Oncotarget

Research Papers:

Gene expression information improves reliability of receptor status in breast cancer patients

PDF |  HTML  |  How to cite

Oncotarget. 2017; 8:77341-77359. https://doi.org/10.18632/oncotarget.20474

Metrics: PDF 1358 views  |   HTML 2606 views  |   ?  

Michael Kenn, Karin Schlangen, Dan Cacsire Castillo-Tong, Christian F. Singer, Michael Cibena, Heinz Koelbl and Wolfgang Schreiner _

Abstract

Michael Kenn1, Karin Schlangen1, Dan Cacsire Castillo-Tong2, Christian F. Singer2, Michael Cibena1, Heinz Koelbl3 and Wolfgang Schreiner1

1Section of Biosimulation and Bioinformatics, Center for Medical Statistics, Informatics and Intelligent Systems (CeMSIIS), Medical University of Vienna, A-1090 Vienna, Austria

2Translational Gynecology Group, Department of Obstetrics and Gynecology, Comprehensive Cancer Center, Medical University of Vienna, A-1090 Vienna, Austria

3Department of General Gynecology and Gynecologic Oncology, Medical University of Vienna, A-1090 Vienna, Austria

Correspondence to:

Wolfgang Schreiner, email: [email protected]

Keywords: gene expression, breast cancer, receptor status, data science, mathematical oncology

Received: April 12, 2017     Accepted: July 06, 2017     Published: August 24, 2017

ABSTRACT

Immunohistochemical (IHC) determination of receptor status in breast cancer patients is frequently inaccurate. Since it directs the choice of systemic therapy, it is essential to increase its reliability.

We increase the validity of IHC receptor expression by additionally considering gene expression (GE) measurements. Crisp therapeutic decisions are based on IHC estimates, even if they are borderline reliable. We further improve decision quality by a responsibility function, defining a critical domain for gene expression. Refined normalization is devised to file any newly diagnosed patient into existing data bases. Our approach renders receptor estimates more reliable by identifying patients with questionable receptor status. The approach is also more efficient since the rate of conclusive samples is increased. We have curated and evaluated gene expression data, together with clinical information, from 2880 breast cancer patients. Combining IHC with gene expression information yields a method more reliable and also more efficient as compared to common practice up to now.

Several types of possibly suboptimal treatment allocations, based on IHC receptor status alone, are enumerated. A ‘therapy allocation check’ identifies patients possibly miss-classified. Estrogen: false negative 8%, false positive 6%. Progesterone: false negative 14%, false positive 11%. HER2: false negative 2%, false positive 50%. Possible implications are discussed.

We propose an ‘expression look-up-plot’, allowing for a significant potential to improve the quality of precision medicine.

Methods are developed and exemplified here for breast cancer patients, but they may readily be transferred to diagnostic data relevant for therapeutic decisions in other fields of oncology.


INTRODUCTION

The selection of an optimum breast cancer therapy has to include the expression of estrogen receptors (ER), progesterone (PGR) and human epidermal growth factor 2 (HER2) receptor proteins in an individual patient. The reliable assessment of these 3 receptor status is hence mandatory for optimum individualized therapy.

Although immunohistochemistry (IHC) is considered the gold standard for status determination, doubts have been raised [13] by reported differences between readers repeatedly examining the very same data. While almost complete concordance was found for ER--status, up to 20% of patients assigned ER+- status may be erroneously classified, according to reports in the literature [4, 5]. Of note, we also found a considerable number of ER- that might be misclassified.

First, correct assignment of ER-status is most important for the individualized choice of treatment [6]. As gene expression measurements can be achieved by different techniques, we investigate if the assessment of receptor status could be improved by additionally exploiting gene expression data.

Second, gene expression signatures for the prediction of individual therapeutic outcome have been established [711] and biomarker discovery methods developed [1214], as recently reviewed [4, 15]. Each of these signature-algorithms includes receptor status as decisive variables upon which calculated prognosis crucially depends. Correct prognostic algorithms can thus be developed only on the basis of reliable receptor status [16].

Hence, it is of paramount value for both, patient treatment and research, to increase the reliability or even impute receptor status by the use of additional information, e.g., gene expression measurements [1720]. In the present work we restrict ourselves to a single measurement platform for gene expression (Affymetrix U133A+2.0), in order to avoid inter-platform batch effects.

The present work deals with three receptors, ER, PGR and HER2.

RESULTS

For the present work we used and curated the datasets listed in Table 1.

Table 1: Expression datasets used

GEO Accession number

Citation

Number of samples

Nsample

ER-

ER+

PGR-

PGR+

HER2-

HER2+

GSE5460

[36]

29

11

18

0

0

21

8

GSE11001

[37]

30

12

18

16

14

23

7

GSE12777

[38]

51

0

0

0

0

34

17

GSE16179

[39]

18

0

0

0

0

0

18

GSE16391

[40]

55

0

55

0

0

42

3

GSE16446

[41]

120

0

0

0

0

90

28

GSE18728

[42]

61

29

32

36

25

44

17

GSE18864

[43]

84

53

31

53

31

64

18

GSE19615

[44]

115

45

70

51

64

79

36

GSE20685

[45]

327

0

0

0

0

0

0

GSE20711

[46]

88

45

42

0

0

62

26

GSE23177

[47]

116

0

116

0

0

116

0

GSE26639

[48]

226

88

138

128

95

145

81

GSE27120

[49]

28

4

24

12

16

26

2

GSE29431

[50]

54

0

0

0

0

12

41

GSE31448

[51]

353

162

188

154

167

234

31

GSE32646

[52]

115

44

71

70

45

0

0

GSE42568

[53]

104

34

67

0

0

0

0

GSE43365

[54]

111

18

93

34

77

96

13

GSE48390

[55]

81

28

53

0

0

47

34

GSE50948

[56]

156

104

52

121

35

42

114

GSE58812

[57]

107

107

0

107

0

107

0

GSE61304

[58, 59]

58

25

28

23

22

0

0

GSE71258

[60]

128

48

51

61

38

77

22

GSE76124

[61] [62]

198

198

0

198

0

198

0

GSE76274

[61] [62]

67

18

49

25

42

28

22

Total

 

2880

1073

1196

1089

671

1587

538

GSE-numbers refer to GEO [25, 26], using Affymetrix platform U133A+2.0. Number of samples are given for each study: in total (Nsample) and also separately for receptor positive and receptor negative patients according to IHC measurements.

Receptor status obtained from gene expression by conventional methods

Given the IHC-estimate of a receptor-status, e.g. ERIHC or ERIHC+, two distributions of expression values (xi, i = 1,…, Nsample, number of patients/samples) of the corresponding gene (e.g. ESR1. see Table 2) were estimated, see Figure 1. These plots show results for the estrogen receptor and also illustrate the computational procedure, see section ‘Methods and models for expression of receptor genes’.

Table 2: Probe sets for receptor genes ER, PGR and HER2 for the Affymetrix platform U133A+2.0

Receptor

ER

PGR

HER2

Probeset ID

205225_at

208305_at

216836_s_at

HUGO Nomenclature

ESR1

PGR

ERBB2

ENTREZID

2099

5241

2064

Distribution of gene expression for estrogen-receptor (ESR1): Data, models and thresholds for Affymetrix platform U133A+2.0.

Figure 1: Distribution of gene expression for estrogen-receptor (ESR1): Data, models and thresholds for Affymetrix platform U133A+2.0. X-axis: gene expression. Upper panel: Boxplots for expression values of receptor gene ESR1 (Affymetrix-probe set 205225_at), grouped according to IHC-estimate (ER+ERIHC+, ER-ERIHC). Lower panel: Y-axis: probability density. Normalized histograms for expression values (blue: ERIHC, yellow: ERIHC+). Estimated probability density functions (PDFs) modelled as sums of two normal distributions. PDF obtained ‘classically’ from means and standard deviations: green curves. Dashed lines: individual distributions for ERIHC+ and ERIHC. Solid: superposition. For methods used see legend. Green dashed curves represent portions for ERIHC+ and ERIHC - patients (integrals < 1), adding up to the solid green curve (integral = 1). Cut-points (vertical lines) resulting from different computational approaches (see legend and text), discriminating receptor negative (left) and receptor positive (right) patients. Note that cut-points for MaxLike and ExMax coincide (only ExMax is shown in the figure).

Obtaining receptor status ROC curves

As a prerequisite, we assumed IHC-estimates (e.g.: ERIHC+, ERIHC) to be ‘true’ and computed a ROC-curve (blue curve in Figure 2) by classifying a patient as receptor positive/negative if the expression value, xi, of gene ESR1 was above/below a running threshold, x^. The area under the curve resulted as AUC = 0.94.

ROC-curve for estrogen-receptor gene (ESR1)

Figure 2: ROC-curve for estrogen-receptor gene (ESR1). ROC-curve from IHC-estimates (e.g.: ERIHC+, ERIHC) and RMA-normalized expression values of gene ESR1 (Affymetrix-probe set 205225_at) yields AUC = 0.94 for U133A+2.0. Several concepts have been applied (see legend) to obtain thresholds for discrimination between ER+ and ER-. Sensitivity and specificity of each threshold can be read off the diagram axes.

Cut-points resulting from models of receptor gene expression

In order to evaluate gene expression we assumed expression values, xi, to be approximately normally distributed (N(μ+,σ+) for ERIHC+ and N(μ,σ) for ERIHC), see Figure 1, lower panel. Adding both normal distributions with proper weights (λ++λ=1) yielded a bimodal distribution, see the stacked bars in Figure 1. Likewise, we obtained bimodal distributions for progesterone (gene PGR) and human epidermal growth factor receptor 2 (HER2, gene ERBB2), see Figure 3.

ROC-curves and probability density (distribution) of PGR and HER2-receptor genes&rsquo; expression (PGR and ERBB2, respectively): Data, models and thresholds.

Figure 3: ROC-curves and probability density (distribution) of PGR and HER2-receptor genes’ expression (PGR and ERBB2, respectively): Data, models and thresholds. Left panels: PGR. Right panels: HER2. Upper panels: ROC-curves constructed from PGR- and HER2-values from IHC and RMA-normalized expression values of genes PGR and ERBB2 (Affymetrix-probe sets 208305_at and 216836_s_at, respectively). The areas under the curves (AUC) are 0.90 (for PGR) and 0.91 (for HER2). To obtain thresholds for discrimination (between PGR+ vs. PGR- and HER2+ vs. HER2-), the same concepts have been applied as described for the estrogen receptor, see legend and text for details. Mid panels: Box plots for gene expression after RMA-normalization for PGR and HER2, with boxes classified according to IHC (+/-). For numbers of samples see Table 1. Lower panels: Normalized histograms of expression values (blue bars). x-axis: gene expression after RMA-normalization over all samples. Y-axis: estimated PDF. PDFs obtained ‘classically’ from means and standard deviations: green curves, see the dashed lines for individual distributions of IHC+ and IHC-, respectively. PDF obtained via maximum likelihood: red curve.

To obtain thresholds discriminating receptor positive versus negative we evaluated 5 different methods: MaxLike, ParEst, LogReg, Youden and ExMax, for maths see the ‘Methods and models for expression of receptor genes’ in ‘Material and methods’. We decided to finally adopt method ExMax, see section ‘Why to adopt ExMax’ in ‘Material and methods’, and obtained the cut-points given in Table 3.

Table 3: Distribution parameters and cut-points for receptors ER, PGR and HER2

Receptor

Method

CP

μ

μ+

σ

σ+

λ

λ+

π+

ER

MaxLike

9.453

6.515

11.624

1.415

0.947

0.483

0.517

n/a

 

ParEst

9.099

6.835

11.300

1.870

1.580

0.483

0.517

n/a

 

LogReg

9.177

n/a

n/a

n/a

n/a

n/a

n/a

n/a

 

Youden

8.832

n/a

n/a

n/a

n/a

n/a

n/a

n/a

 

ExMax

9.363

6.557

11.472

1.534

1.013

n/a

n/a

0.519

PGR

MaxLike

4.046

3.587

5.560

0.197

1.483

0.542

0.458

n/a

 

ParEst

4.399

3.647

5.336

0.409

1.555

0.542

0.458

n/a

 

LogReg

4.145

n/a

n/a

n/a

n/a

n/a

n/a

n/a

 

Youden

3.929

n/a

n/a

n/a

n/a

n/a

n/a

n/a

 

ExMax

4.164

3.619

5.722

0.241

1.486

n/a

n/a

0.454

HER2

MaxLike

11.680

9.481

12.982

0.870

0.585

0.754

0.246

n/a

 

ParEst

11.053

9.396

11.912

0.867

1.484

0.754

0.246

n/a

 

LogReg

11.101

n/a

n/a

n/a

n/a

n/a

n/a

n/a

 

Youden

10.633

n/a

n/a

n/a

n/a

n/a

n/a

n/a

 

ExMax

12.304

9.492

13.209

1.251

0.538

n/a

n/a

0.124

Cut-points (CP) as obtained by 5 different methods (MaxLike, ParEst, LogReg, Youden and ExMax), cf. the colored dots in Figure 2 and Figure 3.

Similar results were obtained for PGR and HER2, see the bottom rows in Figure 3. Computationally, we proceeded along the very same lines as for ER. Note that the majority of patients was HER2-. As a consequence, thresholds shifted towards larger expression values (as compared to ER) and specificity of discrimination resulted fairly large.

Although HER2+ has much lower prevalence then HER2-, the ExMax-estimate performs outstandingly well, as can be seen from Figure 3.

Agreement between receptor status obtained by IHC versus gene expression cut-points

Using the cut-points (CP) shown in Table 3, we obtained receptor status from gene expression as compared to IHC-estimates, see Table 4.

Table 4: Agreement (contingency tables) between receptor status from IHC versus gene expression

 

estrogen ER

progesterone PGR

HER2

GE–

GE+

GE–

GE+

GE–

GE+

IHC−

943

130

892

197

1573

14

IHC+

112

1084

184

487

305

233

log(DOR) ± 95%

4.25±0.27

2.48±0.23

4.45±0.55

Cohen’s Kappa

0.79

0.54

0.52

Kruscal’s Gamma

0.97

0.85

0.98

Rate of discordant samples

11%

22%

15%

Rate of conclusive samples

89%

78%

85%

Cut-points for gene expression were those derived by the ExMax-algorithm, see Table 3. Four measures of agreement are given: log(DOR) (natural logarithm of the Diagnostic Odds Ratio, DOR), Cohen’s Kappa (inter-rater agreement), Kruscal’s Gamma (strength of association) and the rate of discordant samples (conjugate to the rate of conclusive samples).

Association between IHC and gene expression was quantified by Kruscal’s gamma, Cohen’s Kappa (cf. Table 4) and also by the diagnostic odds ratio, DOR, and its 95% confidence intervals [21].

Disagreement between IHC and GE, as quantified by the rate of discordant samples, is highly problematic in the clinical setting, and a clinical decision should not be made on the basis of IHC data only.

PGR showed less agreement than ER due to its lower expression values, entailing a broad distribution of PGR expression values for PGRIHC+. Regarding HER2, the precision of determination suffered from an imbalance in the data: Much more patients are HER2 negative than positive. Note that missing IHC-estimates, as considered in the next chapter, were not included here.

In order to improve this situation we enhanced the decision process by furnishing GE with a responsibility function and a critical domain.

Improved decision quality due to responsibility function and critical domain

Instead of using a single cut-point for each receptor we introduced a ‘critical domain’ [ Xlower,Xupper ] for a probability level α=0.05, see Figure 4 and, for mathematical details, the chapter ‘Material and methods’. For expression values x<Xlower we decided for receptor negative, for x>Xupper receptor positive: In both cases we classified the expression value as ‘informative’. As opposed, expression values within the critical domain XlowerxXupper were classified as ‘undecidable’, and we refrained from a decision. Critical domains thus constructed are given in Table 5 and their practical application is described in section, Expression lookup plot‘ in ‘Material and methods’.

Responsibility functions r&minus; for receptors ER, PGR and HER2.

Figure 4: Responsibility functions r for receptors ER, PGR and HER2. Critical domains, where no decision is possible, are indicated as colored stripes. For PGR, f+is extremely wide (very large σ+) and causes the unexpected shape of r. Borders of critical domains result from the intersections of the respective r with the 5% and 95% limits (dotted lines).

Table 5: Critical domains of gene expression for receptor status determination

Receptor

Probeset

Xlower

Xupper

ER

205225_at

8.517

10.354

PGR

208305_at

3.664

4.407

HER2

216836_s_at

11.783

13.070

The critical domain for expression values x is given by XlowerxXupper, based on the ExMax-algorithm and probe sets listed in Table 2. The section ‘Expression lookup plot’ in ‘Material and methods’ describes how to actually apply the criterion XlowerxXupper

The information conveyed by gene expression was thus refined, yielding the results for GE 0, shown in Table 6 and the yellow sub-bars in Figure 5.

Number of concordant and discordant samples.

Figure 5: Number of concordant and discordant samples. Left panel: Bars according to IHC receptor estimate. Height of bar represents number of samples with gene expression concordant (green), in critical domain (yellow) and discordant (red). Note that samples without IHC estimate are not included. Right panel: Bars according to GE receptor estimate. Height of bar represents number of samples with IHC concordant (green), missing (yellow) and discordant (red).

Table 6: Number of samples for which IHC and gene expression yield receptor positive/unknown/negative

 

gene expression

ER

PGR

HER2

GE –

GE 0

GE +

GE –

GE 0

GE +

GE –

GE 0

GE +

IHC –

887

100

86

590

357

142

1560

20

7

IHC 0

273

81

257

365

282

473

593

88

74

IHC +

70

157

969

75

143

453

271

119

148

Critical GE

 

338

 

 

782

 

 

227

 

Rate of discordant samples

 

5%

 

 

8%

 

 

10%

 

Rate of conclusive samples

 

92%

 

 

79%

 

 

87%

 

IHC 0: no IHC measurement available.

GE 0: expression value within the critical domain, and no decision can be made. Boundaries of critical domains were computed by the ExMax-method.

Bold print: discordant samples.

Note that results shown in this table are obtained step by step via the methods explained in ‘Material and Methods’.

A critical domain regarding GE indicates whether or not to trust in GE expression alone. This is especially useful in cases of lacking IHC estimates. For ER in particular, 81 cases revealed as untrustworthy, since missing IHC was accompanied by GE within the critical domain.

Introducing a critical domain is in particular useful in the case of PGR, since GE estimates of PGR offer only limited power of discrimination (leaving as many as 782 samples in column GE 0). But also for ER, 338 samples were revealed untrustworthy (column GE 0).

HER2 estimates revealed a special problem: Many HER2IHC+ samples had low (i.e. contradicting) GE, suggesting they might be false positives from IHC measurement, see the red part of the rightmost bar in left panel of Figure 5.

Therapy allocation check and consequences

In order to scrutinize if the additional information conveyed by GE might change therapeutic decisions based on IHC only, we compared four groups:

1. (ERIHC+ERGE+,0)(PGRIHC+PGRGE+,0) (see Figure 6, blue curves, left column, 329 Pat.): Receptor positive patients according to IHC, had received hormone treatment. Since GE confirmed IHC, the choice of systemic therapy deems correct in these cases.

Correct and possibly miss-led assignment of systemic therapy influences survival time free of symptoms.

Figure 6: Correct and possibly miss-led assignment of systemic therapy influences survival time free of symptoms. Kaplan Meier estimate of overall survival and 95% confidence intervals. Blue curves: patients with IHC status in accordance with gene expression (correctly assigned therapy). Violet: patients with IHC status contradicted by GE and possibly having received suboptimal therapy.

2. (ERIHCPGRIHC)(ERGE0,PGRGE0,) (see Figure 6, blue curves, right column, 137 Pat.): Receptor negative patients according to IHC, had not received hormone treatment, accordingly. Since GE confirmed IHC, the choice of systemic therapy deems correct in these cases.

3. (ERIHC+PGRIHC+)(ERGEPGRGE) (see Figure 6, violet, upper left, lower right, 18 Pat.): Patients deemed receptor positive according to IHC, had received hormone treatment, accordingly. Since GE contradicted IHC, the choice of systemic therapy might have been incorrect in two possible ways: Those patients who had received neoadjuvant hormone therapy (in addition to adjuvant chemotherapy) would not have benefitted from it (negligible harm). Those patients receiving only hormone therapy without chemotherapy would have been given an ineffective therapy while at the same time being deprived of the adequate, live-saving therapy.

4. (ERIHCPGRIHC)(ERGE+PGRGE+) (see Figure 6, violet, upper right, lower left, 9 pat.): Patients deemed receptor negative according to IHC, not having received hormone treatment, accordingly. Since GE contradicted IHC, the choice of systemic therapy might have been suboptimal in these cases.

For each group, we calculated the Kaplan Meier product limit estimator for time free from relapse (see Figure 6) and obtained 95% confidence limits via the Greenwood formula [22].

In group 1 (blue, left column), a patient’s positive hormone-receptor status according to IHC was not contradicted by GE. Accordingly, these patients are likely to have been allocated the ‘correct’ i.e. anti-hormone treatment and have actually benefitted therefrom.

Patients in group 2 (blue, right column) had been assessed hormone receptor negative via IHC, and this was confirmed by GE: They are likely to have received optimum systemic therapy.

In contrast, patients in group 3 (violet, upper left and lower right) had been assessed hormone receptor positive via IHC, however, this was contradicted by GE: they might have received a hormone therapy but may not have benefitted from it (since they are likely to lack receptors, according to GE). It might even be the case that adjuvant chemotherapy was spared although being mandatory, relying on just hormone therapy (which did not work in these patients).

Patients in group 4 (violet, upper right and lower left) had been assessed hormone receptor negative via IHC, however, this was contradicted by GE: They might have been deprived of a hormone therapy they could have benefitted from.

The above grouping is weakly formulated by intention, in the sense that IHC estimates (used as basis for therapeutic decision) were questioned if and only if GE actually contradicted, not if GE was just within its critical domain ([GE+, GE0] for receptor positive, [GE-, GE0] for receptor negative). Tightening the definition of error-prone groups (not tolerating ‘0’ anymore), might even aggravate the contrasts, concomitantly reducing the number of patients in each group, however. We decided to stick to the broader (and weaker) definition so as not to withhold possibly decisive warnings to patients with borderline receptor status.

DISCUSSION

Achievements due to responsibility function and critical domain

For the majority of patients (89%), the IHC-assessment of estrogen receptors (ERIHC) and gene expression (ERGE, gene ESR1) data yielded compatible results and we may consider such an estimate true. This percentage was similar to the portion of IHC-estimates in the literature suspected to be true [23]. In this work we paved the way to consistently deal with missing as well as contradicting estimates as follows, see Table 6.

In cases of missing IHC-estimates one cannot simply trust GE. Instead, in order to strengthen decision performance, we rather have to introduce a critical region for GE. It effectively flags those cases where GE alone deems untrustworthy – 81, 282 and 88 for ER, PGR and HER2, respectively – and thereby provides a ‘quality filter’.

Available IHC estimates which are contradicted by GE when based on simple cut-points (crisp decision), represent a large number of discordant cases:

(a) For ER, about (11% of patients in our cohort, ERIHC and ERGE disagreed, with about equal numbers (130/112) being found in each of the two possible modes of disagreement see Table 4.

(b) For PGR, discordant cases were similarly balanced (197/184), the rate of discordant samples (22%) being twice that of ER, however.

(c) For HER2 finally, discordant cases were severely imbalanced (14/305), with a rate of 15% of discordant samples, cf. Table 4 and Table 6.

We have to consider, however, that samples with formerly ‘contradicting’ GE (according to a crisp decision based on a simple cut-point for GE) may switch to non-contradicting, i.e. ‘compliant’, if GE lied just within (and not beyond the limits of) its critical domain.

Thus, considering a critical domain for GE, the rate of discordant samples decreased while the rate of conclusive samples increased, as can be seen by comparing Table 4 and Table 6.

Results for patients on top of those for statistics

If the goal were just to build a cohort of patients for developing a gene-expression signature statistically, one could, of course, retreat to the save side and consider only cases in agreement. However, even this might bias the results and call for an improvement of receptor estimate quality.

However, if the goal are decisions upon therapeutic interventions, these have to be made for all patients and in utmost quality - even in cases of disagreement between IHC and gene expression. We therefore tried to reduce the risk of wrong decisions by improving the cut-point-method by the additional concept of a ‘critical domain’.

Improving receptor security for a newly diagnosed patient

For a newly diagnosed patient, we proposed a procedure to improve receptor security, see section ‘Expression lookup plot’ in ‘Material and methods’. The ‘critical domain’ denotes non-decidable cases with responsibilities between 0.05 and 0.95.

Note that such a direct procedure is only possible due to the nature of RMA-normalization of expression data, see section ‘Receptor status obtained from gene expression’ in ‘Results’.

This straight forward procedure for the newly diagnosed patient is easily applicable (and renders any re-normalization on the fly, etc., unnecessary).

Note however, that the ‘expression look-up-plot’ may critically depend on expression chip processing, despite RMA-normalization. We hence recommend building the ‘expression look-up-plot’ on the basis of gene expression measurements performed in the very same environment as data from newly diagnosed patients are analysed.

As a consequence and achievement, treatment quality of newly diagnosed patients will benefit, thus conveying a significant step towards precision medicine.

Balancing the risks of erroneous receptor status

We have shown that additional information from GE-data including a critical domain may improve receptor security in a considerable portion of cases, see Table 6.

a) ERIHC

Out of 1073 patients classified as ERIHC only 92% were not contradicted by gene expression. Without our proposed re-assessment, the remaining 8% may receive chemotherapy although hormone therapy might suffice. Thus, applying our proposed method, unnecessarily severe side-effects could possibly be saved in approximately 8% of that patient cohort.

b) ERIHC+

Out of 1196 patients classified as ERIHC+ only 94% were not contradicted by gene expression. Without our proposed re-assessment, the remaining 6% may erroneously receive non-effective endocrine therapy while chemotherapy is withheld, which may be lethal. Thus, identifying these patients, may save lives in approximately 6% of that patient cohort.

c) PGRIHC

Out of 1089 patients classified PGRIHC only 87% were not contradicted by gene expression. Without our proposed re-assessment, the remaining 13% would possibly receive under-treatment (if also ERIHC), since endocrine therapy would be falsely withheld from this population.

d) PGRIHC+

Out of 671 patients classified PGRIHC+ only 89% were not contradicted by gene expression. Without our proposed re-assessment, the remaining 11% would be falsely misclassified into endocrine sensitive and would thus be exposed to the side effects of endocrine therapy without a therapeutic benefit.

e) HER2IHC

Out of 1587 patients classified HER2IHC, 99.6% were not contradicted by gene expression. Without our proposed re-assessment, the remaining 0.4% would be exposed to Herceptin-associated side effects without deriving a benefit from this expensive and side-effect-related therapy.

f) HER2IHC+

Out of 538 patients classified HER2IHC+ only 50% were not contradicted by gene expression. Without our proposed re-assessment, the remaining 50% would not be able to receive HER2-targeted treatment such as Herceptin, and would be under-treated.

Balancing the consequences of above listed decisions in therapy allocation we adhere to the general consensus that withholding endocrine therapy when it should be delivered is more harmful than applying endocrine therapy when not needed. Hence,

• the therapy allocation error in c) is more serious than it is in d).

• the therapy allocation error in f) is more serious than it is in e).

• the most serious allocation error is listed in b), concerning patients falsely diagnosed as receptor positive.

Characteristics of selected procedures

This section critically enumerates assumptions underlying the results presented here.

Dependence of procedures on IHC - measurements

Expectation maximization (ExMax) does not draw on IHC-measurements at all, i.e. all 5 parameters of the two normal distributions (π+,μ+,σ+,μ,σ)1 are estimated simultaneously from expression data, see Table 3. On the contrary, for methods Youden, ParEst, LogReg and MaxLike, λ+ has to be computed from IHC-values in advance (i.e. these methods depend on IHC-measurements).

Table 7: Distribution parameters of gene expression and their 95% confidence-intervals

Receptor

μ−

μ+

σ−

σ+

ER

6.557 ± 0.047

11.472 ± 0.031

1.534 ± 0.043

1.013 ± 0.027

PGR

3.619 ± 0.007

5.722 ± 0.050

0.241 ± 0.006

1.486 ± 0.031

HER2

9.492 ± 0.047

13.209 ± 0.008

1.251 ± 0.036

0.538 ± 0.008

Parameters have been obtained by the ExMax-method applied to 2880 samples. 95% Confidence intervals have been obtained via expected Fisher information (see ‘Material and methods’, section ‘Obtaining confidence intervals for expression distribution parameters’) and verified by bootstrapping (see chapter 8 in [35]).

1λ+ and λ are normalized to unity, hence only λ+ is free to be adjusted.

Dichotomous IHC-estimates

IHC assays initially produce raw values which are normally dichotomized into ERIHC+ and ERIHC according to some thresholds. Using more than one threshold yields results such as ‘low’, ‘moderate’, ‘high’, etc.

A straight forward improvement of the current methodology could draw on raw values of IHC-assays and file them into the analysis as real values similar to gene expression values. Such an approach could even outperform the one presented here and may be the scope of future investigations.

A drawback of this proposal is, however, that in most cases we only have dichotomized IHC-estimates at hand. No large datasets are available on the net to pursue the issue if gradual IHC estimates could improve precision.

Goals achieved and clinical impact

Receptor status estimated via IHC is a key ingredient of precision medicine for breast cancer patients. However, significant doubts have been raised regarding reliability, when comparing disparate receptor estimates of different readers evaluating the same data [5].

On the other hand, receptor estimates from gene expression, which are usually not used in clinical settings, have been given even more credit than IHC estimates [24].

Taking GE in account in addition to IHC clearly boosts credibility of concordant cases but, at first glance, reduced the number of decidable cases, since contradicting cases also emerged. However, in a second step, concordance could be re-improved by considering a critical domain of GE, thereby again reducing the number of contradictory cases.

In this work we scrutinized possible mathematical ways to enrich receptor information from IHC by gene expression data and thus put decisions on therapies on more solid grounds. To these ends we developed a procedure to incorporate both, IHC estimates and gene expression data.

This work used Affymetrix data together with clinical parameters downloaded from GEO in search for an improved receptor status determination. Translated into clinical reality, the expression status of those few genes involved will most probably be determined via RT-qPCR. Gene expression for single, newly diagnosed patients can be made interpretable and meaningful by our proposed ‘expression-lookup-table’ and lends itself as a valuable tool for improved receptor status assessment.

MATERIALS AND METHODS

Data for gene expression and receptor status

Scanning the gene expression omnibus, GEO [2527], for studies on breast cancer, using Affymetrix U133A+2.0 arrays and also containing receptor data resulted in 28 studies, two of which are redundant. The 26 non-redundant studies comprise a total number of Nsample = 2880 individual breast cancer samples, after excluding samples completely lacking clinical information, control samples as well as duplicate samples.

Affymetrix-probe sets for receptors are given in Table 2.

Normalization

RMA-normalization was performed on all 54675 probe sets on the Affymetrix U133A+2.0 expression chip using the rma-function from the Bioconductor’s affy- package [2830].

For the analyses we restricted ourselves to the subset of the 22215 probe sets U133A+2.0 has in common with U133A, in order to be downwards compatible.

Methods and models for expression of receptor genes

To exploit information from the expression of receptor genes we considered five methods: MaxLike, ParEst, LogReg, Youden and ExMax.

Method 1: Youden point

The conventional, most straight forward approach is to compute the Youden point [31] along the ROC-curve by maximizing

J=sensitivity+specificity1(1)

see the label ‘Youden’ in Figure 1, Figure 2 and Figure 3, lower panels. The expanded formula reads:

J=truepositivestruepositives+falsepositives+truenegativestruenegatives+falsepositives(2)

Method 2: logistic regression (LogReg)

Another simple approach is logistic regression [32] of receptor status versus expression values, xi. The cut-point is set at equal probabilities (0.5, 0.5) for receptor positive and negative, respectively, see the label ‘LogReg’ in Figure 1, Figure 2 and Figure 3, lower panels. Parameters of distributions (N(μ+,σ+) and N(μ,σ)) are not considered.

Method 3: parameter estimation from bimodal distribution (ParEst)

Normal distributions are evaluated classically by computing mean and standard deviation (μ+,σ+) from N+ receptor positive patients and (μ,σ) from N receptor negative patients, see the dashed green curves in Figure 1, Figure 2 and Figure 3. Weights (λ+=N+/(N++N), λ=N/(N++N)) are preset from relative abundances of IHC-measurements (not fitted). This yields the overall probability-density functions (PDF, solid green line).

f(x)=λ+f+(μ+,σ+|x)+λf(μ,σ|x)(3)

The threshold is set at equal Bayesian probabilities Pr(+|x)=Pr(|x)=0.5 [33], where the Bayesian probability is given by:

Pr(A|x)=f(A)(x)λ(A)Af(A)(x)λ(A)(4)

Here f(A) represents the probability density for receptor-status A{ ,+ } and x is the measured expression value of the respective gene.

Equating Pr(+|x)=Pr(|x) yields the following solution for the cut-point x:

a=1(σ)21(σ+)2b=2(μ(σ)2μ+(σ+)2)c=(μσ)2(μ+)2(σ+)2+log(σσ+λ+λ)x=b+b24ac2a(5)

Method 4: maximum likelihood estimator of bimodal distribution (MaxLike)

Weights of both normal distributions are preset (not fitted), according to relative abundance N+/N, as in ‘ParEst’. Distribution parameters (μ+,σ+) and (μ,σ) are then computed as the maximum likelihood estimator [34] via a 4 parameter fit (gradient descent method), directly from the bimodal gene-expression histogram. This fit has to act upon a very flat target function, and therefore we implemented the Newton-gradient algorithm to draw on analytical derivatives.

L(μ+,σ+,μ,σ|x)=j=1Nsample(λ+f+(μ+,σ+|xj)+λf(μ,σ|xj))max(6)

Pr(A|x)=f(A)(x)λ(A)Af(A)(x)λ(A)(7)

Computing the cut-point is achieved again via eq. (4) and eq. (5).

Method 5: expectation maximization (ExMax)

The ExMax-algorithm is a general and very potent mathematical concept, see p. 275 in [35], which can fruitfully be broken down and applied to receptor estimation, as follows.

ExMax not only estimates the parameters of both normal distributions, (μ+,σ+) and (μ,σ), but also their relative weight as an additional parameter, π+, yielding (π=1π+) due to normalization. Hence, ExMax is able to work without any IHC-estimates.

In short the process is as follows: We start from an initial guess for parameters of the two normal distributions ((μ^1,σ^1), (μ^2,σ^2) and π^+=N+/(N++N). It is only this initial guess – not the data used later for the actual estimation – which may be obtained from IHC-data.

Instead of maximizing the likelihood straight away (as in method 4), it is expanded by additional Nsample latent parameters

γ^i,i=1....Nsample(8)

γ^i represents a guess (probability) for sample i to be receptor-positive.

a) These guesses are computed (via a special formula) from the guesses for the ‘real’ parameters of the two normal distributions (μ^1,σ^1), (μ^2,σ^2) and π^+=N+/(N++N).

b) From this expanded maximum likelihood function, new estimates for the ‘real’ parameters are obtained via maximization.

Steps a) and b) are repeated in a loop until convergence. Probability density functions based on these fitted parameters are shown as blue lines in Figure 1, Figure 2 and Figure 3.

Cut-points are obtained via eq. (4) and eq. (5), but λ’s are replaced by calculated π’s.

Note that IHC-results need to be known in advance and individually for each patient to apply methods Youden, LogReg and ParEst (i.e. supervised methods). As opposed, MaxLike draws on the relative abundances λ+=N+/(N++N) only (semi-supervised method). ExMax does not need IHC data at all (unsupervised method), and hence lends itself for analyzing cohorts totally lacking IHC information.

Why to adopt ExMax

Screening the five methods resulted in the following valuation:

• LogReg draws on IHC estimates which may partly be wrong, possibly blurring the decision.

• Youden yields only a cut-point without a confidence interval.

• MaxLike and ExMax yield almost identical results. However, according to our tests, MaxLike seems to perform well only if the abundances of false positive and false negative estimates are about equal, which is incidentally the case in our data. As opposed, ExMax works unaffected by relative abundances. Hence, we preferred ExMax over MaxLike.

• ParEst yields larger confidence intervals than ExMax, see section ‘Obtaining confidence intervals for expression distribution parameters’.

• LogReg and Youden do not draw on normal distributions. Finally, each method yields a specific cut-point for classifying the expression value, xi, see the marks on the ROC-curve (Figure 2), corresponding to the vertical lines in Figure 1 and Figure 3, lower panels.

All proposed methods were found to yield compatible cut-points, see Table 3, and ExMax was finally selected. It yields a PDF (f) for receptor negative and a PDF (f+) for receptor positive.

Obtaining confidence intervals for expression distribution parameters

For the methods ParEst, MaxLike and ExMax, outlined to model receptor gene expression, the accuracy of parameters can be estimated by several methods, such as bootstrap, Leave-One-Out (LOO), cross-validation (CV, via training and validation set) or else by Fisher’s information criterion, FIC, [35]. We will stick to FIC, and it works as follows:

We start from the likelihood function, plug in all parameters (means and standard deviations) as a ‘vector of parameters’, θ, and get:

L(μ+,σ+,μ,σ|x)=L(θ|x)(9)

The so-called Fisher Information matrix is obtained as second partial derivative:

I(θ)=i=1Nsample2logL(θ|xi)θθT(10)

Next, we would like to evaluate the expectation value of I(θ) by integrating over all measured values:

i(θ)=I(θ|xi)f(θ|xi)dx(11)

For a bimodal normal distribution this is not feasible. As an approximation, we instead evaluate I(θ) for the maximum likelihood estimate of the parameters, θ^, as arguments: I(θ^|x). It can be shown mathematically [35] that this converges towards the expectation value.

From statistics one knows that the parameter estimates are normally distributed

θ^N(θ^,i(θ^)1)(12)

We take the main diagonal of this matrix and therefrom construct 95% confidence limits through multiplication by 1.96

θ^j±1.96(i(θ^)1)jj(13)

For results see Tˆable 7

Responsibility functions define lower and upper bounds for gene expression

Each of the four estimation methods (LogReg, ParEst, MaxLike, ExMax) lends itself to construct a ‘responsibility function’, r(x), for the respective sample being receptor-negative (likewise, r+(x) can be constructed for a sample being receptor positive). Each method yields a separate responsibility function, see Figure 7.

Responsibility functions for estrogen receptor-negative.

Figure 7: Responsibility functions for estrogen receptor-negative. Each of the four computational concepts (see legend) yields a separate responsibility function, r, showing the probability that the sample from a given patient is estrogen receptor-negative, based on the gene expression value. A responsibility = 0.5 determines the cut-point in terms of gene expression. Note that the methods MaxLike and ExMax yield coinciding curves (blue and red, appearing as violet). It becomes quite obvious: the steeper the responsibility function declines, the more accurate the limits [ Xlower,Xupper ] can be determined. In any case r(x) is monotonously declining from 1 to 0.

For the methods ParEst, MaxLike and in particular for ExMax, which we finally adopted, responsibility functions are constructed as follows:

Given the two PDFs f and f+ (see Figure 1 and Figure 3), two ‘responsibility functions’ are constructed

r=ff+f+ and r+=f+f+f+(14)

and may be directly expressed in terms of the distribution parameters:

r(x)=(1π+)f(x|μ,σ)(1π+)f(x|μ,σ)+π+f(x|μ+,σ+)r+(x)=π+f(x|μ+,σ+)(1π+)f(x|μ,σ)+π+f(x|μ+,σ+)(15)

The Bayesian concept enters via interpreting π+ as an a priori probability of positive (and 1π+ of negative) receptor status for a given patient. In other words, responsibility functions indicate if the positive or the negative mode (r+(x) or r(x)) is better suited to explain a measured expression value, x. Boundaries of the critical region [ Xlower,Xupper ] are obtained by putting

r(Xlower)=r+(Xupper)=1α(16)

The ‘critical domain’ denotes non-decidable cases with responsibilities between 0.05 and 0.95.

Expression lookup plot

As a prearrangement take a gene expression database of a similar patient cohort and some expression platform, e.g. like the one we have used in this work, as a basis. Perform RMA-normalization as described and generate an ‘expression look-up-plot’ similar to Figure 8. It serves as a reference in the following procedure for the newly diagnosed patient:

Expression look-up-plot: expression versus rank for the estrogen receptor.

Figure 8: Expression look-up-plot: expression versus rank for the estrogen receptor. Probes (Nprobes = 22.215) are ranked according to expression on the x-axis. Arrows indicate the proposed procedure for the newly diagnosed patient: The rank of a receptor gene is embedded within the ranks of all other genes for the very same sample, projected against the curve and the normalized expression value read off the y-axis. This yields a processed value ready for decision making. Values are given for Affymetrix U133A+2.0. Projections for three different ranks are shown: 18.515 → 8.05 (red), 4.319 → 4.75 (blue), 21.917 → 11.19 (green).

a) Take the raw expression values of all probe sets, order them by size and obtain ranks 1 – 22.215 (This number depends on the platform used, here a subset of Affymetrix U133A+2.0 as mentioned in section ‘Normalization’).

b) Locate the rank r of the receptor probe set (e.g. ESR1) among the others e.g. rank 18.515 (within that sample), see Figure 8.

c) Within the ‘expression look-up-plot’ locate the rank of the receptor gene on the rank-axis (x-axis), project it onto the curve and read off the value from the expression axis (y-axis). In math terms, one computes the x = r / Nprobes - quantile.

d) This value, x, comes already in correct dimensions and may directly be compared with the limits of the ‘critical domain’ [ Xlower,Xupper ].

Thus the expression lookup plot yields information directly relevant for improving decision quality based on receptor status.

Abbreviations

IHC: Immunohistochemistry; GE: Gene Expression; ER: Estrogen Receptors; PGR: Progesterone; HER2: Human epidermal growth factor 2; U133A+2.0: Affymetrix Human Genome U133A 2.0 Gene Expression Array; U133A: Affymetrix Human Genome U133A Gene Expression Array; GEO: Gene Expression Omnibus; ESR1: HUGO gene name for ER; RMA- normalization: Robust Multi-array Average normalization; ROC curve: Receiver-Operating-Characteristic curve; CP: Cut-Point; PDFs: Probability Density Functions; DOR: Diagnostic Odds Ratio; ERIHC: Estrogen Receptor, assessed by IHC; ERGE: Estrogen Receptor, assessed by Gene Expression; ExMax: Expectation Maximization; LogReg: Logistic Regression; ParEst: Parameter Estimation of bimodal distribution; MaxLike: Maximum Likelihood estimator of bimodal distribution; RT-qPCR: Real-Time quantitative Polymerase Chain Reaction; LOO: Leave-One-Out; CV: Cross-Validation; AUC: Area Under the Curve HUGO: HUman Genome Organisation; ERBB2: HUGO gene name for HER2.

Author contributions

MK designed the statistical concept, developed the software and performed the calculations.

KS performed data search, retrieval, download and sample evaluations.

DCCT posed the biomedical question and contributed the discussion on IHC measurement quality.

CFS and HK posed the clinical question and formulated the sections on clinical consequences, risk balancing and medical relevance.

MC performed data manipulation and software programming.

WS organized the study and wrote the manuscript.

ACKNOWLEDGMENTS

The software for the analysis is available on request from the authors.

CONFLICTS OF INTEREST

The authors declare that there are no conflicts of interest regarding the publication of this paper.

FUNDING

No financial support was used.

REFERENCES

1. Rhodes A, Jasani B, Balaton A, Miller K. Immunohistochemical demonstration of oestrogen and progesterone receptors: correlation of standards achieved on in house tumours with that achieved on external quality assessment material in over 150 laboratories from 26 countries. J Clin Pathol. 2000; 53:292-301.

2. Hammond ME, Hayes DF, Wolff AC, Mangu PB, Temin S. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Immunohistochemical Testing of Estrogen and Progesterone Receptors in Breast Cancer. J Oncoll Pract. 2010; 6:195-197.

3. Wolff AC, Hammond ME, Schwartz JN, Hagerty KL, Allred DC, Cote RJ, Dowsett M, Fitzgibbons PL, Hanna WM, Langer A, McShane LM, Paik S, Pegram MD, et al. American Society of Clinical Oncology/College of American Pathologists Guideline Recommendations for Human Epidermal Growth Factor Receptor 2 Testing in Breast Cancer. J Clin Oncol. 2006; 25:118-145.

4. Laas E, Mallon P, Duhoux FP, Hamidouche A, Rouzier R, Reyal F. Low concordance between gene expression signatures in ER positive HER2 negative breast carcinoma could impair their clinical application. PLoS One. 2016; 11.

5. Wells CA, Sloane JP, Coleman D, Munt C, Amendoeira I, Apostolikas N, Bellocq JP, Bianchi S, Boecker W, Bussolati G, Connolly CE, Dervan P, Drijkoningen M, et al. Consistency of staining and reporting of oestrogen receptor immunocytochemistry within the European Union - An inter-laboratory study. Virchows Arch. 2004; 445:119-128.

6. Schmidt M, Böhm D, von Törne C, Steiner E, Puhl A, Pilch H, Lehr HA, Hengstler JG, Kölbl H, Gehrmann M. The humoral immune system has a key prognostic impact in node-negative breast cancer. Cancer Res. 2008; 68:5405-5413.

7. Filipits M, Rudas M, Jakesz R, Dubsky P, Fitzal F, Singer CF, Dietze O, Greil R, Jelen A, Sevelda P, Freibauer C, Muller V, Janicke F, et al. A new molecular predictor of distant recurrence in ER-positive, HER2-negative breast cancer adds independent information to conventional clinical risk factors. Clin Cancer Res. 2011; 17:6012-6020.

8. Paik S, Shak S, Tang G, Kim C, Baker J, Cronin M, Baehner FL, Walker MG, Watson D, Park T. A multigene assay to predict recurrence of tamoxifen-treated, node-negative breast cancer. N Engl J Med. 2004; 351:2817-56.

9. Liu MC, Pitcher BN, Mardis ER, Davies SR, Friedman PN, Snider JE, Vickery TL, Reed JP, DeSchryver K, Singh B, Gradishar WJ, Perez EA, Martino S, et al. PAM50 gene signatures and breast cancer prognosis with adjuvant anthracycline- and taxane-based chemotherapy: correlative analysis of C9741 (Alliance). NPJ Breast Cancer. 2016; 2:15023.

10. van 't Veer LJ, Dai H, van de Vijver MJ, He YD, Hart AA, Mao M, Peterse HL, van der Kooy K, Marton MJ, Witteveen AT. Gene expression profiling predicts clinical outcome of breast cancer. Nature. 2002; 415:530-6.

11. Desmedt C, Haibe-Kains B, Wirapati P, Buyse M, Larsimont D, Bontempi G, Delorenzi M, Piccart M, Sotiriou C. Biological processes associated with breast cancer clinical outcome depend on the molecular subtypes. Clin Cancer Res. 2008; 14:5158-5165.

12. Wu G, Stein L. A network module-based method for identifying cancer prognostic signatures. Genome Biology. 2012; 13:R112.

13. Liu R, Guo CX, Zhou HH. Network-based approach to identify prognostic biomarkers for estrogen receptorΓÇôpositive breast cancer treatment with tamoxifen. Cancer Biol Ther. 2015; 16:317-324.

14. Wishart GC, Azzato EM, Greenberg DC, Rashbass J, Kearins O, Lawrence G, Caldas C, Pharoah PDP. PREDICT: A new UK prognostic model that predicts survival following surgery for invasive breast cancer. Breast Cancer Res. 2010; 12: R1.

15. Kaufmann M, Pusztai L, Members BEP. Use of standard markers and incorporation of molecular markers into breast cancer therapy: Consensus recommendations from an International Expert Panel. Cancer. 2011; 117:1575-1582.

16. Zhao X, Rodland EA, Sorlie T, Vollan HK, Russnes HG, Kristensen VN, Lingjaerde OC, Borresen-Dale AL. Systematic assessment of prognostic gene signatures for breast cancer shows distinct influence of time and ER status. BMC Cancer. 2014; 14:211.

17. Gong Y, Yan K, Lin F, Anderson K, Sotiriou C, Andre F, Holmes FA, Valero V, Booser D, Pippen J, Vukelja S, Gomez H, Mejia J, et al. Determination of oestrogen-receptor status and ERBB2 status of breast carcinoma: a gene-expression profiling study. Lancet Oncol. 2007; 8:203-211.

18. Bergqvist J, Ohd JF, Smeds J, Klaar S, Isola J, Nordgren H, Elmberger GP, Hellborg H, Bjohle J, Borg AL, Skoog L, Bergh J. Quantitative real-time PCR analysis and microarray-based RNA expression of HER2 in relation to outcome. Ann Oncol. 2007; 18:845-850.

19. Witzel ID, Milde-Langosch K, Wirtz RM, Roth C, Ihnen M, Mahner S, Zu Eulenburg C, J+ñnicke F, Müller V. Comparison of microarray-based RNA expression with ELISA-based protein determination of HER2, uPA and PAI-1 in tumour tissue of patients with breast cancer and relation to outcome. J Cancer Res Clin Oncol. 2010; 136:1709-1718.

20. Chen X, Li J, Gray WH, Lehmann BD, Bauer JA, Shyr Y, Pietenpol JA. TNBCtype: A subtyping tool for triple-negative breast cancer. Cancer Inform. 2012; 11:147-156.

21. Haynes RB, Sackett OC, Guyatt GH, Tugwell P: Clinical Epidemiology How to do Clinical Practice Research. Ed 3. Philadephia: Lipincott: Williams and Wilkins; 2005.

22. Lawless JF: Statistical Models and Methods for Lifetime Data. New York: John Wiley & Sons; 1982.

23. Regan MM, Viale G, Mastropasqua MG, Maiorano E, Golouh R, Carbone A, Brown B, Suurküla M, Langman G, Mazzucchelli L, Braye S, Grigolato P, Gelber RD, et al. Re-evaluating Adjuvant Breast Cancer Trials: Assessing Hormone Receptor Status by Immunohistochemical Versus Extraction Assays. J Natl Cancer Inst. 2006; 98:1571-1581.

24. Prat A, Ellis MJ, Perou CM. Practical implications of gene-expression-based assays for breast oncologists. Nat Rev Clin Oncol. 2011; 9:48-57.

25. Barrett T, Edgar R. Gene expression omnibus: microarray data storage, submission, retrieval, and analysis. Methods Enzymol. 2006; 411:352-369.

26. Barrett T, Troup DB, Wilhite SE, Ledoux P, Rudnev D, Evangelista C, Kim IF, Soboleva A, Tomashevsky M, Edgar R. NCBI GEO: mining tens of millions of expression profiles—database and tools update. Nucleic Acids Res. 2007; 35:D760-D765.

27. Edgar R, Domrachev M, Lash AE. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 2002; 30:207-210.

28. Irizarry RA, Bolstad BM, Collin F, Cope LM, Hobbs B, Speed TP. Summaries of Affymetrix GeneChip probe level data. Nucleic Acids Res. 2003; 31:e15.

29. Bolstad BM, Irizarry RA, Astrand M, Speed TP. A comparison of normalization methods for high density oligonucleotide array data based on variance and bias. Bioinformatics. 2003; 19:185-193.

30. Gautier L, Cope L, Bolstad BM, Irizarry RA. affy–analysis of Affymetrix GeneChip data at the probe level. Bioinformatics. 2004; 20:307-315.

31. Youden WJ. Index for rating diagnostic tests. Cancer. 1950; 3:32-35.

32. Lu K. On logistic regression analysis of dichotomized responses. Pharm Stat. 2016.

33. van Vliet MH, Reyal F, Horlings HM, van de Vijver MJ, Reinders MJ, Wessels LF. Pooling breast cancer datasets has a synergetic effect on classification performance and improves signature stability. BMC Genomics. 2008; 9:375.

34. Aldrich J. R.A. Fisher and the making of maximum likelihood 1912-1922. Statist Sci. 1997:162-176.

35. Hastie T, Tibshirani R, Friedman J: The Elements of Statistical Learning: Data Mining, Inference, and Prediction. Ed 2: Springer; 2009.

36. Lu X, Lu X, Wang ZC, Iglehart JD, Zhang X, Richardson AL. Predicting features of breast cancer with gene expression patterns. Breast Cancer Res Treat. 2008; 108:191-201.

37. Budczies J, Weichert W, Noske A, Muller BM, Weller C, Wittenberger T, Hofmann HP, Dietel M, Denkert C, Gekeler V. Genome-wide gene expression profiling of formalin-fixed paraffin-embedded breast cancer core biopsies using microarrays. J Histochem Cytochem. 2011; 59:146-157.

38. Hoeflich KP, O'Brien C, Boyd Z, Cavet G, Guerrero S, Jung K, Januario T, Savage H, Punnoose E, Truong T, Zhou W, Berry L, Murray L, et al. in vivo antitumor activity of MEK and phosphatidylinositol 3-kinase inhibitors in basal-like breast cancer models. Clin Cancer Res. 2009; 15:4649-4664.

39. Liu L, Greger J, Shi H, Liu Y, Greshock J, Annan R, Halsey W, Sathe GM, Martin AM, Gilmer TM. Novel mechanism of lapatinib resistance in HER2-positive breast tumor cells: activation of AXL. Cancer Res. 2009; 69:6871-6878.

40. Desmedt C, Giobbie-Hurder A, Neven P, Paridaens R, Christiaens MR, Smeets A, Lallemand F, Haibe-Kains B, Viale G, Gelber RD, Piccart M, Sotiriou C. The Gene expression Grade Index: a potential predictor of relapse for endocrine-treated breast cancer patients in the BIG 1-98 trial. BMC Med Genomics. 2009; 2:40.

41. Desmedt C, Di LA, de AE, Larsimont D, Haibe-Kains B, Selleslags J, Delaloge S, Duhem C, Kains JP, Carly B, Maerevoet M, Vindevoghel A, Rouas G, et al. Multifactorial approach to predicting resistance to anthracyclines. J Clin Oncol. 2011; 29:1578-1586.

42. Korde LA, Lusa L, McShane L, Lebowitz PF, Lukes L, Camphausen K, Parker JS, Swain SM, Hunter K, Zujewski JA. Gene expression pathway analysis to predict response to neoadjuvant docetaxel and capecitabine for breast cancer. Breast Cancer Res Treat. 2010; 119:685-699.

43. Silver DP, Richardson AL, Eklund AC, Wang ZC, Szallasi Z, Li Q, Juul N, Leong CO, Calogrias D, Buraimoh A, Fatima A, Gelman RS, Ryan PD, et al. Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer. J Clin Oncol. 2010; 28:1145-1153.

44. Li Y, Zou L, Li Q, Haibe-Kains B, Tian R, Li Y, Desmedt C, Sotiriou C, Szallasi Z, Iglehart JD, Richardson AL, Wang ZC. Amplification of LAPTM4B and YWHAZ contributes to chemotherapy resistance and recurrence of breast cancer. Nat Med. 2010; 16:214-218.

45. Kao KJ, Chang KM, Hsu HC, Huang AT. Correlation of microarray-based breast cancer molecular subtypes and clinical outcomes: Implications for treatment optimization. BMC Cancer. 2011; 11:143.

46. Dedeurwaerder S, Desmedt C, Calonne E, Singhal SK, Haibe-Kains B, Defrance M, Michiels S, Volkmar M, Deplus R, Luciani J, Lallemand F, Larsimont D, Toussaint J, et al. DNA methylation profiling reveals a predominant immune component in breast cancers. EMBO Mol Med. 2011; 3:726-741.

47. Smeets A, Daemen A, Vanden BI, Gevaert O, Claes B, Wildiers H, Drijkoningen R, Van HP, Lambrechts D, De MB, Neven P, Sotiriou C, Vandorpe T, et al. Prediction of lymph node involvement in breast cancer from primary tumor tissue using gene expression profiling and miRNAs. Breast Cancer Res Treat. 2011; 129:767-776.

48. de CP, Valet F, Gentien D, Lehmann-Che J, Scott V, Tran-Perennou C, Barbaroux C, Servant N, Vacher S, Sigal-Zafrani B, Mathieu MC, Bertheau P, Guinebretiere JM, et al. Importance of pre-analytical steps for transcriptome and RT-qPCR analyses in the context of the phase II randomised multicentre trial REMAGUS02 of neoadjuvant chemotherapy in breast cancer patients. BMC Cancer. 2011; 11:215.

49. Desmedt C, Majjaj S, Kheddoumi N, Singhal SK, Haibe-Kains B, El OF, Chaboteaux C, Michiels S, Lallemand F, Journe F, Duvillier H, Loi S, Quackenbush J, et al. Characterization and clinical evaluation of CD10+ stroma cells in the breast cancer microenvironment. Clin Cancer Res. 2012; 18:1004-1014.

50. Lopez FJ, Cuadros M, Cano C, Concha A, Blanco A. Biomedical application of fuzzy association rules for identifying breast cancer biomarkers. Med Biol Eng Comput. 2012; 50:981-990.

51. Sabatier R, Finetti P, Adelaide J, Guille A, Borg JP, Chaffanet M, Lane L, Birnbaum D, Bertucci F. Down-Regulation of ECRG4, a Candidate Tumor Suppressor Gene, in Human Breast Cancer. PLoS One. 2011; 6:e27656.

52. Miyake T, Nakayama T, Naoi Y, Yamamoto N, Otani Y, Kim SJ, Shimazu K, Shimomura A, Maruyama N, Tamaki Y, Noguchi S. GSTP1 expression predicts poor pathological complete response to neoadjuvant chemotherapy in ER-negative breast cancer. Cancer Sci. 2012; 103:913-920.

53. Clarke C, Madden SF, Doolan P, Aherne ST, Joyce H, O'Driscoll L, Gallagher WM, Hennessy BT, Moriarty M, Crown J, Kennedy S, Clynes M. Correlating transcriptional networks to breast cancer survival: a large-scale coexpression analysis. Carcinogenesis. 2013; 34:2300-2308.

54. Metzger-Filho O, Catteau Al, Michiels S, Buyse M, Ignatiadis M, Saini KS, de Azambuja E, Fasolo V, Naji S, Canon JL, Delrée P, Coibion M, Cusumano P, et al. Genomic Grade Index (GGI): Feasibility in Routine Practice and Impact on Treatment Decisions in Early Breast Cancer. PLoS One. 2013; 8:e66848.

55. Huang CC, Tu SH, Lien HH, Jeng JY, Huang CS, Huang CJ, Lai LC, Chuang EY. Concurrent Gene Signatures for Han Chinese Breast Cancers. PLoS One. 2013; 8:e76421.

56. Prat A, Bianchini G, Thomas M, Belousov A, Cheang MC, Koehler A, Gomez P, Semiglazov V, Eiermann W, Tjulandin S, Byakhow M, Bermejo B, Zambetti M, et al. Research-based PAM50 subtype predictor identifies higher responses and improved survival outcomes in HER2-positive breast cancer in the NOAH study. Clin Cancer Res. 2014; 20:511-521.

57. Jezequel P, Loussouarn D, Guerin-Charbonnel C, Campion L, Vanier A, Gouraud W, Lasla H, Guette C, Valo I, Verriele V, Campone M. Gene-expression molecular subtyping of triple-negative breast cancer tumours: importance of immune response. Breast Cancer Res. 2015; 17:43.

58. Aswad L, Yenamandra SP, Ow GS, Grinchuk O, Ivshina AV, Kuznetsov VA. Genome and transcriptome delineation of two major oncogenic pathways governing invasive ductal breast cancer development. Oncotarget. 2015; 6:36652-36674. http://doi.org/10.18632/oncotarget.5543.

59. Grinchuk OV, Motakis E, Yenamandra SP, Ow GS, Jenjaroenpun P, Tang Z, Yarmishyn AA, Ivshina AV, Kuznetsov VA. Sense-antisense gene-pairs in breast cancer and associated pathological pathways. Oncotarget. 2015; 6:42197-42221. http://doi.org/10.18632/oncotarget.6255.

60. Xiang J, Hurchla MA, Fontana F, Su X, Amend SR, Esser AK, Douglas GJ, Mudalagiriyappa C, Luker KE, Pluard T, Ademuyiwa FO, Romagnoli B, Tuffin G, et al. CXCR4 Protein Epitope Mimetic Antagonist POL5551 Disrupts Metastasis and Enhances Chemotherapy Effect in Triple-Negative Breast Cancer. Mol Cancer Ther. 2015; 14:2473-2485.

61. Burstein MD, Tsimelzon A, Poage GM, Covington KR, Contreras A, Fuqua SA, Savage MI, Osborne CK, Hilsenbeck SG, Chang JC, Mills GB, Lau CC, Brown PH. Comprehensive Genomic Analysis Identifies Novel Subtypes and Targets of Triple-negative Breast Cancer. Clin Cancer Res. 2015; 21:1688-1698.

62. den HP, Rawls K, Tsimelzon A, Shepherd J, Mazumdar A, Hill J, Fuqua SA, Chang JC, Osborne CK, Hilsenbeck SG, Mills GB, Brown PH. Phosphatase PTP4A3 Promotes Triple-Negative Breast Cancer Growth and Predicts Poor Patient Survival. Cancer Res. 2016; 76:1942-1953.


Creative Commons License All site content, except where otherwise noted, is licensed under a Creative Commons Attribution 4.0 License.
PII: 20474