ArticlePDF Available

Efficacy of Neoadjuvant Cisplatin in Triple-Negative Breast Cancer

Authors:

Abstract and Figures

Purpose Cisplatin is a chemotherapeutic agent not used routinely for breast cancer treatment. As a DNA cross-linking agent, cisplatin may be effective treatment for hereditary BRCA1-mutated breast cancers. Because sporadic triple-negative breast cancer (TNBC) and BRCA1-associated breast cancer share features suggesting common pathogenesis, we conducted a neoadjuvant trial of cisplatin in TNBC and explored specific biomarkers to identify predictors of response. Patients and Methods Twenty-eight women with stage II or III breast cancers lacking estrogen and progesterone receptors and HER2/Neu (TNBC) were enrolled and treated with four cycles of cisplatin at 75 mg/m ² every 21 days. After definitive surgery, patients received standard adjuvant chemotherapy and radiation therapy per their treating physicians. Clinical and pathologic treatment response were assessed, and pretreatment tumor samples were evaluated for selected biomarkers. Results Six (22%) of 28 patients achieved pathologic complete responses, including both patients with BRCA1 germline mutations;18 (64%) patients had a clinical complete or partial response. Fourteen (50%) patients showed good pathologic responses (Miller-Payne score of 3, 4, or 5), 10 had minor responses (Miller-Payne score of 1 or 2), and four (14%) progressed. All TNBCs clustered with reference basal-like tumors by hierarchical clustering. Factors associated with good cisplatin response include young age (P = .001), low BRCA1 mRNA expression (P = .03), BRCA1 promoter methylation (P = .04), p53 nonsense or frameshift mutations (P = .01), and a gene expression signature of E2F3 activation (P = .03). Conclusion Single-agent cisplatin induced response in a subset of patients with TNBC. Decreased BRCA1 expression may identify subsets of TNBCs that are cisplatin sensitive. Other biomarkers show promise in predicting cisplatin response.
Predictors of response to cisplatin therapy in triple-negative basal-like tumors. (A) The sample dendrogram of gene expression hierarchical cluster analysis with the intrinsic genes 47 is shown. Cisplatin pretreatment samples (sample numbers in red) are co-clustered with a reference set of breast tumors (sample numbers in black). Intrinsic subtype of the reference cases, determined by an independent hierarchical cluster analysis, is indicated by the color bar below the dendrogram as follows: luminal A, dark blue; luminal B, light blue; ErbB2, green; normal-like, purple; basal-like, red. Cisplatin response of the trial patients is indicated on the lower row as follows: resistant (progression, Miller-Payne score of 1 or 2) in gray; sensitive (Miller-Payne score of 3, 4, or 5) in black. (*) Trial cases with pathologic complete response (pCR; Miller-Payne score of 5). (B) Relationship of BRCA1 biomarkers and p53 family biomarkers to cisplatin sensitivity. Each trial patient is indicated by sample number, and patients are arranged according to relative response to cisplatin chemotherapy. Progression or Miller-Payne response scores are indicated above each sample. Predictive biomarker positivity is indicated with solid circles as follows: i the presence of a BRCA1 germline mutation, ii the lowest quartile of BRCA1 mRNA expression measured by quantitative reverse transcriptase polymerase chain reaction, iii the presence of BRCA1 promoter methylation, iv the ratio of mRNA expression levels of Np63/TAp73 measured by quantitative reverse transcriptase polymerase chain reaction 2, and v the presence of p53 protein-truncating mutations. For each biomarker, samples with no data are indicated by a gray X; in addition, for BRCA1 mRNA expression and promoter methylation, a gray X indicates " not applicable " for the two cases with known BRCA1 germline mutation. NSM, nonsense or frameshift mutations.
… 
Relationship of cisplatin treatment response to clinical and molecular features. (A) The patient age in years (y-axis) and Miller-Payne pathologic response score to neoadjuvant cisplatin therapy (x-axis) are plotted for each patient in the cohort as indicated by solid circles (P .001 based on ordered quartiles of age). (B) Relative BRCA1 mRNA level measured by quantitative reverse transcriptase polymerase chain reaction (PCR; 2 CT ) 48 is plotted for resistant tumors (solid circles) and sensitive tumors (open circles). The average mRNA level of each group is indicated by a black horizontal line. Measurements were performed using PCR primer pairs encompassing exons 1 and 2 (E1/E2), exons 16 and 17 (E16/E17), and exons 19 and 20 (E19/E20) as indicated along the bottom of the plot. The Wilcoxon P values for difference between sensitive and resistant tumors are indicated above each primer pair as follows: (***) P .020; (**) P .048; (*) P .098. (C) Electrophoresis of PCR products spanning the BRCA1 promoter from bisulfite-treated DNA. Each lane contains products generated from separate PCR reactions using primers specific for methylated (m) or unmethylated (u) DNA template. Bacterial methylase-treated lymphocyte DNA was used for the positive control (). DNA from normal lymphocytes was used as a negative control (). The lane marked MW indicates molecular weight markers measured in base pairs (bp). Paired methylated-and unmethylated-specific primer reactions are marked by a line over the paired lanes and labeled corresponding to the template DNA used in the reaction (positive control, negative control, and patient No.). Patients 17 and 23 demonstrate bands in both the unmethylated (u) and methylated (m) lanes indicating the presence of BRCA1 promoter methylation. Patients 16, 20, 21, 22, and 24 lack bands with the methylated primer pair, signifying the absence of BRCA1 promoter methylation. (D) The E2F3 signature score (y-axis) and Miller-Payne pathologic response scores (x-axis) plotted (solid circles) for each patient in the cohort with available gene expression array data (Pearson correlation, 0.46; P .025).
… 
Content may be subject to copyright.
Efficacy of Neoadjuvant Cisplatin in Triple-Negative
Breast Cancer
Daniel P. Silver, Andrea L. Richardson, Aron C. Eklund, Zhigang C. Wang, Zoltan Szallasi, Qiyuan Li,
Nicolai Juul, Chee-Onn Leong, Diana Calogrias, Ayodele Buraimoh, Aquila Fatima, Rebecca S. Gelman,
Paula D. Ryan, Nadine M. Tung, Arcangela De Nicolo, Shridar Ganesan, Alexander Miron, Christian Colin,
Dennis C. Sgroi, Leif W. Ellisen, Eric P. Winer, and Judy E. Garber
From the Dana-Farber Cancer Institute;
Brigham and Women’s Hospital; Chil-
dren’s Hospital Informatics Program at
the Harvard–Massachusetts Institute of
Technology Division of Health Sciences
and Technology; Massachusetts General
Hospital Cancer Center; Beth Israel
Deaconess Hospital, Harvard Medical
School; Harvard School of Public Health,
Boston, MA; Center for Biological
Sequence Analysis, BioCentrum-Technical
University of Denmark, Lyngby, Denmark;
Cancer Institute of New Jersey, Robert
Wood Johnson Medical School, Univer-
sity of Medicine and Dentistry of New
Jersey, New Brunswick, NJ.
Submitted February 13, 2009; accepted
October 23, 2009; published online
ahead of print at www.jco.org on
January 25, 2010.
Supported by Grants No. CA089393
from the National Cancer Institute
Program of Research Excellence
(SPORE) in Breast Cancer at the Dana-
Farber/Harvard Cancer Center and No.
R21LM008823-01A1 from the National
Institutes of Health, and by the Breast
Cancer Research Foundation, Sidney
Kimmel Foundation, Avon supplement
to the Dana-Farber/Harvard Cancer
Center support grant, and Susan G.
Komen for the Cure.
D.P.S. and A.L.R. contributed equally to
this work.
Presented in part at the San Antonio
Breast Cancer Conference, December
14-17, 2006, San Antonio, TX, and the
National Cancer Institute Translational
Science Meeting, November 7-9, 2008,
Washington, DC.
Terms in blue are defined in the glos-
sary, found at the end of this article
and online at www.jco.org.
Authors’ disclosures of potential con-
flicts of interest and author contribu-
tions are found at the end of this
article.
Clinical Trials repository link available on
JCO.org.
Corresponding author: Judy E. Garber,
MD, MPH, Dana-Farber Cancer Insti-
tute, Department of Medical Oncology,
Smith 209, 1 Jimmy Fund Way,
Boston, MA 02115; e-mail: judy_
garber@dfci.harvard.edu.
© 2010 by American Society of Clinical
Oncology
0732-183X/10/2807-1145/$20.00
DOI: 10.1200/JCO.2009.22.4725
ABSTRACT
Purpose
Cisplatin is a chemotherapeutic agent not used routinely for breast cancer treatment. As a DNA
cross-linking agent, cisplatin may be effective treatment for hereditary BRCA1-mutated breast cancers.
Because sporadic triple-negative breast cancer (TNBC) and BRCA1-associated breast cancer share
features suggesting common pathogenesis, we conducted a neoadjuvant trial of cisplatin in TNBC
and explored specific biomarkers to identify predictors of response.
Patients and Methods
Twenty-eight women with stage II or III breast cancers lacking estrogen and progesterone
receptors and HER2/Neu (TNBC) were enrolled and treated with four cycles of cisplatin at 75
mg/m
2
every 21 days. After definitive surgery, patients received standard adjuvant chemotherapy
and radiation therapy per their treating physicians. Clinical and pathologic treatment response
were assessed, and pretreatment tumor samples were evaluated for selected biomarkers.
Results
Six (22%) of 28 patients achieved pathologic complete responses, including both patients with
BRCA1 germline mutations;18 (64%) patients had a clinical complete or partial response. Fourteen
(50%) patients showed good pathologic responses (Miller-Payne score of 3, 4, or 5), 10 had minor
responses (Miller-Payne score of 1 or 2), and four (14%) progressed. All TNBCs clustered with
reference basal-like tumors by hierarchical clustering. Factors associated with good cisplatin
response include young age (P.001), low BRCA1 mRNA expression (P.03), BRCA1 promoter
methylation (P.04), p53 nonsense or frameshift mutations (P.01), and a gene expression
signature of E2F3 activation (P.03).
Conclusion
Single-agent cisplatin induced response in a subset of patients with TNBC. Decreased BRCA1
expression may identify subsets of TNBCs that are cisplatin sensitive. Other biomarkers show
promise in predicting cisplatin response.
J Clin Oncol 28:1145-1153. © 2010 by American Society of Clinical Oncology
INTRODUCTION
Triple-negative breast cancers (TNBCs), those
that do not express estrogen or progesterone re-
ceptors or contain an amplified HER2/Neu gene,
often demonstrate sensitivity to cytotoxic neoad-
juvant treatment regimens
1-4
; however, no specific
molecular targets or chemotherapeutic vulnerabili-
ties have been identified. TNBCs comprise 15% to
20% of breast cancers in Western countries, and
the vast majority are sporadic.
5
Approximately
70% of breast cancers in individuals carrying a
germline BRCA1 mutation are triple negative;
BRCA1-associated and sporadic TNBCs share many
histopathologic features. They are almost always
high grade,
6
with common histologic
7-11
and cyto-
keratin expression patterns,
12,13
and share molecu-
lar features, including frequent p53 mutation
14
and
abnormalities of the inactivated X chromosome.
15
Both BRCA1-associated and sporadic TNBCs are
typically basal-like by hierarchical clustering of tran-
scriptional profiles.
16,17
Further, these tumors share
a pattern of genomic instability characterized by al-
lelic loss.
6,15
These similarities have led to specula-
tion that BRCA1-associated and at least a subset of
sporadic TNBCs may share defects in a BRCA1-
associated pathway; DNA repair has received the
most attention.
18
BRCA1-deficient cells are particularly sus-
ceptible to the interstrand cross-linking agents
JOURNAL OF CLINICAL ONCOLOGY ORIGINAL REPORT
VOLUME 28 NUMBER 7 MARCH 1 2010
© 2010 by American Society of Clinical Oncology 1145
mitomycin and cisplatin. A cell line from a BRCA1-associated breast
tumor was shown to be defective in DNA double-strand break repair
19
and was also cisplatin sensitive
20
; these properties were reversed by
adding wild-type BRCA1.
19,20
BRCA1-deficient tumors in mouse
models also demonstrated cisplatin sensitivity.
21
Some cell lines rep-
resenting sporadic TNBCs show cisplatin and mitomycin sensitivity
(D. Silver and D.M. Livingston, unpublished data), suggesting that
these tumors may have defects in the BRCA1 pathway.
Given these observations, we conducted a neoadjuvant trial of
four cycles of cisplatin in TNBC. The trial end point was pathologic
response. We analyzed pretreatment specimens for predictors of re-
sponse to cisplatin, including BRCA1 expression levels, and BRCA1
promoter methylation. Other features that may predispose to cisplatin
sensitivity, including gene expression patterns, p53 mutation, and the
presence of a cisplatin-specific apoptosis pathway involving the p53
family members p63 and p73, were also explored.
PATIENTS AND METHODS
Patients
Newly diagnosed patients with T1, N1-3, M0 or T2-4, N0-3, M0 breast
cancers (with tumors 1.5 cm), negative for estrogen and progesterone
receptors defined as 1% nuclear staining by immunohistochemistry, HER2/
Neu0or1by immunohistochemistry, or HER2 nonamplified by fluorescent
in situ hybridization were eligible for this trial.
Study Design and Treatment Plan
A core biopsy was performed to obtain tumor tissue for study, and a
radio-opaque clip was placed in the tumor bed; four treatments of cisplatin at
75 mg/m
2
every 21 days were administered. Patients then received definitive
surgery, including an axillary lymph node dissection in patients with positive
sentinel lymph node biopsy. The specimen was evaluated for chemotherapy
response, with focused sampling of the tumor bed marked by the radio-
opaque clip. The Miller-Payne scoring system
21a
was used to assess tumor
response. A score of 3, 4, or 5, with 5 being a pathologic complete response
(pCR), is hereafter termed a good response.
For details of the patients on trial, study design and treatment plan,
specimen analysis, Exon Grouping Analysis genotyping, gene array analysis,
quantitative reverse transcriptase polymerase chain reaction (qRT-PCR), and
microarray data analysis, see Supplemental Methods (online only).
RESULTS
Patient Characteristics
Twenty-nine women were enrolled on the study: one was ineli-
gible and never received protocol treatment. The last two patients
Table 1. Clinical Characteristics and Response to Treatment of the Study Population (N 28)
Patient No. Age (years) BRCA1 Germline
Baseline Tumor
Diameter by MRI (cm)
Baseline
Nodal Status†
Clinical
Response
Pathologic Response
(Miller-Payne scale)‡
15 57 wt 6.5 SD Progression
21 59 wt 2.4 PD Progression
26 39 wt 3.2 PD Progression
27 63 wt 3.3 PD Progression
4 68 wt 2.5 SD 1
6 62 wt Not done cPR 1
12 53 wt 2.7 SD 1
13 56 wt 3.2 cPR 1
16 45 wt 7.0 SD 1
14 43 wt 2.3 cPR 2
20 69 wt 5.0 SD 2
22 67 wt 4.7 cPR 2
24 50 wt 3.3 cPR 2
28 60 wt 3.7 cPR 2
1 59 wt 3.0 cPR 3
11 41 wt 4.0 SD 3
23 29 wt 4.5 cPR 3
25 40 wt 2.4 cPR 3
2 49 wt 4.0 SD 4
7 39 wt 3.7 cCR 4
8 51 wt 4.2 cPR 4
10 43 wt 2.5 cCR 4
3 39 wt 4.5 cPR 5
5 44 mut 5.8 cPR 5
9 31 wt 4.0 cPR 5
17 52 wt 2.0 cCR 5
18 48 mut 2.8 cCR 5
29 44 wt 6.3 cPR 5
Abbreviations: MRI, magnetic resonance imaging; wt, wild type, no germline mutation; SD, stable disease; PD, progressive disease; cPR, clinical partial response;
cCR, clinical complete response; mut, presence of a pathogenic germline BRCA1 mutation.
BRCA1 germline genotype determined as in Patients and Methods section.
†Axillary lymph node status determine at baseline; indicates presence of lymph node metastasis determined by sentinel node biopsy or fine-needle aspiration;
indicates sentinel node biopsy negative for metastasis.
‡Pathologic assessment of response to treatment using Miller-Payne method and grading scale: progression off study prior to surgery for clinical progression
or additional nonprotocol therapy, 1 no or minimal reduction in tumor, 2 up to 30% reduction, 3 30% to 90% reduction, 4 ⫽⬎90% reduction but with some
residual invasive (or axillary) disease, and 5 no residual invasive carcinoma or axillary metastasis (pathologic complete response).
Silver et al
1146 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY
entered signed consent forms concurrently, so a total of 28 instead of
27 women were treated on study. Patient age ranged from 29 to 69
years at diagnosis (Table 1). The median pretreatment tumor size,
determined by magnetic resonance imaging, was 3.7 cm (range, 2.0 to
7.0 cm); 13 patients had axillary metastases. Research genotyping
detected only the two BRCA1 germline mutations previously identi-
fied through clinical testing. Research biopsies of primary tumor pro-
vided adequate material for most molecular analyses from 24 of the
28 patients.
Treatment Response
Eighteen patients had a clinical response (Table 1, 14 had a partial
response and four had a complete response), for an estimated re-
sponse rate of 64% (95% conditional CI, 44% to 81%). The Miller-
Payne score for responses to four cycles of cisplatin are listed in Table
1. Six patients had pCR (21%; 95% conditional CI, 9% to 43%), and
eight additional patients had significant pathologic partial responses
defined as Miller-Payne 3 or 4 (29%, for an overall good response rate
of 50%; 95% conditional CI, 31% to 70%). Four patients had clinical
progression while on cisplatin.
Table 2 lists the distribution of various clinical and pathologic
characteristics by the response outcomes (Miller-Payne 3, 4, and 5
responses, pCR, and overall clinical response). Neither tumor size (by
magnetic resonance imaging) nor axillary lymph node positivity were
significantly related to any of the three response outcomes (P.48
and P.43, respectively).
Table 2. Covariates and Response Variables
Covariate
Miller-Payne 3,4,5 Responses pCR
Clinical Response
(CR and PR)
No. of Patients Observed (%) P
Observed (%) P
Observed (%) P
All 28 50 — 21 64 —
Age, years .001† .13† .46†
29-41 (Q1) 7 86 29 71
42-49 (Q2) 7 71 43 71
50-59 (Q3) 8 38 13 63
60-69 (Q4) 6 0 0 50
Tumor size, cm (by MRI) .75† .48† .83†
Unknown 1 — — —
2.0-2.7 (Q1) 6 50 17 67
2.8-3.7 (Q2) 7 29 14 57
3.8-4.5 (Q3) 8 88 25 75
4.6-7.0 (Q4) 6 33 33 50
Lymph nodes 1.00‡ 1.00‡ .43‡
Negative 15 53 20 73
Positive 13 46 23 54
BRCA1 mRNA levels,
arbitrary relative units
.03† .79† .65†
Unknown/NA 7 57 42 71
0.00-0.03 (Q1) 5 100 0 80
0.04-0.23 (Q2) 6 33 33 50
0.25-0.44 (Q3) 5 40 0 60
0.57-3.69 (Q4) 5 20 20 60
BRCA1 methylation .04‡ 1.00‡ .40‡
Unknown/NA 5 80 60 80
Negative 15 27 13 53
Positive 8 75 13 75
Np63/TAp73 ratio .39‡ .26‡ .66‡
Unknown 6 50 33 67
2 9 67 33 56
21338869
Type of p53 mutation .03‡ .78‡ .09‡
Unknown 6 50 33 67
MSM 10 30 10 60
NSM 6 100 .01§ 33 1.00§ 100 .23§
wt 6 33 .64¶ 17 1.00¶ 33 .14¶
Abbreviations: pCR, pathologic complete response; CR, complete response; PR, partial response; Q1/Q2/Q3/Q4, first, second, third, and fourth quartiles, respectively; MRI,
magnetic resonance imaging; NA, not assessed because of BRCA1 mutation; MSM, missense mutation; NSM, nonsense or frameshift mutation; wt, wild type (no mutation).
Pvalues when patients with unknown values were omitted.
†Fisher’s exact test on four ordered categories of the covariate (equivalent to a Wilcoxon rank sum test using the quartile number as the observation and comparing
responders to nonresponders).
‡Fisher’s exact test on unordered categories of the covariate.
§Fisher’s exact test on NSM vMSM.
¶Fisher’s exact test on wt vMSM NSM.
Neoadjuvant Cisplatin in Triple-Negative Breast Cancer
www.jco.org © 2010 by American Society of Clinical Oncology 1147
Toxicity
Severe toxicity was uncommon. One patient had a grade 4 eleva-
tion of AST/AST. There were nine grade 3 toxicities reported: tinnitus,
neutropenia, fatigue, hyperkalemia, elevation of ALT/ALT, nausea,
myalgia, skin toxicity, and GI toxicity.
Predictors of Response
The TNBCs tend to be classified as basal-like in gene expression
array hierarchical cluster analysis using the intrinsic genes.
12,22
We
determined the intrinsic subtype of all cases with adequate material
(n 24) by co-clustering these cases with a reference set of tumors for
which intrinsic subtype had been determined independently (Supple-
mental Fig 1, Supplemental Methods). All of the trial TNBCs co-
clustered with the reference basal-like tumors (Fig 1A). Hierarchical
clustering with the intrinsic genes did not reveal distinct subclusters of
cisplatin-resistant or -sensitive tumors.
Age
There was a strong association between younger age and good
response (P.001 based on quartiles of age, significant even after
A
B
0.0
0.2
0.4
0.6
0.8
1.0
1.2
Subtype
Response
A22
A70
A72
A47
A96
A187
A43
A98
A5
A40
A71
A1
A1b
A29
A20
A91
A66
A90
A85
A7
A10
A12
A12b
A80
A80b
A28
A17
A44
A30
A54
A2
A55
A117
A115
A63
A63b
A25
A25b
A145
A113
A122
A136
A55b
Ayo2
A120
A124
P10
P28
P11
P4
P3
P22
A125
P25
P14
P1
P15
P27
A11
P17
P26
P6
P5
P16
A116
A116b
P29
A21
A21b
P7
A141
P2
P23
P12
A35
A38
A38b
P21
P8
A147
A134
A140
P24
A118
A123
Resistant Sensitive
Progress 12 345
Response Score
i) BRCA1
Mutation
ii) Low BRCA1 mRNA
iii) BRCA1 methylation
iv) ΔNp63/TAp73 > 2
v) p53 NSM
Sample No. 15 21 26 27 4 6 12 13 16 14 20 22 24 3 5 9 17 181 11 23 25 2 7 8 10
28 29
x x
xx
xx
x
x
x
x
x
x
x
x
xx x
xx x
xx
xx
****
Fig 1. Predictors of response to cisplatin therapy in triple-negative basal-like tumors. (A) The sample dendrogram of gene expression hierarchical cluster analysis with
the intrinsic genes
47
is shown. Cisplatin pretreatment samples (sample numbers in red) are co-clustered with a reference set of breast tumors (sample numbers in
black). Intrinsic subtype of the reference cases, determined by an independent hierarchical cluster analysis, is indicated by the color bar below the dendrogram as
follows: luminal A, dark blue; luminal B, light blue; ErbB2, green; normal-like, purple; basal-like, red. Cisplatin response of the trial patients is indicated on the lower row
as follows: resistant (progression, Miller-Payne score of 1 or 2) in gray; sensitive (Miller-Payne score of 3, 4, or 5) in black. (*) Trial cases with pathologic complete
response (pCR; Miller-Payne score of 5). (B) Relationship of BRCA1 biomarkers and p53 family biomarkers to cisplatin sensitivity. Each trial patient is indicated by sample
number, and patients are arranged according to relative response to cisplatin chemotherapy. Progression or Miller-Payne response scores are indicated above each
sample. Predictive biomarker positivity is indicated with solid circles as follows: i the presence of a BRCA1 germline mutation, ii the lowest quartile of BRCA1
mRNA expression measured by quantitative reverse transcriptase polymerase chain reaction, iii the presence of BRCA1 promoter methylation, iv the ratio of mRNA
expression levels of Np63/TAp73 measured by quantitative reverse transcriptase polymerase chain reaction 2, and v the presence of p53 protein-truncating
mutations. For each biomarker, samples with no data are indicated by a gray X; in addition, for BRCA1 mRNA expression and promoter methylation, a gray X indicates
not applicablefor the two cases with known BRCA1 germline mutation. NSM, nonsense or frameshift mutations.
Silver et al
1148 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY
Bonferroni adjustment for multiple comparisons; when the two
BRCA1 mutations carriers were excluded, P.001 and the Miller-
Payne response rate in patients age 42 to 49 years decreased to 60%;
Table 2; Fig 2A). This effect was not attributable to decreased dose or
dose delays in older patients (data not shown). Age was not signifi-
cantly associated with pCR (P.13) or clinical response (P.46).
BRCA1 Genotype
The two BRCA1 mutation carriers achieved pCR; the other four
patients with pCR were germline BRCA1 wild type (Table 1, Fig 1Bi).
Without the two BRCA1 mutation carriers, the overall clinical re-
sponse rate was 16 (62%) of 26, the good response rate was 12 (46%)
of 26, and the pCR rate was four (15%) of 26.
BRCA1 mRNA Expression
Lower BRCA1 mRNA expression (measured by qRT-PCR) was
significantly associated with a larger percent of patients having good
response (P.03; Table 2 and Figs 1Bii and 2B). All five patients with
the lowest quartile of BRCA1 expression had a good response while
only five of the 16 other patients with BRCA1 expression data had
good responses. Multiple pairs of primers gave similar results. Exclud-
ing the tumors from BRCA1 carriers, the remaining three evaluable
tumors showing pCR did not express BRCA1 at low levels. However,
five of seven evaluable tumors in the Miller-Payne 3 and 4 groups had
BRCA1 expression levels in the lowest quartile. The levels of the
BRCA1 transcriptional repressor, ID4, have been shown to correlate
inversely with BRCA1 expression levels
23
; we did not see an inverse
AB
P 1 E1/E2 E16/E17 E19/E202345
Age (years)
BRCA1 mRNA (2-ΔΔCT)
Miller-Payne Score Primer Pairs
70
10.0
1.0
0.1
0.01
60
50
40
30
***** *
DC
P12345
E2F3 Signature Score
Miller-Payne Score
0.3
0.2
0.0
-0.1
-0.2
-0.3
0.1
200 bp –
100 bp –
–+16
mm mmmmmmmuu uuuuuuu
17 20 21 22 23 24
MW
Fig 2. Relationship of cisplatin treatment response to clinical and molecular features. (A) The patient age in years (y-axis) and Miller-Payne pathologic response score
to neoadjuvant cisplatin therapy (x-axis) are plotted for each patient in the cohort as indicated by solid circles (P.001 based on ordered quartiles of age). (B) Relative
BRCA1 mRNA level measured by quantitative reverse transcriptase polymerase chain reaction (PCR; 2
⫺⌬⌬CT
)
48
is plotted for resistant tumors (solid circles) and
sensitive tumors (open circles). The average mRNA level of each group is indicated by a black horizontal line. Measurements were performed using PCR primer pairs
encompassing exons 1 and 2 (E1/E2), exons 16 and 17 (E16/E17), and exons 19 and 20 (E19/E20) as indicated along the bottom of the plot. The Wilcoxon Pvalues
for difference between sensitive and resistant tumors are indicated above each primer pair as follows: (***) P.020; (**) P.048; (*) P.098. (C) Electrophoresis
of PCR products spanning the BRCA1 promoter from bisulfite-treated DNA. Each lane contains products generated from separate PCR reactions using primers specific
for methylated (m) or unmethylated (u) DNA template. Bacterial methylase-treated lymphocyte DNA was used for the positive control (). DNA from normal
lymphocytes was used as a negative control (). The lane marked MW indicates molecular weight markers measured in base pairs (bp). Paired methylated- and
unmethylated-specific primer reactions are marked by a line over the paired lanes and labeled corresponding to the template DNA used in the reaction (positive control,
negative control, and patient No.). Patients 17 and 23 demonstrate bands in both the unmethylated (u) and methylated (m) lanes indicating the presenceofBRCA1
promoter methylation. Patients 16, 20, 21, 22, and 24 lack bands with the methylated primer pair, signifying the absence of BRCA1 promoter methylation. (D) The E2F3
signature score (y-axis) and Miller-Payne pathologic response scores (x-axis) plotted (solid circles) for each patient in the cohort with available gene expression array
data (Pearson correlation, 0.46; P.025).
Neoadjuvant Cisplatin in Triple-Negative Breast Cancer
www.jco.org © 2010 by American Society of Clinical Oncology 1149
correlation between array-measured ID4 expression and BRCA1 ex-
pression measured by qRT-PCR (Spearman correlation 0.05;
P.84; data not shown).
BRCA1 Promoter Methylation
BRCA1 promoter methylation was analyzed by methylation-
specific PCR, a method sensitive to low levels of methylation (Fig 1Biii
and Fig 2C). BRCA1 expression levels were lower in tumors with
BRCA1 promoter methylation compared with tumors without (me-
dians of 0.025 and 0.33, respectively; Wilcoxon rank sum test P.06;
Supplemental Fig 2). Tumors with BRCA1 promoter methylation
were more likely to have good response, but not pCR or clinical
response, than tumors without methylation (75% v27%; P.04).
Np63/TAp73 Ratio
The transcription factor Np63
promotes survival in a subset
of tumors through its ability to repress the proapoptotic activity of the
related p53 family member TAp73.
24-26
Phosphorylation of TAp73
after cisplatin treatment causes TAp73 release from Np63
, enabling
TAp73 to activate a program of apoptosis.
25
Tumors with this pathway
intact would be predicted to be platinum sensitive and to have high
levels of Np63 relative to TAp73 to repress TAp73 activity in the
absence of cisplatin. For this reason, levels of Np63 and TAp73 were
measured by qRT-PCR using RNA from microdissected primary tu-
mor samples. Nine (41%) of the 22 evaluable tumors had a Np63/
TAp73 ratio 2, the predetermined cutoff value suggesting active
repression of TAp73 by Np63
25
(data not shown). Of these nine
tumors, six (67%) had good responses to cisplatin and three (33%)
had a pCR. Of 13 tumors with Np63/TAp73 2, five (38%) had
good responses and only one (8%) had a pCR (Fig 1Biv; P.39 for
good response; P.26 for pCR; P.66 for clinical response). Of the
complete responders with material available, three of four patients had
aNp63/TAp73 2. These results provide preliminary evidence that
aNp63/TAp73 ratio 2 associates with a greater chance of response
to cisplatin.
p53 Mutation
The sequence of the p53 gene in tumor DNA was determined in
22 patients. Six tumors had nonsense or frameshift mutations (NSM),
10 had missense mutations (MSM), and six were wild type (wt),
confirming the high frequency of p53 mutation in TNBC. There was
no significant association of good response with the presence of a p53
mutation compared with wt (P.64). However, the tumors with
NSM tended to have a higher good response percent than those in the
other two groups (100% v30% v33% in NSM, MSM, and wt groups,
respectively; P.03), and the difference in response rate between
tumors with NSM and MSM was significant (P.01). These tumors
also had a higher rate of pCR (33% v10% and 17%) and clinical
response (100% v60% and 33%) but these differences were not
significant (P.30 and P.11).
Several Predictors of Miller-Payne Response
Used Together
In an exploratory analysis of whether any of the other variables
might add to the prediction based on age, we did step-up logistic
regression for the outcome of good response, omitting the oldest age
quartile. Using this approach, BRCA1 mRNA added significantly to
predictions based on age alone (for details, see Supplemental Data).
Array Mining
We used several approaches to search for genes or gene signatures
associated with cisplatin response. First, we identified candidate genes
reported to have association with cisplatin response (eg, ERCC1,
BIRC5; see Supplemental Data 1) and genes associated with subsets
within TNBC (eg, basal keratins, EGFR,alpha B-crystallin). We eval-
uated the Pearson correlation between the Miller-Payne score and
gene expression array level of these 114 candidate genes. Only a single
gene correlation, AIFM1, remained significant after correction for
multiple hypothesis testing (corrected P.041). However, the ex-
pression level and standard deviation were low for this probe, suggest-
ing random fluctuation. A complete list of candidate genes tested and
Pvalues are listed in Supplemental Data 1.
In addition, we evaluated published gene signatures consisting of
a set of co-regulated gene expression changes in response to a specific
oncogene pathway activation
27
or specific biologic processes, includ-
ing cell cycle,
28
chromosome instability,
29
and core serum response.
30
We also tested an immune response classifier reported to have prog-
nostic value in estrogen receptor–negative breast cancers.
31
For each
signature, we calculated a score estimating the relative level of the gene
signature present in the array data from each tumor. We tested this
score for association with response using Pearson correlation (see
Supplemental Data 2). No signature was statistically significant after
Bonferroni correction for multiple hypothesis testing. The strongest
association was with a signature of E2F3 oncogenic pathway activation
(Fig 2D; r0.46; P.025), which has been associated with general
chemotherapy responsiveness.
27
Finally, we identified all genes with correlation to the Miller-
Payne response score, a standard deviation 0.5, and a Pvalue .01.
No correlation was significant after Bonferroni correction (see Sup-
plemental Data 3). The highest correlations were with inhibitor of
growth family member 3 (ING3;r0.69; P.0002) and metastasis-
associated lung adenocarcinoma transcript 1 (MALAT1), a non-
protein coding gene (r.62; P.001). The reproducibility of these
potential candidate biomarkers will have to be determined by analysis
of independent cohorts of similarly treated patients.
DISCUSSION
Six (21%) of 28 patients with TNBC achieved pCR with single-agent
neoadjuvant cisplatin. Among the 28 patients in this trial were two
BRCA1 carriers, both of whom achieved pCR; four (15%) of the 26
women with sporadic TNBC also achieved pCR to cisplatin. Overall,
50% of the patients had a good response to cisplatin defined by a
Miller-Payne score of 3, 4, or 5. These results are consistent with
preclinical and recent clinical data
32
showing that BRCA1-associated
tumors are responsive to cisplatin, and data that suggest a subset of
basal-like breast cancers with intact BRCA1 share some fundamental
molecular defects with BRCA1-deficient tumors.
The effect of increasing dose, intensity, and/or duration of cispla-
tin on response rates is unknown. Sporadic TNBCs show heterogene-
ity in response to other cytotoxic chemotherapies, with reported pCR
rates of TNBC ranging from 12% for single-agent taxane regimens to
27% to 45% in multiagent neoadjuvant trials.
1,3,4,33
Our data also
suggest heterogeneity among these patients on the basis of their re-
sponse to cisplatin. Clinical progression on cisplatin was also ob-
served: other neoadjuvant studies of TNBC either have used different
Silver et al
1150 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY
criteria or have not reported on progression, making it difficult to
make meaningful comparisons of progression rates.
3,4
We studied
a variety of biomarkers to try to discover those that would distin-
guish patients likely to respond from patients unlikely to respond
to cisplatin.
In our hands, triple negativity using a strict criteria of 1%
nuclear staining for estrogen and progesterone receptor immuno-
histochemistry, criteria for HER2 negativity of 0 or 1by immu-
nohistochemistry, or HER2 nonamplification by fluorescent in situ
hybridization reliably predicted classification into the basal-like sub-
type by hierarchical cluster analysis of the intrinsic genes.
Age correlated with response whether or not the two patients
with BRCA1 mutations were included, with younger patients more
likely to respond. The biologic explanation for this finding is unclear.
It does not seem related to cisplatin dosing or dose intensity because
these did not vary with age. Younger patients may develop subtypes of
TNBC that are more responsive to chemotherapy in general or cispla-
tin in particular. Of note, younger age was a predictor of breast cancer
sensitivity to neoadjuvant therapy with paclitaxel, fluorouracil, doxo-
rubicin, and cyclophosphamide in a recent study.
34
We tested the hypothesis that low BRCA1 expression is an expla-
nation of the phenotypic similarity of sporadic TNBC and BRCA1-
related breast cancer. BRCA1 mRNA levels were lower in patients with
cisplatin sensitivity, consistent with the suggestion that the “BRCA-
ness” of these tumors may be related to decreased BRCA1 expres-
sion.
18,23
However, the lowest BRCA1-expressing tumors were not
those with pCR (excluding the BRCA1 germline mutation tumors),
but rather were those with moderate responses to cisplatin (Miller-
Payne score of 3 or 4). BRCA1 promoter methylation, assessed by a
sensitive methylation-specific PCR assay, was also statistically signifi-
cantly correlated with response to cisplatin (P.04) and was inversely
correlated to BRCA1 mRNA levels (P.06). These data suggest that a
subset of TNBCs may be sensitive to cisplatin on the basis of low
BRCA1 expression levels. The rarity of low BRCA1 levels among
tumors with pCR to cisplatin may be a reflection of the small number
of patients analyzed and emphasizes the need to validate results of
this exploratory trial in an independent cohort. In this regard, it is
interesting that low levels of BRCA1 expression in ovarian tumors
correlate with better survival in patients treated with cisplatin-
containing regimens.
35
Our trial and a recent report
32
of a neoadjuvant trial using the
same cisplatin regimen in women with germline BRCA1 mutations
suggest that tumors from women with hereditary BRCA1 mutations
have a high rate of response to cisplatin. Whether tumors from BRCA1
mutation carriers truly represent a homogeneous group with respect
to cisplatin response or to other cytotoxic agents will require further
experience. The small number of BRCA1 mutation carriers treated to
date does not provide sufficient data for clinical use of neoadjuvant
cisplatin outside of a trial.
p53 mutation status has been investigated as a potential predictor
of chemotherapy responsiveness in solid tumors
36-40
: Nonsense and
truncating p53 mutations have been shown to be common in BRCA1-
mutated breast cancer.
41
We found a significant association of tumor
p53 protein-truncating mutations (NSM) with cisplatin response.
A pathway of apoptosis activated by cisplatin involving the p53
family members TAp73 and Np63
may play a role in cisplatin
sensitivity.
25
We measured the ratio of the mRNA of these two genes as
a marker of the potential integrity of this pathway. Although not
statistically significant, three (75%) of four patients with pCR related
to cisplatin were positive for this biomarker. This finding is consistent
with a recent retrospective analysis of response following neoadjuvant
cisplatin-based chemotherapy.
42
In exploratory analyses (given the small sample size and large
number of measured genes), no single gene on transcriptional arrays
convincingly segregated responders from nonresponders. None of a
smaller set of candidate genes reported to have some relationship to
cisplatin response showed a strong association with response. Notable
negatives include ERCC1, reported prognostic in cisplatin-treated
lung cancer,
43
and alpha B-crystallin.
44
Of several signatures consisting
of responses of many genes to a perturbation relevant for tumorigen-
esis, only one stood out in this analysis, a signature derived by overex-
pression of the transcription factor E2F3.
27
A proportion of TNBCs
have copy number gain on chromosome 6p at the E2F3 locus (Z.C.
Wang, unpublished data), and there is evidence for inactivation of the
Rb pathway in a proportion of TNBCs,
45,46
which leads to E2F3
overexpression. Furthermore, the E2F3 pathway signature is associ-
ated with response of TNBC to neoadjuvant chemotherapy using
drugs other than cisplatin.
40
It is unclear whether any of the biomar-
kers correlated with cisplatin response reported here represent specific
markers of cisplatin response; they may indicate general chemothera-
py responsiveness. However, in cell culture experiments and in a
retrospective clinical analysis, the p63/p73 pathway is related specifi-
cally to cisplatin response and not to response to a variety of other
chemotherapeutic agents.
25,26
We emphasize that patients in this neoadjuvant trial received
standard therapy after surgery, and the relatively low pCR rate to
single-agent cisplatin (21% for all patients, 15% excluding the BRCA1
mutation carriers) argues against administration of single-agent cis-
platin as adjuvant or neoadjuvant therapy for unselected TNBCs.
Multiagent neoadjuvant therapy has achieved higher pCR rates: 45%
to 24 weeks of paclitaxel/fluorouracil, doxorubicin, and cyclophosph-
amide (T/FAC),
4
and 24% to doxorubicin and cyclophosphamide
(AC) often followed by a taxane.
3
In this study, we could not assess
whether the tumors that responded to cisplatin were the same as or
different from tumors that would have responded to established mul-
tiagent regimens. We believe that these results justify exploring thera-
peutic combinations including platinum agents in TNBC and invite
additional efforts at discovering biomarkers predictive of response.
AUTHORS’ DISCLOSURES OF POTENTIAL CONFLICTS
OF INTEREST
Although all authors completed the disclosure declaration, the following
author(s) indicated a financial or other interest that is relevant to the subject
matter under consideration in this article. Certain relationships marked
with a “U” are those for which no compensation was received; those
relationships marked with a “C” were compensated. For a detailed
description of the disclosure categories, or for more information about
ASCO’s conflict of interest policy, please refer to the Author Disclosure
Declaration and the Disclosures of Potential Conflicts of Interest section in
Information for Contributors.
Employment or Leadership Position: None Consultant or Advisory
Role: None Stock Ownership: None Honoraria: None Research
Funding: Judy E. Garber, AstraZeneca Expert Testimony: None Other
Remuneration: None
Neoadjuvant Cisplatin in Triple-Negative Breast Cancer
www.jco.org © 2010 by American Society of Clinical Oncology 1151
AUTHOR CONTRIBUTIONS
Conception and design: Daniel P. Silver, Andrea L. Richardson, Rebecca
S. Gelman, Paula D. Ryan, Shridar Ganesan, Leif W. Ellisen, Eric P.
Winer, Judy E. Garber
Financial support: Daniel P. Silver, Andrea L. Richardson, Leif W.
Ellisen, Eric P. Winer, Judy E. Garber
Administrative support: Daniel P. Silver, Andrea L. Richardson, Eric P.
Winer, Judy E. Garber
Provision of study materials or patients: Daniel P. Silver, Andrea L.
Richardson, Diana Calogrias, Paula D. Ryan, Nadine M. Tung, Dennis C.
Sgroi, Eric P. Winer, Judy E. Garber
Collection and assembly of data: Daniel P. Silver, Andrea L. Richardson,
Zhigang C. Wang, Chee-Onn Leong, Diana Calogrias, Ayodele
Buraimoh, Aquila Fatima, Arcangela De Nicolo, Shridar Ganesan,
Alexander Miron, Christian Colin, Dennis C. Sgroi, Leif W. Ellisen, Eric
P. Winer, Judy E. Garber
Data analysis and interpretation: Daniel P. Silver, Andrea L.
Richardson, Aron C. Eklund, Zhigang C. Wang, Zoltan Szallasi, Qiyuan
Li, Nicolai Juul, Chee-Onn Leong, Rebecca S. Gelman, Arcangela De
Nicolo, Shridar Ganesan, Alexander Miron, Christian Colin, Dennis C.
Sgroi, Leif W. Ellisen, Eric P. Winer, Judy E. Garber
Manuscript writing: Daniel P. Silver, Andrea L. Richardson, Zoltan
Szallasi, Rebecca S. Gelman, Nadine M. Tung, Christian Colin, Eric P.
Winer, Judy E. Garber
Final approval of manuscript: Daniel P. Silver, Andrea L. Richardson,
Aron C. Eklund, Zhigang C. Wang, Zoltan Szallasi, Qiyuan Li, Nicolai
Juul, Chee-Onn Leong, Diana Calogrias, Ayodele Buraimoh, Aquila
Fatima, Rebecca S. Gelman, Paula D. Ryan, Nadine M. Tung, Arcangela
De Nicolo, Shridar Ganesan, Alexander Miron, Christian Colin, Dennis
C. Sgroi, Leif W. Ellisen, Eric P. Winer, Judy E. Garber
REFERENCES
1. Liedtke C, Mazouni C, Hess KR, et al: Re-
sponse to neoadjuvant therapy and long-term sur-
vival in patients with triple-negative breast cancer.
J Clin Oncol 26:1275-1281, 2008
2. Berry DA, Cirrincione C, Henderson IC, et al:
Estrogen-receptor status and outcomes of modern
chemotherapy for patients with node-positive breast
cancer. JAMA 295:1658-1667, 2006
3. Carey LA, Dees EC, Sawyer L, et al: The triple
negative paradox: Primary tumor chemosensitivity of
breast cancer subtypes. Clin Cancer Res 13:2329-
2334, 2007
4. Rouzier R, Perou CM, Symmans WF, et al:
Breast cancer molecular subtypes respond differ-
ently to preoperative chemotherapy. Clin Cancer
Res 11:5678-5685, 2005
5. Foulkes WD, Stefansson IM, Chappuis PO, et
al: Germline BRCA1 mutations and a basal epithelial
phenotype in breast cancer. J Natl Cancer Inst
95:1482-1485, 2003
6. Wang ZC, Lin M, Wei LJ, et al: Loss of
heterozygosity and its correlation with expression
profiles in subclasses of invasive breast cancers.
Cancer Res 64:64-71, 2004
7. Eisinger F, Jacquemier J, Charpin C, et al:
Mutations at BRCA1: The medullary breast carci-
noma revisited. Cancer Res 58:1588-1592, 1998
8. Marcus JN, Watson P, Page DL, et al: Hered-
itary breast cancer: Pathobiology, prognosis, and
BRCA1 and BRCA2 gene linkage. Cancer 77:697-
709, 1996
9. Karp SE, Tonin PN, Begin LR, et al: Influence
of BRCA1 mutations on nuclear grade and estrogen
receptor status of breast carcinoma in Ashkenazi
Jewish women. Cancer 80:435-441, 1997
10. Verhoog LC, Brekelmans CT, Seynaeve C, et
al: Survival and tumour characteristics of breast-
cancer patients with germline mutations of BRCA1.
Lancet 351:316-321, 1998
11. Silver DP: HIN-1 and the nosology of breast
cancer. Cancer Biol Ther 2:564-565, 2003
12. Nielsen TO, Hsu FD, Jensen K, et al: Immu-
nohistochemical and clinical characterization of the
basal-like subtype of invasive breast carcinoma. Clin
Cancer Res 10:5367-5374, 2004
13. Lakhani SR, Reis-Filho JS, Fulford L, et al:
Prediction of BRCA1 status in patients with breast
cancer using estrogen receptor and basal pheno-
type. Clin Cancer Res 11:5175-5180, 2005
14. Feki A, Irminger-Finger I: Mutational spectrum
of p53 mutations in primary breast and ovarian
tumors. Crit Rev Oncol Hematol 52:103-116, 2004
15. Richardson AL, Wang ZC, De Nicolo A, et al: X
chromosomal abnormalities in basal-like human
breast cancer. Cancer Cell 9:121-132, 2006
16. Perou CM, Sorlie T, Eisen MB, et al: Molecular
portraits of human breast tumours. Nature 406:747-
752, 2000
17. Sorlie T, Tibshirani R, Parker J, et al: Repeated
observation of breast tumor subtypes in indepen-
dent gene expression data sets. Proc Natl Acad Sci
U S A 100:8418-8423, 2003
18. Turner N, Tutt A, Ashworth A: Hallmarks of
‘BRCAness’ in sporadic cancers. Nat Rev Cancer
4:814-819, 2004
19. Scully R, Ganesan S, Vlasakova K, et al: Ge-
netic analysis of BRCA1 function in a defined tumor
cell line. Mol Cell 4:1093-1099, 1999
20. Tassone P, Tagliaferri P, Perricelli A, et al:
BRCA1 expression modulates chemosensitivity of
BRCA1-defective HCC1937 human breast cancer
cells. Br J Cancer 88:1285-1291, 2003
21. Rottenberg S, Nygren AO, Pajic M, et al:
Selective induction of chemotherapy resistance of
mammary tumors in a conditional mouse model for
hereditary breast cancer. Proc Natl Acad SciUSA
104:12117-12122, 2007
21a. Ogston KN, Miller ID, Payne S, et al: A new
histological grading system to assess response of
breast cancers to primary chemotherapy: Prognostic
significance and survival. Breast (Edinburgh, Scot-
land) 12:320-327, 2003
22. Kreike B, van Kouwenhove M, Horlings H, et
al: Gene expression profiling and histopathological
characterization of triple-negative/basal-like breast
carcinomas. Breast Cancer Res 9:R65, 2007
23. Turner NC, Reis-Filho JS, Russell AM, et al:
BRCA1 dysfunction in sporadic basal-like breast
cancer. Oncogene 26:2126-2132, 2007
24. DeYoung MP, Johannessen CM, Leong CO,
et al: Tumor-specific p73 up-regulation mediates
p63 dependence in squamous cell carcinoma. Can-
cer Res 66:9362-9368, 2006
25. Leong CO, Vidnovic N, DeYoung MP, et al:
The p63/p73 network mediates chemosensitivity to
cisplatin in a biologically defined subset of primary
breast cancers. J Clin Invest 117:1370-1380, 2007
26. Rocco JW, Leong CO, Kuperwasser N, et al:
p63 mediates survival in squamous cell carcinoma
by suppression of p73-dependent apoptosis. Cancer
Cell 9:45-56, 2006
27. Bild AH, Yao G, Chang JT, et al: Oncogenic
pathway signatures in human cancers as a guide to
targeted therapies. Nature 439:353-357, 2006
28. Whitfield ML, Sherlock G, Saldanha AJ, et al:
Identification of genes periodically expressed in the
human cell cycle and their expression in tumors.
Mol Biol Cell 13:1977-2000, 2002
29. Carter SL, Eklund AC, Kohane IS, et al: A
signature of chromosomal instability inferred from
gene expression profiles predicts clinical outcome in
multiple human cancers. Nat Genet 38:1043-1048,
2006
30. Chang HY, Sneddon JB, Alizadeh AA, et al:
Gene expression signature of fibroblast serum re-
sponse predicts human cancer progression: Similar-
ities between tumors and wounds. PLoS Biol 2:E7,
2004
31. Teschendorff AE, Caldas C: A robust classifier
of high predictive value to identify good prognosis
patients in ER-negative breast cancer. Breast Can-
cer Res 10:R73, 2008
32. Byrski T, Huzarski T, Dent R, et al: Response
to neoadjuvant therapy with cisplatin in BRCA1-
positive breast cancer patients. Breast Cancer Res
Treat, 115:359-363, 2009
33. Hayes DF, Thor AD, Dressler LG, et al: HER2
and response to paclitaxel in node-positive breast
cancer. N Engl J Med 357:1496-1506, 2007
34. Hess KR, Anderson K, Symmans WF, et al:
Pharmacogenomic predictor of sensitivity to preop-
erative chemotherapy with paclitaxel and fluoroura-
cil, doxorubicin, and cyclophosphamide in breast
cancer. J Clin Oncol 24:4236-4244, 2006
35. Quinn JE, James CR, Stewart GE, et al:
BRCA1 mRNA expression levels predict for overall
survival in ovarian cancer after chemotherapy. Clin
Cancer Res 13:7413-7420, 2007
36. Lowe SW, Ruley HE, Jacks T, et al: p53-
dependent apoptosis modulates the cytotoxicity of
anticancer agents. Cell 74:957-967, 1993
37. Di Leo A, Tanner M, Desmedt C, et al: p-53
gene mutations as a predictive marker in a popula-
tion of advanced breast cancer patients randomly
treated with doxorubicin or docetaxel in the context
of a phase III clinical trial. Ann Oncol 18:997-1003,
2007
38. Aas T, Børresen AL, Geisler S, et al: Specific
P53 mutations are associated with de novo resis-
tance to doxorubicin in breast cancer patients. Nat
Med 2:811-814, 1996
39. Bertheau P, Turpin E, Rickman DS, et al: Exquis-
ite sensitivity of TP53 mutant and basal breast cancers
to a dose-dense epirubicin-cyclophosphamide regi-
men. PLoS Med 4:e90, 2007
Silver et al
1152 © 2010 by American Society of Clinical Oncology JOURNAL OF CLINICAL ONCOLOGY
40. Tordai A, Wang J, Andre F, et al: Evaluation of
biological pathways involved in chemotherapy response
in breast cancer. Breast Cancer Res 10:R37, 2008
41. Holstege H, Joosse SA, van Oostrom CT, et
al: High incidence of protein-truncating TP53 muta-
tions in BRCA1-related breast cancer. Cancer Res
69:3625-3633, 2009
42. Rocca A, Viale G, Gelber RD, et al: Pathologic
complete remission rate after cisplatin-based pri-
mary chemotherapy in breast cancer: Correlation
with p63 expression. Cancer Chemother Pharmacol
61:965-971, 2008
43. Olaussen KA, Dunant A, Fouret P, et al: DNA
repair by ERCC1 in non-small-cell lung cancer and
cisplatin-based adjuvant chemotherapy. N Engl
J Med 355:983-991, 2006
44. Ivanov O, Chen F, Wiley EL, et al: alphaB-
crystallin is a novel predictor of resistance to neoad-
juvant chemotherapy in breast cancer. Breast
Cancer Res Treat 111:411-417, 2008
45. Gauthier ML, Berman HK, Miller C, et al: Abro-
gated response to cellular stress identifies DCIS asso-
ciated with subsequent tumor events and defines
basal-like breast tumors. Cancer Cell 12:479-491, 2007
46. Herschkowitz JI, He X, Fan C, et al: The
functional loss of the retinoblastoma tumour sup-
pressor is a common event in basal-like and luminal
B breast carcinomas. Breast Cancer Res 10:R75,
2008
47. Sørlie T, Perou CM, Tibshirani R, et al: Gene
expression patterns of breast carcinomas distin-
guish tumor subclasses with clinical implications.
Proc Natl Acad SciUSA98:10869-10874, 2001
48. Schmittgen TD, Livak KJ: Analyzing real-time
PCR data by the comparative C(T) method. Nat
Protoc 3:1101-1108, 2008
■■■
Glossary Terms
Promoter methylation: Methylation of DNA sequences
within the promoters of genes; this often occurs on the cytosine
residue of CpG dinucleotides and is correlated with decreased
expression of the adjacent gene.
BRCA1 expression: A tumor suppressor gene, the breast
cancer 1 susceptibility gene is known to play a role in repairing
DNA breaks. Mutations in this gene are associated with increased
risks of developing breast or ovarian cancer.
Hierarchical cluster: An analytical tool used to find the clos-
est associations among gene profiles and specimens under evaluation.
PCR (polymerase chain reaction): PCR is a method that
allows exponential amplification of short DNA sequences within a
longer DNA molecule.
Double-strand break repair: Any of several DNA repair pro-
cesses used by organisms to repair breaks in DNA that span both
strands of DNA at a single location.
BRCA1: A tumor suppressor gene that prevents ovarian and
breast cancer.
Gene signature: The coordinated response of many genes to a particu-
lar stimulus; for example, the “myc oncogene signature” is the response of
many genes to the forced overexpression of the myc oncogene.
Intrinsic genes: A set of genes whose level of expression is used to sort
breast cancers into subtypes.
Methylation-specific PCR: A molecular assay that detects methyl-
ation of a particular stretch of DNA.
Gene array: A microchip on which DNA sequences for many genes
are embedded; used to measure gene expression of many genes
simultaneously.
BRCA-ness: A term applied to breast cancers, referring to the
degree of relationship of a given breast cancer to one deficient in the
BRCA1 gene.
Neoadjuvant Cisplatin in Triple-Negative Breast Cancer
www.jco.org © 2010 by American Society of Clinical Oncology 1153
... The HR repair pathway corrects DNA double-strand breaks created by lesions such as platinum-induced interstrand crosslinks. Alterations in HR genes are associated with increased platinum sensitivity in multiple tumor types including breast and ovarian cancer 2,3 . HR deficiency is likely to be present in gastric and esophageal adenocarcinoma (GEA) since mutations in key HR genes, albeit with low frequency, have been detected, for example, in gastric adenocarcinoma 4,5 . ...
... (1) As opposed to WGS, the limitations of using WES samples are fewer available variants for the mutational signature extraction part, decreasing the reliability of the extracted features. (2) The extracted mutational signatures are limited to those previously described within TCGA-STAD, TCGA-ESCA, TCGA-COAD, and TCGA-READ cohorts, which raises the possibility of missing novel signatures. (3) The extracted mutational signatures and genomic scar scores only inform about the historical state of the tumors, and do not engender information about possible acquired drug resistance mechanisms. ...
Article
Full-text available
Homologous recombination (HR) and nucleotide excision repair (NER) are the two most frequently disabled DNA repair pathways in cancer. HR-deficient breast, ovarian, pancreatic and prostate cancers respond well to platinum chemotherapy and PARP inhibitors. However, the frequency of HR deficiency in gastric and esophageal adenocarcinoma (GEA) still lacks diagnostic and functional validation. Using whole exome and genome sequencing data, we found that a significant subset of GEA, but very few colorectal adenocarcinomas, show evidence of HR deficiency by mutational signature analysis (HRD score). High HRD gastric cancer cell lines demonstrated functional HR deficiency by RAD51 foci assay and increased sensitivity to platinum chemotherapy and PARP inhibitors. Of clinical relevance, analysis of three different GEA patient cohorts demonstrated that platinum treated HR deficient cancers had better outcomes. A gastric cancer cell line with strong sensitivity to cisplatin showed HR proficiency but exhibited NER deficiency by two photoproduct repair assays. Single-cell RNA-sequencing revealed that, in addition to inducing apoptosis, cisplatin treatment triggered ferroptosis in a NER-deficient gastric cancer, validated by intracellular GSH assay. Overall, our study provides preclinical evidence that a subset of GEAs harbor genomic features of HR and NER deficiency and may therefore benefit from platinum chemotherapy and PARP inhibitors.
... For the analysis of clinical data, previously published datasets were obtained from ArrayExpress. Microarray data from pre-neoadjuvant trial of cisplatin monotherapy in TNBC tumors were obtained from GEO ID: GSE18864 34 Table S4. GEO2R function of the NCBI-GEO database was used for identifying differentially expressed genes of the clinical datasets. ...
... Previous studies have shown that ZMYND8 levels are significantly lower in aggressive breast cancers, such as basal subtype tumors and higher levels in luminal breast cancers 42 . However, ZMYND8 expression was found to be significantly elevated in TNBC patients with a good pathological response (Miller-Payne score 4 and 5) as compared to reference patient cohort 34 (Fig. 2a). Furthermore, post neoadjuvant chemotherapy treatment, ZMYND8 expression was higher in non-relapsed TNBC patients, compared to relapsed (Fig. 2b), indicating that ZMYND8 expression is higher in chemo-sensitive tumors compared to chemo-resistant tumors. ...
Article
Full-text available
The major challenge in chemotherapy lies in the gain of therapeutic resistance properties of cancer cells. The relatively small fraction of chemo-resistant cancer cells outgrows and are responsible for tumor relapse, with acquired invasiveness and stemness. We demonstrate that zinc-finger MYND type-8 (ZMYND8), a putative chromatin reader, suppresses stemness, drug resistance, and tumor-promoting genes, which are hallmarks of cancer. Reinstating ZMYND8 suppresses chemotherapeutic drug doxorubicin-induced tumorigenic potential (at a sublethal dose) and drug resistance, thereby resetting the transcriptional program of cells to the epithelial state. The ability of ZMYND8 to chemo-sensitize doxorubicin-treated metastatic breast cancer cells by downregulating tumor-associated genes was further confirmed by transcriptome analysis. Interestingly, we observed that ZMYND8 overexpression in doxorubicin-treated cells stimulated those involved in a good prognosis in breast cancer. Consistently, sensitizing the cancer cells with ZMYND8 followed by doxorubicin treatment led to tumor regression in vivo and revert back the phenotypes associated with drug resistance and stemness. Intriguingly, ZMYND8 modulates the bivalent or poised oncogenes through its association with KDM5C and EZH2, thereby chemo-sensitizing the cells to chemotherapy for better disease-free survival. Collectively, our findings indicate that poised chromatin is instrumental for the acquisition of chemo-resistance by cancer cells and propose ZMYND8 as a suitable epigenetic tool that can re-sensitize the chemo-refractory breast carcinoma.
Article
The advancement of renal replacement therapy has significantly enhanced the survival rates of patients with end-stage renal disease (ESRD) over time. However, this prolonged survival has also been associated with a higher likelihood of cancer diagnoses among these patients including breast cancer. Breast cancer treatment typically involves surgery, radiation, and systemic therapies, with approaches tailored to cancer type, stage, and patient preferences. However, renal replacement therapy complicates systemic therapy due to altered drug clearance and the necessity for dialysis sessions. This review emphasizes the need for optimized dosing and administration strategies for systemic breast cancer treatments in dialysis patients, aiming to ensure both efficacy and safety. Additionally, challenges in breast cancer screening and diagnosis in this population, including soft-tissue calcifications, are highlighted.
Article
One of the major issues affecting worldwide public health is cancer. According to the related occurrence and morbidity statistics, it is becoming more common in both economically developed countries and developing countries. Several diagnostics procedures and early treatment methods are essential in order to reduce the incidence rate of breast cancer. In this article, we introduce several reported strategies of treatment based on the tumor progression and breast cancer (BC) molecular subtypes in order to offer the most personalized treatment for BC patients. link: https://dusj.journals.ekb.eg/article_354516_214ec4381367c8acd0e17a4b79e6da58.pdf
Article
Rationale. BRCA1 associated triple-negative breast cancer (TNBC) is one of the most aggressive subtypes of breast cancer. At the same time, carcinomas that develop in carriers of BRCA1 mutations are characterized by extremely high sensitivity to DNA-damaging chemotherapy. Mitomycin C alone or in combination with platinum agents has already demonstrated promising results in the treatment of BRCA-associated ovarian cancer (OC) and metastatic breast cancer. In this article, we present the results of a retrospective study aimed at comparing standard neoadjuvant chemotherapy regimens (NACT) with mitomycin-based regimens for primary locally advanced BRCA1-associated TNBC. The aim of the study is to determine the effectiveness of the combination of mitomycin and platinum compounds during neoadjuvant therapy in patients with primary locally advanced BRCA1 – associated TNBC. Materials and methods . The study included 89 patients diagnosed with primary locally advanced BRCA1-associated TNBC. Patients were divided into three groups depending on the therapy: 1) 4 cycles of anthracycline and cyclophosphamide followed by 12 weekly injections of paclitaxel (n = 48) (AC + T), 2) 4 cycles of anthracycline and cyclophosphamide followed by 12 weekly injections of paclitaxel and carboplatin (n = 27) (AC + TCbP), 3) mitomycin C plus platinum followed by 12 weekly injections of paclitaxel (n = 14) (MR + T). Pathological complete response (pCR) rates were compared. Results. The pCR rate in the MP+T group was 10/14 (71%). In patients with BRCA1-associated breast cancer who received AC + T and AC + TCbP regimens as NACT, the pCR rate was 17/48 (35%) and 19/27 (70%), respectively. The difference in pCR rate between mitomycin-containing therapy and the standard AC + T regimen was statistically significant (p = 0.03); the frequency of regressions was comparable to the frequency in the AC + TCbP group. During the 20-month follow-up period, no relapses were observed in the MR + T group. Relapses were more frequent in the AC + T group compared with the AC + TCbP group (16/48 (33%) vs 1/27 (4%), p = 0.003, Fisher’s exact test). The toxicity profile of the mitomycin-containing regimen included hematologic adverse events, the most common of which were anemia and leukopenia. Compared to standard regimens, nausea was significantly less pronounced. No patients reported alopecia with this regimen. Conclusions. The addition of mitomycin C to neoadjuvant therapy for BRCA1-associated TNBC may be a promising treatment option for this category of patients and merits further study.
Article
Full-text available
Over the past couple of decades, the incidence of breast cancer (BC) has significantly increased among females in comparison to other cancer types. In medicinal terminology, the susceptibility to BC is mainly centered around three hormonal receptors: estrogen (ER), progesterone (PR), and human epidermal growth receptor (HER2). Notably, estrogen- dependent breast cancer has a considerable female demographic, making it treatable with hormonal drugs and less intensive immunotherapy. Conversely, the narrative delves into the ominous type of cancer known as triple-negative breast cancer (TNBC). The orientation of all three receptors falls in a negative direction, which is ineffective for treatments that rely on hormonal or antagonist medicaments. Therefore, the only option available to tackle this type of cancer is chemotherapy, which causes toxicity within the body, is highly expensive, and is non-targeted. To counter this challenge, researchers have pioneered nano-based drug delivery systems (NDDS) owing to their innumerable merits and scientific development. NDDS mainly involves polymeric nanoparticles, liposomes, and dendrimers. This review comprehensively details the advancements in nanoinduced targeted drug delivery systems, with a focus on surface modification techniques for active targeting, enhanced drug release, and improved pharmacokinetics. Critical analysis extends to preclinical and clinical studies, revealing the potential of nano-drug delivery systems in TNBC to surpass traditional therapies, with promising heightened efficacy and reduced side effects. Graphical Abstract
Article
Precise and personalized drug application is crucial in the clinical treatment of complex diseases. Although neural networks offer a new approach to improving drug strategies, their internal structure is difficult to interpret. Here, we propose PBAC (Pathway‐Based Attention Convolution neural network), which integrates a deep learning framework and attention mechanism to address the complex biological pathway information, thereby provide a biology function‐based robust drug responsiveness prediction model. PBAC has four layers: gene‐pathway layer, attention layer, convolution layer and fully connected layer. PBAC improves the performance of predicting drug responsiveness by focusing on important pathways, helping us understand the mechanism of drug action in diseases. We validated the PBAC model using data from four chemotherapy drugs (Bortezomib, Cisplatin, Docetaxel and Paclitaxel) and 11 immunotherapy datasets. In the majority of datasets, PBAC exhibits superior performance compared to traditional machine learning methods and other research approaches (area under curve = 0.81, the area under the precision‐recall curve = 0.73). Using PBAC attention layer output, we identified some pathways as potential core cancer regulators, providing good interpretability for drug treatment prediction. In summary, we presented PBAC, a powerful tool to predict drug responsiveness based on the biology pathway information and explore the potential cancer‐driving pathways.
Preprint
Full-text available
Background Adding platinum to anthracycline- and taxane-based neoadjuvant chemotherapy has improved pathological complete response (pCR) and event-free survival(EFS) in patients with triple-negative breast cancer (TNBC). However, the efficacy for TNBC of combining taxane and platinum without anthracycline remains controversial. Methods The HELEN-001 trial was a randomized, phase 2 controlled, and open-label investigation carried out in China at 6 hospitals. Participants who were aged 18–70 years old, were histologically confirmed for TNBC clinical stage II–III, suitable for potentially curative surgery, and had an Eastern Cooperative Oncology Group performance status (ECOG-PS) of 0 or 1 were selected for this trial. Participants were randomized into two equal groups; those who received docetaxel plus cisplatin (75 mg/m², respectively) and those who received docetaxel plus doxorubicin and cyclophosphamide (docetaxel 75 mg/m², doxorubicin 50 mg/m², and cyclophosphamide 500 mg/m²). These regimens were given every 3 weeks for 6 cycles. Randomization was stratified by tumor size and nodal status. The primary endpoint was the number of individuals achieving a pCR (ypT0/isN0). The trial was registered with chictr.org (number ChiCTR-1800019501). Findings Between November, 2018, and June, 2022, 212 patients were selected (n = 106/treatment arm). The number of individuals who achieved pCR after docetaxel plus cisplatin treatment was 51.9%, and that of those who attained pCR after docetaxel plus doxorubicin and cyclophosphamide was 35.8% (P = 0.019). After median follow-up of 29 months[interquartile range (IQR), 21 to 41], 14 of 106 patients (13.2%) in the docetaxel plus cisplatin group and 18 of 106 patients (17.0%) in the docetaxel plus doxorubicin and cyclophosphamide group had event-free survival (EFS) events [95% confidence interval (CI) = 0.377 to 1.526, hazard ratio (HR) = 0.759, P = 0.492]. The incidence of grade 3 or 4 events was similar in both groups [57 (54%) vs. 51 (48%)]. No treatment-associated deaths were identified in both groups. Interpretation In stage II to III TNBC, the docetaxel plus cisplatin regimen achieved higher pCR rates than docetaxel plus doxorubicin and cyclophosphamide, with a comparable toxicity profile. Consistent with literature, the taxane plus cisplatin regimen demonstrated a favorable risk-to-benefit profile and could serve as an optimal neoadjuvant chemotherapy option for patients with high-risk TNBC.
Article
Full-text available
Cancer invasion and metastasis have been likened to wound healing gone awry. Despite parallels in cellular behavior between cancer progression and wound healing, the molecular relationships between these two processes and their prognostic implications are unclear. In this study, based on gene expression profiles of fibroblasts from ten anatomic sites, we identify a stereotyped gene expression program in response to serum exposure that appears to reflect the multifaceted role of fibroblasts in wound healing. The genes comprising this fibroblast common serum response are coordinately regulated in many human tumors, allowing us to identify tumors with gene expression signatures suggestive of active wounds. Genes induced in the fibroblast serum-response program are expressed in tumors by the tumor cells themselves, by tumor-associated fibroblasts, or both. The molecular features that define this wound-like phenotype are evident at an early clinical stage, persist during treatment, and predict increased risk of metastasis and death in breast, lung, and gastric carcinomas. Thus, the transcriptional signature of the response of fibroblasts to serum provides a possible link between cancer progression and wound healing, as well as a powerful predictor of the clinical course in several common carcinomas.
Article
Full-text available
Ten patients with breast cancer and a breast cancer susceptibility gene 1 (BRCA1) mutation, who presented with stages I to III breast cancer between December 2006 and 2007, were treated with four cycles of neoadjuvant cisplatin, followed by mastectomy and conventional chemotherapy. The excised breast tissue and lymph nodes were examined for the presence of residual disease. Pathologic complete response was observed in nine patients (90%). Platinum-based chemotherapy appears to be effective in a high proportion of patients with BRCA1-associated breast cancers. Clinical trials are now warranted to determine the optimum treatment for this subgroup of breast cancer patients.
Article
Full-text available
Approximately half of all hereditary breast cancers are compromised in their DNA repair mechanisms due to loss of BRCA1 or BRCA2 function. Previous research has found a strong correlation between BRCA mutation and TP53 mutation. However, TP53 mutation status is often indirectly assessed by immunohistochemical staining of accumulated p53 protein. We sequenced TP53 exons 2 to 9 in 21 BRCA1-related breast cancers and 37 sporadic breast tumors. Strikingly, all BRCA1-related breast tumors contained TP53 mutations, whereas only half of these tumors stained positive for p53 accumulation. Positive p53 staining correlates with the presence of TP53 hotspot mutations in both BRCA1-related and sporadic breast tumors. However, whereas the majority of sporadic breast tumors that stained negative for p53 accumulation had wild-type TP53, the majority of BRCA1-associated breast tumors that stained negative for p53 accumulation had protein-truncating TP53 mutations (nonsense, frameshift, and splice mutations). Therefore, the strong selection for p53 loss in BRCA1-related tumors is achieved by an increase of protein-truncating TP53 mutations rather than hotspot mutations. Hence, immunohistochemical detection of TP53 mutation could lead to misdiagnosis in approximately half of all BRCA1-related tumors. The presence of deleterious TP53 mutations in most, if not all, BRCA1-related breast cancers suggests that p53 loss of function is essential for BRCA1-associated tumorigenesis. BRCA1-related tumors may therefore be treated not only with drugs that target BRCA1 deficiency [e.g., poly(ADP-ribose) polymerase inhibitors] but also with drugs that selectively target p53-deficient cells. This raises interesting possibilities for combination therapies against BRCA1-deficient breast cancers and BRCA1-like tumors with homologous recombination deficiency.
Article
Full-text available
Breast cancers can be classified using whole genome expression into distinct subtypes that show differences in prognosis. One of these groups, the basal-like subtype, is poorly differentiated, highly metastatic, genomically unstable, and contains specific genetic alterations such as the loss of tumour protein 53 (TP53). The loss of the retinoblastoma tumour suppressor encoded by the RB1 locus is a well-characterised occurrence in many tumour types; however, its role in breast cancer is less clear with many reports demonstrating a loss of heterozygosity that does not correlate with a loss of RB1 protein expression. We used gene expression analysis for tumour subtyping and polymorphic markers located at the RB1 locus to assess the frequency of loss of heterozygosity in 88 primary human breast carcinomas and their normal tissue genomic DNA samples. RB1 loss of heterozygosity was observed at an overall frequency of 39%, with a high frequency in basal-like (72%) and luminal B (62%) tumours. These tumours also concurrently showed low expression of RB1 mRNA. p16INK4a was highly expressed in basal-like tumours, presumably due to a previously reported feedback loop caused by RB1 loss. An RB1 loss of heterozygosity signature was developed and shown to be highly prognostic, and was potentially a predictive marker of response to neoadjuvant chemotherapy. These results suggest that the functional loss of RB1 is common in basal-like tumours, which may play a key role in dictating their aggressive biology and unique therapeutic responses.
Article
Gene expression array profiles identify subclasses of breast cancers with different clinical outcomes and different molecular features. The present study attempted to correlate genomic alterations (loss of heterozygosity; LOH) with subclasses of breast cancers having distinct gene expression signatures. Hierarchical clustering of expression array data from 89 invasive breast cancers identified four major expression subclasses. Thirty-four of these cases representative of the four subclasses were microdissected and allelotyped using genome-wide single nucleotide polymorphism detection arrays (Affymetrix, Inc.). LOH was determined by comparing tumor and normal single nucleotide polymorphism allelotypes. A newly developed statistical tool was used to determine the chromosomal regions of frequent LOH. We found that breast cancers were highly heterogeneous, with the proportion of LOH ranging widely from 0.3% to >60% of heterozygous markers. The most common sites of LOH were on 17p, 17q, 16q, 11q, and 14q, sites reported in previous LOH studies. Signature LOH events were discovered in certain expression subclasses. Unique regions of LOH on 5q and 4p marked a subclass of breast cancers with “basal-like” expression profiles, distinct from other subclasses. LOH on 1p and 16q occurred preferentially in a subclass of estrogen receptor-positive breast cancers. Finding unique LOH patterns in different groups of breast cancer, in part defined by expression signatures, adds confidence to newer schemes of molecular classification. Furthermore, exclusive association between biological subclasses and restricted LOH events provides rationale to search for targeted genes.
Article
A basal epithelial phenotype is found in not more than 15% of all invasive breast cancers. Microarray studies have shown that this phenotype is associated with breast cancers that express neither estrogen receptor (ER) nor erbB-2 (HER2/neu) (i.e., ER/erbB-2–negative tumors). The ER/erbB-2– negative phenotype is also found in breast cancers occurring in BRCA1 mutation carriers (i.e., BRCA1-related breast cancers). We tested the hypothesis that BRCA1-related breast cancers are more likely than non–BRCA1/ 2-related breast cancer to express a basal epithelial phenotype. Among 292 breast cancer specimens previously analyzed for ER, erbB-2, p53, and germline mutations in BRCA1 and BRCA2, we identified 76 that did not overexpress ER or erbB-2. Of the 72 specimens with sufficient material for testing, 40 expressed stratified epithelial cytokeratin 5 and/or 6 (5/6). In univariate analysis, the expression of cytokeratin 5/6 was statistically significantly associated with BRCA1-related breast cancers (odds ratio = 9.0, 95% confidence interval = 1.9 to 43; P = .002, two-sided Fisher’s exact test). Thus, germline BRCA1 mutations appear to be associated with a distinctive breast cancer phenotype.
Article
BACKGROUND The purpose of this investigation was to determine if there are pathobiologic differences between BRCA1-related and BRCA2-related hereditary breast cancer (HBC) and non-HBC.METHODS On the basis of linkage to chromosomes 17q or 13q and/or the presence of ovarian and male breast cancer, HBC families were classified as either “BRCA1-related” (26 families, 90 breast cancer pathology cases) or “Other” (26 families, 85 cases), in which most BRCA2 cases were likely to reside. Cases were compared with 187 predominantly non-HBC cases. Tumors were assessed for histologic type, grade, and ploidy and S-phase fraction by quantitative DNA flow cytometry. Clinical presentation and available follow-up data were obtained.RESULTSBRCA1-related and Other HBC patients each presented at lower stage (P = 0.003) and earlier age than non-HBC patients (mean, 42.8 years and 47.1 years vs. 62.9 years, P < 0.0001). Compared with non-HBC, invasive BRCA1-related HBC had a lower diploidy rate (13% vs. 35%; P = 0.002), lower mean aneuploid DNA index (1.53 vs. 1.73; P = 0.002), and strikingly higher proliferation rates (mitotic grade 3; odds ratio [OR] = 4.42; P = 0.001; aneuploid mean S-phase fraction 16.5% vs. 9.3%, P < 0.0001). Other HBC patients, including patients in two BRCA2-linked families, had more tubular-lobular group (TLG) carcinomas (OR = 2.56, P = 0.007). All trends were independent of age. A nonsignificant trend toward better crude survival in both HBC groups was age- and stage-dependent. Compared with Other HBC, BRCA1-related HBC patients had fewer recurrences (P = 0.013), a trend toward lower specific death rates, and fared no worse than breast cancer patients at large. Other HBC patients, despite neutral prognostic indicators, fared worse.CONCLUSIONSBRCA1-related HBCs are more frequently aneuploid and have higher tumor cell proliferation rates compared with Other HBC. Despite these adverse prognostic features, BRCA1-related HBC patients have paradoxically lower recurrence rates than Other HBC patients. The excess of TLG cancers in the “Other” HBC group may be associated with BRCA2 linkage. Cancer 1996; 697-709.
Article
All cases were reexamined by a single pathologist experienced in the histology of breast carcinoma (L.B). All pathologic examinations occurred before DNA was extracted, and therefore researchers were blinded to mutation status. Histopathologic typing was based on standard criteria.13 Grading of carcinoma, as an estimate of differentiation, was limited to the invasive portion of the tumor. The score was based on a nuclear grading system with cytologic evaluation of the structural features of tumor nuclei, by comparison with the nuclei of normal mammary epithelium. Nuclear assessment was based on criteria introduced by Black et al.,14 corresponding to the nuclear score of Elston and the nuclear grading scheme of Fisher et al., exclusive of mitotic activity.15, 16 Briefly, in Grade 1 tumors, nuclei were small, had regular outlines and uniform nuclear chromatin, and showed little variation in size. In Grade 2 tumors, the nuclei had an open vesicular pattern, visible nucleoli, and moderate variability in size and shape. Grade 3 tumors had nuclei that were vesicular, often with prominent nucleoli, and exhibited marked variation in size and shape. Axillary lymph node status was determined by conventional hematoxylin and eosin staining of surgical specimens. Nine women did not undergo axillary lymph node dissection.