technique to specifically hybridize RNA from tumor cells. Surgical specimens from other sample sets were reviewed by a pathologist to assess a minimum tumor content of 80% (GSE5206, GSE18088, E-MEXP-1245). However, we did not observe significant differences in signatures performance regarding the tumor-cell enrichment method used.

The gene lists included in this study had little overlap: out of 1.530 genes reported in the 31 profiles, only two were shared by four signatures; 10 were shared by three signatures and 102 were present in two profiles. This result was not unexpected, since it has previously been reported [77], [78]. The lack of gene overlap is generally interpreted as if each signature is random sampling of a small subset of genes from a larger signature that represent the involved pathways [79], [80].

Colon and rectum tumors have been included indiscriminately in this work since in a previous report we showed that no significant differences exist between colon and rectum tumors at transcriptomic level [81]. However, this decision might explain some of the poor performance of the signatures, since it is known that surgery quality is an important prognostic factor in rectal cancer and less important in colon [82]. In the data used in this work, no significant association was found between prognosis and tumor location (data not shown).

The choice of the statistical tools for analyses was an important matter. The intention of this analysis was to test the performance of published prognostic signatures in independent datasets rather than trying to reproduce them using the original methodology. In this context, Random Forest arises as an efficient method that performs very well compared with other competitors [49], [83]. As expected, the signatures tested in their training dataset showed the highest accuracy. Moreover, we succeeded in reproducing the validation results of three out of the five signatures for which data was available (SM09, VL10 and JS09). However, association with prognosis was not observed for profile ST09 in dataset GSE12945, and it was only observed in one out of the three independent validation datasets that are included in this work for profile SL10, although good performance was originally reported (see Table 1, Figure 2 and File S5). A reason could be that the methodology we used does not capture well the prognosis value of some signatures, which might have been developed with more elaborated algorithms to define the risk prediction in the original study. Because this was a recognized limitation of this work, analyses were redone using an alternative methodology (SVM), which provided similar results (Figures S2, S5 and S6, File S7). In ST09 profile, a semi-supervised approach was used while in SL10 a nearest-centroid approach was applied which was not properly described in the paper. SL10 was developed in an Agilent platform and the mapping of the probes to different validation platforms used in the datasets might be an addition source of divergence.

Although some works reported that simpler methods for supervised learning in the context of high-dimensional molecular data could perform equally than those used in this paper which are more elaborated [84], we chose RF and SVM because they are reported to be robust to over-fitting and the presence of noise, and they capable to learn complex classification functions. These properties are especially desirable in our study as we try to capture the hypothetical prediction ability of signatures created with very heterogeneous methodologies [49], [50], [52], [53]. So, our choice of these methodologies reflects our efforts in finding the prognosis information reported in the original works, though we may have failed in some complex signatures.

It is worth noting that two of the analyzed signatures correspond to current available commercial test for CRC prognosis. Oncotype DX was derived from OC10 profile [85] and, interestingly, the reported risk estimations for strata in stage II in their validation study [43] were similar to those obtained in our work (PPTpr = 25% and 22%, NPTpr = 12% and 16% respectively). The algorithm for risk estimation with Oncotype DX implies the use of additional clinical information as tumor extent and mismatch repair status, which substantially improves its risk stratification. To our knowledge, no validation results for stage III patients have been published yet. Coloprint test was derived from SL10, which showed a low performance in our analysis, possibly for the reasons discussed above.

The characteristics of the available test datasets could be other reason of poor performance. Intriguingly, in some datasets (e.g. GSE17537) the performance of signatures was better than for others. This effect was not due to sample size neither tumor cell enrichment: Datasets with the largest number of events (GSE14333 and GSE13294) were not well classified by any of the tested signatures, and datasets with high tumor cell content showed uneven performance (e.g. GSE12945, GSE5206).

Conclusions

Although most of the published signatures of prognosis in CRC tested in this analysis have shown significant statistical association with prognosis, their ability to accurately classify independent samples into high-risk and low-risk groups is limited. Thus, even when prognosis differences exist in expression data, higher accuracy is needed to consider a signature useful for the clinical practice. Well-designed studies, with large sample size, and preferably prospective are needed to accurately identify those patients at risk of recurrence, especially among patients with stage II CRC tumors.

Supporting Information

Figure S1.

Boxplots showing signatures’ MCC values in each dataset and pooled MCC. Dataset GSE2630 was excluded from pooled analysis due to low sample size.

https://doi.org/10.1371/journal.pone.0048877.s001

(PDF)

Figure S2.

Heatmap showing Matthews Correlation Coefficient values (MCC) for each signature in each dataset as result of analyses with Support Vector Machine. Rows correspond to signatures and columns to datasets. Last column shows a pooled MCC across datasets using sample size as weights. Black lines delimit the first five signatures for which training datasets were available (cells highlighted in black). Cells representing signatures and datasets used to validate them are highlighted in blue. Color scale represents the MCC values: the darker the color, the higher MCC (see the legend). Negative values were collapsed to zero.

https://doi.org/10.1371/journal.pone.0048877.s002

(PDF)

Figure S3.

Heatmap showing Matthews Correlation Coefficient (MCC) in stage II tumors as result of analyses with Random Forest. Empty columns are placed in case of no available data and datasets with less than 10 events, which were excluded from analyses.

https://doi.org/10.1371/journal.pone.0048877.s003

(PDF)

Figure S4.

Heatmap showing Matthews Correlation Coefficient (MCC) in stage III tumors as result of analyses with Random Forest. Empty columns are placed in case of no available data and datasets with less than 10 events, which were excluded from analyses.

https://doi.org/10.1371/journal.pone.0048877.s004

(PDF)

Figure S5.

Heatmap showing Matthews Correlation Coefficient (MCC) in stage II tumors as result of analyses with Support Vector Machine. Empty columns are placed in case of no available data and datasets with less than 10 events, which were excluded from analyses.

https://doi.org/10.1371/journal.pone.0048877.s005

(PDF)

Figure S6.

Heatmap showing Matthews Correlation Coefficient (MCC) in stage III tumors as result of analyses with Support Vector Machine. Empty columns are placed in case of no available data and datasets with less than 10 events, which were excluded from analyses.

https://doi.org/10.1371/journal.pone.0048877.s006

(PDF)

Figure S7.

Example of outcome association in stage II samples using disease free survival information: Kaplan-Meier estimates for risk groups predicted by signature YM06 in GSE13294 dataset (Random Forest results).

https://doi.org/10.1371/journal.pone.0048877.s007

(PDF)

Figure S8.

Example of outcome association in stage III samples using disease free survival information: Kaplan-Meier estimates for risk groups predicted by signature AJ08 in GSE14333 dataset (Random Forest results).

https://doi.org/10.1371/journal.pone.0048877.s008

(PDF)

Table S1.

Excluded papers by eligibility criteria in the literature review.

https://doi.org/10.1371/journal.pone.0048877.s009

(PDF)

File S1.

Details on the prognosis signatures studies systematic review containing inclusion and exclusion criteria at each step.

https://doi.org/10.1371/journal.pone.0048877.s010

(PDF)

File S2.

The 27 PRISMA checklist items corresponding to the prognosis signatures studies systematic review.

https://doi.org/10.1371/journal.pone.0048877.s011

(PDF)

File S3.

Signatures translation results. Translation results for each signature to the platforms of public datasets used in this work: Affymetryx, Hs-OperonV2-vB2.2, Human 19 K Oligo array, MWG 30 K Oligo set and Rosetta custom human 23 K array. Translation was performed via Gene Symbol when necessary, using the Universal Protein Resource annotation database, the online repository of HUGO Gene Nomenclature Committee and the chip annotation files from the Affymetrix official web site. Signature: signature name; Platform: platform used to derive the signature; Reported size: size of signature reported in the original paper (genes or features); Extracted size: size of signature after extraction from the original paper (genes or features); Gene Symbols: size of signature in terms of official Gene Symbol when translation was possible; Not found: number of signature features not found in the platform; % not found: percentage of signature features not found in the platform (respect to extracted size); Platform features: signature size in the platform after translation. Signatures are listed in decreasing order of % not found.

https://doi.org/10.1371/journal.pone.0048877.s012

(XLS)

File S4.

Signatures official Gene Symbols and overlapping. Signatures Gene Symbol: signatures in terms of Gene Symbol. For each signature, official Gene Symbols to which some of their original feature was translated are listed. Translation was performed using the Universal Protein Resource annotation database, the online repository of HUGO Gene Nomenclature Committee and the chip annotation files from the Affymetrix official web site. Signatures overlap: official Gene Symbols shared by four, three and two of the signatures used in this work. List of no shared Gene Symbols (Genes in 1 signatures) is also shown.

https://doi.org/10.1371/journal.pone.0048877.s013

(XLS)

File S5.

Random Forest classification results. Random Forest classification results for each signature and dataset are shown for all, stage II, stage III, Microsatellite Stable and Microsatellite instable samples analyses. MCC: Matthews Correlation Coefficient (MCC) and 95% confidence interval; p-value: permutation p-value associated with MCC; Acc: accuracy rate; Sens: sensitivity; Spec: specificity; No events/events: number of samples with good and bad prognosis respectively. Last column shows the same values for the pooled analyses across datasets using sample size as weights.

https://doi.org/10.1371/journal.pone.0048877.s014

(XLS)

File S6.

Global performance of signatures for all, stage II and stage III samples using Random Forest classifier. For each signature, sample size used in the analysis (separately for good and bad prognosis between brackets), pooled Matthews Correlation Coefficients (MCC) with 95% confidence intervals, accuracy rates, sensitivities and specificities are shown. For stage II and stage III analyses, also positive and negative likelihood ratios, negative and positive post-test probabilities, differences between post-test probabilities and 95% confidence interval are reported. Signatures are listed in decreasing order of MCC. Those with significant MCC at 5% level are highlighted in bold letters.

https://doi.org/10.1371/journal.pone.0048877.s015

(XLS)

File S7.

Support Vector Machine classification results. Sheets All samples, Stage 2 and Stage 3: Support Vector Machine classification results for each signature and dataset are shown for all, stage II and, stage III analyses sheets respectively. MCC: Matthews Correlation Coefficient (MCC) and 95% confidence interval; p-value: permutation p-value associated with MCC; Acc: accuracy rate; Sens: sensitivity; Spec: specificity; No events/events: number of samples with good and bad prognosis respectively in that dataset. Sheets All samples global, Stage 2 global and Stage 3 global: Global performance results of signatures for all, stage II and stage III samples using Support Vector Machine are extended. Signatures are listed in decreasing order of MCC. Those with significant MCC at 5% level are highlighted in bold letters.

https://doi.org/10.1371/journal.pone.0048877.s016

(XLS)

Author Contributions

Conceived and designed the experiments: RSP AB CF VM. Analyzed the data: RSP AB DC SR X. Solé MCB EG X. Sanjuan SB GJ AS CF GC VM. Wrote the paper: RSP AB SR CF VM.

References

  1. 1. Ferlay J, Parkin DM, Steliarova-Foucher E (2010) Estimates of cancer incidence and mortality in Europe in 2008. Eur J Cancer 46(4): 765–81.
  2. 2. Saltz LB, Kelsen DP (1997) Adjuvant treatment of colorectal cancer. Annu Rev Med 48: 191–202.
  3. 3. Benson AB 3rd, Schrag D, Somerfield MR, Cohen AM, Figueredo AT, et al (2004) American Society of Clinical Oncology recommendations on adjuvant chemotherapy for stage II colon cancer. J Clin Oncol 22(16): 3408–19.
  4. 4. Gill S, Loprinizi CL, Sargent DJ, Thome SD, Alberts SR, et al. (2004) Pooled analysis of fluorouracil-based adjuvant therapy for stage II and III colon cancer: who benefits and by how much?. J Clin Oncol 22(10): 1797–806.
  5. 5. McMillan DC, McArdle CS, Morrison DS (2010) A clinical risk score to predict 3-, 5- and 10-year survival in patients undergoing surgery for Dukes B colorectal cancer. Br J Cancer. 103(7): 970–4.
  6. 6. Dotan E, Cohen SJ (2011) Challenges in the management of stage II colon cancer. Semin Oncol 38(4): 511–20.
  7. 7. Portera CA Jr, Berman RS, Ellis LM (1998) Molecular determinants of colon cancer metastasis. Surg Oncol 7(3–4): 183–95.
  8. 8. Jones S, Chen WD, Parmigiani G, Diehl F, Beerenwinkel N, et al. (2008) Comparative lesion sequencing provides insights into tumor evolution. Proc Natl Acad Sci U S A 105(11): 4283–8.
  9. 9. George B, Kopetz S (2011) Predictive and prognostic markers in colorectal cancer. Curr Oncol Rep 13(3): 206–15.
  10. 10. Cowin PA, Anglesio M, Etemadmoghadam D, Bowtell DD (2010) Profiling the cancer genome. Annu Rev Genomics Hum Genet 11: 133–59.
  11. 11. Kim C, Paik S (2010) Gene-expression-based prognostic assays for breast cancer. Nat Rev Clin Oncol 7(6): 340–7.
  12. 12. Slodkowska EA, Ross JS (2009) MammaPrint 70-gene signature: another milestone in personalized medical care for breast cancer patients. Expert Rev Mol Diagn 9(5): 417–22.
  13. 13. Moher D, Liberati A, Tetzlaff J (2009) Altman DG; PRISMA Group (2009) Preferred reporting items for systematic reviews and meta-analyses: the PRISMA statement. PLoS Med 21 6(7): e1000097.
  14. 14. Andersen CL, Christensen LL, Thorsen K, Schepeler T, Sorensen FB, et al. (2009) Dysregulation of the transcription factors SOX4, CBFB and SMARCC1 correlates with outcome of colorectal cancer. Br J Cancer 100(3): 511–23.
  15. 15. Anjomshoaa A, Lin YH, Black MA, McCall JL, Humar B, et al. (2008) Reduced expression of a gene proliferation signature is associated with enhanced malignancy in colon cancer. Br J Cancer 99(6): 966–73.
  16. 16. Arango D, Laiho P, Kokko A, Alhopuro P, Sammalkorpi H, et al. (2005) Gene-expression profiling predicts recurrence in Dukes’ C colorectal cancer. Gastroenterology 129(3): 874–84.
  17. 17. Bandres E, Malumbres R, Cubedo E, Honorato B, Zarate R, et al. (2007) A gene signature of 8 genes could identify the risk of recurrence and progression in Dukes’ B colon cancer patients. Oncol Rep 17(5): 1089–94.
  18. 18. Barrier A, Boelle PY, Roser F, Gregg J, Tse C, et al. (2006) Stage II colon cancer prognosis prediction by tumor gene expression profiling.. J Clin Oncol 24(29): 4685–91.
  19. 19. Barrier A, Lemoine A, Boelle PY, Tse C, Brault D, et al. (2005) Colon cancer prognosis prediction by gene expression profiling. Oncogene 24(40): 6155–64.
  20. 20. Bertucci F, Salas S, Eysteries S, Nasser V, Finetti P, et al. (2004) Gene expression profiling of colon cancer by DNA microarrays and correlation with histoclinical parameters. Oncogene 23(7): 1377–91.
  21. 21. Eschrich S, Yang I, Bloom G, Kwong KY, Boulware D, et al. (2005) Molecular staging for survival prediction of colorectal cancer patients. J Clin Oncol 23(15): 3526–35.
  22. 22. Hao JM, Chen JZ, Sui HM, Si-Ma XQ, Li GQ, et al. (2010) Five-gene signature as a potential predictor of metastasis and survival in colorectal cancer. J Pathol 220(4): 475–89.
  23. 23. Jiang Y, Casey G, Lavery IC, Zhang Y, Talantov D, et al. (2008) Development of a clinically feasible molecular assay to predict recurrence of stage II colon cancer. J Mol Diagn 10(4): 346–54.
  24. 24. Jorissen RN, Gibbs P, Christie M, Prakash S, Lipton L, et al. (2009) Metastasis-Associated Gene Expression Changes Predict Poor Outcomes in Patients with Dukes Stage B and C Colorectal Cancer. Clin Cancer Res 15(24): 7642–7651.
  25. 25. Kalady MF, Dejulius K, Church JM, Lavery IC, Fazio VW (2010) Gene signature is associated with early stage rectal cancer recurrence. J Am Coll Surg 211(2): 187–95.
  26. 26. Kennedy RD, Bylesjo M, Kerr P, Davison T, Black JM, et al. (2011) Development and Independent Validation of a Prognostic Assay for Stage II Colon Cancer Using Formalin-Fixed Paraffin-Embedded Tissue. J Clin Oncol 29(35): 4620–6.
  27. 27. Liersch T, Grade M, Gaedcke J, Varma S, Difilippantonio MJ, et al. (2009) Preoperative chemoradiotherapy in locally advanced rectal cancer: correlation of a gene expression-based response signature with recurrence. Cancer Genet Cytogenet. 190(2): 57–65.
  28. 28. Lin YH, Friederichs J, Black MA, Mages J, Rosenberg R, et al. (2007) Multiple gene expression classifiers from different array platforms predict poor prognosis of colorectal cancer. Clin Cancer Res 13(2 Pt 1): 498–507.
  29. 29. Mettu RK, Wan YW, Habermann JK, Ried T, Guo NL (2010) A 12-gene genomic instability signature predicts clinical outcomes in multiple cancer types. Int J Biol Markers 25(4): 219–28.
  30. 30. O’Connell MJ, Lavery I, Yothers G, Paik S, Clark-Langone KM, et al. (2010) Relationship between tumor gene expression and recurrence in four independent studies of patients with stage II/III colon cancer treated with surgery alone or surgery plus adjuvant fluorouracil plus leucovorin. J Clin Oncol 28(25): 3937–44.
  31. 31. Peng J, Wang Z, Chen W, Ding Y, Wang H, et al. (2010) Integration of genetic signature and TNM staging system for predicting the relapse of locally advanced colorectal cancer. Int J Colorectal Dis 25(11): 1277–85.
  32. 32. Pillaire MJ, Selves J, Gordien K, Gourraud PA, Gentil C, et al. (2010) A ‘DNA replication’ signature of progression and negative outcome in colorectal cancer. Oncogene 29(6): 876–87.
  33. 33. Salazar R, Roepman P, Capella G, Moreno V, Simon I, et al. (2011) Gene expression signature to improve prognosis prediction of stage II and III colorectal cancer. J Clin Oncol 29(1): 17–24.
  34. 34. Schetter AJ, Nguyen GH, Bowman ED, Mathe EA, Yuen ST, et al. (2009) Association of inflammation-related and microRNA gene expression with cancer-specific mortality of colon adenocarcinoma. Clin Cancer Res15(18): 5878–87.
  35. 35. Smith JJ, Deane NG, Wu F, Merchant NB, Zhang B, et al. (2010) Experimentally derived metastasis gene expression profile predicts recurrence and death in patients with colon cancer. Gastroenterology 138(3): 958–68.
  36. 36. Staub E, Groene J, Heinze M, Mennerich D, Roepcke S, et al. (2009) An expression module of WIPF1-coexpressed genes identifies patients with favorable prognosis in three tumor types. J Mol Med (Berl) 87(6): 633–44.
  37. 37. Van Laar RK (2010) An online gene expression assay for determining adjuvant therapy eligibility in patients with stage 2 or 3 colon cancer. Br J Cancer 103(12): 1852–7.
  38. 38. Wan YW, Quian Y, Rathanagiriswaran S, Castranova V, Guo NL (2010) A breast cancer prognostic signature predicts clinical outcomes in multiple tumor types. Oncol Rep 24(2): 489–94.
  39. 39. Wang Y, Jatkoe T, Zhang Y, Mutch MG, Talantov D, et al. (2004) Gene expression profiles and molecular markers to predict recurrence of Dukes’ B colon cancer. J Clin Oncol 22(9): 1564–71.
  40. 40. Watanabe T, Kobunai T, Sakamoto E, Yamamoto Y, Konishi T, et al. (2009) Gene expression signature for recurrence in stage III colorectal cancers. Cancer 115(2): 283–92.
  41. 41. Watanabe T, Kobunai T, Yamamoto Y, Kanazawa T, Konishi T, et al. (2010) Prediction of liver metastasis after colorectal cancer using reverse transcription-polymerase chain reaction analysis of 10 genes. Eur J Cancer. 46(11): 2119–26.
  42. 42. Yamasaki M, Takemasa I, Komori T, Watanabe S, Sekimoto M, et al. (2007) The gene expression profile represents the molecular nature of liver metastasis in colorectal cancer. Int J Oncol 30(1): 129–38.
  43. 43. Gray RG, Quirke P, Handley K, Lopatin M, Magill L, et al. (2011) Validation study of a quantitative multigene reverse transcriptase-polymerase chain reaction assay for assessment of recurrence risk in patients with stage II colon cancer. J Clin Oncol 10 29(35): 4611–9.
  44. 44. Barrett T, Edgar R (2006) Gene expression omnibus: microarray data storage, submission, retrieval, and analysis. Methods Enzymol 411: 352–69.
  45. 45. Parkinson H, Sarkans U, Shojatalab M, Abeygunawardena N, Contrino S, et al.. (2005) ArrayExpress–a public repository for microarray gene expression data at the EBI. Nucleic Acids Res 33(Database issue): D553–5.
  46. 46. UniProt Consortium (2011) Ongoing and future developments at the Universal Protein Resource. Nucleic Acids Res 39(Database issue): D214–9.
  47. 47. Povey S, Lovering R, Bruford E, Wright M, Lush M, et al. (2001) The HUGO Gene Nomenclature Committee (HGNC). Hum Genet 109(6): 678–80.
  48. 48. Liu G, Loraine AE, Shigeta R, Cline M, Cheng J, et al. (2003) NetAffx: Affymetrix probesets and annotations. Nucleic Acids Res 31(1): 82–6.
  49. 49. Breiman L (2001) Random Forest. Machine Learning 45(1): 5–32.
  50. 50. Breiman L (2002) Manual On Setting Up, Using, And Understanding Random Forests V3.1. Available: http://oz.berkeley.edu/users/breiman/Using_random_forests_V3.1.pdf.
  51. 51. Mitchell MW (2011) Bias of the Random Forest Out-of-Bag (OOB) Error for Certain Input Parameters. Open Journal of Statistics doi:10.4236/ojs.2011.13024.
  52. 52. Liaw A, Wiene M (2002) Classification and regression by randomForest. R News 2(3): 18–22.
  53. 53. Hastie T, Tibshirani R, Friedman J (2008) The Elements of Statistical Learning (2nd edition). Springer-Verlag.
  54. 54. Vapnik V (1998) Statistical Learning Theory. Wiley, New York.
  55. 55. Liu XY, Wu J, Zhou ZH (2009) Exploratory undersampling for class-imbalance learning. IEEE Trans Syst Man Cybern B Cybern 39(2): 539–50.
  56. 56. Hastie T (2009) svmpath: the SVM Path algorithm. R package version 0.93. Available: http://CRAN.R-project.org/package=svmpath.
  57. 57. Baldi P, Brunak S, Chauvin Y, Andersen CA, Nielsen H (2000) Assessing the accuracy of prediction algorithms for classification: an overview. Bioinformatics 16(5): 412–24.
  58. 58. Shi L, Campbell G, Jones WD, Campagne F, Wen Z, et al. (2010) The MicroArray Quality Control (MAQC)-II study of common practices for the development and validation of microarray-based predictive models. Nat Biotechnol 28(8): 827–38.
  59. 59. Zhou XH, Obuchowsky NA, McClish D (2002) Statistical Mehods in diagnostic Medicine. Wiley.
  60. 60. Braga-Neto UM, Dougherty ER (2004) Is cross-validation valid for small-sample microarray classification? Bioinformatics 12 20(3): 374–80.
  61. 61. Strasser H, Weber C (1999) On the asymptotic theory of permutation statistics. Mathematical Methods of Statistics 8, 220–250.
  62. 62. Hothorn T, Hornik K, van de Wiel MA, Zeileis A (2008) Implementing a class of permutation test: the coin package. Journal of Statistical Software 28(8),1–23.
  63. 63. Efron B, Tibshirani R (1993) An Introduction to the Bootstrap. Chapman & Hall.
  64. 64. Canty A, Ripley B (2011) boot: Bootstrap R (S-Plus) Functions. R package version 1.3–2.
  65. 65. Lusa L, McShane LM, Radmacher MD, Shih JH, Wright GW, et al. (2007) Appropriateness of some resampling-based inference procedures for assessing performance of prognostic classifiers derived from microarray data. Stat Med 28 26(5): 1102–13.
  66. 66. Gray R, Barnwell J, McConkey C, Hills RK, Williams NS, et al. (2007) Adjuvant chemotherapy versus observation in patients with colorectal cancer: a randomised study. Lancet 370(9604): 2020–9.
  67. 67. Lu AT, Salpeter SR, Reeve AE, Escrich S, Johnston PG, et al. (2009) Gene expression profiles as predictors of poor outcomes in stage II colorectal cancer: A systematic review and meta-analysis. Clin Colorectal Cancer 8(4): 207–14.
  68. 68. Simon R (2005) Roadmap for developing and validating therapeutically relevant genomic classifiers. J Clin Oncol 10 23(29): 7332–41.
  69. 69. Dupuy A, Simon RM (2007) Critical review of published microarray studies for cancer outcome and guidelines on statistical analysis and reporting. J Natl Cancer Inst 17 99(2): 147–57.
  70. 70. Raspe E, Decraene C, Berx G (2012) Gene expression profiling to dissect the complexity of cancer biology: Pitfalls and promise. Semin Cancer Biol. Mar 7.
  71. 71. Ioannidis JP, Allison DB, Ball CA, Coulibaly I, Cui X, et al. (2009) Repeatability of published microarray gene expression analyses. Nat Genet. 2009 41(2): 149–55.
  72. 72. Sargent DJ, Wieand HS, Haller DG, Gray R, Benedetti JK, et al. (2005) Disease-free survival versus overall survival as a primary end point for adjuvant colon cancer studies: individual patient data from 20,898 patients on 18 randomized trials. J Clin Oncol 1 23(34): 8664–70.
  73. 73. Sargent DJ, Patiyil S, Yothers G, Haller DG, Gray R, et al. (2007) End points for colon cancer adjuvant trials: observations and recommendations based on individual patient data from 20,898 patients enrolled onto 18 randomized trials from the ACCENT Group. J Clin Oncol 10 25(29): 4569–74.
  74. 74. Hanahan D, Weinberg RA (2000) The hallmarks of cancer. Cell 7 100(1): 57–70.
  75. 75. Barry WT, Kernagis DN, Dressman HK, Griffis RJ, Hunter JD, et al. (2010) Intratumor heterogeneity and precision of microarray-based predictors of breast cancer biology and clinical outcome. J Clin Oncol 1 28(13): 2198–206.
  76. 76. Esposito G (2007) Complementary techniques: laser capture microdissection–increasing specificity of gene expression profiling of cancer specimens. Adv Exp Med Biol 593: 54–65.
  77. 77. Cardoso J, Boer J, Morreau H, Fodde R (2007) Expression and genomic profiling of colorectal cancer. Biochim Biophys Acta 1775(1): 103–37.
  78. 78. Abba MC, Lacunza E, Butti M, Aldaz CM (2010) Breast cancer biomarker discovery in the functional genomic age: a systematic review of 42 gene expression signatures. Biomark Insights 27 5: 103–18.
  79. 79. Lascorz J, Chen B, Hemminki K, Forsti A (2011) Consensus pathways implicated in prognosis of colorectal cancer identified through systematic enrichment analysis of gene expression profiling studies. PLoS One 6(4): e18867.
  80. 80. Sole X, Bonifaci N, Lopez-Bigas N, Berenguer A, Hernandez P, et al.. (2009) Biological convergence of cancer signatures. PLoS One 4(2): p. e4544.
  81. 81. Sanz-Pamplona R, Cordero D, Berenguer A, Lejbkowicz F, Rennert H, et al. (2011) Gene expression differences between colon and rectum tumors. Clin Cancer Res 1 17(23): 7303–12.
  82. 82. Syk E, Glimelius B, Nilsson PJ (2010) Factors influencing local failure in rectal cancer: analysis of 2315 patients from a population-based series. Dis Colon Rectum 53(5): 744–52.
  83. 83. Diaz-Uriarte R, Alvarez de Andres S (2006) Gene selection and classification of microarray data using random forest. BMC Bioinformatics 7: 3.
  84. 84. Dudoit S, Fridlyand J, Speed TP (2002) Comparison of discrimination methods for classification of tumors using gene expression data. J Am Stat Assoc 97: 77–87.
  85. 85. Webber EM, Lin JS, Evelyn P Whitlock (2010) Oncotype DX tumor gene expression profiling in stage II colon cancer. Application: prognostic, risk prediction. PLoS Curr 2;2.