ArticlePDF Available

Therapeutic effect of β-blockers in triple-negative breast cancer postmenopausal women

Authors:

Abstract and Figures

Beta-blockers (BB) drugs have been used for decades worldwide, mainly to treat hypertension. However, in recent epidemiological studies, BBs were suggested to improve cancer prognosis. In the wake of this evidence, we evaluated the possible therapeutic effect of BBs in triple-negative breast cancer (TNBC) patients. We identified 800 postmenopausal women operated between 1997 and 2008 for early primary TNBC. The effect of BB intake on the risk of breast cancer (BC) recurrence and death was evaluated through competing risk and Cox regression survival models. At cancer diagnosis, 74 (9.3 %) women out of 800 were BBs users. Median age was 62 years in BB users and 59 years in non-users (P = 0.02). BB users and non-users were similarly distributed by all tumor characteristics. The 5-year cumulative incidence of BC-related events was 13.6 % in BB users and 27.9 % in non-users (P = 0.02). The beneficial impact of BBs remained statistically significant at multivariable analysis (HR, 0.52; 95 % CI 0.28-0.97), after the adjustment for age, tumor stage, and treatment, peritumoral vascular invasion and use of other antihypertensive drugs, antithrombotics, and statins. Adjusted HRs for metastases and for BC deaths were 0.32 (95 % CI 0.12-0.90) and 0.42 (95 % CI 0.18-0.97), respectively, in favor of BBs. Hypertension, other antihypertensive drugs, antithrombotics, and statins did not impact prognosis. In this series of postmenopausal TNBC patients, BB intake was associated with a significantly decreased risk of BC-related recurrence, metastasis, and BC death. Innovative therapeutic strategies including BBs should be urgently explored in cancer patients.
Content may be subject to copyright.
EPIDEMIOLOGY
Therapeutic effect of b-blockers in triple-negative breast cancer
postmenopausal women
Edoardo Botteri
Elisabetta Munzone
Nicole Rotmensz
Carlo Cipolla
Vincenzo De Giorgi
Barbara Santillo
Arnaldo Zanelotti
Laura Adamoli
Marco Colleoni
Giuseppe Viale
Aron Goldhirsch
Sara Gandini
Received: 1 July 2013 / Accepted: 26 July 2013 / Published online: 3 August 2013
Ó Springer Science+Business Media New York 2013
Abstract Beta-blockers (BB) drugs have been used for
decades worldwide, mainly to treat hypertension. However, in
recent epidemiological studies, BBs were suggested to
improve cancer prognosis. In the wake of this evidence, we
evaluated the possible therapeutic effect of BBs in triple-
negative breast cancer (TNBC) patients. We identified 800
postmenopausal women operated between 1997 and 2008 for
early primary TNBC. The effect of BB intake on the risk of
breast cancer (BC) recurrence and death was evaluated
through competing risk and Cox regression survival models.
At cancer diagnosis, 74 (9.3 %) women out of 800 were BBs
users. Median age was 62 years in BB users and 59 years in
non-users (P = 0.02). BB users and non-users were similarly
distributed by all tumor characteristics. The 5-year cumulative
incidence of BC-related events was 13.6 % in BB users and
27.9 % in non-users (P = 0.02). The beneficial impact of BBs
remained statistically significant at multivariable analysis
(HR, 0.52; 95 % CI 0.28–0.97), after the adjustment for age,
tumor stage, and treatment, peritumoral vascular invasion and
use of other antihypertensive drugs, antithrombotics, and
statins. Adjusted HRs for metastases and for BC deaths were
0.32 (95 % CI 0.12–0.90) and 0.42 (95 % CI 0.18–0.97),
respectively, in favor of BBs. Hypertension, other antihyper-
tensive drugs, antithrombotics, and statins did not impact
prognosis. In this series of postmenopausal TNBC patients,
BB intake was associated with a significantly decreased risk of
BC-related recurrence, metastasis, and BC death. Innovative
therapeutic strategies including BBs should be urgently
explored in cancer patients.
Keywords Beta-blockers Breast cancer
Triple negative Prognosis
Introduction
Beta-blocker (BB) drugs have been used for decades
worldwide, typically to treat hypertension, coronary artery
Electronic supplementary material The online version of this
article (doi:10.1007/s10549-013-2654-3) contains supplementary
material, which is available to authorized users.
E. Botteri (&) N. Rotmensz B. Santillo S. Gandini
Division of Epidemiology and Biostatistics, European Institute
of Oncology, Milan, Italy
e-mail: edoardo.botteri@ieo.it
E. Munzone M. Colleoni
Division of Medical Senology, European Institute of Oncology,
Milan, Italy
C. Cipolla A. Zanelotti
Division of Cardiology, European Institute of Oncology,
Milan, Italy
V. De Giorgi
Department of Dermatology, University of Florence, Florence,
Italy
L. Adamoli A. Goldhirsch
Department of Medicine, European Institute of Oncology, Milan,
Italy
G. Viale
Department of Pathology and Laboratory Medicine, European
Institute of Oncology, Milan, Italy
G. Viale
University of Milan, Milan, Italy
123
Breast Cancer Res Treat (2013) 140:567–575
DOI 10.1007/s10549-013-2654-3
disease, and arrhythmias [1]. Despite the therapeutic indi-
cations, evidence from recent epidemiological studies
suggested that BB intake might potentially improve prog-
nosis in patients affected by prostate [2, 3], breast [47],
lung [8], and ovarian [9] cancer as well as cutaneous
melanoma [10, 11]. Moreover, BB intake was reported to
reduce overall cancer incidence [12] and specific prostate
cancer risk [13]. This observed association is supported by
data coming from preclinical in vivo and in vitro studies,
which showed how b-adrenergic receptors mediate the
effect of stress hormones norepinephrine and epinephrine
on the tumor microenvironment, and how antagonists of
Table 1 Characteristics at
baseline by beta-blocker intake
BB beta-blocker, ACEI
angiotensin-converting enzyme
inhibitor, ARB angiotensin
receptor blocker, CCB calcium
channel blocker, MAST
mastectomy, QUAD
quadrantectomy
a
Clinical stage was used for
patients undergoing
neoadjuvant chemotherapy
b
Among patients not receiving
neoadjuvant chemotherapy.
Numbers might not sum up to
totals due to missing values
Variable Category No BB BB P value
No. (%) No. (%)
Total 726 (90.7) 74 (9.3)
Age Median (range) 59 (30–89) 62 (48–80) 0.02
\65 506 (69.7) 42 (56.8) 0.02
C65 220 (30.3) 32 (43.2)
BMI \25 364 (60.2) 35 (52.2) 0.15
25–30 187 (30.9) 23 (34.3)
[30 54 (8.9) 9 (13.4)
T
a
1 321 (45.5) 41 (56.2) 0.34
2 285 (40.4) 22 (30.1)
3 36 (5.1) 3 (4.1)
4 64 (9.1) 7 (9.6)
N
a
0 374 (53.5) 42 (57.5) 0.43
1 206 (29.5) 21 (28.8)
2/3 119 (17.0) 10 (13.7)
Ki-67 Median (range) 42 (1–95) 38 (4–90) 0.37
B40 337 (48.6) 40 (54.8) 0.32
[40 356 (51.4) 33 (45.2)
Peritumoral vascular invasion Absent 539 (76.0) 61 (83.6) 0.14
Focal 90 (12.7) 7 (9.6)
Extensive 80 (11.3) 5 (6.9)
Neoadjuvant chemotherapy No 598 (82.4) 61 (82.4) 0.99
Yes 128 (17.6) 13 (17.6)
Type of surgery MAST 168 (23.1) 15 (20.3) 0.58
QUAD 558 (76.9) 59 (79.7)
Radiotherapy No 106 (14.6) 8 (10.8) 0.37
Yes 620 (85.4) 66 (89.2)
Adjuvant chemotherapy No 117 (16.1) 10 (13.5) 0.56
Yes 609 (83.9) 64 (86.5)
Hypertension No 533 (73.4) 1 (1.3) \0.01
Yes 193 (26.6) 73 (98.7)
ACEI No use 640 (88.2) 51 (68.9) \0.01
Use 86 (11.8) 23 (31.1)
ARB No use 686 (94.5) 63 (85.1) \0.01
Use 40 (5.5) 11 (14.9)
CCB No use 677 (93.3) 61 (82.4) \0.01
Use 49 (6.7) 13 (17.6)
Diuretic No use 654 (90.1) 49 (66.2) \0.01
Use 72 (9.9) 25 (33.8)
Antithrombotics No use 710 (97.8) 65 (87.8) \0.01
Use 16 (2.2) 9 (12.1)
Statins No use 680 (93.7) 63 (85.1) \0.01
Use 46 (6.3) 11 (14.9)
568 Breast Cancer Res Treat (2013) 140:567–575
123
b-adrenergic receptors, such as BBs, can interfere with
tumor cell proliferation and migration, as well as tumoral
angiogenesis [1417]. The general pattern of transcrip-
tional responses induced by b-adrenergic signaling
includes upregulated expression of metastasis-associated
genes involved in inflammation, angiogenesis, tissue
invasion, and epithelial–mesenchymal transition, and
downregulated expression of genes facilitating antitumor
immune responses [16].
On the basis of all this evidence, we decided to examine
the potential benefit of BB use in the subset of women
affected by triple-negative breast cancer (TNBC), which
represents one of the most aggressive and difficult-to-treat
cancers. Should the beneficial impact of BBs on prognosis
be confirmed in this and other series of TNBC patients,
innovative therapeutic strategies involving the use of BBs
are to be urgently explored.
Patients and methods
We retrospectively analyzed data of all consecutive women
diagnosed and operated for early primary TNBC between
1997 and 2008 at the European Institute of Oncology in
Milan (n = 1,464). Patients with history of any previous
invasive cancer or with metastatic disease at diagnosis
(stage IV) were previously excluded. Given the low prev-
alence (1.1 %) of BB users in premenopausal patients, we
decided to limit the analysis to postmenopausal women
only (n = 800) in order to avoid high heterogeneity
between BB users and non-users. Patients were divided
into two groups: the BB users group, which included
patients who were using any BB at the moment of their
diagnosis of TNBC, and the BB non-users group, which
included all the other patients. The data sources were the
institutional Breast Cancer Database, which collects data
on patients’ characteristics, breast cancer (BC) features and
comorbidities such as hypertension and hypercholesterol-
emia, and the Cardiology Division Database, which col-
lects information on patients’ diseases of the circulatory
system and cardiovascular drugs. In addition to BBs, we
analyzed data on statins, antithrombotics, and other anti-
hypertensive drugs such as angiotensin-converting enzyme
inhibitors, angiotensin receptor blocker, calcium channel
blocker, and diuretics. If a patient took one single drug
containing two or more different antihypertensive agents
(e.g., angiotensin-converting enzyme inhibitor plus diure-
tic), she was considered as taking two or more different
antihypertensive drugs. Use of the data for the present
study was approved by the Institutional Review Board.
TNBC was defined as a BC with negative estrogen and
progesterone receptor expression (i.e., \1 % immunore-
active tumor cells) and without overexpression of HER2/
neu. Immunohistochemical expression of HER2/neu was
scored as follows: 0 (no staining or faint membrane
staining), 1? (faint membrane staining in [10 % of tumor
cells, incomplete membrane staining), 2? (weak to mod-
erate membrane staining in[10 % of tumor cells), and 3?
(intense circumferential membrane staining in [10 % of
tumor cells). For this analysis, we included patients with
negative estrogen and progesterone receptor tumors char-
acterized by HER2/neu scores of 0 and 1?, as well as 2?
resulting negative for gene amplification by fluorescence
in situ hybridization (FISH; Vysis PathVysion; Abbott,
Chicago, IL).
For all the patients receiving pre-surgical chemotherapy
(CT) we reported the clinical stage (T and N). When
pathological complete response was obtained, pathological
data such as ER, PgR, HER2/neu, and Ki-67 were retrieved
from the initial breast lesion biopsy. Responses to pre-
surgical CT were evaluated using the Response Evaluation
Criteria in Solid Tumors (RECIST) guidelines.
Statistical analysis
Association between categorical variables and BB use was
evaluated by the Chi square test, the Fisher exact test and
Table 2 Events and follow-up by beta-blocker intake
Event No BB BB P value
No. (%) No. (%)
At risk 726 74
First events
Local
a
52 (7.2) 6 (8.1) 0.77
Regional
a
28 (3.9) 1 (1.3) 0.51
Distant
a,b
110 (15.2) 3 (4.1) \0.01
Contralateral breast tumor 21 (2.9) 3 (4.1) 0.48
Other non-breast primary
tumor
27 (3.7) 4 (5.4) 0.52
Death from breast cancer
a,b
9 (1.2) 1 (1.4) 1.00
Death from other cancer 0 (0.0) 0 (0.0)
Death from other causes 8 (1.1) 3 (4.1) 0.07
Death from missing causes 16 (2.2) 2 (2.7) 0.68
Overall mortality
Death from breast cancer
c
141 (19.4) 6 (8.1) 0.02
Death from other cancer
d
8 (1.1) 4 (5.4) \0.01
Death from other causes
d
9 (1.2) 3 (4.1) 0.09
Death from missing causes
d
17 (2.3) 2 (2.7) 0.69
Follow-up
Months, median (Q1–Q3) 68 (43–98) 72 (52–100) 0.62
Events were used for the calculation of cumulative incidence of
(a) breast cancer related events; (b) metastases; (c) breast-related
deaths; (d) deaths from causes other than breast cancer. Association
between number of events and BB use was tested by the Chi square or
the Fisher exact test, and the corresponding P value was reported
BB beta-blocker, Q1 lower quartile, Q3 upper quartile
Breast Cancer Res Treat (2013) 140:567–575 569
123
the Chi square test for trend, as appropriate. Differences in
median age and Ki-67 were tested using the non-parametric
median two sample test. Ki-67 was also dichotomized
using the median value of 40 % as cutpoint. For the cal-
culation of cumulative incidences, only first events of
interest were considered. For the cumulative incidence of
BC-related events, BC-related recurrences, and deaths
from BC were counted as events, while contralateral
tumors, non-breast primary tumors, and deaths from other
causes were considered as competing events. For the
cumulative incidence of metastases, metastases and deaths
from BC were counted as events, while loco-regional
events, contralateral tumors, non-breast primary tumors,
and deaths from other causes were considered as compet-
ing events. For the cumulative incidence BC-related
deaths, we considered deaths from other causes as com-
peting events. For the cumulative incidence non-BC
deaths, we considered deaths from BC as competing
events. Cumulative incidences were compared across dif-
ferent subgroups by means of the Gray test. Multivariable
Cox proportional hazards models were applied to evaluate
BB intake as independent prognostic factor, adjusting the
Years
Years
Years
Cumulative Incidence
0.0
0.1
0.2
0.3
0.4
0.5
01234567
Gray test P-value: 0.017
HR*
Beta-Blockers vs Beta-Blocker
: 1.79 (95% CI
0.82-3.90;
P-value
0.146)
* From multivariable analysis
At risk
BB 74 71 67 67 59 50 37 29
No BB 726 702 646 583 513 427 341 257
0.0
0.1
0.2
0.3
0.4
0.5
01234567
Gray test P-value: 0.017
HR*
Beta-Blockers vs Beta-Blocker
: 0.42 (95% CI 0.18-0.97; P-value 0.042)
* From multivariable analysis
At risk
BB 74 71 67 67 59 50 37 29
No BB 726 702 646 583 513 427 341 257
0.0
0.1
0.2
0.3
0.4
0.5
0
1234567
Gray test P-value: 0.011
HR*
Beta-Blockers vs Beta-Blocker
: 0.32 (95% CI 0.12-0.90; P-value 0.031)
* From multivariable analysis
At risk
BB 74 70 65 60 51 43 31 24
No BB 726 659 654 512 434 349 271 201
0.0
0.1
0.2
0.3
0.4
0.5
01234567
Beta-Blockers
Gray test P-value: 0.015
HR*
Beta-Blockers vs Beta-Blocker
: 0.52 (95% CI 0.28-0.97; P-value 0.041)
*From multivariable analysis
At risk
BB 74 70 65 60 51 43 31 24
No BB 726 659 654 512 434 349 271 201
No Beta-Blockers
Years
Cumulative Incidence
Cumulative Incidence
Cumulative Incidence
Beta-Blockers
No Beta-Blockers
Beta-Blockers
No Beta-Blockers
Beta-Blockers
No Beta-Blockers
a
b
c d
Fig. 1 a Cumulative incidence of breast cancer related events by use
of beta-blockers, b cumulative incidence of distant metastases by use
of beta-blockers, c cumulative incidence of deaths from breast cancer
by use of beta-blockers, d cumulative incidence of deaths from causes
other than breast cancer by use of beta-blockers
570 Breast Cancer Res Treat (2013) 140:567–575
123
effect of BB for all the variables that were associated in
univariate analysis with BB use or with the event of
interest. Hypertension was excluded from the multivariable
models in order to avoid multicollinearity with the use of
antihypertensive drugs. Adjusted hazard ratios (HR) with
95 % confidence intervals (CIs) were reported. All analy-
ses were carried out with SAS software (SAS Institute,
Cary, NC) and R software, version 2.12.2 (http://www.r-
project.org). Results associated with P values \0.05 were
considered statistical significant. All reported P values
were two-sided.
Results
At the time of cancer diagnosis, 74 women (9.3 %) out of
800 postmenopausal women were using BBs, while 726
(90.7 %) were not. Among the 74 users, 11 were taking
Carvedilol, 3 Sotalol, 27 Atenolol, 1 Betaxolol, 11 Biso-
prolol, 8 Metoprolol, and 13 Nebivolol. In Table 1 we
reported baseline characteristics in association with BB
use, as well as information on hypertension and use of
statins, antithrombotics and antihypertensive drugs other
than BBs. Median age was 62 years for the BB users and
59 years for the non-users (P = 0.015). BB users and non-
users were similarly distributed by body mass index, cancer
characteristics, and cancer treatments. Quadrantectomy
was performed in 617 (77.1 %) cases, 33 (5.3 %) of which
without radiotherapy. Details of adjuvant CT were as fol-
lows, respectively for BB users and non-users: 23 (31.1 %)
and 199 (27.4 %) received an anthracycline containing
regimen, 26 (35.1 %) and 283 (39.0 %) classical CMF, 15
(20.3 %) and 127 (17.5 %) another regimen, while 10
(13.5 %) and 117 (16.1 %) received no CT (P = 0.763).
As expected, the presence of hypertension and the use of
other antihypertensive drugs, antithrombotics and statins
were significantly more frequent in BBs users (P \ 0.01).
Median follow-up was 72 months (interquartile range
52–100) and 68 months (48–93) in BB users and non-users,
respectively (P = 0.62). Details on first events and
Table 3 Multivariable survival analysis
BC events Metastases BC deaths
HR (95 % CI) HR (95 % CI) HR (95 % CI)
Beta-blockers Use versus No use 0.52 (0.28–0.97) 0.32 (0.12–0.90) 0.42 (0.18–0.97)
Age 1-year increase 1.00 (0.99–1.02) 0.99 (0.97–1.02) 1.01 (0.99–1.03)
Primary tumor (T) 2 versus 1 1.37 (0.96–1.95) 1.28 (0.81–2.03) 1.26 (0.80–1.96)
3/4 versus 1 2.08 (1.11–3.91) 2.53 (1.13–5.63) 2.01 (0.95–4.23)
Positive lymph nodes 1–3 versus 0 1.20 (0.80–1.80) 1.47 (0.88–2.45) 1.77 (1.08–2.90)
[3 versus 0 2.49 (1.64–3.78) 2.33 (1.33–4.07) 2.91 (1.73–4.90)
Peritumoral vascular invasion Present versus absent 1.96 (1.41–2.73) 1.88 (1.22–2.89) 1.99 (1.35–2.94)
Antihypertensive drugs other than Beta-blockers Use versus No use 0.97 (0.69–1.38) 0.83 (0.51–1.35) 0.86 (0.56–1.32)
Antithrombotics Use versus No use 1.02 (0.46–2.28) 1.60 (0.60–4.22) 1.17 (0.45–3.05)
Statins Use versus No use 1.11 (0.66–1.88) 1.12 (0.55–2.24) 0.89 (0.43–1.85)
Response to neoadjuvant chemotherapy CR/PR versus No use 1.11 (0.65–1.90) 1.08 (0.56–2.1) 1.51 (0.81–2.83)
SD/PD versus No use 2.57 (1.49–4.45) 1.30 (0.61–2.76) 2.29 (1.21–4.34)
Loco-regional treatment QUAD versus QUART 3.29 (1.89–5.74) 3.34 (1.59–6.99) 2.99 (1.59–5.66)
MAST versus QUART 1.10 (0.75–1.61) 1.25 (0.76–2.05) 1.37 (0.87–2.14)
BC breast cancer, CR complete response after neoadjuvant chemotherapy, PR partial response after neoadjuvant chemotherapy, SD stable disease
after neoadjuvant chemotherapy, PD progressive disease after neoadjuvant chemotherapy, QUAD quadrantectomy, QUART quadrantectomy and
radiotherapy, MAST mastectomy
Cumulative Incidence
0.0
0.1
0.2
0.3
0.4
0.5
0 1 2 3 4 5 6 7
No hypertension
Hypertension
Gray test P-value: 0.444
Hypertension 266 249 214 190 157 125 95 70
No H
y
pertension 534 480 415 382 328 267 207 155
Years
Atrisk
Fig. 2 Cumulative incidence of breast cancer related events by
hypertension
Breast Cancer Res Treat (2013) 140:567–575 571
123
No Statins
Any Statins
Gray test P-value: 0.923
At risk
Any Statins 57 55 49 45 38 29 24 21
No Statins 743 675 580 528 449 363 278 205
No Antithrombotics
Any Antithrombotics
Gray test P-value: 0.864
At risk
Any Antithrombotics
25 21
21 16 14 10 7 3
No Antithrombotics 608 557 473 382 295 222
No Diuretics
Any Diuretics
Gray test P-value: 0.629
At risk
Any Diuretics 97 94 80 68 58 48 40 29
No Diuretics 703 635 549 505 429 344 262 196
No CCB
Any CCB
Gray test P-value: 0.342
At risk
Any CCB 62 58 51 44 38 24 18 16
No CCB 738 671 578 529 447 367 284 209
Years
Cumulative Incidence
0.0
0.1
0.2
0.3
0.4
0.5
No ARB
Any ARB
Gray test P-value: 0.812
At risk
Any ARB 51 49 40 36 26 21 15 9
No ARB 749 680 589 537 459 371 287 216
Years
Cumulative Incidence
0.0
0.1
0.2
0.3
0.4
0.5
01234567
01234567
No ACEI
Any ACEI
Gray test P-value: 0.920
At risk
Any ACEI 109 101 89 80 66 56 48 37
No ACEI 691 628 540 493 419 336 254 188
0.0
0.1
0.2
0.3
0.4
0.5
01234567
0.0
0.1
0.2
0.3
0.4
0.5
01234567
0.0
0.1
0.2
0.3
0.4
0.5
01 2 345 6 7
0.0
0.1
0.2
0.3
0.4
0.5
01234567
Cumulative Incidence
Cumulative Incidence
Cumulative Incidence
Cumulative Incidence
Years
Years
Years
Years
775
708
a
b
c
d
e
f
572 Breast Cancer Res Treat (2013) 140:567–575
123
mortality were reported in Table 2. Six (8.1 %) local
recurrences were observed in BB users and 52 (7.2 %) in
non-users (P = 0.77), while 1 (1.3 %) regional recurrence
in BB users and 28 (3.9 %) in non-users (P = 0.51). Three
(4.1 %) distant metastases were observed in BB users and
110 (15.2 %) non-users (P \ 0.01).
The 5-year cumulative incidence of BC-related events
was 13.6 % in BB users and 27.9 % in non-users
(P = 0.015; Fig. 1a). Multivariable analysis was reported
in Table 3. After adjusting for age, tumor stage, peritu-
moral vascular invasion, use of other antihypertensive
drugs, antithrombotics and statins, use of and response to
neoadjuvant chemotherapy, and loco-regional BC treat-
ment, the beneficial impact of BB use remained statistically
significant, with a HR of 0.52 (95 % CI 0.28–0.97;
P = 0.041). When analyzing the risk of distant metastases,
the HR adjusted for the same variables was 0.32 (95 % CI
0.12–0.90; P = 0.031; Fig. 1b) in favor of BBs. With
regard to mortality, 6 (8.1 %) BC-related deaths were
observed in BB users and 141 (19.4 %) in non-users, with a
corresponding adjusted HR of 0.42 (95 % CI 0.18–0.97;
P = 0.042; Fig. 1c). Causes of death other than cancer
were as follows: 2 circulatory system disease and 1
myelodysplastic syndrome in BB users; 2 circulatory sys-
tem disease, 1 pulmonary embolism, 1 head trauma, 1 old
age, 1 renal failure, and 3 causes different from BC not
better specified in BB non-users. At univariate analysis, the
risk of death from causes other than BC (other cancer,
other cause and missing cause) was higher in BB users
compared with non-users, but the difference was not sta-
tistically significant after the adjustment for age, antihy-
pertensive and antithrombotic drugs (HR: 1.79; 95 % CI
0.82–3.90; P value 0.146; Fig. 1d). Hypertension, antihy-
pertensive drugs other than BBs, antithrombotics, and
statins did not have any significant effect on the risk of BC-
related events (Figs. 2, 3).
In Supplementary Figure 1 we reported the effect of BB
use on BC-related events in various subgroups of patients.
BBs showed a protective effect in all subgroups (HR \ 1),
even if some HR estimates were not statistically significant,
possibly due to reduced numbers of events. No significant
interaction between BB use and any of the reported vari-
ables was identified. Finally, as shown in Supplementary
Table 1, in the subgroup of patients receiving neoadjuvant
CT, response to therapy was not statistically different by
BB intake.
Discussion
In this large series of postmenopausal women affected by
TNBC, we found that BB intake was associated with a
significantly decreased risk of BC recurrence and death.
Being the TNBC one of the most aggressive and difficult-
to-treat cancers, these findings might be of major clinical
relevance.
Mechanisms through which BB drugs could enhance
cancer patients’ prognosis were explained in preclinical
studies, through in vitro and in vivo models [1822]. The
sympathetic nervous system mediates the acute stress
response by regulating levels of norepinephrine and epi-
nephrine in organ tissues and circulating blood. Those two
stress hormones interfere with the tumor microenvironment
by modulating the function of several cancer-relevant cell
types, such as epithelial, myocytes and pericytes, adipo-
cytes, fibroblasts, neural and glial cells, and most lym-
phoid, and myeloid immune cells [22]. Ligation of
b-receptors by norepinephrine and epinephrine leads to
activation of Protein kinase A (PKA) which in turn regu-
lates a wide variety of cellular processes by activating
transcription factors and downstream kinases with conse-
quent modulation of cell trafficking and motility as well as
cellular resistance to apoptosis. In a preclinical study on
BC, norepinephrine was shown to be a potent inducer of
migratory activity [21]. Also, b-adrenergic antagonists
have been found to block stress-induced enhancement of
tumor progression and metastasis, while b-adrenergic
agonists have also been found to accelerate in vivo tumor
progression and metastasis in the absence of stress [22].
Therefore, BB drugs might have a role in weakening the
pro-migratory and pro-metastatic effects induced by stress
hormones.
Our findings on BB use in TNBC patients are in
agreement with studies reporting a beneficial prognostic
role of BBs in patients with prostate [2, 3], breast [47],
lung [8], and ovarian [9] cancer as well as cutaneous
melanoma [10, 11]. One study reported a null association
between BB use and risk of developing colorectal cancer
[23]. A very recent large study [24] reporting data from the
Danish Breast Cancer Cooperative Group registry showed
a protective effect of BB use on BC recurrence in univar-
iate analysis and a detrimental effect in multivariable
analysis. The switch from protective to detrimental effect
was mainly due to the adjustments for use of simvastatin
and ARB in the multivariable model. That switch was not
observed in our analysis. Unfortunately, the authors did not
Fig. 3 a Cumulative incidence of breast cancer related events by use
of angiotensin-converting enzyme inhibitor, b cumulative incidence
of breast cancer related events by use of angiotensin receptor blocker,
c cumulative incidence of breast cancer related events by use of
calcium channel blocker, d cumulative incidence of breast cancer
related events by use of diuretics, e cumulative incidence of breast
cancer related events by use of antithrombotics, f cumulative
incidence of breast cancer related events by use of statins. ACEI
angiotensin-converting enzyme inhibitor, ARB angiotensin receptor
blocker, CCB calcium channel blocker
b
Breast Cancer Res Treat (2013) 140:567–575 573
123
report results for the subgroups of TNBC patients. Since
the effect of BB on BC prognosis was reported to vary
according to BC molecular subtypes [4], the stratification
according to ER/PgR status as well as HER/neu status
might have possibly led to different conclusions. Finally, of
particular interest, Melhem-Bertrandt et al. [4] showed a
significant protective effect of BB use on relapse-free
survival in a series of 377 TNCB patients, with an adjusted
HR of 0.30 in favor of BB use. That finding was obtained
from a subgroup analysis performed on patients with
TNBC treated with neoadjuvant CT. In accordance to that
finding, when we limited the analysis to TNBC patients
who underwent neoadjuvant CT, we obtained an adjusted
HR of 0.36 in favor of BB (Supplementary Fig. 1). The
authors did not find any potential beneficial effect of BB on
prognosis in estrogen receptor-positive breast cancer
patients. The observed null effect could be due to the
presence of the ER and the use of ER targeting drugs that
may modulate the response to BB.
Interestingly, we did not observe fewer loco-regional
recurrences in BB users in comparison with non-users,
while we found a protective effect of BBs on the risk of
distant metastases and breast-specific mortality. This dif-
ferential effect of BBs on different types of recurrence
could be due to the fact that stress hormone norepinephrine
was shown to be a potent inducer of migratory activity,
with no or little effect on primary tumor growth [21].
Hypothetically, this mechanism might also explain why we
did not find any significant difference between BB users
and non-users in the response to pre-surgical CT. Our
findings on response to pre-surgical CT are consistent with
those reported in the paper by Melhem-Bertrandt et al. [4].
The main strengths of our study are the large size of the
population and its homogeneity, given by the selection of
early primary TNBC diagnosed and operated in one single
institution. An additional strong point of the study was that
BB users and non-users were uniformly distributed by
tumor characteristics and cancer treatments. Also, the
inclusion of TNBC only prevented possible biases due to
interaction between BBs and hormone or anti-HER2/neu
therapies. Moreover, for the analyses of events, we used
competing risk models to take into account the different
types of recurrences as well as the different causes of
death, and these analyses allowed us to highlight the dif-
ferential effect of BBs on different types of recurrence and
the specific effect of BBs on deaths due to BC rather than
deaths due to other causes.
Despite these strengths, the present study has some clear
limitations, apart from the ones linked to the retrospective
design. First of all, we could not retrieve information on
BB use before and after the diagnosis of TNBC. Therefore,
patients that were using BBs at the time of diagnosis might
have quit the medication after BC diagnosis, possibly
switching to different antihypertensive drugs; on the other
hand, patients who were not currently using BB at TNBC
diagnosis, could have started after. Also, the association
between duration of BB use and prognosis could not be
determined. Moreover, possibly not all the patients
receiving BBs at BC diagnosis were correctly identified,
and this would likely have lead to a dilution of the risk
estimates. However, our proportion of BB users was sim-
ilar to the ones reported in other groups of postmenopausal
BC patients [46]. Another potential flaw is that BB users
were older than non-users. It is to say, though, that 3 years
difference in median age has no or little clinical relevance
and that all the reported analyses were adjusted for age.
In conclusion, in this series of women affected by
TNBC, BB intake was associated with a significantly
decreased risk of BC recurrence and death. On the basis of
all the extensive preclinical and clinical evidence, and
given the limited number of treatment options in TNBC
patients, we believe that innovative therapeutic strategies,
including BBs, should be urgently explored.
Conflict of interest None.
References
1. Ram CV (2010) Beta-blockers in hypertension. Am J Cardiol
106(12):1819–1825
2. Grytli HH, Fagerland MW, Fossa
˚
SD et al (2013) Association
between use of b-blockers and prostate cancer-specific survival: a
cohort study of 3561 prostate cancer patients with high-risk or
metastatic disease. Eur Urol 64(1):e11–e12
3. Grytli HH, Fagerland MW, Fossa
˚
SD et al (2013) Use of
b-blockers is associated with prostate cancer-specific survival in
prostate cancer patients on androgen deprivation therapy. Prostate
73(3):250–260
4. Melhem-Bertrandt A, Chavez-Macgregor M, Lei X et al (2011)
Beta-blocker use is associated with improved relapse-free sur-
vival in patients with triple-negative breast cancer. J Clin Oncol
29(19):2645–2652
5. Barron TI, Connolly RM, Sharp L et al (2011) Beta blockers and
breast cancer mortality: a population-based study. J Clin Oncol
29(19):2635–2644
6. Powe DG, Voss MJ, Za
¨
nker KS et al (2010) Beta-blocker drug
therapy reduces secondary cancer formation in breast cancer and
improves cancer specific survival. Oncotarget 1(7):628–638
7. Ganz PA, Habel LA, Weltzien EK et al (2011) Examining the
influence of beta blockers and ACE inhibitors on the risk for
breast cancer recurrence: results from the LACE cohort. Breast
Cancer Res Treat 129(2):549–556
8. Wang HM, Liao ZX, Komaki R et al (2013) Improved survival
outcomes with the incidental use of beta-blockers among patients
with non-small-cell lung cancer treated with definitive radiation
therapy. Ann Oncol 24(5):1312–1319
9. Diaz ES, Karlan BY, Li AJ (2012) Impact of beta blockers on
epithelial ovarian cancer survival. Gynecol Oncol 127(2):
375–378
10. De Giorgi V, Grazzini M, Gandini S et al (2011) Treatment with
b-blockers and reduced disease progression in patients with thick
melanoma. Arch Intern Med 171(8):779–781
574 Breast Cancer Res Treat (2013) 140:567–575
123
11. Lemeshow S, Sørensen HT, Phillips G et al (2011) b-Blockers
and survival among Danish patients with malignant melanoma: a
population-based cohort study. Cancer Epidemiol Biomarkers
Prev 20(10):2273–2279
12. Monami M, Filippi L, Ungar A et al (2013) Further data on beta-
blockers and cancer risk: observational study and meta-analysis
of randomized clinical trials. Curr Med Res Opin 29(4):369–378
13. Perron L, Bairati I, Harel F et al (2004) Antihypertensive drug
use and the risk of prostate cancer (Canada). Cancer Causes
Control 15(6):535–541
14. Powe DG, Entschladen F (2011) Targeted therapies: using b-
blockers to inhibit breast cancer progression. Nat Rev Clin Oncol
8(9):511–512
15. Antoni MH, Lutgendorf SK, Cole SW et al (2006) The influence
of bio-behavioural factors on tumour biology: pathways and
mechanisms. Nat Rev Cancer 6(3):240–248
16. Cole SW, Sood AK (2012) Molecular pathways: beta-adrenergic
signaling in cancer. Clin Cancer Res 18(5):1201–1206
17. Schuller HM (2010) Beta-adrenergic signaling, a novel target for
cancer therapy? Oncotarget 1(7):466–469
18. Campbell JP, Karolak MR, Ma Y et al (2012) Stimulation of host
bone marrow stromal cells by sympathetic nerves promotes breast
cancer bone metastasis in mice. PLoS Biol 10(7):e1001363
19. Benish M, Bartal I, Goldfarb Y et al (2008) Perioperative use of
beta-blockers and COX-2 inhibitors may improve immune
competence and reduce the risk of tumor metastasis. Ann Surg
Oncol 15(7):2042–2052
20. Drell TL 4th, Joseph J, Lang K et al (2003) Effects of neuro-
transmitters on the chemokinesis and chemotaxis of MDA-MB-
468 human breast carcinoma cells. Breast Cancer Res Treat
80(1):63–70
21. Sloan EK, Priceman SJ, Cox BF et al (2010) The sympathetic
nervous system induces a metastatic switch in primary breast
cancer. Cancer Res 70(18):7042–7052
22. Daly CJ, McGrath JC (2011) Previously unsuspected widespread
cellular and tissue distribution of b-adrenoceptors and its rele-
vance to drug action. Trends Pharmacol Sci 32:219–226
23. Jansen L, Below J, Chang-Claude J et al (2012) Beta blocker use
and colorectal cancer risk: population-based case–control study.
Cancer 118(16):3911–3919
24. Sørensen GV, Ganz PA, Cole SW et al (2013) Use of b-blockers,
angiotensin-converting enzyme inhibitors, angiotensin II receptor
blockers, and risk of breast cancer recurrence: a Danish nation-
wide prospective cohort study. J Clin Oncol 31(18):2265–2272
Breast Cancer Res Treat (2013) 140:567–575 575
123
... Beta-blockers have been identified as potential repurposing candidates for cancer due to emerging evidence from preclinical studies (Sloan et al., 2010;Cole and Sood, 2012;Magnon et al., 2013;Creed et al., 2015;Chang et al., 2016;Le et al., 2016), retrospective observational studies across various cancer types (e.g., melanoma , ovarian cancer (Watkins et al., 2015;Couttenier et al., 2019), colorectal cancer (Jansen et al., 2014), prostate cancer (Lu et al., 2015;Malik et al., 2023), and breast cancer (Botteri et al., 2013;Lofling et al., 2022)), and recent prospective clinical trials (Shaashua et al., 2017;Hiller et al., 2020). Despite advances in breast cancer therapeutics, metastasis remains the key prognostic marker for poor survival and the main cause of death (Weigelt et al., 2005;Jung et al., 2011). ...
... However, a metaanalysis of epidemiological and perioperative studies by Yap et al. (2018) raised the possibility that the effects of beta-blockers on cancer outcomes may be tumor type-specific, as significant benefits were observed only in melanoma and ovarian cancer (Yap et al., 2018). Several retrospective observational studies found that betablocker use is associated with improved cancer outcomes in breast cancer (Powe et al., 2010;Barron et al., 2011;Melhem-Bertrandt et al., 2011;Botteri et al., 2013;Sorensen et al., 2013;Choy et al., 2016;Gillis et al., 2021;Lofling et al., 2022), while other studies have reported no significant association (Cardwell et al., 2013;Ganz et al., 2011;Sakellakis et al., 2014;Modi et al., 2020-;Cardwell et al., 2016). Moreover, meta-analyses by Løfling et al. (2022), Caparica et al. (2021) and Spini et al. (2019) have demonstrated more consistent protective effects of beta-blockers amongst women with triple negative breast cancer (TNBC) (Spini et al., 2019;Caparica et al., 2021;Lofling et al., 2022). ...
... A recent meta-analysis of four independent studies by Løfling et al. (2022) reported a 26% reduced risk of breast cancer death in women with TNBC with the use of beta blockers compared to no use (Lofling et al., 2022). For example, Botteri et al. (2013) observed a 48% reduced risk of breast cancer death in women with TNBC who used any beta-blockers (n = 74) over 5 years (Botteri et al., 2013). Løfling et al. (2022) demonstrated a 34% improved breast cancer-survival with beta blocker use in women with TNBC (n = 312) over 5 years (Lofling et al., 2022). ...
Article
Full-text available
Introduction: Cancer registries and hospital electronic medical records are commonly used to investigate drug repurposing candidates for cancer. However, administrative data are often more accessible than data from cancer registries and medical records. Therefore, we evaluated if administrative data could be used to evaluate drug repurposing for cancer by conducting an example study on the association between beta-blocker use and breast cancer mortality. Methods: A retrospective cohort study of women aged ≥50 years with incident breast cancer was conducted using a linked dataset with statewide hospital admission data and nationwide medication claims data. Women receiving beta blockers and first-line anti-hypertensives prior to and at diagnosis were compared. Breast cancer molecular subtypes and metastasis status were inferred by algorithms from commonly prescribed breast cancer antineoplastics and hospitalization diagnosis codes, respectively. Subdistribution hazard ratios (sHR) and corresponding 95% confidence intervals (CIs) for breast cancer mortality were estimated using Fine and Gray’s competing risk models adjusted for age, Charlson comorbidity index, congestive heart failure, myocardial infraction, molecular subtype, presence of metastasis at diagnosis, and breast cancer surgery. Results: 2,758 women were hospitalized for incident breast cancer. 604 received beta-blockers and 1,387 received first-line antihypertensives. In total, 154 breast cancer deaths were identified over a median follow-up time of 2.7 years. We found no significant association between use of any beta-blocker and breast-cancer mortality (sHR 0.86, 95%CI 0.58–1.28), or when stratified by beta-blocker type (non-selective, sHR 0.42, 95%CI 0.14–1.25; selective, sHR 0.95, 95%CI 0.63–1.43). Results were not significant when stratified by molecular subtypes (e.g., triple negative breast cancer (TNBC), any beta blocker, sHR 0.16, 95%CI 0.02–1.51). Discussion: It is possible to use administrative data to explore drug repurposing opportunities. Although non-significant, an indication of an association was found for the TNBC subtype, which aligns with previous studies using registry data. Future studies with larger sample size, longer follow-up are required to confirm the association, and linkage to clinical data sources are required to validate our methodologies.
... A total of 489 studies (after removing duplicates) were retrieved from the selected databases during the literature search and included for initial screening (Fig. 1). 132 reports were assessed in full, and after making various exclusions, 24 studies were deemed to meet the inclusion criteria and were included in this meta-analysis [23,24,36,37,[44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63]. The detailed characteristics of these 24 studies are shown in Table 1. ...
... Overall, this meta-analysis included 24 studies [23,24,36,37,[44][45][46][47][48][49][50][51][52][53][54][55][56][57][58][59][60][61][62][63] with a combined total of 253,082 women with breast cancer. Nineteen of these studies assessed BCD as an outcome, while sixteen assessed BCR as an outcome (eight of these studies assessed both outcomes). ...
Article
Full-text available
Purpose Beta blockers (BBs) are commonly used cardiovascular medications, and their association with breast cancer outcomes has been examined in several previous observational studies and meta-analyses. In this study, an updated meta-analysis was undertaken to ascertain the association between BBs and both breast cancer death (BCD) and breast cancer recurrence (BCR). Methods Articles were sourced from various databases up until the 14th of August 2023. Effect estimates were pooled using the random effects model, and the Higgins I² statistic was computed to ascertain heterogeneity. Subgroup analyses were conducted by the potential for immortal time bias (ITB), the exposure period (prediagnosis vs postdiagnosis), and type of BB (selective vs non-selective). Publication bias was assessed using funnel plots and Egger’s regression tests. Results Twenty-four studies were included. Pooled results showed that there was no statistically significant association between BB use and both BCD (19 studies, hazard ratio = 0.90, 95% CI 0.78–1.04) and BCR (16 studies, HR = 0.87, 95% CI 0.71–1.08). After removing studies with ITB, the associations were attenuated towards the null. There was no effect modification for either outcome when stratifying by the exposure period or type of BB. There was clear evidence of publication bias for both outcomes. Conclusion In this meta-analysis, we found no evidence of an association between BB use and both BCD and BCR. Removing studies with ITB attenuated the associations towards the null, but there was no effect modification by the exposure period or type of BB.
... To date, several retrospective studies as well as two metaanalyses concerning the impact of beta-blocker exposure in breast cancer patients have revealed conflicting [9,[15][16][17][18][19][20][21][22][23] findings. Of note, the majority of these studies included patients with both advanced and early disease stages without specific stratification. ...
Article
Full-text available
Background Retrospective observational studies suggest a potential role of beta-blockers as a protective strategy against progression and metastasis in invasive breast cancer. In this context, we investigated the impact of beta-blocker exposure on risk for progression to invasive breast cancer after diagnosis of ductal cancer in situ (DCIS). Methods The retrospective study population included 2535 women diagnosed with pure DCIS between 2006 and2012 in three healthcare regions in SwedenExposure to beta-blocker was quantified using a time-varying percentage of days with medication available. The absolute risk was quantified using cumulative incidence functions and cox models were applied to quantify the association between beta-blocker exposure and time from DCIS diagnosis to invasive breast cancer, accounting for delayed effects, competing risks and pre-specified confounders. Results The median follow-up was 8.7 years. One third of the patients in our cohort were exposed to beta-blockers post DCIS diagnosis. During the study period, 48 patients experienced an invasive recurrence, giving a cumulative incidence of invasive breast cancer progression of 1.8% at five years. The cumulative exposure to beta-blocker was associated with a reduced risk in a dose-dependent manner, though the effect was not statistically significant. Conclusion Our observational study is suggestive of a protective effect of beta-blockers against invasive breast cancer after primary DCIS diagnosis. These results provide rationales for experimental and clinical follow-up studies in carefully selected DCIS groups.
... This effect was found with very high doses of paclitaxel, which justifies the differences found between their results and those shown herein [40]. Propranolol has been described to have a beneficial effect in particular on triple-negative breast cancer patients [41][42][43]. Moreover, in a very recent work, the implication of a neuromodulatory effect of anthracyclines showed to undermine the potential therapeutic impact. ...
Article
Full-text available
Purpose Globally breast cancer accounts for 24.5% in incidence and 15.5% in cancer deaths in women. The triple-negative subtype lacks any specific therapy and is treated with chemotherapy, resulting in significant side-effects. We aimed to investigate if the dose of chemotherapeutic drugs could be diminished by co-administering it with the β2-agonist salbutamol. Methods Cell proliferation was measured by thymidine incorporation; gene expression, by real-time PCR and protein phosphorylation by WB. Apoptosis was assessed by acridine orange / ethidium bromide and TUNEL tests. Public patient databases were consulted. Cells were inoculated to nude mice and their growth assessed. Results The β2-agonist salbutamol synergizes in MDA-MB-231 cells in vitro with paclitaxel and doxorubicin on cell proliferation through ADRB2 receptors, while the β-blocker propranolol does not. The expression of this receptor was assessed in patient databases and other cell lines. Triple negative samples had the lowest expression. Salbutamol and paclitaxel decreased MDA-MB-231 cell proliferation while their combination further inhibited it. The pathways involved were analyzed. When these cells were inoculated to nude mice, paclitaxel and salbutamol inhibited tumor growth. The combined effect was significantly greater. Paclitaxel increased the expression of MDR1 while salbutamol partially reversed this increase. Conclusion While the effect of salbutamol was mainly on cell proliferation, suboptimal concentrations of paclitaxel provoked a very important enhancement of apoptosis. The latter enhanced transporter proteins as MDR1, whose expression were diminished by salbutamol. The expression of ADRB2 should be assessed in the biopsy or tumor to eventually select patients that could benefit from salbutamol repurposing.
... 11 There is conflicting evidence regarding the effect of BB use on clinical outcomes in patients with breast cancer. Studies have suggested that the use of BBs in patients with breast cancer can reduce the risk of recurrence or mortality [12][13][14][15][16] ; however, some other related studies have suggested the contrary. 10,[17][18][19][20][21][22] Moreover, some meta-analyses have been journals.sagepub.com/home/taw ...
Article
Full-text available
Background: Clinical trials investigating the effects of beta-blockers (BBs) on cancer are underway. Evidence from preclinical research suggests that BBs could serve as anticancer agents and immune boosters. There is conflicting evidence regarding the effect of BB use on clinical outcomes in patients with breast cancer. Objectives: The study aimed to determine whether BB use is associated with progression-free survival (PFS) and overall survival (OS) in patients receiving anti-human epidermal growth factor receptor 2 (HER2) treatment for advanced breast cancer. Design: Retrospective hospital-based study. Methods: The participants enrolled were breast cancer patients with advanced HER2-positive status who initiated trastuzumab monotherapy or concomitant therapy with trastuzumab and any dose of BB. The patients were enrolled between January 2012 and May 2021 and divided into three groups based on whether they received a BB or not in the therapeutic regimen: BB-/trastuzumab+, BB+ (non-selective)/trastuzumab+, and BB+ (selective)/trastuzumab+. PFS and OS were the primary and secondary endpoints, respectively. Results: The estimated median PFS in the BB-/trastuzumab+, BB+ (non-selective)/trastuzumab+, and BB+ (selective)/trastuzumab+ groups was 51.93, 21.50, and 20.77 months, respectively. The corresponding OS was 56.70, 29.10, and 27.17 months. The intergroup differences in these durations were significant. Both PFS [adjusted hazard ratio (HR): 2.21, 95% confidence interval (CI): 1.56-3.12; p < 0.001]) and OS (adjusted HR: 2.46, 95% CI: 1.69-3.57; p < 0.001) were worse when BBs were used. Conclusion: Our study provides important evidence that BB use potentially has a negative effect on patients with HER2-positive advanced breast cancer. Nevertheless, despite the study's results, cardiovascular disease (CVD) should be appropriately treated in patients with HER2-positive advanced breast cancer. Other types of drugs can be used to treat CVD, but BB use should be avoided. Large real-world database and prospective studies should be conducted to validate the results of this study.
Article
Full-text available
Objective Breast cancer is one of the most common causes of death among women. Statins, typically used for cholesterol management, have been hypothesized to reduce recurrence and mortality rates in breast cancer. However, this association remains a subject of debate. This study evaluates the potential impact of statins on breast cancer recurrence and mortality. Methods A comprehensive search was conducted in the PubMed, EMBASE, and Cochrane databases for articles published up to June 2023. These articles examined the effect of statins on breast cancer recurrence and mortality both before and after diagnosis. The analysis was performed using random-effects models, calculating pooled hazard ratios (HR) and their 95% confidence intervals (CI). Results A total of 31 cohort studies, involving 261,834 female breast cancer patients, were included in this analysis. It was found that statin use prior to diagnosis was associated with a decrease in overall mortality (HR, 0.8; 95% CI, 0.69–0.93; I2 = 77.6%; P = 0.001) and breast cancer-specific mortality (HR, 0.76; 95% CI, 0.67–0.87; I2 = 72.7%; P = 0.005). Additionally, statin use after diagnosis was observed to reduce the recurrence of breast cancer (HR, 0.71; 95% CI, 0.61–0.82; I2 = 60%; P = 0.003), overall mortality (HR, 0.81; 95% CI, 0.70–0.92; I2 = 80.7%; P < 0.001), and breast cancer-specific mortality (HR, 0.76; 95% CI, 0.67–0.86; I2 = 74.5%; P < 0.001). Conclusions The findings of this study indicate that statin usage, both before and after breast cancer diagnosis, may be associated with reduced risks of overall and breast cancer-specific mortality, as well as lower recurrence rates.
Article
Full-text available
Triple-negative breast cancer (TNBC) is known for its heterogeneous complexity and is often difficult to treat. TNBC lacks the expression of major hormonal receptors like estrogen receptor, progesterone receptor, and human epidermal growth factor receptor-2 and is further subdivided into androgen receptor (AR) positive and AR negative. In contrast, AR negative is also known as quadruple-negative breast cancer (QNBC). Compared to AR-positive TNBC, QNBC has a great scarcity of prognostic biomarkers and therapeutic targets. QNBC shows excessive cellular growth and proliferation of tumor cells due to increased expression of growth factors like EGF and various surface proteins. This study briefly reviews the limited data available as protein biomarkers that can be used as molecular targets in treating TNBC as well as QNBC. Targeted therapy and immune checkpoint inhibitors have recently changed cancer treatment. Many studies in medicinal chemistry continue to focus on the synthesis of novel compounds to discover new antiproliferative medicines capable of treating TNBC despite the abundance of treatments currently on the market. Drug repurposing is one of the therapeutic methods for TNBC that has been examined. Moreover, some additional micronutrients, nutraceuticals, and functional foods may be able to lower cancer risk or slow the spread of malignant diseases that have already been diagnosed with cancer. Finally, nanomedicines, or applications of nanotechnology in medicine, introduce nanoparticles with variable chemistry and architecture for the treatment of cancer. This review emphasizes the most recent research on nutraceuticals, medication repositioning, and novel therapeutic strategies for the treatment of TNBC.
Chapter
In human, the nervous system governs vital functional activities of many organs. As tumor is a systemic disease, it does not grow as an independent entity, rather, tumor growth and progression is an integral part of the nervous system. The peripheral nervous system (PNS) is a system of autonomic and sensory nerves outside the central nervous system that helps to integrate and regulate physiological processes at molecular, cellular, and organ level to maintain homeostasis. Indicators of homeostasis, such as blood pressure, pH, and metabolism, are constantly adjusted by the PNS in response to internal and external stimuli. Tumors, likewise, must control these neuronal mechanisms in order to survive. An increasing amount of data implies that tumors adopt and reprogram neuronal interactions to support their development and progression, in a way similar to how healthy tissues recruit and sustain PNS innervation. In this chapter, we highlight the importance of the crosstalk between host PNS system and cancer growth.
Article
Full-text available
Background Preclinical studies have shown that norepinephrine can directly stimulate tumor cell migration and that this effect is mediated by the beta-adrenergic receptor.Patients and methodsWe retrospectively reviewed 722 patients with non-small-cell lung cancer (NSCLC) who received definitive radiotherapy (RT). A Cox proportional hazard model was utilized to determine the association between beta-blocker intake and locoregional progression-free survival (LRPFS), distant metastasis-free survival (DMFS), disease-free survival (DFS), and overall survival (OS).ResultsIn univariate analysis, patients taking beta-blockers (n = 155) had improved DMFS (P < 0.01), DFS (P < 0.01), and OS (P = 0.01), but not LRPFS (P = 0.33) compared with patients not taking beta-blockers (n = 567). In multivariate analysis, beta-blocker intake was associated with a significantly better DMFS [hazard ratio (HR), 0.67; P = 0.01], DFS (HR, 0.74; P = 0.02), and OS (HR, 0.78; P = 0.02) with adjustment for age, Karnofsky performance score, stage, histology type, concurrent chemotherapy, radiation dose, gross tumor volume, hypertension, chronic obstructive pulmonary disease and the use of aspirin. There was no association of beta-blocker use with LRPFS (HR = 0.91, P = 0.63).Conclusion Beta-blocker use is associated with improved DMFS, DFS, and OS in this large cohort of NSCLC patients. Future prospective trials can validate these retrospective findings and determine whether the length and timing of beta-blocker use influence survival outcomes.
Article
Full-text available
Bone and lung metastases are responsible for the majority of deaths in patients with breast cancer. Following treatment of the primary cancer, emotional and psychosocial factors within this population precipitate time to recurrence and death, however the underlying mechanism(s) remain unclear. Using a mouse model of bone metastasis, we provide experimental evidence that activation of the sympathetic nervous system, which is one of many pathophysiological consequences of severe stress and depression, promotes MDA-231 breast cancer cell colonization of bone via a neurohormonal effect on the host bone marrow stroma. We demonstrate that induction of RANKL expression in bone marrow osteoblasts, following β2AR stimulation, increases the migration of metastatic MDA-231 cells in vitro, independently of SDF1-CXCR4 signaling. We also show that the stimulatory effect of endogenous (chronic stress) or pharmacologic sympathetic activation on breast cancer bone metastasis in vivo can be blocked with the β-blocker propranolol, and by knockdown of RANK expression in MDA-231 cells. These findings indicate that RANKL promotes breast cancer cell metastasis to bone via its pro-migratory effect on breast cancer cells, independently of its effect on bone turnover. The emerging clinical implication, supported by recent epidemiological studies, is that βAR-blockers and drugs interfering with RANKL signaling, such as Denosumab, could increase patient survival if used as adjuvant therapy to inhibit both the early colonization of bone by metastatic breast cancer cells and the initiation of the "vicious cycle" of bone destruction induced by these cells.
Article
PURPOSETo estimate associations between use of β-blockers, angiotensin-converting enzyme (ACE) inhibitors, or angiotensin receptor blockers (ARBs) and breast cancer recurrence in a large Danish cohort. PATIENTS AND METHODS We enrolled 18,733 women diagnosed with nonmetastatic breast cancer between 1996 and 2003. Patient, treatment, and 10-year recurrence data were ascertained from the Danish Breast Cancer Cooperative Group registry. Prescription and medical histories were ascertained by linkage to the National Prescription Registry and Registry of Patients, respectively. β-Blocker exposure was defined in aggregate and according to solubility, receptor selectivity, and individual drugs. ACE inhibitor and ARB exposures were defined in aggregate. Recurrence associations were estimated with multivariable Cox regression models in which time-varying drug exposures were lagged by 1 year.ResultsCompared with never users, users of any β-blocker had a lower recurrence hazard in unadjusted models (unadjusted hazard ratio [HR] = 0.91; 95% CI, 0.81 to 1.0) and a slightly higher recurrence hazard in adjusted models (adjusted HR = 1.3; 95% CI, 1.1 to 1.5). Associations were similar for exposures defined by receptor selectivity and solubility. Although most individual β-blockers showed no association with recurrence, metoprolol and sotalol were associated with increased recurrence rates (adjusted metoprolol HR = 1.5, 95% CI, 1.2 to 1.8; adjusted sotalol HR = 2.0, 95% CI, 0.99 to 4.0). ACE inhibitors were associated with a slightly increased recurrence hazard, whereas ARBs were not associated with recurrence (adjusted ACE inhibitor HR = 1.2, 95% CI, 0.97 to 1.4; adjusted ARBs HR = 1.1, 95% CI, 0.85 to 1.3). CONCLUSION Our data do not support the hypothesis that β-blockers attenuate breast cancer recurrence risk.
Article
Background: The aim of the present paper is to provide some further data on the relationship between β-blocker treatment and the incidence of cancer, using two different approaches (epidemiological study and meta-analysis of clinical trials). Methods: In a consecutive series of 1340 diabetic patients starting insulin therapy, 112 cases of cancer during a mean follow-up of 75.9 months were identified as first hospital admission or death. For each case, the controls were chosen randomly from those members of the cohort matched for age, sex and BMI. The main predefined analysis was the comparison of cases and controls for length of exposure to β-blockers and proportion of patients exposed using a conditional logistic regression which takes into account the matching structure. For the meta-analytic sub-study, an extensive search of Medline and the Cochrane Library (any date up to December 31st, 2011) was performed for all trials in which a β-blocker was used. Mantel-Haenszel Odds Ratios (MH-OR) with 95% confidence intervals for incident malignancies were calculated using a random effect model. Results: After adjusting for mean daily dose of glargine and metformin, and ischemic heart disease, exposure to β-blockers was associated with a reduced overall risk of cancer (HR 0.33 [0.13; 0.83], p = 0.019; HR for each month of exposure 0.87 [0.77; 0.98], p = 0.025). In the meta-analysis sub-study, performed on nine trials, β-blockers were associated with a non-significant trend toward lower risk of cancer (MH-OR 0.93 [0.86; 1.01], p = 0.070). Study limitation: Limitations of the observational study are the small sample size that limits the statistical power of analyses, that it was performed on diabetic patients only, and that diagnoses of malignancies were derived from administrative data. Conclusions: In conclusion, this research seem to confirm a possible beneficial effect of β-blockers against the risk of cancer development.
Article
Background: We recently reported reduced prostate cancer (PCa)-specific mortality for β-blocker users among patients receiving androgen-deprivation therapy in a health survey cohort including 655 PCa patients. Information on clinical characteristics was limited. Objective: To assess the association between β-blockers and PCa-specific mortality in a cohort of 3561 prostate cancer patients with high-risk or metastatic disease, and to address potential confounding from the use of statins or acetylsalicylic acid (ASA). Design, setting, and participants: Clinical information from all men reported to the Cancer Registry of Norway with a PCa diagnosis between 2004 and 2009 (n=24 571) was coupled with information on filled prescriptions between 2004 and 2011 from the Norwegian Prescription Database. Exclusion criteria were low- or intermediate-risk disease; planned radiotherapy or radical prostatectomy; initiation of β-blocker, ASA, or statin use after diagnosis where applicable; missing information on baseline Gleason score, prostate-specific antigen level, T stage or performance status; and missing follow-up. Outcome measurements and statistical analysis: Cox proportional hazards modelling and competing risk regression modelling were used to analyse the effects of β-blocker use on all-cause and PCa-specific mortality, respectively. Differences between β-blocker users and nonusers regarding baseline clinical characteristics were assessed by the Wilcoxon-Mann-Whitney U test, Pearson chi-square test, and Student t test. Results and limitations: Median follow-up was 39 mo. β-Blocker use was associated with reduced PCa mortality (adjusted subhazard ratio: 0.79; 95% confidence interval [CI], 0.68-0.91; p value: 0.001). The observed reduction in PCa mortality was independent of the use of statins or ASA. We observed no association with all-cause mortality (adjusted hazard ratio: 0.92; 95% CI, 0.83-1.02). The main limitations of the study were the observational study design and short follow-up. Conclusions: β-Blocker use was associated with reduced PCa-specific mortality in patients with high-risk or metastatic disease at the time of diagnosis. Our findings need validation from further observational studies.
Article
Background: Experimental evidence suggests a role for the β(2) -adrenergic receptor pathway in prostate cancer (PCa). We have investigated the association of β-blocker use with PCa incidence and survival in a Norwegian cohort. Methods: Data from the Oslo II study in 2000 (n = 6515) were linked with information from the Cancer Registry of Norway and Statistics Norway. PCa risk and overall- and PCa-specific mortality were analyzed using uni- and multi-variable Cox- and competing risk regression models. Results: At baseline, 776 men (11.9%) reported using a β-blocker. 212 men (3.3%) were diagnosed with PCa before the survey, leaving 6,303 eligible for incidence analysis. During a median follow-up of 122 months, 448 (7.1%) men were diagnosed with PCa. β-blocker use was not associated with PCa risk [hazard ratio (HR): 1.05, 95% CI: 0.79-1.40]. For all patients (n = 655; including med diagnosed before the survey), β-blocker use was not associated with PCa-specific mortality (HR: 0.55, 95% CI 0.24-1.26, P = 0.16). However, in the subgroup of men planned to receive androgen deprivation therapy (ADT), as reported to the Cancer Registry (n = 263), β-blocker use was associated with reduced PCa-specific mortality (HR: 0.14, 95% CI 0.02-0.85, P = 0.032). No effect on overall mortality was seen (HR, all patients: 0.88, 95% CI 0.56-1.38, P = 0.57). β-blocker use did not appear to affect PSA level, Gleason score, or T-stage at diagnosis; however, these variables were missing for many cases. Conclusions: Our findings demonstrate a possible benefit of β-blocker use for men treated with ADT, suggesting the need for investigation in larger cohorts.
Article
Stress may promote ovarian cancer progression through mechanisms including autonomic nervous system mediators such as norepinephrine and epinephrine. Beta blockers, used to treat hypertension, block production of these adrenergic hormones, and have been associated with prolonged survival in several malignancies. We sought to determine the association between beta blocker use and epithelial ovarian cancer (EOC) disease progression and survival. We performed an institutional retrospective review of patients with EOC treated between 1996 and 2006. Patients underwent cytoreductive surgery followed by platinum-based chemotherapy. Women were considered beta blocker users if these medications were documented on at least two records more than 6months apart. Statistical tests included Fisher's exact, Kaplan-Meier, and Cox regression analyses. 248 met inclusion criteria. 68 patients used antihypertensives, and 23 used beta blockers. Median progression-free survival for beta blocker users was 27months, compared with 17months for non-users (p=0.05). Similarly, overall disease-specific survival was longer for beta blocker users (56months) compared with non-users (48months, p=0.02, hazard ratio=0.56). Multivariate analysis identified beta blocker use as an independent positive prognostic factor, after controlling for age, stage, grade, and cytoreduction status (p=0.03). Overall survival remained longer for beta blocker users (56months) when compared with hypertensive patients on other medications (34months) and patients without hypertension (51months) (p=0.007). In this cohort of patients with EOC, beta blocker use was associated with a 54% reduced chance of death compared with that of non-users.