Top Banner
COMPUTATIONAL PATHOLOGY FOR GENOMIC MEDICINE A DISSERTATION SUBMITTED TO THE DEPARTMENT OF BIOMEDICAL INFORMATICS AND THE COMMITTEE ON GRADUATE STUDIES OF STANFORD UNIVERSITY IN PARTIAL FULFILLMENT OF THE REQUIREMENTS FOR THE DEGREE OF DOCTOR OF PHILOSOPHY ANDREW HANNO BECK DECEMBER 2013
153

A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

May 31, 2020

Download

Documents

dariahiddleston
Welcome message from author
This document is posted to help you gain knowledge. Please leave a comment to let me know what you think about it! Share it to your friends and learn new things together.
Transcript
Page 1: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

COMPUTATIONAL PATHOLOGY FOR GENOMIC MEDICINE

A DISSERTATION SUBMITTED TO THE

DEPARTMENT OF BIOMEDICAL INFORMATICS

AND THE COMMITTEE ON GRADUATE STUDIES

OF STANFORD UNIVERSITY

IN PARTIAL FULFILLMENT OF THE REQUIREMENTS

FOR THE DEGREE OF

DOCTOR OF PHILOSOPHY

ANDREW HANNO BECK

DECEMBER 2013

Page 2: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

http://creativecommons.org/licenses/by-nc/3.0/us/

This dissertation is online at: http://purl.stanford.edu/nc361qm2225

© 2013 by Andrew Hanno Beck, MD. All Rights Reserved.

Re-distributed by Stanford University under license with the author.

This work is licensed under a Creative Commons Attribution-Noncommercial 3.0 United States License.

ii

Page 3: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

I certify that I have read this dissertation and that, in my opinion, it is fully adequatein scope and quality as a dissertation for the degree of Doctor of Philosophy.

Daphne Koller, Primary Adviser

I certify that I have read this dissertation and that, in my opinion, it is fully adequatein scope and quality as a dissertation for the degree of Doctor of Philosophy.

Atul Butte

I certify that I have read this dissertation and that, in my opinion, it is fully adequatein scope and quality as a dissertation for the degree of Doctor of Philosophy.

Matt van de Rijn

Approved for the Stanford University Committee on Graduate Studies.

Patricia J. Gumport, Vice Provost for Graduate Education

This signature page was generated electronically upon submission of this dissertation in electronic format. An original signed hard copy of the signature page is on file inUniversity Archives.

iii

Page 4: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

iv

Abstract

The medical specialty of pathology is focused on the transformation of information extracted

from patient tissue samples into biologically informative and clinically useful diagnoses to guide

research and clinical care. Since the mid-19th

century, the primary data type used by surgical

pathologists has been microscopic images of hematoxylin and eosin stained tissue sections. Over

the past several decades, molecular data have been increasingly incorporated into pathological

diagnoses. There is now a need for the development of new computational methods to

systematically model and integrate these complex data to support the development of data-driven

diagnostics for pathology. The overall goal of this dissertation is to develop and apply methods

in this new field of Computational Pathology, which is aimed at: 1) The extraction of

comprehensive integrated sets of data characterizing disease from a patient’s tissue sample; and

2) The application of machine learning-based methods to inform the interpretation of a patient’s

disease state.

The dissertation is centered on three projects, aimed at the development and application

of methods in Computational Pathology for the analysis of three primary data types used in

cancer diagnostics: 1) morphology; 2) biomarker expression; and 3) genomic signatures. First,

we developed the Computational Pathologist (C-Path) system for the quantitative analysis of

cancer morphology from microscopic images. We used the system to build a microscopic

image-based prognostic model in breast cancer. The C-Path prognostic model outperformed

competing approaches and uncovered the prognostic significance of several novel characteristics

of breast cancer morphology. Second, to systematically evaluate the biological informativeness

and clinical utility of the two most commonly used protein biomarkers (estrogen receptor (ER)

and progesterone receptor (PR)) in breast cancer diagnostics, we performed an integrative

Page 5: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

v

analysis over publically available expression profiling data, clinical data, and

immunohistochemistry data collected from over 4,000 breast cancer patients, extracted from 20

published studies. We validated our findings on an independent integrated breast cancer dataset

from over 2,000 breast cancer patients in the Nurses’ Health Study. Our analyses demonstrated

that the ER-/PR+ disease subtype is rare and non-reproducible. Further, in our genomewide

study we identified hundreds of biomarkers more informative than PR for the stratification of

both ER+ and ER- disease. Third, we developed a new computational method, Significance

Analysis of Prognostic Signatures (SAPS), for the identification of robust prognostic signatures

from clinically annotated Omics data. We applied SAPS to publically available clinically

annotated gene expression data obtained from over 3,800 breast cancer patients from 19

published studies and over 1,700 ovarian cancer patients from 11 published studies. Using these

two large meta-datasets, we applied SAPS and performed the largest analysis of subtype-specific

prognostic pathways ever performed in breast or ovarian cancer. Our analyses led to the

identification of a core set of prognostic biological signatures in breast and ovarian cancer and

their molecular subtypes. Further, the SAPS method should be generally useful for future studies

aimed at the identification of biologically informative and clinically useful signatures from

clinically annotated Omics data.

Taken together, these studies provide new insights into the biological factors driving

cancer progression, and our methods and models will support the continuing development of the

field of Computational Pathology.

Page 6: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

vi

Acknowledgements

First, I would like to thank my PhD adviser Daphne Koller. Throughout my PhD

program, Daphne has been an inspiring role model, and her support and encouragement have

been invaluable. Daphne introduced me to the principles of machine learning and taught me how

through careful data modeling, we can both gain new biomedical knowledge and develop data-

driven predictive tools to directly inform clinical decisions. I hope to continue to apply this

important principle throughout my research career. Further, her enthusiasm for learning and

science is contagious, and makes it a joy to do research in her group.

I would like to thank Atul Butte, who has been a great mentor and role model throughout

my time at Stanford. Atul’s boundless enthusiasm, creativity, and passion for translational

bioinformatics drew me to pursue graduate training in this field. Atul taught me many things in

bioinformatics, including the importance of asking interesting questions and coming up with new

ways to answer those questions using data.

I would like to thank Rob Tibshirani, whom I have been very fortunate to collaborate

with on many projects since I began my residency training in Pathology at Stanford and through

my graduate training in Biomedical Informatics. Rob has both taught me an enormous amount

regarding principles of statistical analysis of high dimensional data, and he and his students have

created a set of extremely useful software tools that I continue to apply on an almost daily basis

in my research. It was always a pleasure to work with Rob, who was extremely generous with his

ideas, his attention, and his time.

I would like to thank Matt van de Rijn and Rob West, who have been exceptional

mentors, collaborators, and friends. My relationship with Matt and Rob goes the farthest back;

as I met both while still a medical student, during my interviews for Pathology residency at

Page 7: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

vii

Stanford. I was drawn to the work in their laboratory, which integrates approaches from

genomics and bioinformatics with a deep understanding of disease pathology and

pathophysiology. It was in their laboratory that I gained my first experiences with analyzing

Omics data, and they gave me the opportunity to work on a wide range of interesting and

important projects covering diverse aspects of cancer genomics and bioinformatics. These

experiences were formative in my scientific training, and it was this work, which really cemented

for me the critical (and growing) role of bioinformatics in translational cancer research. The

experiences in their lab played an important role in motivating my choice of pursuing graduate

training in biomedical informatics, and I am fortunate to have had the opportunity to continue to

work with both throughout my PhD.

I would like to thank Stephen Galli, the chairman of the Stanford Department of

Pathology, who was always highly supportive of my research training in Biomedical Informatics,

and helped me to craft an independent residency training program that integrated clinical work

with both protected research time and graduate training in Biomedical Informatics.

I would like to thank Sylvia Plevritis for chairing my oral PhD defense. I am extremely

grateful to Mary Jeanne Oliva, for her support and guidance, at all stages of the PhD program. I

would like to thank Larry Fagan, my BMI academic adviser, for his support of my program and

for his guidance, and Russ Altman for his help in engineering my path to a PhD and for his

support of my training program. I would like to acknowledge the support I received through the

Advanced Residency Training at Stanford fellowship program.

Lastly, I would like to thank my family (Thea, Alma, and Sonya), my parents (Ruth and

Roy), and brother and Sister (Eric and Jody) for their support and encouragement throughout the

PhD.

Page 8: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

viii

Table of Contents

Chapter 1 … Introduction … 1 – 4

Chapter 2 … Morphology … 5 – 43

Chapter 3 … Biomarkers … 44 – 74

Chapter 4 … Signatures … 75 – 123

Chapter 5 … Conclusion … 122 – 124

Appendices … Authorship and Copyright … 125 – 127

List of References … 128 – 142

Page 9: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

ix

List of Tables

Chapter 2

Table 1 (p. 14): Multivariate Cox Proportional Hazards model to predict survival

in NKI (A) and VGH (B).

Table S1 (p. 43): Univariate survival analysis.

Chapter 3

None

Chapter 4

Table 1 (p. 87). Top prognostic signatures in global breast cancer.

Table 2 (p. 90). Top prognostic signatures in ER+/HER2− high proliferation.

Table 3 (p. 94). Top prognostic signatures in ER+/HER2 low proliferation.

Table 4 (p. 95). Top prognostic signatures in HER2+.

Table 5 (p. 96). Top prognostic signatures in ER−/HER2−.

Table 6 (p. 98). Top prognostic signatures in global ovarian cancer.

Table 7(p. 100). Top prognostic signatures in Angiogenic overall.

Table 8 (p. 102). Top prognostic signatures in Non-angiogenic overall.

Table S1 (p. 119). Breast Cancer Datasets.

Table S2 (p. 121). Ovarian Cancer Datasets.

Page 10: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

x

List of Illustrations

Chapter 2

Figure 1 (p. 9). Overview of the image processing pipeline and prognostic model

building Procedure

Figure 2 (p. 11). Kaplan Meier survival curves of the 5YS model predictions and overall

survival on the NKI and VGH datasets.

Figure 3 (p. 17). Top stromal features associated with survival.

Figure 4 (p. 19). Top epithelial features

Figure 5 (p. 20). Kaplan Meier curves of prognostic model build on VGH dataset limited

to top features identified on the NKI dataset

Figure 6 (p. 23). Kaplan Meier survival curves of 5YS model predictions and overall

survival on cases from the VGH cohort stratified according to whether the case

contributed one or multiple TMA cores.

Figure S1 (p. 40). Histologic grade and overall survival.

Figure S2 (p. 41). C-Path 5YS model predictions on VGH with epithelial-stromal

classifications limited to NKI images.

Figure S3 (p. 42). Reproducibility analysis of 5YS model performance.

Chapter 3

Figure 1 (p. 47). Overview of study design and analyses performed.

Figure 2 (p. 57). Genomewide analysis of expression variability in ER+ and ER-negative

breast cancer.

Figure 3 (p. 58). ER and PR subtype frequency and inter-assay concordance.

Page 11: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

xi

Figure 4 (p. 59). Inter-assay agreement confusion matrices for ER/PR subtypes.

Figure 5 (p. 64). Genome-wide survival analysis stratified by ER status.

Figure 6 (p. 65). Cox regression to overall survival.

Chapter 4

Figure 1 (p. 79). Overview of SAPS method.

Figure 2 (p. 86). Global breast cancer Venn diagram and scatterplot.

Figure 3 (p. 89). ER+/HER2− high proliferation Venn diagram and scatterplot.

Figure 4 (p. 91). ER+/HER2− low proliferation Venn diagram and scatterplot.

Figure 5 (p. 94). HER2+ Venn diagram and scatterplot.

Figure 6 (p. 96). ER−/HER2− Venn diagram and scatterplot.

Figure 7 (p. 97). Global ovarian cancer Venn diagram and scatterplot.

Figure 8 (p. 100). Angiogenic subtype Venn diagram and scatterplot.

Figure 9 (p. 102). Non-angiogenic subtype Venn diagram and scatterplot.

Figure 10 (p. 105). Hierarchical clustering of breast and ovarian cancers and their

subtypes based on SAPS scores.

Page 12: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

1

Chapter 1

Introduction

The word pathology is derived from the Ancient Greek πάθος, or pathos, which means

“suffering”, and -λογία, -logia, “the study of”. Thus, pathology is the study of suffering.

Specifically, the study of pathology is focused on the “precise study and diagnosis of disease”.

The practice of pathology dates back to the first half of the third century B.C. (von Staden,

1992). From that time until the mid-19th

century, the primary techniques of the pathologist were

gross analysis of organs removed either during surgery or at autopsy. In the mid-19th

century, led

by the work of Rudolf Virchow and his colleagues, pathology became focused on understanding

the cellular basis of disease through microscopic analyses of diseased tissues and organs

(Ackerknecht, 1953). To this day, the qualitative visual analysis of microscopic images of

disease remains the primary method by which surgical pathologists diagnose disease and guide

treatment decisions (Long, 1962; Malkin, 1993, 1998; Young, 1999).

Despite the importance of morphological data, this is not the only data type currently

available to the pathologist. It is now possible to comprehensively profile alterations in the

genome, transcriptome, and epi-genome from human tissue samples, and major efforts are

underway to use these new data to re-classify disease into more “precise” and data-driven

classifications (NationalResearchCouncil., 2011). Thus, today, and especially in the coming

years, pathologists will be faced with an increasingly complex set of measurements extracted

from patient tissue samples and will need new computational methods for using these

measurements to guide patient care.

Page 13: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

2

In this dissertation, we develop methods for this emerging field of Computational

Pathology. This dissertation makes three primary contributions to this field:

First, we developed a new computational image analysis platform (the Computational

Pathologist). We applied this method to the study of breast cancer and built a highly

accurate image-based prognostic model (Beck et al., 2011). In contrast to previous

approaches to computational image analysis for the analysis of cancer morphology, our

approach was largely unbiased and data-driven. We generated a computational platform

for extracting a rich, quantitative feature set from H&E stained histologic sections of

breast cancer. This feature set includes both standard descriptors of the breast cancer

epithelium and stroma, as well as novel metrics of intra-tumoral heterogeneity and spatial

relationships. In our study, we used the C-Path system to build an accurate image-based

predictor of patient survival. More generally, this platform provides a new means for

transforming cancer morphology into a quantitative high-dimensional data type, which

can be systematically linked with other Omics data types and with treatment response and

clinical outcomes. This work is discussed in Chapter 2 (Morphology: The

Computational Pathologist), based on work published in 2011 in the journal Science

Translational Medicine (Beck et al., 2011).

Second, we performed a systematic re-evaluation of the most commonly used protein

biomarkers used in breast cancer diagnostics (Hefti et al., 2013). We performed an

integrative analysis over a total of greater than 5,000 breast cancer patients, and we

showed that the two most commonly used biomarkers in breast cancer diagnostics

(estrogen receptor and progesterone receptor) show a highly assymetric pattern of co-

expression, with progesterone receptor providing essentially no information for the

Page 14: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

3

stratification of estrogen receptor negative breast cancer. Taking a genomic data-driven

approach, we identify hundreds of protein biomarkers predicted to be more biologically

and clinically informative than progesterone receptor for the stratification of both

estrogen receptor positive and estrogen receptor negative breast cancer. These findings

could both significantly impact breast cancer diagnostics, and more generally, provide a

data-driven framework for the re-evaluation of standard-of-care diagnostics through the

use of publically available clinically annotated Omics data. This approach could be

broadly useful across a range of diseases. This work is discussed in Chapter 3

(Biomarkers: Systematic Re-evaluation of Standard of Care Protein Biomarkers in

Breast Cancer), which is based on work published in 2013 in the journal Breast Cancer

Research (Hefti et al., 2013).

Third, we developed the Significance Analysis of Prognostic Signatures (SAPS) method

for the identification of robust prognostic signatures from clinically annotated Omics

data. A major goal of research in translational cancer genomics is to identify genomic

signatures that are both biologically important and that stratify patients into groups that

show significantly variable survival outcomes. In our study, we show that standard

statistical approaches for identifying biologically important prognostic signatures suffer

from several weaknesses, the most significant being the fact that in certain datasets,

“random” gene sets are able to stratify patients into prognostically variable groups. To

overcome this challenge, we developed a new computational method, which integrates

three significance measures of a signature’s prognostic association, to ensure that a

significant signature is able to: stratify patients into prognostically variable groups, is

enriched for prognostic genes, and performs significantly better than random gene sets at

Page 15: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

4

both of these tasks. In our study, we used SAPS to identify prognostic signatures in breast

and ovarian cancer and their molecular subtypes. More broadly, the SAPS method should

be generally useful for identifying robust prognostic biological signatures from clinically

annotated Omics data. This work is discussed in Chapter 4 (Significance Analysis of

Prognostic Signatures), based on work published in 2013 in the journal PLoS

Computational Biology (Beck et al., 2013).

Page 16: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

5

Chapter 2

Morphology: The Computational Pathologist

This chapter is adapted from work published in the following publication: Beck AH,

Sangoi AR, Leung S, Marinelli RJ, Nielsen TO, van de Vijver MJ, West RB, van de

Rijn M, Koller D. Systematic analysis of breast cancer morphology uncovers stromal

features associated with survival. Sci Transl Med. 2011 Nov 9;3(108):108ra113. doi:

10.1126/scitranslmed.3002564.

Page 17: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

6

Background

In the mid-19th century, it was first appreciated that the process of carcinogenesis

produces characteristic morphologic changes in cancer cells (Müller and West, 1840).

Patey and Scarff showed in 1928 (Patey and Scarff, 1928) that three histologic

features – tubule formation, epithelial nuclear atypia, and epithelial mitotic activity –

could each be scored qualitatively, and the assessments could be combined to stratify

breast cancer patients into three groups that showed significant survival differences.

This semi-quantitative morphological scoring scheme has been refined over the years

(Bloom and Richardson, 1957; Elston and Ellis, 1991; Le Doussal et al., 1989), but

still remains the standard technique for histologic grading in invasive breast cancer.

Although the three epithelial features scored in the current grading systems are

useful in assessing cancer prognosis, valuable prognostic information can also be

derived from other factors, including properties of the cancer stroma, both its

molecular characteristics (Beck et al., 2008; Bergamaschi et al., 2007; Bianchini et al.;

Bissell and Radisky, 2001; Finak et al., 2008; Karnoub et al., 2007; West et al., 2005;

Wiseman and Werb, 2002) and morphological characteristics (such as stromal fibrotic

focus – a scar-like area in the center of a carcinoma (Van den Eynden et al., 2007)).

Although there has been considerable recent effort devoted to molecular profiling for

assessment of prognosis and prediction of treatment response in cancer (Reis-Filho et

al.; Sotiriou and Piccart, 2007), microscopic image assessment is still the most

commonly available, and in some places in the world, the only measurement that is

feasible financially and logistically. Thus, we sought to develop a high-accuracy

image-based predictor to identify new clinically-impactful morphologic phenotypes of

Page 18: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

7

breast cancers, thereby providing new insights into the biological factors driving

breast cancer progression.

The development of such a system could also help address other problems

relevant to the clinical treatment of breast cancer. An important limitation to the

current grading system is that there is considerable variability in histologic grading

among pathologists (Fanshawe et al., 2008), with potentially negative consequences

for determining treatment. An automated system could provide an objective method

for predicting patient prognosis directly from image data. Moreover, once established,

this system could be used in breast cancer clinical trials to provide an accurate,

objective means for assessing breast cancer morphologic characteristics, thus allowing

objective stratification of breast cancer patients based on morphologic criteria and

facilitating the discovery of morphologic features associated with response to specific

therapeutic agents.

Page 19: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

8

Results

We developed the Computational Pathologist (C-Path), a machine learning-based

method for automatically analyzing cancer images and predicting prognosis. To

construct and evaluate the model, we acquired H&E stained histological images from

breast cancer tissue microarrays (TMA). The TMAs contain 0.6 mm diameter cores

(median of 2 cores per case) and represent only a small sample of the full tumor. We

acquired data from two separate and independent cohorts: Netherlands Cancer

Institute (NKI – 248 patients) and Vancouver General Hospital (VGH – 328 patients).

Unlike previous works in cancer morphometry (Baak et al., 1981; Beck et al.,

2007; Cordon-Cardo et al., 2007; Donovan et al., 2008), our image analysis pipeline

was not limited to a pre-defined set of morphometric features selected by pathologists.

Rather, C-Path measures a rich quantitative feature set from the breast cancer

epithelium and the stroma (Fig. 1). Our image processing system first performs an

automated, hierarchical scene segmentation that generates thousands of measurements,

including both standard morphometric descriptors of image objects and higher-level

contextual, relational, and global image features. The pipeline consists of three stages

(Fig. 1A-C). First, we use a set of processing steps to separate tissue from

background; partition the image into small regions of coherent appearance known as

superpixels; find nuclei within the superpixels; and construct nuclear and cytoplasmic

features within the superpixels (Fig. 1A). Within each superpixel, we measured

features of intensity, texture, size, and shape of the superpixel and its neighbors.

Second, to produce more biologically meaningful features, we classified superpixels

as epithelium or stroma (Fig. 1B).

Page 20: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

9

Fig. 1. Overview of the image

processing pipeline and prognostic model building procedure. (A)

Basic image processing and feature

construction. (B) Building an

epithelial/stromal classifier; the

classifier takes as input a set of breast

cancer microscopic images that have

undergone basic image processing and feature construction and that have

had a subset of superpixels hand-

labeled by a pathologist as epithelium

(red) or stroma (green). The super-

pixel labels and feature measurements

are used as input to a supervised

learning algorithm to build an

epithelial/stromal classifier. The

classifier is then applied to new

images to classify superpixels as

epithelium or stroma. (C)

Constructing higher-level

contextual/relational features. After

application of the epithelial/stromal

classifier, all image objects are sub-

classified and colored based on their

tissue region and basic cellular morphologic properties (Epithelial

regular nuclei = red; epithelial

atypical nuclei = pale blue; epithelial

cytoplasm = purple; stromal matrix =

green; stromal round nuclei = dark

green; stromal spindled nuclei = teal

blue; unclassified regions = dark grey;

spindled nuclei in unclassified regions

= yellow; round nuclei in unclassified

regions = grey; background = white

(C, left panel). After the classification

of each image object, a rich feature

set is constructed (D) Learning an

image-based model to predict

survival. Processed images from

patients alive at five years after

surgery and from patients deceased at 5 years after surgery were used to

construct an image-based prognostic

model. After construction of the

model, it was applied to a test set of

breast cancer images (not used in

model building) to classify patients as

high- or low-risk of death by 5 years.

Page 21: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

10

We use a machine learning approach (L1-regularized logistic regression), in which we

hand-labeled super-pixels from 158 images (107 NKI, 51 VGH) and used those to

train the epithelium/stroma classifier. The resulting classifier comprises 31 features

and achieves a classification accuracy of 89% on held-out data. To construct our final

set of features to be used in the prognostic model, we first re-compute the values of the

basic features separately within epithelium and stroma. We sub-classified nuclei as

“typical” or “atypical” and obtained object measurements from contiguous epithelial

and stromal regions, as well as from epithelial nuclei, epithelial atypical nuclei,

epithelial cytoplasm, stromal round nuclei, stromal spindled nuclei, stromal matrix,

and unclassified objects. We computed a range of relational features (Fig. 1C) that

capture the global structure of the sample and the spatial relationships between its

different components, such as mean distance from epithelial nucleus to stromal

nucleus, mean distance of atypical epithelial nucleus to typical epithelial nucleus, or

distance between stromal regions. Overall, this results in a set of 6642 features per

image. For patients with multiple TMA images (208 of 248 NKI patients; 192 of 328

VGH patients), these statistics were summarized as their mean across the images.

Page 22: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

11

The NKI images were used to build an image-feature-based prognostic model

to predict the binary outcome of 5-year survival (5YS model) (Fig. 1D), using L1-

Fig. 2. Kaplan Meier survival curves of the 5YS model predictions and overall survival on the NKI and VGH datasets. Cases classified as high-risk are plotted on the red dotted line

and cases classified as low-risk on the black solid line. The error bars represent 95%

confidence intervals. The y-axis is probability of overall survival, and the x-axis is time in

years. The numbers of patients at risk in the high-risk and low-risk groups at 5 year

intervals are listed beneath the curves. (A) NKI dataset. Patients were stratified into low-

and high-risk groups based on predictions of the 5YS model on held-out cases during cross-

validation. (B)VGH dataset. VGH patients were stratified into low- and high-risk groups

based on predictions of the 5YS model trained on the full NKI data set. In both datasets (A,

B), cases predicted to be high-risk showed significantly worse overall survival than cases

predicted to be low-risk (Log-rank P < 0.001 in both analyses). VGH cases used to train the

epithelial/stromal classifier have been excluded from the analysis.

Page 23: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

12

regularized logistic regression (38). Model performance on the NKI dataset was

assessed by 8-fold cross-validation, where the data set is split into 8 approximately

equal folds, and in each fold, the model was built using 7 of the folds (up to 217 cases)

and evaluated on the held-out fold. For each instance in the data set, we determined

the C-Path model result when that instance was held out during cross-validation,

which allowed this prediction to be used for evaluating model performance on unseen

data. This procedure is known as “pre-validation”(Tibshirani and Efron, 2002). To

further assess performance of the model, we trained the prognostic model on the full

NKI dataset, and tested the model on the VGH data set. We excluded from this

analysis the 42 VGH cases that had been used in training the epithelial-stromal

classifier.

Survival analysis on the NKI Dataset: The pre-validation C-Path 5YS scores were

highly associated with overall survival (Log-rank P <0.001) (Fig. 2A). When cases

were stratified by grade, the C-Path score was significantly associated with survival

within histologic grade 2 tumors (Log-rank P=0.004). The 5YS score did not achieve a

statistically significant association with survival within grade 1 and grade 3 tumors on

the NKI data set.

We next set out to assess the added prognostic value of the C-Path score in

context of other measured prognostic factors, by using a multivariate Cox

proportional-hazards analysis. In addition to standard clinical measurements, the

tumors from all patients in the NKI dataset had previously undergone expression

profiling by microarray, allowing each case to be classified according to several

Page 24: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

13

standard breast cancer molecular signatures: the 70 gene prognosis signature score

(van de Vijver et al., 2002a), the genomic grade index score (Sotiriou et al., 2006a),

the invasiveness gene signature (Liu et al., 2007), the hypoxia gene signature (Chi et

al., 2006a), and intrinsic molecular subtype (Sorlie et al., 2001). The subtype

classifications used in our analysis come from the original publications or from the

supplemental data (Nuyten et al., 2008). The pre-validation C-Path scores were

significantly associated with 5-year survival independent of any of the other clinical or

molecular factors: grade, ER status, age, tumor size, lymph node status, mastectomy,

chemotherapy, 70 gene prognosis signature, hypoxia signature, wound response

signature, genomic grade index, or intrinsic molecular subtypes (P=0.02) (Table 1A).

The only other features significantly associated with survival were the hypoxia

signature and age (Table 1A).

Page 25: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

14

Table 1. Multivariate Cox proportional hazards model to predict survival in NKI (A)

and VGH (B) cohorts.

Table 1A.

The histologic grading scores used in the primary analysis described above

came from manual pathologic interpretation of whole-slide microscopic images by a

centralized review. To directly compare the performance of the C-Path system to

pathological grading on the exact same set of images, we applied standard

pathological grading criteria to the TMA images used in the C-Path analysis (mitotic

. Multivariate Cox Regression (NKI,

n = 248)

exp(coef) exp(-

coef)

Lower

95%

Upper

95%

P value C-Path 5YS Model Score 1.78 0.56 1.11 2.86 0.017 Hypoxia 1.8 0.56 1.09 2.96 0.021 Age 0.79 1.26 0.63 1 0.046 70 gene prognosis signature 2.01 0.5 0.94 4.26 0.070 Size 1.22 0.82 0.94 1.59 0.137 Invasiveness gene signature 1.84 0.54 0.69 4.9 0.226 Mastectomy 1.3 0.77 0.81 2.07 0.276 Wound response siganture 1.44 0.69 0.72 2.89 0.299 ERBB2 molecular subtype 1.88 0.53 0.53 6.67 0.326 Grade 1.19 0.84 0.77 1.83 0.431 Basal molecular subtype 0.68 1.47 0.15 3 0.612 ER 0.82 1.22 0.36 1.87 0.634 LN 1.15 0.87 0.55 2.38 0.715 Genomic grade index 1.09 0.91 0.52 2.32 0.814 Luminal A molecular subtype 1.14 0.87 0.36 3.65 0.819 Luminal B molecular subtype 0.93 1.07 0.29 3.03 0.907 Chemotherapy 1.04 0.96 0.49 2.21 0.925

Table 1B. Multivariate Cox Regression

(VGH, n = 286)

exp(coef) exp(-

coef)

Lower

95%

Upper

95%

P value Age 1.83 0.55 1.49 2.24 <0.0001 Lymph node 1.94 0.52 1.32 2.84 0.0007 C-Path 5YS Model Score 1.54 0.65 1.02 2.32 0.039 ER 0.79 1.27 0.46 1.34 0.376 Mastectomy 0.72 1.39 0.32 1.6 0.417 Size 0.95 1.05 0.85 1.07 0.444 Grade 1.06 0.95 0.74 1.5 0.760

Page 26: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

15

activity, nuclear pleomorphism, and tubule formation were semi-quantitatively scored

from 1 to 3, and the scores were summed with a sum of less than 6 receiving a grade

of 1, sum of 6 to 7 receiving a grade of 2 and sum greater than 7 receiving a grade of

3); the pathologist grading the images was blinded from the survival data. Although

the C-Path predictions were strongly associated with survival, the pathologic grade

derived from TMA cores showed no significant association with survival (Log-rank

P=0.4) on the same TMA images from the NKI data set, highlighting the difficulty of

obtaining accurate prognostic predictions from these small tumor samples.

Survival analysis on the VGH Dataset: We next tested the prognostic model on the

VGH data set, which was not used in constructing the prognostic model. In addition

to being an additional data set, the cases from VGH represented a cohort of patients

with distinct clinical features. The NKI data set was limited to women less than 53

years with Stage I or Stage II breast cancer. In contrast, the VGH data comes from a

population-based cohort with a higher proportion of older women and women with

more advanced disease. A subset of the VGH cases with survival data (51 images from

42 cases) were used for training of the epithelial/stromal classifier, which was built to

classify superpixels as epithelium or stroma and implemented as part of the image

processing pipeline. We excluded these 42 cases from our survival analysis.

The C-Path score was significantly associated with overall survival in this

independent group of cases (Log-rank P=0.001) (Fig. 2B). Notably, the standard

histologic grading scores that had been obtained by routine pathological analysis of

whole slide images using standard grading criteria on the original patient material

Page 27: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

16

showed no significant association with survival on this same cohort of patients (Log-

rank P=0.29), perhaps due to the greater variability of the grading process, in which

grades were assigned independently by individual community pathologists(Fanshawe

et al., 2008). On VGH, significant survival stratification was achieved by the 5YS

model within both grade 2 and 3 tumors (Log-rank P = 0.02 and 0.01, respectively).

We constructed a multivariate Cox proportional hazards model that considered age,

lymph node status, mastectomy, ER status, grade, size, and C-Path 5YS model score.

In this multivariate model, the C-Path 5YS model score, age and lymph node status

were significantly independently associated with patient survival (all P <0.05) (Table

1B). Grade, size, and ER status were not significant independent predictors of survival

in this multivariate model.

Page 28: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

17

To assess the generalizability of the full image processing pipeline, we have

Fig. 3. Top stromal features associated with survival. (A) Variability in absolute difference in

intensity between stromal matrix regions and neighbors. Top panel, high score (24.1); bottom

panel, low score (10.5). (Insets) Top panel, high score; bottom panel; low score. Right panels,

stromal matrix objects colored blue (low), green (medium), or white (high) according to each

object’s absolute difference in intensity to neighbors. (B) Presence of stromal regions without

nuclei. Top panels, high scores; bottom panels, 0 score. Green, stromal contiguous regions with

score 0; red, stromal contiguous regions with high score. (Insets) Red stromal regions are thin and

do not contain nuclei; green regions are larger with nuclei. (C) Average relative border of stromal

spindle nuclei to stromal round nuclei. Top panel, low score; bottom panel, high scoer. (Insets) stromal spindled nuclear objects are green and stromal round nuclear objects are red. Right

panels, higher magnification of a portion of the larger image.

Page 29: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

18

repeated the entire analysis with training of the epithelial/stromal classifier limited

exclusively to the 107 NKI images. This pipeline resulted in decreased performance of

the prognostic model, with statistically significant (Log-rank P < 0.05) survival

stratification observed only on the NKI data set. These findings suggest that a

relatively large, varied set of training images is important for robust performance of

the epithelial/stromal classifier and that accurate epithelial/stromal segmentation is

important for extracting the most prognostically informative morphological features.

Assessing Significance of Features: To identify morphologic features that robustly

contribute to the C-Path model, we performed a bootstrap analysis on the NKI data set

to generate 95% confidence intervals (CI’s) for the coefficient estimates for the image

features in the C-Path model. This analysis revealed 11 features with a 95% CI that

does not include zero. These eleven features included three stromal features (Fig. 3)

and eight epithelial features (Fig. 4).

Page 30: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

19

We assessed correlation of these features with pathological assessment of

Fig. 4. Top epithelial features. The 8 panels in the figure (A-H) each shows one of the top-ranking epithelial features from the

bootstrap analysis. Left panels (improved prognosis), right panels (worse prognosis). (A) Standard deviation of the (standard deviation

of intensity/mean intensity) for pixels within a ring of the center of epithelial nuclei. Left, relatively consistent nuclear intensity pattern

(low score); right, great nuclear intensity diversity (high score). (B) Sum of the number of unclassified objects. Red, epithelial regions;

green, stromal regions; no overlaid color, unclassified region. Left, few unclassified objects (low score); right, higher number of

unclassified objects (high score). (C) Standard deviation of the maximum blue pixel value for atypical epithelial nuclei. Left, high

score; right, low score. (D) Maximum distance between atypical epithelial nuclei. Left, high score; right, low score. (Insets) Red,

atypical epithelial nuclei; black, typical epithelial nuclei. (E) Minimum elliptic fit of epithelial contigiuous regions. Left, high score;

right, low score. (F) Standard deviation of distance between epithelial cytoplasmic and nuclear objects. Left, high score; right, low

score. (G) Average border between epithelial cytoplasmic objects. Left, high score; right, low score. (H) Maximum value of the

minimum green pixel intensity value in epithelial contiguous regions. Left, low score indicating black pixels withi n epithelial region; right, higher score indicating presence of epithelial regions lacking black pixels.

Page 31: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

20

Fig. 5. Kaplan Meier survival curves of prognostic

models built in analysis on VGH data set limited to top

features identified no NKI data set. Cases classified as

high-risk are plotted on the red dotted line and cases

classified as low-risk on the black solid line. The error

bars represent 95% confidence intervals. The y-axis is

probability of overall survival, and the x-axis is time in

years. The numbers of patients at risk in the high-risk

and low-risk groups at 5 year intervals are listed beneath

the curves. (A) Prognostic model built with 3 top stromal

features; (B) Progostic model built with 8 top epithelial

features; (C) Prognostic model built with top epithelial

and stromal features.

epithelial tubule formation, mitotic activity, and nuclear pleomorphism, which are the

standard features used in histologic grading. The top associations of the eleven C-Path

Page 32: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

21

features with pathological grading features were a negative correlation of tubule

formation with “stromal matrix textural variability” (Spearman’s rho = -0.21, p =

0.001) and positive correlation of both mitotic activity and nuclear pleomorphism with

the C-Path feature “number of epithelial nuclei from unclassified regions”

(Spearman’s rho = 0.27 and 0.33 respectively, both p < 0.001).

Seven of the top features in the bootstrap analysis were relational features

characterizing the contextual relationships of epithelial and stromal objects to their

neighbors. Since cancer is a disease of abnormal tumor cell growth and abnormal

cellular relationships between tumor cells and stroma (unlimited replicative potential,

loss of growth inhibition between neighboring transformed cells, cancer cell invasion

of neighboring tissue) (Hanahan and Weinberg, 2000), it is perhaps not surprising that

relational features form key prognostic factors in breast cancer.

To test the prognostic value of the stromal features identified by our analysis,

we tested the predictive performance of stromal features and epithelial features

separately. The model utilizing only stromal features was highly associated with

overall survival in the VGH dataset (Log-rank P=0.004) (Fig. 5) and showed survival

association similar to that of the full C-Path model. For both grade 2 and grade 3

breast cancers, the stromal model predictions were associated with survival (both Log-

rank P <0.05). The predictions from the model comprised solely of epithelial features

was associated with survival overall (Log-rank P =0.02), and this association was

strongest for stratification within histologic grade 3 tumors (Log-rank P =0.002) with

no statistically significant stratification observed in grade 1 and 2 tumors (both Log-

rank P >0.2). Pathologists currently use only epithelial features in the standard

Page 33: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

22

grading scheme for breast cancer and other carcinomas. Our findings suggest that

evaluation of morphologic features of the tumor stroma may offer significant benefits

for assessing prognosis.

The stromal feature with the largest coefficient in the prognostic model was a

measure of the variability of the stromal matrix intensity differences with its neighbors

(Fig. 3A). High values were associated with improved outcome. Breast cancer tissue

that received a high score tended to contain larger contiguous regions of stroma

separated from larger contiguous epithelial regions. This pattern of cancer growth

more closely approximates epithelial-stromal relationships observed in the normal

breast. This pattern results in a high score, because in stroma-rich areas, stromal

matrix regions border exclusively other stromal matrix regions, while in other areas

the stromal matrix directly borders epithelial regions. Cases that receive a low score

tend to have relatively uniform distribution of epithelium and stromal matrix

throughout the image, with thin cords of epithelial cells infiltrating through stroma

across the image, so that each stromal matrix region borders a relatively constant

proportion of epithelial and stromal regions.

Page 34: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

23

The stromal feature

with the second largest

coefficient (Fig. 3B) was

the sum of the minimum

green RGB intensity value

of stromal-contiguous

regions. This feature

received a value of zero

when stromal regions

contained dark pixels (such

as inflammatory nuclei).

The feature received a

positive value when

stromal objects are devoid

of dark pixels. This feature

provides information about

the relationship between

stromal cellular

composition and prognosis and suggests that the presence of inflammatory cells in the

stroma is associated with poor prognosis, a finding consistent with previous

observations (Tan et al., 2011). The third most significant stromal feature (Fig. 3C)

was a measure of the relative border between spindled stromal nuclei to round stromal

nuclei, with an increased relative border of spindled stromal nuclei to round stromal

Fig. 6. Kaplan Meier survival curves of the 5YS model predictions

and overall survival on cases from the VGH cohort, stratified

according to whether the case contributed 1 or multiple TMA cores.

Cases classified as high-risk are plotted on the red dotted line and

cases classified as low-risk on the black solid line. The error bars

represent 95% confidence intervals. The y-axis is probability of

overall survival, and the x-axis is time in years. The numbers of

patients at risk in the high-risk and low-risk groups at 5 year intervals

are listed beneath the curves. (A) C−Path 5YS model predictions on

VGH patients contributing only 1 TMA Core; (B) C-Path 5YS model

predictions on VGH patients contributing multiple TMA cores.

Page 35: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

24

nuclei associated with worse overall survival. While the biological underpinning of

this morphologic feature is currently not known, this analysis suggests that spatial

relationships between different populations of stromal cell types is associated with

breast cancer progression.

Reproducibility of C-Path 5YS Model Predictions on Samples with Multiple

TMA Cores: For the C-Path 5YS model (which was trained on the full NKI dataset),

we assessed the intra-patient agreement of model predictions when predictions were

made separately on each image contributed by patients in the VGH data set. For the

190 VGH patients that contributed 2 images with complete image data, the binary

predictions (high risk or low risk) on the individual images were in agreement with

each other for 69% (131/190) of the cases and agreed with the prediction on the

averaged data for 84% (319/380) of the images. Using the continuous prediction score

(which ranged from 0 to 100), the median of the absolute difference in prediction

score among the patients with replicate images was 5%, and the Spearman correlation

among replicates was 0.27 (p=0.0002). This degree of intra-patient agreement is only

moderate, and these findings suggest significant intra-patient tumor heterogeneity,

which is known to be a cardinal feature of breast carcinomas (Ding et al., 2010;

Marusyk and Polyak, 2010; Shah et al., 2009). Qualitative visual inspection of images

receiving discordant scores suggests that intra-patient variability in both the epithelial

and stromal components is likely to contribute to discordant scores for the individual

images. These differences appeared to relate both to the proportions of epithelium and

stroma as well as to the appearance of the epithelium and stroma. Last, we sought to

Page 36: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

25

analyze whether survival predictions were more accurate on the VGH cases that

contributed multiple cores compared to the cases that contributed only a single core.

This analysis showed that the C-Path 5YS model showed significantly improved

prognostic prediction accuracy on the VGH cases for which we had multiple images,

compared to the cases that contributed only a single image (Fig. 6). Taken together,

these findings show a significant degree of intra-patient variability, and indicate that

increased tumor sampling is associated with improved model performance.

Page 37: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

26

Discussion

We have developed a system for the automatic hierarchical segmentation of

microscopic breast cancer images, and the generation of a rich set of quantitative

features to characterize the image. Based on these features, we built image-based

models to predict patient outcome and to identify clinically significant morphologic

features. Most previous work in quantitative pathology has required laborious steps of

image object identification by skilled pathologists, followed by the measurement of a

small number of expert pre-defined features, primarily characterizing epithelial

nuclear characteristics, such as size, color, and texture (Beck et al., 2007; Mulrane et

al., 2008). In contrast, following initial filtering of images to ensure high-quality

TMA images and training of the C-Path models using expert-derived image

annotations (epithelium vs. stroma labels to build the epithelial/stromal classifier and

survival time and survival status to build the prognostic model), our image analysis

system is automated with no manual steps, which greatly increases its scalability.

Additionally, in contrast to prior approaches, our system measures thousands of

morphologic descriptors of diverse elements of the microscopic cancer image,

including many relational features from both the cancer epithelium and stroma,

allowing identification of prognostic features whose significance was not previously

recognized.

Using our system we built an image-based prognostic model on the NKI dataset and

showed that in this patient cohort the model was a strong predictor of survival and

provided significant additional prognostic information to clinical, molecular, and

pathological prognostic factors in a multivariate model. We also demonstrated that the

Page 38: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

27

image-based prognostic model, built using the NKI data set, is a strong prognostic

factor on another, independent data set with very different characteristics (VGH).

These findings suggest that the C-Path model might be adapted to provide an

objective, quantitative tool for histologic grading of invasive breast cancer in clinical

practice.

A key goal of our project was to use an unbiased data-driven approach to discover

prognostically significant morphologic features in breast cancer. This discovery-based

approach has been widely used in analysis of genomic data, but not yet to the study of

cancer morphology from microscopic images of patient samples. Microscopic images

of cancer samples represent a rich source of biological information, as this level of

resolution facilitates the detailed quantitative assessment of cancer cells’ relationships

with each other, with normal cells, and with the tumor microenvironment, all of which

represent key “hallmarks of cancer”(Hanahan and Weinberg).

Of the top 11 features that were most robustly associated with survival in a

bootstrap analysis, 8 were from the epithelium and 3 from the stroma. A prognostic

model built on only the 3 stromal features was a stronger predictor of patient outcome

than one built from the epithelial features, and is equally as predictive as the model

built from all features. These stromal features included a measure of stromal

inflammation, a process that has previously been implicated in breast cancer

progression (Tan et al., 2011), as well as several stromal morphologic features whose

prognostic significance in breast cancer has not previously been studied. Despite the

growing recognition of stromal molecular characteristics and the tumor

microenvironment in the regulation of carcinogenesis (Beck et al., 2008; Bergamaschi

Page 39: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

28

et al., 2007; Bissell and Radisky, 2001; Finak et al., 2008; Karnoub et al., 2007; West

et al., 2005; Wiseman and Werb, 2002), since the grading of breast cancer began in the

early 20th

century, grading criteria have consisted entirely of epithelial features. Our

analysis suggests that stromal morphologic structure is an important prognostic factor

in breast cancer. Understanding the molecular basis for the prognostically significant

stromal morphologic phenotypes uncovered in our analysis will be informative.

Our study has several limitations, which will need to be addressed prior to

translation of the C-Path system for use in clinical medicine. First, it will be necessary

to establish the effectiveness of the system on whole slide images. All images used in

our study came from breast cancer TMA images. Each TMA image captures only a

minute portion of the full tumor volume, which is much smaller than the multiple

whole-slide images used in routine diagnostic pathology. This fact is both a strength

and limitation of this study. On the one hand, our work demonstrates the ability to

apply image analysis tools within a machine learning framework to build a powerful

microscopic image-based prognostic model from very small samplings of a tumor.

This suggests that C-Path may prove useful for deriving prognostically important

information from small tumor biopsy specimens. On the other hand, it is likely that

we could have derived a more powerful prognostic model by analyzing whole-slide

images, since these might facilitate the generation of additional higher-level features

(such as additional measurements of tumor heterogeneity) and might facilitate more

robust model performance because we would be summarizing our features over a

much larger area of the tumor. Our image processing and machine learning pipeline

is not specific to the use of TMA images and could be adapted and retrained with a

Page 40: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

29

data set of whole-slide images. However, whole slide images will require either

manual or automated identification of breast cancer, since these larger images

typically contain regions of both cancer and normal surrounding breast tissue. The

TMA-based system did not require this step, as the TMA cores tend to sample

exclusively areas of breast cancer. Nevertheless, our results on patients contributing

multiple TMA cores suggest that, once this challenge is addressed, performance of the

prognostic model is likely to improve in the whole-slide regime.

Secondly, the C-Path system must be systematically evaluated on a diverse set

of whole-slide images from different institutions where samples are handled in

different ways. As part of this evaluation, the robustness of the epithelial/stromal

classifier and the prognostic model must be evaluated separately to determine the

robustness of each component of the C-Path system. Our results suggest that, prior to

applying C-Path to additional images from a new institution using a different slide

processing regimen, it may be useful to train the epithelial/stromal classifier on a

subset of images from the new institution. This situation is analogous to standard

pathological evaluation of histologic images from diverse institutions, in which

pathologists use the visual characteristics of known morphologic structures (nuclei,

cytoplasm, epithelium, stroma) from images acquired from a new institution to ‘re-

calibrate’ their visual interpretations prior to applying fixed histologic grading criteria.

Given the ability of our model to generalize across two diverse cohorts, it seems

plausible that only a retraining of the epithelial/stromal classifier will be needed, and

the prognostic features and relative weights in the prognostic model should be robust

across datasets. Based on our experience, re-training of the epithelial-stromal classifier

Page 41: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

30

should require approximately 50-60 images, which can be performed by a trained

pathologist in approximately 1 hour.

Additional validation of our findings in independent cohorts of breast cancer

patients will be useful prior to clinical application of the C-Path system. Our study was

limited to 2 large breast cancer patient cohorts. An important future direction for

research will be testing the model on additional independent cohorts of breast cancer

patients to evaluate more fully the model’s generalizability.

A final critical step for the translation of C-Path to clinical medicine will be the

increased utilization of digital images in routine diagnostic pathology. Even today, the

vast majority of surgical pathology diagnoses are made using images viewed directly

on a light microscope, and digital slide scanners are not routinely used in diagnostic

surgical pathology. Beyond the technical challenges, innovative leadership among

pathologists will be critical for facilitating widespread implementation of quantitative,

digital systems in surgical pathology laboratories (Baak, 2002). However, the

availability of a high-accuracy, robust, automated predictor of cancer prognosis has

significant promise to improve the clinical practice of pathology, especially in parts of

the world where expert pathologists may be in short supply (Hitchcock, 2011).

Although the work reported here has focused on predicting survival for

patients with invasive breast cancer and on discovering morphologic features

associated with prognosis, our unbiased methods are not specific to this setting.

Hence, they can be applied much more broadly. We believe the flexible architecture

of the C-Path system – consisting of the construction of a comprehensive feature set

within a machine learning framework – will enable the application of C-Path to build a

Page 42: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

31

library of image-based models in multiple cancer types, each optimized to predict a

specific clinical outcome, including response to particular pharmacologic agents,

thereby allowing this approach to be used to directly guide treatment decisions.

Page 43: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

32

Methods

Patient Samples: We acquired H&E stained histological images from breast cancer

tissue TMAs from two independent institutions: Netherlands Cancer Institute (NKI –

248 patients represented in TA110-TA116) and Vancouver General Hospital (VGH –

328 patients represented in TA268, TA274, TA280). Images were manually reviewed,

and images were removed that contained out-of-focus areas, less than 10% of tissue

from the TMA core, or folded-over areas of tissue. Approximately 8% of the image

files were removed, leaving a total of 671 NKI and 615 VGH images in the analysis

(images provided on the accompanying website http://tma.stanford.edu/tma_portal/C-

Path/).

Image Processing Pipeline: We developed a customized image processing pipeline

within the Definiens Developer XD image analysis environment (see Supplementary

Methods). The pipeline consists of three stages: basic image processing and feature

construction, training and application of the epithelium/stroma classifier, and

construction of higher-level features. Per image, we computed the mean, standard

deviation (SD), min, max of each feature and ultimately generated a set of 6642

features per image. For patients with multiple images (208 of 248 NKI patients; 192 of

328 VGH patients), these statistics were summarized by their mean across the images

(Supplementary Material).

Learning a Prognostic Model: The NKI images were used to build an image-feature-

based prognostic model to predict the binary outcome of 5-year survival (5YS model).

To focus the model on the most relevant features, we used L1-regularized logistic

regression (38). Model performance on the NKI dataset was assessed by 8-fold cross-

Page 44: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

33

validation; in each fold, the model was built using up to 217 cases of the NKI dataset

and evaluated on the held-out set of 31 cases. If a case from the training set was

censored prior to 5 years (7 of 248 cases), the case was excluded from the training set.

The λ parameter that controls the sparsity of the model was tuned at each fold by

leave-one-out cross validation on the training cases for that fold. During each fold, the

value of lambda was chosen that minimized the binomial deviance on the held out

training cases. The logistic regression model computes a probability of 5 year survival.

To stratify patients into low and high risk groups, we selected the cut-point whose

stratification maximized the statistical significance of the difference in overall survival

between the high- and low-risk groups on the training cases, as indicated by the log

rank test statistic. The model and cut-point were then applied to the held-out cases, so

that all held-out cases received a binary classification. To assess the statistical

significance of the survival stratification observed between cases predicted to be low-

risk vs. high-risk, we computed a Log-Rank p value using the survdiff function in the

R package survival. To assess the statistical significance of feature coefficients in

multivariate cox proportional models, we assessed each feature’s Wald statistic and

associated p value using the function coxph in the R package survival.

To assess the robustness of the logistic regression coefficients, we performed a

bootstrap analysis on the NKI dataset, implemented with the ‘boot’ package (40).

Based on this analysis, for each of the 6642 features, we obtained a 95% CI for the

feature’s coefficient estimate.

To assess performance of the model on the VGH data set, we trained the prognostic

model on the full NKI dataset, and tested the model on the VGH data set.

Page 45: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

34

Image Analysis Procedure:

Image Scanning/Pre-processing:

Images of cores were scanned at 20x using the Ariol scanner (Images provided

at http://tma.stanford.edu/tma_portal/C-Path/). The original images were each

1440 x 2256 pixels. To reduce downstream computation, these images were

cropped by selecting a central region of 720 x 1128 pixels, which removed

most background white-space, while preserving most of the lesional tissue.

Prior to analysis, image contrast was standardized with an auto-contrast

function implemented in Matlab

(http://www.mathworks.com/matlabcentral/fileexchange/10566-auto-contrast).

Image Analysis within Definiens Image Analysis Environment:

The Definiens rule sets used in the analysis are provided at:

http://tma.stanford.edu/tma_portal/C-Path/).

1. Reading in images: Each .jpg image of each core was read into the

workspace with predefined generic image import with one .jpg image per

scene.

2. Segmenting image into super-pixels The epithelial-stromal image layer

was created with the ‘Multiresolution Segmentation’ algorithm applied to

the pixel level. This algorithm applies an optimization procedure that

locally minimizes the average homogeneity for image objects comprised of

Page 46: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

35

pixels. The algorithm takes as input 3 user-defined parameters: a scale

parameter (which influences the size of resulting super-pixels) and shape

and compactness parameters that contribute to the ‘homogeneity criterion’.

We used a scale parameter of 150, shape parameter of 0.5, and

compactness parameter of 0.3. The segmentation algorithm uses a mutual-

best fitting procedure to create image objects that maximize intra-object

homogeneity and inter-object heterogeneity. Additional details describing

the multi-resolution segmentation algorithm are provided in the Definiens

Developer XD 1.3 reference manual. During the training of the

epithelial/stromal classifier, a subset of super-pixels from a total of 158

images (107 NKI, 51 VGH) were manually labeled as epithelium or stroma

(labeled and unlabeled training images provided at

http://tma.stanford.edu/tma_portal/C-Path/).

3. Identifying sub-cellular objects: To identify nuclear regions within the

super-pixels, we first applied an auto threshold algorithm on the layer 1

(Red) pixel values to identify an adaptive threshold for classifying image

objects based on darkness. We then applied a multi-threshold segmentation

algorithm on the pixel level to identify and segment nuclei based on pixel

intensity with a minimum object size of 200 pixels. We then obtained

objects classified as either darker than or lighter than the threshold. This

procedure creates objects based solely on pixel intensity. To use size,

shape, and intensity information to inform segmentation of nuclei, we next

Page 47: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

36

performed multi-resolution segmentation (with a scale parameter of 20,

shape criteria of 0.9, and compactness criteria of 0.1) on the darker objects

obtained from the multi-threshold segmentation. After this step of

segmentation, we sub-classified the object as a “regular nuclei” if its area

was 135 - 750 pixels, roundness less than 0.9, and ratio of length\width less

than or equal to 5. All other darker objects were labeled “atypical nuclei”.

4. Creation of epithelial/stromal classifier: To train the epithelial/stromal

classifier, we exported a total of 112 features from each super-pixel that

had been hand-labeled as either epithelium or stroma. We performed L1

regularized logistic regression to build an epithelial/stromal classifier. The

λ parameter was selected that achieved a classification error within 1

standard error of the minimum classification error on the held-out cases

during 10-fold cross-validation. The resulting model contained 31 features

with non-zero coefficients. This model achieved a cross-validation error of

90% (Supplemental Figure 1).

5. Relabeling of super-pixels: We applied the 31 feature logistic regression

classifier to all super-pixels, which created a probability score indicating

the predicted probability that the super-pixel is epithelial (values > =0.5) or

stromal (values < 0.5). To focus our analysis on high-confidence areas of

epithelium and stroma, we labeled all super-pixels with an epithelial-

stromal classifier score >=0.75 as epithelium and all super-pixels with an

Page 48: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

37

epithelial-stromal classifier score < 0.25 as stroma. We left the remaining

super-pixels as unlabeled.

6. Merging of super-pixels: After the classification of super-pixels as

epithelium or stroma, we merged adjacent super-pixels from the same class

with each other, resulting in the creation of epithelial and stromal super-

pixels, whose size and shape reflected the structure of contiguous epithelial

and stromal regions in the image.

7. Relabeling of sub-cellular objects: We relabeled each sub-cellular

object, based on the classification of its parent super-pixel. This resulted in

the following sub-cellular object classes: epithelial regular nucleus,

epithelial atypical nucleus, epithelial cytoplasm, stromal round nucleus,

stromal spindled nucleus, stromal matrix, unclassified regular nucleus,

unclassified atypical nucleus, and the classification of background for sub-

cellular objects whose parent object had been classified as background.

8. Generation of high-dimensional multi-scale feature set: The preceding

steps carried out a hierarchical segmentation of each image, which broke

the image into two layers of resolution: super-pixel layer and sub-cellular

layer. Each layer comprised a set of objects that each had a classification

label.

Page 49: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

38

a. Super-pixel features: We measured 164 features from each super-

pixel image object. We measured and summarized features

separately for epithelial, stromal, and background super-pixels.

Prior to analysis, we summarized each feature by its mean, min,

max, standard deviation and sum. Measured features include: 1)

standard morphometrical features (super-pixel intensity, size, shape,

and texture); 2) relational features characterizing the local

neighborhood of each super-pixel and distances to each class of

super-pixel; and 3) relational features characterizing the population

of sub-cellular objects underlying each super-pixel.

b. Sub-cellular object features: We measured 188 features from each

sub-cellular image object. We measured and summarized features

separately for epithelial regular nuclei, epithelial atypical nuclei,

epithelial cytoplasm, stromal round nuclei, stromal spindled nuclei,

and background sub-cellular objects. All features were summarized

by mean, min, max, standard deviation, and sum. Measured

features from subcellular objects include: 1) standard

morphometrical features (intensity, size, shape, texture); 2)

relational features characterizing local neighborhood of each sub-

cellular object and typical distance of each object to all classes of

objects; and 3) relational features characterizing relationships

between sub-cellular objects and their parent super-pixel.

Page 50: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

39

c. Global image features: In addition to computing relational

features of each object to each other, we also measured global

image features characterizing the proportion of each image

occupied by the different classes of super-pixel and sub-cellular

objects.

Page 51: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

40

Fig S1.

Page 52: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

41

Fig. S2.

Page 53: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

42

Fig. S3.

Page 54: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

43

Supplemental Tables

Table S1. Univariate survival analysis.

NKI Cohort

Exp(Coef) Exp(-

Coef)

Lower

95%

Upper

95%

P value

5YS Score 2.21 0.45 1.42 3.43 <0.001

Grade 2.30 0.44 1.67 3.16 <0.001

Grade 3 vs.

Rest

2.97 0.34 1.91 4.61 <0.001

Grade 1 vs

Rest

0.25 4.08 0.12 0.51 <0.001

Size 1.39 0.72 1.14 1.69 <0.001

Age 0.95 1.05 0.92 0.99 0.01

Mastectomy 1.17 0.86 0.76 1.80 0.48

Lymph nodes 1.05 0.95 0.68 1.63 0.81

Chemo 1.02 0.98 0.66 1.58 0.92

VGH Cohort

Exp(Coef) Exp(-

Coef)

Lower

95%

Upper

95%

P

5YS Score 1.76 0.57 1.24 2.51 0.002

Age 1.04 0.96 1.03 1.05 <0.001

Mastectomy 0.35 2.9 0.23 0.52 <0.001

Size 1.32 0.76 1.16 1.5 <0.001

Lymph node 1.94 0.51 1.4 2.7 <0.001

Grade 2 vs.

Rest

0.79 1.26 0.59 1.06 0.12

Grade 3 vs.

Rest

1.2 0.83 0.86 1.67 0.28

Grade 1 vs.

Rest

1.15 0.87 0.81 1.64 0.43

Grade 1.03 0.97 0.82 1.28 0.82

Page 55: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

44

Chapter 3

Biomarkers: Data-Driven Re-evaluation of Standard-of-care Breast Cancer

Introduction

This chapter is adapted from: Hefti MM, Hu R, Knoblauch NW, Collins LC, Haibe-

Kains B, Tamimi RM, Beck AH. Estrogen receptor negative/progesterone receptor

positive breast cancer is not a reproducible subtype. Breast Cancer Res. 2013 Aug

23;15(4):R68.

Page 56: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

45

Background

Evaluation of hormone receptor expression is a central component of the pathological

evaluation of breast cancer (Hammond et al., 2010). The biologic, prognostic, and

predictive importance of assessment of estrogen receptor (ER) expression in breast

cancer is well established; however, the added value of progesterone receptor (PR)

assessment is controversial(Colozza et al., 2005; Fuqua et al., 2005; Olivotto et al.,

2004). Despite this uncertainty, the American Society of Clinical Oncology and the

College of American Pathologists recommend testing of both ER and PR on all newly

diagnosed cases of invasive breast cancer (Hammond et al., 2010).

Since the 1970s, it has been hypothesized that PR expression will be associated with

response to hormonal therapies in ER+ breast cancer, as it is thought that ER and PR

co-expression demonstrates a functionally intact estrogen response pathway(Horwitz

et al., 1978; Horwitz and McGuire, 1975, 1978, 1979). Analyses from observational

studies showed that loss of PR expression was associated with worse overall prognosis

among ER+ breast cancers(Bardou et al., 2003; Cancello et al., 2013; Dunnwald et al.,

2007; Grann et al., 2005; Prat et al., 2013). These results suggested that evaluation of

PR status in ER+ breast cancer might be used to help guide clinical management, as

high levels of PR expression may identify a subset of ER+ patients most likely to

benefit from hormonal therapy(Horwitz and McGuire, 1975).

However, a recent meta-analysis of long-term outcomes of 21,457 women with early

stage breast cancer in 20 randomized trials of adjuvant tamoxifen identified ER

Page 57: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

46

expression as the sole pathological factor predictive of response with no significant

independent contribution by PR (relative risk of recurrence following tamoxifen

treatment as compared with placebo or observation was 0.63 (SE 0.03) in the

ER+/PR+ group and 0.60 (S.E. 0.05) in the ER+/PR-neg group)(Davies et al., 2011).

These data show that although PR negativity is associated with a more aggressive

subtype of ER+ breast cancer, evaluation of PR expression cannot be used to identify

ER+ patient subsets most likely to benefit from hormonal therapy. Consequently, the

clinical utility of PR evaluation in ER+ breast cancer is uncertain.

The biological and clinical significance of the ER-neg/PR+ breast cancer subtype is

even more controversial, with some reports claiming it represents a distinct, clinically

useful biologic entity (Rakha et al., 2007; Rhodes and Jasani, 2009), while others posit

that ER-neg/PR+ classification is primarily a technical artifact(De Maeyer et al., 2008;

Nadji et al., 2005) and too rare to be of clinical use(Olivotto et al., 2004). In large

published series, the percentage of ER-neg/PR+ cases has been in the range of

zero(Nadji et al., 2005) to four percent (Bardou et al., 2003; Colditz et al., 2004). In

the Early Breast Cancer Trialists’ Collaborative Group (EBCTCG) meta-analysis, PR

expression was not significantly predictive of tamoxifen treatment response in ER-

negative breast cancer, although there was a slight trend, which failed to reach

statistical significance(Davies et al., 2011). In the EBCTCG analysis, the investigators

noted that as methods for assessment of hormone receptor status have improved, the

proportion of cases reported as ER-neg/PR+ has decreased from ~4% in the early

1990s to only 1% in recent SEER cancer registry data (http://seer.cancer.gov/),

Page 58: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

47

suggesting that as methods of ER testing and interpretation have improved, the rates of

false negative ER results have decreased(Davies et al., 2011). Given the rarity and

uncertain clinical and biological significance of the ER-neg/PR+ classification, it has

been recommended that patients classified as ER-neg/PR+ should undergo repeat ER

testing to rule out a false negative result(Hammond et al., 2010).

Thus, despite the fact that ER and PR evaluation have played central roles in breast

cancer diagnostics and research since the 1970s, it is currently not well established if

the joint assessment of ER and PR stratifies breast cancers into 4 biologically

meaningful and clinically useful subgroups (ER+/PR+, ER+/PR-neg, ER-neg/PR-neg,

and ER-neg/PR+). To provide further insight into the biology of ER and PR

expression and the clinical utility of ER and PR testing in breast cancer, we performed

Figure 1. Overview of study design and analyses performed. MR, medical

record; GEM, gene expression microarray; TMA, tissue microarray.

Page 59: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

48

an integrative analysis, incorporating gene expression profiling data, survival data, and

ER and PR protein expression data from several large cohorts of breast cancer patients

(Figure 1).

The primary aims of our study are to:

1) Determine the frequency and reproducibility of breast cancer subtypes defined by

ER and PR expression levels.

2) Determine the association of PR expression with survival in ER+ and ER-negative

breast cancer and assess the contribution of PR to multivariate prognostic models

including ER and standard clinico-pathologic factors.

Page 60: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

49

Methods

Study overview

An overview of the study design and the set of analyses performed on the GEM and

NHS datasets is shown in Figure 1.

Gene expression microarray (GEM) cohort

We integrated data from a total of 20 previously published gene expression microarray

data-sets. 19 of the data sets were initially provided as supporting material in [20], and

the 20th

data-set comes from The Cancer Genome Atlas (TCGA) breast cancer

cohort(TCGA, 2012). To access the TCGA data, we downloaded the Level 3 loess

normalized Agilent microarray mRNA expression data from the Broad Institute’s

Genome Data Analysis Center

(http://gdac.broadinstitute.org/runs/stddata__2012_12_06/data/BRCA/20121206/).

None of the public gene expression microarray data used in this study required

additional consent to analyze or publish results obtained from the data. Further

description of the data-sets is provided in Additional file 1: Table S1.

Gene expression profiling data scaling and merging

The datasets used in our study were generated using diverse microarray platforms and

originating from different laboratories. We used normalized log2(intensity) for single-

channel platforms and log2(ratio) in dual-channel platforms. Hybridization probes

were mapped to Entrez Gene ID. When multiple probes mapped to the same GeneID,

Page 61: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

50

we used the probe with the highest variance in the dataset under study. We scaled and

centered expression values for each gene to have a mean of zero and standard

deviation of one in the dataset, prior to merging the data from the different data sets.

The complete data-set contains data on 4,111 patients (all with ER and PR

measurements). For the genome-wide analyses, we limited the analysis to the 3,666

patients with valid data from at least 80% of the genes.

ER and PR mRNA expression

We obtained gene expression profiling data on ER and PR mRNA expression from

4,111 patients. Patients were classified as ER+/ER-neg and PR+/PR-neg by modeling

a mixture of 2 Gaussians from the ER mRNA and PR mRNA expression levels

(separately). This procedure was implemented with the Mclust function in the mclust

package in R with equal variance. A similar approach to subtyping was used in(Haibe-

Kains et al., 2012). After subtyping by ER and PR mRNA expression separately,

patients were classified into joint ER/PR categories: ER+/PR+, ER+/PR-neg, ER-

neg/PR-neg, and ER-neg/PR+.

ER and PR protein expression in the GEM dataset

We obtained protein expression data from immunohistochemistry (IHC) from the

clinical data provided in [20] and from the Broad Institute’s Genome Data Analysis

Center

(http://gdac.broadinstitute.org/runs/stddata__2012_12_06/data/BRCA/20121206/) for

Page 62: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

51

patients from TCGA. In total, we obtained matched mRNA and protein expression

data for ER and PR for 1,752 patients in the GEM dataset.

Assessment of agreement between gene expression- and protein-based ER/PR

classifications in the GEM dataset

To assess inter-assay reproducibility, we computed the proportion of cases in each

diagnostic category as determined from the protein expression data in the medical

record (MR) that were classified into the same diagnostic category using the mRNA

expression data. For each binary diagnostic classification schema (ER+/PR+ vs. other;

ER+/PR-neg vs. other; ER-neg/PR-neg vs. other; and ER-neg/PR+ vs. other), we

computed Cohen’s Kappa statistic. The Kappa score is widely used in studies of

diagnostic agreement and interpretation can be aided by published guidelines: (<0 no

agreement; 0–0.2 slight; 0.21-0.40 fair; 0.41-0.60 moderate; 0.61-0.80 substantial;

0.81-1 almost perfect)(Landis and Koch, 1977). Kappa statistics were implemented in

R using the Kappa function in the vcd package.

Survival analyses in the GEM dataset

Univariate survival analysis of gene expression in ER+ and ER-neg breast cancer

We used the survival data and “traditional scaled” breast cancer gene expression

profiling data for 2,731 patients and 13,091 genes provided in(Beck et al., 2013).

Patients were stratified into ER+ (n = 2013, 74%) and ER-neg (n = 718, 26%)

subtypes by modeling a mixture of 2 Gaussians from the ER mRNA expression levels.

Page 63: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

52

Univariate survival analyses were performed using the Cox Proportional Hazards

model, implemented with the coxph function in the survival package in R. The

statistical significance of each gene’s survival association was estimated based on the

gene’s Wald Test P-value in the Cox model. Survival P-Values were adjusted for

multiple hypotheses using the method of Benjamini and Hochberg(Benjamini and

Hochberg, 1995).

Multivariate survival analysis of ER and PR expression levels in breast cancer

We obtained mRNA expression data on ER and PR expression and information on

overall survival, age, grade, lymph node status, and tumor size for 975 patients. We

obtained information on ER and PR protein expression with overall survival, age,

grade, lymph node status, and tumor size for 465 patients. Using these data, we built

multivariate Cox regression models to overall survival.

Data visualization in the GEM dataset

For visualization of the high-dimensional data in our analyses, we produced smoothed

versions of scatterplots with colors representative of the data densities. The smoothed

scatterplots were generated using the smoothScatter function in the graphics package

in R. For our plotting parameters, we used 250 bins for density estimation. The

densities were represented (from least dense to most dense) by the following sequence

of colors: white > beige > grey > black > orange > red.

Page 64: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

53

Nurses’ health study (NHS) cohort

The Nurses’ Health Study cohort was established in 1976 when 121,701 female US

registered nurses ages 30–55 responded to a mail questionnaire that inquired about

risk factors for breast cancer(Colditz and Hankinson, 2005). Every two years, women

are sent a questionnaire and asked whether breast cancer has been diagnosed, and if

so, the date of diagnosis. All women with reported breast cancers (or the next of kin if

deceased) are contacted for permission to review their medical records so as to

confirm the diagnosis. Pathology reports are also reviewed to obtain information on

ER and PR status. Informed consent was obtained from each participant. This study

was approved by the Committee on the Use of Human Subjects in Research at

Brigham and Women’s Hospital.

NHS tissue microarrays and immunohistochemistry

Tissue microarrays (TMAs) have been constructed from paraffin blocks of breast

cancers that developed between 1976 and 2000 among women enrolled in the NHS.

Details of TMA construction and IHC procedures for ER and PR have been previously

described(Tamimi et al., 2008). Briefly, immunohistochemical staining was performed

for ER and PR on 5 μm paraffin sections cut from TMA blocks. Immunostains for

each marker were performed in a single staining run on a Dako Autostainer (Dako

Corporation). The following antibodies and dilutions were used: for ER, a mouse

monoclonal (clone 1D5) from Dako at 1:200 dilution; and for PR, a mouse

monoclonal (PR 636) from Dako at 1:50 dilution. Study pathologists reviewed the

Page 65: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

54

immuno-stained sections under a microscope and estimated the percentage of tumor

cells showing nuclear immuno-reactivity in every tissue core. A case was considered

as positive when there was staining in >1% of the tumor cell nuclei in any of the three

cores from that case, and negative when no nuclear staining was seen in any of the

three cores.

Assessment of agreement between TMA- and medical record-based ER/PR

classifications in NHS

A total of 2011 patients had information on ER and PR status from the medical record

(MR) (28% by IHC, 72% by biochemical assays) and from TMAs (all by IHC). We

computed the proportion of classifications in the MR that received concordant

classifications by TMA and computed Kappa statistics for each of the four ER/PR

subtypes (similar to the analysis in the GEM dataset). We note that in clinical practice

the IHC cut-off for positive ER and PR staining changed from ~10% to 1% over the

course of the study. This change may account for some inflation of the discordance

estimates in the NHS data-set, as the cut-off of 1% was used for interpretation of the

TMAs. We would expect this inflation to affect ER and PR similarly.

Univariate and multivariate survival analyses in NHS

To assess the association of ER and PR expression with survival, we performed

multivariate Cox regression to breast cancer-specific survival, using age, year of

diagnosis, treatment, stage, and grade as co-variates in the models.

Page 66: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

55

Results

PR mRNA tends to be expressed at low-levels in ER-negative breast cancer and

the ER-neg/PR+ subtype is extremely rare

We performed a genome-wide analysis to determine the relative level of PR

expression and variability of PR expression in ER-negative and ER+ breast cancer

(Figure 2). To determine cut-points for ER and PR positivity based on the mRNA

data, we fit a mixture of two Gaussians to the ER mRNA data and PR mRNA data

(separately), which produced a positivity cut-point of −1.3 for ER and 0.4 for PR.

Based on these cut-points, we classified each of 3,666 cancers as ER+ (2,505; 68%) or

ER-negative (1,161; 32%) based on mRNA expression levels. We then computed the

standard deviation of each gene separately in the ER+ and ER-negative cancers. This

analysis demonstrates that PR’s variability is strongly dependent on ER status (Figure

2A). PR shows highly variable expression levels in ER+ breast cancer (PR is more

variable than ~98% of genes in the genome among ER+ cancers). In contrast, PR

expression is highly invariable in ER-negative breast cancer (PR expression is less

variable than >99% of genes in the genome in ER-negative breast cancer). These data

are concordant with the observation that measurement of PR expression can be used to

aid in the stratification of ER+ breast cancer into more- and less-aggressive disease

subtypes(Cancello et al., 2013; Prat et al., 2013; Rakha et al., 2007). The lack of

variation of PR expression in ER-negative breast cancer suggests that it is unlikely PR

will provide clinically or biologically useful information for the stratification of ER-

negative breast cancer.

Page 67: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

56

To gain further insight into the relationship of ER and PR expression, we performed a

scatterplot of ER and PR mRNA expression levels across 4,111 breast cancers (Figure

2B). This analysis shows that ER and PR expression demonstrate a highly asymmetric

relationship, in which PR expression tends to be low/absent in ER-negative breast

cancer, with >95% of ER-negative cases showing relatively low-levels of PR

expression (less than the cut-point of 0.4), while PR expression varies from low-to-

high in ER+ breast cancer, with 43% of ER+ breast cancers showing relatively high

levels of PR expression and 57% of ER+ breast cancers showing relatively low levels

(Figure 2B). Thus, the ER-neg/PR+ subtype is by far the most rare (n = 45; 1%). All

other ER/PR subtypes contain at least 25% of the cancers: ER+/PR+ (n = 1,316; 32%),

ER+/PR-neg (n = 1,720; 42%), and ER-neg/PR-neg (n = 1,030; 25%).

Page 68: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

57

Figure 2. Genomewide analysis

of expression variability in ER+

and ER-negative breast cancer.

This smoothed scatterplot shows the distribution of 11,966 genes

plotted based on their variability

in mRNA levels in ER+ breast

cancer (X axis) and ER-neg breast cancer (Y axis). The color

represents the density of genes

and ranges from white > beige >

grey > black > orange > red, with red the most dense and white the

most sparse. We computed the

standard deviation (SD) of each gene within ER+ cases (n =

2,505) and ER-negative cases (n =

1,161). PR is represented by a red

triangle in the bottom-right portion of the plot, demonstrating

that PR shows highly variable

expression in ER+ breast cancer

(Ranked 157th out of 11,966 genes, 1.3th percentile).

Conversely, PR is one of the least

variable genes in ER-negative

breast cancer (Ranked 11,957th out of 11,966 genes, 99.9th

percentile). B: ER and PR mRNA

expression in GEM dataset. This

smoothed scatterplot shows the distribution of 4,111 breast

tumors. Each tumor is plotted

based on its ER expression level

(X-Axis) and PR expression level (Y-Axis). The color represents the

data density and ranges from

white > beige > grey > black >

orange > red, with red the most dense and white the most sparse.

The jagged black lines represent

the cut-points for converting the

continuous mRNA values into a positive/negative binary score.

The cut-points used were −1.3

and 0.4 for ER and PR,

respectively. Based on these classification boundaries, 1316

(32%) of cases were classified as

ER+/PR+ (+/+), 1720 (42%) as

ER+/PR-neg (+/−), 1030 (25%) as ER-neg/PR-neg (−/−), and 45

(1%) as ER-neg/PR+ (−/+).

Page 69: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

58

We assessed the ER/PR subtypes derived from the protein-based assays in the NHS

and GEM data-sets. The three protein-based analyses showed highly similar

distributions of the ER/PR subtypes (Figure 3), with: 60-66% of cases classified as

ER+/PR+, 13-16% as ER+/PR-neg, 20-21% as ER-neg/PR-neg, and only 1-4% as ER-

neg/PR+. In general, the distributions of ER/PR subtypes were similar in the mRNA

and protein-based analyses, with the exception of a significantly higher proportion of

ER+ cases classified as PR-neg in the microarray data: ~50% of ER+ cases were

classified as PR-neg in the mRNA dataset, compared with only ~20% in the protein

expression data from the GEM dataset (P < 2.2e-16) and 21% and 17% in the NHS

MR and TMA protein-based analyses. In all analyses, the ER-neg/PR+ classification

represented the rarest ER/PR subtype, accounting for between 1 – 4% of cases.

Figure 3. ER and PR subtype frequency and inter-assay concordance. MR,

medical record; GEM, gene expression microarray; TMA, tissue microarray.

Page 70: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

59

Figure 4. Inter-assay agreement confusion matrices for ER/PR subtypes. A and B present 4 ×

4 confusion matrices. A: Gene Expression Microarray (GEM) Dataset. The row and columns

indicate the ER/PR classifications made in the medical record from the GEM dataset (rows) and by

mRNA (columns). The value in each cell in the matrix indicates the proportion of the row’s

subtype that was classified in the column’s subtype. The color represents the proportion agreement

from blue (low) to red (high). B: Nurses’ Health Study (NHS) Dataset. This confusion matrix is

similar to that described in A, but the rows represent the ER/PR classifications from the medical

record in the NHS dataset and the columns represent the classifications made from the NHS TMA

analysis. C: Kappa Values for the gene expression microarray (GEM) and Nurses’ Health Study

(NHS) datasets.

Page 71: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

60

ER-neg/PR+ is the least reproducible breast cancer subtype

To gain further insight into whether ER-neg/PR+ breast cancer represents a true breast

cancer disease subtype, we assessed the inter-assay reproducibility of ER/PR subtypes

for cancers that underwent subtype classification by two methods (mRNA expression

assessment by microarray vs. protein expression reported in the MR in the GEM data-

set; and protein expression recorded in the MR vs. analyzed by IHC on TMAs in the

NHS dataset). For each ER/PR subtype, we computed the proportion of cases in the

MR that received the same classification by the second method, and we computed

Kappa statistics for each ER/PR subtype (Figure 4).

For cases classified as ER+/PR+ by MR in the GEM data-set, 92% were classified as

ER+ by GEM, although this percentage was split between ER+/PR+ (54%) and

ER+/PR-neg (38%). In the NHS dataset, 89% of cases classified as ER+/PR+ by the

MR received the same classification by TMA. The Kappa values for ER+/PR+ were

0.37[95% CI 0.33-0.41] and 0.60[95% CI 0.57-0.64] in the GEM and NHS datasets,

respectively. As would be expected, we see greater inter-assay concordance in the

NHS data-set, as both assays in the NHS dataset are protein-based, while the GEM

dataset analyses are based on the agreement of protein and mRNA expression data.

For cases classified as ER+/PR-neg in the MR in the GEM dataset, 82% were

classified as ER+ in the microarray data, with the ER+/PR-neg category the most

common classification (63%). Similarly, in the NHS for cases classified as ER+/PR-

neg in the MR, 86% were classified as ER+ in the TMA data with a relatively even

Page 72: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

61

split between ER+/PR+ and ER+/PR-neg. The Kappa values for ER+/PR-neg were

0.19[95% CI 0.13-0.24] and 0.37[95% CI 0.30-0.43] in the GEM and NHS datasets,

respectively.

In the GEM dataset, 78% of ER-neg/PR-neg cases in the MR were classified as ER-

neg/PR-neg by microarray. In the NHS dataset, 69% of ER-neg/PR-neg cases in the

MR were classified as ER-neg/PR-neg in the TMA analysis. In both data-sets, the

majority of discordant cases were re-classified as ER+ by the second method (94%

and 86% in the GEM and NHS datasets, respectively), with relatively few ER-neg/PR-

neg cases reclassified as ER-neg/PR+. The Kappa values for ER-neg/PR-neg were

0.65[95% CI 0.61-0.69] and 0.63[95% CI 0.59-0.67] in the GEM and NHS datasets.

The ER-neg/PR+ category showed by far the lowest inter-assay agreement with

concordance of only 2/62 (3%) and 4/71 (6%) of cases classified as ER-neg/PR+ in

the MR in the GEM and NHS datasets, respectively. In both the GEM and NHS data-

sets, the ER-neg/PR+ cases were re-classified relatively evenly into ER+ and ER-

negative subtypes, with a 50/50 and 55/45 split into ER+ and ER-negative subtypes in

the GEM and NHS datasets, respectively. The Kappa values for ER-neg/PR+ were

0.02 [95% CI −0.18 – 0.21] and 0.06[95% CI −0.12 – 0.25] in the GEM and NHS

datasets, indicating no significant agreement (both 95% CI’s include zero).

ER classifications are more reproducible than PR classifications

To gain insight into the individual contributions of ER and PR to the reproducibility of

joint ER/PR assessments, we assessed the inter-assay agreement of ER and PR

Page 73: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

62

separately. In the GEM dataset, there is a higher proportion of concordance for ER

classifications as compared with PR: 1526/1752 [87%] agreement (Kappa = 0.66

[95% CI 0.62 – 0.70]) for ER classifications compared with 1147/1752 [65%]

agreement (Kappa = 0.35 [95% CI 0.31-0.39]) for PR classifications (P for difference

in proportions < 2.2e-16). The NHS dataset shows similar findings, with more

concordance in ER classifications as compared with PR (although the difference are

smaller than seen in the mRNA vs. Protein analysis in the GEM dataset): 1761/2011

[88%] agreement (Kappa = 0.64 [95% CI 0.60-0.69]) for ER vs. 1634/2011 [81%]

agreement (Kappa = 0.59[95% CI 0.55–0.62]) for PR (P for difference in proportions

= 4.3e-8).

We note that these Kappa estimates are likely underestimates of the inter-assay

reproducibility observed in current clinical practice, since: 1) the GEM dataset-based

analysis is comparing mRNA expression with IHC from data obtained across multiple

different institutions; 2) protein expression data in the NHS MR were recorded by

different laboratories, using multiple methods (IHC, biochemical assays), spanning

several decades; and 3) the NHS TMA cases sampled only a subset of the tumor and

did not have the benefit of the whole slide analysis used in routine clinical practice.

Although these factors may produce an underestimate of Kappa values in our study,

we would expect these limitations to affect the Kappa values for ER and PR relatively

similarly, and thus, it is unlikely that these factors confound analyses of the relative

reproducibility of ER compared with PR and of the relative distribution and relative

reproducibility of the combined ER/PR subtypes.

Page 74: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

63

PR mRNA expression and breast cancer prognosis in ER-defined subtypes

Next, we focused our analysis on PR’s prognostic association in ER+ and ER-negative

breast cancer. PR mRNA expression was significantly associated with improved

prognosis in ER+ breast cancer (Adjusted P Value = 0.0003); however, in our

genomewide analysis, we identified hundreds of genes with stronger prognostic

association in ER+ breast cancer (PR’s association was ranked 728th

out of the ~13 K

genes (~6th

percentile), Figure 5, Additional file 1: Table S2). The set of genes more

prognostic than PR in ER+ breast cancer was highly enriched for genes associated

with proliferation and cell cycle (e.g. 12% of this set of genes was associated with the

GO term mitotic cell cycle, FDR for enrichment = 3.4e-32), including the highly

ranked gene AURKA (Adjusted P-Value < 2.4e-13). In agreement with prior studies

[13], we find that (in contrast to PR) ER mRNA expression levels are not associated

with survival in ER+ breast cancer (Figure 5).

~1.3 K genes were identified as significant at an adjusted P-Value of 0.05 in ER-

negative breast cancer. The set of top-ranked prognostic genes in ER-negative breast

cancer was highly enriched for genes involved in the immune response (e.g. 37% of

the genes achieving an adjusted survival P-Value of 1e-4 are associated with the GO

term “immune response”, FDR for enrichment = 1.3e-11). PR expression was not

significantly associated with prognosis in ER-negative breast cancer (Adjusted P-

Value = 0.21).

Page 75: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

64

Figure 5. Genome-wide survival analysis stratified by ER status. This

smoothed scatterplot shows the distribution of the prognostic association of

13,091 genes in ER+ (X-axis) and ER- (Y-axis) breast cancer. The P-values

plotted have been corrected for multiple hypothesis testing using the method of

Benjamini and Hochberg [25]. The color represents the density of genes and

ranges from white > beige > gray > black > orange > red, with red the most

dense and white the most sparse. The dotted black lines represent a significance

threshold of adjusted P = 0.05. The blue triangle represents PGR and the green

triangle represents ESR1. PGR expression is associated with prognosis in ER+

breast cancer; however, 727 genes are more prognostic than PR with the most

prognostic genes showing a prognostic association to the significance level of P

<1 × 10^-12 as compared with the prognostic significance level of 3 × 10–4

achieved by PR.

Page 76: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

65

Survival analyses incorporating ER and PR expression and clinico-pathologic

factors

To further evaluate the clinical significance of ER and PR expression, we built

multivariate prognostic models incorporating ER and PR protein expression and

standard clinico-pathologic factors. In the GEM dataset, a total of 465 patients had ER

and PR protein expression data, covariate data, and overall survival data available.

When either ER or PR was included in multivariate prognostic models considering

age, grade, tumor size (T) and nodal status (N), hormone receptor status was

significantly associated with overall survival (Figure 6). When both ER and PR

protein expression were included in the same multivariate prognostic model, neither

ER nor PR made an independent contribution to the prognostic model. We performed

a similar set of analyses on the NHS data set. To ensure consistent assessment of IHC

staining we used ER and PR as measured on the TMAs, as these were produced and

Figure 6. Cox regression to overall survival. The multivariate regression

analyses to overall survival for the gene expression microarray (GEM) dataset are

adjusted for nodal status, size, age and grade. Nurse’s Health Study (NHS) data

are adjusted for age, year of diagnosis, treatment, stage and grade. Tumor size is

measured in centimeters; nodal status is recorded as positive versus negative.

IHC, immunohistochemistry; OR, odds ratio; TMA, tissue microarray.

Page 77: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

66

interpreted at a central laboratory. Due to the different data points recorded for this

cohort, age, treatment (chemotherapy and endocrine treatment, endocrine treatment

only, chemotherapy only, or no treatment recorded), radiation (present vs. absent),

stage, and grade were included in multivariate models to breast cancer-specific

survival. We found that, as with the protein expression data from the GEM dataset, ER

and PR obtained statistically significant coefficients when included in separate

multivariate Cox models, but neither was significant when both were included in the

same model (Figure 6). To prevent any confounding of inclusion of endocrine

treatment in the prognostic model considering ER and/or PR, we performed the

analysis with the exclusion of the endocrine treatment covariate. We obtained highly

similar results suggesting no significant confounding (Additional file 1: Table S4).

When our analyses were repeated using disease free survival (DFS) in the GEM

dataset, ER by immunohistochemistry was significantly associated with DFS (p =

.002) in a prognostic model considering age, grade, tumor size (T) and nodal status

(N); however PR was not (p = .151) when included in the model (without ER). When

both hormone receptors by IHC were included in a model to DFS, neither obtained a

significant coefficient (p = .67 for PR, .21 for ER), similar to results observed in the

overall survival analyses (Additional file 1: Table S3). When using the mRNA data to

DFS, neither of the hormone receptors achieved significant coefficients when either

one or both were included in prognostic models. However, the GEM dataset was

collected from multiple different institutions, and thus it is possible that different

criteria were used to define DFS at different institutions, which may weaken the DFS

Page 78: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

67

analyses in this meta-dataset. On the NHS dataset, the DFS analysis was largely

concordant with the results from the breast cancer-specific analysis (Additional file 1:

Table S3), with significant (or borderline-significant) coefficients when ER and PR

were included separately in a multivariate model, but non-significant coefficients

when both were included in the same model.

Next, we evaluated the prognostic significance of combined hormone receptor status

(ER+/PR+, ER+/PR-neg, ER-neg/PR-neg). Due to the extremely small sample size of

ER-neg/PR+ cases and to the fact that the ER-neg/PR+ cases did not satisfy the

proportional hazards assumption, we have excluded this classification from the

combined hormonal receptor status multivariate survival analysis. We used the

ER+/PR+ classification as our reference group. In both the GEM and NHS dataset, the

ER+/PR-neg group showed no significant association with decreased survival as

compared with the ER+/PR+ by IHC. By mRNA expression levels in the GEM

dataset, the ER+/PR-neg group was associated with decreased survival.

Page 79: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

68

Discussion

It is recommended that all newly diagnosed breast cancers be evaluated for PR and ER

protein expression by immunohistochemistry(Hammond et al., 2010). The clinical

utility of ER as a predictive biomarker to identify breast cancer patients that will

benefit from hormonal therapy is well established(Davies et al., 2011). The added

clinical value of assessing PR is controversial (Colozza et al., 2005; Fuqua et al.,

2005; Olivotto et al., 2004). The goals of our study were to assess the frequency,

reproducibility, and prognostic association of breast cancer subtypes defined by

ER/PR expression.

Prior work has shown that PR loss in ER+ breast cancer is associated with a more

aggressive subset of ER+ breast cancer (Cancello et al., 2013; Prat et al., 2013; Rakha

et al., 2007). A limitation of most prior studies examining the prognostic significance

of PR expression in ER+ breast cancer is that they have not examined the prognostic

performance of PR relative to other genes, genomewide. It has recently been shown

that a large number of “randomly selected” genes and gene sets obtain statistically

significant associations with patient prognosis in ER+ breast cancer(Venet et al.,

2011), suggesting that prior to inferring the biological significance of a cancer

biomarker (gene or gene signature) based on correlation with survival, it is necessary

to determine the marker’s ability to stratify patients into prognostically variable groups

relative to the performance of randomly selected genes/gene-sets in the dataset(Beck

et al., 2013; Venet et al., 2011).

Page 80: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

69

Our study contributes to the prior literature on the prognostic value of PR expression

in breast cancer, by performing a genomewide survival analysis of ~13 K genes across

~2.7 K patients stratified by ER status. In this analysis, PR expression was associated

with prognosis in ER+ but not ER-negative breast cancer. However, PR was not

among the most strongly prognostic markers in ER+ breast cancer, ranking in the 6th

percentile genomewide, with ~5% of the ~13 K genes in the analysis showing at least

as strong a prognostic association as PR in ER+ breast cancer. Thus, in an unbiased

genomewide search for the most prognostic markers in ER+ breast cancer, PR would

be unlikely to be selected. In our multivariate survival analyses from both the GEM

and NHS datasets, ER and PR were significantly associated with survival in

multivariate survival models considering ER or PR and standard clinco-pathologic

factors; however, when both hormone receptors were included in the same

multivariate model, neither ER nor PR were significant.

The most important attribute of a cancer biomarker is not correlation with patient

prognosis but efficacy in predicting response to specific therapies. It has long been

hypothesized that evaluation of PR expression in ER+ breast cancer could be used to

identify a patient subset most likely to benefit from hormonal therapy(Horwitz and

McGuire, 1975). A recent meta-analysis of 20 randomized clinical trials of tamoxifen

efficacy (n ~ 20 K) demonstrated that both ER+/PR+ and ER+/PR-neg patients show

significant benefit from tamoxifen therapy, and PR is not a useful marker for

predicting tamoxifen response in ER+ breast cancer(Davies et al., 2011). A recent

study evaluating the ability of PR expression to predict benefit from Exemestane vs.

Page 81: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

70

Tamoxifen in ER+ breast cancer similarly identified no association between PR

expression and treatment benefit(Bartlett et al., 2011), providing further evidence to

suggest that PR is a prognostic, but not a predictive biomarker in ER+ breast

cancer(Mackey, 2011). The potential role of PR as a predictive biomarker for

determining benefit from chemotherapy in ER+ breast cancer is also not well defined.

A recent study by Viale et al. (Viale et al., 2008)assessed the added benefit of PR for

predicting response to chemo-endocrine therapy in ER+ breast cancer, and the

investigators did not identify a significant interaction of PR status with

chemotherapeutic regimen in predicting disease free survival. The value of PR for

predicting chemotherapy response in ER+ breast cancer remains uncertain, and this is

an important area for future study.

The biological and clinical significance of PR expression in ER-negative breast cancer

is poorly understood and is controversial. Some studies have suggested that ER-

neg/PR+ breast cancers show distinct clinical and biological features, implying that

ER-neg/PR+ may represent a true breast cancer disease subtype. Other studies have

maintained that ER-neg/PR+ breast cancer is too rare (0 – 0.1% frequency) to

represent a true disease subtype and that as IHC-based methods for ER/PR assessment

improve, the ER-neg/PR+ classification will become even rarer. The recent EBCTG

meta-analysis of randomized trials of tamoxifen efficacy identified a slight trend for

PR expression to be associated with benefit from tamoxifen therapy in ER-negative

breast cancer; although this result did not reach statistical significance(Davies et al.,

2011).

Page 82: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

71

Our study makes two primary contributions to the prior body of literature regarding

ER-neg/PR+ breast cancer. First, we perform a large gene expression microarray-

based analysis incorporating the measurement of mRNA levels of ER and PR from ~4

K breast cancers. We find that PR is one of the least variable genes in ER-neg breast

cancer (ranked 10th

genomewide, <0.1 percentile), and the great majority of ER-

negative cases show low/absent PR expression levels. Thus, ER-neg/PR+ breast

cancer is by far the most rare breast cancer subtype defined by ER/PR expression,

accounting for ~1% of cases in the mRNA-based analyses. We find similar findings in

the protein-based analyses, in which the ER-neg/PR+ subtype is the rarest ER/PR

subtype, accounting for between 1% and 4% of the cases.

The consistency of the observation (both in our study, and in prior studies) that ER-

neg/PR+ breast is by far the most rare breast cancer subtype, accounting for ~1-4% of

cases, establishes that ER and PR show a highly asymmetric pattern of co-expression,

in which ER-negative implies PR-negative, but PR-negative does not imply ER-

negative. These “Boolean implications”(Sahoo et al., 2008) support the long-held

biological model that PR is under the control of ER(Horwitz et al., 1978; Horwitz and

McGuire, 1975, 1978, 1979).

The second major contribution of our study to the characterization of ER-neg/PR+

breast cancer is that we performed an inter-assay reproducibility analysis across two

large and diverse breast cancer datasets, in which ER and PR were assessed by

multiple methods on the same set of tumors. This analysis shows that ER-neg/PR+

breast cancer is by far the least reproducible breast cancer subtype, with the vast

Page 83: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

72

majority (94% and 97% in the 2 datasets) of cases classified as ER-neg/PR+ in the

MR re-classified when testing was performed by a secondary method. The re-

classified cases were relatively evenly split between ER+ and ER-negative subgroups

on repeat testing.

Taken together, our data do not support that ER-neg/PR+ represents a biologically

distinct or clinically useful breast cancer subtype. These data suggest that PR testing is

not warranted in ER-neg breast cancer, as ER-neg/PR+ breast cancer is very rare and

non-reproducible, thus the vast majority of cases classified as ER-neg/PR+ will

represent false classifications. Our data suggests that ER+/PR-neg breast cancer

represents a distinct disease subtype, which accounts for ~15% of breast cancers,

shows fair reproducibility, and is associated with worse prognosis as compared with

ER+/PR+ breast cancer; however, our genomewide analysis identifies hundreds of

genes that are significantly more prognostic than PR in ER+ breast cancer, suggesting

that other candidate prognostic biomarkers are likely to outperform PR for predicting

patient survival in ER+ breast cancer. Further, until there is data to establish that PR is

a predictive (and not merely prognostic) marker in ER+ breast cancer (and

outperforms competing predictive biomarkers in ER+ breast cancer), the clinical

rationale for routine PR testing in ER+ breast cancer will remain uncertain.

Page 84: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

73

Conclusions

The College of American Pathologists and American Society of Clinical Oncology

recommend ER and PR testing for all newly diagnosed cases of invasive breast

cancer(Hammond et al., 2010). While the clinical and biological importance of ER in

breast cancer is well-established, the added clinical benefit of PR evaluation is

uncertain. In our integrative analysis, incorporating gene expression profiling data,

immunohistochemistry data, and clinical data across two large and diverse datasets,

we find that:

1. PR tends to be expressed at low levels in ER-negative breast cancer.

2. PR expression is not associated with prognosis in ER-negative breast cancer.

3. ER-neg/PR+ breast cancer is not a reproducible subtype.

Thus, PR testing is of uncertain clinical utility in ER-negative breast cancer. The

clinical utility of measuring PR expression in ER+ breast cancer is also not well-

defined. Several studies (including ours) show that loss of PR expression is associated

with a more aggressive subset of ER+ breast cancer; however, it is important to note

that testing for PR expression currently provides no clinically actionable information

in ER+ breast cancer, as patients will receive endocrine therapy regardless of PR

status and there is no consensus as to whether knowledge of PR expression by IHC has

a role in informing the use of chemotherapy in ER+ breast cancer. Further, our study

identifies hundreds of genes that are more prognostic than PR in ER+ breast cancer

demonstrating that it is unlikely that PR will emerge as a top-performing prognostic

Page 85: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

74

biomarker in ER+ breast cancer. Therefore, there is currently no strong evidence to

support the clinical utility of routine PR testing in ER+ or ER-negative breast cancer.

Given that breast cancer is the most common cancer diagnosed in women, eliminating

PR testing from the routine diagnostic work-up of invasive breast cancer could save

the health care industry tens of millions of dollars per year, with no loss in the clinical

utility of the pathological evaluation.

Page 86: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

75

Chapter 4

Significance Analysis of Prognostic Signatures

This chapter is adapted from work published in the following publication: Beck AH,

Knoblauch NW, Hefti MM, Kaplan J, Schnitt SJ, Culhane AC, Schroeder MS, Risch

T, Quackenbush J, Haibe-Kains B. Significance analysis of prognostic signatures.

PLoS Comput Biol. 2013;9(1):e1002875.

Page 87: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

76

Background

The identification of pathways that predict prognosis in cancer is important for

enhancing our understanding of the biology of cancer progression and for identifying

new therapeutic targets. There are three widely-recognized breast cancer molecular

subtypes, “luminal” (ER+/HER2−)(Ivshina et al., 2006; Loi et al., 2007a; Paik et al.,

2006; Sotiriou et al., 2006b), “HER2-enriched” (HER2+)(Desmedt et al., 2008; Staaf

et al., 2010) and “basal-like” (ER−/HER2−)(Desmedt et al., 2008; Sabatier et al.,

2011;(Teschendorff and Caldas, 2008; Teschendorff et al., 2007) and a considerable

body of work has focused on defining prognostic signatures in these(Sotiriou and

Pusztai, 2009; Weigelt et al., 2010). Several groups have analyzed prognostic

biological pathways across breast cancer molecular subtypes (Desmedt et al., 2008;

Iwamoto et al., 2011; Wirapati et al., 2008); a tacit assumption is that if a gene

signature is associated with prognosis, it is likely to encode a biological signature

driving carcinogenesis.

Recent work by Venet et al. has questioned the validity of this assumption by

showing that most random gene sets are able to separate breast cancer cases into

groups exhibiting significant survival differences(Venet et al., 2011). This suggests

that it is not valid to infer the biologic significance of a gene set in breast cancer based

on its association with breast cancer prognosis and further, that new rigorous statistical

methods are needed to identify biologically informative prognostic pathways.

Page 88: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

77

To this end, we developed Significance Analysis of Prognostic Signatures

(SAPS). The score derived from SAPS summarizes three distinct significance tests

related to a candidate gene set's association with patient prognosis. The statistical

significance of the SAPSscore is estimated using an empirical permutation-based

procedure to estimate the proportion of random gene sets achieving at least as

significant a SAPS score as the candidate prognostic gene set. We apply SAPS to a

large breast cancer meta-dataset and identify prognostic genes sets in breast cancer

overall, as well as within breast cancer molecular subtypes. Only a small subset of

gene sets that achieve statistical significance using standard statistical measures

achieves significance using SAPS. Further, the gene sets identified by SAPS provide

new insight into the mechanisms driving breast cancer development and progression.

To assess the generalizability of SAPS, we apply it to a large ovarian cancer

meta-dataset and identify significant prognostic gene sets. Lastly, we compare

prognostic gene sets in breast and ovarian cancer molecular subtypes, identifying a

core set of shared biological signatures driving prognosis in ER+ breast cancer

molecular subtypes, a distinct core set of signatures associated with prognosis in ER−

breast cancer and ovarian cancer molecular subtypes, and a set of signatures associated

with improved prognosis across breast and ovarian cancer.

Page 89: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

78

Results

Significance Analysis of Prognostic Signatures

The assumption behind SAPS is that to use a prognostic association to indicate

the biological significance of a gene set, a gene set should achieve three distinct and

complimentary objectives. First, the gene set should cluster patients into groups that

show survival differences. Second, the gene set should perform significantly better

than random gene sets at this task, and third, the gene set should be enriched for genes

that show strong univariate associations with prognosis.

To achieve this end, SAPS computes three p-values (Ppure, Prandom,

and Penrichment) for a candidate prognostic gene set. These individual P-Values are

summarized in the SAPSscore. The statistical significance of the SAPSscore is estimated

by permutation testing involving permuting the gene labels (Figure 1)

Page 90: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

79

Overview of SAPS method

To compute the Ppure, we stratify patients into two groups by performing k-means

clustering (k = 2) of an n×p data matrix, consisting of the n patients in the

Figure 1. Overview of SAPS method.

Page 91: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

80

dataset and the p genes in the candidate prognostic gene set. We then compute a log-

rank P-Value to indicate the probability that the two groups of patients show no

survival difference (Figure 1A).

Next, we assess the probability that a random gene set would perform as well as the

candidate gene set in clustering cases into prognostically variable groups. This P-

Value is the Prandom. To compute the Prandom, we randomly sample genes to create

random gene sets of similar size to the candidate gene set. We randomly sample r gene

sets, and for each random gene set we determine a using the procedure described

above. The Prandom is the proportion of at least as significant as the true

observed Ppure for the candidate gene set (Figure 1B).

Third, we compute the Penrichment to indicate if a candidate gene set is enriched for

prognostic genes. While the procedure to compute the Ppure uses the label determined

by k-means clustering with a candidate gene set as a binary feature to correlate with

survival, the procedure to compute thePenrichment uses the univariate prognostic

association of genes within a candidate gene to produce a gene set enrichment score to

indicate the degree to which a gene set is enriched for genes that show strong

univariate associations with survival (Figure 1C). To compute the Penrichment, we first

rank all the genes in our meta-dataset according to their concordance index by using

the function concordance.index in the survcomp package in R(Schroder et al., 2011).

The concordance index of a gene represents the probability that, for a pair of patients

randomly selected in our dataset, the patient whose tumor expresses that gene at a

Page 92: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

81

higher level will experience the appearance of distant metastasis or death before the

other patient. Based on this genome-wide ranking we perform a pre-ranked

GSEA(Subramanian et al., 2007; Subramanian et al., 2005) to identify the candidate

gene sets that are significantly enriched in genes with either significantly low or high

concordance indices. The GSEA procedure for SAPS has two basic steps. First, an

enrichment score is computed to indicate the overrepresentation of a candidate gene

set at the top or bottom extremes of the ranked list of concordance indices. This

enrichment score is normalized to account for a candidate gene set's size. Second, the

statistical significance of the normalized enrichment score is estimated by permuting

the genes to generate the Penrichment (See (Subramanian et al., 2007; Subramanian et al.,

2005))for further description of pre-ranked GSEA procedure), which indicates the

probability that a similarly sized random gene set would achieve at least as extreme a

normalized enrichment score as the candidate gene set (Figure 1C).

The SAPSscore for each candidate gene set is then computed as the negative log10 of the

maximum of the (Ppure, Prandom, and Penrichment) times the direction of the association

(positive or negative) (Figure 1D). For a given candidate gene set,

the SAPSscore specifies the direction of the prognostic association as well as indicates

the raw P-Value achieved on all 3 of the (Ppure prognosis, Prandom prognosis, and Penrichment).

Since we take the negative log10 of the maximum of the (Ppure prognosis,Prandom prognosis,

and Penrichment), the larger the absolute value of the SAPSscore the more significant the

prognostic association of all 3 P-Values. The statistical significance of the SAPSscore is

determined by permuting genes, generating a null distribution for the SAPSscore and

computing the proportion of similarly sized gene sets from the null distribution

Page 93: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

82

achieving at least as large an absolute value of the SAPSscore as that observed with the

candidate gene set.

When multiple candidate gene sets are evaluated, after generating each gene set's

raw SAPSP-Value by permutation testing, we account for multiple hypotheses and

control the false discovery rate using the method of Benjamini and

Hochberg(Benjamini and Hochberg, 1995) to generate the SAPSq-value (Figure 1E). In

our experiments, we have required a minimum absolute value (SAPSscore) of greater

than 1.3 and a maximum SAPSq-value of less than 0.05 to consider a gene set

prognostically significant. These thresholds ensure that a significant prognostic gene

set will have achieved a raw P-Value of less than or equal to 0.05 for each

of Ppure, Prandom, andPenrichment, and will have achieved an overall SAPSq-Value of less

than or equal to 0.05.

Application and Validation

We chose two model systems to investigate the performance of SAPS. The first is a

curated sample of breast cancer datasets previously described in Haibe-Kains et

al. (Haibe-Kains et al., 2012). Our analysis focused on nineteen datasets with patient

survival information (total n = 3832) (Table S1). The second dataset was a

compendium of twelve ovarian cancer datasets with survival data, as described in

Bentink et al.(Bentink et al., 2012), which includes data from 1735 ovarian cancer

patients for whom overall survival data were available (Table S2).

Identifying Molecular Subtypes

Page 94: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

83

In breast cancer, we used SCMGENE(Haibe-Kains et al., 2012) as implemented in

the R/Bioconductor genefu package to assign patients to one of four molecular

subtypes: ER+/HER2− low proliferation, ER+/HER2− high proliferation,

ER−/HER2− and HER2+. In ovarian cancer, we used the ovcAngiogenic

model [21] as implemented in genefu to classify patients as having disease of either

angiogenic or non-angiogenic subtype.

Data Scaling and Merging

One challenge in the analysis of large published datasets is the heterogeneity of the

platforms used to collect data. To standardize the data, we used normalized

log2(intensity) for single-channel platforms and log2(ratio) in dual-channel platforms.

Hybridization probes were mapped to Entrez GeneID as described in Shi et al.(Shi et

al., 2006) using RefSeq and Entrez whenever possible; otherwise mapping was

performed using IDconverter (http://idconverter.bioinfo.cnio.es)(Alibes et al., 2007).

When multiple probes mapped to the same Entrez GeneID, we used the one with the

highest variance in the dataset under study.

To allow for simultaneous analysis of datasets from multiple institutions, we tested

two data merging protocols. First, we scaled and centered each expression feature

across all patients in each dataset (standard Z scores), and we merged the scaled data

from the different datasets (“traditional scaling”). In a second scaling procedure, we

first assigned each patient in each data set to a breast or ovarian cancer molecular

subtype, using the SCMGENE(Haibe-Kains et al., 2012) and ovcAngiogenic(Bentink

et al., 2012) models, respectively. We then scaled and centered each expression

Page 95: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

84

feature separately within a specific molecular subtype within each dataset, so that each

expression value was transformed into a Z score indicating the level of expression

within patients of a specific molecular subtype within a dataset (“subtype-specific

scaling”).

After merging datasets, we removed genes with missing data in more than half of the

samples and we removed samples that were missing data on more than half of the

genes or for which there was no information on distant metastasis free survival (for

breast) or overall survival (for ovarian). The resulting breast cancer dataset contained

2731 cases with 13091 unique Entrez gene IDs and the ovarian cancer dataset had

1670 cases and 11247 unique Entrez gene IDs for. For each of these reduced data

matrices, we estimated missing values using the function knn.impute in the impute

package in R(Troyanskaya et al., 2001).

Given that breast cancer is an extremely heterogeneous disease with well-defined

disease subtypes, and a primary objective of our work is to identify subtype-specific

prognostic pathways in breast cancer, we focus our subsequent analyses on the

subtype-specific scaled data. Given that ovarian cancer subtypes are more subtle and

less well defined than breast cancer molecular subtypes, we focus our subsequent

analyses in ovarian cancer on the traditional scaled data. SAPS scores in breast and

ovarian cancer generated from the two different scaling procedures showed moderate

to strong correlation across the breast and ovarian cancer molecular subtypes.

Gene Sets

Page 96: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

85

We downloaded gene sets from the Molecular Signatures Database

(MSigDB) (Subramanian et al., 2005)

(http://www.broadinstitute.org/gsea/msigdb/collections.jsp)

(“molsigdb.v3.0.entrez.gmt”). MSigDB contains 5 major collections (positional gene

sets, curated gene sets, motif gene sets, computational gene sets, and GO gene sets)

comprising of a total of 6769 gene sets. We limited our analysis to gene sets with less

than or equal to 250 genes and valid data for genes included in the meta-data sets,

resulting in 5320 gene sets in the breast cancer analysis and 5355 in the ovarian cancer

analysis.

Application of SAPS to Breast Cancer

We first applied SAPS to the entire collection of breast cancer cases independent of

subtype. Of the 5320 gene sets evaluated, 1510 (28%) achieved a raw P-Value of 0.05

by Ppure, 1539 (29%) by Penrichment, 755 (14%) by Prandom, 581 (11%) by all 3 raw P-

Values, and 564 (11%) of these are significant at theSAPSq-value of 0.05 (Figure 2).

Page 97: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

86

Figure 2 Global breast cancer Venn diagram and scatterplot.

Page 98: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

87

The top-ranked gene sets identified by SAPS and associated with poor prognosis in all

breast cancers independent of subtype contained gene sets previously found to be

associated with poor prognosis in breast cancer (Table 1). Thus it is not surprising that

these emerged as the most significant, and this result serves as a measure of validation.

We note that the list of top gene sets associated with poor breast cancer prognosis

identified in our overall analysis includes the gene set

VANTVEER_BREAST_CANCER_METASTASIS_DN, which according to the

Molecular Signatures Database website is defined as “Genes whose expression is

significantly and negatively correlated with poor breast cancer clinical outcome

(defined as developing distant metastases in less than 5 years).” Our analysis suggests

that the set of genes is positively correlated with poor breast cancer clinical outcome.

Comparison the gene list to the published “poor prognosis” gene list from van't Veer

et al. [26] confirms that the gene list is mislabeled in the Molecular Signatures

Database and is in fact the set of genes positively associated with metastasis in van't

Veer et al. [26]

Table 1 Top prognostic signatures in global breast cancer.

Page 99: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

88

The top-ranking gene sets associated with good prognosis were not originally

identified in breast cancers, and represent a range of biological processes. Several

were from analyses of hematolymphoid cells, including: genes down-regulated in

monocytes isolated from peripheral blood samples of patients with mycosis fungoides

compared to those from normal healthy donors, genes associated with the IL-2

receptor beta chain in T cell activation, and genes down-regulated in B2264-19/3 cells

(primary B lymphocytes) within 60–180 min after activation of LMP1 (an oncogene

encoded by Epstein Barr virus). These gene sets suggest that specific subsets of

immune system activation are associated with improved breast cancer prognosis,

consistent with reports that the presence infiltrating lymphocytes is predictive of

outcome in many cancers.

We then applied SAPS to the ER+/HER2− high proliferation subtype. Of the 5320

gene sets evaluated, 1503 (28%) achieved a raw P-Value of 0.05 by Ppure, 1667 (31%)

by Penrichment, 1079 (20%) byPrandom, 675 (13%) by all 3 raw P-Values, and all 675 of

these are significant at the SAPSq-value of 0.05. The top-ranking gene sets by

SAPSscore are associated with cancer and proliferation. One of the top-ranking gene

sets was associated with Ki67, a well-known prognostic marker in Luminal B breast

cancers [27]. Overall, the patterns of significance are highly similar to that seen in

breast cancer analyzed independent of subtype (Figure 3, Table 2).

Page 100: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

89

Figure 3 ER+/HER2− high proliferation Venn diagram and scatterplot.

Page 101: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

90

Table 2 Top prognostic signatures in ER+/HER2− high proliferation.

Next, we used SAPS to analyze the ER+/HER2− low proliferation samples. Of the

5320 gene sets evaluated, 494 (9%) achieved a raw P-Value of 0.05 by Ppure, 1113

(21%) by Penrichment, 939 (18%) byPrandom, 303 (6%) by all 3 raw P-Values, and all 303

of these were significant at the SAPSq-value of 0.05. The top-ranking ER+/HER2− low

proliferation prognostic gene sets by SAPSscore are also highly enriched for genes

involved in proliferation (Figure 4, Table 3). Top ranking gene sets associated with

good prognosis include those highly expressed in lobular breast carcinoma relative to

ductal and inflammation-associated genes up-regulated following infection with

human cytomegalovirus.

Page 102: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

91

Figure 4: ER+/HER2− low proliferation Venn diagram and scatterplot.

Page 103: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

92

Table 3 Top prognostic signatures in ER+/HER2 low proliferation.

Then, we applied SAPS to the HER2+ subset. Of the 5320 gene sets evaluated, 1247

(23%) achieved a raw P-Value of 0.05 by Ppure, 1425 (27%) by Penrichment, 683 (13%)

by Prandom, 439 (8%) by all 3 raw P-Values, and 342 (6%) of these are significant at

the SAPSq-value of 0.05. Most of the top-ranking prognostic pathways in the HER2+

group by SAPSscore are associated with better prognosis and include several gene sets

associated with inflammatory response (Figure 5, Table 4). A gene set containing

genes down-regulated in multiple myeloma cell lines treated with the hypomethylating

agents decitabine and trichostatin A was significantly associated with improved

prognosis in HER2+ breast cancer. The top-ranking gene set associated with decreased

survival is a hypoxia-associated gene set. Hypoxia is a well-known prognostic factor

in breast cancer(Buffa et al., 2010; Chi et al., 2006b), and our analysis suggests it

shows a very strong association with survival in the HER2+ breast cancer molecular

subtype.

Page 105: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

94

Table 4 Top prognostic signatures in HER2+.

Finally, we used SAPS to analyze the poor-prognosis “basal like” subtype which was

classified as being ER−/HER2−. Of the 5320 gene sets evaluated, 786 (15%) achieved

a raw P-Value of 0.05 by Ppure, 1208 (23%) by Penrichment, 304 (6%) by Prandom, 126

(2%) by all 3 raw P-Values, and 25 (0.5%) of these are significant at the SAPSq-value of

0.05. Top-ranking gene sets associated with poor survival include genes up-regulated

in MCF7 breast cancer cells treated with hypoxia mimetic DMOG, genes down-

regulated in MCF7 cells after knockdown of HIF1A and HIF2A, genes regulated by

hypoxia based on literature searches, genes up-regulated in response to both hypoxia

and overexpression of an active form of HIF1A, and genes down-regulated in

fibroblasts with defective XPC (an important DNA damage response protein) in

response to cisplatin (Figure 6, Table 5). This analysis suggests that hypoxia-

associated gene sets are key drivers of poor prognosis in HER2+ and ER−/HER2−

breast cancer subtypes. Interestingly, cisplatin is an agent with activity in ER−/HER2−

breast cancer, and it is has been suggested that ER−/HER2− breast cancers with

Page 106: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

95

defective DNA repair may show increased susceptibility to cisplatin(Silver et al.,

2010).

Page 107: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

96

Figure 6 ER−/HER2− Venn diagram and scatterplot.

Table 5 Top prognostic signatures in ER−/HER2−:

Application of SAPS to Ovarian Cancer

Our analysis for ovarian cancer was similar to that for breast cancer. We began by

applying SAPS to the entire collection of ovarian cancer samples independent of

subtype. Of the 5355 gene sets evaluated, 1190 (22%) achieved a raw P-Value of 0.05

by Ppure, 1391 (26%) by Penrichment, 755 (14%) byPrandom, 497 (9%) by all 3 raw P-

Values (Figure 7, Table 6), and all 497 of these are significant at the SAPSq-value of

0.05. The top gene sets are involved in stem cell-related pathways and pathways

related to epithelial-mesenchymal transition, including genes up-regulated in HMLE

cells (immortalized non-transformed mammary epithelium) after E-cadhedrin (CDH1)

knockdown by RNAi, genes down-regulated in adipose tissue mesenchymal stem cells

vs. bone marrow mesenchymal stem cells, genes down-regulated in medullary breast

cancer relative to ductal breast cancer, genes down-regulated in basal-like breast

Page 108: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

97

cancer cell lines as compared to the mesenchymal-like cell lines, genes up-regulated in

metaplastic carcinoma of the breast subclass 2 compared to the medullary carcinoma

subclass 1, and genes down-regulated in invasive ductal carcinoma compared to

invasive lobular carcinoma.

Figure 7 Global ovarian cancer Venn diagram and scatterplot.

Page 109: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

98

Table 6 Top prognostic signatures in global ovarian cancer.

We then analyzed the angiogenic subtype. Of the 5355 gene sets evaluated, 1153

(22%) achieved a rawP-Value of 0.05 by Ppure, 1377 (26%) by Penrichment, 624 (12%)

by Prandom, 371 (7%) by all 3 raw P-Values (Figure 7, Table 6), and all of these are

significant at the SAPSq-value of 0.05. Top-ranking gene sets associated with poor

prognosis in the angiogenic subtype include: a set of targets of miR-33 (associated

with poor prognosis) (Figure 8, Table 7). This microRNA has not previously been

implicated in ovarian carcinogenesis. Other top hits include several immune response

gene sets, which were associated with improved prognosis.

Page 110: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

99

Figure 8 Angiogenic subtype Venn diagram and scatterplot.

Page 111: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

100

Table 7 Top prognostic signatures in Angiogenic overall.

Finally, we analyzed the non-angiogenic subtype of ovarian cancer. Of the 5355 gene

sets evaluated, 981 (18%) achieved a raw P-Value of 0.05 by Ppure, 957 (18%)

by Penrichment, 658 (12%) by Prandom, 261 (5%) by all 3 raw P-Values (Figure 7, Table

6), and of these, 254 (5%) are significant at the SAPSq-value of 0.05 (Figure 9, Table 8).

The top ranked pathways associated with improved survival are immune-related gene

sets and a gene set found to be negatively associated with metastasis in head and neck

cancers.

Page 113: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

102

Table 8 Top prognostic signatures in Non-angiogenic overall.

Integrated Analysis of Breast and Ovarian Cancer Prognostic Pathways

To assess similarities and differences in prognostic pathways in both breast and

ovarian cancer molecular subtypes, we performed hierarchical clustering of the disease

subtypes using SAPSscores. Specifically, we identified the 1300 gene sets with SAPSq-

value≤0.05 and absolute value (SAPSscore)≥1.3 in at least one of the breast and ovarian

cancer molecular subtypes. We clustered the gene sets and disease subtypes using

hierarchical clustering with complete linkage and distance defined as one minus

Spearman rank correlation (Figure 10). This analysis shows two dominant clusters of

disease subtypes, with one cluster containing ER+/HER2− high proliferation and

ER+/HER2− low proliferation breast cancer molecular subtypes, and the second

cluster containing ovarian cancer molecular subtypes and the ER−/HER2− and

HER2+ breast cancer molecular subtypes. SAPSscores for within ER+ breast cancer

molecular subtypes, within ER−/HER2− and HER2+ breast cancer molecular

subtypes, and within ovarian cancer molecular subtypes show high correlation

Page 114: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

103

(Spearman rho = 0.61, 0.68, and 0.51, respectively, all p<2.2×10−16

).

Interestingly, the SAPSscores for the ER−/HER2− and HER2+ breast cancer subtypes

show far greater correlation with the SAPSscores in the ovarian cancer molecular

subtypes than with the SAPSscores in ER+ molecular subtypes (median Spearman rho is

0.5 for correlation of ER−/HER2− and HER2+ breast cancer molecular subtypes with

ovarian cancer molecular subtypes vs. 0.16 for ER− molecular subtypes with ER+

molecular subtypes (Figure 10). This analysis demonstrates the importance of

performing subtype-specific analyses in breast cancer, as breast cancer is an extremely

heterogeneous disease and prognostic pathways in ER−/HER2− and HER2+ breast

cancer subtypes are far more similar to prognostic pathways in ovarian cancer than

with prognostic pathways in ER+ breast cancer subtypes. Recently, the TCGA breast

cancer analysis demonstrated that the “basal” subtype of breast cancer (ER−/HER2−)

showed genomic alterations far more similar to ovarian cancer than to other breast

cancer molecular subtypes (TCGA, 2012). Our findings show that ER−/HER2− breast

cancers share not only genomic alterations but also prognostic pathways with ovarian

cancer.

Page 115: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

104

Figure 10

Hierarchical clustering of breast and ovarian cancers and their subtypes based

on SAPS scores.

Examining the clusters of gene sets with differential prognostic associations across

breast and ovarian cancer molecular subtypes shows three predominant clusters of

gene sets. The first cluster is predominantly composed of proliferation-associated gene

sets. The second cluster comprised a mixture of EMT-associated gene sets, gene sets

Page 116: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

105

associated with angiogenesis, and with developmental processes. The third is

comprised predominantly of gene sets associated with inflammation.

The proliferation cluster of gene sets is strongly associated with poor prognosis in

breast cancer overall and ER+ breast cancer subtypes. This supports prior studies

demonstrating that proliferation is the strongest factor associated with prognosis in

breast cancer overall(Venet et al., 2011) and in its ER+ molecular subtypes(Desmedt

et al., 2008). Interestingly, the proliferation cluster of gene sets shows little association

with survival in ER−/HER2− and HER2+ breast cancer and ovarian cancer and its

subtypes, and it is the EMT, hypoxia, angiogenesis, and development-associated

cluster of gene sets that are associated with poor prognosis in these diseases/subtypes

with these pathways showing little association with poor prognosis in ER+ breast

cancer. The cluster of immune-related pathways tends to show association with

improved prognosis across breast and ovarian cancer and their subtypes (Figure 10).

Discussion

A significant body of work has focused on identifying prognostic signatures in breast

cancer. Recently, Venet et al. showed that most random signatures are able to stratify

patients into groups that show significantly different survival(Venet et al., 2011). This

work suggests that more sophisticated and statistically rigorous methods are needed to

identify biologically informative gene sets based on observed prognostic associations.

Here we describe such a statistical and computational framework (Significance

Analysis of Prognostic Signature (SAPS)) to allow robust and biologically informative

prognostic gene sets to be identified in disease. The basic premise of SAPS is that in

Page 117: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

106

order for a candidate gene set's association with prognosis to be used to imply its

biological significance, the gene set must satisfy three conditions.

First, the gene set should cluster patients into prognostically variable groups. The p

value generated from this analysis is the standard Ppure, which has been frequently

used in the literature to indicate a gene set's clinical and biological relevance for a

particular disease. A key insight of the SAPS method (building on the work of Venet

et al.(Venet et al., 2011)) is that clinical utility and biological relevance of a gene set

are two very different properties, necessitating distinct statistical tests.

The Ppure assesses the statistical significance of survival differences observed between

two groups of patients stratified using a candidate gene set, and thus this test provides

insight into the potential clinical utility of a gene set for stratifying patients into

prognostically variable groups; however, this statistical test provides no information to

compare the prognostic performance of the candidate gene set with randomly

generated (“biologically null”) gene sets. We believe that it is essential for a candidate

prognostic gene set to not only stratify patients into prognostically variable groups, but

to do so in a way that is significantly superior to a random gene set of similar size.

Therefore, the second condition of the SAPS method is that a gene set must stratify

patients significantly more effectively than a random gene set. This analysis produces

the Prandom. ThePrandom directly compares the prognostic association of a candidate

gene set with the prognostic association of “biologically null” random gene sets.

Lastly, to avoid selecting a gene set that is linked to prognosis solely by the

unsupervised k-means clustering procedure, the SAPS procedure additionally requires

a prognostic gene set to be enriched for genes that show strong univariate associations

Page 118: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

107

with prognosis. Therefore, the third condition of the SAPS method is that a candidate

gene set should achieve a statistically significant Penrichment, which is a measure of the

statistical significance of a candidate gene set's enrichment with genes showing strong

univariate prognostic associations. Our results in breast and ovarian cancer and their

molecular subtypes demonstrate that the Penrichment shows only moderate overall

correlation with the Ppure and Prandom (range Spearman rho = (0.23–0.35),

median Spearman rho = 0.30)) and there is only moderate overlap between

gene sets identified at a raw p value of 0.05 by Ppure, Prandom, and Penrichment (Figures

2A–9A). These data suggest that the Penrichmentprovides useful additional information to

the Ppure and Prandom and allows prioritization of gene sets that are enriched for genes

showing strong univariate prognostic associations.

Summarizing these three distinct statistical tests into a single score is a difficult task as

they were each generated using different methods and they test different hypotheses.

We chose to use the maximum as the summary function (as opposed to a median or

average, for example), as the maximum is a conservative summary measure and it is

easily interpretable. It is important to note that the SAPS method provides users with

the SAPSscore as well as all 3 component P values (and the 3 component q-values

corrected for multiple hypotheses to control the FDR), and therefore the user can

choose to use the SAPSscore or to focus on a particular SAPS component, as desired for

the specific experimental question being evaluated. Importantly, the SAPS method

also performs a permutation-test to estimate the statistical significance of gene

set's SAPSscore.

Page 119: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

108

To test the utility of SAPS in providing insight into prognostic pathways in cancer, we

performed a systematic, comprehensive, and well-powered analysis of prognostic gene

signatures in breast and ovarian cancers and their molecular subtypes. This represents

the largest meta-analysis of subtype-specific prognostic pathways ever performed in

these malignancies. The analysis identified new prognostic gene sets in breast and

ovarian cancer molecular subtypes, and demonstrated significant variability in

prognostic associations across the diseases and their subtypes.

We find that proliferation drives prognosis in ER+ breast cancer, while pathways

related to hypoxia, angiogenesis, development, and expression of extracellular matrix-

associated proteins drive prognosis in ER−/HER2− and HER2+ breast cancer and

ovarian cancer. We see an association of immune-related pathways with improved

prognosis across all subtypes of breast and ovarian cancers. Our analysis demonstrates

that prognostic pathways in HER2+ and ER−/HER2− breast cancer are far more

similar to prognostic pathways in angiogenic and non-angiogenic ovarian cancer than

to prognostic pathways in ER+ breast cancer. This finding parallels the recent

identification of similar genomic alterations in ovarian cancer and basal-like

(ER−/HER2−) breast cancer (TCGA, 2012).

These results demonstrate the importance of performing subtype-specific analyses to

gain insight into the factors driving biology in cancer molecular subtypes. If molecular

subtype is not accounted for, prognostic gene sets identified in breast cancer are

strongly associated with proliferation (Venet et al., 2011); however, when subtype is

accounted for, significant and highly distinct pathways (showing no significant

Page 120: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

109

association with proliferation) are identified as driving prognosis in ER− breast cancer

subtypes. Overall, these data show the utility of performing subtype-specific analyses

and using SAPS to test the significance of prognostic pathways. Furthermore, our data

suggest that ER− breast cancer subtypes and ovarian cancer may share common

therapeutic targets, and future work should address this hypothesis.

In summary, we believe SAPS will be widely useful for the identification of

prognostic and predictive biomarkers from clinically annotated genomic data. The

method is not specific to gene expression data and can be directly applied to other

genomic data types. In the future, we believe that prior to reporting a prognostic gene

set, researchers should be encouraged (and perhaps required) to apply the SAPS (or a

related) method to ensure that their candidate prognostic gene set is significantly

enriched for prognostic genes and stratifies patients into prognostic groups

significantly better than the stratification obtained by random gene sets.

Page 121: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

110

Methods

Breast Cancer Datasets

Our analysis included 19 datasets with survival data (total n = 3832) (Table

S1).

Ovarian Cancer Datasets

Our analysis included 1735 ovarian cancer patients for whom overall survival data

were available (Table S2).

Molecular Subtype Classification

For breast cancer, the SCMGENE model(Haibe-Kains et al., 2012) was used in the

R/Bioconductor genefu packageto stratify patients into four molecular subtypes:

ER+/HER2− low proliferation, ER+/HER2− high proliferation, ER−/HER2− and

HER2+. In the ovarian datasets we used ovcAngiogenic model(Bentink et al., 2012) as

implemented in genefu.

Creation of Meta-Data Sets

For genes with multiple probes, we selected the probe with the highest variance. We

tested two procedures for merging of data: subtype-specific scaling, and traditional

Page 122: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

111

(non subtype-specific scaling) (as described in “Data-Scaling and Merging” portion of

the manuscript). We excluded genes and cases with more than 50% of data missing.

From these reduced data matrices, we imputed missing values using

the impute package in R(Troyanskaya et al., 2001). These pre-processed meta-data

sets are included as Supporting Information in Dataset S1 for both breast and ovarian

cancer using subtype-specific and traditional scaling.

Gene Sets

Gene sets from the Molecular Signatures Database

(MSigDB) [17](http://www.broadinstitute.org/gsea/msigdb/collections.jsp)

(“molsigdb.v3.0.entrez.gmt”). Analyses were limited to gene sets of size greater than 1

and less than or equal to 250 genes.

Application of the Significance Analysis of Prognostic Signatures (SAPS)

Procedure and Visualization of SAPS P Values

The SAPS procedure is described in “Significance Analysis of Prognostic Signatures

(SAPS)” portion of the manuscript. Briefly, for a candidate gene set, SAPS generates

3 component p-values: Ppure,Prandom, and Penrichment. The SAPSscore is the maximum of

these values. The Ppure is the standard log-rank p value, computed by performing K-

means clustering with a k of 2 and assessing the statistical significance of the survival

difference between the 2 resulting clusters, implemented using the survdiff function in

the R package survival and extracting the chi-square statistic for a test of equality of

the 2 survival curves. To compute the Prandom, we generate a distribution of Ppure from

Page 123: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

112

“random” gene sets (we used 10000 random gene sets for a sequence of 8 gene set

sizes ranging from 5 to 250), and we calculate the proportion of random gene sets of a

similar size to the candidate gene sets that achieve aPpure at least as significant as the

true Ppure. To compute the Penrichment, we generate “.rnk” files that include each gene

and its concordance index for survival, implemented with the function

concordance.index in the survcomp R package. These “.rnk” files are used in a pre-

ranked GSEA analysis implemented with the executable jar file gsea2-2.07 (which is

downloadable from:http://www.broadinstitute.org/gsea/downloads.jsp). In our

analyses, we set a maximum gene set size of 250 and used default GSEA parameters.

The SAPSscore for each candidate gene set is then computed as the negative log10 of the

maximum of the (Ppure, Prandom, and Penrichment) times the direction of the association

(positive or negative). The statistical significance of the SAPSscore is determined by

permutation-testing. Specifically, in our experiments, we performed 10000

permutations of the gene labels for each of the sequence of 8 of gene set sizes ranging

from 5 to 250. We performed the full SAPS procedure for each of the 80000 permuted

gene sets and we generated a null distribution of 10000SAPSscores for each of the 8

gene set sizes. The SAPSp-value was computed as the proportion of permuted gene sets

of a similar size to the candidate gene set that achieved at least as extreme aSAPSscore.

The SAPSp-values were then converted to SAPSq-values using the method of Benjamini and

Hochberg (Benjamini and Hochberg, 1995).

Page 124: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

113

Supporting Information

Dataset S1 — Supporting information data files, R scripts, and R workspaces. Data deposited

in the Dryad repository: http://dx.doi.org/10.5061/dryad.mk471

The Dataset S1 files are described below (Additional description of the files and ReadMe files

are provided at datadryad.org).

saps.R – This R script provides R commands for loading data, applying the SAPS method, and

generating the SAPS p values. The script is interactive, and the user must specify the working

directory, and if the analysis is on the ovarian or breast data.

runSAPSonPermutedData.R – This R script generates the P_pure, P_random, and

P_enrichment on random gene sets.

computeSAPS.Permute.PValue.R – This script generates permutation-based p and q values for

the SAPSscores obtained in breast and ovarian cancer.

sapsFigures.R – This R script generates the figures, tables, and file used for clustering

Breast.RData – This R-workspace contains the objects: dat, dat.st, event, st, and time.

Breast.RData

dat Data scaled within each dataset without knowledge of subtype. Data from all data-sets merged into this object, which contains expression data on 2731 patients for 13091 genes. Patients are in rows, and entrezID’s in columns.

dat.st Data scaled within molecular subtype within each dataset. Data from all data-sets merged into this object, which contains expression data on 2731 patients for 13091 genes. Patients are in rows, and entrezID’s in columns.

time Time (days)

event Distant metastasis or death

st Molecular subtype defined by SCMGENE

Ovary.RData – This R-workspace contains the objects: dat, dat.st, event, st, and time.

Page 125: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

114

Ovary.RData

dat Data scaled within each dataset without knowledge of subtype. Data from all data-sets merged into this object, which contains expression data on 1670 patients for 11247 genes. Patients are in rows, and entrezID’s in columns.

dat.st Data scaled within molecular subtype within each dataset. Data from all data-sets merged into this object, which contains expression data on 1670 patients for 11247 genes. Patients are in rows, and entrezID’s in columns.

time Time (days)

event Death

st Molecular subtype defined by SCMGENE

BreastOutput_TradScaled.RData– This R-workspace contains the objects: allPs, allPs.adj,

sumTable.

BreastOutput_TradScaled.RData

allPs Contains raw p values for 5320 genesets in molsigdb.v3.0. The columns indicate the type of p value (P_pure, P_random, P_gsea) and the analysis that generated the p value (Global = “Global analysis”,

ER_H = “ER+ High proliferation”, ER_L = “ER+ Low proliferation”, H2 = “HER2+”,TN = “ER-/HER2-“). These p values were generated on the traditional (non-subtype specific) scaled data.

allPs.adj Matrix contains the adjusted p values using the method of Benajmini and Hochberg on the traditional (non-subtype specific) scaled data.

BreastOutput_SubScaled.RData– This R-workspace contains the objects: allPs, allPs.adj,

sumTable.

BreastOutput_SubScaled.RData

allPs Contains raw p values for 5320 genesets in molsigdb.v3.0. The columns indicate the type of p value (P_pure, P_random, P_gsea) and the analysis that generated the p value (Global = “Global analysis”, ER_H = “ER+ High proliferation”, ER_L = “ER+ Low proliferation”, H2 = “HER2+”,TN = “ER-/HER2-“). These p

values were generated on the subtype-

Page 126: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

115

specific scaled data.

allPs.adj Matrix contains the adjusted p values using the method of Benajmini and Hochberg on the subtype-specific scaled data.

OvaryOutput_TradScaled.RData– This R-workspace contains the objects: allPs, allPs.adj,

sumTable.

OvaryOutput_TradScaled.RData

allPs Contains raw p values for 5355 genesets in molsigdb.v3.0. The columns indicate the type of p value (P_pure, P_random, P_gsea) and the analysis that generated the p value (Global = “Global analysis”, Angio= “Angiogenic subtype” , Non-

Angio = “Non-angiogenic subtype”. These p values were generated on the traditional (non-subtype specific) scaled data.

allPs.adj Matrix contains the adjusted p values

using the method of Benajmini and Hochberg on the traditional (non-subtype specific) scaled data.

OvaryOutput_SubScaled.RData– This R-workspace contains the objects: allPs, allPs.adj,

sumTable.

OvaryOutput_SubScaled.RData

allPs Contains raw p values for 5355 genesets in

molsigdb.v3.0. The columns indicate the type of p value (P_pure, P_random, P_gsea) and the analysis that generated the p value (Global = “Global analysis”, Angio= “Angiogenic subtype” , Non-Angio = “Non-angiogenic subtype”. These p values were generated on the subtype-

specific scaled data.

allPs.adj Matrix contains the adjusted p values using the method of Benajmini and Hochberg on the subtype-specific scaled

Page 127: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

116

data.

FinalOutput_Breast.RData contains the results from the subtype-specific analysis in breast

cancer, including the results of the permutation-based procedure to compute p values and q

values for the SAPSscores.

FinalOutput_Breast.RData

allPs Contains raw p values for 5320 genesets in molsigdb.v3.0. The columns indicate the type of p value (P_pure, P_random,

P_gsea) and the analysis that generated the p value (Global = “Global analysis”, ER_H = “ER+ High proliferation”, ER_L = “ER+ Low proliferation”, H2 = “HER2+”,TN = “ER-/HER2-“). These p values were generated on the subtype-specific scaled data.

allPs.adj Matrix contains the adjusted p values using the method of Benajmini and Hochberg on the subtype-specific scaled data.

saps.p Permutation-based p value for each gene set in molsigdb generated on the subtype-specific scaled data

saps.p.adj Adjusted p value (q-value) to indicate the statistical significance of each gene set’s SAPSScore

saps.score This matrix contains the maximum of each gene set’s raw (P_pure, P_random, P_gsea)

saps.score.adj This matrix contains the maximum of gene set’s adjusted (P_pure, P_random, P_gsea)

saps.score.r Array of dimensions 8 x 10000 x 6. The first dimension is the 8 sizes (from 5 to 250) of the random gene sets. The second dimension is the 10000 permutations. The third dimension is the 6 breast cancer analyses performed (Global and the 5 subtypes). Each cell in the array contains

the SAPSScore obtained with a permuted gene set.

Page 128: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

117

FinalOutput_Ovary.RData contains the results from the traditional scaled data set in ovarian

cancer, including the results of the permutation-based procedure to compute p values and q

values for the SAPSScores.

FinalOutput_Breast.RData

allPs Contains raw p values for 5355 genesets in molsigdb.v3.0. The columns indicate the type of p value (P_pure, P_random, P_gsea) and the analysis that generated the p value (Global = “Global analysis”,

Angio= “Angiogenic subtype” , Non-Angio = “Non-angiogenic subtype”. These p values were generated on the traditional (non-subtype specific) scaled data.

allPs.adj Matrix contains the adjusted p values using the method of Benajmini and Hochberg on the traditional (non-subtype specific) scaled data.

saps.p Permutation-based p value for each gene

set in molsigdb generated on the traditional scaled data

saps.p.adj Adjusted p value (q-value) to indicate the statistical significance of each gene set’s

SAPSScore

saps.score This matrix contains the maximum of each gene set’s raw (P_pure, P_random, P_gsea)

saps.score.adj This matrix contains the maximum of gene set’s adjusted (P_pure, P_random, P_gsea)

saps.score.r Array of dimensions 8 x 10000 x 3. The

first dimension is the 8 sizes (from 5 to 250) of the random gene sets. The second dimension is the 10000 permutations. The third dimension is the 3 ovarian cancer analyses performed (Global and the 2 subtypes). Each cell in the array contains the SAPSScore obtained with a permuted gene set.

Breast.Ps.OnPermutedData.RData contains the results of performing SAPS using permuted

gene sets on the breast data

Breast.Ps.OnPermutedData.RData

P_enrich, p_pure,p_rand 8 x 10000 x 6 arrays with P_enrich,P_pure, and P_random p values from permuted gene sets

Page 129: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

118

Ovary.Ps.OnPermutedData.RData contains the results of performing SAPS using permuted

gene sets on the ovarian data

Ovary.Ps.OnPermutedData.RData

P_enrich, p_pure,p_rand 8 x 10000 x 6 arrays with P_enrich,P_pure, and P_random p values from permuted gene sets

BreastSubtypeSpecScaleRankDir contains the ranked gene lists of concordance indices used

to perform the GSEA in breast cancer

OvaryTradScaleRankDir contains the ranked gene lists used of concordance indices to

perform the GSEA in ovarian cancer

BreastOvary_HCv2 – This directory contains files to generate Figure 10 (Hierarchical

clustering of breast and ovarian cancer subtypes based on SAPS scores) using JavaTreeView

(http://jtreeview.sourceforge.net/)

molsigdb.v3.0.entrezForR – This file is used to read the molsigdb.v3.0 gene sets into R.

GSEA Results: The GSEA results for each cancer subtype are presented in the directories:

Breast_Global, Breast_ERHigh, Breast_ERLow,Breast_ERNegHer2Neg,Breast_Her2,

Ovary_Global, Ovary_Angio, Ovary_NonAngio. These analyses were performed to generate

the P_enrichment as part of the SAPS Procedure. Results can be visualized by clicking the

index.html file in each directory.

Page 130: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

119

Table S1: Breast Cancer Datasets

Dataset Microarray

technology

Survival

data

No. of

patients Source Reference

VDX Affymetrix

HGU

RFS,

DMFS 688

GEO:

GSE2034/GSE5327

(Minn et al.,

2007; Wang

et al., 2005)

NKI Agilent RFS,

DMFS,

OS

319 Rosetta Inpharmatics (Van de

Vijver et al.,

2002b; Van t

Veer et al.,

2002)

UCSF in-house

cDNA

DNFS,

RFS,

OS

162 Authors’ website (Korkola et

al., 2007;

Korkola et al.,

2003) STNO2 in-house

cDNA

RFS, OS 118 SMD (Sorlie et al.,

2003)

NCI in-house

cDNA

RFS 99 Authors’ website (Sotiriou et

al., 2003)

MSK Affymetrix

HGU DMFS 82 GEO: GSE2603 (Minn et al.,

2005)

UPP Affymetrix

HGU

RFS 236 GEO: GSE3494 (Miller et al.,

2005)

STK Affymetrix

HGU RFS 159 GEO: GSE1456

(Pawitan et

al., 2005)

UNT Affymetrix

HGU

RFS,

DMFS

133 GEO: GSE2990 (Loi et al.,

2007b;

Sotiriou et al.,

2006c)

UNC4 Agilent RFS, OS 241 UNC DB (Prat et al.,

2010)

CAL Affymetrix

HGU

RFS,

DMFS,

OS

117 AE: E-TABM-158 (Chin et al.,

2006)

TRANSBIG Affymetrix

HGU

RFS,

DMFS,

OS

198 GEO: GSE7390 (Desmedt et

al., 2007)

MAINZ Affymetrix

HGU DMFS 200 GEO: GSE11121

(Schmidt et

al., 2008)

EMC2 Affymetrix

HGU

DMFS 204 GEO: GSE12276 (Bos et al.,

2009) DFHCC Affymetrix

HGU

DMFS 115 GEO: GSE19615 (Li et al.,

2010) TAM Affymetrix

HGU

DMFS,

RFS

242 GEO:

GSE6532/GSE9195

(Sotiriou et

al., 2005) MDA5 Affymetrix

HGU

DMFS 298 GEO: GSE17705 (Symmans et

al., 2010)

VDX3 Affymetrix

HGU

DMFS 136 GEO: GSE12093 (Zhang et al.,

2008b)

PNC Affymetrix

HGU

OS 85

*Microarray datasets of unique breast cancer patients (3832) used in this study

were retrieved from authors’ websites, Gene Expression Omnibus (GEO;

http://www.ncbi.nlm.nih.gov/geo/), ArrayExpress (AE;

http://www.ebi.ac.uk/arrayexpress/), Stanford Microarray Database (SMD;

http://smd.stanford.edu/), MD Anderson Cancer Center Microarray database

Page 131: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

120

(MDACC DB; http://bioinformatics.mdanderson.org/pubdata.html), University

of North Carolina database (UNC DB; https://genome.unc.edu/), and Rosetta

Inpharmatics (http://www.rosettabio.com/). Each dataset was assigned a short

acronym and an instance number if several datasets were published by the same

institution or consortium: EXPO: expression project for oncology, large dataset

of microarray data published by the International Genomics Consortium (United

States); VDX: Veridex (The Netherlands); NKI: National Kanker Instituut (The

Netherlands); UCSF: University of California, San Francisco (United States);

STNO: Stanford/Norway (United States and Norway); NCI: National Cancer

Institute (United States); MSK Memorial Sloan-Kettering (United States); UPP:

Uppsala hospital (Sweden); STK: Stockholm. Karolinska university hospital

(Sweden); UNT: cohort of untreated breast cancer patients from the Oxford

Radcliffe (United Kingdom) and Karolinska (Sweden) hospitals; UNC:

University of North Carolina (United States); DUKE: Duke university hospital

(United States); CAL: dataset of breast cancer patients from the University of

California, San Francisco and the California Pacific Medical Center (United

States); TRANSBIG: dataset collected by the TransBIG consortium (Europe);

MAINZ: Mainz hospital (Germany); LUND: Lund University Hospital

(Sweden); FNCLCC: Fédération Nationale des Centres de Lutte contre le Cancer

(France); MDA: MD Anderson Cancer Centter (United States); EMC: Erasmus

Medical Center (The Netherlands); MUG: Medical University of Graz (Austria);

NCCS: National Cancer Centre of Singapore (Singapore); MCCC: Peter

MacCallum Cancer Centre (Australia); KOO: Koo Foundation Sun Yat-Sen

Cancer Centre (Taiwan); EORTC10994: Trial number 10994 from the European

Organization for Research and Treatment of Cancer Breast Cancer; (Europe)

HLP: University Hospital La Paz (Spain); DFHCC: Dana-Farber Harvard Cancer

Center (United States); MAQC: Microarray quality control consortium (United

States); JBI: Jules Bordet Institute (Belgium). These datasets were generated

with diverse microarray technologies developed either by Agilent

(http://www.genomics.agilent.com), Affymetrix (HGU GeneChips, which

include chips HG-U133A, HG-U133B and HG-U133PLUS2, and X3P

GeneChip; http://www.affymetrix.com), Swegene

(http://www.genomics.agilent.com), Operon (http://www.operon.com) or

developed in-house (complementary DNA, cDNA, platforms). For most datasets

survival data (distant metastasis-free survival [DMFS], relapse-free survival

[RFS], and overall survival [OS]) and information regarding the adjuvant

treatment (untreated, chemo, hormonal, and heterogeneous standing for no

treatment, chemotherapy, hormonal therapy and heterogeneous combination of

therapies, respectively) was available, otherwise missing information is referred

to as not available (NA). Additional clinical characteristics are provided in Table

2. All untreated patients had surgery, and most of them had radiation therapy,

although information is not available for all datasets.

Page 132: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

121

Table S2: Ovarian Cancer Datasets

Dataset Microarray

technology

Survival

data Treatment No. of

patients

Number

of probes Source Reference

DFCI Ilumina

DASL

RFS, OS Platinum,

chemo

129 12,469 AE: E-MTAB-386 (Bentink et

al., 2012) DUKE Affymetrix

HGU

OS Platinum,

chemo

118 22,283 http://data.cgt.duke.edu/platinum.php (Dressman

et al., 2007) FIGO Agilent

G4112A

OS Platinum,

chemo

110 41,000 GEO: GSE17260 (Yoshihara

et al., 2010) AOCS Affymetrix

HGU

RFS, OS Platinum,

chemo

285 54,675 GEO: GSE9899 (Tothill et

al., 2008) MSKCC Affymetrix

HGU

OS Platinum,

chemo

185 22,283 GEO: GSE26712 (Bonome et

al., 2008) TCGA Affymetrix

HGU

RFS, OS Platinum,

chemo

510 22,283 http://tcga-

data.nci.nih.gov/tcga/tcgaHome2.jsp

(Bell et al.,

2011) BIDMC Affymetrix

HG-U95v2

OS Platinum,

chemo

53 12,625 GEO: GSE19161 (Spentzos et

al., 2004) UPENN Affymetrix

HGU

OS Platinum,

chemo

55 54,675 GEO: GSE19161 (Zhang et

al., 2008a) TOC Affymetrix

HGU

OS Platinum,

chemo

80 22,283 GEO: GSE14764 (Denkert et

al., 2009) UMCG Operon

human v3

35K

OS Platinum,

chemo

157 15,909 GEO: GSE13876 (Crijns et

al., 2009) BWH Affymetrix

HGU

OS Platinum,

chemo

53 54,675 GEO: GSE18520 (Mok et al.,

2009)

*Microarray datasets of unique breast cancer patients (5715) used in this study

were retrieved from authors’ websites, Gene Expression Omnibus (GEO;

http://www.ncbi.nlm.nih.gov/geo/), ArrayExpress (AE;

http://www.ebi.ac.uk/arrayexpress/). Each dataset was assigned a short acronym

and an instance number if several datasets were published by the same institution

or consortium: DFCI: Dana-Farber Cancer Institute (United States); DUKE:

Duke university hospital (United States); FIGO: International Federation of

Gynecology and Obstetrics (Japan); AOCS: Australian Ovarian Cancer Study

(Australia); MSKCC: Memorial Sloan-Kettering Cancer Center (United States);

TCGA: The Cancer Genome Atlas (United States); BIDMC: Beth Israel

Deaconess Medical Center (United States); UPENN: University of Pennsylvania

(United States); TOC: tumour bank ovarian cancer (Europe); UMCG: University

Medical Center Groningen (The Netherlands); BWH: Brigham and Women’s

Hospital (United States). These datasets were generated with diverse microarray

technologies developed either by Agilent (http://www.genomics.agilent.com),

Affymetrix (HGU GeneChips, which include chips HG-U133A, HG-U133B and

HG-U133PLUS2 GeneChips; http://www.affymetrix.com), or Operon

(http://www.operon.com). For most datasets survival data (relapse-free survival

[RFS], and overall survival [OS]) and information regarding the adjuvant

treatment (platinum, chemo standing for platinum and chemotherapy

respectively) was available, otherwise missing information is referred to as not

available (NA).

Page 133: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

122

Chapter 5

Conclusions and Future Directions

The field of pathology is rapidly changing. For the entire twentieth century – and

continuing to this day for most diseases – the primary data type used in surgical

pathology has been visual analysis of hematoxylin and eosin stained microscopic

images. Tremendous growth in three major areas of biomedicine is causing a dramatic

shift in the practice of pathology. These three areas of growth are:

1. New technologies to extract near comprehensive genomic, transcriptomic, and

epi-genomic profiles from tissue samples (See, for example, (TCGA, 2012)).

2. The presence of increasingly massive publicly available clinically annotated

Omics data (Butte, 2008).

3. The growth of an increasingly large set of potential therapies to prevent and

treat disease (Hurle et al., 2013).

In the field of oncology, the changing landscape of pathology is particularly

advanced. First, projects such as The Cancer Genome Atlas have demonstrated the

ability to perform integrated Omics profiling on tumor samples in a streamlined,

highly standardized approach (TCGA, 2012). Further, in the past several years there

have been major developments in technologies to profile DNA mutations, DNA copy

number alterations, DNA methylation patterns, coding RNA expression, non-coding

RNA expression, targeted and highly multi-plexed protein expression from routine

formalin fixed paraffin embedded patient tissue specimens (Beck et al., 2010;

Frampton et al., 2013; Gerdes et al., 2013), which will facilitate the clinical

Page 134: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

123

application of Omics technologies for cancer diagnostics. One of the major challenges

going forward will be how to translate this massive quantity of data that can now be

extracted from patient tissue samples into clinically useful biomedical knowledge and

into clinically applicable data-driven diagnostics.

In this dissertation, we have developed and applied new computational methods to

begin to address these challenges. Our studies focused on breast cancer; however, our

methods are general and should be adaptable to many other biomedical domains.

In Chapter 2, we developed the Computational Pathologist (C-Path) system, and

we used it to build an accurate image-based predictor of breast cancer patient survival.

Future work in this area will aim to apply this system to early breast neoplasia, both to

automatically accurately classify early breast neoplastic lesions, and to use the method

to dissect the biology of breast cancer tissue-based risk factors. Further, this method

should be generally useful beyond breast cancer, and we would like to extend the

approach to additional solid cancers (e.g. lung, prostate, and brain).

In Chapter 3, we developed a new method for assessing the biological

informativeness and clinical utility of the two most commonly used protein

biomarkers in breast cancer diagnostics (estrogen receptor and progesterone receptor).

Our analysis showed that progesterone receptor contributes essentially no biological or

clinical information for the stratification of estrogen receptor negative breast cancer,

and we identified thousands of candidate biomarkers far more informative than

progesterone receptor. Future studies will be aimed at the validation of these

biomarkers on additional large cohorts of breast cancer patients. If successful, these

Page 135: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

124

studies could lead to the clinical application of improved biomarkers for breast cancer

stratification, leading to improved individualized treatment regimens.

In Chapter 4, we developed a new method for the identification of robust and

biologically informative prognostic signatures from clinically annotated Omics data.

We applied this method to identify biological signatures in breast and ovarian cancer

and their molecular subtypes. Our analyses uncovered highly diverse prognostic

signatures across disease subtypes, and our studies reveal that the biological factors

driving prognosis in estrogen receptor negative breast cancer subtypes are more

similar to those in ovarian cancer than to those in estrogen receptor positive breast

cancer. Further, we expect the SAPS method to be generally useful for studies aiming

to identify robust biological signatures predictive of clinically relevant phenotypes

from annotated Omics datasets. Future work will aim to extend this method to the

identification of robust biological signatures predictive of drug response to an array of

targeted therapies.

Page 136: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

125

Appendices

Information on Copyright and Author Contribution for

Chapters Adapted from Published Manuscripts

Chapter 2: Morphology: The Computational Pathologist

This chapter is adapted from work published in the following publication: Beck AH,

Sangoi AR, Leung S, Marinelli RJ, Nielsen TO, van de Vijver MJ, West RB, van de

Rijn M, Koller D. Systematic analysis of breast cancer morphology uncovers stromal

features associated with survival. Sci Transl Med. 2011 Nov 9;3(108):108ra113. doi:

10.1126/scitranslmed.3002564.

Copyright Statement: The American Association for the Advancement of Science

(AAAS), the non-profit publisher of Science Translational Medicine specifies in their

“AAAS Author License to Publish Policy” that:

Authors can immediately use final works for non-profit purposes—no

permission needed. The author retains the non-exclusive right to use the final,

published version of the work, immediately after it is made public by AAAS

and without further permission, for educational and other non-commercial

purposes. Such purposes include, for example, print collections of the author’s

own writings; completion of the author’s thesis or dissertation.

Author Contributions: I made a major contribution to all aspects of this project,

including: experimental design, development of the Computational Pathology (C-Path)

image analysis platform, construction and evaluation of the C-Path prognostic models,

and manuscript preparation.

Page 137: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

126

Chapter 3: Biomarkers: Systematic Re-evaluation of Standard of Care Protein

Biomarkers in Breast Cancer. This chapter is adapted from: Hefti MM, Hu R,

Knoblauch NW, Collins LC, Haibe-Kains B, Tamimi RM, Beck AH. Estrogen

receptor negative/progesterone receptor positive breast cancer is not a reproducible

subtype. Breast Cancer Res. 2013 Aug 23;15(4):R68.

Copyright Statement: The journal Breast Cancer Research is part of the Biomed

Central collection of journals, which states in its copyright policy that:

Copyright on any research article in a journal published by BioMed Central is

retained by the author(s),” and: Anyone is free: to copy, distribute, and display

the work; to make derivative works; to make commercial use of the work; Under

the following conditions: Attribution the original author must be given credit; for

any reuse or distribution, it must be made clear to others what the license terms of

this work are; any of these conditions can be waived if the authors gives

permission.

Author Contributions: I made a major contribution to all aspects of this project,

including: experimental design, construction and analysis of the gene expression

microarray meta-data set, identification of prognostic genes in ER+ and ER- breast

cancer, performance of reproducibility and multivariate survival analyses, and

manuscript preparation.

Chapter 4: Genomic Signatures: Significance Analysis of Prognostic Signatures.

This chapter is adapted from work published in the following publication: Beck AH,

Knoblauch NW, Hefti MM, Kaplan J, Schnitt SJ, Culhane AC, Schroeder MS, Risch

T, Quackenbush J, Haibe-Kains B. Significance analysis of prognostic signatures.

PLoS Comput Biol. 2013;9(1):e1002875.

Copyright Statement: PLoS Computational Biology is published by the Public Library

of Science (PLoS), which applies the Creative Commons Attribution License (CCAL)

Page 138: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

127

to all works published. Under the CCAL, authors retain ownership of the copyright for

their article, but authors allow anyone to download, reuse, reprint, modify, distribute,

and/or copy articles in PLOS journals, so long as the original authors and source are

cited. No permission is required from the authors or the publishers

(http://creativecommons.org/licenses/by/2.5/legalcode).

Author Contributions: I made a major contribution to all aspects of this project,

including: design of the Significance Analysis of Prognostic Signatures (SAPS)

procedure, implementation of SAPS in R, application of the method to perform

subtype-specific analyses in breast and ovarian cancer meta-data sets, and manuscript

preparation.

Page 139: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

128

List of References

Ackerknecht, E.H. (1953). Rudolf Virchow: Doctor, Statesman, Anthropologist.

Rudolf Virchow: Doctor, Statesman, Anthropologist.

Alibes, A., Yankilevich, P., Canada, A., and Diaz-Uriarte, R. (2007). IDconverter and

IDClight: conversion and annotation of gene and protein IDs. BMC Bioinformatics 8, 9.

Baak, J.P. (2002). The framework of pathology: good laboratory practice by

quantitative and molecular methods. J Pathol 198, 277-283.

Baak, J.P., Kurver, P.H., de Graaf, S., and Boon, M.E. (1981). Morphometry for prognosis prediction in breast cancer. Lancet 2, 315.

Bardou, V.J., Arpino, G., Elledge, R.M., Osborne, C.K., and Clark, G.M. (2003).

Progesterone receptor status significantly improves outcome prediction over estrogen

receptor status alone for adjuvant endocrine therapy in two large breast cancer databases. J Clin Oncol 21, 1973-1979.

Bartlett, J.M., Brookes, C.L., Robson, T., van de Velde, C.J., Billingham, L.J.,

Campbell, F.M., Grant, M., Hasenburg, A., Hille, E.T., Kay, C., et al. (2011).

Estrogen receptor and progesterone receptor as predictive biomarkers of response to

endocrine therapy: a prospectively powered pathology study in the Tamoxifen and Exemestane Adjuvant Multinational trial. J Clin Oncol 29, 1531-1538.

Beck, A.H., Espinosa, I., Gilks, C.B., van de Rijn, M., and West, R.B. (2008). The

fibromatosis signature defines a robust stromal response in breast carcinoma. Lab Invest 88, 591-601.

Beck, A.H., Knoblauch, N.W., Hefti, M.M., Kaplan, J., Schnitt, S.J., Culhane, A.C.,

Schroeder, M.S., Risch, T., Quackenbush, J., and Haibe-Kains, B. (2013). Significance

analysis of prognostic signatures. PLoS Comput Biol 9, e1002875.

Beck, A.H., Resnick, M.B., Drumea, K.C., and Sabo, E. (2007). Quantitative Image

Analysis for the Classification of Epithelial Neoplasia. In Image Analysis in Medical

Microscopy and Pathology, H.S. Wu, and A. Einstein, eds. (Kerala, Research

Signpost).

Page 140: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

129

Beck, A.H., Sangoi, A.R., Leung, S., Marinelli, R.J., Nielsen, T.O., van de Vijver,

M.J., West, R.B., van de Rijn, M., and Koller, D. (2011). Systematic analysis of breast

cancer morphology uncovers stromal features associated with survival. Sci Transl Med 3, 108ra113.

Beck, A.H., Weng, Z., Witten, D.M., Zhu, S., Foley, J.W., Lacroute, P., Smith, C.L.,

Tibshirani, R., van de Rijn, M., Sidow, A., et al. (2010). 3'-end sequencing for

expression quantification (3SEQ) from archival tumor samples. PLoS One 5, e8768.

Bell, D., Berchuck, A., Birrer, M., Chien, J., Cramer, D.W., Dao, F., Dhir, R., DiSaia,

P., Gabra, H., Glenn, P., et al. (2011). Integrated genomic analyses of ovarian carcinoma. Nature 474, 609-615.

Benjamini, Y., and Hochberg, Y. (1995). Controlling the false discovery rate: a

practical and powerful approach to multiple testing. Journal of the Royal Statistical Society Series B (Methodological), 289-300.

Bentink, S., Haibe-Kains, B., Risch, T., Fan, J.B., Hirsch, M.S., Holton, K., Rubio, R.,

April, C., Chen, J., Wickham-Garcia, E., et al. (2012). Angiogenic mRNA and

microRNA gene expression signature predicts a novel subtype of serous ovarian cancer. PLoS One 7, e30269.

Bergamaschi, A., Tagliabue, E., Sorlie, T., Naume, B., Triulzi, T., Orlandi, R.,

Russnes, H., Nesland, J., Tammi, R., Auvinen, P., et al. (2007). Extracellular matrix

signature identifies breast cancer subgroups with different clinical outcome. J Pathol.

Bianchini, G., Qi, Y., Alvarez, R.H., Iwamoto, T., Coutant, C., Ibrahim, N.K., Valero,

V., Cristofanilli, M., Green, M.C., Radvanyi, L., et al. Molecular anatomy of breast

cancer stroma and its prognostic value in estrogen receptor-positive and -negative

cancers. J Clin Oncol 28, 4316-4323.

Bissell, M.J., and Radisky, D. (2001). Putting tumours in context. Nat Rev Cancer 1, 46-54.

Bloom, H.J.G., and Richardson, W.W. (1957). Histological grading and prognosis in breast cancer. Br J Cancer 11, 359-377.

Bonome, T., Levine, D.A., Shih, J., Randonovich, M., Pise-Masison, C.A.,

Bogomolniy, F., Ozbun, L., Brady, J., Barrett, J.C., Boyd, J., et al. (2008). A gene

Page 141: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

130

signature predicting for survival in suboptimally debulked patients with ovarian cancer. Cancer Res 68, 5478-5486.

Bos, P.D., Zhang, X.H.-F., Nadal, C., Shu, W., Gomis, R.R., Nguyen, D.X., Minn,

A.J., Van de Vijver, M.J., Gerald, W.L., Foekens, J.A., et al. (2009). Genes that mediate breast cancer metastasis to the brain. Nature 459, 1005-1009.

Buffa, F.M., Harris, A.L., West, C.M., and Miller, C.J. (2010). Large meta-analysis of

multiple cancers reveals a common, compact and highly prognostic hypoxia metagene.

Br J Cancer 102, 428-435.

Butte, A.J. (2008). Translational bioinformatics: coming of age. J Am Med Inform Assoc 15, 709-714.

Cancello, G., Maisonneuve, P., Rotmensz, N., Viale, G., Mastropasqua, M.G., Pruneri,

G., Montagna, E., Iorfida, M., Mazza, M., Balduzzi, A., et al. (2013). Progesterone

receptor loss identifies Luminal B breast cancer subgroups at higher risk of relapse. Ann Oncol 24, 661-668.

Chi, J., Wang, Z., Nuyten, D.S.A., Rodriguez, E.H., Schaner, M.E., Salim, A., Wang,

Y., Kristensen, G.B., Helland, A., and Borresen-Dale, A. (2006a). Gene expression

programs in response to hypoxia: cell type specificity and prognostic significance in human cancers. PLoS Medicine 3, 395.

Chi, J.T., Wang, Z., Nuyten, D.S., Rodriguez, E.H., Schaner, M.E., Salim, A., Wang,

Y., Kristensen, G.B., Helland, A., Borresen-Dale, A.L., et al. (2006b). Gene

expression programs in response to hypoxia: cell type specificity and prognostic significance in human cancers. PLoS Med 3, e47.

Chin, K., DeVries, S., Fridlyand, J., Spellman, P., Roydasgupta, R., Kuo, W.L.,

Lapuk, A., Neve, R., Qian, Z., Ryder, T., et al. (2006). Genomic and transcriptional

aberrations linked to breast cancer pathophysiologies. Cancer cell 10, 529-541.

Colditz, G.A., and Hankinson, S.E. (2005). The Nurses' Health Study: lifestyle and health among women. Nat Rev Cancer 5, 388-396.

Colditz, G.A., Rosner, B.A., Chen, W.Y., Holmes, M.D., and Hankinson, S.E. (2004).

Risk factors for breast cancer according to estrogen and progesterone receptor status. J

Natl Cancer Inst 96, 218-228.

Page 142: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

131

Colozza, M., Larsimont, D., and Piccart, M.J. (2005). Progesterone receptor testing: not the right time to be buried. J Clin Oncol 23, 3867-3868; author reply 3869-3870.

Cordon-Cardo, C., Kotsianti, A., Verbel, D.A., Teverovskiy, M., Capodieci, P.,

Hamann, S., Jeffers, Y., Clayton, M., Elkhettabi, F., Khan, F.M., et al. (2007).

Improved prediction of prostate cancer recurrence through systems pathology. J Clin Invest 117, 1876-1883.

Crijns, A.P., Fehrmann, R.S., de Jong, S., Gerbens, F., Meersma, G.J., Klip, H.G.,

Hollema, H., Hofstra, R.M., te Meerman, G.J., de Vries, E.G., et al. (2009). Survival-

related profile, pathways, and transcription factors in ovarian cancer. PLoS medicine 6, e24.

Davies, C., Godwin, J., Gray, R., Clarke, M., Cutter, D., Darby, S., McGale, P., Pan,

H.C., Taylor, C., Wang, Y.C., et al. (2011). Relevance of breast cancer hormone

receptors and other factors to the efficacy of adjuvant tamoxifen: patient-level meta-analysis of randomised trials. Lancet 378, 771-784.

De Maeyer, L., Van Limbergen, E., De Nys, K., Moerman, P., Pochet, N., Hendrickx,

W., Wildiers, H., Paridaens, R., Smeets, A., Christiaens, M.R., et al. (2008). Does

estrogen receptor negative/progesterone receptor positive breast carcinoma exist? J Clin Oncol 26, 335-336; author reply 336-338.

Denkert, C., Budczies, J., Darb-Esfahani, S., Gyorffy, B., Sehouli, J., Konsgen, D.,

Zeillinger, R., Weichert, W., Noske, A., Buckendahl, A.C., et al. (2009). A prognostic

gene expression index in ovarian cancer - validation across different independent data sets. J Pathol 218, 273-280.

Desmedt, C., Haibe-Kains, B., Wirapati, P., Buyse, M., Larsimont, D., Bontempi, G.,

Delorenzi, M., Piccart, M., and Sotiriou, C. (2008). Biological processes associated

with breast cancer clinical outcome depend on the molecular subtypes. Clin Cancer Res 14, 5158-5165.

Desmedt, C., Piette, F., Loi, S.M., Wang, Y., Lallemand, F., Haibe-Kains, B., Viale,

G., Delorenzi, M., Zhang, Y., d&apos;Assignies, M.S., et al. (2007). Strong time

dependence of the 76-gene prognostic signature for node-negative breast cancer

patients in the TRANSBIG multicenter independent validation series. Clinical cancer

research : an official journal of the American Association for Cancer Research 13, 3207-3214.

Page 143: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

132

Ding, L., Ellis, M.J., Li, S., Larson, D.E., Chen, K., Wallis, J.W., Harris, C.C.,

McLellan, M.D., Fulton, R.S., and Fulton, L.L. (2010). Genome remodelling in a

basal-like breast cancer metastasis and xenograft. Nature 464, 999-1005.

Donovan, M.J., Hamann, S., Clayton, M., Khan, F.M., Sapir, M., Bayer-Zubek, V.,

Fernandez, G., Mesa-Tejada, R., Teverovskiy, M., Reuter, V.E., et al. (2008). Systems

pathology approach for the prediction of prostate cancer progression after radical

prostatectomy. J Clin Oncol 26, 3923-3929.

Dressman, H.K., Berchuck, A., Chan, G., Zhai, J., Bild, A., Sayer, R., Cragun, J.,

Clarke, J., Whitaker, R.S., Li, L., et al. (2007). An integrated genomic-based approach

to individualized treatment of patients with advanced-stage ovarian cancer. J Clin

Oncol 25, 517-525.

Dunnwald, L.K., Rossing, M.A., and Li, C.I. (2007). Hormone receptor status, tumor

characteristics, and prognosis: a prospective cohort of breast cancer patients. Breast Cancer Res 9, R6.

Elston, C.W., and Ellis, I.O. (1991). Pathological prognostic factors in breast cancer. I.

The value of histological grade in breast cancer: experience from a large study with long-term follow-up. Histopathology 19, 403-410.

Fanshawe, T.R., Lynch, A.G., Ellis, I.O., Green, A.R., and Hanka, R. (2008).

Assessing Agreement between Multiple Raters with Missing Rating Information,

Applied to Breast Cancer Tumour Grading. PLoS One 3, 2925.

Finak, G., Bertos, N., Pepin, F., Sadekova, S., Souleimanova, M., Zhao, H., Chen, H.,

Omeroglu, G., Meterissian, S., Omeroglu, A., et al. (2008). Stromal gene expression predicts clinical outcome in breast cancer. Nat Med 14, 518-527.

Frampton, G.M., Fichtenholtz, A., Otto, G.A., Wang, K., Downing, S.R., He, J.,

Schnall-Levin, M., White, J., Sanford, E.M., An, P., et al. (2013). Development and

validation of a clinical cancer genomic profiling test based on massively parallel DNA sequencing. Nat Biotechnol.

Fuqua, S.A., Cui, Y., Lee, A.V., Osborne, C.K., and Horwitz, K.B. (2005). Insights

into the role of progesterone receptors in breast cancer. J Clin Oncol 23, 931-932; author reply 932-933.

Page 144: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

133

Gerdes, M.J., Sevinsky, C.J., Sood, A., Adak, S., Bello, M.O., Bordwell, A., Can, A.,

Corwin, A., Dinn, S., Filkins, R.J., et al. (2013). Highly multiplexed single-cell

analysis of formalin-fixed, paraffin-embedded cancer tissue. Proc Natl Acad Sci U S A 110, 11982-11987.

Grann, V.R., Troxel, A.B., Zojwalla, N.J., Jacobson, J.S., Hershman, D., and Neugut,

A.I. (2005). Hormone receptor status and survival in a population-based cohort of

patients with breast carcinoma. Cancer 103, 2241-2251.

Haibe-Kains, B., Desmedt, C., Loi, S., Culhane, A.C., Bontempi, G., Quackenbush, J.,

and Sotiriou, C. (2012). A three-gene model to robustly identify breast cancer molecular subtypes. J Natl Cancer Inst 104, 311-325.

Hammond, M.E., Hayes, D.F., Dowsett, M., Allred, D.C., Hagerty, K.L., Badve, S.,

Fitzgibbons, P.L., Francis, G., Goldstein, N.S., Hayes, M., et al. (2010). American

Society of Clinical Oncology/College of American Pathologists guideline

recommendations for immunohistochemical testing of estrogen and progesterone

receptors in breast cancer. Arch Pathol Lab Med 134, 907-922.

Hanahan, D., and Weinberg, R.A. Hallmarks of cancer: the next generation. Cell 144, 646-674.

Hanahan, D., and Weinberg, R.A. (2000). The hallmarks of cancer. Cell 100, 57-70.

Hefti, M.M., Hu, R., Knoblauch, N.W., Collins, L.C., Haibe-Kains, B., Tamimi, R.M.,

and Beck, A.H. (2013). Estrogen receptor negative/progesterone receptor positive

breast cancer is not a reproducible subtype. Breast Cancer Res 15, R68.

Hitchcock, C.L. (2011). The future of telepathology for the developing world. Arch Pathol Lab Med 135, 211-214.

Horwitz, K.B., Koseki, Y., and McGuire, W.L. (1978). Estrogen control of

progesterone receptor in human breast cancer: role of estradiol and antiestrogen.

Endocrinology 103, 1742-1751.

Horwitz, K.B., and McGuire, W.L. (1975). Predicting response to endocrine therapy in human breast cancer: a hypothesis. Science 189, 726-727.

Page 145: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

134

Horwitz, K.B., and McGuire, W.L. (1978). Estrogen control of progesterone receptor

in human breast cancer. Correlation with nuclear processing of estrogen receptor. J

Biol Chem 253, 2223-2228.

Horwitz, K.B., and McGuire, W.L. (1979). Estrogen control of progesterone receptor

induction in human breast cancer: role of nuclear estrogen receptor. Adv Exp Med Biol 117, 95-110.

Hurle, M.R., Yang, L., Xie, Q., Rajpal, D.K., Sanseau, P., and Agarwal, P. (2013).

Computational drug repositioning: from data to therapeutics. Clin Pharmacol Ther 93, 335-341.

Ivshina, A.V., George, J., Senko, O., Mow, B., Putti, T.C., Smeds, J., Lindahl, T.,

Pawitan, Y., Hall, P., Nordgren, H., et al. (2006). Genetic reclassification of histologic

grade delineates new clinical subtypes of breast cancer. Cancer Res 66, 10292-10301.

Iwamoto, T., Bianchini, G., Booser, D., Qi, Y., Coutant, C., Shiang, C.Y., Santarpia,

L., Matsuoka, J., Hortobagyi, G.N., Symmans, W.F., et al. (2011). Gene pathways

associated with prognosis and chemotherapy sensitivity in molecular subtypes of

breast cancer. J Natl Cancer Inst 103, 264-272.

Karnoub, A.E., Dash, A.B., Vo, A.P., Sullivan, A., Brooks, M.W., Bell, G.W.,

Richardson, A.L., Polyak, K., Tubo, R., and Weinberg, R.A. (2007). Mesenchymal

stem cells within tumour stroma promote breast cancer metastasis. Nature 449, 557-

563.

Korkola, J.E., Blaveri, E., DeVries, S., Moore, D.H., 2nd, Hwang, E.S., Chen, Y.Y.,

Estep, A.L., Chew, K.L., Jensen, R.H., and Waldman, F.M. (2007). Identification of a

robust gene signature that predicts breast cancer outcome in independent data sets.

BMC cancer 7, 61.

Korkola, J.E., DeVries, S., Fridlyand, J., Hwang, E.S., Estep, A.L., Chen, Y.Y., Chew,

K.L., Dairkee, S.H., Jensen, R.M., and Waldman, F.M. (2003). Differentiation of

lobular versus ductal breast carcinomas by expression microarray analysis. Cancer

research 63, 7167-7175.

Landis, J.R., and Koch, G.G. (1977). The measurement of observer agreement for categorical data. Biometrics 33, 159-174.

Page 146: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

135

Le Doussal, V., Tubiana-Hulin, M., Friedman, S., Hacene, K., Spyratos, F., and

Brunet, M. (1989). Prognostic value of histologic grade nuclear components of Scarff-

Bloom-Richardson (SBR). An improved score modification based on a multivariate analysis of 1262 invasive ductal breast carcinomas. Cancer 64, 1914-1921.

Li, Q., Eklund, A.C., Juul, N., Haibe-Kains, B., Workman, C.T., Richardson, A.L.,

Szallasi, Z., and Swanton, C. (2010). Minimising immunohistochemical false negative

ER classification using a complementary 23 gene expression signature of ER status. PloS one 5, e15031.

Liu, R., Wang, X., Chen, G.Y., Dalerba, P., Gurney, A., Hoey, T., Sherlock, G.,

Lewicki, J., Shedden, K., and Clarke, M.F. (2007). The prognostic role of a gene

signature from tumorigenic breast-cancer cells. New England Journal of Medicine 356, 217.

Loi, S., Haibe-Kains, B., Desmedt, C., Lallemand, F., Tutt, A.M., Gillet, C., Ellis, P.,

Harris, A., Bergh, J., Foekens, J.A., et al. (2007a). Definition of clinically distinct

molecular subtypes in estrogen receptor-positive breast carcinomas through genomic grade. J Clin Oncol 25, 1239-1246.

Loi, S.M., Haibe-Kains, B., Desmedt, C., Lallemand, F., Tutt, A.M., Gillet, C., Ellis,

P., Harris, A., Bergh, J., Foekens, J.A., et al. (2007b). Definition of clinically distinct

molecular subtypes in estrogen receptor-positive breast carcinomas through genomic

grade. Journal of clinical oncology : official journal of the American Society of Clinical Oncology 25, 1239-1246.

Long, E.R. (1962). A history of American pathology (Thomas).

Mackey, J.R. (2011). Can quantifying hormone receptor levels guide the choice of

adjuvant endocrine therapy for breast cancer? J Clin Oncol 29, 1504-1506.

Malkin, H.M. (1993). Out of the mist: the foundation of modern pathology and medicine during the nineteenth century (Vesalius Books).

Malkin, H.M. (1998). Comparison of the use of the microscope in pathology in

germany and the united states during the nineteenth century. Annals of diagnostic

pathology 2, 79-88.

Page 147: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

136

Marusyk, A., and Polyak, K. (2010). Tumor heterogeneity: causes and consequences. Biochimica et Biophysica Acta (BBA)-Reviews on Cancer 1805, 105-117.

Miller, L.D., Smeds, J., George, J., Vega, V.B., Vergara, L., Ploner, A., Pawitan, Y.,

Hall, P., Klaar, S., Liu, E.T., et al. (2005). An expression signature for p53 status in

human breast cancer predicts mutation status, transcriptional effects, and patient

survival. Proceedings of the National Academy of Sciences of the United States of

America 102, 13550-13555.

Minn, A.J., Gupta, G.P., Padua, D., Bos, P., Nguyen, D.X., Nuyten, D., Kreike, B.,

Zhang, Y., Wang, Y., Ishwaran, H., et al. (2007). Lung metastasis genes couple breast

tumor size and metastatic spread. Proceedings of the National Academy of Sciences

104, 6740-6745.

Minn, A.J., Gupta, G.P., Siegel, P.M., Bos, P.D., Shu, W., Giri, D.D., Viale, A.,

Olshen, A.B., Gerald, W.L., and Massague, J. (2005). Genes that mediate breast cancer metastasis to lung. Nature 436, 518-524.

Mok, S.C., Bonome, T., Vathipadiekal, V., Bell, A., Johnson, M.E., Wong, K.K.,

Park, D.C., Hao, K., Yip, D.K., Donninger, H., et al. (2009). A gene signature

predictive for outcome in advanced ovarian cancer identifies a survival factor: microfibril-associated glycoprotein 2. Cancer Cell 16, 521-532.

Müller, J., and West, C. (1840). On the Nature and Structural Characteristics of

Cancer: And of Those Morbid Growths which May be Confounded with it (Sherwood, Gilbert, and Piper).

Mulrane, L., Rexhepaj, E., Penney, S., Callanan, J.J., and Gallagher, W.M. (2008).

Automated image analysis in histopathology: a valuable tool in medical diagnostics.

Expert Rev Mol Diagn 8, 707-725.

Nadji, M., Gomez-Fernandez, C., Ganjei-Azar, P., and Morales, A.R. (2005).

Immunohistochemistry of estrogen and progesterone receptors reconsidered: experience with 5,993 breast cancers. Am J Clin Pathol 123, 21-27.

NationalResearchCouncil. (2011). Toward Precision Medicine: Building a Knowledge

Network for Biomedical Research and a New Taxonomy of Disease (National Academies Press).

Page 148: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

137

Nuyten, D.S., Hastie, T., Chi, J.T., Chang, H.Y., and van de Vijver, M.J. (2008).

Combining biological gene expression signatures in predicting outcome in breast

cancer: An alternative to supervised classification. Eur J Cancer 44, 2319-2329.

Olivotto, I.A., Truong, P.T., Speers, C.H., Bernstein, V., Allan, S.J., Kelly, S.J., and

Lesperance, M.L. (2004). Time to stop progesterone receptor testing in breast cancer management. J Clin Oncol 22, 1769-1770.

Paik, S., Tang, G., Shak, S., Kim, C., Baker, J., Kim, W., Cronin, M., Baehner, F.L.,

Watson, D., Bryant, J., et al. (2006). Gene expression and benefit of chemotherapy in

women with node-negative, estrogen receptor-positive breast cancer. J Clin Oncol 24, 3726-3734.

Patey, D.H., and Scarff, R.W. (1928). THE POSITION OF HISTOLOGY IN THE

PROGNOSIS OF CARCINOMA OF THE BREAST. The Lancet 211, 801-804.

Pawitan, Y., Bjohle, J., Amler, L., Borg, A.L., Egyhazi, S., Hall, P., Han, X.,

Holmberg, L., Huang, F., Klaar, S., et al. (2005). Gene expression profiling spares

early breast cancer patients from adjuvant therapy: derived and validated in two

population-based cohorts. Breast cancer research : BCR 7, R953-964.

Prat, A., Cheang, M.C., Martin, M., Parker, J.S., Carrasco, E., Caballero, R.,

Tyldesley, S., Gelmon, K., Bernard, P.S., Nielsen, T.O., et al. (2013). Prognostic

significance of progesterone receptor-positive tumor cells within

immunohistochemically defined luminal A breast cancer. J Clin Oncol 31, 203-209.

Prat, A., Parker, J.S., Karginova, O., Fan, C., Livasy, C., Herschkowitz, J.I., He, X.,

and Perou, C.M. (2010). Phenotypic and molecular characterization of the claudin-low intrinsic subtype of breast cancer. Breast cancer research : BCR 12, R68.

Rakha, E.A., El-Sayed, M.E., Green, A.R., Paish, E.C., Powe, D.G., Gee, J.,

Nicholson, R.I., Lee, A.H., Robertson, J.F., and Ellis, I.O. (2007). Biologic and

clinical characteristics of breast cancer with single hormone receptor positive phenotype. J Clin Oncol 25, 4772-4778.

Reis-Filho, J.S., Weigelt, B., Fumagalli, D., and Sotiriou, C. Molecular Profiling:

Moving Away from Tumor Philately. Science Translational Medicine 2, 47ps43.

Page 149: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

138

Rhodes, A., and Jasani, B. (2009). The oestrogen receptor-negative/progesterone

receptor-positive breast tumour: a biological entity or a technical artefact? J Clin

Pathol 62, 95-96.

Sahoo, D., Dill, D.L., Gentles, A.J., Tibshirani, R., and Plevritis, S.K. (2008). Boolean

implication networks derived from large scale, whole genome microarray datasets. Genome Biol 9, R157.

Schmidt, M., Bohm, D., von Torne, C., Steiner, E., Puhl, A., Pilch, H., Lehr, H.A.,

Hengstler, J.G., Kolbl, H., and Gehrmann, M. (2008). The Humoral Immune System

Has a Key Prognostic Impact in Node-Negative Breast Cancer. Cancer research 68, 5405-5413.

Schroder, M.S., Culhane, A.C., Quackenbush, J., and Haibe-Kains, B. (2011).

survcomp: an R/Bioconductor package for performance assessment and comparison of survival models. Bioinformatics 27, 3206-3208.

Shah, S.P., Morin, R.D., Khattra, J., Prentice, L., Pugh, T., Burleigh, A., Delaney, A.,

Gelmon, K., Guliany, R., and Senz, J. (2009). Mutational evolution in a lobular breast

tumour profiled at single nucleotide resolution. Nature 461, 809-813.

Shi, L., Reid, L.H., Jones, W.D., Shippy, R., Warrington, J.A., Baker, S.C., Collins,

P.J., de Longueville, F., Kawasaki, E.S., Lee, K.Y., et al. (2006). The MicroArray

Quality Control (MAQC) project shows inter- and intraplatform reproducibility of

gene expression measurements. Nat Biotechnol 24, 1151-1161.

Silver, D.P., Richardson, A.L., Eklund, A.C., Wang, Z.C., Szallasi, Z., Li, Q., Juul, N.,

Leong, C.O., Calogrias, D., Buraimoh, A., et al. (2010). Efficacy of neoadjuvant Cisplatin in triple-negative breast cancer. J Clin Oncol 28, 1145-1153.

Sorlie, T., Perou, C.M., Tibshirani, R., Aas, T., Geisler, S., Johnsen, H., Hastie, T.,

Eisen, M.B., van de Rijn, M., Jeffrey, S.S., et al. (2001). Gene expression patterns of

breast carcinomas distinguish tumor subclasses with clinical implications. Proc Natl Acad Sci U S A 98, 10869-10874.

Sorlie, T., Tibshirani, R., Parker, J., Hastie, T., Marron, J.S., Nobel, A., Deng, S.,

Johnsen, H., Pesich, R., Geister, S., et al. (2003). Repeated Observation of Breast

Tumor Subtypes in Independent Gene Expression Data Sets. Proc Natl Acad Sci USA 1, 8418-8423.

Page 150: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

139

Sotiriou, C., Neo, S.Y., McShane, L.M., Korn, E.L., Long, P.M., Jazaeri, A., Martiat,

P., Fox, S.B., Harris, A.L., and Liu, E.T. (2003). Breast Cancer Classification and

Prognosis Based on Gene Expression Profiles from a Population-Based Study. Proc Natl Acad Sci 100, 10393-10398.

Sotiriou, C., and Piccart, M.J. (2007). Taking gene-expression profiling to the clinic:

when will molecular signatures become relevant to patient care? Nat Rev Cancer 7,

545-553.

Sotiriou, C., and Pusztai, L. (2009). Gene-expression signatures in breast cancer. N Engl J Med 360, 790-800.

Sotiriou, C., Wirapati, P., Loi, S., Harris, A., Fox, S., Smeds, J., Nordgren, H., Farmer,

P., Praz, V., and Haibe-Kains, B. (2006a). Gene expression profiling in breast cancer:

understanding the molecular basis of histologic grade to improve prognosis. JNCI Cancer Spectrum 98, 262.

Sotiriou, C., Wirapati, P., Loi, S., Harris, A., Fox, S., Smeds, J., Nordgren, H., Farmer,

P., Praz, V., Haibe-Kains, B., et al. (2006b). Gene expression profiling in breast

cancer: understanding the molecular basis of histologic grade to improve prognosis. J Natl Cancer Inst 98, 262-272.

Sotiriou, C., Wirapati, P., Loi, S.M., Desmedt, C., Durbecq, V., Harris, A., Bergh, J.,

Smeds, J., Haibe-Kains, B., Larsimont, D., et al. (2005). Breast tumours with

intermediate histological grade can be reclassified into prognostically distinct groups by gene expression profiling. In Breast cancer research and treatment, pp. S30.

Sotiriou, C., Wirapati, P., Loi, S.M., Harris, A., Fox, S., Smeds, J., Nordgren, H.,

Farmer, P., Praz, V., Haibe-Kains, B., et al. (2006c). Gene expression profiling in

breast cancer: understanding the molecular basis of histologic grade to improve prognosis. Journal of the National Cancer Institute 98, 262-272.

Spentzos, D., Levine, D.A., Ramoni, M.F., Joseph, M., Gu, X., Boyd, J., Libermann,

T.A., and Cannistra, S.A. (2004). Gene expression signature with independent

prognostic significance in epithelial ovarian cancer. J Clin Oncol 22, 4700-4710.

Staaf, J., Ringner, M., Vallon-Christersson, J., Jonsson, G., Bendahl, P.O., Holm, K.,

Arason, A., Gunnarsson, H., Hegardt, C., Agnarsson, B.A., et al. (2010). Identification

of subtypes in human epidermal growth factor receptor 2--positive breast cancer

reveals a gene signature prognostic of outcome. J Clin Oncol 28, 1813-1820.

Page 151: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

140

Subramanian, A., Kuehn, H., Gould, J., Tamayo, P., and Mesirov, J.P. (2007). GSEA-

P: a desktop application for Gene Set Enrichment Analysis. Bioinformatics 23, 3251-

3253.

Subramanian, A., Tamayo, P., Mootha, V.K., Mukherjee, S., Ebert, B.L., Gillette,

M.A., Paulovich, A., Pomeroy, S.L., Golub, T.R., Lander, E.S., et al. (2005). Gene set

enrichment analysis: a knowledge-based approach for interpreting genome-wide

expression profiles. Proc Natl Acad Sci U S A 102, 15545-15550.

Symmans, W.F., Hatzis, C., Sotiriou, C., Andre, F., Peintinger, F., Regitnig, P.,

Daxenbichler, G., Desmedt, C., Domont, J., Marth, C., et al. (2010). Genomic index of

sensitivity to endocrine therapy for breast cancer. Journal of clinical oncology :

official journal of the American Society of Clinical Oncology 28, 4111-4119.

Tamimi, R.M., Baer, H.J., Marotti, J., Galan, M., Galaburda, L., Fu, Y., Deitz, A.C.,

Connolly, J.L., Schnitt, S.J., Colditz, G.A., et al. (2008). Comparison of molecular

phenotypes of ductal carcinoma in situ and invasive breast cancer. Breast Cancer Res

10, R67.

Tan, W., Zhang, W., Strasner, A., Grivennikov, S., Cheng, J.Q., Hoffman, R.M., and

Karin, M. (2011). Tumour-infiltrating regulatory T cells stimulate mammary cancer metastasis through RANKL-RANK signalling. Nature 470, 548-553.

TCGA (2012). Comprehensive molecular portraits of human breast tumours. Nature

490, 61-70.

Teschendorff, A.E., and Caldas, C. (2008). A robust classifier of high predictive value

to identify good prognosis patients in ER-negative breast cancer. Breast Cancer Res 10, R73.

Teschendorff, A.E., Miremadi, A., Pinder, S.E., Ellis, I.O., and Caldas, C. (2007). An

immune response gene expression module identifies a good prognosis subtype in estrogen receptor negative breast cancer. Genome Biol 8, R157.

Tibshirani, R.J., and Efron, B. (2002). Pre-validation and inference in microarrays. Stat Appl Genet Mol Biol 1, Article1.

Tothill, R.W., Tinker, A.V., George, J., Brown, R., Fox, S.B., Lade, S., Johnson, D.S.,

Trivett, M.K., Etemadmoghadam, D., Locandro, B., et al. (2008). Novel molecular

Page 152: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

141

subtypes of serous and endometrioid ovarian cancer linked to clinical outcome. Clin Cancer Res 14, 5198-5208.

Troyanskaya, O., Cantor, M., Sherlock, G., Brown, P., Hastie, T., Tibshirani, R.,

Botstein, D., and Altman, R.B. (2001). Missing value estimation methods for DNA microarrays. Bioinformatics 17, 520-525.

van de Vijver, M.J., He, Y.D., van't Veer, L.J., Dai, H., Hart, A.A., Voskuil, D.W.,

Schreiber, G.J., Peterse, J.L., Roberts, C., Marton, M.J., et al. (2002a). A gene-

expression signature as a predictor of survival in breast cancer. N Engl J Med 347, 1999-2009.

Van de Vijver, M.J., He, Y.D., van't Veer, L.J., Dai, H., Hart, A.A.M., Voskuil, D.W.,

Schreiber, G.J., Peterse, J.L., Roberts, C., Marton, M.J., et al. (2002b). A gene-

expression signature as a predictor of survival in breast cancer. The New England journal of medicine 347, 1999-2009.

Van den Eynden, G.G., Colpaert, C.G., Couvelard, A., Pezzella, F., Dirix, L.Y.,

Vermeulen, P.B., Van Marck, E.A., and Hasebe, T. (2007). A fibrotic focus is a

prognostic factor and a surrogate marker for hypoxia and (lymph)angiogenesis in

breast cancer: review of the literature and proposal on the criteria of evaluation. Histopathology 51, 440-451.

Van t Veer, L.J., Dai, H., Van de Vijver, M.J., He, Y.D., Hart, A.A.M., Mao, M.,

Peterse, H.L., van der Kooy, K., Marton, M.J., Witteveen, A.T., et al. (2002). Gene expression profiling predicts clinical outcome of breast cancer. Nature 415, 530-536.

Venet, D., Dumont, J.E., and Detours, V. (2011). Most random gene expression

signatures are significantly associated with breast cancer outcome. PLoS Comput Biol

7, e1002240.

Viale, G., Regan, M.M., Maiorano, E., Mastropasqua, M.G., Golouh, R., Perin, T.,

Brown, R.W., Kovacs, A., Pillay, K., Ohlschlegel, C., et al. (2008). Chemoendocrine

compared with endocrine adjuvant therapies for node-negative breast cancer:

predictive value of centrally reviewed expression of estrogen and progesterone receptors--International Breast Cancer Study Group. J Clin Oncol 26, 1404-1410.

von Staden, H. (1992). The discovery of the body: human dissection and its cultural contexts in ancient Greece. Yale J Biol Med 65, 223-241.

Page 153: A DISSERTATION SUBMITTED TO THE …nc361qm2225/...I certify that I have read this dissertation and that, in my opinion, it is fully adequate in scope and quality as a dissertation

142

Wang, Y., Klijn, J.G.M., Zhang, Y., Sieuwerts, A.M., Look, M.P., Yang, F., Talantov,

D., Timmermans, M., Meijer-van Gelder, M.E., Yu, J., et al. (2005). Gene-expression

profiles to predict distant metastasis of lymph-node-negative primary breast cancer. Lancet 365, 671-679.

Weigelt, B., Baehner, F.L., and Reis-Filho, J.S. (2010). The contribution of gene

expression profiling to breast cancer classification, prognostication and prediction: a

retrospective of the last decade. J Pathol 220, 263-280.

West, R.B., Nuyten, D.S., Subramanian, S., Nielsen, T.O., Corless, C.L., Rubin, B.P.,

Montgomery, K., Zhu, S., Patel, R., Hernandez-Boussard, T., et al. (2005). Determination of stromal signatures in breast carcinoma. PLoS Biol 3, e187.

Wirapati, P., Sotiriou, C., Kunkel, S., Farmer, P., Pradervand, S., Haibe-Kains, B.,

Desmedt, C., Ignatiadis, M., Sengstag, T., Schutz, F., et al. (2008). Meta-analysis of

gene expression profiles in breast cancer: toward a unified understanding of breast cancer subtyping and prognosis signatures. Breast Cancer Res 10, R65.

Wiseman, B., and Werb, Z. (2002). Stromal effects on mammary gland development

and breast cancer. Science 296, 1046.

Yoshihara, K., Tajima, A., Yahata, T., Kodama, S., Fujiwara, H., Suzuki, M., Onishi,

Y., Hatae, M., Sueyoshi, K., Kudo, Y., et al. (2010). Gene expression profile for

predicting survival in advanced-stage serous ovarian cancer across two independent

datasets. PLoS ONE 5, e9615.

Young, R.H. (1999). Guiding the surgeon's hand. The history of american surgical pathology (LWW).

Zhang, L., Volinia, S., Bonome, T., Calin, G.A., Greshock, J., Yang, N., Liu, C.G.,

Giannakakis, A., Alexiou, P., Hasegawa, K., et al. (2008a). Genomic and epigenetic

alterations deregulate microRNA expression in human epithelial ovarian cancer. Proc Natl Acad Sci U S A 105, 7004-7009.

Zhang, Y., Sieuwerts, A., McGreevy, M., Casey, G., Cufer, T., Paradiso, A., Harbeck,

N., Span, P., Hicks, D., Crowe, J., et al. (2008b). The 76-gene signature defines high-

risk patients that benefit from adjuvant tamoxifen therapy. Breast cancer research and treatment.