ArticlePDF Available

Expression of RUNX1 Correlates with Poor Patient Prognosis in Triple Negative Breast Cancer

PLOS
PLOS ONE
Authors:

Abstract and Figures

The RUNX1 transcription factor is widely recognised for its tumour suppressor effects in leukaemia. Recently a putative link to breast cancer has started to emerge, however the function of RUNX1 in breast cancer is still unknown. To investigate if RUNX1 expression was important to clinical outcome in primary breast tumours a tissue microarray (TMA) containing biopsies from 483 patients with primary operable invasive ductal breast cancer was stained by immunohistochemistry. RUNX1 was associated with progesterone receptor (PR)-positive tumours (P<0.05), more tumour CD4+(P<0.05) and CD8+(P<0.01) T-lymphocytic infiltrate, increased tumour CD138+plasma cell (P<0.01) and more CD68+macrophage infiltrate (P<0.001). RUNX1 expression did not influence outcome of oestrogen receptor (ER)-positive or HER2-positive disease, however on univariate analysis a high RUNX1 protein was significantly associated with poorer cancer-specific survival in patients with ER-negative (P<0.05) and with triple negative (TN) invasive breast cancer (P<0.05). Furthermore, multivariate Cox regression analysis of cancer-specific survival showed a trend towards significance in ER-negative patients (P<0.1) and was significant in triple negative patients (P<0.05). Of relevance, triple negative breast cancer currently lacks good biomarkers and patients with this subtype do not benefit from the option of targeted therapy unlike patients with ER-positive or HER2-positive disease. Using multivariate analysis RUNX1 was identified as an independent prognostic marker in the triple negative subgroup. Overall, our study identifies RUNX1 as a new prognostic indicator correlating with poor prognosis specifically in the triple negative subtype of human breast cancer.
Content may be subject to copyright.
Expression of RUNX1 Correlates with Poor Patient
Prognosis in Triple Negative Breast Cancer
Nicola Ferrari
1
, Zahra M. A. Mohammed
, Colin Nixon
1
, Susan M. Mason
1
, Elizabeth Mallon
3
,
Donald C. McMillan
2
, Joanna S. Morris
4
, Ewan R. Cameron
4
, Joanne Edwards
5
, Karen Blyth
1
*
1Transgenic Models Lab, Cancer Research UK Beatson Institute, Glasgow, Scotland, United Kingdom, 2Academic Unit of Surgery, College of Medical, Veterinary and Life
Sciences, University of Glasgow, Glasgow, Scotland, United Kingdom, 3University Pathology Unit, Southern General Hospital, Glasgow, Scotland, United Kingdom,
4School of Veterinary Medicine, University of Glasgow, Glasgow, Scotland, United Kingdom, 5Institute of Cancer Sciences, University of Glasgow, Glasgow, Scotland,
United Kingdom
Abstract
The RUNX1 transcription factor is widely recognised for its tumour suppressor effects in leukaemia. Recently a putative link
to breast cancer has started to emerge, however the function of RUNX1 in breast cancer is still unknown. To investigate if
RUNX1 expression was important to clinical outcome in primary breast tumours a tissue microarray (TMA) containing
biopsies from 483 patients with primary operable invasive ductal breast cancer was stained by immunohistochemistry.
RUNX1 was associated with progesterone receptor (PR)-positive tumours (P,0.05), more tumour CD4+(P,0.05) and CD8+
(P,0.01) T-lymphocytic infiltrate, increased tumour CD138+plasma cell (P,0.01) and more CD68+macrophage infiltrate (P,
0.001). RUNX1 expression did not influence outcome of oestrogen receptor (ER)-positive or HER2-positive disease, however
on univariate analysis a high RUNX1 protein was significantly associated with poorer cancer-specific survival in patients with
ER-negative (P,0.05) and with triple negative (TN) invasive breast cancer (P,0.05). Furthermore, multivariate Cox
regression analysis of cancer-specific survival showed a trend towards significance in ER-negative patients (P,0.1) and was
significant in triple negative patients (P,0.05). Of relevance, triple negative breast cancer currently lacks good biomarkers
and patients with this subtype do not benefit from the option of targeted therapy unlike patients with ER-positive or HER2-
positive disease. Using multivariate analysis RUNX1 was identified as an independent prognostic marker in the triple
negative subgroup. Overall, our study identifies RUNX1 as a new prognostic indicator correlating with poor prognosis
specifically in the triple negative subtype of human breast cancer.
Citation: Ferrari N, Mohammed ZMA, Nixon C, Mason SM, Mallon E, et al. (2014) Expression of RUNX1 Correlates with Poor Patient Prognosis in Triple Negative
Breast Cancer. PLOS ONE 9(6): e100759. doi:10.1371/journal.pone.0100759
Editor: Arto Mannermaa, University of Eastern Finland, Finland
Received February 24, 2014; Accepted May 28, 2014; Published June 26, 2014
Copyright: ß2014 Ferrari et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits
unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Funding: This work was funded by Cancer Research UK. The funders had no role in study design, data collection and analysis, decision to publish, or preparation
of the manuscript.
Competing Interests: The authors have declared that no competing interests exist.
* Email: K.Blyth@beatson.gla.ac.uk
¤ Current address: University Department of Pathology, Omar Almukhtar University, Al bayda, Libya
Introduction
Breast cancer is the third most common cause of cancer death in
the UK, accountable for more than 11,000 deaths in 2011 alone
(www.cancerresearchuk.org) and an estimated 39,620 female
deaths in the USA in 2013 (www.cancer.gov). In human breast
cancer, oestrogen receptor (ER), progesterone receptor (PR), and
human epidermal growth factor receptor 2 (HER2) are well-
established prognostic and predictive markers, and testing for
them is now considered standard of care [1]. Based on the receptor
status, human breast cancer can be subdivided into three main
groups: oestrogen receptor positive (ER+), human epidermal
growth factor receptor 2 positive (HER2+) and triple negative
(ER2/PR2/HER2–). ER+and HER2+patients benefit from
targeted treatments such as Tamoxifen and/or Trastuzumab
which have consistently improved disease outcome [2]. On the
other hand, the triple negative (TN) subtype lacks any specific
targeted therapy and is associated with worse overall prognosis in
comparison with the other subtypes [3]. This underlines the urgent
need for new prognostic and therapeutic targets specific for this
group of patients.
The RUNX genes are a family of three transcription factors
(RUNX1, 2 and 3) known to play essential roles in haematopoiesis,
osteogenesis and neurogenesis [4]. Besides being key developmen-
tal regulators, RUNX genes are also important in cancer, acting
both as oncogenes or tumour suppressors in different systems [5].
RUNX1 is the most frequently mutated gene in human leukaemia
and many studies have focused on its tumour suppressive function
in haematopoietic malignancies [6]. However, in recent years, a
new role for RUNX1 outside the haematopoietic system has
started to emerge with several studies indicating how this
transcription factor could be more broadly implicated in cancer
[7,8]. In particular RUNX1 has been identified as a key regulator
of tumourigenesis in various epithelial cancers [9–11]. However
little is known about the role of RUNX1 in human breast cancer
[12]. Wang and colleagues using 3D culture models showed that
RUNX1 deletion in MCF10A acini resulted in increased cell
proliferation and abnormal morphogenesis [13]. In addition, three
independent large scale sequencing studies on human breast
PLOS ONE | www.plosone.org 1 June 2014 | Volume 9 | Issue 6 | e100759
cancers discovered recurrent RUNX1 mutations and deletions in
human tumours [14–16] while Kadota et al showed by qRT-PCR
on a small breast cancer cohort (29 samples) that RUNX1
downregulation is associated with high-grade primary breast
tumours [17]. Here we have carried out the first comprehensive
characterization of RUNX1 expression in tissues from a large
cohort of human breast cancers and demonstrate its prognostic
value in different tumour subtypes.
Materials and Methods
Patients
The expression studies in human tissues were ethically approved
from West of Scotland Research Ethics Service West of Scotland
REC4 (REC Ref: Project Number 02/SG007(10), R and D
project: RN07PA001). Consent was not obtained, but all patient
information is anonymised with all patient identifiers removed.
Patients diagnosed with invasive breast cancer at three Glasgow
hospitals (Royal Infirmary, Western Infirmary and Stobhill
Figure 1. Expression of RUNX1 in human breast cancer cell lines. RUNX1 expression by western blot on a panel of human breast cancer cell
lines with basal–like (HCC-70, BT-549, BT-20, MDA-MB-231, MDA-MB-436, MDA-MB-468) and luminal-like (BT-474, MCF-7, T47D, MDA-MB-361)
features. HDAC2 used as a loading control. hMEC-TERT; immortalized human mammary epithelial cells.
doi:10.1371/journal.pone.0100759.g001
Table 1. Clinico-pathological characteristics of patients with primary operable invasive ductal breast cancer.
Clinico-pathological characteristics (total) Patients (n%)
Age (#50/.50 years) (n= 483) 141 (29%)/342 (71%)
Size (#20/21–50/.50 mm) (n= 481) 280 (58%)/186 (39%)/15 (3%)
Tumour type (Special type/lobular/ductal) (n = 483) 23 (5%)/33 (7%)/427 (88%)
Grade (I/II/III) (n = 481) 88 (18%)/202 (42%)/191 (40%)
Involved lymph node (Negative/positive) (n = 478) 268 (56%)/210 (44%)
Oestrogen -receptor status (ER2/ER+) (n = 481) 184 (38%)/297 (62%)
Progesterone -receptor status (PR2/PR+) (n = 480) 266 (55%)/214 (45%)
HER2 status (HER22/HER2+) (n = 466) 393 (84%)/73 (16%)
Lymphovascular invasion (Absent/present) (n = 372) 198 (53%)/174 (47%)
Microvessel density (CD34+) (Low/medium/high) (n = 450) 157 (35%)/150 (33%)/143 (32%)
Ki-67 status (Low Ki-67/high Ki-67) (n= 468) 353 (75%)/115 (25%)
Tumour necrosis (Absent/present) (n = 473) 213 (45%)/260 (55%)
TUNEL (Low/high) (n = 417) 235 (56%)/182 (44%)
General inflammatory infiltrate (Low high) (n = 473) 334 (71%)/139 (29%)
Tumour CD4+T- lymphocytic infiltrate (Low/medium/high) (n = 474) 217 (46%)/93 (20%)/164 (34%)
Tumour CD8+T- lymphocytic infiltrate (Low/medium/high) (n = 474) 162 (34%)/154 (32.5%)/158 (33%)
Tumour CD138+B- lymphocytic infiltrate (Low/medium/high) (n = 473) 265 (56%)/60 (13%)/148 (31%)
Tumour CD68+macrophages infiltrate (Low/medium/high) (n = 471) 141 (30%)/164 (35%)/166 (35%)
Loco-regional treatment (Lumpectomy+radiotherapy/mastectomy+radiotherapy) (n = 483) 170 (35%)/313 (65%)
Systemic treatment (ER-based treatment) (hormonal/hormonal+chemotherapy/chemotherapy/none) (n =476) 252 (53%)/98 (20%)/103 (22%)/23 (5%)
RUNX1 (Negative/positive) (n = 483) 117 (24%)/366 (76%)
(n = 483).
doi:10.1371/journal.pone.0100759.t001
RUNX1 in Triple Negative Breast Cancer
PLOS ONE | www.plosone.org 2 June 2014 | Volume 9 | Issue 6 | e100759
Hospital) between 1995 and 1998 were studied (n = 483). Clinical
and pathological data including age, histological tumour type,
grade, tumour size, lymph node status, lymphovascular invasion,
type of surgery and use of adjuvant treatment (chemotherapy,
hormonal therapy and radiotherapy) were retrieved from the
patient records and histopathology reports.
Tissue microarray (TMA) construction and
immunohistochemistry
Tissue microarrays (TMA) were already available for use in this
study. 0.6 mm
2
cores of breast cancer tissue, identified by the
pathologist (EM), were removed from representative areas of the
tumour taken from breast cancer patients at the time of surgical
resection. All tissue microarray blocks were constructed in
triplicate and were utilized to assess ER, PR, HER2 status, Ki-
67 and microvessel density by immunohistochemical analyses as
previously described [18–21]. Immunohistochemistry was used to
quantify cellular infiltrate of macrophages [19], CD4+, CD8+
lymphocytes and CD138+plasma cells as previously reported [22].
Immunohistochemistry for RUNX1. RUNX1 antibody
(Sigma, HPA004176) was validated to confirm its specificity by
western blot (Figure S1). Expression was detected in a positive
control (T6i) but not in a leukaemia cell line deleted for RUNX1
(3SS cells). Human mammary epithelial cells (hMEC) transfected
with a RUNX1 overexpression vector (hMEC-RUNX1) or empty
vector (hMEC-Puro) were used as an independent validation.
TMAs were stained for RUNX1 by immunohistochemistry. Heat
induced epitope retrieval for RUNX1 was performed at 98uC for
25 minutes in citrate buffer (pH 6). Endogenous peroxidase was
blocked by incubation in 3% hydrogen peroxide (DAKO, UK) for
5 minutes. The cores were then incubated with primary antibody
for RUNX1 added at dilution of 1:100 for 40 minutes at 25uC.
Sites of binding were detected using the appropriate Envision
secondary antibody (DAKO code K4003) and visualized using
DAB (3-39diaminobenzidine, DAKO, UK) according to the
manufacturer’s instruction. Cores were counterstained with
haematoxylin, dehydrated and coverslipped with DPX.
Weighted histoscore method. RUNX1 staining was quan-
tified using the weighted histoscore method to give a value of 0–
300 [23]. One hundred and fifty cores (10% of total core number)
were scored independently for epithelial RUNX1 expression by
two observers (NF and ZM) blind to patient’s outcome and the
other observer’s score. Interclass Correlation Coefficient (ICC),
measure of inter-observer agreement, was 0.82. NF then scored all
cores and this data was used in subsequent analysis.
Statistical analysis
Inter-relationships between variables were assessed using
contingency tables with the chi-squared test for trend as
appropriate. Univariate analysis and multivariate survival analysis
with calculation of hazard ratios (HR) were performed using Cox’s
proportional-hazards model. A stepwise backward procedure was
used to derive a final model of the variables that had a significant
independent relationship with survival. Mortality incidences up to
March 2010 were included in the analysis. Analysis was performed
using SPSS software version 19 (SPSS Inc., Chicago, IL, USA).
Table 2. The relationship between RUNX1 and clinico-pathological characteristics of patients with primary operable invasive
ductal breast cancer.
Clinico-pathological characteristics (total)
RUNX1 Negative
(n = 117)
RUNX1 Positive
(n = 366) p-value
Age (#50/.50 years) (n = 483) 25/92 116/250 0.033
Size (#20/21–50/.50 mm) (n = 481) 70/43/4 210/143/11 0.769
Tumour type (Special type/lobular/ductal) (n = 483) 8/9/100 15/24/327 0.197
Grade (I/II/III) (n = 481) 17/57/43 71/145/148 0.891
Involved lymph node (Negative/positive) (n = 478) 70/46 198/164 0.287
Oestrogen -receptor status (ER2/ER+) (n = 481) 53/64 131/233 0.072
Progesterone -receptor status (PR2/PR+) (n = 480) 75/42 191/172 0.03
HER2 status (HER22/HER2+) (n = 466) 97/16 296/57 0.613
Lymphovascular invasion (Absent/present) (n = 372) 48/36 150/138 0.414
Microvessel density (CD34+) (Low/medium/high) (n = 450) 44/31/30 113/119/113 0.143
Ki-67 status (Low Ki-67/high Ki-67) (n = 468) 87/26 266/89 0.658
Tumour necrosis (Absent/present) (n = 473) 52/63 161/197 0.963
TUNEL (Low/high) (n = 417) 51/42 184/140 0.738
General inflammatory infiltrate (Low high) (n = 473) 81/34 253/105 0.962
Tumour CD4+T- lymphocytic infiltrate (Low/medium/high) (n = 474) 63/22/30 154/71/134 0.015
Tumour CD8+T- lymphocytic infiltrate (Low/medium/high) (n = 474) 55/28/32 107/126/126 0.004
Tumour CD138+B- lymphocytic infiltrate (Low/medium/high) (n = 473) 80/11/24 185/49/124 0.001
Tumour CD68+macrophages infiltrate (Low/medium/high) (n = 471) 50/39/24 91/125/142
,
0.001
Loco-regional treatment (Lumpectomy+radiotherapy/mastectomy+radiotherapy) (n = 483) 39/78 131/235 0.628
Systemic treatment (ER-based treatment) (hormonal/hormonal+chemotherapy/chemotherapy/none)
(n = 476)
65/23/21/6 187/75/82/17 0.42
Cancer specific survival (months)* 156 (146–165) 149 (143–155) 0.158
*Mean (95%CI).
doi:10.1371/journal.pone.0100759.t002
RUNX1 in Triple Negative Breast Cancer
PLOS ONE | www.plosone.org 3 June 2014 | Volume 9 | Issue 6 | e100759
Cell lines
T6i leukaemia cell line overexpressing RUNX1 [24] and 3SS (a
cell line generated from a murine lymphoma which is genetically
deleted for RUNX1 and kindly provided by Gillian Borland in
ERC’s lab) were used respectively as positive and negative controls
in RUNX1 western blots. The genetically altered mouse used to
generate 3SS was covered by University of Glasgow ethical review
process and project licence PPL60/4408. hMEC-TERT cell line
[25] (a kind gift of Barbara Chaneton) was grown in HuMEC
complete media (Gibco). MDA-MB-231, MDA-MB-436, MDA-
MB-468, HCC-70, BT-20, BT-549, T47D, MDA-MB-361, MCF-
7 and BT-474 were originally sourced from the American Type
Culture Collection (ATCC). All cell lines were grown in a Galaxy+
incubator (RS Biotech) at 37uC with 5% CO
2
.
To generate RUNX1 overexpressing cells, hMEC-TERT were
transfected with pBABE-Puro-RUNX1 or pBABE-Puro (kindly
provided by Dr Anna Kilbey) through electroporation using
Nucleofector Kit V, program T-013 (Amaxa, Lonza). After
electroporation, cells were allowed to recover for 24 h and then
selected in puromycin selection media (10 mg/ml) for 2 weeks.
Western blot
Nuclear extracts were prepared from mammary cell lines using
NE-PER Nuclear and Cytoplasmic Extraction Reagents (Thermo
Scientific, Cat No 78833) as per kit instructions. Protein extracts
were resolved on 10% NuPAGE Novex Bis-Tris gels (Life
Technologies) and transferred to Hybond-ECL nitrocellulose
membranes (Amersham). Membranes were probed with antibod-
ies to RUNX1 (HPA004176, Sigma), HDAC2 (sc-6296, Santa
Cruz) and GAPDH (Cell Signalling).
Results
Characterisation of RUNX1 expression in human breast
cancer
RUNX1 expression was first tested on a panel of breast cancer
cell lines. The chosen cell lines included normal human mammary
epithelial cells derived from primary tissue and immortalised with
TERT expression (hMEC-TERT), 6 basal-like (HCC-70, BT-549,
BT-20, MDA-MB-231, MDA-MB-436, MDA-MB-468) and 4
luminal-like (BT-474, MCF-7, T47D, MDA-MB-361) breast
cancer cell lines. Significantly, RUNX1 expression was not
detectable in normal hMEC-TERT but was expressed in all
breast cancer cell lines tested with the exception of BT-549
(Figure 1). These results suggest that RUNX1 expression could be
dysregulated in human breast cancer.
To investigate if RUNX1 expression influenced clinical
outcome in primary breast tumours, a tissue microarray (TMA)
containing biopsies from 483 patients with operable invasive
ductal breast cancer [18] was stained for RUNX1. Baseline
clinico-pathological characteristics of the patients included in the
TMA are shown in Table 1. The invasive cancers showed different
degrees of RUNX1 expression, predominantly localized to the
nucleus (Figure 2A). RUNX1 expression in the tumour epithelium
was determined by histoscore which takes into account the
percentage of positive signal and staining intensity. Patients were
divided into two groups: RUNX1 negative (histoscore = 0,
n = 117) and RUNX1 positive (histoscore .0, n = 366). The
relationships between RUNX1 expression and clinico-pathological
characteristics in patients with primary operable ductal invasive
breast cancer are shown in Table 2. In the whole cohort a number
of factors were identified to be associated with positive RUNX1
Table 3. The relationship between RUNX1 and clinico-pathological characteristics of patients with triple negative primary
operable invasive ductal breast cancer.
Clinico-pathological characteristics (total) RUNX1 Negative RUNX1 Positive
p-
value
Age (#50/.50 years) (n = 483) 10/22 33/53 0.477
Size (#20/21–50/.50 mm) (n = 481) 17/15/0 43/38/4 0.537
Tumour type (Special type/lobular/ductal) (n = 483) 5/0/27 6/2/78 0.223
Grade (I/II/III) (n = 481) 0/9/23 4/14/66 0.857
Involved lymph node (Negative/positive) (n = 478) 20/12 48/38 0.515
Lymphovascular invasion (Absent/present) (n = 372) 16/9 37/35 0.278
Microvessel density (CD34+) (Low/medium/high) (n = 450) 8/11/11 30/17/37 0.928
Ki-67 status (Low Ki-67/high Ki-67) (n = 468) 22/9 69/16 0.239
Tumour necrosis (Absent/present) (n = 473) 7/25 16/70 0.691
TUNEL (Low/high) (n = 417) 14/8 48/17 0.363
General inflammatory infiltrate (Low high) (n = 473) 10/22 40/46 0.137
Tumour CD4+T- lymphocytic infiltrate (Low/medium/high) (n = 474) 18/3/10 25/17/42 0.016
Tumour CD8+T- lymphocytic infiltrate (Low/medium/high) (n = 474) 11/7/13 28/16/40 0.675
Tumour CD138+B- lymphocytic infiltrate (Low/medium/high) (n = 473) 19/3/9 42/8/34 0.251
Tumour CD68+macrophages infiltrate
(Low/medium/high) (n = 471)
14/7/10 36/17/31 0.71
Loco-regional treatment (Lumpectomy+radiotherapy/mastectomy+radiotherapy) (n = 483) 16/16 35/51 0.367
Systemic treatment (ER-based treatment) (hormonal/hormonal+chemotherapy/chemotherapy/none) (n = 476) 8/7/14/2 21/8/46/9 0.356
Cancer specific survival (months)* 163 (148–179) 129 (114–144) 0.013
*Mean (95%CI).
(n = 118).
doi:10.1371/journal.pone.0100759.t003
RUNX1 in Triple Negative Breast Cancer
PLOS ONE | www.plosone.org 4 June 2014 | Volume 9 | Issue 6 | e100759
protein levels including age (P,0.05), ER status (P,0.10), PR
status (P,0.05), tumour lymphocyte and macrophage infiltrate (all
P,0.05). In contrast, in those patients with triple negative receptor
status only the tumour CD4 lymphocytic infiltrate was significantly
associated with positive RUNX1 protein levels (Table 3; P,0.05).
The relationship between RUNX1 expression and clinical
outcome was then assessed by looking at cancer-specific survival in
the full cohort as shown in Figure 2B. Survival analyses showed no
significant difference between RUNX1 negative (mean of 156
months - 95% confidence interval, 146–165 months) and RUNX1
positive tumours (mean of 149 months - 95% confidence interval,
143–155 months) (Figure 2B). Minimum follow-up was 142
months; the median follow-up of the survivors was 164 months.
110 patients developed recurrence; 18 local, 71 distant, 6 with
both and 15 with no information available. During the follow up
period, 207 patients died and of these, 95 deaths could be directly
attributed to their disease.
Impact of RUNX1 expression on survival in breast cancer
according to hormonal status
To define the prognostic impact of RUNX1 expression in
different breast cancer subtypes, the patient cohort was divided
into 4 subgroups accordingly to their receptor status (ER+, ER–,
HER2+and ER2/PR2/HER22). The distribution of RUNX1
positive and negative samples in relation to hormonal status (ER/
PR/HER2) of the full cohort is shown in Table S1. No specific
enrichment of RUNX1 was detected in any one of the hormonally
defined subgroups, similar to what has been shown at a
transcriptomic level [26]. The relationship between RUNX1
expression and clinical outcome was then assessed by looking at
cancer-specific survival in each breast cancer subtype. Survival
analyses showed no difference between the RUNX1 positive and
negative groups in the ER+and HER2+patients (Figure 3A, 3C).
However, RUNX1 showed a positive association with worse
prognosis in the ER2(Figure 3B) and in the triple negative (TN)
(Figure 3D) patients. In the TN subgroup mean cancer-specific
survival of RUNX1 positive patients was 129 months (95% CI,
114–144 months) compared to 163 months (95% CI, 148–179
months) of the RUNX1 negative group. The relationships between
RUNX1 and clinico-pathological characteristics were examined in
patients with ER– (Table S2) and TN tumours (Table 3). In
addition to a significant increase in CD4+T-lymphocytic infiltrate
(P,0.05), RUNX1 positive tumours showed a significant increase
in CD138+B- lymphocytic infiltrate (P,0.05) in ER- patients
(Table S2). In a univariate analysis the presence of RUNX1 was
associated with poorer cancer-specific survival for patients with
ER- tumours (Table 4, p,0.05) and showed a tendency towards
significance as an independent prognostic marker in multivariate
Figure 2. RUNX1 expression and cancer-specific survival in primary operable breast cancer. (A) Representative examples of invasive
breast carcinomas in a tissue microarray containing 483 breast cancers which were positive (left) and negative (right) for RUNX1 expression. Note the
nuclear staining in the tumour epithelium. Scale bar represents 100 mm. (B) The association between the absence and the presence of RUNX1 and
cancer-specific survival in primary operable breast cancer (n = 483). Survival curves are plotted for patients with cancers scored positive for RUNX1
(solid line), or negative for RUNX1 expression (dotted line). P.0.1, P-value calculated using Log Rank (Mantel-Cox) test.
doi:10.1371/journal.pone.0100759.g002
RUNX1 in Triple Negative Breast Cancer
PLOS ONE | www.plosone.org 5 June 2014 | Volume 9 | Issue 6 | e100759
Table 4. The relationship between clinic-pathological characteristics of patients with ER- negative primary operable invasive ductal breast cancer and recurrence-free/cancer-
specific survival.
Recurrence-free survival Cancer-specific survival
Univariate
survival analysis
Multivariate
survival analysis
Univariate
survival analysis
Multivariate
survival analysis
Clinico-pathological
characteristics (total)
Hazard ratio
(95% CI)
P
-value
Hazard ratio
(95% CI)
P
-value
Hazard ratio
(95% CI)
P
-value
Hazard ratio
(95% CI)
P
-value
Age (#50/.50 years) 0.79 (0.45–1.38) 0.401 1.02 (0.57–1.83) 0.948
Size (#20/21–50/.50 mm) 2.01 (1.20–3.38) 0.008 1.98 (1.18–3.33) 0.01 2.02 (1.20–3.40) 0.008 1.81 (1.04–3.13) 0.035
Tumour type (Special type/lobular/ductal) 2.26 (0.81–6.36) 0.121 3.48 (0.78–15.56) 0.103
Grade (I/II/III) 1.10 (0.69–1.74) 0.701 1.71 (0.94–3.11) 0.077 0.435
Involved lymph node (Negative/positive) 1.91 (1.07–3.39) 0.027 0.108 2.83 (1.54–5.21) 0.001 2.24 (1.19–4.24) 0.013
RUNX1 (Negative/positive) 1.77 (0.88–3.54) 0.108 2.29 (1.07–4.88) 0.033 2.09 (0.97–4.48) 0.058
doi:10.1371/journal.pone.0100759.t004
Table 5. The relationship between clinic-pathological characteristics of patients with triple negative primary operable invasive ductal breast cancer and recurrence-free/cancer-
specific survival.
Recurrence-free survival Cancer-specific survival
Univariate
survival analysis
Multivariate
survival analysis
Univariate
survival analysis
Multivariate
survival analysis
Clinico-pathological
characteristics (total)
Hazard ratio
(95% CI)
P
-value
Hazard ratio
(95% CI)
P
-value
Hazard ratio
(95% CI)
P
-value
Hazard ratio
(95% CI)
P
-value
Age (#50/.50 years) 0.95 (0.44–2.04) 0.888 1.18 (0.57–2.46) 0.659
Size (#20/21–50/.50 mm) 2.53 (1.23–5.21) 0.012 2.31 (1.14–4.65) 0.019 2.76 (1.45–5.25) 0.002 2.63 (1.36–5.09) 0.004
Tumour type (Special type/lobular/ductal) 5.33 (0.36–78.77) 0.223 3.18 (0.73–13.83) 0.122
Grade (I/II/III) 0.97 (0.49–1.93) 0.926 1.40 (0.64–3.07) 0.404
Involved lymph node (Negative/positive) 2.48 (1.15–5.35) 0.021 2.19 (1.00–4.81) 0.05 4.15 (1.91–9.02) ,0.001 4.01 (1.83–8.81) 0.001
RUNX1 (Negative/positive) 3.40 (1.02–11.28) 0.046 3.00 (0.90–10.05) 0.075 4.03 (1.23–13.27) 0.022 3.83 (1.16–12.67) 0.028
doi:10.1371/journal.pone.0100759.t005
RUNX1 in Triple Negative Breast Cancer
PLOS ONE | www.plosone.org 6 June 2014 | Volume 9 | Issue 6 | e100759
analysis (p = 0.058). More interestingly, RUNX1 was significantly
associated with poorer recurrence-free survival and cancer-specific
survival for patients with triple negative disease (Table 5, p = 0.046
and p = 0.022 respectively). Using multivariate analysis RUNX1
expression was an independent prognostic marker for cancer
specific-survival in the TN subtype when assessed against
established pathological prognostic factors such as tumour size,
grade, tumour type and lymph node status (Table 5).
Discussion
Recent studies have highlighted a novel link for RUNX1 with
breast cancer [13–16] but to date no direct assessment of RUNX1
protein has been carried out. We have now addressed this need
and show that 366/483 (76%) of invasive breast carcinomas in a
tumour tissue microarray were positive for RUNX1 protein. Our
analysis reveals that there was no difference in overall survival of
the full patient cohort, or in ER+,PR+and HER2+subgroups,
when stratified on RUNX1 expression. However on univariate
analysis, positive RUNX1 expression was significantly associated
with poorer cancer-specific survival in the ER2(P,0.05) and
triple negative (ER2/PR2/HER22)(P,0.05) groups of patients.
There was also a trend towards significance on multivariate Cox
regression analysis of cancer-specific survival in ER2breast
cancer (P,0.10) which reached significance in triple negative
breast cancer (TNBC) (P,0.05). TNBC, which accounts for 15%
to 20% of breast cancers, is an aggressive disease, associated with a
significantly higher probability of relapse and poorer overall
survival when compared with other breast cancer subtypes [27].
The lack of identified molecular targets in the majority of TNBCs
means that chemotherapy remains the treatment of choice for
these patients and unfortunately early relapse after chemotherapy
is common [28]. Hence there is an urgent need for identification of
better prognostic markers and novel therapeutic targets for this
subtype [3,29]. Only a few markers have so far been identified as
having a predictive role for the prognosis of TNBC patients
Figure 3. RUNX1 expression and cancer-specific survival in different subtypes of breast cancer. The association between the absence
and the presence of RUNX1 and cancer-specific survival in patients with (A) ER-positive, (B) ER-negative, (C) HER2-positive and (D) triple negative (TN)
primary operable breast cancer. Survival is plotted for patients with cancers positive for RUNX1 (solid line), or those with no RUNX1 expression
(dotted line). ER+cohort (n = 297), p = 0.974; ER– (n = 184), p = 0.028; HER2+cohort (n = 73), p = 0.406; TN cohort (n = 118), p = 0.013. P-values
calculated using Log Rank (Mantel-Cox) test.
doi:10.1371/journal.pone.0100759.g003
RUNX1 in Triple Negative Breast Cancer
PLOS ONE | www.plosone.org 7 June 2014 | Volume 9 | Issue 6 | e100759
[30,31]. Our results now suggest the utility of RUNX1 as a novel
biomarker. In fact, regression analysis using the Cox’s propor-
tional Hazards model confirmed that RUNX1 has prognostic
value together with tumour size and lymph node status in the
TNBC subgroup. Furthermore, multivariate analysis indicated
that RUNX1 expression was independent of the established
pathological prognostic factors currently used in the clinic making
it a new putative prognostic indicator for TN tumours.
It is intriguing that even though RUNX1 was expressed in most
breast cancer cell lines (Figure 1) and the majority of patients
(Table S1) regardless of hormonal status, it was only in the
hormone-negative patients that RUNX1 expression correlated
with patient outcome. Our data therefore indicate that TNBCs
expressing RUNX1 represent a group of tumours with the poorest
prognosis and suggest that in this subtype RUNX1 may be
contributing to tumour progression. If RUNX1 has a pro-
oncogenic role in TNBCs, the question arises as to why this effect
is not observed in tumours expressing the oestrogen receptor. It is
possible that this is being masked by the capacity of RUNX1 to
attenuate or distort ER signalling [26]. In this scenario RUNX1
would be exerting opposing effects; dampening ER driven growth
yet inducing latent tumour aggression. These results may explain
why in a recent flurry of papers ascribing a tumour suppressor role
for RUNX1 in breast cancer [12–16], mutations were found
almost exclusively in ER+cancers. Of course it will be important
to definitively establish a pro-oncogenic role for RUNX1 in
TNBC and understand why expression is maintained in the most
aggressive subtype. Interestingly our data are supported by several
transcriptomic studies that have identified RUNX1 as a possible
oncogene in TNBC [32–34]. In particular RUNX1 is among a 264
gene signature which correlates with a poor prognosis in TNBC
[33] whilst another study demonstrated an inverse correlation
between RUNX1 expression and survival in the claudin low group
of TNBCs [32]. RUNX1 was also among the top 20% differentially
expressed genes in two TN subtypes identified by cluster analysis,
namely the ‘mesenchymal stem-like’ (MSL), and ‘luminal andro-
gen receptor’ (LAR) subtypes [34]. The MSL subtype also displays
low expression of claudins 3, 4, and 7, supporting a possible link
between RUNX1 expression and the claudin-low subtype.
Inflammation has been shown to represent a critical component
of tumour progression [35]. Of significance, RUNX1 expression
correlates with the presence of lymphocytic CD4+infiltrate in
TNBC. RUNX1 is one of the key factors that drives various
aspects of T-cell differentiation including regulation of cytokine
production [36]. We could speculate that in TNBC highly positive
for RUNX1, that RUNX1 would drive a transcriptional
programme in breast cancer cells resulting in production and
secretion of high levels of cytokines which would then lead to
recruitment of lymphocytic cells at the tumour site. Further studies
will clarify the significance of the correlation between RUNX1
and CD4 lymphocytes in TNBC.
The widespread expression of RUNX1 in TNBC also suggests
new therapeutic avenues for the treatment of TNBCs; for example
the development of small-molecule inhibitors which bind to CBFb
and inhibit RUNX1 activity opens the possibility of a RUNX1-
specific targeted therapy [37,38]. In addition, work from
Tumbar’s laboratory has shown that RUNX1 overexpression
leads to STAT3 activation and is necessary for skin and oral
cancer growth [39]. STAT3 is involved in human breast cancer
with high STAT3 levels correlating with poorer survival [40]. If
further studies can establish if RUNX1 overexpression and
STAT3 activation are conserved in human breast cancer, and in
TNBC in particular, this could pave the way for new treatment
options based on the use of STAT3 inhibitors. Taken together our
results identify RUNX1 as a new biomarker in TNBC and are
opening exciting possibilities for the development of novel targeted
therapies for this subgroup.
Supporting Information
Figure S1 Validation of the RUNX1 antibody. RUNX1
antibody specificity was confirmed by western blot using known
positive (T6i, hMEC-RUNX1) and negative (3SS, hMEC-Puro)
controls. GAPDH used as a loading control. T6i; leukaemia cell
line overexpressing RUNX1. 3SS; leukaemia cell line deleted for
RUNX1. hMEC-TERT (immortalized human mammary epithe-
lial cells) transfected with RUNX1 (hMEC-RUNX1) or empty
vector (hMEC-Puro).
(TIF)
Table S1 Distribution of RUNX1 expression in relation to
breast cancer hormonal status.
(DOCX)
Table S2 The relationship between RUNX1 and clinico-
pathological characteristics of ER- patients with primary operable
invasive ductal breast cancer (n = 184).
(DOCX)
Acknowledgments
The authors wish to thank Barbara Chaneton, Anna Kilbey and Gillian
Borland for the kind gifts of cell lines and vectors. We thank Catherine
Winchester for critical reading of the manuscript. The image analysis
system used in the study was gifted by the Think Pink charity and Western
Infirmary Breast Endowment Fund.
Author Contributions
Conceived and designed the experiments: NF EC JE KB. Performed the
experiments: NF ZM CN SM. Analyzed the data: NF ZM EM DM JM EC
JE KB. Contributed reagents/materials/analysis tools: ZM EM DM JE.
Wrote the paper: NF ZM JE KB.
References
1. Taneja P, Maglic D, Kai F, Zhu S, Kendig RD, et al. (2010) Classical and Novel
Prognostic Markers for Breast Cancer and their Clinical Significance. Clin Med
Insights Oncol 4: 15–34.
2. Higgins MJ, Baselga J (2011) Targeted therapies for breast cancer. J Clin Invest
121: 3797–3803.
3. Foulkes WD, Smith IE, Reis-Filho JS (2010) Triple-negative breast cancer.
N Engl J Med 363: 1938–1948.
4. Chuang LSH, Ito K, Ito Y (2013) RUNX family: Regulation and diversification
of roles through interacting proteins. Int J Cancer 132: 1260–1271.
5. Blyth K, Cameron ER, Neil JC (2005) The RUNX genes: gain or loss of
function in cancer. Nat Rev Cancer 5: 376–387.
6. Lam K, Zhang D-E (2012) RUNX1 and RUNX1-ETO: roles in hematopoiesis
and leukemogenesis. Front Biosci 17: 1120–1139.
7. Taniuchi I, Osato M, Ito Y (2012) Runx1 : no longer just for leukemia. EMBO J
31: 4098–4099.
8. Scheitz CJF, Tumbar T (2013) New insights into the role of Runx1 in epithelial
stem cell biology and pathology. J Cell Biochem 114: 985–993.
9. Yeh H-Y, Cheng S-W, Lin Y-C, Yeh C-Y, Lin S-F, et al. (2009) Identifying
significant genetic regulatory networks in the prostate cancer from microarray
data based on transcription factor analysis and conditional independency. BMC
Med Genomics 2: 70.
10. Hoi CSL, Lee SE, Lu S-Y, McDermitt DJ, Osorio KM, et al. (2010) Runx1
directly promotes proliferation of hair follicle stem cells and epithelial tumor
formation in mouse skin. Mol Cell Biol 30: 2518–2536.
11. Keita M, Bachvarova M, Morin C, Plante M, Gregoire J, et al. (2013) The
RUNX1 transcription factor is expressed in serous epithelial ovarian carcinoma
RUNX1 in Triple Negative Breast Cancer
PLOS ONE | www.plosone.org 8 June 2014 | Volume 9 | Issue 6 | e100759
and contributes to cell proliferation, migration and invasion. Cell Cycle 12: 972–
986.
12. Janes KA (2011) RUNX1 and its understudied role in breast cancer. Cell Cycle
10: 3461–3465.
13. Wang L, Brugge JS, Janes KA (2011) Intersection of FOXO- and RUNX1-
mediated gene expression programs in single breast epithelial cells during
morphogenesis and tumor progression. Proc Natl Acad Sci U S A 108: E803–12.
14. Ellis MJ, Ding L, Shen D, Luo J, Suman VJ, et al. (2012) Whole-genome analysis
informs breast cancer response to aromatase inhibition. Nature 486: 353–360.
15. Banerji S, Cibulskis K, Rangel-Escareno C, Brown KK, Carter SL, et al. (2012)
Sequence analysis of mutations and translocations across breast cancer subtypes.
Nature 486: 405–409.
16. Cancer Genome Atlas Network (2012) Comprehensive molecular portraits of
human breast tumours. Nature 490: 61–70.
17. Kadota M, Yang HH, Gomez B, Sato M, Clifford RJ, et al. (2010) Delineating
genetic alterations for tumor progression in the MCF10A series of breast cancer
cell lines. PLoS One 5: e9201.
18. Mohammed ZMA, Edwards J, Orange C, Mallon E, Doughty JC, et al. (2012)
Breast cancer outcomes by steroid hormone receptor status assessed visually and
by computer image analysis. Histopathology 61: 283–292.
19. Mohammed ZMA, Going JJ, Edwards J, Elsberger B, Doughty JC, et al. (2012)
The relationship between components of tumour inflammatory cell infiltrate and
clinicopathological factors and survival in patients with primary operable
invasive ductal breast cancer. Br J Cancer 107: 864–873.
20. Mohammed ZMA, Going JJ, McMillan DC, Orange C, Mallon E, et al. (2012)
Comparison of visual and automated assessment of HER2 status and their
impact on outcome in primary ope rable invasive ductal breast ca ncer.
Histopathology 61: 675–684.
21. Mohammed ZMA, Orange C, McMillan DC, Mallon E, Doughty JC, et al.
(2013) Comparison of visual and automated assessment of microvessel density
and their impact on outcome in primary operable invasive ductal breast cancer.
Hum Pathol 44: 1688–1695.
22. Mohammed ZMA, Going JJ, Edwards J, Elsberger B, McMillan DC (2013) The
relationship between lymphocyte subsets and clinico-pathological determinants
of survival in patients with primary operable invasive ductal breast cancer.
Br J Cancer 109: 1676–1684.
23. Kirkegaard T, Edwards J, Tovey S, McGlynn LM, Krishna SN, et al. (2006)
Observer variation in immunohistochemical analysis of protein expression, time
for a change? Histopathology 48: 787–794.
24. Wotton S, Stewart M, Blyth K, Vaillant F, Kilbey A, et al. (2002) Proviral
insertion indicates a dominant oncogenic role for Runx1/AML-1 in T-cell
lymphoma. Cancer Res 62: 7181–7185.
25. Thibodeaux CA, Liu X, Disbrow GL, Zhang Y, Rone JD, et al. (2009)
Immortalization and transformation of human mammary epithelial cells by a
tumor-derived Myc mutant. Breast Cancer Res Treat 116: 281–294.
26. Chimge N-O, Frenkel B (2013) The RUNX family in breast cancer:
relationships with estrogen signaling. Oncogene 32: 2121–2130.
27. Arnedos M, Bihan C, Delaloge S, Andre F (2012) Triple-negative breast cancer:
are we making headway at least? Ther Adv Med Oncol 4: 195–210.
28. Gluz O, Liedtke C, Gottschalk N, Pusztai L, Nitz U, et al. (2009) Triple-nega tive
breast cancer–current status and future directions. Ann Oncol 20: 1913–1927.
29. Lehmann BD, Pietenpol JA (2014) Identification and use of biomarkers in
treatment strategies for triple-negative breast cancer subtypes. J Pathol 232: 142–
150.
30. Zhou L, Li K, Luo Y, Tian L, Wang M, et al. (2013) Novel prognostic markers
for patients with triple-negative breast cancer. Hum Pathol 44: 2180–2187.
31. Abdel-Fatah TMA, Perry C, Dickinson P, Ball G, Moseley P, et al. (2013) Bcl2 is
an independent prognostic marker of triple negative breast cancer (TNBC ) and
predicts response to anthracycline combination (ATC) chemotherapy (CT) in
adjuvant and neoadjuvant settings. Ann Oncol 24: 2801–2807.
32. Rody A, Karn T, Liedtke C, Pusztai L, Ruckhaeberle E, et al. (2011) A clinically
relevant gene signature in triple negative and basal-like breast cancer. Breast
Cancer Res 13: R97.
33. Karn T, Pusztai L, Holtrich U, Iwamoto T, Shiang CY, et al. (2011)
Homogeneous Datasets of Triple Negative Breast Cancers Enable the
Identification of Novel Prognostic and Predictive Signatures. PLoS One 6:
e28403.
34. Lehmann BD, Bauer JA, Chen X, Sanders ME, Chakravarthy AB, et al. (2011)
Identification of human triple-negative breast cancer subtypes and preclinical
models for selection of targeted therapies. J Clin Invest 121: 2750–2767.
35. Grivennikov SI, Greten FR, Karin M (2010) Immunity, inflammation, and
cancer. Cell 140: 883–899.
36. Wong WF, Kohu K, Nakamura A, Ebina M, Kikuchi T, et al. (2012) Runx1
Deficiency in CD4+T Cells Causes Fatal Autoimmune Inflammatory Lung
Disease Due to Spontaneous Hyperactivation of Cells. J Immunol 188: 5408–
5420.
37. Gorczynski MJ, Grembecka J, Zhou Y, Kong Y, Roudaia L, et al. (2007)
Allosteric Inhibition of the Protein-Protein Interaction between the Leukemia-
Associated Proteins Runx1 and CBFb. Chem Biol 14: 1186–1197.
38. Cunningham L, Finckbeiner S, Hyde RK, Southall N, Marugan J, et al. (2012)
Identification of benzodiazepine Ro5-3335 as an inhibitor of CBF leukemia
through quantitative high throughput screen against RUNX1-CBFbinteraction.
Proc Natl Acad Sci U S A 109: 14592–14597.
39. Scheitz CJF, Lee TS, McDe rmitt DJ, Tumbar T (2012) Defining a tissue stem
cell-driven Runx1/Stat3 signalling axis in epithelial cancer. EMBO J 31: 4124–
4139.
40. Diaz N, Minton S, Cox C, Bowman T, Gritsko T, et al. (2006) Activation of
stat3 in primary tumors from high-risk breast cancer patients is associated with
elevated levels of activated SRC and survivin expression. Clin Canc er Res 12:
20–28.
RUNX1 in Triple Negative Breast Cancer
PLOS ONE | www.plosone.org 9 June 2014 | Volume 9 | Issue 6 | e100759
... Hypothetically, this makes Auger electron emitters an ideal source for internal radiotherapy as, under a perfect drug delivery scenario, there would be minimal adverse normal tissue effects. There have been many attempts to develop TRT agents labeled with Auger-electron-emitting radionuclides, especially with 125 I and 111 In (Table 1 [ [16][17][18][19][20][21][22][23][24][25]) (e.g., [ 125 I]IUdR [16], 125 I-labeled huA33 [17], 111 In-DTPA-octreotide [19], and 111 In-DTPA-EGFR [21]). However, the therapeutic efficacy of these agents has been modest or low in the clinical trials performed to date. ...
... Hypothetically, this makes Auger electron emitters an ideal source for internal radiotherapy as, under a perfect drug delivery scenario, there would be minimal adverse normal tissue effects. There have been many attempts to develop TRT agents labeled with Auger-electron-emitting radionuclides, especially with 125 I and 111 In (Table 1 [ [16][17][18][19][20][21][22][23][24][25]) (e.g., [ 125 I]IUdR [16], 125 I-labeled huA33 [17], 111 In-DTPA-octreotide [19], and 111 In-DTPA-EGFR [21]). However, the therapeutic efficacy of these agents has been modest or low in the clinical trials performed to date. ...
... Hypothetically, this makes Auger electron emitters an ideal source for internal radiotherapy as, under a perfect drug delivery scenario, there would be minimal adverse normal tissue effects. There have been many attempts to develop TRT agents labeled with Auger-electron-emitting radionuclides, especially with 125 I and 111 In (Table 1 [ [16][17][18][19][20][21][22][23][24][25]) (e.g., [ 125 I]IUdR [16], 125 I-labeled huA33 [17], 111 In-DTPA-octreotide [19], and 111 In-DTPA-EGFR [21]). However, the therapeutic efficacy of these agents has been modest or low in the clinical trials performed to date. ...
Article
Full-text available
The present review aims to explore the potential targets/partners for future targeted radionuclide therapy (TRT) strategies, wherein cancer cells often are not killed effectively, despite receiving a high average tumor radiation dose. Here, we shall discuss the key factors in the cancer genome, especially those related to DNA damage response/repair and maintenance systems for escaping cell death in cancer cells. To overcome the current limitations of TRT effectiveness due to radiation/drug-tolerant cells and tumor heterogeneity, and to make TRT more effective, we propose that a promising strategy would be to target the DNA maintenance factors that are crucial for cancer survival. Considering their cancer-specific DNA damage response/repair ability and dysregulated transcription/epigenetic system, key factors such as PARP, ATM/ATR, amplified/overexpressed transcription factors, and DNA methyltransferases have the potential to be molecular targets for Auger electron therapy; moreover, their inhibition by non-radioactive molecules could be a partnering component for enhancing the therapeutic response of TRT.
... A follow-up study supported the idea that RUNX1 mainly acts as a tumor suppressor in ER+ breast cancer, and it can exert oncogenic effects by suppressing the estrogen-mediated inhibition of AXIN1 and activation of the Wnt/β-catenin signaling pathway [41]. In contrast, it has also been shown that RUNX1 levels are abnormally elevated in triple-negative breast cancer (TNBC) and this is associated with a poor prognosis, indicating that RUNX1 plays a pro-tumor role in TNBC [42]. An analysis of data from multiple databases confirmed the abnormally high expression levels of RUNX1 in cervical cancer [43]. ...
... Furthermore, multivariate Cox regression analyses have substantiated that elevated RUNX1 levels serve as an independent prognostic factor for poor overall survival (OS) in this patient population [96]. In TNBC patients, elevated levels of RUNX1 are associated with poor prognosis and have been established as an independent prognostic marker through multivariate Cox regression analysis [42]. However, certain studies have also reached the opposite conclusion, finding that the absence of RUNX1 expression in breast cancer is associated with activation of the TGF-β and WNT signaling pathways, and that a low RUNX1 expression level suggests a poor prognosis in breast cancer [97]. ...
Article
Full-text available
Runt-related transcription factors (RUNX) are a family of transcription factors that are essential for normal and malignant hematopoietic processes. Their most widely recognized role in malignancy is to promote the occurrence and development of acute myeloid leukemia. However, it is worth noting that during the last decade, studies of RUNX proteins in solid tumors have made considerable progress, suggesting that these proteins are directly involved in different stages of tumor development, including tumor initiation, progression, and invasion. RUNX proteins also play a role in tumor angiogenesis, the maintenance of tumor cell stemness, and resistance to antitumor drugs. These findings have led to the consideration of RUNX as a tumor biomarker. All RUNX proteins are involved in the occurrence and development of solid tumors, but the role of each RUNX protein in different tumors and the major signaling pathways involved are complicated by tumor heterogeneity and the interacting tumor microenvironment. Understanding how the dysregulation of RUNX in tumors affects normal biological processes is important to elucidate the molecular mechanisms by which RUNX affects malignant tumors.
... Conversely, there is evidence of RUNX1 over-expression in various cases of ER negative and triple negative breast cancer, supporting a pro-oncogenic role of RUNX1 correlated with poor prognosis and survival [104][105][106][107][108]. RUNX1 expression was noted to be significantly high in some cases of the more invasive subtypes of breast cancer [96]. ...
... Conversely, there is evidence of RUNX1 over-expression in various cases of ER negative and triple negative breast cancer, supporting a pro-oncogenic role of RUNX1 correlated with poor prognosis and survival [104][105][106][107][108]. RUNX1 expression was noted to be significantly high in some cases of the more invasive subtypes of breast cancer [96]. Studies analyzing tissue microarrays (TMA) from TN breast cancer patients have correlated elevated RUNX1 expression to poor survival outcome [108][109][110]. In vitro studies using ER negative breast cancer cell lines have shown that RUNX1 associates with super-enhancers known to interact with other oncogenic transcription factors, such as MYC [111]. ...
Article
Full-text available
Dissecting and identifying the major actors and pathways in the genesis, progression and aggressive advancement of breast cancer is challenging, in part because neoplasms arising in this tissue represent distinct diseases and in part because the tumors themselves evolve. This review attempts to illustrate the complexity of this mutational landscape as it pertains to the RUNX genes and their transcription co-factor CBFβ. Large-scale genomic studies that characterize genetic alterations across a disease subtype are a useful starting point and as such have identified recurring alterations in CBFB and in the RUNX genes (particularly RUNX1). Intriguingly, the functional output of these mutations is often context dependent with regards to the estrogen receptor (ER) status of the breast cancer. Therefore, such studies need to be integrated with an in-depth understanding of both the normal and corrupted function in mammary cells to begin to tease out how loss or gain of function can alter the cell phenotype and contribute to disease progression. We review how alterations to RUNX/CBFβ function contextually ascribe to breast cancer subtypes and discuss how the in vitro analyses and mouse model systems have contributed to our current understanding of these proteins in the pathogenesis of this complex set of diseases.
... 17 Some TFs, such as RARA, SMAD3, KLF7, BCL6, FOXO3, RUNX1, and NR2F2, were also classified as common TFs, which have been implicated in the regulation of gene activity, cell proliferation, cell death, and tumor survival. [18][19][20][21][22][23] However, AR in prostate cells, 24 POU1F1 in brain tissue, 25 SHOX in bone, 26 ZSCAN4 in placenta tissue, 23 and VSX2 in retina 27 were considered specific TFs in this study. Those TFs have been shown to function in specific tissues by TF marker, which is a comprehensive manually curated database of TFs and related markers with experimental evidence from humanspecific cell/tissue types. ...
Article
Full-text available
A core transcription regulatory circuitry (CRC) is an interconnected self-regulatory circuitry that is formed by a group of core transcription factors (TFs). These core TFs collectively regulate gene expression by binding not only to their own super enhancers (SEs) but also to the SEs of one another. For most human tissue/cell types, a global view of CRCs and core TFs has not been generated. Here, we identified numerous CRCs using two identification methods and detailed the landscape of the CRCs driven by SEs in large cell/tissue samples. The comprehensive biological analyses, including sequence conservation, CRC activity and genome binding affinity were conducted for common TFs, moderate TFs, and specific TFs, which exhibit different biological features. The local module located from the common CRC network highlighted the essential functions and prognostic performance. The tissue-specific CRC network was highly related to cell identity. Core TFs in tissue-specific CRC networks exhibited disease markers, and had regulatory potential for cancer immunotherapy. Moreover, a user-friendly resource named CRCdb (http://www.licpathway.net/crcdb/index.html) was developed, which contained the detailed information of CRCs and core TFs used in this study, as well as other interesting results, such as the most representative CRC, frequency of TFs, and indegree/outdegree of TFs.
Article
Full-text available
Introduction Angiogenesis is one of the hallmarks of cancer and can impact the processes of cancer initiation, progression, and response to therapy. Background Anti-angiogenic therapy is thus an encouraging therapeutic option to treat cancers, but the detailed angiogenic mechanisms and the association between angiogenesis and clinical outcome remain unknown in different cancers. Methods Here, we systematically assess the impacts of 82 angiogenesis-associated genes (AAGs) in tumor tissue specificity and prognosis across 16 cancer types. Results Results demonstrate that the expression patterns of the 82 AAGs can reflect the tumor tissue specificity, and high expressions of up-regulated AAGs are significantly associated with poor prognosis of cancer. We further define a prognostic score for predicting overall survival (OS) based on the expressions of up-regulated AAGs and confirm its reliable predictive ability. Results indicate that a low prognostic score demonstrates a superior OS and vice versa. Conclusion The results of this study will contribute to the understanding of different tumor angiogenesis mechanisms in various tissues and cancer-personalized anti-angiogenic treatment. The code of our analysis can be accessed at https://github.com/ZhenshenBao/AAGs_analysis.git.
Article
Full-text available
Background Opioids are the primary analgesics for cancer pain. Recent clinical evidence suggests opioids may counteract the effect of immune checkpoint inhibition (ICI) immunotherapy, but the mechanism for this interaction is unknown. The following experiments study how opioids and immunotherapy modulate a common RNA expression pathway in triple negative breast cancer (TNBC), a cancer subtype in which immunotherapy is increasingly used. This study identifies a mechanism by which opioids may decrease ICI efficacy, and compares ketamine, a non-opioid analgesic with emerging use in cancer pain, for potential ICI interaction. Methods Tumor RNA expression and clinicopathologic data from a large cohort with TNBC (N=286) was used to identify RNA expression signatures of disease. Various drug-induced RNA expression profiles were extracted from multimodal RNA expression datasets and analyzed to estimate the RNA expression effects of ICI, opioids, and ketamine on TNBC. Results We identified a RNA expression network in CD8⁺ T-cells that was relevant to TNBC pathogenesis and prognosis. Both opioids and anti-PD-L1 ICI regulated RNA expression in this network, suggesting a nexus for opioid-ICI interaction. Morphine and anti-PD-L1 therapy regulated RNA expression in opposing directions. By contrast, there was little overlap between the effect of ketamine and anti-PD-L1 therapy on RNA expression. Conclusions Opioids and ICI may target a common immune network in TNBC and regulate gene expression in opposing fashion. No available evidence supports a similar interaction between ketamine and ICI.
Article
Full-text available
Background Gemcitabine (GEM)-based chemotherapy is the first-line option for pancreatic ductal adenocarcinoma (PDAC). However, the development of drug resistance limits its efficacy, and the specific mechanisms remain largely unknown. RUNX1, a key transcription factor in hematopoiesis, also involved in the malignant progression of PDAC, but was unclear in the chemoresistance of PDAC. Methods Comparative analysis was performed to screen GEM-resistance related genes using our single-cell RNA sequencing(scRNA-seq) data and two public RNA-sequencing datasets (GSE223463, GSE183795) for PDAC. The expression of RUNX1 in PDAC tissues was detected by qRT-PCR, immunohistochemistry (IHC) and western blot. The clinical significance of RUNX1 in PDAC was determined by single-or multivariate analysis and survival analysis. We constructed the stably expressing cell lines with shRUNX1 and RUNX1, and successfully established GEM-resistant cell line. The role of RUNX1 in GEM resistance was determined by CCK8 assay, plate colony formation assay and apoptosis analysis in vitro and in vivo. To explore the mechanism, we performed bioinformatic analysis using the scRNA-seq data to screen for the endoplasm reticulum (ER) stress signaling that was indispensable for RUNX1 in GEM resistance. We observed the cell morphology in ER stress by transmission electron microscopy and validated RUNX1 in gemcitabine resistance depended on the BiP/PERK/eIF2α pathway by in vitro and in vivo oncogenic experiments, using ER stress inhibitor(4-PBA) and PERK inhibitor (GSK2606414). The correlation between RUNX1 and BiP expression was assessed using the scRNA-seq data and TCGA dataset, and validated by RT-PCR, immunostaining and western blot. The mechanism of RUNX1 regulation of BiP was confirmed by ChIP-PCR and dual luciferase assay. Finally, the effect of RUNX1 inhibitor on PDAC was conducted in vivo mouse models, including subcutaneous xenograft and patient-derived xenograft (PDX) mouse models. Results RUNX1 was aberrant high expressed in PDAC and closely associated with GEM resistance. Silencing of RUNX1 could attenuate resistance in GEM-resistant cell line, and its inhibitor Ro5-3335 displayed an enhanced effect in inhibiting tumor growth, combined with GEM treatment, in PDX mouse models and GEM-resistant xenografts. In detail, forced expression of RUNX1 in PDAC cells suppressed apoptosis induced by GEM exposure, which was reversed by the ER stress inhibitor 4-PBA and PERK phosphorylation inhibitor GSK2606414. RUNX1 modulation of ER stress signaling mediated GEM resistance was supported by the analysis of scRNA-seq data. Consistently, silencing of RUNX1 strongly inhibited the GEM-induced activation of BiP and PERK/eIF2α signaling, one of the major pathways involved in ER stress. It was identified that RUNX1 directly bound to the promoter region of BiP, a primary ER stress sensor, and stimulated BiP expression to enhance the reserve capacity for cell adaptation, which in turn facilitated GEM resistance in PDAC cells. Conclusions This study identifies RUNX1 as a predictive biomarker for response to GEM-based chemotherapy. RUNX1 inhibition may represent an effective strategy for overcoming GEM resistance in PDAC cells.
Article
Full-text available
Among patients with triple-negative breast cancer (TNBC), several studies have suggested that deregulated microRNA (miRNA) expression may be associated with a more aggressive phenotype. Although tumor molecular signatures may be race- and/or ethnicity-specific, there is limited information on the molecular profiles in women with TNBC of Hispanic and Latin American ancestry. We simultaneously profiled TNBC biopsies for the genome-wide copy number and miRNA global expression from 28 Latina women and identified a panel of 28 miRNAs associated with copy number alterations (CNAs). Four selected miRNAs (miR-141-3p, miR-150-5p, miR-182-5p, and miR-661) were validated in a subset of tumor and adjacent non-tumor tissue samples, with miR-182-5p being the most discriminatory among tissue groups (AUC value > 0.8). MiR-141-3p up-regulation was associated with increased cancer recurrence; miR-661 down-regulation with larger tumor size; and down-regulation of miR-150-5p with larger tumor size, high p53 expression, increased cancer recurrence, presence of distant metastasis, and deceased status. This study reinforces the importance of integration analysis of CNAs and miRNAs in TNBC, allowing for the identification of interactions among molecular mechanisms. Additionally, this study emphasizes the significance of considering the patients ancestral background when examining TNBC, as it can influence the relationship between intrinsic tumor molecular characteristics and clinical manifestations of the disease.
Article
Background and objective: Many classification tasks in translational bioinformatics and genomics are characterised by the high dimensionality of potential features and unbalanced sample distribution among classes. This can affect classifier robustness and increase the risk of overfitting, curse of dimensionality and generalization leaks; furthermore and most importantly, this can prevent obtaining adequate patient stratification required for precision medicine in facing complex diseases, like cancer. Setting up a feature selection strategy able to extract only proper predictive features by removing irrelevant, redundant, and noisy ones is crucial to achieving valuable results on the desired task. Methods: We propose a new feature selection approach, called ReRa, based on supervised Relevance-Redundancy assessments. ReRa consists of a customized step of relevance-based filtering, to identify a reduced subset of meaningful features, followed by a supervised similarity-based procedure to minimize redundancy. This latter step innovatively uses a combination of global and class-specific similarity assessments to remove redundant features while preserving those differentiated across classes, even when these classes are strongly unbalanced. Results: We compared ReRa with several existing feature selection methods to obtain feature spaces on which performing breast cancer patient subtyping using several classifiers: we considered two use cases based on gene or transcript isoform expression. In the vast majority of the assessed scenarios, when using ReRa-selected feature spaces, the performances were significantly increased compared to simple feature filtering, LASSO regularization, or even MRmr - another Relevance-Redundancy method. The two use cases represent an insightful example of translational application, taking advantage of ReRa capabilities to investigate and enhance a clinically-relevant patient stratification task, which could be easily applied also to other cancer types and diseases. Conclusions: ReRa approach has the potential to improve the performance of machine learning models used in an unbalanced classification scenario. Compared to another Relevance-Redundancy approach like MRmr, ReRa does not require tuning the number of preserved features, ensures efficiency and scalability over huge initial dimensionalities and allows re-evaluation of all previously selected features at each iteration of the redundancy assessment, to ultimately preserve only the most relevant and class-differentiated features.
Article
Full-text available
We analysed primary breast cancers by genomic DNA copy number arrays, DNA methylation, exome sequencing, messenger RNA arrays, microRNA sequencing and reverse-phase protein arrays. Our ability to integrate information across platforms provided key insights into previously defined gene expression subtypes and demonstrated the existence of four main breast cancer classes when combining data from five platforms, each of which shows significant molecular heterogeneity. Somatic mutations in only three genes (TP53, PIK3CA and GATA3) occurred at .10% incidence across all breast cancers; however, there were numerous subtype-associated and novel gene mutations including the enrichment of specific mutations in GATA3, PIK3CA and MAP3K1 with the luminal A subtype. We identified two novel protein-expression-defined subgroups, possibly produced by stromal/microenvironmental elements, and integrated analyses identified specific signalling pathways dominant in each molecular subtype including a HER2/phosphorylated HER2/EGFR/phosphorylated EGFR signature within the HER2-enriched expression subtype. Comparison of basal-like breast tumours with high-grade serous ovarian tumours showed many molecular commonalities, indicating a related aetiology and similar therapeutic opportunities. The biological finding of the four main breast cancer subtypes caused by different subsets of genetic and epigenetic abnormalities raises the hypothesis that much of the clinically observable plasticity and heterogeneity occurs within, and not across, these major biological subtypes of breast cancer.
Article
Full-text available
Background: The importance of lymphocyte subtypes in determining outcome in primary operable ductal invasive breast cancer remains unclear. The aim of present study was to examine the relationship between tumour lymphocyte subsets infiltrate and standard clinico-pathological factors and survival in patients with primary operable invasive ductal breast cancer. Methods: The analysis of the inflammatory cell infiltrate, including lymphocyte subtypes, was undertaken using immunohistochemical techniques and visual quantitative and semi-quantitative techniques in 338 patients with ductal breast cancer. Results: The majority (91%) of patients had high grade inflammatory cell infiltrate. The median follow-up of the survivors was 164 months. During this period, 65 died of their cancer. On univariate analysis, tumour inflammatory cell infiltrate, macrophages infiltrate (P<0.05), lymphocytic infiltrate (P<0.001) and CD8+ T-lymphocytic infiltrate (P<0.01) were associated with improved cancer-specific survival, whereas neutrophil (P<0.05) and CD138+ B-lymphocytic infiltrate (P<0.001) were associated with poorer cancer-specific survival. On multivariate analysis, tumour lymphocytic infiltrate (P<0.001), macrophage infiltrate (P<0.05), CD8+ T-lymphocytic infiltrate (P<0.01) and CD138+ B-lymphocytic infiltrate (P<0.001) were independently associated with cancer survival. When the significant inflammatory cell types were included with tumour-based factors in multivariate analysis only tumour size (Hazard ratios (HR): 2.55, 95% confidence interval (CI): 1.53–4.27, P<0.001), Ki-67 index (HR: 2.08, 95% CI: 1.08–4.00, P<0.05), lymphovascular invasion (HR: 4.40, 95% CI: 2.07–9.35, P<0.001), macrophage infiltrate (HR: 0.49, 95% CI: 0.33–0.73, P<0.001), lymphocytic infiltrate (HR: 0.11, 95% CI: 0.05–0.23, P<0.001), CD8+ T-lymphocytic infiltrate (HR: 0.57, 95% CI: 0.38–0.87, P<0.001) and CD138+ B-lymphocytic infiltrate (HR: 2.86, 95% CI: 1.79–4.56, P<0.001) were independently associated with cancer survival. Conclusion: The majority of patients with invasive ductal breast cancer had high-grade inflammatory cell infiltrate. In these patients, inflammatory cells including macrophage and lymphocytic infiltrate, and subsets CD8+ T-lymphocytic infiltrate and CD138+ B-lymphocytic infiltrate had superior prognostic value, compared with hormone status and lymph node involvement in patients with primary operable invasive ductal breast cancer.
Article
Full-text available
TNBC represents a heterogeneous subgroup of BC with poor prognosis and frequently resistant to CT. The relationship between Bcl2 immunohistochemical protein expression and clinico-pathological outcomes was assessed in 736 TNBC-patients: 635 patients had early primary-TNBC (EP-TNBC) and 101 had primary locally advanced (PLA)-TNBC treated with neo-adjuvant- ATC-CT. Negative Bcl2 (Bcl2-) was observed in 70% of EP-TNBC and was significantly associated with high proliferation, high levels of P-Cadherin, E-Cadherin and HER3 (P's < 0.01), while Bcl2+ was significantly associated with high levels of p27, MDM4 and SPAG5 (P < 0.01). After controlling for chemotherapy and other prognostic factors, Bcl2- was associated with 2-fold increased risk of death (P = 0.006) and recurrence (P = 0.0004). Furthermore, the prognosis of EP-TNBC/Bcl2- patients had improved both BC-specific survival (P = 0.002) and disease-free survival (P = 0.003), if they received adjuvant-ATC-CT. Moreover, Bcl2- expression was an independent predictor of pathological complete response of primary locally advanced triple negative breast cancer (PLA-TNBC) treated with neoadjuvant-ATC-CT (P = 0.008). Adding Bcl2 to the panel of markers used in current clinical practice could provide both prognostic and predictive information in TNBC. TNBC/Bcl2- patients appear to benefit from ATC-CT, whereas Bcl2+ TNBC seems to be resistant to ATC-CT and may benefit from a trial of different type of chemotherapy with/without novel-targeted agents.
Article
The Runx1 transcription factor is abundantly expressed in naïve T cells but rapidly down-regulated in activated T cells, suggesting that it plays an important role in a naïve stage. In the present study, we showed that mice harboring Runx1-deleted CD4+ T cells developed a fatal autoimmune lung disease. CD4+ T cells from these mice were spontaneously activated and preferentially homed to the lung. At early age, lung peribronchiovascular regions of the mice were infiltrated with CD4+ lymphocytes, while at later stage, interstitial regions were massively occupied with immune cells and alveolar spaces were filled with granular exudates that resembled pulmonary alveolar proteinosis in humans. Mice suffered from respiratory failure as well as systemic inflammatory responses which caused early dead at around 7-8 months old. Runx1-deleted CD4+ T cells in the mice expressed various cytokines including IL-17 and IL-21 which promoted neutrophils and monocytes activation and triggered formation of IgG+ plasma B cells, respectively. Among these, the deregulation of IL-21 transcription was likely to be associated with Runx-binding sites located in an IL-21 intron. Our data indicate that Runx1 plays an essential role in repressing the transcription of cytokine genes in naïve CD4+ T cells and thereby maintains cell quiescence.
Article
Triple negative breast cancer (TNBC) is a heterogeneous disease with distinct molecular subtypes that differentially respond to chemotherapy and targeted agents. The absence of high-frequency molecular alterations and a limited number of known biomarkers have limited the development of therapeutic strategies for the disease. Herein, we summarize the results of the first round of targeted therapy approaches in TNBC and discuss new preclinical strategies. Common themes emerge from the proposed strategies, such as the use of biomarkers to identify tumors with genomic instability, targeting adapted molecular states resulting from tumor suppressor loss, and targeting altered metabolic pathways.
Article
Triple-negative breast cancer (TNBC) is a subtype of breast cancer characterized by poor prognosis. Currently, no reliable markers have been identified as having a predictive role for the prognosis of TNBC patients. In this study, 119 breast cancer samples, including 31 TNBC and 89 non-TNBC subtypes, were collected. The protein levels of cleaved caspase-3 (CC3), aldehyde dehydrogenase 1 (ALDH1), cyclooxygenase-2, Ki-67, H2A histone family member X, and phosphorylated protein kinase B protein were measured by immunohistochemical staining. The percentage of positive CC3 (P = .017), ALDH1 (P = .015), Ki-67 (P = .001), and H2A histone family member X (P = .016) staining was significantly higher in TNBC than in non-TNBC cases. Positive CC3 and ALDH1 staining significantly correlated with poor prognosis of breast cancer, the TNBC subtype and non-TNBC subtype. Positive cyclooxygenase-2 expression significantly correlated with the survival of patients with TNBC. Multivariate analysis demonstrated that CC3 and ALDH1 are independent prognostic factors for BC and non-TNBC. ALDH1 is a prognostic marker for TNBC patients.
Article
Angiogenesis is essential for tumor growth and metastasis, and several studies have reported increased angiogenesis, as quantified by microvessel density, to be a powerful prognostic tool in breast cancer. Therefore, there is considerable interest in automated assessment of microvessel density with possible benefits in improved accuracy, increased precision, and handling workload. Visual and automated assessment of microvessel density (CD34(+)) and survival were examined in patients with primary operable invasive ductal breast cancer. Tissue microarrays (n = 356 patients) immunostained for microvessel density (CD34(+)) were scored visually and automatically with the Slidepath Tissue IA system (Dublin, Ireland). Visual and automated microvessel density (CD34(+)) were in agreement (interclass correlation coefficient = 0.69, P < .001). Visual but not the automated method for microvessel density (CD34(+)) was associated with locoregional treatment and metastasis. On univariate survival analysis, visual but not automated method for microvessel density (CD34(+)) was associated with recurrence-free and cancer-specific survival in patients with invasive ductal breast cancer (P < .01). Although automated assessment of microvessel density (CD34(+)) is in reasonable agreement, it poorly predicts outcome in patients with operable invasive ductal breast cancer.