ArticlePDF Available

Abstract and Figures

We used data obtained by Caris Life Sciences, to evaluate the benefits of tailoring treatments for a breast carcinoma cohort by using tumor molecular profiles to inform decisions. Data for 92 breast cancer patients from the commercial Caris Molecular Intelligence database was retrospectively divided into two groups, so that the first always followed treatment recommendations, whereas in the second group all patients received at least one drug after profiling that was predicted to lack benefit. The biomarker and drug associations were based on tests including fluorescent in situ hybridization and DNA sequencing, although immunohistochemistry was the main test used. Patients whose drugs matched those recommended according to their tumor profile had an average overall survival of 667 days, compared to 510 days for patients that did not (P=0.0316). In the matched treatment group, 26% of patients were deceased by the last time of monitoring, whereas this was 41% in the unmatched group (P=0.1257). We therefore confirm the ability of tumor molecular profiling to improve survival of breast cancer patients. Immunohistochemistry biomarkers for the androgen, estrogen and progesterone receptors were found to be prognostic for survival.
Content may be subject to copyright.
Oncotarget1
www.impactjournals.com/oncotarget
Molecular profiling of advanced breast cancer tumors is beneficial
in assisting clinical treatment plans
Philip Carter1, Costi Alifrangis2, Biancastella Cereser1, Pramodh Chandrasinghe1,3,
Lisa Del Bel Belluz1, Nina Moderau1, Fotini Poyia1, Lee S. Schwartzberg4, Neha
Tabassum1, Jinrui Wen1, Jonathan Krell1 and Justin Stebbing1
1Department of Surgery and Cancer, Imperial College, London, UK
2Department of Oncology, University College Hospital, London, UK
3Department of Surgery, University of Kelaniya, Kelaniya, Sri Lanka
4WEST Cancer Center, The University of Tennessee, Memphis, USA
Correspondence to: Philip Carter, email: phil.carter@imperial.ac.uk
Keywords: tumor profiling; breast cancer; cancer treatment
Received: July 29, 2017 Accepted: October 28, 2017 Published: February 24, 2018
Copyright: Carter et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License 3.0
(CC BY 3.0), which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source
are credited.
ABSTRACT
We used data obtained by Caris Life Sciences, to evaluate the benefits of tailoring
treatments for a breast carcinoma cohort by using tumor molecular profiles to inform
decisions. Data for 92 breast cancer patients from the commercial Caris Molecular
Intelligence database was retrospectively divided into two groups, so that the first
always followed treatment recommendations, whereas in the second group all
patients received at least one drug after profiling that was predicted to lack benefit.
The biomarker and drug associations were based on tests including fluorescent in
situ hybridization and DNA sequencing, although immunohistochemistry was the main
test used.
Patients whose drugs matched those recommended according to their tumor
profile had an average overall survival of 667 days, compared to 510 days for patients
that did not (P=0.0316). In the matched treatment group, 26% of patients were
deceased by the last time of monitoring, whereas this was 41% in the unmatched
group (P=0.1257). We therefore confirm the ability of tumor molecular profiling to
improve survival of breast cancer patients. Immunohistochemistry biomarkers for
the androgen, estrogen and progesterone receptors were found to be prognostic for
survival.
INTRODUCTION
Breast cancer is the most prevalent form of cancer
in women, causing approximately one in four of all cases
worldwide. In 2012, there were 1.68 million diagnoses and
522,000 deaths according to the World Health Organization,
and around 80% of cases occur within patients over the age
of 50. Risk factors include obesity, lack of exercise, alcohol
consumption, age, family history and age at menarche. The
long-term outcome for patients depends on the stage of the
tumor and its characteristics at diagnosis.
Established evidence-based treatments for advanced
disease includes radiation, chemotherapy, hormonal
therapy, and targeted therapies. Due to the application
of these treatments, in the developed world survival is
relatively high, with between 80% and 90% of those in
England and the USA surviving for at least five years.
Hereditary genetic factors are thought to play
a minor role in sporadic breast carcinoma, but in
approximately 5% of cases it is significant. Germline
mutations in the genes BRCA1, BRCA2, p53, PTEN,
STK11, CHEK2, ATM, BRIP1 and PALB2 are all
considered important in breast cancer tumorigenesis.
The genetics of sporadic breast cancer is now better
understood, due to genomic sequencing of many such
tumors [1]. Somatic driver variants and the mutational
www.impactjournals.com/oncotarget/ Oncotarget, Advance Publications 2018
Oncotarget2
www.impactjournals.com/oncotarget
processes underlying them have now been identified [2],
and the sequencing of 560 breast cancer genomes [3] has
furthered progression towards a complete description of
the molecular events that cause these tumors. In total, 93
protein-coding cancer genes were found to have probable
driver mutations. This and other data including exon
sequencing [4], whole genome [5], transcriptional [6], and
methylation-based studies [7] have been used to develop a
molecular taxonomy of breast cancer.
Molecularly defined characteristics have been
used as predictive and prognostic biomarkers in breast
cancer to define therapeutic approaches. The earliest such
example is the identification of the estrogen receptor
(ER) overexpression in a subset of breast carcinomas in
the 1970s, and its subsequent targeting with ER directed
therapies [8]. Similarly HER2 (human epidermal growth
factor receptor 2) overexpression and its targeting with
Herceptin [9] have further defined a subset of this disease
that behaves and responds uniquely to HER2-directed
therapies. Gene chip technologies that use gene expression
profiling of the primary tumor such as OncotypeDX, have
been FDA approved as a decision aid in early breast cancer
to help define prognostic features [10]. Several preclinical
studies have identified drug-genome interactions [11].
These have been borne through with great successes in
specific situations, such as the EML4-ALK translocation in
non-small cell carcinoma of the lung. These specific tissue
and gene scenarios have been validated in prospective
clinical studies, and have transformed clinical practice [12].
An approach that has gained traction in recent years
is the application of molecular characterization beyond
established immunohistochemistry (IHC) biomarkers.
This has been used to guide therapeutic decision making
across many tissue types, after failure of standard
therapies. Genomic sequencing of the cancer [13] enables
identification of somatic driver variants, which have
Table 1: A summary of patient information comparing the matched and unmatched groups against all patients
overall
Patient & Tumor Information
Group Age Ethnicity Histology Grade Stage Survival (days) Mortality
All patients
(92) 57
White: 71
Black/African
American: 13
Asian: 4
Hawaiian/Pacific
Islander: 2
American Indian/
Alaskan Native: 1
Other/Unknown: 1
Infiltrating duct carcinoma, NOS: 50
Infiltrating ductular carcinoma: 11
Carcinoma, NOS: 10
Lobular carcinoma, NOS: 5
Adenocarcinoma, NOS: 4
Infiltrating lobular carcinoma, NOS: 3
Metaplastic carcinoma, NOS: 2
Ductal carcinoma, NOS: 2
Intraductal papillary adenocarcinoma with
invasion: 1
Infiltrating duct and lobular carcinoma: 1
Infiltrating duct mixed with other types of
carcinoma, in situ: 1
Intraductal papillary-mucinous carcinoma,
invasive: 1
Infiltrating lobular mixed with other types of
carcinoma: 1
Grade 3/
Poorly
differentiated:
41 (45%)
Grade 2 /
Moderately
differentiated:
44 (48%)
Grade 1 / Well
differentiated:
2 (2%)
Unknown / Not
determined: 4
(4%)
None / Not
applicable: 1
(1%)
IV: 18 (19%)
III no IIIC: 21 (23%)
IIIC: 9 (10%)
II: 31 (34%)
I: 10 (11%)
Unknown: 3 (3%)
583 34%
Matched
only (43) 55.8
White: 34
Asian: 3
Black/African
American: 3
American Indian/
Alaskan Native: 1
Other/Unknown: 1
Hawaiian/Pacific
Islander: 1
Infiltrating duct carcinoma, NOS: 21
Carcinoma, NOS: 8
Infiltrating ductular carcinoma: 5
Adenocarcinoma, NOS: 2
Infiltrating lobular carcinoma, NOS: 2
Lobular carcinoma, NOS: 2
Ductal carcinoma, NOS: 1
Metaplastic carcinoma, NOS: 1
Infiltrating duct and lobular carcinoma: 1
Grade 3/
Poorly
differentiated:
15 (35%)
Grade 2 /
Moderately
differentiated:
28 (65%)
II: 13 (30%)
III no IIIC: 10 (23%)
IV: 10 (23%)
IIIC: 4 (10%)
I: 3 (7%)
Unknown: 3 (7%)
667 26%
Unmatched
(49) 58.1
White: 37
Black/African American:
10
Asian: 1
Hawaiian/Pacific
Islander: 1
Infiltrating duct carcinoma, NOS: 29
Infiltrating ductular carcinoma: 6
Lobular carcinoma, NOS: 3
Adenocarcinoma, NOS: 2
Carcinoma, NOS: 2
Ductal carcinoma, NOS: 1
Infiltrating lobular carcinoma, NOS: 1
Infiltrating duct mixed with other types of
carcinoma, in situ: 1
Intraductal papillary adenocarcinoma with
invasion: 1
Metaplastic carcinoma, NOS: 1
Infiltrating lobular mixed with other types of
carcinoma: 1
Intraductal papillary-mucinous carcinoma,
invasive: 1
Grade 3/
Poorly
differentiated:
26 (53%)
Grade 2 /
Moderately
differentiated:
16 (33%)
Grade 1 / Well
differentiated:
2 (4%)
Unknown / Not
determined: 4
(8%)
None / Not
applicable: 1
(2%)
IV: 8 (16%)
III no IIIC: 11 (23%)
IIIC: 5 (10%)
II: 18 (37%)
I: 7 (14%)
510 41%
Oncotarget3
www.impactjournals.com/oncotarget
been associated with therapeutic outcomes in preclinical
or clinical studies [14]. The efficacy of this approach is
currently unclear across tumor types, as some mutations
are only known to be prognostic for response in particular
situations; some early attempts at matching therapies
failed for this reason, e.g. the use of BRAF inhibitors in
BRAF mutant colorectal cancer [15].
It has been shown that tumor profiling of non-
responsive breast cancer resulted in better clinical
treatments [14], while other studies have demonstrated
the benefit of profiling in other tumor types [16]. To
investigate the effectiveness of one such profiling method,
we evaluated data provided by Caris Life Sciences
from their CODE database (version 1.0). This resource
describes molecular profiling data that has been used to
recommend clinical treatments, and drug regimens used
before and after clinicians received this information along
with their outcomes. The impact of profiling on drug usage
and survival was evaluated here.
RESULTS
Patient characteristics
Data from the Caris CODE database of 92 advanced
stage breast cancer patients who underwent treatment was
analyzed. These patients were retrospectively divided
into two groups, based on their matching of treatments
to recommendations that had been generated according to
their profiles. In the matched treatment group, 43 patients
received at least one recommended drug after collection of
tumor sample for profiling and none that were predicted
to lack benefit, whereas in the unmatched treatment
group 49 patients were given one or more drugs that were
classified as having a lack of benefit at any time following
profiling. Information about the patients in both groups is
summarized in Table 1 (age, ethnicity, histology, tumor
grade and stage, and survival information).
Treatment analysis
Do patients whose treatments consistently follow
profile-based recommendations fare better than patients
whose treatments that do not? To compare the overall
survival of the two groups that will be referred to as
matched and unmatched, waterfall plots for both are shown
in Figure 1, where each bar represents a treatment schedule
for a breast cancer patient. The 92 bars shown denote 43
matched and 49 unmatched patients (on the left and right
respectively). Each set is ordered by survival time following
profiling, so that from left to right in the plots patients are
displayed as their post-profiling survival time increases.
Green lines indicate administration of drugs predicted to
be of benefit (and therefore more prevalent in the matched
group), red lines are drugs that have a lack of benefit, and
yellow corresponds to times when both of these types of
drug were received by the patient. The recommendations
from Caris are mostly based on the literature.
Table 2 shows the drugs most frequently given to all
patients compared to the matched and unmatched groups.
Figure 1: On the left (darker grey background) - treatment regimens followed by 43 matched patients, in ascending
post-profiling survival time; on the right (lighter grey background) - 49 unmatched patients ordered by post-profiling
survival time. Each column represents one patient. The y-axis is time (days) where zero is the time of profiling. Dark grey within a
column shows the total time monitored from diagnosis to either death or last follow-up; a black line at the top of a column indicates death;
green bars represents time on a drug of benefit; red is a lack of benefit drug; yellow is time on a combination therapy associated with both
benefit and lack of benefit. Blue bars represent time on a neutral therapy associated with neither benefit nor lack of benefit.
Oncotarget4
www.impactjournals.com/oncotarget
The number of patients treated with a drug is shown in
the first column, and the number of continuous treatment
periods is shown in all other columns i.e. treatments of
the same patient with intervening periods are counted
separately. The drugs given to the most number of patients
were cyclophosphamide (70 patients), doxorubicin
hydrochloride (58) and docetaxel (56). Overall the most
commonly administered drugs were cyclophosphamide
(given for 76 time periods), doxorubicin hydrochloride
(61), and docetaxel (58). In the matched group docetaxel
was given more often than doxorubicin hydrochloride,
although cyclophosphamide was still given most
often. However, in the matched group after profiling,
cyclophosphamide, doxorubicin hydrochloride, paclitaxel
and trastuzumab were given less frequently.
On average patients received 5.8 drug treatments.
Of these, 40% (2.3 drugs) were predicted to be of
benefit, 19% (1.1 drugs) lacked benefit, and 41% (2.4)
being neither. Matched patients on average had 5.6 drug
treatments – 49% (2.7 drugs) of these were profiled to be
of benefit, 6% (0.4) lacked benefit, and 45% (2.5) being
neither. Unmatched patients received an average of 5.9
drug treatments; 33% (1.9 drugs) of these were of benefit,
30% (1.8) lacked benefit, and 38% (2.2) neither.
In the unmatched set, 76% of patients received at
least one drug treatment predicted to be of benefit, and
49% received two or more drug treatments of this type.
The most commonly given drugs of benefit
were letrozole (28), doxorubicin hydrochloride (22),
trastuzumab (22), and docetaxel (21). The most
commonly given lack of benefit drugs were doxorubicin
hydrochloride (32), trastuzumab (11) and docetaxel
(11). Some of the administered drugs did not have a
recommendation for or against, and appear in the “neither”
category. This neither class makes up 45% of drugs
administered in the matched cohort versus 38% in the
unmatched cohort. The most common agent by far in the
neither category was cyclophosphamide (given for 73 time
periods, i.e. 13% of all drug treatments for this cohort).
As might be expected, some of the drugs that were
most commonly used were administered at similar rates
whether or not they were predicted to be of benefit to
Table 2: Most frequently given drug treatments in the matched and unmatched groups, compared with all patients,
and the most popular drugs overall that were predicted to be of benefit, lacking benefit, or neither of these
Number of
Patients Treated Most Frequently Administered Drugs (Total Treatment Periods)
All Patients
Treated
All Patients –
Treatment Periods
Matched Only
Patients, All
Treatments
Matched,
After Profiling
Treatments Only
Unmatched
Patients, All
Treatments
Unmatched,
After Profiling
Treatments Only
Drugs Predicted of
Benefit
Drugs Predicted to
Lack Benefit
Drugs with No
Prediction (Neither
of Benefit or Lack
of Benefit)
cyclophosphamide
– 70 patients
cyclophosphamide
(76)
cyclophosphamide
(32)
letrozole; docetaxel
(11)
cyclophosphamide
(44) docetaxel (13) letrozole (28) doxorubicin
hydrochloride (32)
cyclophosphamide
(73)
doxorubicin
hydrochloride – 58
patients
doxorubicin
hydrochloride (61) docetaxel (29) - doxorubicin
hydrochloride (36) letrozole (11)
doxorubicin
hydrochloride;
trastuzumab (22)
trastuzumab;
docetaxel (11) docetaxel (24)
docetaxel – 56
patients docetaxel (58) doxorubicin
hydrochloride (25)
carboplatin;
capecitabine (7) docetaxel (29) gemcitabine
hydrochloride (10) - - paclitaxel (18)
carboplatin – 32
patients carboplatin (36) carboplatin (20) - trastuzumab (22) capecitabine (9) docetaxel (21) carboplatin (10) capecitabine (13)
letrozole – 31
patients trastuzumab (35) paclitaxel (17)
exemestane;
gemcitabine
hydrochloride (6)
letrozole (17) anastrozole (8) tamoxifen citrate
(18) capecitabine (6) carboplatin (12)
paclitaxel – 29
patients
letrozole; paclitaxel
(32) letrozole (15) -
capecitabine;
carboplatin;
gemcitabine
hydrochloride (16)
cyclophosphamide
(6) anastrozole (17) gemcitabine
hydrochloride (5)
gemcitabine
hydrochloride (9)
capecitabine – 27
patients - trastuzumab (13) nab-paclitaxel (5) -
methotrexate;
doxorubicin
hydrochloride (5)
carboplatin (13) methotrexate (5) fulvestrant (8)
gemcitabine
hydrochloride;
trastuzumab – 22
patients
capecitabine;
gemcitabine
hydrochloride (27)
gemcitabine
hydrochloride;
capecitabine; nab-
paclitaxel (11)
cyclophosphamide;
tamoxifen citrate;
anastrozole (4)
- - gemcitabine
hydrochloride (12) nab-paclitaxel (4) nab-paclitaxel (8)
- - - - paclitaxel (15)
carboplatin;
fluorouracil;
vinorelbine tartrate
(4)
exemestane (9)
anastrozole;
pegylated liposomal
doxorubicin
hydrochloride;
paclitaxel (3)
bevacizumab;
vinorelbine tartrate
(7)
anastrozole;
nab-paclitaxel;
tamoxifen citrate –
19 patients
anastrozole; nab-
paclitaxel (21) - - anastrozole (11) - fluorouracil (8) - -
Most commonly given drugs are listed in descending order going down, with the total number of treatments shown in parentheses.
Oncotarget5
www.impactjournals.com/oncotarget
Figure 2: Differences between matched and unmatched groups in biomarker statuses, survival, demographics and
tumour grade. Left: Comparison of biomarkers between matched and unmatched groups; positive ratio represents the percentage of the
cases that have “positive” biomarker results. Specifically, for IHC, positive is defined as protein expression being above a predetermined
threshold. For sequencing biomarkers, positive is defined as a gene mutation (usually pathogenic). The size of the circle indicates the
number of cases. Top-right: A Kaplan-Meier curve showing the increase in overall survival from time of profiling for those patients treated
only with therapies predicted to be of benefit by their molecular profile, compared to those patients who received at least one therapy
predicted to lack benefit. Middle-right and lower-right: Comparison of age of patients, survival time, treatment numbers, grade of samples,
between matched and unmatched. Blue denotes matched patients and red is unmatched patients in all plots.
Figure 3: Volcano plot of biomarkers’ prognostic value for a Caris breast cancer dataset. Biomarkers of significance that
can be used to indicate differences in survival are found in a cluster on the top right – these are the immunohistochemistry androgen receptor
(AR), estrogen receptor (ER) and progesterone receptor (PR) markers. Color code: green = the hazard rate of a positive biomarker result
is significantly lower than that of a negative biomarker result; grey = the difference between a positive biomarker result and a negative
biomarker result is not significant.
Oncotarget6
www.impactjournals.com/oncotarget
the patient. However, 31% of the times that trastuzumab
was given it was expected to lack benefit. Doxorubicin
hydrochloride was only given 38% of the time when it
was thought to be beneficial, while 55% of the time it
was predicted to lack benefit. Letrozole was used when
profiled to be of help 88% of the time. We note that
palbociclib was not used in combination with letrozole in
any of the cases in the breast cohort studied here, but most
of these cases were prior to the FDA approval of this drug.
Tamoxifen citrate was prescribed 95% of the time when
it was expected to be favorable, and anastrozole was also
well matched, being given 85% of the time that it was
predicted to be of value.
Survival analysis
Patients in the matched group on average survived
for 667 days after the day of profiling, compared to 510
days for patients whose treatments did not match their
molecular profile (P=0.0316); this is an increase of 31%.
In the matched group 26% of patients were deceased
by the end of the time of monitoring compared to 41% of
the unmatched group of patients (P=0.1257).
Patients who received more than one drug in the
lack-of-benefit category trended towards worse overall
survival (OS) than patients who received only a single
drug in this category: 550 days versus 461 days.
A Kaplan-Meier curve (Figure 2, top-right) shows
the improvement in OS from time of profiling for patients
treated only with therapies predicted to be of benefit
by their molecular profile, and separately, patients who
received at least one drug predicted to lack benefit. Figure
2 (left) also gives a comparison of biomarkers between
matched and unmatched groups, and (middle-right and
lower-right plots) matched versus unmatched: age of
patients, survival time, treatment numbers and grade of
samples.
DISCUSSION
Predictive biomarkers – matched treatments
better than unmatched
This report looked at data from a breast carcinoma
cohort made available from Caris Life Sciences via their
CODE database. This was a retrospective review of a
cohort of patients that were profiled using established
IHC biomarkers, along with fragment analysis, in situ
hybridization and sequencing. Their treatments were either
matched or unmatched based on whether the treatment
chosen by their physician was predicted to be beneficial
by Caris Life Sciences using the molecular profile of the
tumor. Patients whose treatments subsequently agreed
with these recommendations were compared to those
who received at least one drug that was predicted to lack
benefit, i.e. their regimen did not agree with their tumor
profile-based treatment predictions. Comparing these two
groups showed that the matched treatment group had an
increase of 31% in survival compared to the average for
the unmatched group, an increase of 157 days from 510 to
667 days (P=0.0316).
When comparing the matched and unmatched
groups, in terms of HER2 and ER directed therapies,
there was a similar level of use – in the matched group
87% of treatments were of either of these types, and in
the unmatched group 83% of the treatments were one of
these two types.
The unmatched group received 0.32 more lines of
therapy on average than the matched group, survived for
less time, and had a higher mortality rate. This could have
been influenced by the tendency for the unmatched group
to have tumors that were generally more advanced than in
the matched group, as shown in Table 1. The unmatched
group may have tended to receive more treatments and
be less adherent to the recommended treatments, due to
clinicians trying all possible options as a last resort as the
disease advanced, although this is speculative.
Interestingly, across this cohort of patients, the
expression of ER and PR was found to be prognostic
for overall survival (see Figure 3). The expression of the
androgen receptor (AR) was also found to be prognostic;
this agrees with previously published data that shows
improved long-term survival with co-expression of AR in
ER positive breast cancers [16].
The survival curves from time of diagnosis initially
overlap and then diverge after profiling occurs. This
may suggest that basing therapy on tumor profiling has
an effect on selecting optimal therapies and improving
outcome. Combined with the increase in survival and
lowering of death rates, this leads to the conclusion that
there is a beneficent role of tumor molecular profiling in
this cohort.
MATERIALS AND METHODS
The Caris CODE database (Comprehensive
Oncology Database Explorer) contains tumor molecular
profile data for 841 patients with solid tumors in version
1.0. It also contains demographic information about the
patients, their drug treatments that they received before
and after molecular profiling, and records of their clinical
outcomes. There are 92 breast cancer patients recorded,
and this breast cancer cohort was mined after web scraping
the data from the Caris website, to determine if molecular
characterization recommendations influenced drug
selection by their physicians after the time of profiling,
and if any molecular subsets had different outcomes. Table
1 describes the clinical characteristics of the patients in
this breast cancer cohort. According to Caris, 33% of
the samples were from metastatic samples; 50% of these
Oncotarget7
www.impactjournals.com/oncotarget
metastatic breast samples were from the lymph nodes, and
the rest were from other sites.
The amount of time that patients were monitored
varied, as shown in Figure 1. On average patients’
treatment records were available for 1327 days after
diagnosis (1342 for matched treatment patients and 131
for unmatched), and on average the time of monitoring
after profiling was 583 days. The longest period of
monitoring after tumor profiling (the patient represented
on the furthest right of Figure 1) was 1317 days; this was
1407 days after diagnosis. The longest amount of time that
records were available, i.e. after diagnosis up until the last
contact day, was 9427 days.
Abbreviations
AR: androgen receptor; HER2: human epidermal
growth factor receptor 2; IHC: immunohistochemistry;
ER: estrogen receptor; OS: overall survival; PR:
progesterone receptor.
Author contributions
All authors contributed equally to the work
described in this paper and were involved in writing it.
ACKNOWLEDGMENTS
Thanks to Caris Life Sciences for the provision
of their CODE database and the data contained within,
and assistance by Dr David Spetzler, Todd Maney, Dr Jia
Zeng, Dr Tina Lui, Dr Nick Xiao and Stephanie Ratliff in
the use of this resource. We are also grateful to the patients
involved in this study.
CONFLICTS OF INTEREST
We have no conflicts of interest.
FUNDING
This work was supported by the Action Against
Cancer charity (http://www.aacancer.org/). Infrastructure
support was provided by the Imperial Experimental
Cancer Medicine Centre, the CRUK Imperial Centre, and
the Imperial NIHR Biomedical Research Centre.
REFERENCES
1.
Stephens, P.J., Stephens P.J., Tarpey P.S., Davies H., Van
Loo P., Greenman C., Wedge D.C., Nik-Zainal S., Martin
S., Varela I., Bignell G.R., Yates L.R., Papaemmanuil E., et
al., The landscape of cancer genes and mutational processes
in breast cancer. Nature, 2012. 486: p. 400-4.
2.
Alexandrov L.B., Nik-Zainal S., Wedge D.C., Aparicio
S.A., Behjati S., Biankin A.V., Bignell G.R., Bolli N., Borg
A., Børresen-Dale A.L., Boyault S., Burkhardt B., Butler
A.P., et al., Signatures of mutational processes in human
cancer. Nature, 2013. 500: p. 415-21.
3.
Nik-Zainal S., Davies H., Staaf J., Ramakrishna M., Glodzik
D., Zou X., Martincorena I., Alexandrov L.B., Martin S.,
Wedge D.C., Van Loo P., Ju Y.S., Smid M., et al., Landscape
of somatic mutations in 560 breast cancer whole-genome
sequences. Nature, 2016. 534: p. 47-54.
4.
Pereira B., Chin S.F., Rueda O.M., Vollan H.K., Provenzano
E., Bardwell H.A., Pugh M., Jones L., Russell R., Sammut
S.J., Tsui D.W., Liu B., Dawson S.J., et al., The somatic
mutation profiles of 2,433 breast cancers refines their
genomic and transcriptomic landscapes. Nat Commun,
2016. 7: p. 11479.
5.
Nik-Zainal, S., Alexandrov L.B., Wedge D.C., Van Loo
P.,Greenman C.D., Raine K., Jones D., Hinton J., Marshall
J.,Stebbings L.A., Menzies A., Martin S., Leung K., et al.,
Mutational processes molding the genomes of 21 breast
cancers. Cell, 2012. 149: p. 979-93.
6.
Shlien A., Raine K., Fuligni F., Arnold R., Nik-Zainal S.,
Dronov S., Mamanova L., Rosic A., Ju Y.S., Cooke S.L.,
Ramakrishna M., Papaemmanuil E., Davies H.R., et al.,
Direct Transcriptional Consequences of Somatic Mutation
in Breast Cancer. Cell Rep, 2016. 16: p. 2032-46.
7.
Smid M., Rodríguez-González F.G., Sieuwerts A.M.,
Salgado R., Prager-Van der Smissen W.J., Vlugt-Daane
M.V., van Galen A., Nik-Zainal S., Staaf J., Brinkman
A.B., van de Vijver M.J., Richardson A.L., Fatima A.,
et al., Breast cancer genome and transcriptome integration
implicates specific mutational signatures with immune cell
infiltration. Nat Commun, 2016. 7: p. 12910.
8.
Ward, H.W., Anti-oestrogen therapy for breast cancer: a trial
of tamoxifen at two dose levels. Br Med J, 1973. 1: p. 13-4.
9.
Bozovic-Spasojevic I., Azim H.A. Jr, Paesmans M., Suter
T., Piccart M.J., de Azambuja E., Neoadjuvant anthracycline
and trastuzumab for breast cancer: is concurrent treatment
safe? Lancet Oncol, 2011. 12: p. 209-11.
10.
Baxter E., Gondara L., Lohrisch C., Chia S., Gelmon K.,
Hayes M., Davidson A., Tyldesley S., Using proliferative
markers and Oncotype DX in therapeutic decision-making
for breast cancer: the B.C. experience. Curr Oncol, 2015.
22: p. 192-8.
11.
Garnett M.J., Edelman E.J., Heidorn S.J., Greenman C.D.,
Dastur A., Lau K.W., Greninger P., Thompson I.R., Luo
X., Soares J., Liu Q., Iorio F., Surdez D., et al., Systematic
identification of genomic markers of drug sensitivity in
cancer cells. Nature, 2012. 483: p. 570-5.
12.
Shaw A.T., Kim D.W., Nakagawa K., Seto T., Crinó L., Ahn
M.J., De Pas T., Besse B., Solomon B.J., Blackhall F., Wu
Y.L., Thomas M., O'Byrne K.J., et al., Crizotinib versus
chemotherapy in advanced ALK-positive lung cancer. N
Engl J Med, 2013. 368: p. 2385-94.
Oncotarget8
www.impactjournals.com/oncotarget
13.
Frampton G.M., Fichtenholtz A., Otto G.A., Wang
K., Downing S.R., He J., Schnall-Levin M., White J.,
Sanford E.M., An P., Sun J., Juhn F., Brennan K., et al.,
Development and validation of a clinical cancer genomic
profiling test based on massively parallel DNA sequencing.
Nat Biotechnol, 2013. 31: p. 1023-31.
14.
Khirade, M.F., G. Lal, S.A. Bapat, Derivation of a fifteen gene
prognostic panel for six cancers. Sci Rep, 2015. 5: p. 13248.
15.
Corcoran, R.B. New therapeutic strategies for BRAF
mutant colorectal cancers. J Gastrointest Oncol. 2015
Dec;6:650-9
16.
Hu R., Dawood S., Holmes M.D., Collins L.C., Schnitt
S.J., Cole K., Marotti J.D., Hankinson S.E., Colditz G.A.,
Tamimi R.M. Androgen receptor expression and breast
cancer survival in postmenopausal women. Clin Cancer
Res, 2011. 17: p. 1867-74.
... In a retrospective study, 92 breast cancer patients (33% with metastatic disease) from the Caris Molecular Intelligence database were retrospectively divided based on whether they received treatment that aligned with the Caris recommendation (matched, n = 43) or if they received at least one therapy that was predicted to lack benefit (unmatched, n = 49). 65 The matched group had an average overall survival of 667 days, whereas the unmatched group had average overall survival of 510 days (p = 0.0316). 65 While this data is promising, there is still a need for a larger clinical trial to validate a mortality benefit from the Caris test. ...
... 65 The matched group had an average overall survival of 667 days, whereas the unmatched group had average overall survival of 510 days (p = 0.0316). 65 While this data is promising, there is still a need for a larger clinical trial to validate a mortality benefit from the Caris test. ...
Article
As genetic testing becomes increasingly more accessible and more applicable with a broader range of clinical implications, it may also become more challenging for breast cancer providers to remain up‐to‐date. This review outlines some of the current clinical guidelines and recent literature surrounding germline genetic testing, as well as genomic testing, in the screening, prevention, diagnosis, and treatment of breast cancer, while identifying potential areas of further research.
... Based on some of these factors, we previously developed a novel staging system to stratify patients with de novo MBC into 3 subgroups (IVA, IVB, and IVC) in order to better discriminate prognosis [16•] and validation studies are ongoing. Others are investigating the significance of circulating tumor cells [17] and genomic assays to help tailor treatments [18,19], although tumor genomic profiling in particular may not always impact clinical management decisions [20]. ...
Article
Full-text available
Purpose of Review Among individuals with metastatic breast cancer, surgical resection of the primary tumor remains controversial, and its benefit is unclear. In this review, we highlight select retrospective and prospective studies which have sought to address this clinical scenario. In addition, we discuss further considerations that may be relevant. Recent Findings Numerous retrospective studies have suggested a potential survival benefit associated with surgical resection of the primary breast tumor in women with metastatic disease; however, three randomized controlled trials more recently have challenged these findings. Mixed results have demonstrated no survival benefit with locoregional treatment versus limited benefit in select patient groups. Summary Prospective studies suggest that most patients with metastatic breast cancer are unlikely to experience a survival benefit related to resection of their primary tumor. However, ongoing work seeks to further define if there may be select subgroups that could benefit from surgery.
... Researchers have been working to understand cancer-driving mechanisms of each BCa subtype to improve tools for diagnostic and treatment purposes. New research focuses on elucidating molecular pathways that are associated with cancer signatures of advanced BCa tumors [8]. For instance, pathways related to protein synthesis, degradation and metabolic changes that facilitate energy production of cancer cells have been identified to be enriched in BCa tissues [9]. ...
Article
Breast cancer (BCa) is the second most common cancer worldwide and the most prevalent cancer in women. The majority of BCa cases are positive (+) for the estrogen receptor (ER⁺, 80%) and progesterone receptor (PR⁺, 65%). Estrogen and progesterone hormones are known to be involved in cancer progression, and thus hormonal deprivation is used as an effective treatment for ER⁺PR⁺ BCa subtypes. However, some ER⁺PR⁺ BCa patients develop resistance to such therapies. Meanwhile, chemotherapy is the only available treatment for ER⁻PR⁻ BCa tumors. Another hormone receptor known as the androgen receptor (AR) has also been found to be widely expressed in human breast carcinomas. However, the mechanisms of AR and its endogenous androgen ligands is not well-understood in BCa and its biological role in this hormone-related disease remains unclear. In this review, we aim to address the importance of the AR in BCa diagnosis and prognosis, current AR-targeting approaches in BCa, and the potential for AR-downstream molecules to serve as therapeutic targets.
Article
Full-text available
Multigene sequencing technologies provide a foundation for targeted therapy and precision oncology by identifying actionable alterations and enabling the development of treatments that substantially improve clinical outcomes. This review emphasizes the importance of having a molecular compass guiding treatment decision-making through the multitude of alterations and genetic mutations, showcasing why NGS plays a pivotal role in modern oncology.
Article
Full-text available
Purpose: This review summarizes the published evidence on the clinical impact of using next-generation sequencing (NGS) tests to guide management of patients with cancer in the United States. Methods: We performed a comprehensive literature review to identify recent English language publications that presented progression-free survival (PFS) and overall survival (OS) of patients with advanced cancer receiving NGS testing. Results: Among 6,475 publications identified, 31 evaluated PFS and OS among subgroups of patients who received NGS-informed cancer management. PFS and OS were significantly longer among patients who were matched to targeted treatment in 11 and 16 publications across tumor types, respectively. Conclusion: Our review indicates that NGS-informed treatment can have an impact on survival across tumor types.
Article
Full-text available
Background: Tamoxifen is one of the medicines for adjuvant endocrine therapy of hormone-dependent breast cancer. However, development of resistance to tamoxifen occurs inevitably during treatment. This study aimed to determine whether sensitivity of tamoxifen-resistant breast cancer cells (TAM-R) could be reinstated by tetrandrine (Tet). Methods: All experiments were conducted in TAM-R cells derived from the MCF-7 breast cancer cell line by long-term tamoxifen exposure. Cell growth, apoptosis, and autophagy were end-points that evaluated the effect of Tet (0.9 μg/ml, 1.8 μg/ml, and 3.75 μg/ml) alone or in combination with TAM (1 μM). Cell apoptosis was determined by an ELISA assay and autophagy was determined by fluorescent staining using the Enzo autophagy detection kit. Immunoblotting was used to evaluate markers for apoptosis, autophagy, and related signal pathway molecules. Results: Growth of TAM-R cells was significantly inhibited by Tet. Combination of Tet with tamoxifen induced a greater inhibition on cell growth than tamoxifen alone, which was predominantly due to enhancement of pro-apoptotic effect of TAM by Tet. Autophagy was significantly inhibited in TAM-R cells treated with Tet plus TAM as shown by increased autophagosomes and the levels of LC3-II and p62. At 0.9 μg/ml, Tet increased the levels of both apoptosis and autophagy markers. Among them increase in p53 levels was more dramatic. Conclusions: Tet as a monotherapy inhibits TAM-R cells. Tet potentiates the pro-apoptotic effect of TAM via inhibition of autophagy.
Article
Full-text available
A recent comprehensive whole genome analysis of a large breast cancer cohort was used to link known and novel drivers and substitution signatures to the transcriptome of 266 cases. Here, we validate that subtype-specific aberrations show concordant expression changes for, for example, TP53, PIK3CA, PTEN, CCND1 and CDH1. We find that CCND3 expression levels do not correlate with amplification, while increased GATA3 expression in mutant GATA3 cancers suggests GATA3 is an oncogene. In luminal cases the total number of substitutions, irrespective of type, associates with cell cycle gene expression and adverse outcome, whereas the number of mutations of signatures 3 and 13 associates with immune-response specific gene expression, increased numbers of tumour-infiltrating lymphocytes and better outcome. Thus, while earlier reports imply that the sheer number of somatic aberrations could trigger an immune-response, our data suggests that substitutions of a particular type are more effective in doing so than others.
Article
Full-text available
Disordered transcriptomes of cancer encompass direct effects of somatic mutation on transcription, coordinated secondary pathway alterations, and increased transcriptional noise. To catalog the rules governing how somatic mutation exerts direct transcriptional effects, we developed an exhaustive pipeline for analyzing RNA sequencing data, which we integrated with whole genomes from 23 breast cancers. Using X-inactivation analyses, we found that cancer cells are more transcriptionally active than intermixed stromal cells. This is especially true in estrogen receptor (ER)-negative tumors. Overall, 59% of substitutions were expressed. Nonsense mutations showed lower expression levels than expected, with patterns characteristic of nonsense-mediated decay. 14% of 4,234 rearrangements caused transcriptional abnormalities, including exon skips, exon reusage, fusions, and premature polyadenylation. We found productive, stable transcription from sense-to-antisense gene fusions and gene-to-intergenic rearrangements, suggesting that these mutation classes drive more transcriptional disruption than previously suspected. Systematic integration of transcriptome with genome data reveals the rules by which transcriptional machinery interprets somatic mutation.
Article
Full-text available
The genomic landscape of breast cancer is complex, and inter- and intra-tumour heterogeneity are important challenges in treating the disease. In this study, we sequence 173 genes in 2,433 primary breast tumours that have copy number aberration (CNA), gene expression and long-term clinical follow-up data. We identify 40 mutation-driver (Mut-driver) genes, and determine associations between mutations, driver CNA profiles, clinical-pathological parameters and survival. We assess the clonal states of Mut-driver mutations, and estimate levels of intra-tumour heterogeneity using mutant-allele fractions. Associations between PIK3CA mutations and reduced survival are identified in three subgroups of ER-positive cancer (defined by amplification of 17q23, 11q13–14 or 8q24). High levels of intra-tumour heterogeneity are in general associated with a worse outcome, but highly aggressive tumours with 11q13–14 amplification have low levels of intra-tumour heterogeneity. These results emphasize the importance of genome-based stratification of breast cancer, and have important implications for designing therapeutic strategies.
Article
Full-text available
We analysed whole-genome sequences of 560 breast cancers to advance understanding of the driver mutations conferring clonal advantage and the mutational processes generating somatic mutations. We found that 93 protein-coding cancer genes carried probable driver mutations. Some non-coding regions exhibited high mutation frequencies, but most have distinctive structural features probably causing elevated mutation rates and do not contain driver mutations. Mutational signature analysis was extended to genome rearrangements and revealed twelve base substitution and six rearrangement signatures. Three rearrangement signatures, characterized by tandem duplications or deletions, appear associated with defective homologous-recombination-based DNA repair: one with deficient BRCA1 function, another with deficient BRCA1 or BRCA2 function, the cause of the third is unknown. This analysis of all classes of somatic mutation across exons, introns and intergenic regions highlights the repertoire of cancer genes and mutational processes operating, and progresses towards a comprehensive account of the somatic genetic basis of breast cancer.
Article
Full-text available
The hallmarks of cancer deem biological pathways and molecules to be conserved. This approach may be useful for deriving a prognostic gene signature. Weighted Gene Co-expression Network Analysis of gene expression datasets in eleven cancer types identified modules of highly correlated genes and interactive networks conserved across glioblastoma, breast, ovary, colon, rectal and lung cancers, from which a universal classifier for tumor stratification was extracted. Specific conserved gene modules were validated across different microarray platforms and datasets. Strikingly, preserved genes within these modules defined regulatory networks associated with immune regulation, cell differentiation, metastases, cell migration, metastases, oncogenic transformation, and resistance to apoptosis and senescence, with AIF1 and PRRX1 being suggested to be master regulators governing these biological processes. A universal classifier from these conserved networks enabled execution of common set of principles across different cancers that revealed distinct, differential correlation of biological functions with patient survival in a cancer-specific manner. Correlation analysis further identified a panel of 15 risk genes with potential prognostic value, termed as the GBOCRL-IIPr panel [(GBM-Breast-Ovary-Colon-Rectal-Lung)-Immune-Invasion-Prognosis], that surprisingly, were not amongst the master regulators or important network hubs. This panel may now be integrated in predicting patient outcomes in the six cancers.
Article
Full-text available
Proliferative scoring of breast tumours can guide treatment recommendations, particularly for estrogen receptor (er)-positive, her2-negative, T1-2, N0 disease. Our objectives were to □ estimate the proportion of such patients for whom proliferative indices [mitotic count (mc), Ki-67 immunostain, and Oncotype dx (Genomic Health, Redwood City, CA, U.S.A.) recurrence score (rs)] were obtained.□ compare the indices preferred by oncologists with the indices available to them.□ correlate Nottingham grade (ng) and its subcomponents with Oncotype dx.□ assess interobserver variation. All of the er-positive, her2-negative, T1-2, N0 breast cancers diagnosed from 2007 to 2011 (n = 5110) were linked to a dataset of all provincial breast cancers with a rs. A 5% random sample of the 5110 cancers was reviewed to estimate the proportion that had a mc, Ki-67 index, and rs. Correlation coefficients were calculated for the rs with ng subcomponent scores. Interobserver variation in histologic grading between outside and central review pathology reports was assessed using a weighted kappa test. During 2007-2011, most cancers were histologically graded and assigned a mc; few had a Ki-67 index or rs. The ng and mc were significantly positively correlated with rs. The level of agreement in histologic scoring between outside and central pathology reports was good or very good. Very few cases with a low mc had a high rs (1.8%). Patients with low ng and mc scores are unlikely to have a high rs, and thus are less likely to benefit from chemotherapy. In the context of limited resources, that finding can guide clinicians about when a rs adds the most value.
Article
Full-text available
As more clinically relevant cancer genes are identified, comprehensive diagnostic approaches are needed to match patients to therapies, raising the challenge of optimization and analytical validation of assays that interrogate millions of bases of cancer genomes altered by multiple mechanisms. Here we describe a test based on massively parallel DNA sequencing to characterize base substitutions, short insertions and deletions (indels), copy number alterations and selected fusions across 287 cancer-related genes from routine formalin-fixed and paraffin-embedded (FFPE) clinical specimens. We implemented a practical validation strategy with reference samples of pooled cell lines that model key determinants of accuracy, including mutant allele frequency, indel length and amplitude of copy change. Test sensitivity achieved was 95-99% across alteration types, with high specificity (positive predictive value >99%). We confirmed accuracy using 249 FFPE cancer specimens characterized by established assays. Application of the test to 2,221 clinical cases revealed clinically actionable alterations in 76% of tumors, three times the number of actionable alterations detected by current diagnostic tests.
Article
Full-text available
All cancers are caused by somatic mutations; however, understanding of the biological processes generating these mutations is limited. The catalogue of somatic mutations from a cancer genome bears the signatures of the mutational processes that have been operative. Here we analysed 4,938,362 mutations from 7,042 cancers and extracted more than 20 distinct mutational signatures. Some are present in many cancer types, notably a signature attributed to the APOBEC family of cytidine deaminases, whereas others are confined to a single cancer class. Certain signatures are associated with age of the patient at cancer diagnosis, known mutagenic exposures or defects in DNA maintenance, but many are of cryptic origin. In addition to these genome-wide mutational signatures, hypermutation localized to small genomic regions, ‘kataegis’, is found in many cancer types. The results reveal the diversity of mutational processes underlying the development of cancer, with potential implications for understanding of cancer aetiology, prevention and therapy.
Article
Full-text available
Background: In single-group studies, chromosomal rearrangements of the anaplastic lymphoma kinase gene (ALK) have been associated with marked clinical responses to crizotinib, an oral tyrosine kinase inhibitor targeting ALK. Whether crizotinib is superior to standard chemotherapy with respect to efficacy is unknown. Methods: We conducted a phase 3, open-label trial comparing crizotinib with chemotherapy in 347 patients with locally advanced or metastatic ALK-positive lung cancer who had received one prior platinum-based regimen. Patients were randomly assigned to receive oral treatment with crizotinib (250 mg) twice daily or intravenous chemotherapy with either pemetrexed (500 mg per square meter of body-surface area) or docetaxel (75 mg per square meter) every 3 weeks. Patients in the chemotherapy group who had disease progression were permitted to cross over to crizotinib as part of a separate study. The primary end point was progression-free survival. Results: The median progression-free survival was 7.7 months in the crizotinib group and 3.0 months in the chemotherapy group (hazard ratio for progression or death with crizotinib, 0.49; 95% confidence interval [CI], 0.37 to 0.64; P<0.001). The response rates were 65% (95% CI, 58 to 72) with crizotinib, as compared with 20% (95% CI, 14 to 26) with chemotherapy (P<0.001). An interim analysis of overall survival showed no significant improvement with crizotinib as compared with chemotherapy (hazard ratio for death in the crizotinib group, 1.02; 95% CI, 0.68 to 1.54; P=0.54). Common adverse events associated with crizotinib were visual disorder, gastrointestinal side effects, and elevated liver aminotransferase levels, whereas common adverse events with chemotherapy were fatigue, alopecia, and dyspnea. Patients reported greater reductions in symptoms of lung cancer and greater improvement in global quality of life with crizotinib than with chemotherapy. Conclusions: Crizotinib is superior to standard chemotherapy in patients with previously treated, advanced non-small-cell lung cancer with ALK rearrangement. (Funded by Pfizer; ClinicalTrials.gov number, NCT00932893.).
Article
Oncogenic BRAF mutations are found in ~10% of colorectal cancers (CRCs) and predict poor prognosis. Although BRAF inhibitors have demonstrated striking efficacy in BRAF mutant melanomas, BRAF inhibitor monotherapy is ineffective in BRAF mutant CRC. Over the past few years, studies have begun to define the molecular mechanisms underlying the relative resistance of BRAF mutant CRC to BRAF inhibitors, leading to the development of novel therapeutic strategies that are showing promising clinical activity in initial clinical trials. Our current understanding of the mechanisms of BRAF inhibitor resistance in BRAF mutant CRC and the therapeutic approaches currently in clinical trials for BRAF mutant CRC are reviewed herein.