ALL Metrics
-
Views
-
Downloads
Get PDF
Get XML
Cite
Export
Track
Research Note
Revised

Predicting Outcomes of Hormone and Chemotherapy in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) Study by Biochemically-inspired Machine Learning

[version 3; peer review: 2 approved]
PUBLISHED 12 May 2017
Author details Author details
OPEN PEER REVIEW
REVIEWER STATUS

This article is included in the Bioinformatics gateway.

This article is included in the Machine learning: life sciences collection.

Abstract

Genomic aberrations and gene expression-defined subtypes in the large METABRIC patient cohort have been used to stratify and predict survival. The present study used normalized gene expression signatures of paclitaxel drug response to predict outcome for different survival times in METABRIC patients receiving hormone (HT) and, in some cases, chemotherapy (CT) agents. This machine learning method, which distinguishes sensitivity vs. resistance in breast cancer cell lines and validates predictions in patients; was also used to derive gene signatures of other HT  (tamoxifen) and CT agents (methotrexate, epirubicin, doxorubicin, and 5-fluorouracil) used in METABRIC. Paclitaxel gene signatures exhibited the best performance, however the other agents also predicted survival with acceptable accuracies. A support vector machine (SVM) model of paclitaxel response containing genes ABCB1, ABCB11, ABCC1, ABCC10, BAD, BBC3, BCL2, BCL2L1, BMF, CYP2C8, CYP3A4, MAP2, MAP4, MAPT, NR1I2, SLCO1B3, TUBB1, TUBB4A, and TUBB4B was 78.6% accurate in predicting survival of 84 patients treated with both HT and CT (median survival ≥ 4.4 yr). Accuracy was lower (73.4%) in 304 untreated patients. The performance of other machine learning approaches was also evaluated at different survival thresholds. Minimum redundancy maximum relevance feature selection of a paclitaxel-based SVM classifier based on expression of genes BCL2L1, BBC3, FGF2, FN1, and TWIST1 was 81.1% accurate in 53 CT patients. In addition, a random forest (RF) classifier using a gene signature (ABCB1, ABCB11, ABCC1, ABCC10, BAD, BBC3, BCL2, BCL2L1, BMF, CYP2C8, CYP3A4, MAP2, MAP4, MAPT, NR1I2,SLCO1B3, TUBB1, TUBB4A, and TUBB4B) predicted >3-year survival with 85.5% accuracy in 420 HT patients. A similar RF gene signature showed 82.7% accuracy in 504 patients treated with CT and/or HT. These results suggest that tumor gene expression signatures refined by machine learning techniques can be useful for predicting survival after drug therapies.

Keywords

Gene expression signatures, breast cancer, chemotherapy resistance, hormone therapy, machine learning, support vector machine, random forest

Revised Amendments from Version 2

We have addressed the reviewers' comments regarding overfitting by 1) deriving and validation biochemically inspired machine learning models using the METABRIC Validation patient dataset independently of the Discovery data and 2) assessing the accuracy of the Discovery dataset-based models with patient data derived from an independent source (reference 5). In addition, we have stratified the patients by breast cancer subtype and evaluated each subtype with the combined Discovery+Validation dataset-based models using all of the feature selection methods (Supplementary File 1).

See the authors' detailed response to the review by Elana J. Fertig
See the authors' detailed response to the review by Chun-Wei Tung

Introduction

Current pharmacogenetic analysis of chemotherapy makes qualitative decisions about drug efficacy in patients (determination of good, intermediate or poor metabolizer phenotypes) based on variants present in genes involved in the transport, biotransformation, or disposition of a drug. We have applied a supervised machine learning (ML) approach to derive accurate gene signatures, based on the biochemically-guided response to chemotherapies with breast cancer cell lines1, which show variable responses to growth inhibition by paclitaxel and gemcitabine therapies2,3. We analyzed stable4 and linked unstable genes in pathways that determine their disposition. This involved investigating the correspondence between 50% growth inhibitory concentrations (GI50) of paclitaxel and gemcitabine and gene copy number, mutation, and expression first in breast cancer cell lines and then in patients1. Genes encoding direct targets of these drugs, metabolizing enzymes, transporters, and those previously associated with chemo-resistance to paclitaxel (n=31 genes) were then pruned by multiple factor analysis (MFA), which indicated that expression levels of genes ABCC10, BCL2, BCL2L1, BIRC5, BMF, FGF2, FN1, MAP4, MAPT, NKFB2, SLCO1B3, TLR6, TMEM243, TWIST1, and CSAG2 could predict sensitivity in breast cancer cell lines with 84% accuracy. The cell line-based paclitaxel-gene signature predicted sensitivity in 84% of patients with no or minimal residual disease (n=56; data from 5). The present study derives related gene signatures with ML approaches that predict outcome of hormone- and chemotherapies in the large METABRIC breast cancer cohort6.

Methods

SVM (Support Vector Machine) learning: Previously, paclitaxel-related response genes were identified from peer-reviewed literature, and their expression and copy number in breast cancer cell lines were analyzed by multiple factor analysis of GI50 values of these lines2 (Figure 1). Given the expression levels of each gene, a SVM is evaluated on patients by classifying those with shorter survival time as resistant and longer survival as sensitive to hormone and/or chemotherapy using paclitaxel, tamoxifen, methotrexate, 5-fluorouracil, epirubicin, and doxorubicin. The SVM was trained using the function fitcsvm in MATLAB R2014a7 and tested with either leave-one-out or 9 fold cross-validation (indicated in Table 1). The Gaussian kernel was used for this study, unlike Dorman et al.1 which used the linear kernel. The SVM requires selection of two different parameters, C (misclassification cost) and sigma (which controls the flexibility and smoothness of Gaussians)8; these parameters determine how strictly the SVM learns the training set, and hence if not selected properly, can lead to overfitting. A grid search evaluates a wide range of combinations of these values by parallelization. A Gaussian kernel selects the C and sigma combination that lead to the lowest cross-validation misclassification rate. A backwards feature selection (greedy) algorithm was designed and implemented in MATLAB in which one gene of the set is left out in a reduced gene set and the classification is then assessed; genes that maintain or lower the misclassification rate are kept in the signature. The procedure is repeated until the subset with the lowest misclassification rate is selected as the optimal subset of genes. These SVMs were then assessed for their ability to predict patient outcomes based on available metadata (see Figure 1 and reference 1). Interactive prediction using normalized expression values as input is available at http://chemotherapy.cytognomix.com.

ea764b20-98b8-4220-99ba-ff2a348add06_figure1.gif

Figure 1. Biochemically-inspired SVM gene signature derivation workflow.

The initial set of genes is carefully selected through the understanding of the drug and the pathways associated with it. A multiple factor analysis of the GI50 values of a training set of breast cancer cell lines and the corresponding expression levels of each gene in the initial set reduces the list of genes.

Table 1. SVM gene expression signature performance on METABRIC patients.

Patient
treatment
# of patientsAgent:
final gene
signature (C
and sigma)
Accuracy (%)PrecisionF-MeasureMCC1AUC2
Both CT
and HT3
84Paclitaxel: ABCC1, ABCC10, BAD,
BIRC5, FN1, GBP1, MAPT, SLCO1B3,
TMEM243, TUBB3, TUBB4B
(C=10000, σ=10)
78.60.7870.7820.5590.814
Tamoxifen: ABCC2, ALB, CCNA2,
E2F7, FLAD1, FMO1, NCOA2, NR1I2,
PIAS4, SULT1E1 (C=100000, σ=100)
76.20.7610.7600.5100.701
Methotrexate: ABCC2, ABCG2,
CDK2, DHFRL1 (C=10, σ=1)
71.40.7120.7110.4100.766
Epirubicin: ABCB1, CDA, CYP1B1,
ERBB3, ERCC1, MTHFR, PON1,
SEMA4D, TFDP2 (C=1000, σ=10)
72.60.7250.7230.4340.686
Doxorubicin: ABCC2, ABCD3, CBR1,
FTH1, GPX1, NCF4, RAC2, TXNRD1
(C=100000, σ=100)
75.00.7490.7500.4880.701
5-Fluorouracil: ABCB1, ABCC3,
MTHFR, TP53 (C=10000, σ=100)
71.40.7140.7140.4170.718
CT and/or
HT3,4,5,6
735Paclitaxel: BAD, BCAP29, BCL2,
BMF, CNGA3, CYP2C8, CYP3A4,
FGF2, FN1, NFKB2, NR1I2, OPRK1,
SLCO1B3, TLR6, TUBB1, TUBB3,
TUBB4A, TUBB4B, TWIST1
(C=10000, σ=100)
66.10.6520.6430.2870.660
Deceased
only2,6,7
(CT and/or
HT)
327Paclitaxel: ABCB11, BAD, BBC3,
BCL2, BCL2L1, BIRC5, CYP2C8,
FGF2, FN1, GBP1, MAPT, NFKB2,
OPRK1, SLCO1B3, TMEM243
(C=100, σ=10)
75.30.7520.7520.5050.763
No
treatment3
304Paclitaxel: ABCB1, ABCB11, BBC3,
BCL2L1, BMF, CYP3A4, FGF2,
GBP1, MAP4, MAPT, NR1I2, OPRK1,
SLCO1B3, TUBB4A, TUBB4B,
TWIST2 (C=100, σ=10)
73.40.7340.7330.4670.769

Initial gene sets preceding feature selection: Paclitaxel - ABCB1, ABCB11, ABCC1, ABCC10, BAD, BBC3, BCAP29, BCL2, BCL2L1, BIRC5, BMF, CNGA3, CYP2C8, CYP3A4, FGF2, FN1, GBP1, MAP2, MAP4, MAPT, NFKB2, NR1I2, OPRK1, SLCO1B3, TLR6, TUBB1, TWIST1. Tamoxifen - ABCB1, ABCC2, ALB, C10ORF11, CCNA2, CYP3A4, E2F7, F5, FLAD1, FMO1, IGF1, IGFBP3, IRS2, NCOA2, NR1H4, NR1I2, PIAS4, PPARA, PROC, RXRA, SMARCD3, SULT1B1, SULT1E1, SULT2A1. Methotrexate - ABCB1, ABCC2, ABCG2, CDK18, CDK2, CDK6, CDK8, CENPA, DHFRL1. Epirubicin - ABCB1, CDA, CYP1B1, ERBB3, ERCC1, GSTP1, MTHFR, NOS3, ODC1, PON1, RAD50, SEMA4D, TFDP2. Doxorubicin - ABCB1, ABCC2, ABCD3, AKR1B1, AKR1C1, CBR1, CYBA, FTH1, FTL, GPX1, MT2A, NCF4, RAC2, SLC22A16, TXNRD1. 5-Fluorouracil - ABCB1, ABCC3, CFLAR, IL6, MTHFR, TP53, UCK2. 1MCC: Matthews Correlation Coefficient. 2AUC: Area under receiver operating curve. 3 Surviving patients; 4 Analysis included patients in the METABRIC ‘discovery’ dataset only; 5 SVMs tested with 9 fold cross-validation, all others tested with leave-one-out cross-validation; 6 Includes all patients treated with HT,CT, combination CT/HT, either with or without combination radiotherapy; 7 Median time after treatment until death (> 4.4 years) was used to distinguish favorable outcome, ie. sensitivity to therapy.

RF (Random Forest) learning: RF was trained using the WEKA 3.79 data mining tool. This classifier uses multiple random trees for classification, which are combined via a voting scheme to make a decision on the given input gene set. A grid search was used to optimize the maximum number of randomly selected genes for each tree in RF, where k (maximum number of selected genes for each tree) was set from 1 to 19. Figure 2 depicts the therapy outcome prediction process of a given patient using a RF consisting of a series of decision trees derived from different subsets of paclitaxel-related genes.

ea764b20-98b8-4220-99ba-ff2a348add06_figure2.gif

Figure 2. RF decision tree diagram depicts the therapy outcome prediction process of a given patient, using a RF consisting of k decision trees.

Several DTs are built using different subsets of paclitaxel-related genes. The process starts from the root of each tree and if the expression of the gene corresponding to that node is greater than a specific value, the process continues through the right branch, otherwise it continues through the left branch until it reaches a leaf node; that leaf represents the prediction of the tree for that specific input. The decisions of all trees are considered and the one with the largest number of votes is selected as the patient outcome.

Augmented Gene Selection: The most relevant genes (features) for therapy outcome prediction were found using the Minimum Redundancy and Maximum Relevance (mRMR) approach10. mRMR is a wrapper approach that incrementally selects genes by maximizing the average mutual information between gene expression features and classes, while minimizing their redundancies:

mRMR=maxs[1|s|fiSI(fi,C)1|s|2fi,fjSI(fi,fj)]

where fi corresponds to a feature in gene set S, I(fi,C) is the mutual information between fi and class C, and I(fi,fj) is the mutual information between features fi and fj.

For this experiment, we used a 26-gene signature (genes ABCB1, ABCB11, ABCC1, ABCC10, BAD, BBC3, BCL2, BCL2L1, BMF, CYP2C8, CYP3A4, MAP2, MAP4, MAPT, NR1I2, SLCO1B3, TUBB1, TUBB4A, TUBB4B, FGF2, FN1, GBP1, NFKB2, OPRK1, TLR6, and TWIST1) as the base feature set. These genes were selected (in Dorman et al.1) based either on their known involvement in paclitaxel metabolism, or evidence that their expression levels and/or copy numbers correlate with paclitaxel GI50 values. mRMR and SVM were combined to obtain a subset of genes that can accurately predict patient survival outcomes; here, we considered 3, 4 and 5 years as survival thresholds for breast cancer patients.

Performance was evaluated with several metrics. WEKA determined accuracy (ACC), the weighted average of precision and F-measure, the Matthews Correlation Coefficient (MCC) and the area under ROC curve (AUC).

Results and discussion

Dataset 1.Predicted treatment response for each individual METABRIC patient11.
The predicted and expected response to treatment for each individual METABRIC patient for each analyses listed in Table 1, Table 2 and Table 3 are indexed. Patients sensitive to treatment are labeled with ‘0’ while resistant patients are labeled ‘1’.

Table 2. Results of applying RF to predict outcome of paclitaxel therapy.

Type of treatmentSurvival years (as
threshold)
# PatientsK (number of genes
to be used in
random selection)
Accuracy (True
Positive - TP) (%)
PrecisionF-MeasureMCC1AUC2
Chemotherapy
(CT)
353756.60.5100.524-0.0950.441
4769.80.6980.6980.3960.700
51966.00.6450.6360.2300.653
Hormone therapy
(HT)
34201985.50.7310.7880.0000.606
4978.60.7150.7060.0690.559
5971.00.6340.6270.0590.632
CT and/or HT3504982.70.6850.7490.0000.506
41973.60.6470.6480.0390.527
5765.30.6020.5930.0860.588

1MCC: Matthews Correlation Coefficient. 2AUC: Area under receiver operating curve; both Discovery and Validation patient datasets analyzed. RF predictions done using a gene panel consisting of 19 genes (ABCB1, ABCB11, ABCC1, ABCC10, BAD, BBC3, BCL2, BCL2L1, BMF, CYP2C8, CYP3A4, MAP2, MAP4, MAPT, NR1I2, SLCO1B3, TUBB1, TUBB4A, TUBB4B).

Table 3. Results of mRMR feature selection for an SVM for predicting outcome of paclitaxel therapy.

DataCT1HTCT+HT
Survival years
(as threshold)
345345345
# patients253420504
Accuracy (TP)
(%)
81.181.184.985.779.572.983.174.867.9
Precision0.8090.8130.8520.8780.7650.6920.7950.7030.662
F-Measure0.8090.8110.8450.7940.7260.6630.7720.6720.666
MCC 0.5820.6250.6750.1190.170.1730.1610.1370.238
AUC 0.7830.8120.820.5080.5330.5480.530.5310.61
SVM Par.
(gamma)
0.00.51.01.00.751.50.750.51.0
SVM Par.
(cost)
64128826421622
Selected
genes
MAP4,
GBP1,
FN1,
MAPT,
BBC3,
FGF2,
NFKB2,
TUBB4B
TWIST1,
FN1,
BBC3,
FGF2,
BCL2L1
ABCB11,
BCL2,
GBP1,
SLCO1B3,
ABCB1,
BAD,
TUBB4A,
MAPT,
NFKB2,
TUBB4B
ABCB11,
BCL2,
MAP4,
TUBB1,
GBP1,
SLCO1B3,
ABCB1,
BAD,
TWIST1,
FN1,
TUBB4A,
MAPT,
OPRK1,
BBC3,
FGF2,
NFKB2,
ABCC1,
NR1I2
BAD,
GBP1,
MAPT,
BBC3
ABCB11,
MAP4,
SLCO1B3,
BAD,
FN1,
OPRK1,
BBC3,
NFKB2,
NR1I2,
TUBB4B
ABCB11,
SLCO1B3,
BAD,
TUBB4A,
MAPT,
BBC3,
FGF2,
NFKB2,
ABCC1,
NR1I2
ABCB11,
BMF,
BCL2,
MAP4,
TUBB1,
GBP1,
SLCO1B3,
ABCB1,
BAD,
TWIST1,
FN1,
MAPT,
OPRK1,
BBC3,
FGF2,
NFKB2,
ABCC1,
NR1I2,
TUBB4B
MAP4,
GBP1,
SLCO1B3,
BAD,
MAPT,
OPRK1,
BBC3,
NFKB2,
ABCC1,
NR1I2,
TUBB4B

1For patients treated with CT with ≥4 Yr survival and CT+ HT for ≥ 5 Yr, the cost for the mRMR model was set to 64. Of those treated with CT for ≥ 4 Yr, genes were selected using a greedy, stepwise forward search, while in other cases, greedy stepwise backward search was used. Also, gamma = 0 in all cases. 2Predicted responses for individual METABRIC patients are provided in Dataset 1.

The performances of several ML techniques have been compared such that they distinguish paclitaxel sensitivity and resistance in METABRIC patients using its tumour gene expression datasets. We used mRMR to generate gene signatures and determine which genes are important for treatment response in METABRIC patients. The paclitaxel models are more accurate for prediction of outcomes in patients receiving HT and/or CT compared to other patient groups.

SVMs and RF were trained using expression of genes associated with paclitaxel response, mechanism of action and stable genes in the biological pathways of these targets (Figure 3). Pair-wise comparisons of these genes with those from MammaPrint and Oncotype Dx (other genomic classifiers for breast cancer) find that these signatures are nearly independent of each other, with only a single gene overlap. The distinct differences of these signatures are due to their methodology of derivation, based on different principles and for different purposes (i.e. drug response for a specific reagent). SVM models for drugs used to treat these patients were derived by backwards feature selection on patient subsets stratified by treatment or outcome (Table 1). The highest SVM accuracy was found for the paclitaxel signature in patients treated with HT and/or adjuvant chemotherapy (78.6%). Since some CT patients were also treated with tamoxifen, methotraxate, epirubicin, doxorubicin and 5-fluorouracil, we also evaluated the performance of models developed for these drugs using the same algorithm. These gene signatures also had acceptable performance (accuracies between 71–76%; AUCs between 0.686 – 0.766). Leave-one-out validation (CT and HT, no treatment, and deceased patients) exhibited higher model performance than 9-fold crossvalidation (CT and/or HT, including patients treated with radiation).

ea764b20-98b8-4220-99ba-ff2a348add06_figure3.gif

Figure 3. Schematic elements of gene expression changes associated with response to paclitaxel.

Red boxes indicate genes with a positive correlation between gene expression or copy number, and resistance using multiple factor analysis. Blue demonstrates a negative correlation. Genes outlined in dark grey are those in a previously published paclitaxel SVM model (reproduced from reference 1 with permission).

The RF classifier was used to predict paclitaxel therapy outcome for patients that underwent CT and/or HT (Table 2). The best performance achieved with RF showed an 85.5% overall accuracy using a 3-year survival threshold for distinguishing therapeutic resistance vs. sensitivity for those patients that underwent HT.

The best overall accuracy and AUC (sensitivity and specificity) for CT/HT patients using mRMR feature selection for SVM predicting outcome of paclitaxel therapy was obtained for CT patients with 4-year survival (Table 3). Outcomes for HT patients with 3-year survival were predicted with 85.7% accuracy; however, the specificity was lower in this group. SVM combined with mRMR further improved accuracy of feature selection and prediction of response to hormone and/or chemotherapy based on survival time than either SVM or RF alone. Predicted treatment responses for individual METABRIC patients using the described ML techniques are indicated in Dataset 1.

Tumor co-variate information was provided by METABRIC, which included Estrogen receptors (ER), Progesterone Receptor (PR), HER2, Lymph Node (LN) and PAM50 subtypes. To assess model co-variate accuracy, predictions described in Table 1Table 3 were broken down by subtype (available in Supplementary file 1). Subtypes with <20 individuals for a particular treatment combination were not analyzed. The deviation in classification accuracy between subtypes was mostly consistent with the average. One exception involved the RF and mRMR analyses, which was 8.3 to 23.0% below the average for (ER)-negative, (HER2)-positive and basal subtypes in patients treated with HT. However, this deviation was not observed for CT-treated patients with the (ER)-negative subtype, which was consistent with the fact that CT response was derived from the paclitaxel gene set. (ER)-negative patients primarily received CT6. Further, the accuracy of the SVM models tested with CT and HT-treated patients was significantly higher for (HER2)-positive patients (26 correct, 3 misclassified; 90% accurate) compared to (HER2)-negative patients (40 correct, 15 misclassified; 73% accurate). MAPT expression (present in reduced ‘CT and HT’ paclitaxel model; Table 1) has been shown to segregate well with PAM50 luminal and basal subtypes1. When analyzing METABRIC patients, however, the accuracy of these two subtypes are nearly identical to the average (78.6%, where basal and luminal classification accuracy is 76.7% [n=30] and 76.2% [n=21], respectively).

We assessed the separate Discovery and Validation datasets, respectively, as training and test sets and repeated the previous experiments. In this scenario, the performance of the model was poor (slightly better than random). This occurred because the gene expression distributions of many of the paclitaxel-related genes in our signature were not reproducible between these two sets (based on Wilcoxon rank sum test, Kruskal-Wallis test and t-tests; Supplementary file 2). Cross-study validation allows for the comparison of classification accuracy between the generated gene signatures. The observed heterogeneity in gene expression highlights one of the many challenges of cross-validation of gene signatures between these data from the same study exhibit drastic differences (for example, BCL2L1; Supplementary file 2). Furthermore, these gene expression differences also affect the performance of these methods when these datasets were combined (compare Table 2 and Table 4 for RF; Table 3 and Table 5 for mRMR). We considered the possibility that the Discovery model might be subject to overfitting. We therefore performed cross-study validation of the Discovery set-signature with an independently-derived dataset (319 invasive breast cancer patients treated with paclitaxel and anthracycline chemotherapy5). The mRMR+SVM CT-models performed well (4-year threshold model had an overall accuracy of 68.7%; 3-year threshold model exhibited lower overall accuracy [52%], but was significantly better at predicting patients in remission [74.2%]).

To evaluate the paclitaxel models without relying on the Validation dataset, the Discovery set was split into two distinct parts, consisting of 70% of the patient samples randomly selected for training, and a different set of 30% of samples for testing. This procedure was repeated 100 times using different combinations of training and test samples, and the median performance of these runs is reported (Table 4 and Table 5). We also compared the performance of our mRMR+SVM model with the K-TSP model12 (Table 6). In most cases, our method outperformed K-TSP, based on its accuracy in classifying new patients. Starting with the same set of Discovery genes, we also trained a separate model using the Validation data, and tested this data by 70/30% cross-validation (accuracy for RF: 56–67% [CT], 67–83% [HT], 56–81% [CT-HT]; accuracy for mRMR: 33–56% [CT], 70–84% [HT], 64–82% [CT-HT]). In addition, we evaluated the performance of the model derived from the Discovery set on a different set of patients treated with paclitaxel5. These results suggest that the aforementioned issue with Discovery training and Validation testing was primarily due to a batch effect, rather than to overfitting.

Table 4. Results of applying RF to predict outcome of the paclitaxel signature for the METABRIC Discovery patient set.

Type of
treatment
Survival
years (as
threshold)
# PatientsK (number
of genes
to be used
in random
selection)
Accuracy
(True
Positive -
TP) (%)
PrecisionF-MeasureMCCAUC
Chemotherapy
(CT)
322761.10.6170.6120.2240.444
4766.70.6430.6460.1890.715
51966.70.7220.6870.1890.571
Hormone therapy
(HT)
31851977.00.7800.7750.0180.524
4979.10.7330.7100.0840.527
5968.90.5330.601-0.1330.594
CT and/or HT3221980.20.6770.734-0.070.389
41954.80.5540.551-0.1430.395
5760.50.5670.5790.0160.479

Paclitaxel gene panel consisted of 19 genes (ABCB1, ABCB11, ABCC1, ABCC10, BAD, BBC3, BCL2, BCL2L1, BMF, CYP2C8, CYP3A4, MAP2, MAP4, MAPT, NR1I2, SLCO1B3, TUBB1, TUBB4A, TUBB4B).

Table 5. Results of mRMR feature selection for an SVM for predicting outcome of the paclitaxel signature for the METABRIC Discovery patient set.

TreatmentCT1HTCT+HT
Survival
years (as
threshold)
345345345
# patients22185221
Accuracy
(TP) (%)
57.1457.1485.781.870.963.671.269.771.2
Precision0.5950.6860.7350.7260.6700.5320.6470.6290.693
F-Measure0.5710.6230.7910.7690.6860.5620.6680.6280.666
MCC0.167-0.2580.000-0.0800.032-0.0750.0350.0710.245
AUC0.5830.3330.5000.4790.5140.4770.5130.5210.586
SVM Par.
(gamma)
0.00.51.01.00.751.50.750.51.0
SVM Par.
(cost)
64128826421622
Selected
genes
TWIST1
BMF
CYP2C8
CYP3A4
BCL2L1
BBC3
BAD
MAP2
MAPT
NFKB2
FN1
BCL2
BMF
CYP2C8
CYP3A4
BAD
ABCC10
NFKB2
MAP2
BCL2
BCL2L1
BBC3
MAPT
GBP1
NFKB2
TWIST1
BCL2
BMF
CYP2C8
CYP3A4
BCL2L1
BBC3
TLR6
BAD
ABCB11
ABCC1
ABCC10
MAP4
MAPT
NR1I2
GBP1
NFKB2
OPRK1
FN1
TWIST1
CYP2C8
CYP3A4
BCL2L1
BBC3
TLR6
ABCB11
ABCC1
ABCC10
MAP2
MAPT
NR1I2
GBP1
NFKB2
FN1
TWIST1
BMF
CYP2C8
CYP3A4
BCL2L1
BBC3
ABCB11
ABCC1
ABCC10
MAP2
MAP4
MAPT
NR1I2
GBP1
NFKB2
OPRK1
BMF
CYP2C8
BCL2L1
BBC3
BAD
ABCC1
ABCC10
MAP4
NR1I2
GBP1
NFKB2
OPRK1
FN1
TWIST1
BMF
CYP2C8
CYP3A4
BCL2L1
BBC3
TLR6
ABCB11
ABCC1
ABCC10
MAP2
MAP4
MAPT
NR1I2
GBP1
NFKB2
OPRK1
FN1
TWIST1
BMF
CYP3A4
BCL2L1
BBC3
TLR6
BAD
ABCB11
ABCC1
MAP2
MAP4
MAPT
NR1I2
GBP1
NFKB2
OPRK1
FN1

1For patients treated with CT with ≥4 Yr survival and CT+ HT for ≥ 5 Yr, the cost for the mRMR model was set to 64. Of those treated with CT for ≥ 4 Yr, genes were selected using a greedy, stepwise forward search, while in other cases, greedy stepwise backward search was used. Also, gamma = 0 in all cases.

Table 6. Comparison between our mRMR+SVM method and K-TSP method on Discovery patient set of the METABRIC data.

DataCTHTCT+HT
Survival years345345345
# patients22185221
mRMR+SVM Accuracy (%)57.1457.1485.781.870.963.671.2169.7071.21
K-TSP12 Accuracy (%) 57.1428.5728.5780.9168.1869.1971.2154.5553.03

The performances of several ML techniques have been compared such that they distinguish paclitaxel sensitivity and resistance in METABRIC patients using its tumour gene expression datasets. We used mRMR to generate gene signatures and determine which genes are important for treatment response in METABRIC patients. The paclitaxel models are more accurate for prediction of outcomes in patients receiving HT and/or CT compared to other patient groups.

While not a replication study sensu stricto, the initial paclitaxel gene set used for feature selection was the same as in our previous study1. Predictions for the METABRIC patient cohort, which was independent of the previous validation set5 used in Dorman et al.1, of the either same (SVM) or different ML methods (RF and SVM with mRMR) exhibited comparable or better accuracies than our previous gene signature1.

These techniques are powerful tools which can be used to identify genes that may be involved in drug resistance, as well as predict patient survival after treatment. Future efforts to expand these models to other drugs may assist in suggesting preferred treatments in specific patients, with the potential impact of improving efficacy and reducing duration of therapy.

Conclusion

In this study we used METABRIC dataset to predict outcome for different survival times in patients receiving hormone (HT) and, in some cases, chemotherapy (CT) agents. We used published literature and various machine learning methods in order to identify optimal subsets of genes from a biologically-relevant initial gene set that can accurately predict therapeutic response of patients who have received chemotherapy, hormone therapy or a combination of both treatments. The SVM methodology has been previously shown to outperform randomized gene sets1. The predictions made by our method are based on the level of an individual drug. Genomic information has been shown to correlate with tumor therapy response in previous studies5,1317. From these studies, analytical methods have been used to develop gene signatures for chemotherapy resistance prediction5, subtypes (PAM50), and metastatic risk stratification (Oncotype DX™, MammaPrint®). We also examined the method exhibiting the best performance in the Sage Bionetworks / DREAM Breast Cancer Prognosis Challenge18, which was also phenotype-based, however it produces outcome signatures based on molecular processes rather than the cancer drugs themselves. While interesting and informative, the results cannot be directly compared. Our approach may be useful for selecting specific therapies in patients that would be expected to produce a favorable response.

Data availability

Patient data: The METABRIC datasets are accessible from the European Genome-Phenome Archive (EGA) using the accession number EGAS00000000083 (https://www.ebi.ac.uk/ega/studies/EGAS00000000083). Normalized patient expression data for the Discovery (EGAD00010000210) and Validation sets (EGAD00010000211) were retrieved with permission from EGA. Corresponding clinical data was obtained from the literature6. While not individually curated, HT patients were treated with tamoxifen and/or aromatase inhibitors, while CT patients were most commonly treated with cyclophosphamide-methotrexate-fluorouracil (CMF), epirubicin-CMF, or doxorubicin-cyclophosphamide.

F1000Research: Dataset 1. Predicted treatment response for each individual METABRIC patient, 10.5256/f1000research.9417.d14986411

Comments on this article Comments (0)

Version 3
VERSION 3 PUBLISHED 31 Aug 2016
Comment
Author details Author details
Competing interests
Grant information
Copyright
Download
 
Export To
metrics
Views Downloads
F1000Research - -
PubMed Central
Data from PMC are received and updated monthly.
- -
Citations
CITE
how to cite this article
Mucaki EJ, Baranova K, Pham HQ et al. Predicting Outcomes of Hormone and Chemotherapy in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) Study by Biochemically-inspired Machine Learning [version 3; peer review: 2 approved] F1000Research 2017, 5:2124 (https://doi.org/10.12688/f1000research.9417.3)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
track
receive updates on this article
Track an article to receive email alerts on any updates to this article.

Open Peer Review

Current Reviewer Status: ?
Key to Reviewer Statuses VIEW
ApprovedThe paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approvedFundamental flaws in the paper seriously undermine the findings and conclusions
Version 3
VERSION 3
PUBLISHED 12 May 2017
Revised
Views
16
Cite
Reviewer Report 31 May 2017
Elana J. Fertig, Division of Oncology Biostatistics and Bioinformatics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA 
Approved
VIEWS 16
The authors have ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Fertig EJ. Reviewer Report For: Predicting Outcomes of Hormone and Chemotherapy in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) Study by Biochemically-inspired Machine Learning [version 3; peer review: 2 approved]. F1000Research 2017, 5:2124 (https://doi.org/10.5256/f1000research.12412.r22653)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
Version 2
VERSION 2
PUBLISHED 27 Jan 2017
Revised
Views
23
Cite
Reviewer Report 21 Feb 2017
Chun-Wei Tung, School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan 
Approved
VIEWS 23
The authors have addressed all the concerns raised from the previous review. A minor comment for the batch effects is given in the follows. As batch effects are expected for heterogeneous datasets, the direct application of prediction model built on ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Tung CW. Reviewer Report For: Predicting Outcomes of Hormone and Chemotherapy in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) Study by Biochemically-inspired Machine Learning [version 3; peer review: 2 approved]. F1000Research 2017, 5:2124 (https://doi.org/10.5256/f1000research.11525.r19726)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
  • Author Response 12 May 2017
    Peter Rogan, Department of Biochemistry, University of Western Ontario, London, Canada
    12 May 2017
    Author Response
    Thank you for your suggestion. As recommended, we have repeated the 70/30% cross-validation analysis performed in the manuscript (Tables 4 and 5) with the same genes obtained from the Discovery ... Continue reading
COMMENTS ON THIS REPORT
  • Author Response 12 May 2017
    Peter Rogan, Department of Biochemistry, University of Western Ontario, London, Canada
    12 May 2017
    Author Response
    Thank you for your suggestion. As recommended, we have repeated the 70/30% cross-validation analysis performed in the manuscript (Tables 4 and 5) with the same genes obtained from the Discovery ... Continue reading
Views
41
Cite
Reviewer Report 03 Feb 2017
Elana J. Fertig, Division of Oncology Biostatistics and Bioinformatics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA 
Approved with Reservations
VIEWS 41
The authors were very responsive to the previous round of reviews, including more robust cross-validation and cross-study validation and comparison with other classifiers. Particular concerns remain that the author’s conclusions that it is inappropriate to perform cross-study validation due to ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Fertig EJ. Reviewer Report For: Predicting Outcomes of Hormone and Chemotherapy in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) Study by Biochemically-inspired Machine Learning [version 3; peer review: 2 approved]. F1000Research 2017, 5:2124 (https://doi.org/10.5256/f1000research.11525.r19727)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
  • Author Response 12 May 2017
    Peter Rogan, Department of Biochemistry, University of Western Ontario, London, Canada
    12 May 2017
    Author Response
    Methods
    Comment 1. Abbreviations SVM and RF must be spelled out as Support Vector Machine and
    Random Forest on first use. This was not addressed in the revised methods section.


    Response: These abbreviations ... Continue reading
COMMENTS ON THIS REPORT
  • Author Response 12 May 2017
    Peter Rogan, Department of Biochemistry, University of Western Ontario, London, Canada
    12 May 2017
    Author Response
    Methods
    Comment 1. Abbreviations SVM and RF must be spelled out as Support Vector Machine and
    Random Forest on first use. This was not addressed in the revised methods section.


    Response: These abbreviations ... Continue reading
Version 1
VERSION 1
PUBLISHED 31 Aug 2016
Views
32
Cite
Reviewer Report 03 Oct 2016
Chun-Wei Tung, School of Pharmacy, Kaohsiung Medical University, Kaohsiung, Taiwan 
Approved with Reservations
VIEWS 32
This study proposed prediction methods using SVM and RF classifiers with mRMR selected feature sets from cell line data and demonstrate its prediction ability for outcomes from METABRIC patient cohort. The classifiers with good prediction performance show the usefulness of ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Tung CW. Reviewer Report For: Predicting Outcomes of Hormone and Chemotherapy in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) Study by Biochemically-inspired Machine Learning [version 3; peer review: 2 approved]. F1000Research 2017, 5:2124 (https://doi.org/10.5256/f1000research.10141.r16345)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
  • Author Response 27 Jan 2017
    Peter Rogan, CytoGnomix Inc, London, Canada
    27 Jan 2017
    Author Response
    Comment 1:What are the values of parameters for SVM and RF classifiers and the methods for parameter selection (by default or other selection methods)?

    Response: The parameter values for ... Continue reading
COMMENTS ON THIS REPORT
  • Author Response 27 Jan 2017
    Peter Rogan, CytoGnomix Inc, London, Canada
    27 Jan 2017
    Author Response
    Comment 1:What are the values of parameters for SVM and RF classifiers and the methods for parameter selection (by default or other selection methods)?

    Response: The parameter values for ... Continue reading
Views
45
Cite
Reviewer Report 30 Sep 2016
Elana J. Fertig, Division of Oncology Biostatistics and Bioinformatics, School of Medicine, Johns Hopkins University, Baltimore, MD, USA 
Approved with Reservations
VIEWS 45
This study develops SVM and RF algorithms built upon previously learned gene signatures of therapeutic response to breast cancer. The algorithms are applied and compared to predict patient survival under different treatment conditions in METABRIC data. The analyses and comparisons ... Continue reading
CITE
CITE
HOW TO CITE THIS REPORT
Fertig EJ. Reviewer Report For: Predicting Outcomes of Hormone and Chemotherapy in the Molecular Taxonomy of Breast Cancer International Consortium (METABRIC) Study by Biochemically-inspired Machine Learning [version 3; peer review: 2 approved]. F1000Research 2017, 5:2124 (https://doi.org/10.5256/f1000research.10141.r16733)
NOTE: it is important to ensure the information in square brackets after the title is included in all citations of this article.
  • Author Response 27 Jan 2017
    Peter Rogan, CytoGnomix Inc, London, Canada
    27 Jan 2017
    Author Response
    Comment 1: The methods require further clarification to distinguish differences between this study and the previous study as well as the parameters of the machine learning algorithms.
     
    Response: The ... Continue reading
COMMENTS ON THIS REPORT
  • Author Response 27 Jan 2017
    Peter Rogan, CytoGnomix Inc, London, Canada
    27 Jan 2017
    Author Response
    Comment 1: The methods require further clarification to distinguish differences between this study and the previous study as well as the parameters of the machine learning algorithms.
     
    Response: The ... Continue reading

Comments on this article Comments (0)

Version 3
VERSION 3 PUBLISHED 31 Aug 2016
Comment
Alongside their report, reviewers assign a status to the article:
Approved - the paper is scientifically sound in its current form and only minor, if any, improvements are suggested
Approved with reservations - A number of small changes, sometimes more significant revisions are required to address specific details and improve the papers academic merit.
Not approved - fundamental flaws in the paper seriously undermine the findings and conclusions
Sign In
If you've forgotten your password, please enter your email address below and we'll send you instructions on how to reset your password.

The email address should be the one you originally registered with F1000.

Email address not valid, please try again

You registered with F1000 via Google, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Google account password, please click here.

You registered with F1000 via Facebook, so we cannot reset your password.

To sign in, please click here.

If you still need help with your Facebook account password, please click here.

Code not correct, please try again
Email us for further assistance.
Server error, please try again.